CA2459622A1 - Compositions and methods for restoring sensitivity to treatment with her2 antagonists - Google Patents
Compositions and methods for restoring sensitivity to treatment with her2 antagonists Download PDFInfo
- Publication number
- CA2459622A1 CA2459622A1 CA 2459622 CA2459622A CA2459622A1 CA 2459622 A1 CA2459622 A1 CA 2459622A1 CA 2459622 CA2459622 CA 2459622 CA 2459622 A CA2459622 A CA 2459622A CA 2459622 A1 CA2459622 A1 CA 2459622A1
- Authority
- CA
- Canada
- Prior art keywords
- pcdgf
- antagonist
- her2
- erbb2
- cells
- Prior art date
- Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
- Granted
Links
- 239000005557 antagonist Substances 0.000 title claims abstract description 113
- 238000000034 method Methods 0.000 title claims abstract description 20
- 230000035945 sensitivity Effects 0.000 title claims abstract description 15
- 239000000203 mixture Substances 0.000 title claims abstract description 13
- 238000011282 treatment Methods 0.000 title description 22
- 101100314454 Caenorhabditis elegans tra-1 gene Proteins 0.000 title 1
- 102100037632 Progranulin Human genes 0.000 claims abstract description 105
- 101710114165 Progranulin Proteins 0.000 claims abstract description 105
- 102100030086 Receptor tyrosine-protein kinase erbB-2 Human genes 0.000 claims abstract description 85
- 101001012157 Homo sapiens Receptor tyrosine-protein kinase erbB-2 Proteins 0.000 claims abstract description 83
- 230000009036 growth inhibition Effects 0.000 claims abstract description 10
- 229940022353 herceptin Drugs 0.000 claims description 19
- 206010028980 Neoplasm Diseases 0.000 claims description 16
- 108020004459 Small interfering RNA Proteins 0.000 claims description 5
- 239000008194 pharmaceutical composition Substances 0.000 claims description 5
- 230000002401 inhibitory effect Effects 0.000 claims description 4
- 150000003384 small molecules Chemical class 0.000 claims description 4
- 230000004565 tumor cell growth Effects 0.000 claims description 3
- 230000004614 tumor growth Effects 0.000 claims description 3
- 108091034117 Oligonucleotide Proteins 0.000 claims 1
- 239000000074 antisense oligonucleotide Substances 0.000 claims 1
- 238000012230 antisense oligonucleotides Methods 0.000 claims 1
- 238000012544 monitoring process Methods 0.000 claims 1
- 210000004027 cell Anatomy 0.000 abstract description 40
- 210000004881 tumor cell Anatomy 0.000 abstract description 8
- 230000001225 therapeutic effect Effects 0.000 abstract description 3
- 238000011269 treatment regimen Methods 0.000 abstract description 2
- 206010006187 Breast cancer Diseases 0.000 description 14
- 208000026310 Breast neoplasm Diseases 0.000 description 14
- 238000002560 therapeutic procedure Methods 0.000 description 13
- 230000000118 anti-neoplastic effect Effects 0.000 description 10
- 230000010261 cell growth Effects 0.000 description 8
- 230000002018 overexpression Effects 0.000 description 7
- 102000005962 receptors Human genes 0.000 description 6
- 108020003175 receptors Proteins 0.000 description 6
- 206010055113 Breast cancer metastatic Diseases 0.000 description 5
- -1 antisense Proteins 0.000 description 5
- 201000011510 cancer Diseases 0.000 description 5
- 230000000694 effects Effects 0.000 description 5
- 108020004707 nucleic acids Proteins 0.000 description 5
- 102000039446 nucleic acids Human genes 0.000 description 5
- 150000007523 nucleic acids Chemical class 0.000 description 5
- 230000026731 phosphorylation Effects 0.000 description 5
- 238000006366 phosphorylation reaction Methods 0.000 description 5
- 230000003442 weekly effect Effects 0.000 description 5
- 230000000692 anti-sense effect Effects 0.000 description 4
- 230000004071 biological effect Effects 0.000 description 4
- 239000012472 biological sample Substances 0.000 description 4
- 239000012634 fragment Substances 0.000 description 4
- 230000014509 gene expression Effects 0.000 description 4
- 230000003472 neutralizing effect Effects 0.000 description 4
- 210000001519 tissue Anatomy 0.000 description 4
- 208000005623 Carcinogenesis Diseases 0.000 description 3
- 241001465754 Metazoa Species 0.000 description 3
- 229930012538 Paclitaxel Natural products 0.000 description 3
- 239000000427 antigen Substances 0.000 description 3
- 102000036639 antigens Human genes 0.000 description 3
- 108091007433 antigens Proteins 0.000 description 3
- 238000001574 biopsy Methods 0.000 description 3
- 230000036952 cancer formation Effects 0.000 description 3
- 231100000504 carcinogenesis Toxicity 0.000 description 3
- 238000011260 co-administration Methods 0.000 description 3
- 238000001990 intravenous administration Methods 0.000 description 3
- 229940043355 kinase inhibitor Drugs 0.000 description 3
- 229960001592 paclitaxel Drugs 0.000 description 3
- 239000003757 phosphotransferase inhibitor Substances 0.000 description 3
- 108090000765 processed proteins & peptides Proteins 0.000 description 3
- 230000035755 proliferation Effects 0.000 description 3
- 239000000523 sample Substances 0.000 description 3
- 230000004083 survival effect Effects 0.000 description 3
- RCINICONZNJXQF-MZXODVADSA-N taxol Chemical compound O([C@@H]1[C@@]2(C[C@@H](C(C)=C(C2(C)C)[C@H](C([C@]2(C)[C@@H](O)C[C@H]3OC[C@]3([C@H]21)OC(C)=O)=O)OC(=O)C)OC(=O)[C@H](O)[C@@H](NC(=O)C=1C=CC=CC=1)C=1C=CC=CC=1)O)C(=O)C1=CC=CC=C1 RCINICONZNJXQF-MZXODVADSA-N 0.000 description 3
- 239000003053 toxin Substances 0.000 description 3
- 231100000765 toxin Toxicity 0.000 description 3
- 206010003571 Astrocytoma Diseases 0.000 description 2
- AOJJSUZBOXZQNB-TZSSRYMLSA-N Doxorubicin Chemical compound O([C@H]1C[C@@](O)(CC=2C(O)=C3C(=O)C=4C=CC=C(C=4C(=O)C3=C(O)C=21)OC)C(=O)CO)[C@H]1C[C@H](N)[C@H](O)[C@H](C)O1 AOJJSUZBOXZQNB-TZSSRYMLSA-N 0.000 description 2
- WQZGKKKJIJFFOK-GASJEMHNSA-N Glucose Chemical compound OC[C@H]1OC(O)[C@H](O)[C@@H](O)[C@@H]1O WQZGKKKJIJFFOK-GASJEMHNSA-N 0.000 description 2
- 102000008100 Human Serum Albumin Human genes 0.000 description 2
- 108091006905 Human Serum Albumin Proteins 0.000 description 2
- 206010029260 Neuroblastoma Diseases 0.000 description 2
- 101710100968 Receptor tyrosine-protein kinase erbB-2 Proteins 0.000 description 2
- 206010039491 Sarcoma Diseases 0.000 description 2
- FAPWRFPIFSIZLT-UHFFFAOYSA-M Sodium chloride Chemical compound [Na+].[Cl-] FAPWRFPIFSIZLT-UHFFFAOYSA-M 0.000 description 2
- NKANXQFJJICGDU-QPLCGJKRSA-N Tamoxifen Chemical compound C=1C=CC=CC=1C(/CC)=C(C=1C=CC(OCCN(C)C)=CC=1)/C1=CC=CC=C1 NKANXQFJJICGDU-QPLCGJKRSA-N 0.000 description 2
- IQFYYKKMVGJFEH-XLPZGREQSA-N Thymidine Chemical compound O=C1NC(=O)C(C)=CN1[C@@H]1O[C@H](CO)[C@@H](O)C1 IQFYYKKMVGJFEH-XLPZGREQSA-N 0.000 description 2
- RJURFGZVJUQBHK-UHFFFAOYSA-N actinomycin D Natural products CC1OC(=O)C(C(C)C)N(C)C(=O)CN(C)C(=O)C2CCCN2C(=O)C(C(C)C)NC(=O)C1NC(=O)C1=C(N)C(=O)C(C)=C2OC(C(C)=CC=C3C(=O)NC4C(=O)NC(C(N5CCCC5C(=O)N(C)CC(=O)N(C)C(C(C)C)C(=O)OC4C)=O)C(C)C)=C3N=C21 RJURFGZVJUQBHK-UHFFFAOYSA-N 0.000 description 2
- 230000010056 antibody-dependent cellular cytotoxicity Effects 0.000 description 2
- 230000003305 autocrine Effects 0.000 description 2
- 230000008901 benefit Effects 0.000 description 2
- 210000004369 blood Anatomy 0.000 description 2
- 239000008280 blood Substances 0.000 description 2
- 210000000988 bone and bone Anatomy 0.000 description 2
- 210000001185 bone marrow Anatomy 0.000 description 2
- 210000004556 brain Anatomy 0.000 description 2
- 210000000481 breast Anatomy 0.000 description 2
- 210000003679 cervix uteri Anatomy 0.000 description 2
- 239000003795 chemical substances by application Substances 0.000 description 2
- 238000002512 chemotherapy Methods 0.000 description 2
- 210000001072 colon Anatomy 0.000 description 2
- 238000002648 combination therapy Methods 0.000 description 2
- 239000002299 complementary DNA Substances 0.000 description 2
- 150000001875 compounds Chemical class 0.000 description 2
- 230000003247 decreasing effect Effects 0.000 description 2
- 239000003937 drug carrier Substances 0.000 description 2
- 208000005017 glioblastoma Diseases 0.000 description 2
