NZ614427B2 - Use of inhibitors of egfr-family receptors in the treatment of hormone refractory breast cancers - Google Patents
Use of inhibitors of egfr-family receptors in the treatment of hormone refractory breast cancers Download PDFInfo
- Publication number
- NZ614427B2 NZ614427B2 NZ614427A NZ61442712A NZ614427B2 NZ 614427 B2 NZ614427 B2 NZ 614427B2 NZ 614427 A NZ614427 A NZ 614427A NZ 61442712 A NZ61442712 A NZ 61442712A NZ 614427 B2 NZ614427 B2 NZ 614427B2
- Authority
- NZ
- New Zealand
- Prior art keywords
- antibody
- erbb3
- tumor
- seq
- estrogen
- Prior art date
Links
- 230000002401 inhibitory effect Effects 0.000 title claims description 79
- 239000003112 inhibitor Substances 0.000 title claims description 67
- 206010006187 Breast cancer Diseases 0.000 title description 61
- 229940088597 Hormone Drugs 0.000 title description 49
- 239000005556 hormone Substances 0.000 title description 49
- 102000004965 antibodies Human genes 0.000 claims abstract description 146
- 108090001123 antibodies Proteins 0.000 claims abstract description 146
- 108010038795 estrogen receptors Proteins 0.000 claims abstract description 51
- 102000015694 estrogen receptors Human genes 0.000 claims abstract description 51
- 201000011510 cancer Diseases 0.000 claims abstract description 39
- 102100016662 ERBB2 Human genes 0.000 claims abstract description 36
- 101700025368 ERBB2 Proteins 0.000 claims abstract description 36
- 101710037934 QRSL1 Proteins 0.000 claims abstract description 35
- 239000003814 drug Substances 0.000 claims abstract description 30
- 230000027455 binding Effects 0.000 claims abstract description 28
- 230000004913 activation Effects 0.000 claims abstract description 18
- 238000006366 phosphorylation reaction Methods 0.000 claims abstract description 16
- 230000000865 phosphorylative Effects 0.000 claims abstract description 16
- 238000002560 therapeutic procedure Methods 0.000 claims abstract description 13
- 230000001404 mediated Effects 0.000 claims abstract description 9
- 238000004519 manufacturing process Methods 0.000 claims abstract description 8
- 238000001574 biopsy Methods 0.000 claims abstract description 7
- 239000000328 estrogen antagonist Substances 0.000 claims abstract description 6
- 239000000833 heterodimer Substances 0.000 claims abstract description 6
- 230000036210 malignancy Effects 0.000 claims abstract description 6
- 230000001833 anti-estrogenic Effects 0.000 claims abstract description 5
- 238000000338 in vitro Methods 0.000 claims abstract description 5
- 238000004458 analytical method Methods 0.000 claims abstract description 4
- 206010028980 Neoplasm Diseases 0.000 claims description 99
- 210000004027 cells Anatomy 0.000 claims description 61
- HPJKCIUCZWXJDR-UHFFFAOYSA-N Letrozole Chemical compound C1=CC(C#N)=CC=C1C(N1N=CN=C1)C1=CC=C(C#N)C=C1 HPJKCIUCZWXJDR-UHFFFAOYSA-N 0.000 claims description 34
- 229960003881 letrozole Drugs 0.000 claims description 34
- 239000003886 aromatase inhibitor Substances 0.000 claims description 18
- 230000012010 growth Effects 0.000 claims description 16
- BFYIZQONLCFLEV-DAELLWKTSA-N Aromasine Chemical compound O=C1C=C[C@]2(C)[C@H]3CC[C@](C)(C(CC4)=O)[C@@H]4[C@@H]3CC(=C)C2=C1 BFYIZQONLCFLEV-DAELLWKTSA-N 0.000 claims description 15
- 229960000255 exemestane Drugs 0.000 claims description 15
- NKANXQFJJICGDU-QPLCGJKRSA-N Tamoxifen Chemical compound C=1C=CC=CC=1C(/CC)=C(C=1C=CC(OCCN(C)C)=CC=1)/C1=CC=CC=C1 NKANXQFJJICGDU-QPLCGJKRSA-N 0.000 claims description 11
- 229960001603 Tamoxifen Drugs 0.000 claims description 11
- -1 lasofoxone Chemical compound 0.000 claims description 11
- 210000000481 Breast Anatomy 0.000 claims description 9
- 210000004881 tumor cells Anatomy 0.000 claims description 9
- 230000014509 gene expression Effects 0.000 claims description 7
- 238000001802 infusion Methods 0.000 claims description 7
- 239000002464 receptor antagonist Substances 0.000 claims description 7
- YBBLVLTVTVSKRW-UHFFFAOYSA-N Anastrozole Chemical compound N#CC(C)(C)C1=CC(C(C)(C#N)C)=CC(CN2N=CN=C2)=C1 YBBLVLTVTVSKRW-UHFFFAOYSA-N 0.000 claims description 5
- 229950011548 FADROZOLE Drugs 0.000 claims description 5
- CLPFFLWZZBQMAO-UHFFFAOYSA-N Fadrozole Chemical compound C1=CC(C#N)=CC=C1C1N2C=NC=C2CCC1 CLPFFLWZZBQMAO-UHFFFAOYSA-N 0.000 claims description 5
- OSVMTWJCGUFAOD-KZQROQTASA-N Formestane Chemical compound O=C1CC[C@]2(C)[C@H]3CC[C@](C)(C(CC4)=O)[C@@H]4[C@@H]3CCC2=C1O OSVMTWJCGUFAOD-KZQROQTASA-N 0.000 claims description 5
- XLMPPFTZALNBFS-INIZCTEOSA-N Vorozole Chemical compound C1([C@@H](C2=CC=C3N=NN(C3=C2)C)N2N=CN=C2)=CC=C(Cl)C=C1 XLMPPFTZALNBFS-INIZCTEOSA-N 0.000 claims description 5
- 229960002932 anastrozole Drugs 0.000 claims description 5
- 229960004421 formestane Drugs 0.000 claims description 5
- 229960001771 vorozole Drugs 0.000 claims description 5
- VWUXBMIQPBEWFH-WCCTWKNTSA-N (7R,8R,9S,13S,14S,17S)-13-methyl-7-[9-(4,4,5,5,5-pentafluoropentylsulfinyl)nonyl]-6,7,8,9,11,12,14,15,16,17-decahydrocyclopenta[a]phenanthrene-3,17-diol Chemical compound OC1=CC=C2[C@H]3CC[C@](C)([C@H](CC4)O)[C@@H]4[C@@H]3[C@H](CCCCCCCCCS(=O)CCCC(F)(F)C(F)(F)F)CC2=C1 VWUXBMIQPBEWFH-WCCTWKNTSA-N 0.000 claims description 4
- TXUZVZSFRXZGTL-QPLCGJKRSA-N Afimoxifene Chemical compound C=1C=CC=CC=1C(/CC)=C(C=1C=CC(OCCN(C)C)=CC=1)/C1=CC=C(O)C=C1 TXUZVZSFRXZGTL-QPLCGJKRSA-N 0.000 claims description 4
- 229960003437 Aminoglutethimide Drugs 0.000 claims description 4
- ROBVIMPUHSLWNV-UHFFFAOYSA-N Aminoglutethimide Chemical compound C=1C=C(N)C=CC=1C1(CC)CCC(=O)NC1=O ROBVIMPUHSLWNV-UHFFFAOYSA-N 0.000 claims description 4
- 229960002258 Fulvestrant Drugs 0.000 claims description 4
- 210000004072 Lung Anatomy 0.000 claims description 4
- 206010027476 Metastasis Diseases 0.000 claims description 4
- XFCLJVABOIYOMF-QPLCGJKRSA-N Toremifene Chemical compound C1=CC(OCCN(C)C)=CC=C1C(\C=1C=CC=CC=1)=C(\CCCl)C1=CC=CC=C1 XFCLJVABOIYOMF-QPLCGJKRSA-N 0.000 claims description 4
- 229960005026 toremifene Drugs 0.000 claims description 4
- 229950003105 Afimoxifene Drugs 0.000 claims description 3
- 210000003491 Skin Anatomy 0.000 claims description 3
- 238000010253 intravenous injection Methods 0.000 claims description 3
- MCGDSOGUHLTADD-UHFFFAOYSA-N Arzoxifene Chemical compound C1=CC(OC)=CC=C1C1=C(OC=2C=CC(OCCN3CCCCC3)=CC=2)C2=CC=C(O)C=C2S1 MCGDSOGUHLTADD-UHFFFAOYSA-N 0.000 claims description 2
- 229950005529 Arzoxifene Drugs 0.000 claims description 2
- 210000001672 Ovary Anatomy 0.000 claims description 2
- 229960005353 Testolactone Drugs 0.000 claims description 2
- 230000002018 overexpression Effects 0.000 claims description 2
- BPEWUONYVDABNZ-DZBHQSCQSA-N testolactone Chemical compound O=C1C=C[C@]2(C)[C@H]3CC[C@](C)(OC(=O)CC4)[C@@H]4[C@@H]3CCC2=C1 BPEWUONYVDABNZ-DZBHQSCQSA-N 0.000 claims description 2
- 125000003275 alpha amino acid group Chemical group 0.000 claims 2
- 239000000203 mixture Substances 0.000 description 60
- 150000003384 small molecules Chemical class 0.000 description 20
- 101700073818 CDR1 Proteins 0.000 description 18
- 102100002977 CDR1 Human genes 0.000 description 18
- 239000002246 antineoplastic agent Substances 0.000 description 18
- 102000005962 receptors Human genes 0.000 description 18
- 108020003175 receptors Proteins 0.000 description 18
- 150000001413 amino acids Chemical class 0.000 description 15
- 239000003795 chemical substances by application Substances 0.000 description 15
- 150000001875 compounds Chemical class 0.000 description 15
- 230000000694 effects Effects 0.000 description 15
- 239000008194 pharmaceutical composition Substances 0.000 description 13
- 229940079593 drugs Drugs 0.000 description 12
- HKVAMNSJSFKALM-XJFKSLPYSA-N Everolimus Chemical compound C1C[C@@H](OCCO)[C@H](OC)C[C@@H]1C[C@@H](C)[C@H]1OC(=O)[C@@H]2CCCCN2C(=O)C(=O)[C@](O)(O2)[C@H](C)CC[C@H]2C[C@H](OC)C(C)=C/C=C/C=C/[C@@H](C)C[C@@H](C)C(=O)[C@H](OC)[C@H](O)C(C)=C[C@@H](C)C(=O)C1 HKVAMNSJSFKALM-XJFKSLPYSA-N 0.000 description 11
- 235000001014 amino acid Nutrition 0.000 description 11
- 229960005167 everolimus Drugs 0.000 description 11
- 230000001225 therapeutic Effects 0.000 description 11
- 239000000470 constituent Substances 0.000 description 10
- 230000011664 signaling Effects 0.000 description 10
- 229920000160 (ribonucleotides)n+m Polymers 0.000 description 9
- 102100001432 BTC Human genes 0.000 description 9
- 101700038892 BTC Proteins 0.000 description 9
- 230000004614 tumor growth Effects 0.000 description 9
- 241000699666 Mus <mouse, genus> Species 0.000 description 8
- 239000000427 antigen Substances 0.000 description 8
- 102000038129 antigens Human genes 0.000 description 8
- 108091007172 antigens Proteins 0.000 description 8
- 230000001629 suppression Effects 0.000 description 8
- 102000018358 Immunoglobulins Human genes 0.000 description 7
- 108060003951 Immunoglobulins Proteins 0.000 description 7
- 101710034060 RUNX1T1 Proteins 0.000 description 7
- 108020004459 Small Interfering RNA Proteins 0.000 description 7
- 229920001891 Small hairpin RNA Polymers 0.000 description 7
- 238000004166 bioassay Methods 0.000 description 7
- 239000000262 estrogen Substances 0.000 description 7
- 238000003364 immunohistochemistry Methods 0.000 description 7
- 239000003628 mammalian target of rapamycin inhibitor Substances 0.000 description 7
- 108010078554 Aromatase Proteins 0.000 description 6
- 102100005573 CYP19A1 Human genes 0.000 description 6
- 102000003993 Phosphatidylinositol 3-Kinases Human genes 0.000 description 6
- 108090000430 Phosphatidylinositol 3-Kinases Proteins 0.000 description 6
- 239000003124 biologic agent Substances 0.000 description 6
- 239000003937 drug carrier Substances 0.000 description 6
- 239000002924 silencing RNA Substances 0.000 description 6
- 239000004055 small Interfering RNA Substances 0.000 description 6
- 239000000243 solution Substances 0.000 description 6
- 230000003442 weekly Effects 0.000 description 6
- 229940046844 Aromatase inhibitors Drugs 0.000 description 5
- 238000002648 combination therapy Methods 0.000 description 5
- 238000011161 development Methods 0.000 description 5
- 230000018109 developmental process Effects 0.000 description 5
- 238000002347 injection Methods 0.000 description 5
- 239000007924 injection Substances 0.000 description 5
- 238000001990 intravenous administration Methods 0.000 description 5
- 101710007041 let-363 Proteins 0.000 description 5
- 108010045030 monoclonal antibodies Proteins 0.000 description 5
- 102000005614 monoclonal antibodies Human genes 0.000 description 5
- 235000018102 proteins Nutrition 0.000 description 5
- 102000004169 proteins and genes Human genes 0.000 description 5
- 108090000623 proteins and genes Proteins 0.000 description 5
- 102000027776 ERBB3 Human genes 0.000 description 4
- 101700041204 ERBB3 Proteins 0.000 description 4
- 102100014263 IGF1R Human genes 0.000 description 4
- 101700025802 IGF1R Proteins 0.000 description 4
- 206010046766 Uterine cancer Diseases 0.000 description 4
- 230000000996 additive Effects 0.000 description 4
- 239000003963 antioxidant agent Substances 0.000 description 4
- 235000006708 antioxidants Nutrition 0.000 description 4
- 239000000969 carrier Substances 0.000 description 4
- 201000010099 disease Diseases 0.000 description 4
- 239000006185 dispersion Substances 0.000 description 4
- 230000002452 interceptive Effects 0.000 description 4
- 239000003446 ligand Substances 0.000 description 4
- 231100000682 maximum tolerated dose Toxicity 0.000 description 4
- 239000012528 membrane Substances 0.000 description 4
- 239000003757 phosphotransferase inhibitor Substances 0.000 description 4
- 230000002829 reduced Effects 0.000 description 4
- 238000010186 staining Methods 0.000 description 4
- 239000005483 tyrosine kinase inhibitor Substances 0.000 description 4
- 229940121358 tyrosine kinase inhibitors Drugs 0.000 description 4
- 239000002525 vasculotropin inhibitor Substances 0.000 description 4
- XLYOFNOQVPJJNP-UHFFFAOYSA-N water Substances O XLYOFNOQVPJJNP-UHFFFAOYSA-N 0.000 description 4
- 238000001262 western blot Methods 0.000 description 4
- LQVXSNNAFNGRAH-QHCPKHFHSA-N (2S)-1-[4-[(5-cyclopropyl-1H-pyrazol-3-yl)amino]pyrrolo[2,1-f][1,2,4]triazin-2-yl]-N-(6-fluoropyridin-3-yl)-2-methylpyrrolidine-2-carboxamide Chemical compound C([C@@]1(C)C(=O)NC=2C=NC(F)=CC=2)CCN1C(=NN1C=CC=C11)N=C1NC(=NN1)C=C1C1CC1 LQVXSNNAFNGRAH-QHCPKHFHSA-N 0.000 description 3
- 108010071919 Bispecific Antibodies Proteins 0.000 description 3
- 206010008342 Cervix carcinoma Diseases 0.000 description 3
- 229940121647 EGFR inhibitors Drugs 0.000 description 3
- 229960001433 Erlotinib Drugs 0.000 description 3
- AAKJLRGGTJKAMG-UHFFFAOYSA-N Erlotinib Chemical compound C=12C=C(OCCOC)C(OCCOC)=CC2=NC=NC=1NC1=CC=CC(C#C)=C1 AAKJLRGGTJKAMG-UHFFFAOYSA-N 0.000 description 3
- 102000008100 Human Serum Albumin Human genes 0.000 description 3
- 108091006822 Human Serum Albumin Proteins 0.000 description 3
- 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 3
- 241000229754 Iva xanthiifolia Species 0.000 description 3
- 239000002136 L01XE07 - Lapatinib Substances 0.000 description 3
- BCFGMOOMADDAQU-UHFFFAOYSA-N Lapatinib Chemical compound O1C(CNCCS(=O)(=O)C)=CC=C1C1=CC=C(N=CN=C2NC=3C=C(Cl)C(OCC=4C=C(F)C=CC=4)=CC=3)C2=C1 BCFGMOOMADDAQU-UHFFFAOYSA-N 0.000 description 3
- 101700064507 MARK2 Proteins 0.000 description 3
- 102100000541 MARK2 Human genes 0.000 description 3
- 206010033128 Ovarian cancer Diseases 0.000 description 3
- 229960001592 Paclitaxel Drugs 0.000 description 3
- GZUITABIAKMVPG-UHFFFAOYSA-N Raloxifene Chemical compound C1=CC(O)=CC=C1C1=C(C(=O)C=2C=CC(OCCN3CCCCC3)=CC=2)C2=CC=C(O)C=C2S1 GZUITABIAKMVPG-UHFFFAOYSA-N 0.000 description 3
- 229960004622 Raloxifene Drugs 0.000 description 3
- 210000002966 Serum Anatomy 0.000 description 3
- 102100015249 VEGFA Human genes 0.000 description 3
- 108010073929 Vascular Endothelial Growth Factor A Proteins 0.000 description 3
- 239000004480 active ingredient Substances 0.000 description 3
- ULXXDDBFHOBEHA-CWDCEQMOSA-N afatinib Chemical compound N1=CN=C2C=C(O[C@@H]3COCC3)C(NC(=O)/C=C/CN(C)C)=CC2=C1NC1=CC=C(F)C(Cl)=C1 ULXXDDBFHOBEHA-CWDCEQMOSA-N 0.000 description 3
- 230000003042 antagnostic Effects 0.000 description 3
- 239000005557 antagonist Substances 0.000 description 3
- 230000000111 anti-oxidant Effects 0.000 description 3
- 201000010881 cervical cancer Diseases 0.000 description 3
- KRKNYBCHXYNGOX-UHFFFAOYSA-N citric acid Chemical compound OC(=O)CC(O)(C(O)=O)CC(O)=O KRKNYBCHXYNGOX-UHFFFAOYSA-N 0.000 description 3
- 229910003460 diamond Inorganic materials 0.000 description 3
- 239000010432 diamond Substances 0.000 description 3
- 239000002552 dosage form Substances 0.000 description 3
- 238000009472 formulation Methods 0.000 description 3
- PEDCQBHIVMGVHV-UHFFFAOYSA-N glycerine Chemical compound OCC(O)CO PEDCQBHIVMGVHV-UHFFFAOYSA-N 0.000 description 3
- 230000000670 limiting Effects 0.000 description 3
- 239000006166 lysate Substances 0.000 description 3
- 239000002829 mitogen activated protein kinase inhibitor Substances 0.000 description 3
- 229920001184 polypeptide Polymers 0.000 description 3
- 239000000843 powder Substances 0.000 description 3
- 238000002360 preparation method Methods 0.000 description 3
- 230000004044 response Effects 0.000 description 3
- 238000009097 single-agent therapy Methods 0.000 description 3
- 230000004083 survival Effects 0.000 description 3
- 239000003981 vehicle Substances 0.000 description 3
