WO2008112470A1 - Increased effectiveness of mda-7 with mdr-1 over-expression - Google Patents
Increased effectiveness of mda-7 with mdr-1 over-expression Download PDFInfo
- Publication number
- WO2008112470A1 WO2008112470A1 PCT/US2008/055877 US2008055877W WO2008112470A1 WO 2008112470 A1 WO2008112470 A1 WO 2008112470A1 US 2008055877 W US2008055877 W US 2008055877W WO 2008112470 A1 WO2008112470 A1 WO 2008112470A1
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- mda
- protein
- mdr
- cells
- dox
- Prior art date
Links
- 108090000237 interleukin-24 Proteins 0.000 title claims abstract description 180
- 102000003898 interleukin-24 Human genes 0.000 title claims abstract description 177
- 230000002018 overexpression Effects 0.000 title abstract description 17
- 108090000623 proteins and genes Proteins 0.000 claims abstract description 68
- 102000004169 proteins and genes Human genes 0.000 claims abstract description 37
- 239000002246 antineoplastic agent Substances 0.000 claims abstract description 25
- 229940127089 cytotoxic agent Drugs 0.000 claims abstract description 23
- 238000002512 chemotherapy Methods 0.000 claims abstract description 17
- 150000007523 nucleic acids Chemical class 0.000 claims description 55
- 108020004707 nucleic acids Proteins 0.000 claims description 54
- 102000039446 nucleic acids Human genes 0.000 claims description 54
- 206010028980 Neoplasm Diseases 0.000 claims description 37
- 201000011510 cancer Diseases 0.000 claims description 33
- 208000037265 diseases, disorders, signs and symptoms Diseases 0.000 claims description 24
- 238000000034 method Methods 0.000 claims description 24
- 101000839071 Rattus norvegicus Heterogeneous nuclear ribonucleoprotein M Proteins 0.000 claims description 21
- 230000004797 therapeutic response Effects 0.000 claims description 11
- 230000014509 gene expression Effects 0.000 abstract description 15
- 238000011282 treatment Methods 0.000 abstract description 11
- 201000001441 melanoma Diseases 0.000 abstract description 9
- 230000004044 response Effects 0.000 abstract description 7
- 230000008901 benefit Effects 0.000 abstract description 6
- 230000004069 differentiation Effects 0.000 abstract description 6
- 108700041567 MDR Genes Proteins 0.000 abstract description 2
- 210000004027 cell Anatomy 0.000 description 110
- 208000015181 infectious disease Diseases 0.000 description 29
- 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 27
- 230000036457 multidrug resistance Effects 0.000 description 24
- 239000013598 vector Substances 0.000 description 24
- 230000006907 apoptotic process Effects 0.000 description 20
- 208000035475 disorder Diseases 0.000 description 19
- 102100033350 ATP-dependent translocase ABCB1 Human genes 0.000 description 18
- 108090000672 Annexin A5 Proteins 0.000 description 16
- 102000004121 Annexin A5 Human genes 0.000 description 16
- 230000003247 decreasing effect Effects 0.000 description 16
- 230000000694 effects Effects 0.000 description 16
- 101001017818 Homo sapiens ATP-dependent translocase ABCB1 Proteins 0.000 description 14
- 125000003275 alpha amino acid group Chemical group 0.000 description 13
- 241000700605 Viruses Species 0.000 description 11
- 108700020796 Oncogene Proteins 0.000 description 10
- 229960004679 doxorubicin Drugs 0.000 description 10
- 238000001262 western blot Methods 0.000 description 10
- 238000002474 experimental method Methods 0.000 description 9
- 108010076504 Protein Sorting Signals Proteins 0.000 description 8
- 238000000159 protein binding assay Methods 0.000 description 8
- 239000003814 drug Substances 0.000 description 7
- 238000000684 flow cytometry Methods 0.000 description 7
- 230000006698 induction Effects 0.000 description 7
- 230000001939 inductive effect Effects 0.000 description 7
- 238000002560 therapeutic procedure Methods 0.000 description 7
- 241000701161 unidentified adenovirus Species 0.000 description 7
- 230000001640 apoptogenic effect Effects 0.000 description 6
- 229940079593 drug Drugs 0.000 description 6
- 239000002773 nucleotide Substances 0.000 description 6
- 125000003729 nucleotide group Chemical group 0.000 description 6
- 230000035508 accumulation Effects 0.000 description 5
- 238000009825 accumulation Methods 0.000 description 5
- 230000035945 sensitivity Effects 0.000 description 5
- 229920001817 Agar Polymers 0.000 description 4
- 241000725303 Human immunodeficiency virus Species 0.000 description 4
- 108010047230 Member 1 Subfamily B ATP Binding Cassette Transporter Proteins 0.000 description 4
- 108020004459 Small interfering RNA Proteins 0.000 description 4
- 239000008272 agar Substances 0.000 description 4
- 201000010099 disease Diseases 0.000 description 4
- 239000013604 expression vector Substances 0.000 description 4
- 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 4
- 230000036210 malignancy Effects 0.000 description 4
- 230000003211 malignant effect Effects 0.000 description 4
- 239000013603 viral vector Substances 0.000 description 4
- 208000023275 Autoimmune disease Diseases 0.000 description 3
- 206010006187 Breast cancer Diseases 0.000 description 3
- 108091026890 Coding region Proteins 0.000 description 3
- 206010009944 Colon cancer Diseases 0.000 description 3
- 208000001333 Colorectal Neoplasms Diseases 0.000 description 3
- 108020004414 DNA Proteins 0.000 description 3
- 238000000134 MTT assay Methods 0.000 description 3
- 231100000002 MTT assay Toxicity 0.000 description 3
- 230000010261 cell growth Effects 0.000 description 3
- 230000003828 downregulation Effects 0.000 description 3
- 238000009472 formulation Methods 0.000 description 3
- 238000001415 gene therapy Methods 0.000 description 3
- 239000002502 liposome Substances 0.000 description 3
- 230000004807 localization Effects 0.000 description 3
- 230000014759 maintenance of location Effects 0.000 description 3
- 239000000203 mixture Substances 0.000 description 3
- 201000006417 multiple sclerosis Diseases 0.000 description 3
- 210000004881 tumor cell Anatomy 0.000 description 3
- 230000035899 viability Effects 0.000 description 3
- 230000009385 viral infection Effects 0.000 description 3
- 108091032973 (ribonucleotides)n+m Proteins 0.000 description 2
- 208000026310 Breast neoplasm Diseases 0.000 description 2
- 201000009030 Carcinoma Diseases 0.000 description 2
- 108010078791 Carrier Proteins Proteins 0.000 description 2
- 102100025064 Cellular tumor antigen p53 Human genes 0.000 description 2
- 102000004127 Cytokines Human genes 0.000 description 2
- 108090000695 Cytokines Proteins 0.000 description 2
- 101710113436 GTPase KRas Proteins 0.000 description 2
- 208000005176 Hepatitis C Diseases 0.000 description 2
- 241000598171 Human adenovirus sp. Species 0.000 description 2
- 206010061218 Inflammation Diseases 0.000 description 2
- 102000004877 Insulin Human genes 0.000 description 2
- 108090001061 Insulin Proteins 0.000 description 2
- 102000015696 Interleukins Human genes 0.000 description 2
- 108010063738 Interleukins Proteins 0.000 description 2
- 206010058467 Lung neoplasm malignant Diseases 0.000 description 2
- 108700019961 Neoplasm Genes Proteins 0.000 description 2
- 102000048850 Neoplasm Genes Human genes 0.000 description 2
- 241000714474 Rous sarcoma virus Species 0.000 description 2
- 208000000102 Squamous Cell Carcinoma of Head and Neck Diseases 0.000 description 2