- 230000012010 growth Effects 0.000 description 2
- 239000003102 growth factor Substances 0.000 description 2
- 210000003128 head Anatomy 0.000 description 2
- 230000005764 inhibitory process Effects 0.000 description 2
- NOESYZHRGYRDHS-UHFFFAOYSA-N insulin Chemical compound N1C(=O)C(NC(=O)C(CCC(N)=O)NC(=O)C(CCC(O)=O)NC(=O)C(C(C)C)NC(=O)C(NC(=O)CN)C(C)CC)CSSCC(C(NC(CO)C(=O)NC(CC(C)C)C(=O)NC(CC=2C=CC(O)=CC=2)C(=O)NC(CCC(N)=O)C(=O)NC(CC(C)C)C(=O)NC(CCC(O)=O)C(=O)NC(CC(N)=O)C(=O)NC(CC=2C=CC(O)=CC=2)C(=O)NC(CSSCC(NC(=O)C(C(C)C)NC(=O)C(CC(C)C)NC(=O)C(CC=2C=CC(O)=CC=2)NC(=O)C(CC(C)C)NC(=O)C(C)NC(=O)C(CCC(O)=O)NC(=O)C(C(C)C)NC(=O)C(CC(C)C)NC(=O)C(CC=2NC=NC=2)NC(=O)C(CO)NC(=O)CNC2=O)C(=O)NCC(=O)NC(CCC(O)=O)C(=O)NC(CCCNC(N)=N)C(=O)NCC(=O)NC(CC=3C=CC=CC=3)C(=O)NC(CC=3C=CC=CC=3)C(=O)NC(CC=3C=CC(O)=CC=3)C(=O)NC(C(C)O)C(=O)N3C(CCC3)C(=O)NC(CCCCN)C(=O)NC(C)C(O)=O)C(=O)NC(CC(N)=O)C(O)=O)=O)NC(=O)C(C(C)CC)NC(=O)C(CO)NC(=O)C(C(C)O)NC(=O)C1CSSCC2NC(=O)C(CC(C)C)NC(=O)C(NC(=O)C(CCC(N)=O)NC(=O)C(CC(N)=O)NC(=O)C(NC(=O)C(N)CC=1C=CC=CC=1)C(C)C)CC1=CN=CN1 NOESYZHRGYRDHS-UHFFFAOYSA-N 0.000 description 2
- 230000002452 interceptive effect Effects 0.000 description 2
- 210000003734 kidney Anatomy 0.000 description 2
- 210000004185 liver Anatomy 0.000 description 2
- 210000004072 lung Anatomy 0.000 description 2
- 206010061289 metastatic neoplasm Diseases 0.000 description 2
- 210000003739 neck Anatomy 0.000 description 2
- 230000001613 neoplastic effect Effects 0.000 description 2
- 230000001537 neural effect Effects 0.000 description 2
- 210000001672 ovary Anatomy 0.000 description 2
- 210000000496 pancreas Anatomy 0.000 description 2
- 239000012188 paraffin wax Substances 0.000 description 2
- 238000007911 parenteral administration Methods 0.000 description 2
- 210000003800 pharynx Anatomy 0.000 description 2
- 210000002307 prostate Anatomy 0.000 description 2
- 102000004169 proteins and genes Human genes 0.000 description 2
- 108090000623 proteins and genes Proteins 0.000 description 2
- 210000003491 skin Anatomy 0.000 description 2
- 239000011780 sodium chloride Substances 0.000 description 2
- 239000000243 solution Substances 0.000 description 2
- 210000002784 stomach Anatomy 0.000 description 2
- 238000012360 testing method Methods 0.000 description 2
- 210000001550 testis Anatomy 0.000 description 2
- 210000001685 thyroid gland Anatomy 0.000 description 2
- 230000000381 tumorigenic effect Effects 0.000 description 2
- 210000004291 uterus Anatomy 0.000 description 2
- XLYOFNOQVPJJNP-UHFFFAOYSA-N water Substances O XLYOFNOQVPJJNP-UHFFFAOYSA-N 0.000 description 2
- STQGQHZAVUOBTE-UHFFFAOYSA-N 7-Cyan-hept-2t-en-4,6-diinsaeure Natural products C1=2C(O)=C3C(=O)C=4C(OC)=CC=CC=4C(=O)C3=C(O)C=2CC(O)(C(C)=O)CC1OC1CC(N)C(O)C(C)O1 STQGQHZAVUOBTE-UHFFFAOYSA-N 0.000 description 1
- 229920001817 Agar Polymers 0.000 description 1
- DWRXFEITVBNRMK-UHFFFAOYSA-N Beta-D-1-Arabinofuranosylthymine Natural products O=C1NC(=O)C(C)=CN1C1C(O)C(O)C(CO)O1 DWRXFEITVBNRMK-UHFFFAOYSA-N 0.000 description 1
- 108010006654 Bleomycin Proteins 0.000 description 1
- COVZYZSDYWQREU-UHFFFAOYSA-N Busulfan Chemical compound CS(=O)(=O)OCCCCOS(C)(=O)=O COVZYZSDYWQREU-UHFFFAOYSA-N 0.000 description 1
- KLWPJMFMVPTNCC-UHFFFAOYSA-N Camptothecin Natural products CCC1(O)C(=O)OCC2=C1C=C3C4Nc5ccccc5C=C4CN3C2=O KLWPJMFMVPTNCC-UHFFFAOYSA-N 0.000 description 1
- 102000000844 Cell Surface Receptors Human genes 0.000 description 1
- 108010001857 Cell Surface Receptors Proteins 0.000 description 1
- 108010047041 Complementarity Determining Regions Proteins 0.000 description 1
- CMSMOCZEIVJLDB-UHFFFAOYSA-N Cyclophosphamide Chemical compound ClCCN(CCCl)P1(=O)NCCCO1 CMSMOCZEIVJLDB-UHFFFAOYSA-N 0.000 description 1
- 108010092160 Dactinomycin Proteins 0.000 description 1
- 101150029707 ERBB2 gene Proteins 0.000 description 1
- 206010014733 Endometrial cancer Diseases 0.000 description 1
- 206010014759 Endometrial neoplasm Diseases 0.000 description 1
- 102000004190 Enzymes Human genes 0.000 description 1
- 108090000790 Enzymes Proteins 0.000 description 1
- GHASVSINZRGABV-UHFFFAOYSA-N Fluorouracil Chemical compound FC1=CNC(=O)NC1=O GHASVSINZRGABV-UHFFFAOYSA-N 0.000 description 1
- VWUXBMIQPBEWFH-WCCTWKNTSA-N Fulvestrant Chemical compound OC1=CC=C2[C@H]3CC[C@](C)([C@H](CC4)O)[C@@H]4[C@@H]3[C@H](CCCCCCCCCS(=O)CCCC(F)(F)C(F)(F)F)CC2=C1 VWUXBMIQPBEWFH-WCCTWKNTSA-N 0.000 description 1
- 241000287828 Gallus gallus Species 0.000 description 1
- 241000282412 Homo Species 0.000 description 1
- 108010054477 Immunoglobulin Fab Fragments Proteins 0.000 description 1
- 102000001706 Immunoglobulin Fab Fragments Human genes 0.000 description 1
- 102000018071 Immunoglobulin Fc Fragments Human genes 0.000 description 1
- 108010091135 Immunoglobulin Fc Fragments Proteins 0.000 description 1
- 102000004877 Insulin Human genes 0.000 description 1
- 108090001061 Insulin Proteins 0.000 description 1
- 208000008839 Kidney Neoplasms Diseases 0.000 description 1
- 229930192392 Mitomycin Natural products 0.000 description 1
- 241001529936 Murinae Species 0.000 description 1
- NWIBSHFKIJFRCO-WUDYKRTCSA-N Mytomycin Chemical compound C1N2C(C(C(C)=C(N)C3=O)=O)=C3[C@@H](COC(N)=O)[C@@]2(OC)[C@@H]2[C@H]1N2 NWIBSHFKIJFRCO-WUDYKRTCSA-N 0.000 description 1
- 108700020796 Oncogene Proteins 0.000 description 1
- 108090000526 Papain Proteins 0.000 description 1
- 102000057297 Pepsin A Human genes 0.000 description 1
- 108090000284 Pepsin A Proteins 0.000 description 1
- 102000019204 Progranulins Human genes 0.000 description 1
- 108010012809 Progranulins Proteins 0.000 description 1
- 206010060862 Prostate cancer Diseases 0.000 description 1
- 208000000236 Prostatic Neoplasms Diseases 0.000 description 1
- 239000004365 Protease Substances 0.000 description 1
- 108020004511 Recombinant DNA Proteins 0.000 description 1
- 206010038389 Renal cancer Diseases 0.000 description 1
- 208000005718 Stomach Neoplasms Diseases 0.000 description 1
- RJURFGZVJUQBHK-IIXSONLDSA-N actinomycin D Chemical compound C[C@H]1OC(=O)[C@H](C(C)C)N(C)C(=O)CN(C)C(=O)[C@@H]2CCCN2C(=O)[C@@H](C(C)C)NC(=O)[C@H]1NC(=O)C1=C(N)C(=O)C(C)=C2OC(C(C)=CC=C3C(=O)N[C@@H]4C(=O)N[C@@H](C(N5CCC[C@H]5C(=O)N(C)CC(=O)N(C)[C@@H](C(C)C)C(=O)O[C@@H]4C)=O)C(C)C)=C3N=C21 RJURFGZVJUQBHK-IIXSONLDSA-N 0.000 description 1
- 230000009471 action Effects 0.000 description 1
- 239000008272 agar Substances 0.000 description 1
- 230000001093 anti-cancer Effects 0.000 description 1
- 229940046836 anti-estrogen Drugs 0.000 description 1
- 230000001833 anti-estrogenic effect Effects 0.000 description 1
- 239000002246 antineoplastic agent Substances 0.000 description 1
- 230000006907 apoptotic process Effects 0.000 description 1
- 239000007864 aqueous solution Substances 0.000 description 1
- 238000003556 assay Methods 0.000 description 1
- 239000012752 auxiliary agent Substances 0.000 description 1
- 230000009286 beneficial effect Effects 0.000 description 1
- IQFYYKKMVGJFEH-UHFFFAOYSA-N beta-L-thymidine Natural products O=C1NC(=O)C(C)=CN1C1OC(CO)C(O)C1 IQFYYKKMVGJFEH-UHFFFAOYSA-N 0.000 description 1
- 239000011230 binding agent Substances 0.000 description 1
- 229960001561 bleomycin Drugs 0.000 description 1
- OYVAGSVQBOHSSS-UAPAGMARSA-O bleomycin A2 Chemical compound N([C@H](C(=O)N[C@H](C)[C@@H](O)[C@H](C)C(=O)N[C@@H]([C@H](O)C)C(=O)NCCC=1SC=C(N=1)C=1SC=C(N=1)C(=O)NCCC[S+](C)C)[C@@H](O[C@H]1[C@H]([C@@H](O)[C@H](O)[C@H](CO)O1)O[C@@H]1[C@H]([C@@H](OC(N)=O)[C@H](O)[C@@H](CO)O1)O)C=1N=CNC=1)C(=O)C1=NC([C@H](CC(N)=O)NC[C@H](N)C(N)=O)=NC(N)=C1C OYVAGSVQBOHSSS-UAPAGMARSA-O 0.000 description 1
- 230000000903 blocking effect Effects 0.000 description 1
- 229960002092 busulfan Drugs 0.000 description 1
- 229940127093 camptothecin Drugs 0.000 description 1
- VSJKWCGYPAHWDS-FQEVSTJZSA-N camptothecin Chemical compound C1=CC=C2C=C(CN3C4=CC5=C(C3=O)COC(=O)[C@]5(O)CC)C4=NC2=C1 VSJKWCGYPAHWDS-FQEVSTJZSA-N 0.000 description 1
- 239000002775 capsule Substances 0.000 description 1
- 229960004562 carboplatin Drugs 0.000 description 1
- 190000008236 carboplatin Chemical compound 0.000 description 1