- QNDVLZJODHBUFM-FCXWGWLASA-N (2R)-3-[(2S,6R,8S,11R)-2-[(E,2R)-4-[(2S,2'S,4R,4aS,6R,8aR)-4-hydroxy-2-[(1S,3S)-1-hydroxy-3-[(2S,3R,6S)-3-methyl-1,7-dioxaspiro[5.5]undecan-2-yl]butyl]-3-methylidenespiro[4a,7,8,8a-tetrahydro-4H-pyrano[3,2-b]pyran-6,5'-oxolane]-2'-yl]but-3-en-2-yl]-11-hyd Chemical compound C([C@H](O1)[C@H](C)/C=C/[C@@H]2CC[C@@]3(CC[C@H]4O[C@@H](C([C@@H](O)[C@@H]4O3)=C)[C@@H](O)C[C@H](C)[C@@H]3[C@@H](CC[C@@]4(OCCCC4)O3)C)O2)C(C)=C[C@]21O[C@H](C[C@@](C)(O)C(O)=O)CC[C@H]2O QNDVLZJODHBUFM-FCXWGWLASA-N 0.000 description 2
- JLPULHDHAOZNQI-ZTIMHPMXSA-N 1-hexadecanoyl-2-(9Z,12Z-octadecadienoyl)-sn-glycero-3-phosphocholine Chemical compound CCCCCCCCCCCCCCCC(=O)OC[C@H](COP([O-])(=O)OCC[N+](C)(C)C)OC(=O)CCCCCCC\C=C/C\C=C/CCCCC JLPULHDHAOZNQI-ZTIMHPMXSA-N 0.000 description 2
- 101710025088 66 Proteins 0.000 description 2
- 101710034857 ATIC Proteins 0.000 description 2
- 229960001686 Afatinib Drugs 0.000 description 2
- 108010005144 Bevacizumab Proteins 0.000 description 2
- 210000000988 Bone and Bones Anatomy 0.000 description 2
- 208000000409 Breast Neoplasms Diseases 0.000 description 2
- 229940095259 Butylated Hydroxytoluene Drugs 0.000 description 2
- 239000004322 Butylated hydroxytoluene Substances 0.000 description 2
- 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 2
- 101700024634 CDK16 Proteins 0.000 description 2
- 229950002826 Canertinib Drugs 0.000 description 2
- 239000005461 Canertinib Substances 0.000 description 2
- OMZCMEYTWSXEPZ-UHFFFAOYSA-N Canertinib Chemical compound C1=C(Cl)C(F)=CC=C1NC1=NC=NC2=CC(OCCCN3CCOCC3)=C(NC(=O)C=C)C=C12 OMZCMEYTWSXEPZ-UHFFFAOYSA-N 0.000 description 2
- 108010022830 Cetuximab Proteins 0.000 description 2
- ZBNZXTGUTAYRHI-UHFFFAOYSA-N Dasatinib Chemical compound C=1C(N2CCN(CCO)CC2)=NC(C)=NC=1NC(S1)=NC=C1C(=O)NC1=C(C)C=CC=C1Cl ZBNZXTGUTAYRHI-UHFFFAOYSA-N 0.000 description 2
- 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 2
- 101700033006 EGF Proteins 0.000 description 2
- 102100010813 EGF Human genes 0.000 description 2
- 102100010782 EGFR Human genes 0.000 description 2
- 238000002965 ELISA Methods 0.000 description 2
- 229940034984 ENDOCRINE THERAPY ANTINEOPLASTIC AND IMMUNOMODULATING AGENTS Drugs 0.000 description 2
- 101700036757 ERN1 Proteins 0.000 description 2
- 102100016655 ERN1 Human genes 0.000 description 2
- 101700014948 ERN2 Proteins 0.000 description 2
- 229950008085 Figitumumab Drugs 0.000 description 2
- 101710037135 GAPC2 Proteins 0.000 description 2
- 101710037116 GAPC3 Proteins 0.000 description 2
- 101710025049 GAPDG Proteins 0.000 description 2
- 101710008404 GAPDH Proteins 0.000 description 2
- 102100006425 GAPDH Human genes 0.000 description 2
- 101710010461 Gapdh1 Proteins 0.000 description 2
- XGALLCVXEZPNRQ-UHFFFAOYSA-N Gefitinib Chemical compound C=12C=C(OCCCN3CCOCC3)C(OC)=CC2=NC=NC=1NC1=CC=C(F)C(Cl)=C1 XGALLCVXEZPNRQ-UHFFFAOYSA-N 0.000 description 2
- 108010010803 Gelatin Proteins 0.000 description 2
- 229920002153 Hydroxypropyl cellulose Polymers 0.000 description 2
- 101700085586 IRE1A Proteins 0.000 description 2
- 101700019719 IRE1B Proteins 0.000 description 2
- KTUFNOKKBVMGRW-UHFFFAOYSA-N Imatinib Chemical compound C1CN(C)CCN1CC1=CC=C(C(=O)NC=2C=C(NC=3N=C(C=CN=3)C=3C=NC=CC=3)C(C)=CC=2)C=C1 KTUFNOKKBVMGRW-UHFFFAOYSA-N 0.000 description 2
- MTCFGRXMJLQNBG-REOHCLBHSA-N L-serine Chemical compound OC[C@H](N)C(O)=O MTCFGRXMJLQNBG-REOHCLBHSA-N 0.000 description 2
- 239000005517 L01XE01 - Imatinib Substances 0.000 description 2
- 239000005411 L01XE02 - Gefitinib Substances 0.000 description 2
- 239000002147 L01XE04 - Sunitinib Substances 0.000 description 2
- 229940067606 Lecithin Drugs 0.000 description 2
- 101710007526 MAP3K14 Proteins 0.000 description 2
- 101710025050 MK0970 Proteins 0.000 description 2
- 108010029626 MM-151 Proteins 0.000 description 2
- 229950008001 Matuzumab Drugs 0.000 description 2
- 241001465754 Metazoa Species 0.000 description 2
- 229950010203 Nimotuzumab Drugs 0.000 description 2
- XAPRFLSJBSXESP-UHFFFAOYSA-N Oxycinchophen Chemical compound N=1C2=CC=CC=C2C(C(=O)O)=C(O)C=1C1=CC=CC=C1 XAPRFLSJBSXESP-UHFFFAOYSA-N 0.000 description 2
- 101700044505 PUB33 Proteins 0.000 description 2
- 101700045570 PUB34 Proteins 0.000 description 2
- 101700046887 PUB35 Proteins 0.000 description 2
- 101700066160 PUB51 Proteins 0.000 description 2
- 101700067511 PUB52 Proteins 0.000 description 2
- 101700068819 PUB53 Proteins 0.000 description 2
- 101700086326 PUB70 Proteins 0.000 description 2
- 108010061219 Panitumumab Proteins 0.000 description 2
- 229950006299 Pelitinib Drugs 0.000 description 2
- 102000004022 Protein-Tyrosine Kinases Human genes 0.000 description 2
- 108090000412 Protein-Tyrosine Kinases Proteins 0.000 description 2
- BUROJSBIWGDYCN-OZFXDLPUSA-N Ridaforolimus Chemical compound C1C[C@@H](OP(C)(C)=O)[C@H](OC)C[C@@H]1C[C@@H](C)[C@H]1OC(=O)[C@@H]2CCCCN2C(=O)C(=O)[C@](O)(O2)[C@H](C)CC[C@H]2C[C@H](OC)C(C)=CC=C/C=C/[C@@H](C)C[C@@H](C)C(=O)[C@H](OC)[C@H](O)/C(C)=C/[C@@H](C)C(=O)C1 BUROJSBIWGDYCN-OZFXDLPUSA-N 0.000 description 2
- 108010070144 Single-Chain Antibodies Proteins 0.000 description 2
- 102000005632 Single-Chain Antibodies Human genes 0.000 description 2
- QFJCIRLUMZQUOT-HPLJOQBZSA-N Sirolimus Chemical compound C1C[C@@H](O)[C@H](OC)C[C@@H]1C[C@@H](C)[C@H]1OC(=O)[C@@H]2CCCCN2C(=O)C(=O)[C@](O)(O2)[C@H](C)CC[C@H]2C[C@H](OC)/C(C)=C/C=C/C=C/[C@@H](C)C[C@@H](C)C(=O)[C@H](OC)[C@H](O)/C(C)=C/[C@@H](C)C(=O)C1 QFJCIRLUMZQUOT-HPLJOQBZSA-N 0.000 description 2
- GEHJYWRUCIMESM-UHFFFAOYSA-L Sodium sulfite Chemical compound [Na+].[Na+].[O-]S([O-])=O GEHJYWRUCIMESM-UHFFFAOYSA-L 0.000 description 2
- WINHZLLDWRZWRT-ATVHPVEESA-N Sunitinib Chemical compound CCN(CC)CCNC(=O)C1=C(C)NC(\C=C/2C3=CC(F)=CC=C3NC\2=O)=C1C WINHZLLDWRZWRT-ATVHPVEESA-N 0.000 description 2
- 102000013530 TOR Serine-Threonine Kinases Human genes 0.000 description 2
- 108010065917 TOR Serine-Threonine Kinases Proteins 0.000 description 2
- CBPNZQVSJQDFBE-FUXHJELOSA-N Temsirolimus Chemical compound C1C[C@@H](OC(=O)C(C)(CO)CO)[C@H](OC)C[C@@H]1C[C@@H](C)[C@H]1OC(=O)[C@@H]2CCCCN2C(=O)C(=O)[C@](O)(O2)[C@H](C)CC[C@H]2C[C@H](OC)/C(C)=C/C=C/C=C/[C@@H](C)C[C@@H](C)C(=O)[C@H](OC)[C@H](O)/C(C)=C/[C@@H](C)C(=O)C1 CBPNZQVSJQDFBE-FUXHJELOSA-N 0.000 description 2
- 238000010521 absorption reaction Methods 0.000 description 2
- 239000002253 acid Substances 0.000 description 2
- 239000000654 additive Substances 0.000 description 2
- 230000000259 anti-tumor Effects 0.000 description 2
- 229940045698 antineoplastic Taxanes Drugs 0.000 description 2
- 230000033228 biological regulation Effects 0.000 description 2
- 230000015572 biosynthetic process Effects 0.000 description 2
- 230000037396 body weight Effects 0.000 description 2
- 201000008275 breast carcinoma Diseases 0.000 description 2
- UIIMBOGNXHQVGW-UHFFFAOYSA-M buffer Substances [Na+].OC([O-])=O UIIMBOGNXHQVGW-UHFFFAOYSA-M 0.000 description 2
- 235000010354 butylated hydroxytoluene Nutrition 0.000 description 2
- 125000003178 carboxy group Chemical group [H]OC(*)=O 0.000 description 2
- 231100000504 carcinogenesis Toxicity 0.000 description 2
- 101710025091 cbbGC Proteins 0.000 description 2
- 229960005395 cetuximab Drugs 0.000 description 2
- 238000002512 chemotherapy Methods 0.000 description 2
- 108010020644 dalotuzumab Proteins 0.000 description 2
- 229960002482 dalotuzumab Drugs 0.000 description 2
- 238000000326 densiometry Methods 0.000 description 2
- 230000001419 dependent Effects 0.000 description 2
- 239000002612 dispersion media Substances 0.000 description 2
- 238000009261 endocrine therapy Methods 0.000 description 2
- 230000002357 endometrial Effects 0.000 description 2
- LFQSCWFLJHTTHZ-UHFFFAOYSA-N ethanol Chemical compound CCO LFQSCWFLJHTTHZ-UHFFFAOYSA-N 0.000 description 2
- 230000001747 exhibiting Effects 0.000 description 2
- 238000002474 experimental method Methods 0.000 description 2
- 108010025050 figitumumab Proteins 0.000 description 2
- 235000013305 food Nutrition 0.000 description 2
- 238000007710 freezing Methods 0.000 description 2
- 101710025070 gapdh-2 Proteins 0.000 description 2
- 229960002584 gefitinib Drugs 0.000 description 2
- 239000008273 gelatin Substances 0.000 description 2
- 229920000159 gelatin Polymers 0.000 description 2
- 235000019322 gelatine Nutrition 0.000 description 2
- 235000011852 gelatine desserts Nutrition 0.000 description 2
- 239000001863 hydroxypropyl cellulose Substances 0.000 description 2
- 235000010977 hydroxypropyl cellulose Nutrition 0.000 description 2
- 229940071676 hydroxypropylcellulose Drugs 0.000 description 2
- 229960002411 imatinib Drugs 0.000 description 2
- 239000004615 ingredient Substances 0.000 description 2
- 238000001361 intraarterial administration Methods 0.000 description 2
- 230000003834 intracellular Effects 0.000 description 2
- 238000007918 intramuscular administration Methods 0.000 description 2
- 238000007912 intraperitoneal administration Methods 0.000 description 2
- 238000007913 intrathecal administration Methods 0.000 description 2
- 101700052395 ire-1 Proteins 0.000 description 2
- 229960004891 lapatinib Drugs 0.000 description 2
- 239000000787 lecithin Substances 0.000 description 2
- 235000010445 lecithin Nutrition 0.000 description 2
- 238000011068 load Methods 0.000 description 2
- 108010029633 matuzumab Proteins 0.000 description 2
- 238000005259 measurement Methods 0.000 description 2
- 239000002609 media Substances 0.000 description 2
- 238000000034 method Methods 0.000 description 2
- 230000000394 mitotic Effects 0.000 description 2
- 108010001830 monoclonal antibody 806 Proteins 0.000 description 2
- 108010043585 nimotuzumab Proteins 0.000 description 2
- 230000002611 ovarian Effects 0.000 description 2
- 229960001972 panitumumab Drugs 0.000 description 2
- 239000012188 paraffin wax Substances 0.000 description 2
- WVUNYSQLFKLYNI-AATRIKPKSA-N pelitinib Chemical compound C=12C=C(NC(=O)\C=C\CN(C)C)C(OCC)=CC2=NC=C(C#N)C=1NC1=CC=C(F)C(Cl)=C1 WVUNYSQLFKLYNI-AATRIKPKSA-N 0.000 description 2
- NBIIXXVUZAFLBC-UHFFFAOYSA-N phosphoric acid Chemical compound OP(O)(O)=O NBIIXXVUZAFLBC-UHFFFAOYSA-N 0.000 description 2
- BASFCYQUMIYNBI-UHFFFAOYSA-N platinum Chemical compound [Pt] BASFCYQUMIYNBI-UHFFFAOYSA-N 0.000 description 2
- 229920000515 polycarbonate Polymers 0.000 description 2
- 230000002335 preservative Effects 0.000 description 2
- 239000003755 preservative agent Substances 0.000 description 2
- 125000002924 primary amino group Chemical group [H]N([H])* 0.000 description 2
- 230000002035 prolonged Effects 0.000 description 2
- DNIAPMSPPWPWGF-UHFFFAOYSA-N propylene glycol Chemical compound CC(O)CO DNIAPMSPPWPWGF-UHFFFAOYSA-N 0.000 description 2
- 238000000159 protein binding assay Methods 0.000 description 2
- 239000003909 protein kinase inhibitor Substances 0.000 description 2
- 238000011160 research Methods 0.000 description 2
- 229960001302 ridaforolimus Drugs 0.000 description 2
- 230000035945 sensitivity Effects 0.000 description 2
- 229960002930 sirolimus Drugs 0.000 description 2
- FAPWRFPIFSIZLT-UHFFFAOYSA-M sodium chloride Chemical compound [Na+].[Cl-] FAPWRFPIFSIZLT-UHFFFAOYSA-M 0.000 description 2
- 239000011780 sodium chloride Substances 0.000 description 2
- 239000002904 solvent Substances 0.000 description 2
- 230000001954 sterilising Effects 0.000 description 2
- 238000004659 sterilization and disinfection Methods 0.000 description 2
- 238000003860 storage Methods 0.000 description 2
- 238000007920 subcutaneous administration Methods 0.000 description 2
- 235000000346 sugar Nutrition 0.000 description 2
- 150000008163 sugars Chemical class 0.000 description 2
- 229960001796 sunitinib Drugs 0.000 description 2
- 238000001356 surgical procedure Methods 0.000 description 2
- 229930003347 taxol Natural products 0.000 description 2
- 229960000235 temsirolimus Drugs 0.000 description 2
- 230000004797 therapeutic response Effects 0.000 description 2
- 210000001519 tissues Anatomy 0.000 description 2
- 230000000699 topical Effects 0.000 description 2
- 230000002588 toxic Effects 0.000 description 2
- XRYJULCDUUATMC-CYBMUJFWSA-N 4-[4-[[(1R)-1-phenylethyl]amino]-7H-pyrrolo[2,3-d]pyrimidin-6-yl]phenol Chemical compound N([C@H](C)C=1C=CC=CC=1)C(C=1C=2)=NC=NC=1NC=2C1=CC=C(O)C=C1 XRYJULCDUUATMC-CYBMUJFWSA-N 0.000 description 1
- HRZFUMHJMZEROT-UHFFFAOYSA-L 7681-57-4 Chemical compound [Na+].[Na+].[O-]S(=O)S([O-])(=O)=O HRZFUMHJMZEROT-UHFFFAOYSA-L 0.000 description 1
- 102100001249 ALB Human genes 0.000 description 1
- 101710027066 ALB Proteins 0.000 description 1
- 101700047950 ASK9 Proteins 0.000 description 1
- 108010083275 AV-203 Proteins 0.000 description 1
- 229940028652 Abraxane Drugs 0.000 description 1
- 210000000577 Adipose Tissue Anatomy 0.000 description 1
- 102000007299 Amphiregulin Human genes 0.000 description 1
- 108010033760 Amphiregulin Proteins 0.000 description 1
- AEMFNILZOJDQLW-QAGGRKNESA-N Androstenedione Chemical compound O=C1CC[C@]2(C)[C@H]3CC[C@](C)(C(CC4)=O)[C@@H]4[C@@H]3CCC2=C1 AEMFNILZOJDQLW-QAGGRKNESA-N 0.000 description 1
- 229960005471 Androstenedione Drugs 0.000 description 1
- 210000004369 Blood Anatomy 0.000 description 1
- 210000004556 Brain Anatomy 0.000 description 1
- 206010055113 Breast cancer metastatic Diseases 0.000 description 1
- OLESAACUTLOWQZ-UHFFFAOYSA-L Carboplatin Chemical compound O=C1O[Pt]([N]([H])([H])[H])([N]([H])([H])[H])OC(=O)C11CCC1 OLESAACUTLOWQZ-UHFFFAOYSA-L 0.000 description 1
- 229960004562 Carboplatin Drugs 0.000 description 1
- 208000005623 Carcinogenesis Diseases 0.000 description 1
- 210000003855 Cell Nucleus Anatomy 0.000 description 1
- OSASVXMJTNOKOY-UHFFFAOYSA-N Chlorobutanol Chemical compound CC(C)(O)C(Cl)(Cl)Cl OSASVXMJTNOKOY-UHFFFAOYSA-N 0.000 description 1
- 229960004926 Chlorobutanol Drugs 0.000 description 1
- 229960001305 Cysteine Hydrochloride Drugs 0.000 description 1
- FBPFZTCFMRRESA-FSIIMWSLSA-N D-Glucitol Natural products OC[C@H](O)[C@H](O)[C@@H](O)[C@H](O)CO FBPFZTCFMRRESA-FSIIMWSLSA-N 0.000 description 1
- FBPFZTCFMRRESA-KAZBKCHUSA-N D-Mannitol Natural products OC[C@@H](O)[C@H](O)[C@H](O)[C@H](O)CO FBPFZTCFMRRESA-KAZBKCHUSA-N 0.000 description 1
- FBPFZTCFMRRESA-JGWLITMVSA-N D-glucitol Chemical compound OC[C@H](O)[C@@H](O)[C@H](O)[C@H](O)CO FBPFZTCFMRRESA-JGWLITMVSA-N 0.000 description 1
- 230000004544 DNA amplification Effects 0.000 description 1
- 101700039191 EGFR Proteins 0.000 description 1
- 102100016629 EPGN Human genes 0.000 description 1
- 101000011882 ERBB3 Proteins 0.000 description 1
- 102100009851 ERBB4 Human genes 0.000 description 1
- 101700023619 ERBB4 Proteins 0.000 description 1
- 101700072570 EREG Proteins 0.000 description 1
- 102100019146 EREG Human genes 0.000 description 1
- 108010016906 Epigen Proteins 0.000 description 1
- 229940082789 Erbitux Drugs 0.000 description 1
- LVGKNOAMLMIIKO-QXMHVHEDSA-N Ethyl oleate Chemical compound CCCCCCCC\C=C/CCCCCCCC(=O)OCC LVGKNOAMLMIIKO-QXMHVHEDSA-N 0.000 description 1
- 210000003722 Extracellular Fluid Anatomy 0.000 description 1
- 230000035693 Fab Effects 0.000 description 1
- 241000272184 Falconiformes Species 0.000 description 1
- 101700085894 GSK1 Proteins 0.000 description 1
- 210000001035 Gastrointestinal Tract Anatomy 0.000 description 1
- 229940080856 Gleevec Drugs 0.000 description 1
- 102000006771 Gonadotropins Human genes 0.000 description 1
- 108010086677 Gonadotropins Proteins 0.000 description 1
- 241001009045 Grapevine virus I Species 0.000 description 1
- 101700029736 HBEGF Proteins 0.000 description 1
- 102100016400 HBEGF Human genes 0.000 description 1
- 101700075868 HER1 Proteins 0.000 description 1
- 241000282412 Homo Species 0.000 description 1
- 210000004408 Hybridomas Anatomy 0.000 description 1
- 229940072221 IMMUNOGLOBULINS Drugs 0.000 description 1
- 229960003445 Idelalisib Drugs 0.000 description 1
- YKLIKGKUANLGSB-HNNXBMFYSA-N Idelalisib Chemical compound C1([C@@H](NC=2[C]3N=CN=C3N=CN=2)CC)=NC2=CC=CC(F)=C2C(=O)N1C1=CC=CC=C1 YKLIKGKUANLGSB-HNNXBMFYSA-N 0.000 description 1