- XNKLLVCARDGLGL-JGVFFNPUSA-N Stavudine Chemical compound O=C1NC(=O)C(C)=CN1[C@H]1C=C[C@@H](CO)O1 XNKLLVCARDGLGL-JGVFFNPUSA-N 0.000 description 2
- 241000700618 Vaccinia virus Species 0.000 description 2
- WREGKURFCTUGRC-POYBYMJQSA-N Zalcitabine Chemical compound O=C1N=C(N)C=CN1[C@@H]1O[C@H](CO)CC1 WREGKURFCTUGRC-POYBYMJQSA-N 0.000 description 2
- 150000001413 amino acids Chemical group 0.000 description 2
- 230000001093 anti-cancer Effects 0.000 description 2
- 239000013592 cell lysate Substances 0.000 description 2
- 230000003833 cell viability Effects 0.000 description 2
- 230000005757 colony formation Effects 0.000 description 2
- 201000010989 colorectal carcinoma Diseases 0.000 description 2
- WHBIGIKBNXZKFE-UHFFFAOYSA-N delavirdine Chemical compound CC(C)NC1=CC=CN=C1N1CCN(C(=O)C=2NC3=CC=C(NS(C)(=O)=O)C=C3C=2)CC1 WHBIGIKBNXZKFE-UHFFFAOYSA-N 0.000 description 2
- NOPFSRXAKWQILS-UHFFFAOYSA-N docosan-1-ol Chemical compound CCCCCCCCCCCCCCCCCCCCCCO NOPFSRXAKWQILS-UHFFFAOYSA-N 0.000 description 2
- 239000012634 fragment Substances 0.000 description 2
- 208000002672 hepatitis B Diseases 0.000 description 2
- 208000010710 hepatitis C virus infection Diseases 0.000 description 2
- 238000001727 in vivo Methods 0.000 description 2
- 230000004054 inflammatory process Effects 0.000 description 2
- 229940125396 insulin Drugs 0.000 description 2
- 230000007154 intracellular accumulation Effects 0.000 description 2
- 229960001627 lamivudine Drugs 0.000 description 2
- JTEGQNOMFQHVDC-NKWVEPMBSA-N lamivudine Chemical compound O=C1N=C(N)C=CN1[C@H]1O[C@@H](CO)SC1 JTEGQNOMFQHVDC-NKWVEPMBSA-N 0.000 description 2
- 229960004961 mechlorethamine Drugs 0.000 description 2
- HAWPXGHAZFHHAD-UHFFFAOYSA-N mechlorethamine Chemical compound ClCCN(C)CCCl HAWPXGHAZFHHAD-UHFFFAOYSA-N 0.000 description 2
- GLVAUDGFNGKCSF-UHFFFAOYSA-N mercaptopurine Chemical compound S=C1NC=NC2=C1NC=N2 GLVAUDGFNGKCSF-UHFFFAOYSA-N 0.000 description 2
- KKZJGLLVHKMTCM-UHFFFAOYSA-N mitoxantrone Chemical compound O=C1C2=C(O)C=CC(O)=C2C(=O)C2=C1C(NCCNCCO)=CC=C2NCCNCCO KKZJGLLVHKMTCM-UHFFFAOYSA-N 0.000 description 2
- -1 neviripine Chemical compound 0.000 description 2
- ZJAOAACCNHFJAH-UHFFFAOYSA-N phosphonoformic acid Chemical compound OC(=O)P(O)(O)=O ZJAOAACCNHFJAH-UHFFFAOYSA-N 0.000 description 2
- 239000013612 plasmid Substances 0.000 description 2
- 230000003389 potentiating effect Effects 0.000 description 2
- 208000016691 refractory malignant neoplasm Diseases 0.000 description 2
- 230000010076 replication Effects 0.000 description 2
- 239000003419 rna directed dna polymerase inhibitor Substances 0.000 description 2
- 208000000649 small cell carcinoma Diseases 0.000 description 2
- 230000004083 survival effect Effects 0.000 description 2
- 229960002555 zidovudine Drugs 0.000 description 2
- HBOMLICNUCNMMY-XLPZGREQSA-N zidovudine Chemical compound O=C1NC(=O)C(C)=CN1[C@@H]1O[C@H](CO)[C@@H](N=[N+]=[N-])C1 HBOMLICNUCNMMY-XLPZGREQSA-N 0.000 description 2
- FDKXTQMXEQVLRF-ZHACJKMWSA-N (E)-dacarbazine Chemical compound CN(C)\N=N\c1[nH]cnc1C(N)=O FDKXTQMXEQVLRF-ZHACJKMWSA-N 0.000 description 1
- AOJJSUZBOXZQNB-VTZDEGQISA-N 4'-epidoxorubicin Chemical compound O([C@H]1C[C@@](O)(CC=2C(O)=C3C(=O)C=4C=CC=C(C=4C(=O)C3=C(O)C=21)OC)C(=O)CO)[C@H]1C[C@H](N)[C@@H](O)[C@H](C)O1 AOJJSUZBOXZQNB-VTZDEGQISA-N 0.000 description 1
- 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
- 208000030507 AIDS Diseases 0.000 description 1
- 208000024893 Acute lymphoblastic leukemia Diseases 0.000 description 1
- 208000014697 Acute lymphocytic leukaemia Diseases 0.000 description 1
- 241000702423 Adeno-associated virus - 2 Species 0.000 description 1
- 241000710929 Alphavirus Species 0.000 description 1
- 102000000412 Annexin Human genes 0.000 description 1
- 108050008874 Annexin Proteins 0.000 description 1
- IYMAXBFPHPZYIK-BQBZGAKWSA-N Arg-Gly-Asp Chemical group NC(N)=NCCC[C@H](N)C(=O)NCC(=O)N[C@@H](CC(O)=O)C(O)=O IYMAXBFPHPZYIK-BQBZGAKWSA-N 0.000 description 1
- AXRYRYVKAWYZBR-UHFFFAOYSA-N Atazanavir Natural products C=1C=C(C=2N=CC=CC=2)C=CC=1CN(NC(=O)C(NC(=O)OC)C(C)(C)C)CC(O)C(NC(=O)C(NC(=O)OC)C(C)(C)C)CC1=CC=CC=C1 AXRYRYVKAWYZBR-UHFFFAOYSA-N 0.000 description 1
- 108010019625 Atazanavir Sulfate Proteins 0.000 description 1
- 241000201370 Autographa californica nucleopolyhedrovirus Species 0.000 description 1
- 208000010839 B-cell chronic lymphocytic leukemia Diseases 0.000 description 1
- 206010005003 Bladder cancer Diseases 0.000 description 1
- COVZYZSDYWQREU-UHFFFAOYSA-N Busulfan Chemical compound CS(=O)(=O)OCCCCOS(C)(=O)=O COVZYZSDYWQREU-UHFFFAOYSA-N 0.000 description 1
- 108010074051 C-Reactive Protein Proteins 0.000 description 1
- 102100032752 C-reactive protein Human genes 0.000 description 1
- GAGWJHPBXLXJQN-UORFTKCHSA-N Capecitabine Chemical compound C1=C(F)C(NC(=O)OCCCCC)=NC(=O)N1[C@H]1[C@H](O)[C@H](O)[C@@H](C)O1 GAGWJHPBXLXJQN-UORFTKCHSA-N 0.000 description 1
- GAGWJHPBXLXJQN-UHFFFAOYSA-N Capecitabine Natural products C1=C(F)C(NC(=O)OCCCCC)=NC(=O)N1C1C(O)C(O)C(C)O1 GAGWJHPBXLXJQN-UHFFFAOYSA-N 0.000 description 1
- DLGOEMSEDOSKAD-UHFFFAOYSA-N Carmustine Chemical compound ClCCNC(=O)N(N=O)CCCl DLGOEMSEDOSKAD-UHFFFAOYSA-N 0.000 description 1
- 102000014914 Carrier Proteins Human genes 0.000 description 1
- VWFCHDSQECPREK-LURJTMIESA-N Cidofovir Chemical compound NC=1C=CN(C[C@@H](CO)OCP(O)(O)=O)C(=O)N=1 VWFCHDSQECPREK-LURJTMIESA-N 0.000 description 1
- 208000003322 Coinfection Diseases 0.000 description 1
- CMSMOCZEIVJLDB-UHFFFAOYSA-N Cyclophosphamide Chemical compound ClCCN(CCCl)P1(=O)NCCCO1 CMSMOCZEIVJLDB-UHFFFAOYSA-N 0.000 description 1
- UHDGCWIWMRVCDJ-CCXZUQQUSA-N Cytarabine Chemical compound O=C1N=C(N)C=CN1[C@H]1[C@@H](O)[C@H](O)[C@@H](CO)O1 UHDGCWIWMRVCDJ-CCXZUQQUSA-N 0.000 description 1
- 241000701022 Cytomegalovirus Species 0.000 description 1
- 229940123780 DNA topoisomerase I inhibitor Drugs 0.000 description 1
- 229940124087 DNA topoisomerase II inhibitor Drugs 0.000 description 1
- 241000702421 Dependoparvovirus Species 0.000 description 1
- BXZVVICBKDXVGW-NKWVEPMBSA-N Didanosine Chemical compound O1[C@H](CO)CC[C@@H]1N1C(NC=NC2=O)=C2N=C1 BXZVVICBKDXVGW-NKWVEPMBSA-N 0.000 description 1
- XPOQHMRABVBWPR-UHFFFAOYSA-N Efavirenz Natural products O1C(=O)NC2=CC=C(Cl)C=C2C1(C(F)(F)F)C#CC1CC1 XPOQHMRABVBWPR-UHFFFAOYSA-N 0.000 description 1
- XQSPYNMVSIKCOC-NTSWFWBYSA-N Emtricitabine Chemical compound C1=C(F)C(N)=NC(=O)N1[C@H]1O[C@@H](CO)SC1 XQSPYNMVSIKCOC-NTSWFWBYSA-N 0.000 description 1
- 108010032976 Enfuvirtide Proteins 0.000 description 1
- HTIJFSOGRVMCQR-UHFFFAOYSA-N Epirubicin Natural products COc1cccc2C(=O)c3c(O)c4CC(O)(CC(OC5CC(N)C(=O)C(C)O5)c4c(O)c3C(=O)c12)C(=O)CO HTIJFSOGRVMCQR-UHFFFAOYSA-N 0.000 description 1
- 206010015108 Epstein-Barr virus infection Diseases 0.000 description 1
- 241000713730 Equine infectious anemia virus Species 0.000 description 1
- 241000714165 Feline leukemia virus Species 0.000 description 1
- GHASVSINZRGABV-UHFFFAOYSA-N Fluorouracil Chemical compound FC1=CNC(=O)NC1=O GHASVSINZRGABV-UHFFFAOYSA-N 0.000 description 1
- 206010053759 Growth retardation Diseases 0.000 description 1
- 108091005904 Hemoglobin subunit beta Proteins 0.000 description 1
- 206010073069 Hepatic cancer Diseases 0.000 description 1
- 241000700721 Hepatitis B virus Species 0.000 description 1
- 208000009889 Herpes Simplex Diseases 0.000 description 1