- 238000000423 cell based assay Methods 0.000 description 1
- 230000004663 cell proliferation Effects 0.000 description 1
- 230000001413 cellular effect Effects 0.000 description 1
- 210000001175 cerebrospinal fluid Anatomy 0.000 description 1
- 238000009104 chemotherapy regimen Methods 0.000 description 1
- 229960004630 chlorambucil Drugs 0.000 description 1
- JCKYGMPEJWAADB-UHFFFAOYSA-N chlorambucil Chemical compound OC(=O)CCCC1=CC=C(N(CCCl)CCCl)C=C1 JCKYGMPEJWAADB-UHFFFAOYSA-N 0.000 description 1
- DQLATGHUWYMOKM-UHFFFAOYSA-L cisplatin Chemical compound N[Pt](N)(Cl)Cl DQLATGHUWYMOKM-UHFFFAOYSA-L 0.000 description 1
- 229960004316 cisplatin Drugs 0.000 description 1
- 239000003636 conditioned culture medium Substances 0.000 description 1
- 229960004397 cyclophosphamide Drugs 0.000 description 1
- 229940127089 cytotoxic agent Drugs 0.000 description 1
- 229960000640 dactinomycin Drugs 0.000 description 1
- STQGQHZAVUOBTE-VGBVRHCVSA-N daunorubicin Chemical compound O([C@H]1C[C@@](O)(CC=2C(O)=C3C(=O)C=4C=CC=C(C=4C(=O)C3=C(O)C=21)OC)C(C)=O)[C@H]1C[C@H](N)[C@H](O)[C@H](C)O1 STQGQHZAVUOBTE-VGBVRHCVSA-N 0.000 description 1
- 229960000975 daunorubicin Drugs 0.000 description 1
- 230000001419 dependent effect Effects 0.000 description 1
- 238000011161 development Methods 0.000 description 1
- 238000007435 diagnostic evaluation Methods 0.000 description 1
- 201000010099 disease Diseases 0.000 description 1
- 208000037265 diseases, disorders, signs and symptoms Diseases 0.000 description 1
- VSJKWCGYPAHWDS-UHFFFAOYSA-N dl-camptothecin Natural products C1=CC=C2C=C(CN3C4=CC5=C(C3=O)COC(=O)C5(O)CC)C4=NC2=C1 VSJKWCGYPAHWDS-UHFFFAOYSA-N 0.000 description 1
- 239000002552 dosage form Substances 0.000 description 1
- 231100000673 dose–response relationship Toxicity 0.000 description 1
- 229960004679 doxorubicin Drugs 0.000 description 1
- 239000003814 drug Substances 0.000 description 1
- 230000009977 dual effect Effects 0.000 description 1
- 235000013601 eggs Nutrition 0.000 description 1
- 239000000839 emulsion Substances 0.000 description 1
- 229940088598 enzyme Drugs 0.000 description 1
- 229940011871 estrogen Drugs 0.000 description 1
- 239000000262 estrogen Substances 0.000 description 1
- 239000000328 estrogen antagonist Substances 0.000 description 1
- 102000015694 estrogen receptors Human genes 0.000 description 1
- 108010038795 estrogen receptors Proteins 0.000 description 1
- VJJPUSNTGOMMGY-MRVIYFEKSA-N etoposide Chemical compound COC1=C(O)C(OC)=CC([C@@H]2C3=CC=4OCOC=4C=C3[C@@H](O[C@H]3[C@@H]([C@@H](O)[C@@H]4O[C@H](C)OC[C@H]4O3)O)[C@@H]3[C@@H]2C(OC3)=O)=C1 VJJPUSNTGOMMGY-MRVIYFEKSA-N 0.000 description 1
- 229960002949 fluorouracil Drugs 0.000 description 1
- 229960002258 fulvestrant Drugs 0.000 description 1
- 206010017758 gastric cancer Diseases 0.000 description 1
- 229960001101 ifosfamide Drugs 0.000 description 1
- HOMGKSMUEGBAAB-UHFFFAOYSA-N ifosfamide Chemical compound ClCCNP1(=O)OCCCN1CCCl HOMGKSMUEGBAAB-UHFFFAOYSA-N 0.000 description 1
- 230000001900 immune effect Effects 0.000 description 1
- 230000028993 immune response Effects 0.000 description 1
- 238000003364 immunohistochemistry Methods 0.000 description 1
- 238000000338 in vitro Methods 0.000 description 1
- 238000001727 in vivo Methods 0.000 description 1
- 238000011065 in-situ storage Methods 0.000 description 1
- 238000010348 incorporation Methods 0.000 description 1
- 239000003112 inhibitor Substances 0.000 description 1
- 238000002347 injection Methods 0.000 description 1
- 239000007924 injection Substances 0.000 description 1
- 229940125396 insulin Drugs 0.000 description 1
- 238000007918 intramuscular administration Methods 0.000 description 1
- 238000007912 intraperitoneal administration Methods 0.000 description 1
- 206010073095 invasive ductal breast carcinoma Diseases 0.000 description 1
- 201000010985 invasive ductal carcinoma Diseases 0.000 description 1
- 201000010982 kidney cancer Diseases 0.000 description 1
- 230000007774 longterm Effects 0.000 description 1
- 230000036210 malignancy Effects 0.000 description 1
- 238000004519 manufacturing process Methods 0.000 description 1
- 229960004961 mechlorethamine Drugs 0.000 description 1
- HAWPXGHAZFHHAD-UHFFFAOYSA-N mechlorethamine Chemical compound ClCCN(C)CCCl HAWPXGHAZFHHAD-UHFFFAOYSA-N 0.000 description 1
- 229960001924 melphalan Drugs 0.000 description 1
- SGDBTWWWUNNDEQ-LBPRGKRZSA-N melphalan Chemical compound OC(=O)[C@@H](N)CC1=CC=C(N(CCCl)CCCl)C=C1 SGDBTWWWUNNDEQ-LBPRGKRZSA-N 0.000 description 1
- 229960004857 mitomycin Drugs 0.000 description 1
- 229940086322 navelbine Drugs 0.000 description 1
- 239000012457 nonaqueous media Substances 0.000 description 1
- 229940055729 papain Drugs 0.000 description 1
- 235000019834 papain Nutrition 0.000 description 1
- 230000001575 pathological effect Effects 0.000 description 1
- 229940111202 pepsin Drugs 0.000 description 1
- 230000010412 perfusion Effects 0.000 description 1
- 239000000546 pharmaceutical excipient Substances 0.000 description 1
- 238000002360 preparation method Methods 0.000 description 1
- 229960000624 procarbazine Drugs 0.000 description 1
- CPTBDICYNRMXFX-UHFFFAOYSA-N procarbazine Chemical compound CNNCC1=CC=C(C(=O)NC(C)C)C=C1 CPTBDICYNRMXFX-UHFFFAOYSA-N 0.000 description 1
- 102000004196 processed proteins & peptides Human genes 0.000 description 1
- 230000002035 prolonged effect Effects 0.000 description 1
- 230000006337 proteolytic cleavage Effects 0.000 description 1
- 229960004622 raloxifene Drugs 0.000 description 1
- 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 1
- 230000001105 regulatory effect Effects 0.000 description 1
- 230000004044 response Effects 0.000 description 1
- 230000004043 responsiveness Effects 0.000 description 1
- 210000002966 serum Anatomy 0.000 description 1
- 238000003153 stable transfection Methods 0.000 description 1
- 238000010186 staining Methods 0.000 description 1
- 230000000638 stimulation Effects 0.000 description 1
- 201000011549 stomach cancer Diseases 0.000 description 1
- 238000007920 subcutaneous administration Methods 0.000 description 1
- 239000000725 suspension Substances 0.000 description 1
- 229960001603 tamoxifen Drugs 0.000 description 1
- 229940104230 thymidine Drugs 0.000 description 1
- 229960000575 trastuzumab Drugs 0.000 description 1
- JXLYSJRDGCGARV-CFWMRBGOSA-N vinblastine Chemical compound C([C@H](C[C@]1(C(=O)OC)C=2C(=CC3=C([C@]45[C@H]([C@@]([C@H](OC(C)=O)[C@]6(CC)C=CCN([C@H]56)CC4)(O)C(=O)OC)N3C)C=2)OC)C[C@@](C2)(O)CC)N2CCC2=C1NC1=CC=CC=C21 JXLYSJRDGCGARV-CFWMRBGOSA-N 0.000 description 1
- 229960003048 vinblastine Drugs 0.000 description 1
- 229960004528 vincristine Drugs 0.000 description 1
- OGWKCGZFUXNPDA-XQKSVPLYSA-N vincristine Chemical compound C([N@]1C[C@@H](C[C@]2(C(=O)OC)C=3C(=CC4=C([C@]56[C@H]([C@@]([C@H](OC(C)=O)[C@]7(CC)C=CCN([C@H]67)CC5)(O)C(=O)OC)N4C=O)C=3)OC)C[C@@](C1)(O)CC)CC1=C2NC2=CC=CC=C12 OGWKCGZFUXNPDA-XQKSVPLYSA-N 0.000 description 1
- OGWKCGZFUXNPDA-UHFFFAOYSA-N vincristine Natural products C1C(CC)(O)CC(CC2(C(=O)OC)C=3C(=CC4=C(C56C(C(C(OC(C)=O)C7(CC)C=CCN(C67)CC5)(O)C(=O)OC)N4C=O)C=3)OC)CN1CCC1=C2NC2=CC=CC=C12 OGWKCGZFUXNPDA-UHFFFAOYSA-N 0.000 description 1
- CILBMBUYJCWATM-PYGJLNRPSA-N vinorelbine ditartrate Chemical compound OC(=O)[C@H](O)[C@@H](O)C(O)=O.OC(=O)[C@H](O)[C@@H](O)C(O)=O.C1N(CC=2C3=CC=CC=C3NC=22)CC(CC)=C[C@H]1C[C@]2(C(=O)OC)C1=CC([C@]23[C@H]([C@@]([C@H](OC(C)=O)[C@]4(CC)C=CCN([C@H]34)CC2)(O)C(=O)OC)N2C)=C2C=C1OC CILBMBUYJCWATM-PYGJLNRPSA-N 0.000 description 1
Landscapes
- Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
- Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
- Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
Abstract
Methods and compositions for restoring growth inhibition sensitivity to a tumor cell resistant to growth inhibition by HER2 antagonists. The methods involve administering a PCDGF antagonist to the cell in an amount effective to restore growth inhibition sensitivity to HER2 antagonists. The invention also provides treatment regimens, and therapeutic compositions comprising an HER2 antagonist and a PCDGF antagonist.