- 108090000539 Immunoglobulin Isotypes Proteins 0.000 description 1
- 102000004090 Immunoglobulin Isotypes Human genes 0.000 description 1
- 102000017727 Immunoglobulin Variable Region Human genes 0.000 description 1
- 108010067060 Immunoglobulin Variable Region Proteins 0.000 description 1
- 229940084651 Iressa Drugs 0.000 description 1
- 210000003734 Kidney Anatomy 0.000 description 1
- 239000005551 L01XE03 - Erlotinib Substances 0.000 description 1
- 239000002067 L01XE06 - Dasatinib Substances 0.000 description 1
- 102100000087 LARP6 Human genes 0.000 description 1
- 101700066207 LARP6 Proteins 0.000 description 1
- 210000004185 Liver Anatomy 0.000 description 1
- 239000004425 Makrolon Substances 0.000 description 1
- 206010025650 Malignant melanoma Diseases 0.000 description 1
- FBPFZTCFMRRESA-KVTDHHQDSA-N Mannitol Chemical compound OC[C@@H](O)[C@@H](O)[C@H](O)[C@H](O)CO FBPFZTCFMRRESA-KVTDHHQDSA-N 0.000 description 1
- FEWJPZIEWOKRBE-XIXRPRMCSA-N Mesotartaric acid Chemical compound OC(=O)[C@@H](O)[C@@H](O)C(O)=O FEWJPZIEWOKRBE-XIXRPRMCSA-N 0.000 description 1
- 206010061289 Metastatic neoplasm Diseases 0.000 description 1
- LXCFILQKKLGQFO-UHFFFAOYSA-N Methylparaben Chemical compound COC(=O)C1=CC=C(O)C=C1 LXCFILQKKLGQFO-UHFFFAOYSA-N 0.000 description 1
- 102000004232 Mitogen-activated protein kinase kinases Human genes 0.000 description 1
- 108090000744 Mitogen-activated protein kinase kinases Proteins 0.000 description 1
- 241000699660 Mus musculus Species 0.000 description 1
- JZZFDCXSFTVOJY-UHFFFAOYSA-N N-[4-(3-chloro-4-fluoroanilino)-7-(3-morpholin-4-ylpropoxy)quinazolin-6-yl]prop-2-enamide;hydron;dichloride Chemical compound Cl.Cl.C1=C(Cl)C(F)=CC=C1NC1=NC=NC2=CC(OCCCN3CCOCC3)=C(NC(=O)C=C)C=C12 JZZFDCXSFTVOJY-UHFFFAOYSA-N 0.000 description 1
- 108009000118 Nuclear Receptors Proteins 0.000 description 1
- 206010070577 Oestrogen receptor positive breast cancer Diseases 0.000 description 1
- 101700067249 POP2 Proteins 0.000 description 1
- 241000724284 Peanut stunt virus Species 0.000 description 1
- 102000017343 Phosphatidylinositol kinase Human genes 0.000 description 1
- 108050005377 Phosphatidylinositol kinase Proteins 0.000 description 1
- 108091000081 Phosphotransferases Proteins 0.000 description 1
- 241000186704 Pinales Species 0.000 description 1
- 239000002202 Polyethylene glycol Substances 0.000 description 1
- 229940075579 Propyl Gallate Drugs 0.000 description 1
- 210000002307 Prostate Anatomy 0.000 description 1
- 102000001253 Protein Kinases Human genes 0.000 description 1
- 108020004511 Recombinant DNA Proteins 0.000 description 1
- 206010070308 Refractory cancer Diseases 0.000 description 1
- 229940100996 SODIUM BISULFATE Drugs 0.000 description 1
- 210000003079 Salivary Glands Anatomy 0.000 description 1
- CYOHGALHFOKKQC-UHFFFAOYSA-N Selumetinib Chemical compound OCCONC(=O)C=1C=C2N(C)C=NC2=C(F)C=1NC1=CC=C(Br)C=C1Cl CYOHGALHFOKKQC-UHFFFAOYSA-N 0.000 description 1
- 229950010746 Selumetinib Drugs 0.000 description 1
- WBHQBSYUUJJSRZ-UHFFFAOYSA-M Sodium bisulfate Chemical compound [Na+].OS([O-])(=O)=O WBHQBSYUUJJSRZ-UHFFFAOYSA-M 0.000 description 1
- 229940075582 Sorbic Acid Drugs 0.000 description 1
- 229940035718 Sular Drugs 0.000 description 1
- 229940034785 Sutent Drugs 0.000 description 1
- 101700038204 TGFA Proteins 0.000 description 1
- 229940120982 Tarceva Drugs 0.000 description 1
- 229940035295 Ting Drugs 0.000 description 1
- AOBORMOPSGHCAX-DGHZZKTQSA-N Tocofersolan Chemical compound OCCOC(=O)CCC(=O)OC1=C(C)C(C)=C2O[C@](CCC[C@H](C)CCC[C@H](C)CCCC(C)C)(C)CCC2=C1C AOBORMOPSGHCAX-DGHZZKTQSA-N 0.000 description 1
- 108010010691 Trastuzumab Proteins 0.000 description 1
- 229940094060 Tykerb Drugs 0.000 description 1
- 210000003932 Urinary Bladder Anatomy 0.000 description 1
- 229940058865 Vectibix Drugs 0.000 description 1
- 241000711975 Vesicular stomatitis virus Species 0.000 description 1
- CIWBSHSKHKDKBQ-JLAZNSOCSA-N Vitamin C Chemical compound OC[C@H](O)[C@H]1OC(=O)C(O)=C1O CIWBSHSKHKDKBQ-JLAZNSOCSA-N 0.000 description 1
- QIJRTFXNRTXDIP-JIZZDEOASA-N [(1R)-1-carboxy-2-sulfanylethyl]azanium;chloride;hydrate Chemical compound O.Cl.SC[C@H](N)C(O)=O QIJRTFXNRTXDIP-JIZZDEOASA-N 0.000 description 1
- 150000007513 acids Chemical class 0.000 description 1
- 230000003213 activating Effects 0.000 description 1
- 239000000556 agonist Substances 0.000 description 1
- 229940050528 albumin Drugs 0.000 description 1
- 229940087168 alpha Tocopherol Drugs 0.000 description 1
- GVJHHUAWPYXKBD-IEOSBIPESA-N alpha-Tocopherol Natural products OC1=C(C)C(C)=C2O[C@@](CCC[C@H](C)CCC[C@H](C)CCCC(C)C)(C)CCC2=C1C GVJHHUAWPYXKBD-IEOSBIPESA-N 0.000 description 1
- 229910000147 aluminium phosphate Inorganic materials 0.000 description 1
- 239000003242 anti bacterial agent Substances 0.000 description 1
- 230000000844 anti-bacterial Effects 0.000 description 1
- 230000003388 anti-hormone Effects 0.000 description 1
- 239000003429 antifungal agent Substances 0.000 description 1
- 230000000890 antigenic Effects 0.000 description 1
- 229940045988 antineoplastic drugs Protein kinase inhibitors Drugs 0.000 description 1
- 230000003078 antioxidant Effects 0.000 description 1
- 239000007864 aqueous solution Substances 0.000 description 1
- 238000005899 aromatization reaction Methods 0.000 description 1
- 235000010323 ascorbic acid Nutrition 0.000 description 1
- 229960005070 ascorbic acid Drugs 0.000 description 1
- 239000011668 ascorbic acid Substances 0.000 description 1
- 125000003289 ascorbyl group Chemical group [H]O[C@@]([H])(C([H])([H])O*)[C@@]1([H])OC(=O)C(O*)=C1O* 0.000 description 1
- 229960000397 bevacizumab Drugs 0.000 description 1
- 108010080308 biregulin Proteins 0.000 description 1
- 230000000903 blocking Effects 0.000 description 1
- 239000008280 blood Substances 0.000 description 1
- 201000008274 breast adenocarcinoma Diseases 0.000 description 1
- 230000001488 breeding Effects 0.000 description 1
- 235000019282 butylated hydroxyanisole Nutrition 0.000 description 1
- 230000036952 cancer formation Effects 0.000 description 1
- 239000002775 capsule Substances 0.000 description 1
- 201000009030 carcinoma Diseases 0.000 description 1
- 101710014509 celF Proteins 0.000 description 1
- 239000006285 cell suspension Substances 0.000 description 1
- 239000002738 chelating agent Substances 0.000 description 1
- 230000000973 chemotherapeutic Effects 0.000 description 1
- 239000011248 coating agent Substances 0.000 description 1
- 238000000576 coating method Methods 0.000 description 1
- 229960002448 dasatinib Drugs 0.000 description 1
- 230000034994 death Effects 0.000 description 1
- 231100000517 death Toxicity 0.000 description 1
- 230000003247 decreasing Effects 0.000 description 1
- 238000006297 dehydration reaction Methods 0.000 description 1
- 230000001934 delay Effects 0.000 description 1
- 230000001627 detrimental Effects 0.000 description 1
- 230000029087 digestion Effects 0.000 description 1
- UGMCXQCYOVCMTB-UHFFFAOYSA-K dihydroxy(stearato)aluminium Chemical compound CCCCCCCCCCCCCCCCCC(=O)O[Al](O)O UGMCXQCYOVCMTB-UHFFFAOYSA-K 0.000 description 1
- 239000000539 dimer Substances 0.000 description 1
- 239000002270 dispersing agent Substances 0.000 description 1
- 238000009826 distribution Methods 0.000 description 1
- 229960003668 docetaxel Drugs 0.000 description 1
- 238000002651 drug therapy Methods 0.000 description 1
- KCXVZYZYPLLWCC-UHFFFAOYSA-N edta Chemical compound OC(=O)CN(CC(O)=O)CCN(CC(O)=O)CC(O)=O KCXVZYZYPLLWCC-UHFFFAOYSA-N 0.000 description 1
- 239000003995 emulsifying agent Substances 0.000 description 1
- 238000005516 engineering process Methods 0.000 description 1
- 239000002834 estrogen receptor modulator Substances 0.000 description 1
- 201000007281 estrogen-receptor positive breast cancer Diseases 0.000 description 1
- 229940093471 ethyl oleate Drugs 0.000 description 1
- 238000011156 evaluation Methods 0.000 description 1
- 239000012091 fetal bovine serum Substances 0.000 description 1
- 238000009093 first-line therapy Methods 0.000 description 1
- WSFSSNUMVMOOMR-UHFFFAOYSA-N formaldehyde Chemical compound O=C WSFSSNUMVMOOMR-UHFFFAOYSA-N 0.000 description 1
- 238000005755 formation reaction Methods 0.000 description 1
- 238000004108 freeze drying Methods 0.000 description 1
- 239000000499 gel Substances 0.000 description 1
- 238000002695 general anesthesia Methods 0.000 description 1
- 235000019410 glycyrrhizin Nutrition 0.000 description 1
- 239000002622 gonadotropin Substances 0.000 description 1
- 239000001963 growth media Substances 0.000 description 1
- 108010084091 heregulin beta1 Proteins 0.000 description 1
- 238000005734 heterodimerization reaction Methods 0.000 description 1
- 238000007489 histopathology method Methods 0.000 description 1
- 230000003054 hormonal Effects 0.000 description 1
- 239000003667 hormone antagonist Substances 0.000 description 1
- 102000023886 human ERBB3 protein Human genes 0.000 description 1
- 101500002601 human Epidermal growth factor Proteins 0.000 description 1
- 229940116978 human epidermal growth factor Drugs 0.000 description 1
- 230000002055 immunohistochemical Effects 0.000 description 1
- 238000007901 in situ hybridization Methods 0.000 description 1
- 238000010348 incorporation Methods 0.000 description 1
- 239000007972 injectable composition Substances 0.000 description 1
- 230000002601 intratumoral Effects 0.000 description 1
- 238000002665 ion therapy Methods 0.000 description 1
- 150000002500 ions Chemical class 0.000 description 1
- 239000007951 isotonicity adjuster Substances 0.000 description 1
- 230000003902 lesions Effects 0.000 description 1
- 239000002502 liposome Substances 0.000 description 1
- 239000007788 liquid Substances 0.000 description 1
- 239000000594 mannitol Substances 0.000 description 1
- 235000010355 mannitol Nutrition 0.000 description 1
- 239000000463 material Substances 0.000 description 1
- 201000001441 melanoma Diseases 0.000 description 1
- 230000003340 mental Effects 0.000 description 1
- 229910052751 metal Inorganic materials 0.000 description 1
- 239000002184 metal Substances 0.000 description 1
- 230000001394 metastastic Effects 0.000 description 1
- VKQFCGNPDRICFG-UHFFFAOYSA-N methyl 2-methylpropyl 2,6-dimethyl-4-(2-nitrophenyl)-1,4-dihydropyridine-3,5-dicarboxylate Chemical compound COC(=O)C1=C(C)NC(C)=C(C(=O)OCC(C)C)C1C1=CC=CC=C1[N+]([O-])=O VKQFCGNPDRICFG-UHFFFAOYSA-N 0.000 description 1
- 239000004530 micro-emulsion Substances 0.000 description 1
- 238000001471 micro-filtration Methods 0.000 description 1
- 244000005700 microbiome Species 0.000 description 1
- 239000006956 minimum essential medium Substances 0.000 description 1
- 239000003226 mitogen Substances 0.000 description 1
- 229960004927 neomycin Drugs 0.000 description 1
- 102000006255 nuclear receptors Human genes 0.000 description 1
- 108020004707 nucleic acids Proteins 0.000 description 1
- 150000007523 nucleic acids Chemical class 0.000 description 1
- 150000002895 organic esters Chemical class 0.000 description 1
- 201000002528 pancreatic cancer Diseases 0.000 description 1
- 238000007911 parenteral administration Methods 0.000 description 1
- 230000036961 partial Effects 0.000 description 1
- 239000002245 particle Substances 0.000 description 1
- 230000037361 pathway Effects 0.000 description 1
- 108010042024 pertuzumab Proteins 0.000 description 1
- 229960002087 pertuzumab Drugs 0.000 description 1
- 238000009521 phase II clinical trial Methods 0.000 description 1
- ISWSIDIOOBJBQZ-UHFFFAOYSA-N phenol Chemical compound OC1=CC=CC=C1 ISWSIDIOOBJBQZ-UHFFFAOYSA-N 0.000 description 1
- 239000006187 pill Substances 0.000 description 1
- 230000003169 placental Effects 0.000 description 1
- 229910052697 platinum Inorganic materials 0.000 description 1
- 239000004417 polycarbonate Substances 0.000 description 1
- 229920001223 polyethylene glycol Polymers 0.000 description 1
- 229920005862 polyol Polymers 0.000 description 1
- 150000003077 polyols Chemical class 0.000 description 1
- 230000002265 prevention Effects 0.000 description 1
- 238000004393 prognosis Methods 0.000 description 1
- 239000003380 propellant Substances 0.000 description 1
- ZTHYODDOHIVTJV-UHFFFAOYSA-N propyl 3,4,5-trihydroxybenzoate Chemical compound CCCOC(=O)C1=CC(O)=C(O)C(O)=C1 ZTHYODDOHIVTJV-UHFFFAOYSA-N 0.000 description 1
- 239000000473 propyl gallate Substances 0.000 description 1
- 235000010388 propyl gallate Nutrition 0.000 description 1
- 230000001698 pyrogenic Effects 0.000 description 1
- 102000027656 receptor tyrosine kinases Human genes 0.000 description 1
- 108091007921 receptor tyrosine kinases Proteins 0.000 description 1
- 238000003757 reverse transcription PCR Methods 0.000 description 1
- 150000003839 salts Chemical class 0.000 description 1
- 238000010187 selection method Methods 0.000 description 1
- 230000019491 signal transduction Effects 0.000 description 1
- KEAYESYHFKHZAL-UHFFFAOYSA-N sodium Chemical compound [Na] KEAYESYHFKHZAL-UHFFFAOYSA-N 0.000 description 1
- 239000011734 sodium Substances 0.000 description 1
- 229910052708 sodium Inorganic materials 0.000 description 1
- 229910000342 sodium bisulfate Inorganic materials 0.000 description 1
- 229940001584 sodium metabisulfite Drugs 0.000 description 1
- 235000010262 sodium metabisulphite Nutrition 0.000 description 1
- 229940001482 sodium sulfite Drugs 0.000 description 1
- 235000010265 sodium sulphite Nutrition 0.000 description 1
- 235000010199 sorbic acid Nutrition 0.000 description 1
- 239000004334 sorbic acid Substances 0.000 description 1
- WSWCOQWTEOXDQX-UHFFFAOYSA-N sorbic acid Chemical compound CC=CC=CC(O)=O WSWCOQWTEOXDQX-UHFFFAOYSA-N 0.000 description 1
- 239000000600 sorbitol Substances 0.000 description 1
- 230000000638 stimulation Effects 0.000 description 1
- 239000000126 substance Substances 0.000 description 1
- 230000000576 supplementary Effects 0.000 description 1
- 238000002198 surface plasmon resonance spectroscopy Methods 0.000 description 1
- 238000003786 synthesis reaction Methods 0.000 description 1
- 230000002194 synthesizing Effects 0.000 description 1
- 239000003826 tablet Substances 0.000 description 1
- 239000011975 tartaric acid Substances 0.000 description 1
- 229960001367 tartaric acid Drugs 0.000 description 1
- 235000002906 tartaric acid Nutrition 0.000 description 1
- 229910052718 tin Inorganic materials 0.000 description 1
- ATJFFYVFTNAWJD-UHFFFAOYSA-N tin hydride Chemical compound [Sn] ATJFFYVFTNAWJD-UHFFFAOYSA-N 0.000 description 1
- 229960000984 tocofersolan Drugs 0.000 description 1
- 231100000730 tolerability Toxicity 0.000 description 1
- 231100000331 toxic Toxicity 0.000 description 1
- 102000027575 transmembrane receptors Human genes 0.000 description 1
- 108091007901 transmembrane receptors Proteins 0.000 description 1
- 229960000575 trastuzumab Drugs 0.000 description 1
- 238000004450 types of analysis Methods 0.000 description 1
- 150000004917 tyrosine kinase inhibitor derivatives Chemical class 0.000 description 1
- 238000001291 vacuum drying Methods 0.000 description 1
- 238000009777 vacuum freeze-drying Methods 0.000 description 1
- 239000000080 wetting agent Substances 0.000 description 1
- 239000002076 α-tocopherol Substances 0.000 description 1
- 235000004835 α-tocopherol Nutrition 0.000 description 1
Classifications
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K39/00—Medicinal preparations containing antigens or antibodies
- A61K2039/505—Medicinal preparations containing antigens or antibodies comprising antibodies
-
- 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