- 208000029433 Herpesviridae infectious disease Diseases 0.000 description 1
- 208000017604 Hodgkin disease Diseases 0.000 description 1
- 208000021519 Hodgkin lymphoma Diseases 0.000 description 1
- 208000010747 Hodgkins lymphoma Diseases 0.000 description 1
- 101000733249 Homo sapiens Tumor suppressor ARF Proteins 0.000 description 1
- 241000701044 Human gammaherpesvirus 4 Species 0.000 description 1
- XQFRJNBWHJMXHO-RRKCRQDMSA-N IDUR Chemical compound C1[C@H](O)[C@@H](CO)O[C@H]1N1C(=O)NC(=O)C(I)=C1 XQFRJNBWHJMXHO-RRKCRQDMSA-N 0.000 description 1
- XDXDZDZNSLXDNA-TZNDIEGXSA-N Idarubicin Chemical compound C1[C@H](N)[C@H](O)[C@H](C)O[C@H]1O[C@@H]1C2=C(O)C(C(=O)C3=CC=CC=C3C3=O)=C3C(O)=C2C[C@@](O)(C(C)=O)C1 XDXDZDZNSLXDNA-TZNDIEGXSA-N 0.000 description 1
- XDXDZDZNSLXDNA-UHFFFAOYSA-N Idarubicin Natural products C1C(N)C(O)C(C)OC1OC1C2=C(O)C(C(=O)C3=CC=CC=C3C3=O)=C3C(O)=C2CC(O)(C(C)=O)C1 XDXDZDZNSLXDNA-UHFFFAOYSA-N 0.000 description 1
- 206010061598 Immunodeficiency Diseases 0.000 description 1
- 208000029462 Immunodeficiency disease Diseases 0.000 description 1
- 208000003456 Juvenile Arthritis Diseases 0.000 description 1
- 206010059176 Juvenile idiopathic arthritis Diseases 0.000 description 1
- KJHKTHWMRKYKJE-SUGCFTRWSA-N Kaletra Chemical compound N1([C@@H](C(C)C)C(=O)N[C@H](C[C@H](O)[C@H](CC=2C=CC=CC=2)NC(=O)COC=2C(=CC=CC=2C)C)CC=2C=CC=CC=2)CCCNC1=O KJHKTHWMRKYKJE-SUGCFTRWSA-N 0.000 description 1
- FBOZXECLQNJBKD-ZDUSSCGKSA-N L-methotrexate Chemical compound C=1N=C2N=C(N)N=C(N)C2=NC=1CN(C)C1=CC=C(C(=O)N[C@@H](CCC(O)=O)C(O)=O)C=C1 FBOZXECLQNJBKD-ZDUSSCGKSA-N 0.000 description 1
- 241000713666 Lentivirus Species 0.000 description 1
- GQYIWUVLTXOXAJ-UHFFFAOYSA-N Lomustine Chemical compound ClCCN(N=O)C(=O)NC1CCCCC1 GQYIWUVLTXOXAJ-UHFFFAOYSA-N 0.000 description 1
- 208000031422 Lymphocytic Chronic B-Cell Leukemia Diseases 0.000 description 1
- 206010025323 Lymphomas Diseases 0.000 description 1
- 108010052285 Membrane Proteins Proteins 0.000 description 1
- 102000018697 Membrane Proteins Human genes 0.000 description 1
- 108090000301 Membrane transport proteins Proteins 0.000 description 1
- 102000003939 Membrane transport proteins Human genes 0.000 description 1
- 206010027406 Mesothelioma Diseases 0.000 description 1
- 206010027476 Metastases Diseases 0.000 description 1
- 229940121849 Mitotic inhibitor Drugs 0.000 description 1
- 241000713869 Moloney murine leukemia virus Species 0.000 description 1
- 102000007474 Multiprotein Complexes Human genes 0.000 description 1
- 108010085220 Multiprotein Complexes Proteins 0.000 description 1
- 241001529936 Murinae Species 0.000 description 1
- ZDZOTLJHXYCWBA-VCVYQWHSSA-N N-debenzoyl-N-(tert-butoxycarbonyl)-10-deacetyltaxol Chemical compound O([C@H]1[C@H]2[C@@](C([C@H](O)C3=C(C)[C@@H](OC(=O)[C@H](O)[C@@H](NC(=O)OC(C)(C)C)C=4C=CC=CC=4)C[C@]1(O)C3(C)C)=O)(C)[C@@H](O)C[C@H]1OC[C@]12OC(=O)C)C(=O)C1=CC=CC=C1 ZDZOTLJHXYCWBA-VCVYQWHSSA-N 0.000 description 1
- 206010029260 Neuroblastoma Diseases 0.000 description 1
- 244000061176 Nicotiana tabacum Species 0.000 description 1
- 235000002637 Nicotiana tabacum Nutrition 0.000 description 1
- 208000015914 Non-Hodgkin lymphomas Diseases 0.000 description 1
- 238000000636 Northern blotting Methods 0.000 description 1
- 108091028043 Nucleic acid sequence Proteins 0.000 description 1
- 229940122313 Nucleoside reverse transcriptase inhibitor Drugs 0.000 description 1
- 206010033128 Ovarian cancer Diseases 0.000 description 1
- 206010061535 Ovarian neoplasm Diseases 0.000 description 1
- 238000010222 PCR analysis Methods 0.000 description 1
- 229930012538 Paclitaxel Natural products 0.000 description 1
- 206010061902 Pancreatic neoplasm Diseases 0.000 description 1
- 208000009608 Papillomavirus Infections Diseases 0.000 description 1
- JNTOCHDNEULJHD-UHFFFAOYSA-N Penciclovir Chemical compound N1C(N)=NC(=O)C2=C1N(CCC(CO)CO)C=N2 JNTOCHDNEULJHD-UHFFFAOYSA-N 0.000 description 1
- 102000010292 Peptide Elongation Factor 1 Human genes 0.000 description 1
- 108010077524 Peptide Elongation Factor 1 Proteins 0.000 description 1
- 208000006664 Precursor Cell Lymphoblastic Leukemia-Lymphoma Diseases 0.000 description 1
- 206010060862 Prostate cancer Diseases 0.000 description 1
- 208000000236 Prostatic Neoplasms Diseases 0.000 description 1
- 102000007327 Protamines Human genes 0.000 description 1
- 108010007568 Protamines Proteins 0.000 description 1
- 229940124158 Protease/peptidase inhibitor Drugs 0.000 description 1
- 101000611641 Rattus norvegicus Protein phosphatase 1 regulatory subunit 15A Proteins 0.000 description 1
- 108020004511 Recombinant DNA Proteins 0.000 description 1
- 206010038389 Renal cancer Diseases 0.000 description 1
- 208000006265 Renal cell carcinoma Diseases 0.000 description 1
- 206010039710 Scleroderma Diseases 0.000 description 1
- 241000700584 Simplexvirus Species 0.000 description 1
- 241000710960 Sindbis virus Species 0.000 description 1
- 208000021386 Sjogren Syndrome Diseases 0.000 description 1
- 208000005718 Stomach Neoplasms Diseases 0.000 description 1
- 210000001744 T-lymphocyte Anatomy 0.000 description 1
- 229940123237 Taxane Drugs 0.000 description 1
- BPEGJWRSRHCHSN-UHFFFAOYSA-N Temozolomide Chemical compound O=C1N(C)N=NC2=C(C(N)=O)N=CN21 BPEGJWRSRHCHSN-UHFFFAOYSA-N 0.000 description 1
- 208000024313 Testicular Neoplasms Diseases 0.000 description 1
- 206010057644 Testis cancer Diseases 0.000 description 1
- 239000004098 Tetracycline Substances 0.000 description 1
- SUJUHGSWHZTSEU-UHFFFAOYSA-N Tipranavir Natural products C1C(O)=C(C(CC)C=2C=C(NS(=O)(=O)C=3N=CC(=CC=3)C(F)(F)F)C=CC=2)C(=O)OC1(CCC)CCC1=CC=CC=C1 SUJUHGSWHZTSEU-UHFFFAOYSA-N 0.000 description 1
- 239000000365 Topoisomerase I Inhibitor Substances 0.000 description 1
- 239000000317 Topoisomerase II Inhibitor Substances 0.000 description 1
- 108700009124 Transcription Initiation Site Proteins 0.000 description 1
- 102100033254 Tumor suppressor ARF Human genes 0.000 description 1
- 102000044159 Ubiquitin Human genes 0.000 description 1
- 108090000848 Ubiquitin Proteins 0.000 description 1
- 208000007097 Urinary Bladder Neoplasms Diseases 0.000 description 1
- HDOVUKNUBWVHOX-QMMMGPOBSA-N Valacyclovir Chemical compound N1C(N)=NC(=O)C2=C1N(COCCOC(=O)[C@@H](N)C(C)C)C=N2 HDOVUKNUBWVHOX-QMMMGPOBSA-N 0.000 description 1
- WPVFJKSGQUFQAP-GKAPJAKFSA-N Valcyte Chemical compound N1C(N)=NC(=O)C2=C1N(COC(CO)COC(=O)[C@@H](N)C(C)C)C=N2 WPVFJKSGQUFQAP-GKAPJAKFSA-N 0.000 description 1
- OIRDTQYFTABQOQ-UHTZMRCNSA-N Vidarabine Chemical compound C1=NC=2C(N)=NC=NC=2N1[C@@H]1O[C@H](CO)[C@@H](O)[C@@H]1O OIRDTQYFTABQOQ-UHTZMRCNSA-N 0.000 description 1
- JXLYSJRDGCGARV-WWYNWVTFSA-N Vinblastine Natural products O=C(O[C@H]1[C@](O)(C(=O)OC)[C@@H]2N(C)c3c(cc(c(OC)c3)[C@]3(C(=O)OC)c4[nH]c5c(c4CCN4C[C@](O)(CC)C[C@H](C3)C4)cccc5)[C@@]32[C@H]2[C@@]1(CC)C=CCN2CC3)C JXLYSJRDGCGARV-WWYNWVTFSA-N 0.000 description 1
- 229940122803 Vinca alkaloid Drugs 0.000 description 1
- 208000036142 Viral infection Diseases 0.000 description 1
- HCHKCACWOHOZIP-UHFFFAOYSA-N Zinc Chemical compound [Zn] HCHKCACWOHOZIP-UHFFFAOYSA-N 0.000 description 1
- 229960004748 abacavir Drugs 0.000 description 1
- MCGSCOLBFJQGHM-SCZZXKLOSA-N abacavir Chemical compound C=12N=CN([C@H]3C=C[C@@H](CO)C3)C2=NC(N)=NC=1NC1CC1 MCGSCOLBFJQGHM-SCZZXKLOSA-N 0.000 description 1
- 229960004150 aciclovir Drugs 0.000 description 1