Description
COMPOSITIONS AND METHODS FOR RESTORING SENSITIVITY TO
BACKGROUND OF THE INVENTION
~ooo y Approximately 25-30% of breast cancer patients overexpress the proto-oncoprotein and cell surface receptor c-erbB2 (human epidermal growth factor receptor 2 protein), also known as HER2/neu. Overexpression of the c-erbB2 oncogene has been linked to poor outcome and decreased survival for patients. The HER-2/neu proto-oncogene is overexpressed in 20-30% of metastatic breast cancers, and is associated with decreased survival and increased recurrence of breast cancer. HER-2/neu is also overexpressed in other cancers types including endometrial cancer, kidney cancer, gastric cancer, and prostate cancer. Presently, the most common form of treatment for these patients is the use of the humanized monoclonal antibody Trastuzumab, also known as Herceptin~.
[ooo~~ Herceptin~ is a recombinant DNA-derived humanized monoclonal antibody that selectively binds with high affinity (Kd = 5 nM) to the extracellular domain of c-erbB2 in a cell-based assay. See Science 1985;230:1132-9 and Cancer Res 1993;53:4960-70 hereby incorporated by reference in its entirety.
Herceptin~ is an IgG1 kappa antibody that binds to HER2 and contains human framework regions with the complementarity-determining regions of a murine antibody (4D5). Id. However, only 25% of the patients treated with Herceptin~
or any other antibody to c-erbB2/HER2 are responsive to this therapy. Several models have been postulated to explain resistance to treatment with c-erbB2/HER2 antibodies.
BRIEF SUMMARY OF THE INVENTION
The present invention is based in part on the discovery that an autocrine growth factor, PC-Cell Derived Growth Factor ("PCDGF"), confers resistance to the antineoplastic effects of c-erbB2/HER2 ("HER2") antagonists.
Preferred embodiments of this invention are directed to therapeutic compositions and methods for restoring growth inhibition sensitivity to tumor cells resistant to the anHneoplastic effects of HER2 antagonists by administering a PCDGF
antagonist in an amount effective to restore growth inhibition sensitivity to HERZ
antagonists. Another embodiment of the invention provides therapeutic compositions and methods for inhibiting tumor cell growth comprising administering a PCDGF antagonist and a HER2 antagonist in an amount effective to inhibit tumor cell growth.
The invention also provides preferred compositions comprising a HER2 antagonist and a PCDGF antagonist. In another embodiment, the invention provides a pharmaceutical composition comprising a HER2 antagonist, a PCDGF antagonist, and a pharmaceutically-acceptable carrier (e.g., water, saline, Ringer's solution, dextrose solution, and human serum albumin).
CoooS~ Further embodiments of the invention provide methods of determining whether a patient is resistant to the antineoplastic effects of antagonists, comprising obtaining a biological sample containing cells from a patient; detecting PCDGF in the biological sample; and determining the amount of PCDGF in said sample wherein the amount of PCDGF is indicative of resistance to the antineoplastic effects of HER2 antagonists.
BRIEF DESCRIPTION OF THE DRAWINGS
~ooos~ FIG. 1 summarizes pathological studies in paraffin embedded human breast cancer biopsies. PCDGF and erbB2 staining were monitored by immunohistochemisty. Thus, overexpression of PCDGF in cells overexpressing erbB2 renders the cells HER2 antagonist resistant. It is known that 25% of patients with tumors that overexpress erbB2 will be responsive to HER2 antagonist therapy.
~ooo ~ ~ FIG. 2 shows erbB2 levels in MCF7 cells (breast cancer cells) transfected with erbB2.
Cooos~ FIG. 3 shows PCDGF expression in MCF7 cells transfected with erbB2.
FIG. 4 shows PCDGF protein levels in conditioned media of cells transfected with both PCDGF and erbB2 (D.c2) and erbB2 alone (erbB2.c1).
FIG. 5 shows that PCDGF stimulates erbB2 phosphorylation in erbB2 overexpressing cells.
Coo 1 y FIG. 6 shows that PCDGF stimulates erbB2 phosphorylation in other breast cancer cells that are known to overexpress erbB2 such as BT474 and SKBR3 (see FIG. 12).
Cooiz~ FIG. 7 shows the dose response of PCDGF stimulation of erbB2 in BT474 cells.
Coo~ga FIG. 8 shows the long term growth of erbB2 cells in response to Herceptin~. Herceptin~ inhibits proliferation of erbB2 overexpressing cells as it is a monoclonal antibody that neutralizes erbB2 and inhibits proliferation in erbB2 overexpressing breast cancer cells.
BACKGROUND OF THE INVENTION
~ooo y Approximately 25-30% of breast cancer patients overexpress the proto-oncoprotein and cell surface receptor c-erbB2 (human epidermal growth factor receptor 2 protein), also known as HER2/neu. Overexpression of the c-erbB2 oncogene has been linked to poor outcome and decreased survival for patients. The HER-2/neu proto-oncogene is overexpressed in 20-30% of metastatic breast cancers, and is associated with decreased survival and increased recurrence of breast cancer. HER-2/neu is also overexpressed in other cancers types including endometrial cancer, kidney cancer, gastric cancer, and prostate cancer. Presently, the most common form of treatment for these patients is the use of the humanized monoclonal antibody Trastuzumab, also known as Herceptin~.
[ooo~~ Herceptin~ is a recombinant DNA-derived humanized monoclonal antibody that selectively binds with high affinity (Kd = 5 nM) to the extracellular domain of c-erbB2 in a cell-based assay. See Science 1985;230:1132-9 and Cancer Res 1993;53:4960-70 hereby incorporated by reference in its entirety.
Herceptin~ is an IgG1 kappa antibody that binds to HER2 and contains human framework regions with the complementarity-determining regions of a murine antibody (4D5). Id. However, only 25% of the patients treated with Herceptin~
or any other antibody to c-erbB2/HER2 are responsive to this therapy. Several models have been postulated to explain resistance to treatment with c-erbB2/HER2 antibodies.
BRIEF SUMMARY OF THE INVENTION
The present invention is based in part on the discovery that an autocrine growth factor, PC-Cell Derived Growth Factor ("PCDGF"), confers resistance to the antineoplastic effects of c-erbB2/HER2 ("HER2") antagonists.
Preferred embodiments of this invention are directed to therapeutic compositions and methods for restoring growth inhibition sensitivity to tumor cells resistant to the anHneoplastic effects of HER2 antagonists by administering a PCDGF
antagonist in an amount effective to restore growth inhibition sensitivity to HERZ
antagonists. Another embodiment of the invention provides therapeutic compositions and methods for inhibiting tumor cell growth comprising administering a PCDGF antagonist and a HER2 antagonist in an amount effective to inhibit tumor cell growth.
The invention also provides preferred compositions comprising a HER2 antagonist and a PCDGF antagonist. In another embodiment, the invention provides a pharmaceutical composition comprising a HER2 antagonist, a PCDGF antagonist, and a pharmaceutically-acceptable carrier (e.g., water, saline, Ringer's solution, dextrose solution, and human serum albumin).
CoooS~ Further embodiments of the invention provide methods of determining whether a patient is resistant to the antineoplastic effects of antagonists, comprising obtaining a biological sample containing cells from a patient; detecting PCDGF in the biological sample; and determining the amount of PCDGF in said sample wherein the amount of PCDGF is indicative of resistance to the antineoplastic effects of HER2 antagonists.
BRIEF DESCRIPTION OF THE DRAWINGS
~ooos~ FIG. 1 summarizes pathological studies in paraffin embedded human breast cancer biopsies. PCDGF and erbB2 staining were monitored by immunohistochemisty. Thus, overexpression of PCDGF in cells overexpressing erbB2 renders the cells HER2 antagonist resistant. It is known that 25% of patients with tumors that overexpress erbB2 will be responsive to HER2 antagonist therapy.
~ooo ~ ~ FIG. 2 shows erbB2 levels in MCF7 cells (breast cancer cells) transfected with erbB2.
Cooos~ FIG. 3 shows PCDGF expression in MCF7 cells transfected with erbB2.
FIG. 4 shows PCDGF protein levels in conditioned media of cells transfected with both PCDGF and erbB2 (D.c2) and erbB2 alone (erbB2.c1).
FIG. 5 shows that PCDGF stimulates erbB2 phosphorylation in erbB2 overexpressing cells.
Coo 1 y FIG. 6 shows that PCDGF stimulates erbB2 phosphorylation in other breast cancer cells that are known to overexpress erbB2 such as BT474 and SKBR3 (see FIG. 12).
Cooiz~ FIG. 7 shows the dose response of PCDGF stimulation of erbB2 in BT474 cells.
Coo~ga FIG. 8 shows the long term growth of erbB2 cells in response to Herceptin~. Herceptin~ inhibits proliferation of erbB2 overexpressing cells as it is a monoclonal antibody that neutralizes erbB2 and inhibits proliferation in erbB2 overexpressing breast cancer cells.