- A61K31/4196—1,2,4-Triazoles
-
- 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
- A61K31/56—Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids
- A61K31/565—Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids not substituted in position 17 beta by a carbon atom, e.g. estrane, estradiol
- A61K31/568—Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids not substituted in position 17 beta by a carbon atom, e.g. estrane, estradiol substituted in positions 10 and 13 by a chain having at least one carbon atom, e.g. androstanes, e.g. testosterone
- A61K31/5685—Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids not substituted in position 17 beta by a carbon atom, e.g. estrane, estradiol substituted in positions 10 and 13 by a chain having at least one carbon atom, e.g. androstanes, e.g. testosterone having an oxo group in position 17, e.g. androsterone
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K39/00—Medicinal preparations containing antigens or antibodies
- A61K39/395—Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
- A61K39/39533—Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals
- A61K39/39558—Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals against tumor tissues, cells, antigens
-
- 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
- A61P11/00—Drugs for disorders of the respiratory system
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P15/00—Drugs for genital or sexual disorders; Contraceptives
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P17/00—Drugs for dermatological disorders
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P35/00—Antineoplastic agents
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K16/00—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
- C07K16/18—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
- C07K16/32—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against translation products of oncogenes
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K16/00—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
- C07K16/46—Hybrid immunoglobulins
- C07K16/468—Immunoglobulins having two or more different antigen binding sites, e.g. multifunctional antibodies
Abstract
Disclosed is the use of an antibody that inhibits heregulin binding to ErbB2/ErbB3 heterodimer for manufacture of a medicament for treating a malignant tumour in a human patient, wherein the patient has been treated for malignancy with an anti-estrogen therapy and has become resistant to such therapy, wherein the malignant tumour, by analysis of a tumour biopsy taken from the patent after the patient has become resistant, is estrogen receptor positive and expresses HER2, wherein the antibody inhibits heregulin-mediated activation of estrogen receptors in malignant tumour cells, which activation comprises phosphorylation of estrogen receptors, wherein the antibody is formulated for administration at a dosage that yields a concentration of the antibody in the patient’s bloodstream that is a sufficient concentration to inhibit heregulin-induced estrogen receptor phosphorylation in MCF7 cells in vitro by at least 20%, at least 30%, at least 40%, at least 50%, at least 60%, or at least 70%, and wherein the antibody comprises, VH CDR1, 2 and 3 sequences as shown in SEQ ID NOs: 3-5, respectively, and, VL CDR1, 2 and 3 sequences as shown in SEQ ID NOs: 6-8, respectively. py, wherein the malignant tumour, by analysis of a tumour biopsy taken from the patent after the patient has become resistant, is estrogen receptor positive and expresses HER2, wherein the antibody inhibits heregulin-mediated activation of estrogen receptors in malignant tumour cells, which activation comprises phosphorylation of estrogen receptors, wherein the antibody is formulated for administration at a dosage that yields a concentration of the antibody in the patient’s bloodstream that is a sufficient concentration to inhibit heregulin-induced estrogen receptor phosphorylation in MCF7 cells in vitro by at least 20%, at least 30%, at least 40%, at least 50%, at least 60%, or at least 70%, and wherein the antibody comprises, VH CDR1, 2 and 3 sequences as shown in SEQ ID NOs: 3-5, respectively, and, VL CDR1, 2 and 3 sequences as shown in SEQ ID NOs: 6-8, respectively.
Description
USE OF INHIBITORS OF EGFR-FAMILY RECEPTORS IN THE TREATMENT OF
HORMONE REFRACTORY BREAST CANCERS
Background
In women, breast cancer is among the most common s and is the fifth most common
cause of cancer deaths. Due to the heterogeneity of breast cancers, 10-year progression free survival
can vary widely with stage and type, from 98% to 10%. Different forms of breast s can have
remarkably different biological characteristics and clinical behavior. Thus, classification of a
patient's breast cancer has become a critical component for determining a treatment regimen. For
example, along with classification of ogical type and grade, breast cancers now are routinely
evaluated for expression of hormone receptors (estrogen receptor (ER) and terone receptor (PR)
and for expression of HER2 (ErbB2), since a number of ent modalities are currently available
that target hormone receptors or HER2. Other cancers, e.g., uterine or ovarian cancers, may be
similarly characterized. ER and PR are both nuclear receptors (i.e., they are predominantly located at
cell nuclei, rather than the cell surface) and small molecule inhibitors that directly or indirectly target
ER and/or PR have been ped. HER2, or human epidermal growth factor receptor type 2, is a
receptor normally located on the cell surface and antibodies that target HER2 have been developed as
therapeutics. HER2 is the only member of the EGFR family (which also includes HER1 (EGFR),
HER3 (ErbB3) and HER4 (ErbB4)) that is not e of binding to an ting ligand on its own.
Thus HER2 is only functional as a receptor when incorporated into a heterodimeric receptor complex
with another EGFR family member, such as HER3. Cancers classified as expressing the estrogen
receptor (estrogen receptor positive, "ER+") may be treated with an ER nist such as tamoxifen.
Similarly, s classified as expressing high levels of the HER2 may be treated with an anti-HER2
antibody, such as zumab, or with a HER2-active receptor tyrosine kinase inhibitor such as
lapatinib (which also inhibits EGFR tyrosine kinase) or AG879.
Tamoxifen has been used as therapy against ER+ breast cancer for decades and now represent
a standard component of front-line therapy for ER+ breast cancers. Tamoxifen is a member of the
class of ive estrogen receptor modulators (e.g., raloxifene, fene and fulvestrant), of which
tamoxifen, toremifene and fulvestrant are estrogen receptor antagonists and raloxifene has agonist
activity in bone and antagonist ty in breast and uterine cancers. These antagonist drugs
specifically block the hormonal activation of the estrogen receptor and are effective therapeutic agents
for the treatment of ER+ breast cancers that have not become hormone refractory. Tamoxifen, for
example, induces remissions in over half of ER+ breast cancer patients upon l treatment. The
long term utility of hormone receptor de is limited by the phenomenon of the development of
e refractory tumor teristics following extended treatment. Most treated tumors
eventually become hormone refractory in that they become tamoxifen resistant.
Thus, al blockade with hormone antagonists and other hormone modulatory drugs
such as aromatase inhibitors (e.g., exemestane, anastrozole, letrozole, anastrozole, vorozole,
formestane and fadrozole), which block estrogen synthesis, can delay progression of ER+ tumors, but
the frequent pment of resistance to such hormone modulatory drugs has created a longstanding
need for anti-cancer therapeutic agents that are effective against hormone refractory ER+ cancers.
The present disclosure addresses this need and provides onal benefits.
Summary
Provided herein are methods for treating hormone refractory breast s (e.g., tumors),
including estrogen receptor positive and estrogen receptor ve e refractory breast cancers,
as well as pharmaceutical compositions that can be used in such s. The methods and
compositions are based, at least in part, on the discovery that ErbB3 inhibition can suppress the
growth of hormone refractory breast cancer cells. In particular, administration of anti-ErbB3 antibody
is believed to suppress the growth of hormone refractory breast cancer cells. Furthermore, it has now
been discovered that heregulin activation of ErbB2/ErbB3 heterodimers can in turn activate (by
causing the phosphorylation of) estrogen receptors, a phenomenon that is believed to play a role in the
development of resistance to hormone modulatory drugs in ER+ tumors. Thus, also provided herein
are methods and compositions for inhibiting the activation of estrogen receptors by inhibiting the
binding of heregulin to ErbB2/ErbB3 heterodimers. Such s may be beneficially practiced in
combination with co-administration of one or more estrogen receptor modulatory drugs as bed
herein.
Accordingly, use of an ErbB3 inhibitor (e.g., use thereof for the manufacture of a
medicament) for the treatment of hormone refractory breast cancer is provided. In another aspect, a
method is disclosed of suppressing growth of a hormone refractory breast cancer tumor (optionally an
estrogen receptor positive hormone refractory breast cancer tumor), the method comprising contacting
the tumor with an effective amount of an ErbB3 inhibitor. In another aspect, a method of suppressing
growth of a hormone refractory breast cancer tumor (optionally an en receptor positive hormone
refractory breast cancer tumor) in a patient is provided, the method comprising administering to the
patient an effective amount of an ErbB3 inhibitor. In yet another aspect, a method of treating a
patient for a hormone refractory breast cancer tumor (optionally an estrogen receptor positive
hormone refractory breast cancer tumor) is provided, the method comprising administering to the
patient an ive amount of an ErbB3 inhibitor. In still r , a method of treating a breast
cancer tumor in a patient is provided, the method comprising: selecting a patient with a hormone
refractory breast cancer tumor (optionally an en receptor positive e refractory breast
cancer tumor); and administering to the patient an ive amount of an ErbB3 tor.
In an exemplary embodiment, the ErbB3 tor is an anti-ErbB3 antibody. An exemplary
rbB3 antibody is Ab #6, sing VH and/or VL regions comprising the amino acid ces
set forth in SEQ ID NOs: 1 and 2, respectively. Another exemplary anti-ErbB3 antibody is an
antibody comprising, optionally in amino al to carboxy terminal order, VH CDR1, 2 and 3
sequences as shown in SEQ ID NOs: 3-5, respectively, and, optionally in amino terminal to carboxy
terminal order, VL CDR1, 2 and 3 sequences as shown in SEQ ID NOs: 6-8, respectively. In another
embodiment, the anti-ErbB3 antibody has heavy and light chains comprising the amino acid
sequences set forth in SEQ ID NOs 42 and 43, respectively. In other embodiments, the anti-ErbB3
antibody is Ab #3 (comprising VH and VL sequences as shown in SEQ ID NOs: 9 and 10,
respectively), Ab #14 (comprising VH and VL sequences as shown in SEQ ID NOs: 17 and 18,
respectively), Ab #17 (comprising VH and VL sequences as shown in SEQ ID NOs: 25 and 26,
respectively) or Ab #19 (comprising VH and VL sequences as shown in SEQ ID NOs: 33 and 34,
respectively). In another embodiment, administration of the anti-ErbB3 antibody inhibits growth or
invasiveness or metastasis of the tumor.
In another aspect, the treatment methods provided herein further comprise inistering to
the patient at least one additional anti-cancer agent that is not an ErbB3 inhibitor. In one embodiment,
the at least one additional ancer agent comprises at least one chemotherapeutic drug, such as a
drug(s) ed from the group consisting of platinum-based chemotherapy drugs, taxanes, tyrosine
kinase inhibitors, serine/threonine protein kinase inhibitors, anti-EGFR antibodies, anti-ErbB2
antibodies, bispecific anti-ErbB2/ErbB3 dies, and combinations thereof.
In another embodiment, the at least one additional anti-cancer agent comprises an EGFR
inhibitor, such as an anti-EGFR antibody or a small molecule inhibitor of EGFR signaling. A
preferred anti-EGFR antibody comprises cetuximab. Other examples of anti-EGFR antibodies
include MM-151, Sym004, matuzumab, panitumumab, nimotuzumab and mAb 806. An exemplary
small molecule inhibitor of EGFR signaling comprises gefitinib. Other examples of useful small
molecule inhibitors of EGFR signaling include but are not d to afatinib, nib, canertinib,
erlotinib HCL, pelitinib, 6, PD-158780, and AG 1478.
In yet another embodiment, the at least one additional anti-cancer agent comprises a vascular
endothelial growth factor (VEGF) inhibitor. An ary VEGF inhibitor comprises an anti-VEGF
dy, such as the bevacizumab antibody. In still r embodiment, the at least one additional
ancer agent comprises either or both of an estrogen receptor nist and an aromatase
inhibitor. Examples of en receptor antagonists include fene, tamoxifen, afimoxifene (4-
hydroxytamoxifen), arzoxifene, lasofoxone, toremifene and trant. Examples of aromatase
inhibitors include but are not limited to exemestane, ozole, letrozole, aminoglutethimide,
actone, vorozole, formestane and fadrozole. In one embodiment, the aromatase inhibitor is
letrozole. In still another embodiment, the at least one additional anti-cancer agent comprises a
serine/threonine protein kinase inhibitor, such as a ian target of rapamycin (mTOR) tor,
a phosphatidylinositolkinase (PI3K) inhibitor, or a mitogen activated kinase kinase (MEK)
inhibitor. Examples of mTOR inhibitors include but are not limited to temsirolimus, everolimus,
sirolimus, or ridaforolimus. Examples of PI3K inhibitors include but are not limited to CAL101 and
PX-866, both of which are currently being tested in clinical trials. Examples of MEK inhibitors
include but are not limited to XL518, CI-1040, 01, selumetinib, and GSK1 120212. In one
ment, the at least one additional anti-cancer agent comprises either or both of an mTOR
inhibitor and an aromatase inhibitor. In one embodiment, the at least one anti-cancer agent comprises
imus and exemestane. In yet another embodiment, the at least one additional anti-cancer agent
comprises an IGF1R inhibitor. Examples of IGF1R inhibitors include dalotuzumab, AMG-479,
R1507, figitumumab, 2, XL228, BMS-754807 and MM- 141.
In one embodiment, the hormone refractory breast cancer is ER+.
In a r aspect, provided herein are methods for inhibiting lin-mediated activation
of estrogen receptors in tumor cells, said method comprising 1) selecting a human patient who has
been treated for a malignancy with an anti-estrogen therapy and has become resistant to such therapy,
which patient has a malignant tumor, which tumor, by is of a tumor biopsy taken from the
patent after the patient has become resistant, is estrogen receptor positive and overexpresses HER2,
and which activation comprises phosphorylation of estrogen receptors, and 2) administering to the
t so selected an antibody that inhibits heregulin g to ErbB2/ErbB3 dimer, wherein
the antibody is administered at a dosage that yields a concentration of the antibody in the patient's
bloodstream that is a sufficient concentration to inhibit heregulin-induced estrogen receptor
phosphorylation in MCF7 cells in vitro by at least 20%, at least 30%, at least 40%, at least 50%, at
least 60%, or at least 70%, wherein said stration at said dosage is effective to treat the tumor.