- MKUXAQIIEYXACX-UHFFFAOYSA-N aciclovir Chemical compound N1C(N)=NC(=O)C2=C1N(COCCO)C=N2 MKUXAQIIEYXACX-UHFFFAOYSA-N 0.000 description 1
- 229960001997 adefovir Drugs 0.000 description 1
- WOZSCQDILHKSGG-UHFFFAOYSA-N adefovir depivoxil Chemical compound N1=CN=C2N(CCOCP(=O)(OCOC(=O)C(C)(C)C)OCOC(=O)C(C)(C)C)C=NC2=C1N WOZSCQDILHKSGG-UHFFFAOYSA-N 0.000 description 1
- 208000009956 adenocarcinoma Diseases 0.000 description 1
- 229940100198 alkylating agent Drugs 0.000 description 1
- 239000002168 alkylating agent Substances 0.000 description 1
- 230000004075 alteration Effects 0.000 description 1
- 125000000539 amino acid group Chemical group 0.000 description 1
- 229960001830 amprenavir Drugs 0.000 description 1
- YMARZQAQMVYCKC-OEMFJLHTSA-N amprenavir Chemical compound C([C@@H]([C@H](O)CN(CC(C)C)S(=O)(=O)C=1C=CC(N)=CC=1)NC(=O)O[C@@H]1COCC1)C1=CC=CC=C1 YMARZQAQMVYCKC-OEMFJLHTSA-N 0.000 description 1
- 230000001772 anti-angiogenic effect Effects 0.000 description 1
- 230000003110 anti-inflammatory effect Effects 0.000 description 1
- 230000000340 anti-metabolite Effects 0.000 description 1
- 239000000043 antiallergic agent Substances 0.000 description 1
- 229940100197 antimetabolite Drugs 0.000 description 1
- 239000002256 antimetabolite Substances 0.000 description 1
- 229940041181 antineoplastic drug Drugs 0.000 description 1
- 229940124522 antiretrovirals Drugs 0.000 description 1
- 239000003903 antiretrovirus agent Substances 0.000 description 1
- 238000013459 approach Methods 0.000 description 1
- 206010003246 arthritis Diseases 0.000 description 1
- 210000001130 astrocyte Anatomy 0.000 description 1
- 229960003277 atazanavir Drugs 0.000 description 1
- AXRYRYVKAWYZBR-GASGPIRDSA-N atazanavir Chemical compound C([C@H](NC(=O)[C@@H](NC(=O)OC)C(C)(C)C)[C@@H](O)CN(CC=1C=CC(=CC=1)C=1N=CC=CC=1)NC(=O)[C@@H](NC(=O)OC)C(C)(C)C)C1=CC=CC=C1 AXRYRYVKAWYZBR-GASGPIRDSA-N 0.000 description 1
- 210000000481 breast Anatomy 0.000 description 1
- 201000008275 breast carcinoma Diseases 0.000 description 1
- 229960002092 busulfan Drugs 0.000 description 1
- 230000000981 bystander Effects 0.000 description 1
- 229960004117 capecitabine Drugs 0.000 description 1
- 210000000234 capsid Anatomy 0.000 description 1
- 229960004562 carboplatin Drugs 0.000 description 1
- 190000008236 carboplatin Chemical compound 0.000 description 1
- 229960005243 carmustine Drugs 0.000 description 1
- 230000015556 catabolic process Effects 0.000 description 1
- 210000000170 cell membrane Anatomy 0.000 description 1
- 230000000973 chemotherapeutic effect Effects 0.000 description 1
- 229940044683 chemotherapy drug Drugs 0.000 description 1
- 229960004630 chlorambucil Drugs 0.000 description 1
- JCKYGMPEJWAADB-UHFFFAOYSA-N chlorambucil Chemical compound OC(=O)CCCC1=CC=C(N(CCCl)CCCl)C=C1 JCKYGMPEJWAADB-UHFFFAOYSA-N 0.000 description 1
- 208000019069 chronic childhood arthritis Diseases 0.000 description 1
- 208000032852 chronic lymphocytic leukemia Diseases 0.000 description 1
- 229960000724 cidofovir Drugs 0.000 description 1
- 229960004316 cisplatin Drugs 0.000 description 1
- DQLATGHUWYMOKM-UHFFFAOYSA-L cisplatin Chemical compound N[Pt](N)(Cl)Cl DQLATGHUWYMOKM-UHFFFAOYSA-L 0.000 description 1
- 230000007012 clinical effect Effects 0.000 description 1
- 238000010367 cloning Methods 0.000 description 1
- 239000002299 complementary DNA Substances 0.000 description 1
- 229960004397 cyclophosphamide Drugs 0.000 description 1
- 229960000684 cytarabine Drugs 0.000 description 1
- 231100000433 cytotoxic Toxicity 0.000 description 1
- 230000001472 cytotoxic effect Effects 0.000 description 1
- 229960003901 dacarbazine Drugs 0.000 description 1
- 229960000975 daunorubicin 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
- 230000034994 death Effects 0.000 description 1
- 230000002950 deficient Effects 0.000 description 1
- 238000006731 degradation reaction Methods 0.000 description 1
- 229960005319 delavirdine Drugs 0.000 description 1
- 230000001419 dependent effect Effects 0.000 description 1
- UREBDLICKHMUKA-CXSFZGCWSA-N dexamethasone Chemical compound C1CC2=CC(=O)C=C[C@]2(C)[C@]2(F)[C@@H]1[C@@H]1C[C@@H](C)[C@@](C(=O)CO)(O)[C@@]1(C)C[C@@H]2O UREBDLICKHMUKA-CXSFZGCWSA-N 0.000 description 1
- 229960003957 dexamethasone Drugs 0.000 description 1
- 206010012601 diabetes mellitus Diseases 0.000 description 1
- 229960002656 didanosine Drugs 0.000 description 1
- 238000002224 dissection Methods 0.000 description 1
- 229960003668 docetaxel Drugs 0.000 description 1
- 229960000735 docosanol Drugs 0.000 description 1
- 231100000673 dose–response relationship Toxicity 0.000 description 1
- 230000005014 ectopic expression Effects 0.000 description 1
- 229960003804 efavirenz Drugs 0.000 description 1
- XPOQHMRABVBWPR-ZDUSSCGKSA-N efavirenz Chemical compound C([C@]1(C2=CC(Cl)=CC=C2NC(=O)O1)C(F)(F)F)#CC1CC1 XPOQHMRABVBWPR-ZDUSSCGKSA-N 0.000 description 1
- 229960003586 elvitegravir Drugs 0.000 description 1
- JUZYLCPPVHEVSV-LJQANCHMSA-N elvitegravir Chemical compound COC1=CC=2N([C@H](CO)C(C)C)C=C(C(O)=O)C(=O)C=2C=C1CC1=CC=CC(Cl)=C1F JUZYLCPPVHEVSV-LJQANCHMSA-N 0.000 description 1
- 229960000366 emtricitabine Drugs 0.000 description 1
- 210000002889 endothelial cell Anatomy 0.000 description 1
- 230000003511 endothelial effect Effects 0.000 description 1
- PEASPLKKXBYDKL-FXEVSJAOSA-N enfuvirtide Chemical compound C([C@@H](C(=O)N[C@@H](CO)C(=O)N[C@@H](CC(C)C)C(=O)N[C@@H]([C@@H](C)CC)C(=O)N[C@@H](CCC(O)=O)C(=O)N[C@@H](CCC(O)=O)C(=O)N[C@@H](CO)C(=O)N[C@@H](CCC(N)=O)C(=O)N[C@@H](CC(N)=O)C(=O)N[C@@H](CCC(N)=O)C(=O)N[C@@H](CCC(N)=O)C(=O)N[C@@H](CCC(O)=O)C(=O)N[C@@H](CCCCN)C(=O)N[C@@H](CC(N)=O)C(=O)N[C@@H](CCC(O)=O)C(=O)N[C@@H](CCC(N)=O)C(=O)N[C@@H](CCC(O)=O)C(=O)N[C@@H](CC(C)C)C(=O)N[C@@H](CC(C)C)C(=O)N[C@@H](CCC(O)=O)C(=O)N[C@@H](CC(C)C)C(=O)N[C@@H](CC(O)=O)C(=O)N[C@@H](CCCCN)C(=O)N[C@@H](CC=1C2=CC=CC=C2NC=1)C(=O)N[C@@H](C)C(=O)N[C@@H](CO)C(=O)N[C@@H](CC(C)C)C(=O)N[C@@H](CC=1C2=CC=CC=C2NC=1)C(=O)N[C@@H](CC(N)=O)C(=O)N[C@@H](CC=1C2=CC=CC=C2NC=1)C(=O)N[C@@H](CC=1C=CC=CC=1)C(N)=O)NC(=O)[C@@H](NC(=O)[C@H](CC(C)C)NC(=O)[C@H](CO)NC(=O)[C@@H](NC(=O)[C@H](CC=1C=CC(O)=CC=1)NC(C)=O)[C@@H](C)O)[C@@H](C)CC)C1=CN=CN1 PEASPLKKXBYDKL-FXEVSJAOSA-N 0.000 description 1
- 229960002062 enfuvirtide Drugs 0.000 description 1
- 229960001904 epirubicin Drugs 0.000 description 1
- 210000002919 epithelial cell Anatomy 0.000 description 1
- 210000003743 erythrocyte Anatomy 0.000 description 1
- VJJPUSNTGOMMGY-MRVIYFEKSA-N etoposide Chemical compound COC1=C(O)C(OC)=CC([C@@H]2C3=CC=4OCOC=4C=C3[C@@H](O[C@H]3[C@@H]([C@@H](O)[C@@H]4O[C@H](C)OC[C@H]4O3)O)[C@@H]3[C@@H]2C(OC3)=O)=C1 VJJPUSNTGOMMGY-MRVIYFEKSA-N 0.000 description 1
- 229960005420 etoposide Drugs 0.000 description 1
- 229960002049 etravirine Drugs 0.000 description 1
- PYGWGZALEOIKDF-UHFFFAOYSA-N etravirine Chemical compound CC1=CC(C#N)=CC(C)=C1OC1=NC(NC=2C=CC(=CC=2)C#N)=NC(N)=C1Br PYGWGZALEOIKDF-UHFFFAOYSA-N 0.000 description 1
- 239000000835 fiber Substances 0.000 description 1
- 210000002950 fibroblast Anatomy 0.000 description 1
- 229960000390 fludarabine Drugs 0.000 description 1
- GIUYCYHIANZCFB-FJFJXFQQSA-N fludarabine phosphate Chemical compound C1=NC=2C(N)=NC(F)=NC=2N1[C@@H]1O[C@H](COP(O)(O)=O)[C@@H](O)[C@@H]1O GIUYCYHIANZCFB-FJFJXFQQSA-N 0.000 description 1
- 229960002949 fluorouracil Drugs 0.000 description 1
- 229960001447 fomivirsen Drugs 0.000 description 1