[oo i ~~ FIG. 9 shows that when PCDGF is overexpressed in the erbB2 overexpressing cells (e.g., D.c2 cells), Herceptin~ no longer inhibits the proliferation of the breast cancer cells.
Cooi5~ FIG. 10 shows the same results as FIG. 9 using thymidine incorporation as a measure of cell growth.
~oois~ FIG. 11 shows that PCDGF overexpression prevents Herceptin0 inhibition of breast cancer cells in a soft agar (tumorigenesis) assay in erbB2 overexpressing cells. These results demonstrate that overexpression of PCDGF
confers Herceptin~ resistance in erbB2 overexpressing cells.
Cool ~~ FIG. 22 shows that PCDGF stimulates erbB2 phosphorylation in SKBR3 cells. Overexpression of PCDGF confers Herceptin~ resistance in SKBR3 cells overexpressing erbB2.
DETAILED DESCRIPTION OF THE INVENTION
Coo i s~ PCDGF is an 88 kDa autocrine growth factor characterized in our laboratory and shown to be overexpressed in and induce tumorigenesis of a wide variety of human and animal tumor cells (e.g., neuroblastoma, glioblastoma, astrocytoma, sarcomas, and cancers of the prostate, blood, cerebrospinal fluid, liver, kidney, breast, head and neck, pharynx, thyroid, pancreas, stomach, colon, colorectal, uterus, cervix, bone, bone marrow, testes, brain, neural tissue, ovary, skin, and lung). See, ~ U.S. Patent Number 6,309,826, hereby incorporated by reference in its entirety.
~oois~ PCDGF also confers resistance to the antineoplastic effects HER2 antagonists (e.g., Herceptin~, HER2 kinase inhibitors) on tumor cells. As described in U.S. Patent Number 6,309,826, overexpression of PCDGF leads to uncontrolled cell growth and increased tumorigenesis. The degree of PCDGF
Cooi5~ FIG. 10 shows the same results as FIG. 9 using thymidine incorporation as a measure of cell growth.
~oois~ FIG. 11 shows that PCDGF overexpression prevents Herceptin0 inhibition of breast cancer cells in a soft agar (tumorigenesis) assay in erbB2 overexpressing cells. These results demonstrate that overexpression of PCDGF
confers Herceptin~ resistance in erbB2 overexpressing cells.
Cool ~~ FIG. 22 shows that PCDGF stimulates erbB2 phosphorylation in SKBR3 cells. Overexpression of PCDGF confers Herceptin~ resistance in SKBR3 cells overexpressing erbB2.
DETAILED DESCRIPTION OF THE INVENTION
Coo i s~ PCDGF is an 88 kDa autocrine growth factor characterized in our laboratory and shown to be overexpressed in and induce tumorigenesis of a wide variety of human and animal tumor cells (e.g., neuroblastoma, glioblastoma, astrocytoma, sarcomas, and cancers of the prostate, blood, cerebrospinal fluid, liver, kidney, breast, head and neck, pharynx, thyroid, pancreas, stomach, colon, colorectal, uterus, cervix, bone, bone marrow, testes, brain, neural tissue, ovary, skin, and lung). See, ~ U.S. Patent Number 6,309,826, hereby incorporated by reference in its entirety.
~oois~ PCDGF also confers resistance to the antineoplastic effects HER2 antagonists (e.g., Herceptin~, HER2 kinase inhibitors) on tumor cells. As described in U.S. Patent Number 6,309,826, overexpression of PCDGF leads to uncontrolled cell growth and increased tumorigenesis. The degree of PCDGF
overexpression directly correlates with the degree of cellular tumorigenicity.
Cells overexpressing PCDGF do not require external signals to maintain uncontrolled cell growth. Loss of regulated cell growth, such as a loss in responsiveness to insulin and/or estrogen, leads to increased malignancy and excessive unregulated cell growth. Development of methods and compositions that interfere with the tumorigenic activity of PCDGF is therefore of great interest for the treatment of cancer.
HER2 antagonist therapy (e.g., but not limited to, Herceptin~
and HER2 kinase inhibitors) is useful for treatment of patients with metastatic breast cancer, including patients whose tumors overexpress the HER2 protein and who have received one or more chemotherapy regimens for their metastatic disease. Herceptin~, in particular, also is approved for combination therapy with paclitaxel for treatment of patients with metastatic breast cancer whose tumors overexpress the HER2 protein and who have not received chemotherapy for their metastatic disease. However, only 25% of the patients treated with Herceptin~ or any other antibody to HER2 are responsive to such therapy.
PCDGF antagonists, such as anti-PCDGF antibodies, interfere with the biological activity of PCDGF (e.g., tumorigenic activity) by binding PCDGF directly and preventing PCDGF from transmitting cell growth signals to a target cell (e.g., breast cancer cell). An anti-PCDGF antibody may bind the active site of PCDGF (e.g., the PCDGF receptor binding site) and prevent PCDGF
from binding to its receptor. Alternatively, anti-PCDGF antibodies may bind to a site on PCDGF other than the active site, alter the conformation of the active site, and thus render PCDGF incapable of binding to its receptor. Anti-PCDGF
antibodies include PCDGF neutralizing antibodies. "Neutralizing" antibodies have the ability to inhibit or block the normal biological activity of PCDGF, including PCDGF's ability to stimulate cell proliferation, increase cell survival, block apoptosis, or induce tumor growth in animals and in humans. PCDGF
antagonists also include nucleic acids (antisense, siRNA etc.) and anti-PCDGF
receptor antibodies. PCDGF confers resistance to the antineoplastic effects of HER2 antagonists. Immunohistochemistry studies in paraffin embedded human breast cancer biopsies showed that PCDGF was highly expressed in 25% of c-erbB2 positive (+3) invasive ductal carcinomas.
CoozZa Preferred embodiments of the invention are directed to methods of restoring growth inhibition sensitivity to tumor cells resistant to the antineoplastic effects of HER2 antagonists by administering a PCDGF antagonist to a human patient in an amount effective to restore the patient's growth inhibition sensitivity to HER2 antagonists.
[00~3~ The term "HER2 antagonist" refers to any molecule (e.g., protein, peptide, small molecule, nucleic acid, antisense, or siRNA) that is capable of binding, interfering with, or inhibiting the activity of HER2 or any analogs or derivatives of HER2 that retain the neoplastic properties of HER2 (e.g., but not limited to Herceptin~ and HER2 kinase inhibitors).
In one embodiment, a HER2 antagonist includes a molecule that can target or selectively bind to HER2 and, for example, deliver a toxin or other compound or molecule to kill a cell or inhibit cell growth. For example, a antibody can be coupled to a toxin or chemotherapeutic agent that is delivered to a tumor cell after the antibody binds to HER2. HER2 antagonists also include molecules (e.g., peptides, small molecules, antisense molecules, and siRNA) that modulate the biological activity of molecules that regulate the activity of HER2.
A HER2 antagonist can be an antibody that recruits an immune response, e.g., through ADCC (antibody dependent cell cytotoxicity).
Coo~5~ The term "PCDGF antagonist" refers to any molecule (e.g., protein, peptide, small molecule, nucleic acid, antisense, or siRNA) that is capable of selectively binding, interfering with, or inhibiting the biological activity of PCDGF including, but not limited to, the HER2 antagonist resistance-conferring activity of PCDGF or any analogs or derivatives of PCDGF that retain the properties of PCDGF. In one embodiment, a PCDGF antagonist includes PCDGF receptor antibodies that can target or selectively bind to the PCDGF
receptor and, for example, deliver a toxin or other compound or molecule to kill a cell or inhibit cell growth.
The invention also provides pharmaceutical compositions containing one or more HER2 antagonists and one or more PCDGF antagonists.
Patients receiving both HER2 and PCDGF antagonists will benefit from the antineoplastic effects of each antagonist in that the PCDGF antagonist can restore the HER2 antagonist sensitivity of patients resistant to HER therapy.
Pharmaceutical compositions comprising a HER2 antagonist, a PCDGF
antagonist, and an acceptable pharmaceutical carrier (such as water, saline, Ringer's solution, dextrose solution, and human serum albumin) are also provided.
Preferred embodiments of the invention also include methods of determining whether a patient is resistant to the antineoplastic effects of antagonist therapy by obtaining a biological sample containing cells from the patient; detecting PCDGF in the biological sample; and determining the amount of PCDGF in the sample wherein the amount of PCDGF is indicative of resistance to the antineoplastic effects of HER2 antagonist therapy.
~00~8~ The term antibody herein includes but is not limited to human and non-human polyclonal antibodies, human and non-human monoclonal antibodies (mAbs), chimeric antibodies, anti-idiotypic antibodies (anti-IdAb), neutralizing antibodies, non-neutralizing antibodies, and humanized antibodies.
Polyclonal antibodies are heterogeneous populations of antibody molecules derived either from sera of animals immunized with an antigen or from chicken eggs. Monoclonal antibodies ("mAbs") are substantially homogeneous populations of antibodies to specific antigens. mAbs may be obtained by any suitable method. Such antibodies may be of any immunological class including IgG, IgM, IgE, IgA, IgD and any subclass thereof. The term antibody is also meant to include both intact molecules as well as fragments thereof such as, for example, Fab and F(ab')2, which are capable of binding to the antigen. Fab and F(ab')2 fragments Lack the Fc fragment of intact antibody, clear more rapidly from the circulation, and may have less non-specific tissue binding than an intact antibody. Such fragments are typically produced by proteolytic cleavage, using enzymes such as papain (to generate Fab fragments) and pepsin (to generate F(ab')2 fragments).
[00z9~ In yet another embodiment, a subject's cells (e.g., tumor cells) are removed from the body, transfected with a polynucleodde encoding a HER2 antagonist and a PCDGF antagonist, and injected at the site of the tumor.
Expression of the polynucleoddes encoding the HER2 antagonist and PCDGF
antagonist localizes the HER2 and PCDGF antagonists at the tumor site. A
subject's cells (e.g., tumor cells) can also be directly transfected in the body (e.g., in situ) with a construct containing a nucleic acid encoding a HER2 antagonist and/or a PCDGF antagonist. Expression of the nucleic acid results in production of the HER2 antagonist and/or PCDGF antagonist inside the transfected cell.