The cell may be in a tumor that, by biopsy, is ER+ and HER2++ or HER2+++, or that contains at
least 0.02 pg HRG^g of n (e.g., by ELISA), or is HER2 FISH-positive. The inhibition is
accomplished by introducing into the extracellular fluid an antibody that inhibits heregulin binding to
ErbB3 heterodimer. In one embodiment the tumor is a malignant tumor.
Non-limiting examples of types of tumors to be treated include cancers of the breast, ovary,
lung, or skin (e.g., melanoma).
The tumor may be in a patient and the antibody introduced into the tream by
administration to the patient of an amount of the antibody that is effective to yield the sufficient
concentration of the antibody in the bloodstream. The administration may be by intravenous injection
or infusion. In one embodiment the antibody may be an anti-HER3 (anti-ErbB3) antibody, e.g., an
antibody having VH and/or VL regions comprising the amino acid sequences set forth in SEQ ID NOs:
1 and 2, tively. In another embodiment the antibody may be an anti-ErbB3 antibody comprising
VH and/or VL regions comprising the amino acid sequences set forth in SEQ ID NOs: 42 and 43,
respectively. The antibody may be an anti-HER2 (anti-ErbB2) antibody, e.g., C6.5, C6.5 diabody, or
umab. The antibody may also be an anti-ErbB2/anti-ErbB3 bispecific antibody. A number of
bispecific rbB2/anti-ErbB3 dies that are scFv human serum albumin (HSA) conjugates
are described in US patent publication 201 10059076, and PCT publication number WO2009/126920,
each of which discloses B2B3-1 and other bispecific anti-ErbB2/antiErbB3 antibodies that are scFv
HSA conjugates and that are suitable for use in the methods and compositions provided herein,
including ALM, A5-HSA-ML3.9, A5-HSA-B1D2, B12-HSA-B1D2, A5-HSA-F5B6H2, H3-HSAF5B6H2
, F4-HSA-F5B6H2, and -B1D2. In one embodiment, the bispecific antibody
comprises SEQ ID NO:44. Other le bispecific anti-ErbB2/antiErbB3 antibodies are disclosed
and claimed in US Patent Nos. 7,332,580 and 7,332,585. Preferably, administration of the dy
inhibits growth or invasiveness or metastasis of the tumor.
ingly, an ErbB3 inhibitor (e.g., an anti-ErbB3 antibody) or an anti-ErbB2 antibody or a
ific anti-ErbB2/ErbB3 antibody is provided (e.g., use thereof for the manufacture of a
medicament) for the inhibition of heregulin mediated estrogen receptor activation, and also or
alternately for the treatment of hormone refractory breast cancer (or another hormone refractory
cancer such as ovarian cancer, uterine cancer, or cervical cancer) or of aromatase resistant estrogen
receptor positive cancer such as breast cancer, n cancer, e cancer, or cervical cancer, is
disclosed. In an additional embodiment, an ErbB3 inhibitor, e.g., an anti-ErbB3 antibody or an anti-
ErbB2 antibody or a bispecific anti-ErbB2/ErbB3 antibody is provided (e.g., use thereof for the
manufacture of a medicament) for use in the treatment of an estrogen receptor positive cancer (e.g.,
breast , ovarian cancer, uterine cancer, or cervical cancer) in combination therapy with an
aromatase inhibitor. Such combinations retard or prevent the development of hormone resistance in
cancers treated with such combinations. In additional embodiments the method r comprises co
stration of either or both of an estrogen receptor antagonist and an aromatase inhibitor. In
further aspects herein provided are compositions for tion of heregulin-mediated activation of
estrogen receptor, said inhibition following selection of a human patient who has been treated for
malignancy with an anti-estrogen therapy and has become resistant to such therapy, which patient has
a malignant tumor, which tumor, by analysis of a tumor biopsy taken from the patent after the patient
has become resistant, is estrogen receptor positive and overexpresses ErbB2, and which activation
comprises phosphorylation of en receptors, said composition comprising: an anti-ErbB3
antibody that inhibits heregulin binding to ErbB2/ErbB3 heterodimer; an anti-ErbB2 antibody that
binds to ErbB2 and ts heregulin binding to ErbB3 heterodimer (e.g., pertuzumab); or
comprising an anti-ErbB2/antiErbB3 bispecific dy that ts heregulin binding to
ErbB2/ErbB3 heterodimer (e.g., the antibody comprising SEQ ID NO:44 (also referred to as SEQ ID
NO: 16 in U.S. patent publication No. 20110059076) . In some embodiments the cancer is a hormone
refractory en-receptor positive cancer.
In one embodiment, each of these compositions optionally comprises one or more of an
estrogen receptor antagonist and an aromatase inhibitor. Examples of estrogen receptor entagonists
include raloxifene, tamoxifen, afimoxifene roxytamoxifen), fene, lasofoxone, toremifene
and fulvestrant. es of aromatase inhibitors include exemestane, anastrozole, letrozole,
aminoglutethimide, testolactone, vorozole, formestane and fadrozole. In an exemplary embodiment,
the aromatase inhibitor is letrozole.
In r embodiment, each of these compositions optionally comprises one or more of an
mTOR inhibitor and an aromatase inhibitor. Examples of mTOR inhibitors include temsirolimus,
everolimus, sirolimus, or ridaforolimus. In an exemplary embodiment, the mTOR inhibitor is
everolimus. Examples of aromatase inhibitors include exemestane, anastrozole, letrozole,
aminoglutethimide, actone, vorozole, formestane and fadrozole. In an exemplary embodiment,
the aromatase inhibitor is tane.
In another embodiment, each of these itions optionally comprises one or more of a
MEK inhibitor, a PI3K inhibitor, and an IGF-1R inhibitor.
Brief Description of the Drawings
Figure 1 comprises images of western blots of a gel of lysates from untreated control cells
("C"), cells pretreated with ("MM121") or t pretreatment with MM-121 that were stimulated
with heregulin beta 1 ("HRG"), betacellulin ("BTC"), or estrogen ("E2"). The top panel shows
results from a blot probed with an dy specific to phosphorylated ErbB3 3), the middle
panel shows a blot probed with an antibody specific to phosphorylated (ser 167 and ser 118) en
receptor alpha (pER), and the bottom panel shows a blot probed with an antibody specific to
aldehydephosphate ogenase (GAPDH) as a g control.
Figure 2 is a graph showing densitometry results from the relevant (pER) band in each of the
control ("Con."), lin ("HRG"), betacellulin ("BTC"), heregulin plus MM-121
("HRG+MM121") and betacellulin plus MM-121 ("BTC+MM121") lanes in Figure 1. Band density
was normalized to GAPDH density and the normalized density (Y-axis) is shown for control and
stimulated cells (both with and without MM-121 pretreatment) as indicated on the X-axis. The
arrowhead between the HRG and BTC lanes indicates the pER band in the HRG lane.
Figure 3 is a graph showing tumor volume (y-axis) over time (in weeks, x axis) in a letrozole
resistant mouse xenograft model. Data are shown for mice treated with: PBS as a l (square),
MM-121 alone ("MM", triangle), letrozole alone ("Let", upside down triangle), and the combination
of MM-121 and letrozole (MM+Let, diamond). At the 14 week mark, the letrozole mice were split
into three groups: letrozole alone ("Let", upside down triangle), MM-121 alone ("Let->MM", circle)
and the combination of MM-121 and letrozole
Detailed Description
Provided herein are methods for treating hormone refractory breast cancers and other ER+
cancers, particularly those that overexpress HER2. Also ed are pharmaceutical compositions
for, and uses thereof in, such treatment. As described r in the Examples, it is believed that
ErbB3 tors, e.g., anti-ErbB3 antibodies, or other antibodies that can inhibit the binding of
heregulin to ErbB2/ErbB3 heterodimers, are able to suppress one or more of the growth, invasiveness
and asis of hormone refractory breast cancer cells in vivo. Accordingly, provided are methods
and compositions and uses thereof for suppressing the growth invasiveness or metastasis of hormone
refractory breast s (e.g., estrogen receptor positive hormone refractory breast cancers), as well
as methods and compositions for treating such breast cancers in patients, e.g., with an ErbB3
inhibitor.
ER+ cancers exemplify candidates for therapy regimens that include anti-estrogen agents.
Such cancers may include but are not limited to certain breast, ovarian, uterine, endometrial, lung,
bone, brain, bladder, liver and urogenital cancers.
A cancer may be an ErbB2 gene amplified cancer and/or an ErbB2 expressing (HER2+) or
overexpressing (HER2++, HER2+++) cancer. ErbB2, also known as HER2 or Neu, is a cell surface
transmembrane receptor n that tes intracellular signals (e.g., upon ligand activation) via
its intracellular tyrosine kinase activity. In excess, such signals can promote oncogenesis e.g., by
triggering cell on. The ErbB2 gene is amplified and/or overexpressed in many types of human
malignancies, including but not limited to breast, n, endometrial, pancreatic, colorectal,
prostate, salivary gland, skin, kidney, and lung. ErbB2 overexpressing cancers are designated a
+ or HER2++ depending on the level of ErbB2 overexpression, with HER2+++ indicating
the highest levels of HER2 expression. HER2+++ and HER2++ status are typically determined by an
assay such as immunohistochemistry (IHC), e.g., Herceptest® . ing to guidelines
provided by the College of American Pathologists (CAP) and the American Society of Clinical
Oncology (ASCO), a tumor designated HER2 negative is a tumor in which an IHC test shows no
staining or membrane staining in <30% of tumor cells; a tumor is designated "HER2"+ if an IHC test
results in faint membrane ng in > 30% of tumor cells, wherein only part of membrane is stained;
a tumor is designated "HER2++" if an IHC assay results in weak or moderate (complete) membrane
staining in >30% of tumor cells; and a tumor is designated "HER2+++" if an IHC test results in a
uniform, intense stain of >30% of the tumor cells. ErbB2 gene amplification is may be determined
by, e.g., FISH escence in situ hybridization), with HER2-amplified cancer cells being those that
have more than two HER2 gene copies being HER2-amplified, and cells and/or tumors comprising
HER2-amplified cancer cells being referred to as "FISH positive."
Definitions:
As used herein, the term "hormone refractory breast cancer" refers to breast cancer that is
resistant to the effects of anti-hormone y. A hormone refractory breast cancer is an estrogen
receptor positive breast cancer that is either de novo resistant to endocrine therapy or acquires
resistance while on treatment. About 25-50% of hormone-receptor-positive breast cancers are de
novo ant to endocrine therapy, and ially all metastatic breast cancers p acquired
resistance.
As used , the term "estrogen receptor positive" (ER+) refers to tumors (e.g.,
carcinomas), typically breast tumors, in which the tumor cells score positive (i.e., using conventional
histopathology methods) for en receptor (ER). According to recommendations provided by
CAP and ASCO, a tumor is ER+ if at least 1% of the tumor cells tested (e.g., by
immunohistochemistry) score ER positive.
The terms "ErbB3" and "HER3," as used interchangeably herein, refer to human ErbB3
protein, as described in U.S. Patent No. 5,480,968.
The terms ," "HER2," and "HER2 receptor," as used hangeably herein, refer to
the protein t of the human neu oncogene, also ed to as the ErbB2 oncogene or the HER2
oncogene.
As used herein, the term "ErbB3 inhibitor" is intended to include therapeutic agents that
inhibit, downmodulate, suppress or downregulate activity of ErbB3. The term is intended to include
chemical compounds, such as small molecule inhibitors, and biologic agents, such as antibodies,
interfering RNA (shRNA, siRNA), soluble receptors and the like. An exemplary ErbB3 inhibitor is
an anti-ErbB3 antibody.
An "antibody," as used herein is a protein consisting of one or more ptides comprising
g domains substantially encoded by immunoglobulin genes or fragments of immunoglobulin
genes, wherein the protein immunospecifically binds to an antigen. The recognized immunoglobulin
genes include the kappa, lambda, alpha, gamma, delta, epsilon and mu constant region genes, as well
as myriad immunoglobulin variable region genes. Light chains are classified as either kappa or
lambda. Heavy chains are classified as gamma, mu, alpha, delta, or epsilon, which in turn define the
immunoglobulin classes, IgG, IgM, IgA, IgD and IgE, respectively. A typical immunoglobulin
structural unit comprises a tetramer that is composed of two identical pairs of polypeptide chains,
each pair having one "light" (about 25 kD) and one "heavy" chain (about 50-70 kD). "VL" and VH"
refer to the variable regions of these light and heavy chains respectively.
Antibodies include intact immunoglobulins as well as antigen-binding fragments thereof,
which may be ed by digestion with various ases, or synthesized de novo either
chemically or using recombinant DNA sion technology. Such fragments include, for e,
F(ab)2 dimers and Fab monomers. Useful antibodies include single chain antibodies (antibodies that
exist as a single polypeptide chain), e.g., single chain Fv antibodies (scFv) in which a VH and a VL
chain are joined together (directly or through a peptide ) to form a continuous polypeptide.
"Immunospecific" or "immunospecifically" refer to antibodies that bind via domains
ntially encoded by immunoglobulin genes or fragments of immunoglobulin genes to one or
more epitopes of a protein of interest, but which do not substantially recognize and bind other
les in a sample containing a mixed population of antigenic molecules. Typically, an dy
binds immunospecifically to a cognate antigen with a Kd with a value of no greater than 50 nM, as
measured by a e plasmon resonance assay or a cell binding assay. The use of such assays is
well known in the art, and is described in e 3, below.
An "anti-ErbB3 antibody" is an antibody that immunospecifically binds to the ectodomain of
ErbB3 and an "anti-ErbB2 antibody" is an antibody that immunospecifically binds to the ectodomain
of ErbB2. The antibody may be an isolated antibody. Such binding to ErbB3 or ErB2 exhibits a Kd
with a value of no greater than 50 nM as measured by a surface plasmon resonance assay or a cell
binding assay. Exemplary anti-ErbB3 antibodies inhibit EGF-like ligand mediated phosphorylation of
ErbB3, e.g., anti-ErbB2 antibodies that t the binding of heregulin to ErbB2/ErbB3 heterodimers.
EGF-like ligands include EGF, TGFa, llulin, heparin-binding epidermal growth factor,
biregulin, epigen, epiregulin, and amphiregulin, which lly bind to ErbB 1 and induce
heterodimerization of ErbB 1 with ErbB3.
The term "bispecific antibody" as used herein refers to a protein comprising two antigenbinding
sites, a first binding site exhibiting immunospecific binding to a first antigen or epitope and a
second binding site exhibiting immunospecific binding to a second antigen or epitope ct from
the first. An anti-ErbB3/anti-ErbB2 bispecific antibody is an antibody that comprises two g
sites, one that immunospecifically binds to the ectodomain of ErbB3 and another that
immunospecifically binds to the ectodomain of ErbB2.
As used , the term "EGFR inhibitor" or "inhibitor of EGFR signaling" is intended to
include therapeutic agents that inhibit, downmodulate, suppress or gulate EGFR signaling
activity. The term is intended to include al compounds, such as small molecule inhibitors
(e.g., small le tyrosine kinase inhibitors) and biologic agents, such as antibodies, ering
RNA (shRNA, siRNA), e receptors and the like.
As used herein, the term "VEGF inhibitor" is intended to include therapeutic agents that
t, downmodulate, suppress or downregulate VEGF signaling activity. The term is intended to
include chemical compounds, such as small molecule inhibitors (e.g., small molecule tyrosine kinase
inhibitors) and biologic , such as antibodies, interfering RNA (shRNA, siRNA), soluble
receptors and the like.
As used herein, the term "mTOR inhibitor" is intended to include therapeutic agents that
inhibit, downmodulate, suppress or downregulate mammalian target of rapamycin . The term
is intended to include chemical compounds, such as small le inhibitors (e.g., small molecule
serine/threonine kinase inhibitors) and biologic agents, such as dies, ering RNA (shRNA,
siRNA), soluble receptors and the like.
As used herein, the term "MEK inhibitor" is intended to include therapeutic agents that
inhibit, downmodulate, suppress or downregulate mitogen activated protein kinase kinase (MEK).
The term is intended to include chemical compounds, such as small molecule inhibitors (e.g., small
molecule serine/threonine kinase inhibitors) and biologic , such as antibodies, interfering RNA
(shRNA, siRNA), soluble ors and the like.
As used herein, the term "PI3K inhibitor" is intended to include therapeutic agents that
inhibit, downmodulate, suppress or downregulate phosphatidylinositol-3 -kinase (PI3K). The term is
ed to include chemical compounds, such as small molecule inhibitors (e.g., small molecule
serine/threonine kinase inhibitors) and biologic agents, such as antibodies, ering RNA (shRNA,
siRNA), soluble receptors and the like.
The terms "suppress", "suppression", "inhibit" and "inhibition" as used interchangeably
herein, refer to any statistically significant decrease in ical activity (e.g., tumor cell ),
ing full blocking of the activity. For example, "inhibition" can refer to a decrease of about
%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, or 100% in biological activity.
The term "patient" includes a human or other mammalian animal that receives either
prophylactic or therapeutic treatment.
The terms "treat," "treating," and "treatment," as used herein, refer to therapeutic or
tative measures described herein. The methods of "treatment" employ administration to a
patient of an ErbB3 inhibitor such as those described herein, for example, a patient having a hormone
refractory breast cancer tumor, in order to cure, delay, reduce the severity of, or ameliorate one or
more symptoms of the e or er or ing disease or disorder, or in order to prolong the
survival of a patient beyond that expected in the absence of such treatment.