- XCWFZHPEARLXJI-UHFFFAOYSA-N fomivirsen Chemical compound C1C(N2C3=C(C(NC(N)=N3)=O)N=C2)OC(CO)C1OP(O)(=S)OCC1OC(N(C)C(=O)\N=C(\N)C=C)CC1OP(O)(=S)OCC1OC(N2C3=C(C(NC(N)=N3)=O)N=C2)CC1OP(O)(=S)OCC1OC(N2C(NC(=O)C(C)=C2)=O)CC1OP(O)(=S)OCC1OC(N2C(NC(=O)C(C)=C2)=O)CC1OP(O)(=S)OCC1OC(N2C(NC(=O)C(C)=C2)=O)CC1OP(O)(=S)OCC1OC(N2C3=C(C(NC(N)=N3)=O)N=C2)CC1OP(O)(=S)OCC1OC(N2C(N=C(N)C=C2)=O)CC1OP(O)(=S)OCC(C(C1)OP(S)(=O)OCC2C(CC(O2)N2C(N=C(N)C=C2)=O)OP(O)(=S)OCC2C(CC(O2)N2C(NC(=O)C(C)=C2)=O)OP(O)(=S)OCC2C(CC(O2)N2C(NC(=O)C(C)=C2)=O)OP(O)(=S)OCC2C(CC(O2)N2C(N=C(N)C=C2)=O)OP(O)(=S)OCC2C(CC(O2)N2C(NC(=O)C(C)=C2)=O)OP(O)(=S)OCC2C(CC(O2)N2C(NC(=O)C(C)=C2)=O)OP(O)(=S)OCC2C(CC(O2)N2C(N=C(N)C=C2)=O)OP(O)(=S)OCC2C(CC(O2)N2C(NC(=O)C(C)=C2)=O)OP(O)(=S)OCC2C(CC(O2)N2C(NC(=O)C(C)=C2)=O)OP(O)(=S)OCC2C(CC(O2)N2C3=C(C(NC(N)=N3)=O)N=C2)OP(O)(=S)OCC2C(CC(O2)N2C(N=C(N)C=C2)=O)OP(O)(=S)OCC2C(CC(O2)N2C3=C(C(NC(N)=N3)=O)N=C2)O)OC1N1C=C(C)C(=O)NC1=O XCWFZHPEARLXJI-UHFFFAOYSA-N 0.000 description 1
- 229960003142 fosamprenavir Drugs 0.000 description 1
- MLBVMOWEQCZNCC-OEMFJLHTSA-N fosamprenavir Chemical compound C([C@@H]([C@H](OP(O)(O)=O)CN(CC(C)C)S(=O)(=O)C=1C=CC(N)=CC=1)NC(=O)O[C@@H]1COCC1)C1=CC=CC=C1 MLBVMOWEQCZNCC-OEMFJLHTSA-N 0.000 description 1
- 229960005102 foscarnet Drugs 0.000 description 1
- 229940125777 fusion inhibitor Drugs 0.000 description 1
- 108020001507 fusion proteins Proteins 0.000 description 1
- 102000037865 fusion proteins Human genes 0.000 description 1
- 229960002963 ganciclovir Drugs 0.000 description 1
- IRSCQMHQWWYFCW-UHFFFAOYSA-N ganciclovir Chemical compound O=C1NC(N)=NC2=C1N=CN2COC(CO)CO IRSCQMHQWWYFCW-UHFFFAOYSA-N 0.000 description 1
- 206010017758 gastric cancer Diseases 0.000 description 1
- SDUQYLNIPVEERB-QPPQHZFASA-N gemcitabine Chemical compound O=C1N=C(N)C=CN1[C@H]1C(F)(F)[C@H](O)[C@@H](CO)O1 SDUQYLNIPVEERB-QPPQHZFASA-N 0.000 description 1
- 229960005277 gemcitabine Drugs 0.000 description 1
- 230000009395 genetic defect Effects 0.000 description 1
- 208000005017 glioblastoma Diseases 0.000 description 1
- 230000012010 growth Effects 0.000 description 1
- 239000003102 growth factor Substances 0.000 description 1
- 230000009931 harmful effect Effects 0.000 description 1
- 201000000459 head and neck squamous cell carcinoma Diseases 0.000 description 1
- 230000036541 health Effects 0.000 description 1
- 231100000086 high toxicity Toxicity 0.000 description 1
- 230000006801 homologous recombination Effects 0.000 description 1
- 238000002744 homologous recombination Methods 0.000 description 1
- 210000005260 human cell Anatomy 0.000 description 1
- 208000021145 human papilloma virus infection Diseases 0.000 description 1
- 229960000908 idarubicin Drugs 0.000 description 1
- 229960004716 idoxuridine Drugs 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
- 238000003384 imaging method Methods 0.000 description 1
- 230000007813 immunodeficiency Effects 0.000 description 1
- 230000002163 immunogen Effects 0.000 description 1
- 238000003364 immunohistochemistry Methods 0.000 description 1
- 230000003308 immunostimulating effect Effects 0.000 description 1
- 238000000338 in vitro Methods 0.000 description 1
- 230000002458 infectious effect Effects 0.000 description 1
- 239000003112 inhibitor Substances 0.000 description 1
- 230000005764 inhibitory process Effects 0.000 description 1
- 238000013383 initial experiment Methods 0.000 description 1
- 238000002347 injection Methods 0.000 description 1
- 239000007924 injection Substances 0.000 description 1
- 229940124524 integrase inhibitor Drugs 0.000 description 1
- 239000002850 integrase inhibitor Substances 0.000 description 1
- 238000001361 intraarterial administration Methods 0.000 description 1
- 230000003834 intracellular effect Effects 0.000 description 1
- 238000007918 intramuscular administration Methods 0.000 description 1
- 238000007913 intrathecal administration Methods 0.000 description 1
- 230000002601 intratumoral effect Effects 0.000 description 1
- 238000001990 intravenous administration Methods 0.000 description 1
- 229960004768 irinotecan Drugs 0.000 description 1
- UWKQSNNFCGGAFS-XIFFEERXSA-N irinotecan Chemical compound C1=C2C(CC)=C3CN(C(C4=C([C@@](C(=O)OC4)(O)CC)C=4)=O)C=4C3=NC2=CC=C1OC(=O)N(CC1)CCC1N1CCCCC1 UWKQSNNFCGGAFS-XIFFEERXSA-N 0.000 description 1
- 201000002215 juvenile rheumatoid arthritis Diseases 0.000 description 1
- 230000003902 lesion Effects 0.000 description 1
- 150000002632 lipids Chemical class 0.000 description 1
- 230000033001 locomotion Effects 0.000 description 1
- 229960002247 lomustine Drugs 0.000 description 1
- 229960004525 lopinavir Drugs 0.000 description 1
- 201000005202 lung cancer Diseases 0.000 description 1
- 201000005296 lung carcinoma Diseases 0.000 description 1
- 208000020816 lung neoplasm Diseases 0.000 description 1
- 206010025135 lupus erythematosus Diseases 0.000 description 1
- 208000015486 malignant pancreatic neoplasm Diseases 0.000 description 1
- 230000007246 mechanism Effects 0.000 description 1
- 230000001404 mediated effect Effects 0.000 description 1
- 210000002752 melanocyte Anatomy 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
- 229960001428 mercaptopurine Drugs 0.000 description 1
- 108020004999 messenger RNA Proteins 0.000 description 1
- 230000004060 metabolic process Effects 0.000 description 1
- 230000009401 metastasis Effects 0.000 description 1
- 229960000485 methotrexate Drugs 0.000 description 1
- 239000011859 microparticle Substances 0.000 description 1
- 229960001156 mitoxantrone Drugs 0.000 description 1
- 239000002105 nanoparticle Substances 0.000 description 1
- 230000007830 nerve conduction Effects 0.000 description 1
- OSTGTTZJOCZWJG-UHFFFAOYSA-N nitrosourea Chemical compound NC(=O)N=NO OSTGTTZJOCZWJG-UHFFFAOYSA-N 0.000 description 1
- 229940042402 non-nucleoside reverse transcriptase inhibitor Drugs 0.000 description 1
- 239000002726 nonnucleoside reverse transcriptase inhibitor Substances 0.000 description 1
- 230000000414 obstructive effect Effects 0.000 description 1
- 238000011275 oncology therapy Methods 0.000 description 1
- 201000002740 oral squamous cell carcinoma Diseases 0.000 description 1
- 229960001592 paclitaxel Drugs 0.000 description 1
- 229940124583 pain medication Drugs 0.000 description 1
- 201000002528 pancreatic cancer Diseases 0.000 description 1
- 208000008443 pancreatic carcinoma Diseases 0.000 description 1
- 239000002245 particle Substances 0.000 description 1
- 239000008188 pellet Substances 0.000 description 1
- 229960005079 pemetrexed Drugs 0.000 description 1
- QOFFJEBXNKRSPX-ZDUSSCGKSA-N pemetrexed Chemical compound C1=N[C]2NC(N)=NC(=O)C2=C1CCC1=CC=C(C(=O)N[C@@H](CCC(O)=O)C(O)=O)C=C1 QOFFJEBXNKRSPX-ZDUSSCGKSA-N 0.000 description 1
- 229960001179 penciclovir Drugs 0.000 description 1
- 239000000137 peptide hydrolase inhibitor Substances 0.000 description 1
- 238000009520 phase I clinical trial Methods 0.000 description 1
- 230000008488 polyadenylation Effects 0.000 description 1
- 208000005987 polymyositis Diseases 0.000 description 1
- 108090000765 processed proteins & peptides Proteins 0.000 description 1
- 238000012545 processing Methods 0.000 description 1
- 238000004393 prognosis Methods 0.000 description 1
- 230000001737 promoting effect Effects 0.000 description 1
- 230000000644 propagated effect Effects 0.000 description 1