[ooso~ For in vivo applications, PCDGF antagonists, and HER2 antagonists can be provided to a subject by a variety of administration routes and dosage forms. A subject, preferably a human subject, suffering from a neoplastic condition, including but not limited to breast cancer, or other disease condition associated with increased HER2 and/or PCDGF expression, is treated consecutively or simultaneously with an HER2 antagonist and a PCDGF
antagonist. In one embodiment, the HER2 antagonist and PCDGF antagonist are co-administered. In another embodiment, the HERZ antagonist and PCDGF
antagonist are sequentially administered, preferably the Ievel of PCDGF
ascertained and, if elevated, the PCDGF antagonist being administered first.
[oo~ 1~ Treatment with a PCDGF antagonist can precede, follow, or be conducted concurrently with treatment with a HER2 antagonist. Treatment with a PCDGF antagonist may precede or follow treatment with a HER2 antagonist by intervals ranging from minutes to weeks. In another embodiment, a PCDGF
antagonist and a HER2 antagonist are administered in a way to ensure that a prolonged period of time does not elapse between the time of administration of each agent. For example, each antagonist can be administered to a patient within seconds, minutes, or hours of the other antagonist.
Coo3z~ In a preferred embodiment, treatment with a PCDGF antagonist increases c-erbB2 phosphorylation by at Ieast 10%, more preferably by at least 50%. In a further preferred embodiment, treatment with a PCDGF antagonist increases HER2 sensitivity by at least two-fold, more preferably by at least three-fold.
~ooss~ 'The antagonists of the present invention may be administered by any means that achieves their intended purpose. For example, antibody administration may be by various routes including but not limited to subcutaneous, intravenous, intradermal, intramuscular, intraperitoneal, and oral.
Parenteral administration can be by bolus injection or by gradual perfusion over time. Preparations for parenteral administration include sterile aqueous or non-aqueous solutions, suspensions and emulsions, which may contain auxiliary agents or excipients. Pharmaceutical compositions such as tablets and capsules can also be prepared. It is understood that the dosage will be dependent upon the age, sex and weight of the recipient, kind of concurrent treatment, if any, frequency of treatment and the nature of the effect desired.
[oos4~ In another embodiment of the invention, a HER2 antagonist in combination with a PCDGF antagonist and/or chemotherapy (e.g., paclitaxel, cisplatin (CDDP), carboplatin, procarbazine, mechlorethamine, cyclophosphamide, camptothecin, ifosfamide, melphalan, chlorambucil, busulfan, nitrosurea, dactinomycin, daunorubicin, doxorubicin, bleomycin, plicomycin, mitomycin, etoposide (VP16), tamoxifen, raloxifene, estrogen receptor binding agents, taxol, gemcitabien, navelbine, farnesyl-protein tansferase inhibitors, transplatinum, 5-fluorouracil, vincristin, vinblastin andmethotrexate, or any analog or derivative or variant of the foregoing) can be used to treat metastatic breast cancer or cancers of a variety of tissues (e.g., neuroblastoma, glioblastoma, astrocytoma, sarcomas, and cancers of the prostate, blood, liver, kidney, breast, head and neck, pharynx, thyroid, pancreas, stomach, colon, colorectal, uterus, cervix, bone, bone marrow, testes, brain, neural tissue, ovary, skin, and lung). Combination therapy can be administered independently of diagnostic evaluation.
~oos5~ In a preferred modality of the invention, a breast cancer patient (e.g., having metastadc breast cancer and overexpressing HER2) is given HER2 antagonist treatment with, for example, Herceptin. Although, the patient exhibits some degree of tumor growth inhibition, her progress declines, and she becomes resistant to HER2 antagonist therapy. A biopsy or serum sample is conducted and reveals that the patient has elevated levels of PCDGF. In another embodiment, the patient's PCDGF levels are determined prior to beginning HER2 antagonist therapy. The patient is then treated with a PCDGF antagonist alone, or co-administration of PCDGF antagonist and HER2 antagonist, to restore sensitivity to HER2 antagonist therapy. Following treatment with PCDGF
antagonist, the patient is again responsive to HER2 antagonist therapy. The PCDGF level of the patient is periodically monitored, for example, weekly or monthly throughout the HER2 therapy and a PCDGF antagonist provided to again restore HER2 sensitivity if needed. Alternatively, the patient can continue to receive co-administration of PCDGF antagonist and HER2 antagonist.
[oo3s~ The ranges of effective doses provided below are not intended to limit the invention and merely represent illustrative dose ranges. However the most preferred dosage will be tailored to the individual subject as is understood and determinable by one of ordinary skill in the art given the teachings herein.
The total dose required for each treatment may be administered by multiple doses or in a single dose. In one embodiment, effective amounts of each of a HER2 antibody and a PCDGF antibody are from about 0.01 ng to about 500 qg/ml and preferably from about 10 ng to about 100 ~ug/ml. An HER2 antagonist and a PCDGF antagonist may be administered alone, administered together, or in conjunction with other therapeutics. In another embodiment, the amount of each antagonist administered will typically be in the range of about 0.1 to about mg/kg of patient weight, so long as the HER2 and PCDGF antagonists are administered to the patient in therapeutically effective amounts (i.e., amounts that eliminate and/or reduce the patient's tumor burden or restore sensitivity to the antineoplastic effects of any HER2 antagonists).
Coos ~ ~ A preferred treatment regimen comprises co-administration of an effective amount of a HER2 antagonist and a PCDGF antagonist over a period of one or several weeks and including between about one week and six months.
In another embodiment, a HER2 antagonist can be provided in an initial dose of 4 mg/ kg followed by 2 mg/kg intravenous (i.v.) weekly; or dose of 8 mg/kg initial dose followed by 4 mg/kg i.v. weekly. Treatment with a PCDGF
antagonist can be provided, for example, in an initial dose of 4 mg/ kg followed by 2 mg/kg intravenous (i.v.) weekly; or a dose of 8 mg/kg initial dose followed by 4 mg/kg i.v. weekly. Additional examples of regimens for cancer treatment that can be used for treatment with a HER2 antagonist, a PCDGF antagonist, and/or chemotherapeuHc agents are disclosed in the following articles, hereby incorporated by reference in their entirety: Hamid, O., J Am Pharm Assoc, 2004 Jan-Feb;44(1):52-8; Kubo et al., Anticancer Res. 2003 Nov-Dec; 23(6a):4443-9;
Slamon et al., N Engl J Med. 2001;344:783-792.; Baselga et al., Cancer Res 1998;58:2825-2831; and Jones, et al., Ann Oncol. 2003 Dec;l4(12):1697-704.
ANTAGONIST THERAPY
~ooss~ To investigate the role of PCDGF in HER2 antagonist resistance of c-erbB2 overexpressing cells, MCF-7 breast cancer cells were stably transfected with c-erbB2 cDNA. A c-erbB2 overexpressing clone (erbB2.c1) was selected, and used for the subsequent stable transfection with PCDGF cDNA. Clones overexpressing both c-erbB2 and PCDGF were obtained and further examined in comparison to single erbB2 overexpressing cells. We show in the appended Figures that dual-overexpressing clones (D.c2) (i.e., clones overexpressing both PCDGF and erbB2) were HER2 antagonist resistant with respect to their ability to proliferate in vitro, whereas the erbB2.c1 cells were sensitive to Herceptin~
treatment. These Herceptin~-sensitive erbB2.c1 cells also had a growth advantage when treated with PCDGF. Moreover PCDGF stimulates c-erbB2 phosphorylation in these cells. We also show that the dual expressing D.c2 cells were more resistant than the erbB2.c1 cells to the antiestrogen ICI 182,780.
Our findings demonstrate that PCDGF confers Herceptin~ resistance in c-erbB2 overexpressing tumors. See Figures. Blocking PCDGF action would be beneficial for patients undergoing HER2 antagonist treatment by reversing the initial resistance.
Gi t ',..' O
v ~ .C ~ .._ C~ .~
4~ O
O ~ ~ O
t_I~ +~ ~ _O 4 ~i _ O .~
.O ~ ~ ~ O
V ~ ~ .
O ~ ~ ~ .O
3 ~, 'O O
v 00 ._ ._G~~ tn .tAO
G~ tI~
V t/~ ~ V ._ ~ ai O L ~ v ,~ ~ = O .. d' ~ tl~ O
o~ >,N~ ~, ~
V ~ ca ~: ~ V
v~
~ 00 .~ try ca O c.~ = o tIf t~ .'~ ~ .O
O -O .~ ~ 4-N ~ .~- ~ t/~
N
O ;~ N ~i L tI~
V O LL
G~ O. ~ L ~ ~ ~ 'O
O ~ ~ .= O
c~t0~ .-~t/~.~~i t LZ ~ V D ~ >
Ll.l ~ N
N ~j G~ C' ~ m Lt a~
~ ~ ~ >
4.. O ~ N t/~ ~ ~ O
. _ IJ~
L. ~ L ~ O _~ ~ ' Q (I~
N = ~ ~ ~ ~ ~ +~
ap ~ '~"' ~ .O
V _~ ' . _ "~ ~ O .t~ ~ c~
_LNt~i t/~ ~ ~ '~~'_ V~ ~,0~._ t~ ~~~~~'O
= L~ ~ Qo._ ca tn ~ '~ .~ >. a~ u.~ a~ a> > ~ a~ t,~l m O
tL ~ - Ls. G~ .O 4~ ~c o ~ ~ a- 3 = ca ~ . ._ ~ tt> > c~ v ~. a~ L: _v~ c ~_ 3 ~ ~ (,~ ~; ~ L ~
L N r ~ 4~ ~ .. ~ ~ G~ O = ,O
~- =- Z3 o ~ ~ ~ ,~ ~ ~ s L Q ~ c~a v ~J ~ t c~ ~ ~ oo tn 'd" tn - 'O . _ ._~.*.~E- >,3 O ~~ ~ ~ ai -_ ~~'"' ~'~~ O~L~=~O~tli V ~ ~ ~ ~ ~ ~-. _ .~ ~ p . _ .,,", ca LL >
- L ~ ~ >
._ G~ ~. '~ ~ Q V ~ d; ~ '= ~ ~ O
/- O. t~ ~ ~ ~- t~ Q "~ ~ ~ ~ ~ ~r0- O.