The term "effective amount," as used herein, refers to that amount of an agent, such as an
ErbB3 inhibitor, e.g., an anti-ErbB3 antibody, which is sufficient to effect treatment, prognosis or
sis of a hormone refractory breast cancer, when administered to a patient. A therapeutically
effective amount will vary depending upon the patient and disease condition being treated, the weight
and age of the patient, the severity of the e condition, the manner of administration and the like,
which can readily be determined by one of ordinary skill in the art. The dosages for administration
can range from, for example, about 1 ng to about 10,000 mg, about 5 ng to about 9,500 mg, about 10
ng to about 9,000 mg, about 20 ng to about 8,500 mg, about 30 ng to about 7,500 mg, about 40 ng to
about 7,000 mg, about 50 ng to about 6,500 mg, about 100 ng to about 6,000 mg, about 200 ng to
about 5,500 mg, about 300 ng to about 5,000 mg, about 400 ng to about 4,500 mg, about 500 ng to
about 4,000 mg, about 1 g to about 3,500 mg, about 5 g to about 3,000 mg, about 10 g to about
2,600 mg, about 20 g to about 2,575 mg, about 30 mg to about 2,550 mg, about 40 mg to about 2,500
mg, about 50 mg to about 2,475 mg, about 100 mg to about 2,450 mg, about 200 mg to about 2,425 mg,
about 300 mg to about 2,000, about 400 mg to about 1,175 mg, about 500 mg to about 1,150 mg, about
0.5 mg to about 1,125 mg, about 1 mg to about 1,100 mg, about 1.25 mg to about 1,075 mg, about 1.5
mg to about 1,050 mg, about 2.0 mg to about 1,025 mg, about 2.5 mg to about 1,000 mg, about 3.0
mg to about 975 mg, about 3.5 mg to about 950 mg, about 4.0 mg to about 925 mg, about 4.5 mg to
about 900 mg, about 5 mg to about 875 mg, about 10 mg to about 850 mg, about 20 mg to about 825
mg, about 30 mg to about 800 mg, about 40 mg to about 775 mg, about 50 mg to about 750 mg, about
100 mg to about 725 mg, about 200 mg to about 700 mg, about 300 mg to about 675 mg, about 400
mg to about 650 mg, about 500 mg, or about 525 mg to about 625 mg, of an antibody or antigen
binding portion thereof, as ed herein. Dosing may be, e.g., every week, every 10 days, every 2
weeks, every 18 days, every three weeks, every 4 weeks, every 5 weeks or every 6 weeks. Dosage
regimens may be adjusted to provide the optimum therapeutic response. An effective amount is also
one in which any toxic or detrimental effects (side s) of the ErbB3 inhibitor are minimized
and/or outweighed by the beneficial effects. For MM- 121, administration may be intravenous at
exactly or about 6 mg/kg or 12 mg/kg weekly, or 12 mg/kg or 24 mg/kg biweekly. For MM-1 11,
dosing may be intravenous at exactly or about every x days with an initial loading dose of exactly or
about y mg/kg and subsequent maintenance doses of exactly or about z mg/kg, where x, y and z are:
7, 25, and 20, or 10, 40 and 30, or 14, 60, and 44, or 18, 90, and 75, or 21, 120, and 105. Additional
preferred dosing regimens are described below.
The terms "anti-cancer agent" and "antineoplastic agent" refer to drugs used to treat
malignancies, such as cancerous growths. Drug therapy may be used alone, or in combination with
other treatments such as surgery or radiation y.
"Therapeutic synergy" refers to a phenomenon where treatment of patients with a
combination of therapeutic agents manifests a therapeutically superior outcome to the outcome
achieved by each individual constituent of the combination used at its optimum dose (T. H . Corbett et
al., 1982, Cancer Treatment Reports, 66, 1187). In this context a eutically superior outcome is
one in which the patients either a) exhibit fewer incidences of adverse events while receiving a
eutic t that is equal to or greater than that where dual constituents of the
combination are each administered as monotherapy at the same dose as in the combination, or b) do
not exhibit dose-limiting ties while ing a therapeutic benefit that is greater than that of
treatment with each individual constituent of the combination when each constituent is administered
in at the same doses in the combination(s) as is administered as individual ents. In xenograft
models, a ation, used at its maximum tolerated dose, in which each of the constituents will be
present at a dose generally not exceeding its individual maximum tolerated dose, manifests
therapeutic synergy when decrease in tumor growth achieved by administration of the combination is
greater than the value of the decrease in tumor growth of the best constituent when the constituent is
administered alone.
Thus, in combination, the components of such combinations have an additive or superadditive
effect on suppressing atic tumor , as compared to monotherapy with the anti-ErbB3
antibody or treatment with the chemotherapeutic(s) in the absence of dy therapy. By "additive"
is meant a result that is greater in extent (e.g., in the degree of reduction of tumor mitotic index or of
tumor growth or in the degree of tumor shrinkage or the frequency and/or duration of symptom-free
or symptom-reduced periods) than the best separate result ed by erapy with each
individual component, while "superadditive" is used to indicate a result that exceeds in extent the sum
of such separate results. In one embodiment, the additive effect is measured as slowing or stopping of
atic tumor growth. The additive effect can also be measured as, e.g., reduction in size of a
pancreatic tumor, reduction of tumor mitotic index, reduction in number of metastatic lesions over
time, increase in overall response rate, or increase in median or overall survival.
One non-limiting example of a measure by which effectiveness of a eutic treatment can
be quantified is by calculating the log 10 cell kill, which is ined according to the ing
equation:
log10 cell kill = T C (days)/3.32 x Td
in which T C ents the delay in growth of the cells, which is the average time, in days,
for the tumors of the treated group (T) and the tumors of the control group (C) to have reached a
predetermined value ( 1 g, or 10 mL, for example), and Td represents the time, in days necessary for
the volume of the tumor to double in the control s. When applying this measure, a product is
considered to be active if loglO cell kill is greater than or equal to 0.7 and a product is considered to
be very active if loglO cell kill is greater than 2.8. Using this measure, a ation, used at its own
maximum tolerated dose, in which each of the constituents is present at a dose generally less than or
equal to its maximum tolerated dose, exhibits therapeutic synergy when the loglO cell kill is greater
than the value of the loglO cell kill of the best constituent when it is administered alone. In an
exemplary case, the loglO cell kill of the combination exceeds the value of the loglO cell kill of the
best constituent of the combination by at least 0 .1 log cell kill, at least 0.5 log cell kill, or at least 1.0
log cell kill. Various aspects and embodiments are described in r detail in the following
subsections.
I . ErbB3 Inhibitors
As described in further detail herein, the methods and itions provided herein involve
the use of one or more ErbB3 inhibitors.
In one embodiment, the ErbB3 inhibitor is an anti-ErbB3 antibody, e.g., a monoclonal
antibody. Useful rbB3 antibodies (or VH/VL domains derived therefrom) can be made using
methods well known in the art. Alternatively, art recognized anti-ErbB3 antibodies can be used. For
example, Ab#3, Ab #14, Ab #17, Ab # 19, described in U.S. 7,846,440, can be used. Antibodies that
compete with any of these antibodies for binding to ErbB3 also can be used. onal art-
recognized anti-ErbB3 antibodies which can be used include those disclosed in US 7,285,649;
US202003 10557; US20100255010, as well as antibodies IB4C3 and 2D1D12 (U3 Pharma Ag), both
of which are bed in e.g., US20040197332 and are produced by hybridoma cell lines DSM ACC
2527 or DSM ACC 2517 (deposited at DSMZ); anti-ErbB3 antibody referred to as AMG888 (U3 U3 Pharma Ag and Amgen) described in U.S. patent No. 7,705,130; and onal antibody
8B8 (ATCC® HB-12070™), described in US 5,968,511, and the anti-ErbB3 antibody referred to as
AV-203 (Aveo Pharmaceuticals) which is described in US patent publication No. 20110256154.
Other useful rbB3 antibodies are disclosed in the art in the context of a bispecific antibody (see
e.g., B2B3-1 or B2B3-2 in WO/2009126920 and those described in US 7,846,440, US 20090291085,
US 20100056761, and US 66584. An exemplary anti-ErbB3 monoclonal antibody comprises
MM-121, a fully human rbB3 antibody currently undergoing Phase II clinical trials. MM-121 is
described further in PCT Publication No. and U.S. Patent No. 7,846,440, and
comprises VH and VL sequences as shown in SEQ ID NOs: 1 and 2, tively. MM-121 is referred
to as "Ab #6" in US 7,846,440. Alternately, the anti-ErbB3 monoclonal antibody is an antibody that
competes with MM-121 for binding to ErbB3. In another embodiment, the anti-ErbB3 antibody is an
dy comprising the VH and VL CDR sequences of MM-121, which are shown in SEQ ID NOs: 3-
(VH CDRl, 2, 3) and 6-8 (VL CDR1, 2, 3), respectively. In another embodiment, the anti-ErbB3
antibody has heavy and light chains sing the amino acid sequences set forth in SEQ ID NOs 42
and 43, respectively. Other examples of anti-ErbB3 antibodies e Ab #3, Ab #14, Ab #17 and
Ab #19, also described further in and having VH and VL sequences as shown in
SEQ ID NOs: 9 and 10, 17 and 18, 25 and 26, and 33 and 34 respectively. In another embodiment,
the anti-ErbB3 antibody is an antibody sing the VH and VL CDR sequences of Ab # 3 (shown
in SEQ ID NOs: 11-13 and 14-18, respectively) or antibody comprising the VH and VL CDR
sequences of Ab # 14 (shown in SEQ ID NOs: 19-21 and 22-24, respectively) or an dy
comprising the VH and VL CDR sequences of Ab # 17 (shown in SEQ ID NOs: 27-29 and 30-32,
respectively) or an antibody comprising the VH and VL CDR sequences of Ab # 19 (shown in SEQ ID
NOs: 35-37 and 38-40, tively).
Alternately, the anti-ErbB3 antibody is a monoclonal antibody or antigen binding portion
thereof which binds an epitope of human ErbB3 comprising residues 92-104 of SEQ ID NO:41 and is
characterized by inhibition of eration of a cancer cell expressing ErbB3. The cancer cell may be
a 3M cell, an AdrR cell, or an ACHN cell and the eration may be reduced by at least
% relative to control. In an additional ment this isolated monoclonal antibody or antigen
binding portion thereof binds an epitope comprising es 92-104 and 129 of SEQ ID NO:41.
In yet another embodiment, the anti-ErbB3 antibody can comprise a mixture, or cocktail, of
two or more anti-ErbB3 antibodies, each of which binds to a different epitope on ErbB3. In one
embodiment, the mixture, or cocktail, comprises three anti-ErbB3 antibodies, each of which binds to a
different epitope on ErbB3.
In another embodiment, the ErbB3 inhibitor ses a nucleic acid molecule, such as an
RNA molecule, that inhibits the expression or activity of ErbB3. RNA antagonists of ErbB3 have
been described in the art (see e.g., U.S. Patent Application Publication No. 20080318894). Moreover,
interfering RNAs specific for ErbB3, such as shRNAs or siRNAs that specifically inhibits the
expression and/or activity of ErbB3, have been described in the art.
In yet another embodiment, the ErbB3 inhibitor comprises a soluble form of ErbB3 that
inhibits signaling through the ErbB3 pathway. Such e ErbB3 molecules have been described in
the art (see e.g., U.S. Patent No. 7,390,632, U.S. Patent No. 7,638,303 and U.S. Patent No. 302 ,
each by Maihle et al, and U.S. Patent No. 7,919,098 by Zhou).
II. Anti-ErbB2 antibodies
The methods and compositions provided herein may involve the use of one or more anti-
ErbB2 antibodies that can inhibit the binding of heregulin to ErbB2/ErbB3 heterodimers. Suitable
anti-ErbB2 antibodies e C6.5 (and the numerous derivatives thereof) described in U.S. Patent
No. 5,977,322, as well as trastuzumab, as described in U.S. Patent No. 6,054,297, or umab, as
described in U.S. Patent No. 6,949,245.
III. Bispecific antibodies
The methods and compositions provided herein may involve the use of one or more bispecific
antibodies, preferably ones that can inhibit the binding of heregulin to ErbB2/ErbB3 heterodimers.
Such bispecific dies include ALM, as described in US patent 580, as well as A5-HSA-
ML3.9, A5-HSA-B1D2, B12-HSA-B1D2, A5-HSA-F5B6H2, H3-HSA-F5B6H2, F4-HSA-F5B6H2,
and H3-HSA-B1D2, as described in U.S. Patent Application Publication No. 20110059076, and PCT
publication number WO2009/126920, each of which, as described therein, have t forms such as
those comprising mHSA. In one embodiment, the bispecific antibody comprises SEQ ID NO:44.
IV. Methods
In one aspect, use of an ErbB3 inhibitor for the manufacture of a medicament for the
treatment of hormone refractory breast cancer is ed, in certain embodiments the breast cancer is
estrogen receptor positive hormone refractory breast cancer.
In another aspect, a method of suppressing growth of a hormone tory breast cancer cell
(optionally an ER+ hormone refractory breast cancer cell) is provided, the method comprising
contacting the cell with an effective amount of an ErbB3 inhibitor.
In another aspect, a method of suppressing growth of a hormone refractory breast cancer
tumor (optionally an ER+ e tory breast cancer tumor) in a patient is provided, the
method sing administering to the patient an effective amount of an ErbB3 inhibitor.
In still another aspect, a method of treating a breast cancer tumor (optionally an estrogen
receptor ve hormone tory breast cancer tumor) in a patient is provided, the method
comprising:
selecting a patient with a hormone refractory breast cancer tumor; and
administering to the patient an effective amount of an ErbB3 inhibitor.
In another aspect, the patient with a hormone refractory breast cancer tumor is a patient
r selected by use of the selection methods disclosed in pending international application
The hormone refractory breast cancer to be treated with ErbB3 inhibitor may co-express
ErbB 1 (EGFR), ErbB3, and heregulin (HRG). Expression of EGFR and HRG can be identified by
RT-PCR or by standard assay techniques, such as ELISA assay, immunohistochemical
staining of in-fixed, in-embedded tissues (e.g., breast cancer tissues routinely processed
for histological evaluation), using an anti-EGFR antibody, anti-ErbB3 antibody or an anti-HRG
antibody. Additional characteristics of preferred tumors for treatment in accordance with the
disclosure herein are set forth in pending U.S. Patent Publication No. 20110027291, which claims
priority to PCT application No. 2009/05405 1.
In one embodiment, the ErbB3 inhibitor administered to the patient is an anti-ErbB3 antibody.
An exemplary anti-ErbB3 antibody is MM- 121, comprising VH and VL sequences as shown in SEQ
ID NOs: 1 and 2, respectively, or an antibody comprising VH CDR1, 2 and 3 sequences as shown in
SEQ ID NOs: 3-5, respectively, and VL CDR1, 2 and 3 ces as shown in SEQ ID NOs: 6-8,
respectively (i.e., the VH and VL CDRs of MM-121). Additional non-limiting exemplary anti-ErbB3
antibodies and other forms of ErbB3 inhibitors are described in detail in Subsection I above.
The ErbB3 inhibitor can be administered to the patient by any route suitable for the effective
delivery of the inhibitor to the patient. For example, many small molecule inhibitors are suitable for
oral administration. Antibodies and other biologic agents lly are administered parenterally, e.g.,
intravenously, intraperitoneally, aneously or intramuscularly. Various routes of administration,
s and pharmaceutical formulations suitable for use in the methods provided herein are
described in further detail below.
In further aspects, the methods described herein include s inhibition (e.g., at least
partial blockade) of heregulin-mediated activating phosphorylation of estrogen receptors. These
methods involve the use of one or more antibodies that can inhibit the g of heregulin to
ErbB2/ErbB3 heterodimers to inhibit such phosphorylation. In certain embodiments, such methods
further include optional inistration of hormone modulatory drugs, including estrogen receptor
antagonists and aromatase inhibitors.
V. Pharmaceutical Compositions
In another aspect, pharmaceutical compositions are provided that can be used in the methods
sed herein, i.e., pharmaceutical compositions for treating e refractory breast cancer
In one embodiment, the pharmaceutical composition for treating hormone refractory breast
cancer ses an ErbB3 inhibitor and a pharmaceutical carrier. The ErbB3 inhibitor can be
formulated with the ceutical carrier into a pharmaceutical composition. Additionally, the
pharmaceutical composition can include, for example, instructions for use of the composition for the
treatment of patients for e refractory breast cancer tumors.
In one embodiment, the ErbB3 inhibitor in the composition is an anti-ErbB3 antibody, e.g.,
MM-121 or an antibody comprising the VH and VL CDRs of MM-121 positioned in the antibody in
the same relative order as they are present in MM-121 so as to provide immunospecific binding of
ErbB3. Additional non-limiting exemplary anti-ErbB3 antibodies and other forms of ErbB3 inhibitors
are described in detail in Subsection I above.
As used herein, "pharmaceutically acceptable carrier" includes any and all solvents,
dispersion media, coatings, antibacterial and ngal agents, isotonic and tion delaying
agents, buffers, and other excipients that are physiologically compatible. Preferably, the carrier is
suitable for parenteral, oral, or topical administration. Depending on the route of administration, the
active compound, e.g., small molecule or biologic agent, may be coated in a material to t the
compound from the action of acids and other natural conditions that may inactivate the nd.
Pharmaceutically able carriers include sterile aqueous solutions or dispersions and
sterile powders for the extemporaneous ation of sterile injectable solutions or dispersion, as
well as conventional excipients for the preparation of tablets, pills, capsules and the like. The use of
such media and agents for the formulation of pharmaceutically active substances is known in the art.
Except insofar as any conventional media or agent is incompatible with the active compound, use
thereof in the pharmaceutical compositions provided herein is contemplated. Supplementary active
compounds can also be incorporated into the compositions.
A pharmaceutically acceptable carrier can include a pharmaceutically acceptable antioxidant.
Examples of ceutically-acceptable antioxidants include: (1) water soluble antioxidants, such as
ascorbic acid, cysteine hydrochloride, sodium bisulfate, sodium metabisulfite, sodium sulfite and the
like; (2) luble antioxidants, such as ascorbyl ate, butylated hydroxyanisole (BHA),
butylated hydroxytoluene (BHT), lecithin, propyl gallate, alpha-tocopherol, and the like; and (3) metal
chelating agents, such as citric acid, nediamine tetraacetic acid (EDTA), ol, tartaric acid,
phosphoric acid, and the like.
Examples of suitable aqueous and nonaqueous carriers which may be employed in the
pharmaceutical itions provided herein include water, ethanol, polyols (such as glycerol,
propylene glycol, polyethylene glycol, and the like), and suitable es thereof, and injectable
organic esters, such as ethyl oleate. When required, proper fluidity can be maintained, for example,
by the use of coating als, such as lecithin, by the maintenance of the ed particle size in the
case of dispersions, and by the use of tants. In many cases, it will be preferable to include
isotonic agents, for example, sugars, polyalcohols such as mannitol, sorbitol, or sodium de in the
composition. Prolonged tion of the injectable compositions can be brought about by including
in the composition an agent that delays absorption, for e, monostearate salts and gelatin.