- 229940048914 protamine Drugs 0.000 description 1
- 238000003259 recombinant expression Methods 0.000 description 1
- 230000006798 recombination Effects 0.000 description 1
- 238000005215 recombination Methods 0.000 description 1
- 201000010174 renal carcinoma Diseases 0.000 description 1
- 238000011160 research Methods 0.000 description 1
- 230000004043 responsiveness Effects 0.000 description 1
- 239000012313 reversal agent Substances 0.000 description 1
- 201000009410 rhabdomyosarcoma Diseases 0.000 description 1
- 206010039073 rheumatoid arthritis Diseases 0.000 description 1
- 210000003705 ribosome Anatomy 0.000 description 1
- 238000004062 sedimentation Methods 0.000 description 1
- 239000000243 solution Substances 0.000 description 1
- 206010041823 squamous cell carcinoma Diseases 0.000 description 1
- 239000003381 stabilizer Substances 0.000 description 1
- 238000010186 staining Methods 0.000 description 1
- 238000010561 standard procedure Methods 0.000 description 1
- 229960001203 stavudine Drugs 0.000 description 1
- 201000011549 stomach cancer Diseases 0.000 description 1
- 229960001052 streptozocin Drugs 0.000 description 1
- ZSJLQEPLLKMAKR-GKHCUFPYSA-N streptozocin Chemical compound O=NN(C)C(=O)N[C@H]1[C@@H](O)O[C@H](CO)[C@@H](O)[C@@H]1O ZSJLQEPLLKMAKR-GKHCUFPYSA-N 0.000 description 1
- 238000007920 subcutaneous administration Methods 0.000 description 1
- 239000000758 substrate Substances 0.000 description 1
- 239000000725 suspension Substances 0.000 description 1
- 208000024891 symptom Diseases 0.000 description 1
- 238000003786 synthesis reaction Methods 0.000 description 1
- DKPFODGZWDEEBT-QFIAKTPHSA-N taxane Chemical class C([C@]1(C)CCC[C@@H](C)[C@H]1C1)C[C@H]2[C@H](C)CC[C@@H]1C2(C)C DKPFODGZWDEEBT-QFIAKTPHSA-N 0.000 description 1
- RCINICONZNJXQF-MZXODVADSA-N taxol Chemical compound O([C@@H]1[C@@]2(C[C@@H](C(C)=C(C2(C)C)[C@H](C([C@]2(C)[C@@H](O)C[C@H]3OC[C@]3([C@H]21)OC(C)=O)=O)OC(=O)C)OC(=O)[C@H](O)[C@@H](NC(=O)C=1C=CC=CC=1)C=1C=CC=CC=1)O)C(=O)C1=CC=CC=C1 RCINICONZNJXQF-MZXODVADSA-N 0.000 description 1
- 229960004964 temozolomide Drugs 0.000 description 1
- NRUKOCRGYNPUPR-QBPJDGROSA-N teniposide 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](OC[C@H]4O3)C=3SC=CC=3)O)[C@@H]3[C@@H]2C(OC3)=O)=C1 NRUKOCRGYNPUPR-QBPJDGROSA-N 0.000 description 1
- 229960001278 teniposide Drugs 0.000 description 1
- 201000003120 testicular cancer Diseases 0.000 description 1
- 229930101283 tetracycline Natural products 0.000 description 1
- 229960002180 tetracycline Drugs 0.000 description 1
- 235000019364 tetracycline Nutrition 0.000 description 1
- 150000003522 tetracyclines Chemical class 0.000 description 1
- 229940124597 therapeutic agent Drugs 0.000 description 1
- 230000001225 therapeutic effect Effects 0.000 description 1
- 229960000838 tipranavir Drugs 0.000 description 1
- SUJUHGSWHZTSEU-FYBSXPHGSA-N tipranavir Chemical compound C([C@@]1(CCC)OC(=O)C([C@H](CC)C=2C=C(NS(=O)(=O)C=3N=CC(=CC=3)C(F)(F)F)C=CC=2)=C(O)C1)CC1=CC=CC=C1 SUJUHGSWHZTSEU-FYBSXPHGSA-N 0.000 description 1
- 229960000303 topotecan Drugs 0.000 description 1
- UCFGDBYHRUNTLO-QHCPKHFHSA-N topotecan Chemical compound C1=C(O)C(CN(C)C)=C2C=C(CN3C4=CC5=C(C3=O)COC(=O)[C@]5(O)CC)C4=NC2=C1 UCFGDBYHRUNTLO-QHCPKHFHSA-N 0.000 description 1
- 231100000419 toxicity Toxicity 0.000 description 1
- 230000001988 toxicity Effects 0.000 description 1
- 230000035897 transcription Effects 0.000 description 1
- 238000013518 transcription Methods 0.000 description 1
- 238000001890 transfection Methods 0.000 description 1
- 229960003962 trifluridine Drugs 0.000 description 1
- VSQQQLOSPVPRAZ-RRKCRQDMSA-N trifluridine Chemical compound C1[C@H](O)[C@@H](CO)O[C@H]1N1C(=O)NC(=O)C(C(F)(F)F)=C1 VSQQQLOSPVPRAZ-RRKCRQDMSA-N 0.000 description 1
- 229960000832 tromantadine Drugs 0.000 description 1
- UXQDWARBDDDTKG-UHFFFAOYSA-N tromantadine Chemical compound C1C(C2)CC3CC2CC1(NC(=O)COCCN(C)C)C3 UXQDWARBDDDTKG-UHFFFAOYSA-N 0.000 description 1
- 230000004614 tumor growth Effects 0.000 description 1
- 241000701447 unidentified baculovirus Species 0.000 description 1
- 241001430294 unidentified retrovirus Species 0.000 description 1
- 201000005112 urinary bladder cancer Diseases 0.000 description 1
- 229940093257 valacyclovir Drugs 0.000 description 1
- 229960002149 valganciclovir Drugs 0.000 description 1
- 229960003636 vidarabine Drugs 0.000 description 1
- 229960003048 vinblastine Drugs 0.000 description 1
- JXLYSJRDGCGARV-XQKSVPLYSA-N vincaleukoblastine Chemical compound C([C@@H](C[C@]1(C(=O)OC)C=2C(=CC3=C([C@]45[C@H]([C@@]([C@H](OC(C)=O)[C@]6(CC)C=CCN([C@H]56)CC4)(O)C(=O)OC)N3C)C=2)OC)C[C@@](C2)(O)CC)N2CCC2=C1NC1=CC=CC=C21 JXLYSJRDGCGARV-XQKSVPLYSA-N 0.000 description 1
- 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
- GBABOYUKABKIAF-GHYRFKGUSA-N vinorelbine Chemical compound 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 GBABOYUKABKIAF-GHYRFKGUSA-N 0.000 description 1
- 229960002066 vinorelbine Drugs 0.000 description 1
- 230000003612 virological effect Effects 0.000 description 1
- 229960000523 zalcitabine Drugs 0.000 description 1
- 239000011701 zinc Substances 0.000 description 1
- 229910052725 zinc Inorganic materials 0.000 description 1
Classifications
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K48/00—Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
- A61K48/005—Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy characterised by an aspect of the 'active' part of the composition delivered, i.e. the nucleic acid delivered
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K38/00—Medicinal preparations containing peptides
- A61K38/16—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
- A61K38/17—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
- A61K38/19—Cytokines; Lymphokines; Interferons
- A61K38/20—Interleukins [IL]
- A61K38/2066—IL-10
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N15/00—Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
- C12N15/09—Recombinant DNA-technology
- C12N15/63—Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
- C12N15/79—Vectors or expression systems specially adapted for eukaryotic hosts
- C12N15/85—Vectors or expression systems specially adapted for eukaryotic hosts for animal cells
- C12N15/86—Viral vectors
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2710/00—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA dsDNA viruses
- C12N2710/00011—Details
- C12N2710/10011—Adenoviridae
- C12N2710/10311—Mastadenovirus, e.g. human or simian adenoviruses
- C12N2710/10341—Use of virus, viral particle or viral elements as a vector
- C12N2710/10343—Use of virus, viral particle or viral elements as a vector viral genome or elements thereof as genetic vector
Definitions
- the present invention relates to the use of melanoma differentiation associated gene-7, its encoded protein, and related molecules ( Collectively, "Mda-7 Molecules”) for improving the response of a subject to chemotherapy and the particular benefit of Mda-7 treatment in a subject resistant to chemotherapy. It is based, at least in part, on the discovery that mda-7 expression increases the response of hitherto resistant cells to chemotherapeutic agent, as well as on the discovery that mda-7 expression is itself enhanced in the context of over- expression of the multidrug resistance gene (mdr-l).