Cells overexpressing PCDGF do not require external signals to maintain uncontrolled cell growth. Loss of regulated cell growth, such as a loss in responsiveness to insulin and/or estrogen, leads to increased malignancy and excessive unregulated cell growth. Development of methods and compositions that interfere with the tumorigenic activity of PCDGF is therefore of great interest for the treatment of cancer.
HER2 antagonist therapy (e.g., but not limited to, Herceptin~
and HER2 kinase inhibitors) is useful for treatment of patients with metastatic breast cancer, including patients whose tumors overexpress the HER2 protein and who have received one or more chemotherapy regimens for their metastatic disease. Herceptin~, in particular, also is approved for combination therapy with paclitaxel for treatment of patients with metastatic breast cancer whose tumors overexpress the HER2 protein and who have not received chemotherapy for their metastatic disease. However, only 25% of the patients treated with Herceptin~ or any other antibody to HER2 are responsive to such therapy.
PCDGF antagonists, such as anti-PCDGF antibodies, interfere with the biological activity of PCDGF (e.g., tumorigenic activity) by binding PCDGF directly and preventing PCDGF from transmitting cell growth signals to a target cell (e.g., breast cancer cell). An anti-PCDGF antibody may bind the active site of PCDGF (e.g., the PCDGF receptor binding site) and prevent PCDGF
from binding to its receptor. Alternatively, anti-PCDGF antibodies may bind to a site on PCDGF other than the active site, alter the conformation of the active site, and thus render PCDGF incapable of binding to its receptor. Anti-PCDGF
antibodies include PCDGF neutralizing antibodies. "Neutralizing" antibodies have the ability to inhibit or block the normal biological activity of PCDGF, including PCDGF's ability to stimulate cell proliferation, increase cell survival, block apoptosis, or induce tumor growth in animals and in humans. PCDGF
antagonists also include nucleic acids (antisense, siRNA etc.) and anti-PCDGF
receptor antibodies. PCDGF confers resistance to the antineoplastic effects of HER2 antagonists. Immunohistochemistry studies in paraffin embedded human breast cancer biopsies showed that PCDGF was highly expressed in 25% of c-erbB2 positive (+3) invasive ductal carcinomas.
CoozZa Preferred embodiments of the invention are directed to methods of restoring growth inhibition sensitivity to tumor cells resistant to the antineoplastic effects of HER2 antagonists by administering a PCDGF antagonist to a human patient in an amount effective to restore the patient's growth inhibition sensitivity to HER2 antagonists.
[00~3~ The term "HER2 antagonist" refers to any molecule (e.g., protein, peptide, small molecule, nucleic acid, antisense, or siRNA) that is capable of binding, interfering with, or inhibiting the activity of HER2 or any analogs or derivatives of HER2 that retain the neoplastic properties of HER2 (e.g., but not limited to Herceptin~ and HER2 kinase inhibitors).
In one embodiment, a HER2 antagonist includes a molecule that can target or selectively bind to HER2 and, for example, deliver a toxin or other compound or molecule to kill a cell or inhibit cell growth. For example, a antibody can be coupled to a toxin or chemotherapeutic agent that is delivered to a tumor cell after the antibody binds to HER2. HER2 antagonists also include molecules (e.g., peptides, small molecules, antisense molecules, and siRNA) that modulate the biological activity of molecules that regulate the activity of HER2.
A HER2 antagonist can be an antibody that recruits an immune response, e.g., through ADCC (antibody dependent cell cytotoxicity).
Coo~5~ The term "PCDGF antagonist" refers to any molecule (e.g., protein, peptide, small molecule, nucleic acid, antisense, or siRNA) that is capable of selectively binding, interfering with, or inhibiting the biological activity of PCDGF including, but not limited to, the HER2 antagonist resistance-conferring activity of PCDGF or any analogs or derivatives of PCDGF that retain the properties of PCDGF. In one embodiment, a PCDGF antagonist includes PCDGF receptor antibodies that can target or selectively bind to the PCDGF
receptor and, for example, deliver a toxin or other compound or molecule to kill a cell or inhibit cell growth.
The invention also provides pharmaceutical compositions containing one or more HER2 antagonists and one or more PCDGF antagonists.
Patients receiving both HER2 and PCDGF antagonists will benefit from the antineoplastic effects of each antagonist in that the PCDGF antagonist can restore the HER2 antagonist sensitivity of patients resistant to HER therapy.
Pharmaceutical compositions comprising a HER2 antagonist, a PCDGF
antagonist, and an acceptable pharmaceutical carrier (such as water, saline, Ringer's solution, dextrose solution, and human serum albumin) are also provided.
Preferred embodiments of the invention also include methods of determining whether a patient is resistant to the antineoplastic effects of antagonist therapy by obtaining a biological sample containing cells from the patient; detecting PCDGF in the biological sample; and determining the amount of PCDGF in the sample wherein the amount of PCDGF is indicative of resistance to the antineoplastic effects of HER2 antagonist therapy.
~00~8~ The term antibody herein includes but is not limited to human and non-human polyclonal antibodies, human and non-human monoclonal antibodies (mAbs), chimeric antibodies, anti-idiotypic antibodies (anti-IdAb), neutralizing antibodies, non-neutralizing antibodies, and humanized antibodies.
Polyclonal antibodies are heterogeneous populations of antibody molecules derived either from sera of animals immunized with an antigen or from chicken eggs. Monoclonal antibodies ("mAbs") are substantially homogeneous populations of antibodies to specific antigens. mAbs may be obtained by any suitable method. Such antibodies may be of any immunological class including IgG, IgM, IgE, IgA, IgD and any subclass thereof. The term antibody is also meant to include both intact molecules as well as fragments thereof such as, for example, Fab and F(ab')2, which are capable of binding to the antigen. Fab and F(ab')2 fragments Lack the Fc fragment of intact antibody, clear more rapidly from the circulation, and may have less non-specific tissue binding than an intact antibody. Such fragments are typically produced by proteolytic cleavage, using enzymes such as papain (to generate Fab fragments) and pepsin (to generate F(ab')2 fragments).
[00z9~ In yet another embodiment, a subject's cells (e.g., tumor cells) are removed from the body, transfected with a polynucleodde encoding a HER2 antagonist and a PCDGF antagonist, and injected at the site of the tumor.
Expression of the polynucleoddes encoding the HER2 antagonist and PCDGF
antagonist localizes the HER2 and PCDGF antagonists at the tumor site. A
subject's cells (e.g., tumor cells) can also be directly transfected in the body (e.g., in situ) with a construct containing a nucleic acid encoding a HER2 antagonist and/or a PCDGF antagonist. Expression of the nucleic acid results in production of the HER2 antagonist and/or PCDGF antagonist inside the transfected cell.
[ooso~ For in vivo applications, PCDGF antagonists, and HER2 antagonists can be provided to a subject by a variety of administration routes and dosage forms. A subject, preferably a human subject, suffering from a neoplastic condition, including but not limited to breast cancer, or other disease condition associated with increased HER2 and/or PCDGF expression, is treated consecutively or simultaneously with an HER2 antagonist and a PCDGF
antagonist. In one embodiment, the HER2 antagonist and PCDGF antagonist are co-administered. In another embodiment, the HERZ antagonist and PCDGF
antagonist are sequentially administered, preferably the Ievel of PCDGF
ascertained and, if elevated, the PCDGF antagonist being administered first.
[oo~ 1~ Treatment with a PCDGF antagonist can precede, follow, or be conducted concurrently with treatment with a HER2 antagonist. Treatment with a PCDGF antagonist may precede or follow treatment with a HER2 antagonist by intervals ranging from minutes to weeks. In another embodiment, a PCDGF
antagonist and a HER2 antagonist are administered in a way to ensure that a prolonged period of time does not elapse between the time of administration of each agent. For example, each antagonist can be administered to a patient within seconds, minutes, or hours of the other antagonist.
Coo3z~ In a preferred embodiment, treatment with a PCDGF antagonist increases c-erbB2 phosphorylation by at Ieast 10%, more preferably by at least 50%. In a further preferred embodiment, treatment with a PCDGF antagonist increases HER2 sensitivity by at least two-fold, more preferably by at least three-fold.
~ooss~ 'The antagonists of the present invention may be administered by any means that achieves their intended purpose. For example, antibody administration may be by various routes including but not limited to subcutaneous, intravenous, intradermal, intramuscular, intraperitoneal, and oral.
Parenteral administration can be by bolus injection or by gradual perfusion over time. Preparations for parenteral administration include sterile aqueous or non-aqueous solutions, suspensions and emulsions, which may contain auxiliary agents or excipients. Pharmaceutical compositions such as tablets and capsules can also be prepared. It is understood that the dosage will be dependent upon the age, sex and weight of the recipient, kind of concurrent treatment, if any, frequency of treatment and the nature of the effect desired.
[oos4~ In another embodiment of the invention, a HER2 antagonist in combination with a PCDGF antagonist and/or chemotherapy (e.g., paclitaxel, cisplatin (CDDP), carboplatin, procarbazine, mechlorethamine, cyclophosphamide, camptothecin, ifosfamide, melphalan, chlorambucil, busulfan, nitrosurea, dactinomycin, daunorubicin, doxorubicin, bleomycin, plicomycin, mitomycin, etoposide (VP16), tamoxifen, raloxifene, estrogen receptor binding agents, taxol, gemcitabien, navelbine, farnesyl-protein tansferase inhibitors, transplatinum, 5-fluorouracil, vincristin, vinblastin andmethotrexate, or any analog or derivative or variant of the foregoing) can be used to treat metastatic breast cancer or cancers of a variety of tissues (e.g., neuroblastoma, glioblastoma, astrocytoma, sarcomas, and cancers of the prostate, blood, liver, kidney, breast, head and neck, pharynx, thyroid, pancreas, stomach, colon, colorectal, uterus, cervix, bone, bone marrow, testes, brain, neural tissue, ovary, skin, and lung). Combination therapy can be administered independently of diagnostic evaluation.