These compositions may also contain functional excipients such as preservatives, wetting
agents, emulsifying agents and dispersing agents.
Therapeutic compositions typically must be sterile, non-pyrogenic, and stable under the
conditions of manufacture and storage. The composition can be formulated as a solution,
microemulsion, liposome, or other ordered structure suitable to high drug tration.
Sterile injectable solutions can be prepared by incorporating the active compound in the
required amount in an appropriate solvent with one or a combination of ients enumerated
above, as required, followed by sterilization, e.g., by microfiltration. Generally, dispersions are
prepared by incorporating the active compound into a sterile vehicle that contains a basic dispersion
medium and the required other ingredients from those enumerated above. In the case of e
powders for the preparation of sterile injectable ons, the preferred methods of ation are
vacuum drying and freeze-drying (lyophilization) that yield a powder of the active ingredient plus any
additional desired ingredient from a previously sterile-filtered solution thereof. The active agent(s)
may be mixed under sterile conditions with additional pharmaceutically acceptable carrier(s), and
with any preservatives, buffers, or propellants which may be required.
Prevention of presence of microorganisms may be ensured both by sterilization procedures,
supra, and by the inclusion of various antibacterial and antifungal agents, for example, paraben,
chlorobutanol, phenol sorbic acid, and the like. It may also be desirable to include ic agents,
such as sugars, sodium chloride, and the like into the compositions. In addition, prolonged tion
of the injectable pharmaceutical form may be brought about by the inclusion of agents which delay
absorption such as aluminum monostearate and gelatin.
Pharmaceutical compositions sing an ErbB3 inhibitor can be administered alone or in
combination therapy. For example, the combination therapy can include a composition provided
herein comprising an ErbB3 inhibitor and at least one or more additional therapeutic agents, such as
one or more chemotherapeutic agents known in the art, discussed in further detail in Subsection IV
below. ceutical compositions can also be administered in conjunction with ion therapy
and/or surgery.
Dosage regimens are adjusted to provide the optimum desired response (e.g., a therapeutic
response). For example, a single bolus may be administered, l divided doses may be
administered over time or the dose may be proportionally reduced or increased as indicated by the
exigencies of the therapeutic situation.
ary dosage ranges for administration of an antibody include: 10-1000 mg
(antibody)/kg (body weight of the patient), 10-800 mg/kg, 10-600 mg/kg, 10-400 mg/kg, 10-200
mg/kg, 30-1000 mg/kg, 30-800 mg/kg, 30-600 mg/kg, 30-400 mg/kg, 30-200 mg/kg, 50-1000 mg/kg,
50-800 mg/kg, 50-600 mg/kg, 50-400 mg/kg, 50-200 mg/kg, 100-1000 mg/kg, 100-900 mg/kg, 100-
800 mg/kg, 100-700 mg/kg, 100-600 mg/kg, 100-500 mg/kg, 100-400 mg/kg, 100-300 mg/kg and
100-200 mg/kg. Exemplary dosage schedules include once every three days, once every five days,
once every seven days (i.e., once a week), once every 10 days, once every 14 days (i.e., once every
two weeks), once every 2 1 days (i.e., once every three , once every 28 days (i.e., once every
four weeks) and once a month.
It may be advantageous to ate parenteral itions in unit dosage form for ease of
stration and uniformity of . Unit dosage form as used herein refers to physically
discrete units suited as unitary dosages for the patients to be treated; each unit contains a
predetermined quantity of active agent calculated to produce the desired therapeutic effect in
association with any required pharmaceutical carrier. The specification for unit dosage forms are
ed by and directly dependent on (a) the unique characteristics of the active compound and the
particular therapeutic effect to be achieved, and (b) the limitations inherent in the art of compounding
such an active compound for the treatment of sensitivity in individuals.
Actual dosage levels of the active ingredients in the pharmaceutical compositions disclosed
herein may be varied so as to obtain an amount of the active ient which is effective to achieve
the desired eutic response for a particular patient, composition, and mode of administration,
without being toxic to the patient. teral" as used herein in the context of administration means
modes of administration other than enteral and l administration, usually by injection, and
includes, t limitation, intravenous, intramuscular, intraarterial, intrathecal, intracapsular,
intraorbital, intracardiac, intradermal, intraperitoneal, transtracheal, subcutaneous, subcuticular,
intraarticular, subcapsular, subarachnoid, pinal, epidural and intrasternal injection and infusion.
The phrases "parenteral administration" and "administered parenterally" as used herein refer
to modes of administration other than enteral (i.e., via the digestive tract) and topical administration,
usually by injection or infusion, and includes, without limitation, intravenous, intramuscular,
intraarterial, intrathecal, intracapsular, intraorbital, intracardiac, intradermal, intraperitoneal,
transtracheal, subcutaneous, subcuticular, intraarticular, sular, subarachnoid, intraspinal,
epidural and intrasternal injection and infusion. Intravenous injection and infusion are often (but not
exclusively) used for antibody administration.
When agents provided herein are administered as pharmaceuticals, to humans or animals, they
can be given alone or as a pharmaceutical composition containing, for e, 0.001 to 90% (more
preferably, 0.005 to 70%, such as 0.01 to 30%) of active ingredient in combination with a
pharmaceutically acceptable carrier.
VI. Combination Therapy
In certain embodiments, the methods and uses provided herein for suppressing growth of
e refractory breast cancer cells or for treating a patient with a hormone refractory breast tumor
or can se administration of an ErbB3 inhibitor and at least one additional ancer agent that
is not an ErbB3 inhibitor.
In one embodiment, the at least one additional anti-cancer agent comprises at least one
chemotherapeutic drug. Non-limiting examples of such chemotherapeutic drugs include platinumbased
chemotherapy drugs (e.g., tin, carboplatin), taxanes (e.g., axel (Taxol®), docetaxel
(Taxotere®), EndoTAG-1™ (a formulation of axel encapsulated in positively d lipid-
based complexes; MediGene®), Abraxane® (a formulation of paclitaxel bound to albumin), ne
kinase inhibitors (e.g., imatinib/Gleevec ® , sunitinib/Sutent ® , dasatinib/Sprycel ®), and combinations
thereof.
In another embodiment, the at least one additional anti-cancer agent comprises an EGFR
inhibitor, such as an anti-EGFR dy or a small molecule inhibitor of EGFR signaling. An
exemplary anti-EGFR antibody is cetuximab (Erbitux® , ImClone Systems). Other examples of anti-
EGFR antibodies include MM-151 er described in Bukhalid et al., copending commonly
assigned U.S. Patent ation Serial No. 61/504,633, filed on July 5, 2011), Sym004 (Symphogen,
Pederson et al., Cancer Research January 15, 2010 70; 588, also see U.S. Patent Serial No.
7,887,805), matuzumab (EMD72000), panitumumab (Vectibix® ; Amgen); nimotuzumab
(TheraCIM™) and mAb 806. An exemplary small molecule inhibitor of the EGFR signaling pathway
is nib (Iressa®), which is commercially ble from AstraZeneca and Teva. Other examples
of small molecule inhibitors of EGFR signaling include erlotinib HCL 74; Tarceva® ; OSI
Pharma), lapatinib (Tykerb® , GlaxoSmithKline), canertinib (canertinib dihydrochloride, ),
pelitinib (Pfizer); PKI-166 (Novartis); PD158780; afatinib (Tomtovok® , Boehringer Ingleheim); and
AG 1478 (4-(3-Chloroanillino)-6,7-dimethoxyquinazoline).
In yet another embodiment, the at least one additional anti-cancer agent comprises a VEGF
inhibitor. An exemplary VEGF inhibitor comprises an anti-VEGF antibody, such as bevacizumab
(Avastatin® ; Genentech).
In another embodiment, the at least one additional anti-cancer agent comprises an IGF1R
inhibitor, such as an anti-EGFR antibody or a small molecule inhibitor of EGFR signaling. Examples
of anti-IGFIR inhibitors include dalotuzumab (Merck, also MK-0646), AMG-479 ), R1507
(Roche), figitumumab (Pfizer), IMC-A12 (Imclone/Lilly), and MM-141, a bispecific ErbB3/IGFlR
inhibitor (further described in Lugovskoy et al., copending commonly assigned U.S. Patent
Application Serial No. 61/558,192, filed 201 1). es of small molecule IGF1R inhibitors
include XL228 xis) and BMS-754807 (BMS).
Examples
Example 1 : MM-121 treatment of ER+, hormone refractory mammary tumors
Analyses of the anti-tumor efficacy and tolerability of MM- 121 treatment of ER+ hormone
tory mammary tumor-bearing mice are carried out using xenografts of tamoxifen-resistant
variants of MCF7 human mammary carcinoma cells. Tamoxifen-resistant human mammary
carcinoma cell lines TAMR-1, TAMR-7, and TAMR-8 cells are obtained from the laboratory of A . E .
Lykkesfeldt (Department of Tumor Endocrinology, Division for Cancer Biology, Danish Cancer
y. Strandboulevarden 49, DK-2100 Copenhagen 0, Denmark). These are grown as xenografts
in female athymic nu+/nu+ nude mice obtained from Charles River Laboratories International. SCID
mice (C.B.-17/IcrACCscid ) obtained from the Arizona Cancer Center breeding colony, Tucson, AZ,
are also le. The mice are housed in Tecniplast® Individually Ventilated polycarbonate
(Makrolon®) Cages (IVC) set in climate-controlled rooms and have free access to food and acidified
water. Mice are injected under general anesthesia with ~ (about) 107 TAMR-1, TAMR-7, or TAMR-8
cells either subcutaneously in the flank or into the mammary fat pad.
To investigate anti-tumor efficacy in monotherapy, MM-121 or e control (IOOm ) is
given to tumor-bearing mice (i.e., after 14 days of tumor growth following injection of cells) at 600 g
per mouse (MM-121 as a 6 mg/mL solution in PBS) by IP ion every three days. Control mice
receive the PBS vehicle only. Efficacy is determined by comparing tumor growth between the
antibody-treated mice and the vehicle l mice and is expressed as the mental to control
ratio of median relative tumor s (T/C value). A minimum T/C value below 50% is a
uisite for rating a treatment as effective. The control and experimental groups each contain 10
mice bearing one tumor each. To obtain 30 mice bearing tumors of similar sizes for randomization,
40 mice per tumor are implanted erally.
Mice are randomized and therapy begins when a sufficient number of individual tumors have
grown to a volume of approximately 200 mm3. Tumors are measured (L x W) by digital caliper
measurement and the tumor volume is calculated using the formula p/6 (W2 x L). The first dose is
administered either on Day 0 (day of randomization) or one day later.
Approximately 24 hours after administration of the final dose all mice are bled to prepare
serum; in addition, tumors are collected from the same mice for freezing and paraffin
embedding (FFPE) (1/2 tumor each).
According to regulations for animal experiments, mice are sacrificed if the tumor volume
exceeds 1800 mm3 (one tumor per mouse). Mice are monitored and dosed until their tumors have
grown to that size but no longer than 60 days. Thereafter, they are sacrificed for sample collection.
At the end of the study, approximately 24 hours after administration of the final dose, all mice
on study are bled sublingually to obtain a maximum amount of blood for the preparation of serum.
Serum is aliquoted in 2 tubes with imately 250 E in each.
In addition, tumors from all mice are excised without delay for snap-freezing in liquid
en (1/2 tumor, COVARIS bags for the storage of samples are provided) and for fixation in 10%
buffered formalin for <24 hours, subsequent dehydration and paraffin embedding (1/2 tumor).
Animal weights and tumor ers (W and L) are ed twice weekly and tumor
volumes are calculated using the formula p/6 (W2xL). Tumor growth curves are plotted. Tumor
tion and absolute growth delay for 2 and 4 doubling times are ated.
Results of experiments carried out substantially as described will demonstrate that MM- 121
treatment inhibits or stops tumor growth, and in some cases reduces tumor size.
Example 2 : MM-121 inhibition of HRG-induced ER phosphorylation in vitro
MCF7 cells are either untreated or pretreated with MM- 121 (250nM) for 1 hour. Cells are
then stimulated with heregulin betal (EGF , lOnM R&D systems), betacellulin (20nM, R&D
systems) or estrogen (beta estradiol- lOOnM, Sigma) for 30 minutes, or left unstimulated. Lysates of
the cells are ed by western blot probed for pER and for pErbB3.
To demonstrate the ability of MM- 121 to reduce heregulin-induced activation of the estrogen
receptor, treatments were tested in the ER+, PR+, ErbB2 + cancer cell line MCF7 using the methods
described above or trivial variations thereof. Cells were either ted or pretreated with MM- 121.
Untreated and pretreated cells were stimulated with heregulin, betacellulin, or estrogen. Cell lysates
were analyzed by western blot for phosphorylated forms of ErbB3 and estrogen receptor.
As shown in Figure 1 (western blot) and Figure 2 (densitometry of the data in Figure 1),
untreated heregulin-stimulated cells, and to a lesser extent (about 2/3 less) untreated betacellulin-
stimulated cells, exhibited phosphorylation of both the estrogen receptor and ErbB3. In contrast,
heregulin-stimulated cells pretreated with MM- 121 exhibited a substantial (about 2/3) reduction in the
amount of pErbB3 and pER. The results demonstrate that heregulin-induced activation of the
estrogen receptor is mediated by ErbB3 and that MM- 121 can inhibit this mode of estrogen receptor
activation. Surprisingly, heregulin stimulation induced a dramatically (at least four-fold) higher level
of ER phosphorylation than did estrogen.
e 3 : Restoring sensitivity and/or preventing resistance to aromatase tors
by co-administration with MM-121
Aromatase tor (AI) ent is well tolerated by patients, and the therapy is effective
for a relatively long period. However, patients who are initially responsive to AI treatment can
become resistant to the drug. To investigate the mechanism of AI resistance, a xenograft model was
developed that corresponds to ER + postmenopausal breast cancer. Tumors for this intratumoral
aromatase xenograft model are grown from MCF7 human breast adenocarcinoma cells that have been
stably transfected with a human placental aromatase gene to provide a non-ovarian source of estrogen
production in ovariectomized athymic mice (MCF-7CA, see e.g. Brodie et al., Clinical Cancer
Research 884s Vol. 11, 884s - 888s, January 15, 2005 (Suppl.)). Sufficient estrogen is produced
(from aromatization of injected androstenedione) by the MCF7-CA cells to stimulate their
eration and tumor formation. This is a model of a postmenopausal breast cancer patient with
tumors that express aromatase and are free of gonadotropin feedback regulation.
MCF-7CA tumors: MCF-7CA cells were cultured in Eagle's minimum essential medium
containing 5% fetal bovine serum and neomycin. The culture medium was changed twice weekly.
Subconfluent MCF-7CA cells were scraped into Hank's solution and fuged at 1,000 rpm for 2
min at 4°C. The cells were then resuspended in el™ (lOmg/ml) to make a cell sion of 2-
x 107 cells/ml. Ovariectomized female BALB/c athymic mice 4-6 weeks of age (20-22g body
weight) were housed in a pathogen-free environment under lled conditions of light and
humidity and received food and water ad libitum. Each mouse was inoculated subcutaneously (s.c.)
with 0.1 ml of the cell suspension. Animals were then ed s.c. daily with 0.1 mg
tenedione/mouse. Growth rates were determined by measuring the tumors with calipers
weekly. Tumor volumes were calculated according to the formula for a sphere (4/3 rl2. r 2). When
tumors d a measurable size, mice were divided into groups of 10 s with equivalent tumor
volumes (300mm3). Mice were d with 0.1 ml compounds in 0.3% hydroxypropylcellulose
(HPC) for 6 weeks.
Letrozole vs. ole + MM121 in Letrozole Resistant Model
To demonstrate the efficacy of MM-121 in combination with letrozole in a letrozole-resistant
xenograft model, tumor bearing mice were prepared as described above and randomized into 3 groups
of 10 mice each and one group of 30 mice, each containing mice with a similar size distribution of
tumors. For initial ent during the period when development of resistance was expected, one
group was d with PBS, Q3D, i.p. as a control; one group was treated with MM-121 alone
(600 g MM121 in .2ml PBS/mouse every 3 days i.p.); the se group a was treated with
letrozole alone (10 g/mouse/day); and a final group was treated with both letrozole and MM-121.
Tumors were measured weekly and tumor volume was calculated. Mice were sacrificed if tumors
continued to grow to volumes of greater than about 1400-1700 mm3. When the tumors in the group of
mice treated with letrozole alone became resistant and exceeded about 600 mm3 in volume, the group
was subdivided into 3 groups of ten mice each. To investigate the effect of MM-121 combination
therapy on these resistant tumors, treatment of these three groups continued as follows: one group
continued to receive letrozole alone (10 e/day), one group stopped receiving daily ole
and was treated with MM-121 (600 g Q3D, i.p.) alone, and one group was treated with a
combination of letrozole mouse/day) and MM-121(600mg Q3D, i.p.).
Results are shown in Figure 3. As indicated therein, the tumors in the MM-121 -treated
(triangle) and PBS only control (square) mice grew rapidly over the course of 5 weeks, which was the
final measurement for these groups. Tumor growth progressed more slowly in the three groups
receiving letrozole treatment alone (upside down arrow) and the group receiving combination
treatment with MM-121 and ole (diamond).
Letrozole resistance in the groups of mice receiving letrozole ent was defined as the
point where tumor volume increased to 600mm3. This tumor volume was reached in the groups
receiving letrozole alone after about 14 weeks of daily letrozole treatment. While the tumors in the
mice receiving either letrozole alone or MM-121 alone continued to increase in volume over time, the
tumors of the mice receiving the ation of MM-121 and letrozole decreased to well below the
600mm3 resistance threshold and maintained a reduced volume hout the rest of the study (19
weeks), thus demonstrating that the combination treatment overcomes acquired letrozole resistance.