- mdr-l multidrug resistance gene
- mda-7 Melanoma differentiation associated gene-7 (mda-7) (Jiang et al., 1995, Oncogene 11 :2477-2486) is a secreted cytokine belonging to the interleukin (IL)-IO family designated as IL-24 (Pestka et al., 2004, Annu Rev Immunol 22:929- 979).
- IL-24 interleukin-IO family designated as IL-24
- mda-7/IL-24 does not induce toxicity in normal endothelial and epithelial cells, fibroblasts, melanocytes and astrocytes (Ekmekcioglu et al., 2001, Int J Cancer 94(l):54-59; Ellerhorst et al., 2002, J CHn Oncol 20(4):1069-1074; Fisher, 2005, Cancer Res 65(22):10128-10138; Fisher et al., 2003, Cancer Biol Ther 2 (4 Suppl l):S23-37; Gupta et al., 2006, Pharmacol Ther 111 : 596-628; Huang et al., 2001, Oncogene 20(48):7051-7063; Jiang et al., 1996, Proc Natl Acad Sci U S A
- mda-7/IL-24 possesses potent anti-angiogenic, immunostimulatory and bystander activities (Fisher, 2005, Cancer Res 65(22):10128-10138; Fisher et al., 2003, Cancer Biol Ther 2 (4 Suppl l):S23-37; Gupta et al., 2006, Pharmacol Ther 111 : 596-628; Lebedeva et al., 2005, MoI Ther 11(1):4-18).
- the sum of these attributes makes mda-7/IL-24 a significant candidate for cancer gene therapy (Fisher, 2005, Cancer Res 65(22): 10128- 10138).
- Ad.mda-7 has been successfully used for a Phase I clinical trial for advanced carcinomas and melanomas and has shown promising results in tumor growth inhibition and induction of cancer apoptosis (Cunningham et al., 2005, MoI Ther 11(1):149-159 ; Fisher, 2005, Cancer Res 65(22):10128-10138; Fisher et al., 2003, Cancer Biol Ther 2 (4 Suppl l):S23-37, ; Lebedeva et al., 2005, MoI Ther
- Ad.mda-7 induces growth suppression and apoptosis in histologically diverse cancer cells containing single or multiple genetic defects, including alterations in p53, pl6/INK4a and/or Rb (Emdad et al., 2006, J Cell Physiol 208(2):298-306; Huang et al., 2001, Oncogene 20(48):7051-7063; Jiang et al., 1996, Proc Natl Acad Sci U S A 93(17):9160-9165; Lebedeva et al., 2002, Oncogene 21(5):708-718; Mhashilkar et al., 2001, MoI Med 7(4):271-282; Saeki et al., 2000, Gene Ther 7(23):2051-2057; Su et al., 2001, Proc Natl Acad
- Ad.mda-7 is equally effective in inducing apoptosis in breast and lung carcinoma and melanoma cells containing wtp53, mutp53 or which are null for p53 expression (Lebedeva et al., 2002, Lebedeva et al., 2002, Oncogene 21(5):708-718; Madireddi et al., 2000, Adv Exp Med Biol 465:239-261; Saeki et al., 2000, Gene Ther 7(23):2051-2057; Saeki et al., 2002, Oncogene 21(29):4558-4566; Su et al., 1998, Proc Natl Acad Sci U S A 95(24): 14400-14405).
- a fragment of MDA-7, retaining amino acid residues 104 to 206 of the wild-type sequence, has been found to retain the cancer-specific growth suppressive and apoptosis-inducing properties of the full-length protein (Gupta et al., 2006, Cancer Res. 66(16 ⁇ :8182-8191).
- the mdr-1 gene encodes a multi-drug resistant transporter protein, MDR-1/P-glycoprotein (also referred to herein as "P-gp"), a 170 kDa glycosylated membrane protein which is a member of the ATP-binding cassette superfamily of membrane transporters (Meijer et al., 1999, J. Clin. Pathol. 52:450-454; Sakeada et al., 2002, Biol. Pharma Cull. 25:1391-1400). MDR-1/P-glycoprotein acts as a molecular pump which can expel chemotherapeutic agents from tumor cells.
- MDR-1/P-glycoprotein acts as a molecular pump which can expel chemotherapeutic agents from tumor cells.
- MDR-1 In addition to its obstructive role in cancer therapy, over-expression of mdr-1 gene and protein also interferes with chemotherapy of diseases other than cancer.
- MDR-1 is believed to function in the transport and metabolism of antiretroviral agents, and variants of MDR-1 have been associated with different levels of therapeutic response in human immunodeficiency virus infection (Saitoh et al., 2005, AIDS Jj9 ⁇ 4 ⁇ :371-380).
- Hepatitis B virus has been shown to transactivate mdr-1 gene expression (Doong et al., 1998, J. Hepatol. 29(6): 872-878).
- Mdr- 1 over-expression has been associated with various autoimmune diseases (Levy et al., 2002, Br. J. Haematol. 118(3):836-838: Tsujimura et al., 2004, Genes Cells 9(121:1265-1273).
- the present invention relates to a method for improving the therapeutic response of a subject to chemotherapy, where the subject had previously been resistant to chemotherapy and/or where the mdr-1 gene and/or MDR-I protein are shown to be over-expressed, comprising administering, to the subject, an effective amount of a Mda-7 molecule (mda-7 nucleic acid, MDA-7 protein, M4 nucleic acid or M4 protein, defined below) in conjunction with one or more chemotherapeutic agent. It is based, at least in part, on the discovery that expression of mda-7 in drug-resistant cancer cells resulted in increased accumulation of, and sensitivity to, the drug.
- the present invention provides for a method of treating a subject suffering from a cancer found to be resistant to chemotherapy and/or where the mdr-1 gene and/or MDR-I protein are shown to be over-expressed comprising administering, to the subject, an effective amount of a Mda-7 molecule.
- This aspect is based, at least in part, on the discovery that the anticancer effect of the melanoma differentiation associated gene -7 (mda-7) was increased in the context of over-expression of the multidrug resistance (MDR-I) gene.
- FIGURE IA-C A. Western blot analysis of endogenous P-gp levels in SW620/WT and SW620/DOX cells.
- B Effect of different doses of doxorubicin (Dox) on cell growth in SW620/WT and SW620/DOX cells. Graphs show the average ⁇ SD of results of 3 independent experiments.
- C Effect of Dox on apoptosis induction in SW620/WT and SW620/DOX cells by Annexin V binding assay. Results are the average ⁇ SD from replicate studies.
- Dox doxorubicin
- FIGURE 2A-C A. SW620/WT (also referred to herein as "SW620") and SW620/DOX cells were infected with either Ad.vec or Ad.mda7 (25 pfu/cell) and then treated with the indicated concentrations of Dox for 48 h. Percentage of apoptotic cells were determined by Annexin V binding assay and flow cytometry.
- FIGURE 3 A-B Effect of Ad.mda7 on the efflux of Dox from
- SW620/WT and SW620/DOX cells were SW620/WT and SW620/DOX cells.
- FIGURE 4A-C A. SW620/WT and SW620/DOX cells were infected with either Ad.vec or Ad.mda7 with the indicated MOI and cell viability was determined by MTT assay 5 days after infection. Values are the average ⁇ SD of three independent experiments.
- FIGURE 5A-D A.
- SW620/WT cells were transiently transfected with either empty vector (pCDNA3.1) or MDR and then infected with either Ad.vec or Ad.mda7 at the indicated MOI. Apoptosis induction was evaluated by Annexin V binding assay 48 h after infection.
- B Effect of MDR over-expression on MDA-7/IL- 24 protein expression. Western blot Analysis was performed with the cell lysates harvested at 24 h after infection.
- C. Colony formation in soft agar as a function of Ad.vec or Ad.mda-7 in the presence or absence of MDR. Data represents mean ⁇ SD of triplicate plates in three independent experiments.