~oos5~ In a preferred modality of the invention, a breast cancer patient (e.g., having metastadc breast cancer and overexpressing HER2) is given HER2 antagonist treatment with, for example, Herceptin. Although, the patient exhibits some degree of tumor growth inhibition, her progress declines, and she becomes resistant to HER2 antagonist therapy. A biopsy or serum sample is conducted and reveals that the patient has elevated levels of PCDGF. In another embodiment, the patient's PCDGF levels are determined prior to beginning HER2 antagonist therapy. The patient is then treated with a PCDGF antagonist alone, or co-administration of PCDGF antagonist and HER2 antagonist, to restore sensitivity to HER2 antagonist therapy. Following treatment with PCDGF
antagonist, the patient is again responsive to HER2 antagonist therapy. The PCDGF level of the patient is periodically monitored, for example, weekly or monthly throughout the HER2 therapy and a PCDGF antagonist provided to again restore HER2 sensitivity if needed. Alternatively, the patient can continue to receive co-administration of PCDGF antagonist and HER2 antagonist.
[oo3s~ The ranges of effective doses provided below are not intended to limit the invention and merely represent illustrative dose ranges. However the most preferred dosage will be tailored to the individual subject as is understood and determinable by one of ordinary skill in the art given the teachings herein.
The total dose required for each treatment may be administered by multiple doses or in a single dose. In one embodiment, effective amounts of each of a HER2 antibody and a PCDGF antibody are from about 0.01 ng to about 500 qg/ml and preferably from about 10 ng to about 100 ~ug/ml. An HER2 antagonist and a PCDGF antagonist may be administered alone, administered together, or in conjunction with other therapeutics. In another embodiment, the amount of each antagonist administered will typically be in the range of about 0.1 to about mg/kg of patient weight, so long as the HER2 and PCDGF antagonists are administered to the patient in therapeutically effective amounts (i.e., amounts that eliminate and/or reduce the patient's tumor burden or restore sensitivity to the antineoplastic effects of any HER2 antagonists).
Coos ~ ~ A preferred treatment regimen comprises co-administration of an effective amount of a HER2 antagonist and a PCDGF antagonist over a period of one or several weeks and including between about one week and six months.
In another embodiment, a HER2 antagonist can be provided in an initial dose of 4 mg/ kg followed by 2 mg/kg intravenous (i.v.) weekly; or dose of 8 mg/kg initial dose followed by 4 mg/kg i.v. weekly. Treatment with a PCDGF
antagonist can be provided, for example, in an initial dose of 4 mg/ kg followed by 2 mg/kg intravenous (i.v.) weekly; or a dose of 8 mg/kg initial dose followed by 4 mg/kg i.v. weekly. Additional examples of regimens for cancer treatment that can be used for treatment with a HER2 antagonist, a PCDGF antagonist, and/or chemotherapeuHc agents are disclosed in the following articles, hereby incorporated by reference in their entirety: Hamid, O., J Am Pharm Assoc, 2004 Jan-Feb;44(1):52-8; Kubo et al., Anticancer Res. 2003 Nov-Dec; 23(6a):4443-9;
Slamon et al., N Engl J Med. 2001;344:783-792.; Baselga et al., Cancer Res 1998;58:2825-2831; and Jones, et al., Ann Oncol. 2003 Dec;l4(12):1697-704.
ANTAGONIST THERAPY
~ooss~ To investigate the role of PCDGF in HER2 antagonist resistance of c-erbB2 overexpressing cells, MCF-7 breast cancer cells were stably transfected with c-erbB2 cDNA. A c-erbB2 overexpressing clone (erbB2.c1) was selected, and used for the subsequent stable transfection with PCDGF cDNA. Clones overexpressing both c-erbB2 and PCDGF were obtained and further examined in comparison to single erbB2 overexpressing cells. We show in the appended Figures that dual-overexpressing clones (D.c2) (i.e., clones overexpressing both PCDGF and erbB2) were HER2 antagonist resistant with respect to their ability to proliferate in vitro, whereas the erbB2.c1 cells were sensitive to Herceptin~
treatment. These Herceptin~-sensitive erbB2.c1 cells also had a growth advantage when treated with PCDGF. Moreover PCDGF stimulates c-erbB2 phosphorylation in these cells. We also show that the dual expressing D.c2 cells were more resistant than the erbB2.c1 cells to the antiestrogen ICI 182,780.
Our findings demonstrate that PCDGF confers Herceptin~ resistance in c-erbB2 overexpressing tumors. See Figures. Blocking PCDGF action would be beneficial for patients undergoing HER2 antagonist treatment by reversing the initial resistance.
Gi t ',..' O
v ~ .C ~ .._ C~ .~
4~ O
O ~ ~ O
t_I~ +~ ~ _O 4 ~i _ O .~
.O ~ ~ ~ O
V ~ ~ .
O ~ ~ ~ .O
3 ~, 'O O
v 00 ._ ._G~~ tn .tAO
G~ tI~
V t/~ ~ V ._ ~ ai O L ~ v ,~ ~ = O .. d' ~ tl~ O
o~ >,N~ ~, ~
V ~ ca ~: ~ V
v~
~ 00 .~ try ca O c.~ = o tIf t~ .'~ ~ .O
O -O .~ ~ 4-N ~ .~- ~ t/~
N
O ;~ N ~i L tI~
V O LL
G~ O. ~ L ~ ~ ~ 'O
O ~ ~ .= O
c~t0~ .-~t/~.~~i t LZ ~ V D ~ >
Ll.l ~ N
N ~j G~ C' ~ m Lt a~
~ ~ ~ >
4.. O ~ N t/~ ~ ~ O
. _ IJ~
L. ~ L ~ O _~ ~ ' Q (I~
N = ~ ~ ~ ~ ~ +~
ap ~ '~"' ~ .O
V _~ ' . _ "~ ~ O .t~ ~ c~
_LNt~i t/~ ~ ~ '~~'_ V~ ~,0~._ t~ ~~~~~'O
= L~ ~ Qo._ ca tn ~ '~ .~ >. a~ u.~ a~ a> > ~ a~ t,~l m O
tL ~ - Ls. G~ .O 4~ ~c o ~ ~ a- 3 = ca ~ . ._ ~ tt> > c~ v ~. a~ L: _v~ c ~_ 3 ~ ~ (,~ ~; ~ L ~
L N r ~ 4~ ~ .. ~ ~ G~ O = ,O
~- =- Z3 o ~ ~ ~ ,~ ~ ~ s L Q ~ c~a v ~J ~ t c~ ~ ~ oo tn 'd" tn - 'O . _ ._~.*.~E- >,3 O ~~ ~ ~ ai -_ ~~'"' ~'~~ O~L~=~O~tli V ~ ~ ~ ~ ~ ~-. _ .~ ~ p . _ .,,", ca LL >
- L ~ ~ >
._ G~ ~. '~ ~ Q V ~ d; ~ '= ~ ~ O
/- O. t~ ~ ~ ~- t~ Q "~ ~ ~ ~ ~ ~r0- O.
Claims (10)
1. A method of restoring growth inhibition sensitivity to a tumor patient resistant to growth inhibition by an HER2 antagonist, comprising administering a PCDGF
antagonist to the patient in an amount effective to restore growth inhibition sensitivity to an HER2 antagonist.
antagonist to the patient in an amount effective to restore growth inhibition sensitivity to an HER2 antagonist.
2. A method of inhibiting tumor cell growth comprising administering a PCDGF antagonist and an HER2 antagonist to the patient in an amount effective to inhibit tumor growth.
3. The method of claim 2, wherein the antagonists are co-administered.
4. A pharmaceutical composition comprising an HER2 antagonist, and a PCDGF antagonist.
5. The method/composition of claims 1-4, wherein the HER2 antagonist is Herceptin.
6. The method/composition of claims 1-4, wherein the PCDGF antagonist is an antibody.
7. The method/composition of claims 1-4 is an antisense oligonucleotide.
8. The method/composition of claims 1-4 is a small molecule.
9. The method/composition of claims 1-4 is siRNA.
10. The method of claim 2, further comprising monitoring the patient's level of PCDGF.
Applications Claiming Priority (2)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
US49153603P | 2003-08-01 | 2003-08-01 | |
US60/491,536 | 2003-08-01 |
Publications (2)
Publication Number | Publication Date |
---|---|
CA2459622A1 true CA2459622A1 (en) | 2005-02-01 |
CA2459622C CA2459622C (en) | 2016-12-13 |
Family
ID=34135125
Family Applications (1)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
CA2459622A Expired - Fee Related CA2459622C (en) | 2003-08-01 | 2004-02-27 | Compositions and methods for restoring sensitivity to treatment with her2 antagonists |
Country Status (1)
Country | Link |
---|---|
CA (1) | CA2459622C (en) |
-
2004
- 2004-02-27 CA CA2459622A patent/CA2459622C/en not_active Expired - Fee Related
Also Published As
Publication number | Publication date |
---|---|
CA2459622C (en) | 2016-12-13 |
Similar Documents
Publication | Publication Date | Title |
---|---|---|
BR112019025188A2 (en) | ACTIVABLE ANTI-PDL1 ANTIBODIES AND METHODS OF USE OF THE SAME | |
Friedländer et al. | ErbB-directed immunotherapy: antibodies in current practice and promising new agents | |
US20180251536A1 (en) | Tumor therapy with an anti-vegf antibody | |
RU2689779C2 (en) | Duocarmicin adc showing improved anti-tumour activity in vivo | |
KR101280267B1 (en) | EGF Receptor epitope peptides and uses thereof | |
TWI630916B (en) | Materials and methods for treating or preventing her-3 associated diseases | |
BR112019020508A2 (en) | bispecific antibodies binding to erbb-2 and erbb3 for use in the treatment of cells that have an nrg1 fusion gene | |
US6927203B1 (en) | Treatment of metastatic disease | |
US8445219B2 (en) | Method of determination of receptor binding saturation effected by monoclonal antibodies | |
KR102477179B1 (en) | Compositions and methods for detecting and treating gastric cancer | |
ES2426916T3 (en) | Compositions and procedures to restore sensitivity to treatment with HER2 antagonists | |
KR20050072744A (en) | Methods for regulating cancer | |
KR102428254B1 (en) | Compositions and methods for detection and treatment of ovarian cancer | |
WO2020221198A1 (en) | Bispecific antibody with double her2 sites for tumor immunotherapy | |
CA2459622C (en) | Compositions and methods for restoring sensitivity to treatment with her2 antagonists | |
EA046932B1 (en) | TREATMENT OF METASTATIC BRAIN TUMOR BY ADMINISTRATION OF ANTIBODY-DRUG CONJUGATE |
Legal Events
Date | Code | Title | Description |
---|---|---|---|
EEER | Examination request | ||
MKLA | Lapsed |
Effective date: 20200227 |