In contrast to the mice receiving letrozole alone, the group receiving both ole and MM-
121 (diamond shape) from the start of treatment did not develop letrozole resistant tumors (i.e. the
tumors never reached a volume of 600mm3), thus demonstrating that treatment with the combination
ts the development of letrozole resistance.
e 4 : inistration of MM-121 with an mTOR inhibitor and an aromatase
inhibitor
To demonstrate whether the triple combination of MM-121 + exemestane + everolimus is
more effective than either exemestane alone or the combination of everolimus and exemestane in the
treatment of ER+ breast cancer, patients will be dosed with MM-121 alone, tane alone,
everolimus alone, the ation of everolimus and exemestane, and the combination of MM-121,
everolimus, and exemestane. MM-121 will be dosed, e.g., at a 40mg/kg loading dose on week 1,
followed by 20 mg/kg weekly maintenance dose administered over 60 minutes as an intravenous
infusion once per week; exemestane will be dosed at 25 mg administered orally once per day;
everolimus will be dosed at 10 mg administered orally once per day. ts will be treated until
radiologic or al progression of their disease is documented. The results will demonstrate that the
triple combination of MM- 121 + exemestane + everolimus is more effective than exemestane alone or
the combination of everolimus and exemestane in the treatment of ER+ breast cancer ts.
Equivalents
Those skilled in the art will recognize, or be able to ascertain using no more than routine
experimentation, many equivalents of the specific embodiments described herein. Such equivalents
are intended to be encompassed by the following claims. Any combinations of the embodiments
disclosed in the dependent claims are contemplated to be within the scope of the invention.
Incorporation by Reference
Each and every, issued patent, patent application and publication referred to herein is hereby
orated herein by reference in its entirety.
SUMMARY OF SEQUENCE LISTING
MM-121 V_1 CDR3 {SEQ ID NO:8{
CSYAGSSIFVI
Ab # 3 V_H amino acid segucncc {SEQ ID NO:9{
SGGGLVQPGGSLRLSCAASGFTFSAYNMRWVRQAPGKGLEWVSVIYPSGG
ATRYADSVKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYCARGYYYYGMDVWGQGTLVT
Ab # 3 V_1 amino acid scgucncc {SEQ ID NO:10{
QSVLTQPPSASGTPGQRVTISCSGSDSNIGRNYIYWYQQFPGTAPKLLIYRNNQRPSGV
PDRISGSKSGTSASLAISGLRSEDEAEYHCGTWDDSLSGPVFGGGTKLTVL
Ab # 3 V_H CDRI {SEQ ID NO:11{
AYNMR
Ab # 3 V_H CDR2 {SEQ ID NO:12{
VIYPSGGATRYADSVKG
Ab # 3 V_H CDR3 {SEQ ID NO:13{
GYYYYGMDV
Ab # 3 V_I CDRI {SEQ ID NO:14{
SGSDSNIGRNYIY
Ab # 3 V_I CDR2 {SEQ ID NO:15{
RNNQRPS
Ab # 3 V_1CDR3 {SEQ ID NO:16{
GTWDDSLSGPV
Ab # 14 V_H amino acid segucncc {SEQ ID NO:17{
EVQLLESGGGLVQPGGSLRLSCAASGFTFSAYGMGWVRQAPGKGLEWVSYISPSGG
HTKYADSVKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYCAKVLETGLLVDAFDIWGQGT
MVTVSS
Ab # 14 V_I amino acid scgucncc {SEQ ID NO: 18)
QYELTQPPSVSVYPGQTASITCSGDQLGSKFVSWYQQRPGQSPVLVMYKDKRRPSEIP
ERFSGSNSGNTATLTISGTQAIDEADYYCQAWDSSTYVFGTGTKVTVL
Ab #14 V_H CDR1 {SEQ ID NO:19{
AYGMG
Ab # 14 V_H CDR2 {SEQ ID N020!
YISPSGGHTKYADSVKG
Ab #14 V_H CDR3 {SEQ ID N021!
LVDAFDI
Ab # 14 V_I CDR1 {SEQ ID NO:22{
SGDQLGSKFVS
Ab # 14 V_I CDR2 {SEQ ID N023!
YKDKRRPS
Ab # 14 V_I CDR3 {SEQ ID N024)
QAWDSSTYV
Ab # 17 V_H amino acid segucncc {SEQ ID N025)
EVQLLESGGGLVQPGGSLRLSCAASGFTFSWYGMGWVRQAPGKGLEWVSYISPSGG
ITVYADSVKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYCARLNYYYGLDVWGQGTTVTVS
Ab # 17 V_1 amino acid scgucncc {SEQ ID N026)
QDIQMTQSPSSLSASVGDRITITCQASQDIGDSLNWYQQKPGKAPRLLIYDASNLETG
VPPRFSGSGSGTDFTFTFRSLQPEDIATYFCQQSANAPFTFGPGTKVDIK
Ab #17 V_H CDR1 {SEQ ID NO:27{
WYGMG
Ab # 17 V_H CDR2 {SEQ ID N028!
YISPSGGITVYADSVKG
Ab # 17 V_H CDR3 !SEQ ID N0:29!
LNYYYGLDV
Ab #17 V_1 CDR1 {SEQ ID N030!
GDSLN
Ab #17 V_I CDR2 gSEQ ID N031!
DASNLET
Ab #17 V_I CDR3 {SEQ ID N032!
QQSANAPFT
Ab # 19 V_H amino acid seguence {SEQ ID N033!
EVQLLESGGGLVQPGGSLRLSCAASGFTFSRYGMW WVRQAPGKGLEWVSYIGSSGG
PTYYVDSVKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYCAGGRGTPYYFDSWGQGTLVTV
Ab # 19 V_I amino acid seguence {SEQ ID N034!
QYELTQPASVSGSPGQSITISCTGTSSDIGRWNIVSWYQQ
GVSNRF
SGSKSGNTASLTISGLQAEDEADYYCSSYTSSSTWVFGGGTKLTVL
Ab #19 V_H CDR1 {SEQ ID N035!
RYGMW
Ab #19 V_H CDR2 {SEQ ID N036!
YIGSSGGPTYYVDSVKG
Ab #19 V_H CDR3 gSEQ ID N037!
GRGTPYYFDS
Ab #19 V_1 CDR1 {SEQ ID N038
TGTSSDIGRWNIVS
Ab #19 V_1 CDR2 {SEQ ID N039!
—26—
DVSNRPS
Ab # 19 V_1 CDR3 {SEQ ID NO:401
SSYTSSSTWV
ErbB3 {SEQ ID NO:41!
SEVGNSQAVCPGTLNGLSVTGDAENQYQTLYKLYERCEVVMGNLEIVLTGHNADLSFLQWI
REVTGYVLVAMNEFSTLPLPNLRVVRGTQVYDGKFAIFVMLNYNTNSSHALRQLRLTQLTEI
LSGGVYIEKNDKLCHMDTIDWRDIVRDRDAEIVVKDNGRSCPPCHEVCKGRCWGPGSEDCQ
TLTKTICAPQCNGHCFGPNPNQCCHDECAGGCSGPQDTDCFACRHFNDSGACVPRCPQPLVY
NKLTFQLEPNPHTKYQYGGVCVASCPHNFVVDQTSCVRACPPDKMEVDKNGLKMCEPCGG
LCPKACEGTGSGSRFQTVDSSNIDGFVNCTKILGNLDFLITGLNGDPWHKIPALDPEKLNVFR
TVREITGYLNIQSWPPHMHNFSVFSNLTTIGGRSLYNRGFSLLIMKNLNVTSLGFRSLKEISAG
RIYISANRQLCY
GQCLSCRNYSRGGVCVTHCNFLNGEPREFAHEAECFSCHPECQPMEGTATCNGSGSDTCAQ
CAHFRDGPHCVSSCPHGVLGAKGPIYKYPDVQNECRPCHENCTQGCKGPELQDCLGQTLVLI
GKTHLTMALTVIAGLVVIFMMLGGTFLYWRGRRIQNKRAMRRYLERGESIEPLDPSEKANK
VLARIFKETELRKLKVLGSGVFGTVHKGVWIPEGESIKIPVCIKVIEDKSGRQSFQAVTDHML
AIGSLDHAHIVRLLGLCPGSSLQLVTQYLPLGSLLDHVRQHRGALGPQLLLNWGVQIAKGMY
YLEEHGMVHRNLAARNVLLKSPSQVQVADFGVADLLPPDDKQLLYSEAKTPIKWMALESIH
FGKYTHQSDVWSYGVTVWELMTFGAEPYAGLRLAEVPDLLEKGERLAQPQICTIDVYMVM
VKCWMIDENIRPTFKELANEFTRMARDPPRYLVIKRESGPGIAPGPEPHGLTNKKLEEVELEP
DLEAEEDNLATTTLGSALSLPVGTLNRPRGSQSLLSPSSGYMPMNQGNLGESCQES
AVSGSSERCPRPVSLHPMPRGCLASESSEGHVTGSEAELQEKVSMCRSRSRSRSPRPRGDSAY
LLTPVTPLSPPGLEEEDVNGYVMPDTHLKGTPSSREGTLSSVGLSSVLGTEEEDEDE
EYEYMNRRRRHSPPHPPRPSSLEELGYEYMDVGSDLSASLGSTQSCPLHPVPIMPTAGTTPDE
DYEYMNRQRDGGGPGGDYAAMGACPASEQGYEEMRAFQGPGHQAPHVHYARLKTLRSLE
ATDSAFDNPDYWHSRLFPKANAQRT
MM-121 Heavy Chain Amino Acid Seguence {SEQ ID N042)
4VQ T4SGGG LVQPGGSLRA SCAASGFTFS {YVMAWVRQA PGKGLEWVSS
GWTLY FT: SRDVS<NTLY QMNST.RATD TAVYYCTRGA
_ (MAT.FDYWG .4VTVSSA SI<GBSVFEL ABCSRSIS.£S IAALGC4V<D
YFPTPVTVSW GVI TFPAVAQSSG AYSLSSVVTV PSSNFGTQTY
TCWVDI<PSV <VDKTV.3R< CCVjCBBCBA BBVAGBSVE'S EBBKBKDI
VICV {4'3Bfi VQEVWYVDGV EVHNA<IKPQ £4QbNSIEQV
34- VSV.TVVHQD .VGKTY<C< VSN<G.BAB fiKI S<IKGQ 'jBQVYI_IB
TL' BSTfiT IKNQ IC V<Gb' YBSD AV4'Wfi SNGQPENNY< DSDG
401 SFF_IYSKLTV '(SRWQQGNV FSCSVMHTA {NHYTQKSLS
MM-121 Light Chain Amino Acid Seguence {SEQ ID NO:431
PCT/L S2012/028792
QSALTQPASV SGSBGQS i SCiGiSSDVG SYVVVSWYQQ HBGKABKT
PSGV SNRFSGSKSG NTAST."T SG . Qi*'.,D*ZADYYC CSYAGSS._FV
:FGGGTKVTV LGQB {AABSV KQSNNKYAAS1 Lb' J:’J:’S S4' QA\I {ATLVCL VSDFYPGAVT
DGSPV KVGVETTKPS SY .S .TP'LQW KS {RSYSCRV
1GSiV1K1 VAL’AL'CS
Bis ecific antibod bed in US atent tion 20110059076 as SE ID NO:16 SE ID
QVQLQESGGGLVKPGGSLRLSCAASGFTFSSYWMSWVRQAPGKGLEWVANINRDGSASYY
VDSVKGRFTISRDDAKNSLYLQMNSLRAEDTAVYYCARDRGVGYFDLWGRGTLVTVSSAST
GGGSGGGGSQSALTQPASVSGSPGQSITISCTGTSSDVGGYNFVSWYQQHPGKAPK
LMIYDVSDRPSGVSDRFSGSKSGNTASLIISGLQADDEADYYCSSYGSSSTHVIFGGGTKVTVL
GAASDAHKSEVAHRFKDLGEENFKALVLIAFAQYLQQSPFEDHVKLVNEVTEFAKTCVADE
SAENCDKSLHTLFGDKLCTVATLRETYGEMADCCAKQEPERNECFLQHKDDNPNLPRLVRP
EVDVMCTAFHDNEETFLKKYLYEIARRHPYFYAPELLFFAKRYKAAFTECCQAADKAACLLP
KLDELRDEGKASSAKQRLKCASLQKFGERAFKAWAVARLSQRFPKAEFAEVSKLVTDLTKV
HTECCHGDLLECADDRADLAKYICENQDSISSKLKECCEKPLLEKSHCIAEVENDEMPADLPS
LAADFVESKDVCKNYAEAKDVFLGMFLYEYARRHPDYSVVLLLRLAKTYETTLEKCCAAA
DPHECYAKVFDEFKPLVEEPQNLIKQNCELFEQLGEYKFQNALLVRYTKKVPQVSTPTLVEV
SRNLGKVGSKCCKHPEAKRMPCAEDYLSVVLNQLCVLHEKTPVSDRVTKCCTESLVNRRPC
FSALEVDETYVPKEFQAETFTFHADICTLSEKERQIKKQTALVELVKHKPKATKEQLKAVMD
DFAAFVEKCCKADDKETCFAEEGKKLVAASQAALGLAAALQVQLVQSGAEVKKPGESLKIS
CKGSGYSFTSYWIAWVRQMPGKGLEYMGLIYPGDSDTKYSPSFQGQVTISVDKSVSTAYLQ
WSSLKPSDSAVYFCARHDVGYCTDRTCAKWPEWLGVWGQGTLVTVSSGGGGSSGGGSGG
GGSQSVLTQPPSVSAAPGQKVTISCSGSSSNIGNNYVSWYQQLPGTAPKLLIYD
DRFSGSKSGTSASLAISGFRSEDEADYYCASWDYTLSGWVFGGGTKLTVLG
—28—
Claims (15)
1. Use of an antibody that inhibits heregulin binding to ErbB2/ErbB3 heterodimer for manufacture of a medicament for treating a malignant tumor in a human t, wherein the patient has been treated for malignancy with an anti-estrogen y and has become resistant to such therapy, wherein the ant tumor, by analysis of a tumor biopsy taken from the patent after the t has become resistant, is estrogen receptor positive and expresses HER2, wherein the dy inhibits heregulin-mediated activation of estrogen receptors in malignant tumor cells, which activation comprises phosphorylation of estrogen receptors, wherein the antibody is formulated for stration at a dosage that yields a tration of the antibody in the patient’s bloodstream that is a sufficient concentration to inhibit heregulin-induced estrogen receptor phosphorylation in MCF7 cells in vitro by at least 20%, at least 30%, at least 40%, at least 50%, at least 60%, or at least 70%, and wherein the antibody comprises, VH CDR1, 2 and 3 sequences as shown in SEQ ID NOs: 3-5, respectively, and, VL CDR1, 2 and 3 sequences as shown in SEQ ID NOs: 6-8, respectively.
2. The use of claim 1, wherein the malignant tumor is a tumor of the breast, ovary lung, or skin.
3. The use of claim 1 or claim 2, wherein the expression of HER2 is overexpression as indicated by the tumor biopsy testing HER2++, or HER2+++ or HER2 FISH-positive.
4. The use of any one of claims 1 to 3, wherein the anti-estrogen therapy is either, or both of, a treatment with an estrogen receptor r and a treatment with an aromatase inhibitor.
5. The use of any one of claims 1 to 4, wherein the antibody is formulated for stration by intravenous injection or infusion.
6. The use of any one of claims 1 to 5, wherein the antibody is an rbB2 antibody.
7. The use of any one of claims 1 to 6, wherein the antibody is an anti-ErbB3 antibody.
8. The use of claim 7, wherein the antibody comprises VH and VL regions sing the amino acid sequences set forth in SEQ ID NOs: 1 and 2, respectively.
9. The use of claim 7 or claim 8, wherein the anti-ErbB3 antibody comprises heavy and light chains sing the amino acid sequences set forth in SEQ ID NOs: 42 and 43, respectively.
10. The use of any one of claims 1 to 5, wherein the antibody is an rbB2/anti-ErbB3 bispecific antibody.
11. The use of claim 10, wherein the bispecific antibody is selected from the group consisting of ALM, A5-HSA-ML3.9, -B1D2, B12-HSA-B1D2, A5-HSA-F5B6H2, H3-HSAF5B6H2 , F4-HSA-F5B6H2 and H3-HSA-B1D2.
12. The use of any one of claims 1 to 11, wherein the antibody is formulated for inhibition of one or more of growth, invasiveness, and metastasis of the tumor.
13. The use of any one of claims 1 to 12, wherein the antibody is formulated for coadministration of either, or both of, an estrogen receptor antagonist and an aromatase inhibitor.
14. The use of claim 13, wherein the estrogen receptor antagonist is selected from the group consisting of fene, tamoxifen, afimoxifene roxytamoxifen), arzoxifene, lasofoxone, toremifene and fulvestrant; and the aromatase inhibitor is selected from exemestane, anastrozole, letrozole, aminoglutethimide, testolactone, vorozole, formestane and fadrozole.
15. The use of claim 13 or claim 14, wherein the ase inhibitor is letrozole.
Applications Claiming Priority (5)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
US201161451848P | 2011-03-11 | 2011-03-11 | |
US61/451,848 | 2011-03-11 | ||
US201261604281P | 2012-02-28 | 2012-02-28 | |
US61/604,281 | 2012-02-28 | ||
PCT/US2012/028792 WO2012125573A2 (en) | 2011-03-11 | 2012-03-12 | Use of inhibitors of egfr-family receptors in the treatment of hormone refractory breast cancers |
Publications (2)
Publication Number | Publication Date |
---|---|
NZ614427A NZ614427A (en) | 2015-08-28 |
NZ614427B2 true NZ614427B2 (en) | 2015-12-01 |
Family
ID=
Similar Documents
Publication | Publication Date | Title |
---|---|---|
AU2012229147B2 (en) | Use of inhibitors of EGFR-family receptors in the treatment of hormone refractory breast cancers | |
JP6185102B2 (en) | Use of ERBB3 inhibitors in the treatment of triple negative and basal-like breast cancer | |
Viloria-Petit et al. | Acquired resistance to EGFR inhibitors: mechanisms and prevention strategies | |
Gandullo-Sánchez et al. | HER3 in cancer: from the bench to the bedside | |
US20120156130A1 (en) | Use of her3 binding agents in prostate treatment | |
JP2017534574A (en) | Compositions and methods for treating cancer resistant to tyrosine kinase inhibitors (TKI) | |
AU2010236818B2 (en) | Combination therapy using an anti-EGFR agent(s) and IGF-1R specific inhibitors | |
WO2022223006A1 (en) | Use of anti-pd-1 antibody in combination with first-line chemotherapy for treating advanced non-small cell lung cancer | |
NZ614427B2 (en) | Use of inhibitors of egfr-family receptors in the treatment of hormone refractory breast cancers | |
BR112019020507A2 (en) | erbb-2 targeting agent and a bispecific antibody with antigen binding sites that bind an epitope to an extracellular part of erbb-2 and erbb-3 for treating an individual with an erbb-2, erbb-2 / positive tumor erbb-3 |