- D Graph shows cloning efficiency.
- FIGURE 6 Effect of down-regulation of MDR (by siRNA) on
- FIGURE 7A-B K-ras localization in (A) SW620/WT and (B) SW620/DOX cells. Confocal images of SW620/WT and SW620/DOX cells immunofluorescently stained with anti-k-Ras antibody.
- Mda-7 molecules (i) Mda-7 molecules; (ii) methods of administering Mda-7 molecules;
- nucleic acid is referred to by lower case letters (e.g., mda-7 or mdr-1)
- a protein is referred to by capital letters (e.g., MDA-7 or MDR-I) and both are generically referred to by an initial capital letter followed by lower case (e.g. Mda-7 or Mdr-1).
- a "mda-7 nucleic acid” as defined herein is a nucleic acid which may be:
- nucleic acid having a nucleotide sequence comprising SEQ ID NO:1 (nucleotides 275 to 895 of GenBank Accession No. U 16261; Jiang et al., 1995, Oncogene 11 -.2477-2486), which encodes wild-type human MDA-7 protein;
- nucleic acid which encodes an MDA-7 protein having an amino acid sequence comprising SEQ ID NO:2 (GenBank Accession No. Q13007; Jiang et al., 1995, Oncogene 11 :2477-2486; the human wild-type MDA-7 amino acid sequence); (iii) a nucleic acid having a nucleotide sequence comprising SEQ ID NO:3, which encodes a version of wild-type human MDA-7 protein lacking the signal sequence;
- nucleic acid which encodes a protein (lacking a signal peptide) having an amino acid sequence comprising SEQ ID NO:4 (the human wild-type MDA-7 amino acid sequence lacking the signal peptide);
- nucleic acid having a nucleotide sequence which is at least 90 percent or at least 95 percent homologous to SEQ ID NO: 1 ;
- a nucleic acid which encodes a protein having an amino acid sequence which is at least 90 percent or at least 95 percent homologous to SEQ ID NO:4 where homology may be determined, for example, using standard software such as BLAST or FASTA).
- a MDA-7 protein as defined herein is a protein which may be: (i) a protein having an amino acid sequence comprising SEQ ID NO:2 (the human wild-type MDA-7 amino acid sequence); (ii) a protein (lacking a signal peptide) having an amino acid sequence comprising SEQ ID NO: 4 (the human wild-type MDA-7 amino acid sequence lacking the signal peptide);
- the M4 fragment of wild-type MDA-7 may be used, having the sequence of residues 104 to 206 of SEQ ID NO:2, which is identified herein as SEQ ID NO:5, or a protein which is at least 90 percent, at least 95 percent, or at least 98 percent homologous thereto (all of which are defined herein to be "M4 proteins").
- a nucleic acid encoding a M4 protein is referred to as a "M4 nucleic acid” herein, and includes, but is not limited to, a nucleic acid, the sequence of which is SEQ ID NO:6.
- Mda-7 molecules is defined herein as the class of molecules including mda-7 nucleic acids, MDA-7 proteins, M4 nucleic acids, and M4 proteins.
- the present invention provides for mda-7 nucleic acids or M4 nucleic acids operably linked to a promoter element and optionally comprised in an expression vector.
- the mda-7 nucleic acid or M4 nucleic acid may be operably linked to a suitable promoter element, such as, but not limited to, the cytomegalovirus immediate early (CMV) promoter, the Rous sarcoma virus (RSV) long terminal repeat promoter, the human elongation factor 1 ⁇ promoter, the human ubiquitin c promoter, etc.
- CMV cytomegalovirus immediate early
- RSV Rous sarcoma virus
- inducible promoters include the murine mammary tumor virus promoter (inducible with dexamethasone), commercially-available tetracycline-responsive or ecdysone- responsive promoters, etc. It may also be desirable to utilize a promoter which is selectively active in the cancer cell to be treated, for example the PEG-3 gene promoter (U.S. No. 6,472,520). Examples of tissue- and cancer cell-specific promoters are well known to those of ordinary skill in the art.
- mda-7 or M4 nucleic acid - bearing vector include transcription start sites, stop sites, polyadenylation sites, ribosomal binding sites, etc.
- Suitable expression vectors include virus-based vectors and non- virus based DNA or RNA delivery systems.
- appropriate virus-based vectors include, but are not limited to, those derived from retroviruses, for example Moloney murine leukemia-virus based vectors such as LX, LNSX, LNCX or LXSN (Miller and Rosman, 1989, Biotechniques 7:980-989); lentiviruses, for example human immunodeficiency virus (“HIV”), feline leukemia virus (“FIV”) or equine infectious anemia virus ( ⁇ IAV)-based vectors (Case et al, 1999, Proc. Natl. Acad. Sci. U.S.A.
- HIV human immunodeficiency virus
- FMV feline leukemia virus
- ⁇ IAV equine infectious anemia virus
- herpes simplex viruses for example vectors based on HSV-I (Geller and Freese, 1990, Proc. Natl. Acad. Sci. U.S.A. 87:1149- 1153); baculoviruses, for example AcMNPV-based vectors (Boyce and Bucher, 1996, Proc. Natl. Acad. Sci. U.S.A. 93:2348-2352); SV40, for example SVluc (Strayer and Milano, 1996, Gene Ther. 2:581-587); Epstein-Barr viruses, for example EBV-based replicon vectors (Hambor et al, 1988, Proc.
- alphaviruses for example Semliki Forest virus- or Sindbis virus-based vectors (Polo et al, 1999, Proc. Natl. Acad. Sci. U.S.A. 96:4598-4603); vaccinia viruses, for example modified vaccinia virus (MVA)-based vectors (Sutter and Moss, 1992, Proc. Natl. Acad. Sci. U.S.A. 89:10847-10851) or any other class of viruses that can efficiently transduce human tumor cells and that can accommodate the nucleic acid sequences required for therapeutic efficacy.
- VMA modified vaccinia virus
- Non-limiting examples of non- virus-based delivery systems which may be used according to the invention include, but are not limited to, so-called naked nucleic acids (Wolff et al. , 1990, Science 247: 1465- 1468), nucleic acids encapsulated in liposomes (Nicolau et al, 1987, Methods in Enzymology 149:157-176), nucleic acid/lipid complexes (Legendre and Szoka, 1992, Pharmaceutical Research 9:1235- 1242), and nucleic acid/protein complexes (Wu and Wu, 1991, Biother. 3:87-95).
- naked nucleic acids Wilff et al. , 1990, Science 247: 1465- 1468
- nucleic acids encapsulated in liposomes Nicolau et al, 1987, Methods in Enzymology 149:157-176
- nucleic acid/lipid complexes Legendre and Szoka, 1992, Pharmaceutical Research 9:1235-
- the expression vector is an El -deleted human adenovirus vector of serotype 5, although those of ordinary skill in the art would recognize that many of the different naturally-occurring human Ad serotypes or Ad vectors derived from non-human adenoviruses may substitute for human Ad 5 -derived vectors.
- a recombinant replication-defective Ad.mda-7 virus for use as an mda-7 vector may be created in two steps as described in Su et al. , 1998, Proc. Natl. Acad. Sci. U.S.A. 95:14400-14405.
- the coding region of the mda-7 gene may be cloned into a modified Ad expression vector pAd.CMV (Falck-Pedersen et al. , 1994, MoI. Pharmacol. 45:684-689).
- This vector contains, in order, the first 355 bp from the left end of the Ad genome, the CMV promoter, DNA encoding splice donor and acceptor sites, the coding region of the mda-7 cDNA, DNA encoding a polyA signal sequence from the ⁇ globin gene, and ⁇ 3 kbp of adenovirus sequence extending from within the ElB coding region.
- the recombinant virus may be created in vitro in 293 cells (Graham et al, 1977, J. Gen. Virol. 36:59-72) by homologous recombination between an mda-7-containing version of pAd.CMV and plasmid pJM17, which contains the whole of the Ad genome cloned into a modified version of pBR322 (McGrory et al, 1988, Virology 163:614-617).
- pJM17 gives rise to Ad genomes in vivo, but they are too large to be packaged in mature Ad capsids.
- This constraint is relieved by recombination with the vector to create a packageable genome ⁇ Id.) containing the mda-7 gene.
- the recombinant virus is replication defective in human cells except 293 cells, which express adenovirus ElA and ElB. Following transfection of the two plasmids, infectious virus may be recovered, and the genomes may be analyzed to confirm the recombinant structure, and then virus may be plaque purified by standard procedures (Volkert and Young, 1983, Virology 125: 175- 193).
- the infectivity of an adenovirus vector carrying an mda-7 gene may be improved by inserting an Arg-Gly-Asp motif into the fiber know (Ad5-Delta24RGD), as described in Lamfers et al, 2002, Cancer Res. 62:5736-5742.
- MDA-7 or M4 protein may be produced in vivo in an organism or in an isolated cell which may be part of a cell population.
- MDA-7 or M4 protein may be produced in a recombinant expression and then collected and purified using methods known in the art.
- Such proteins may be administered comprised in liposomes, nanoparticles, with a carrier, etc. (see below).
- a mda-7 nucleic acid or a M4 nucleic acid may be operably linked to a suitable promoter and introduced into a cell in need of such treatment (a "target cell"), which, as defined herein, is a cell which exhibits multidrug resistance and may have been demonstrated to over express the mdr-1 gene.
- a target cell a cell in need of such treatment
- the mda-7 nucleic acid or the M4 nucleic acid may be contained in a viral vector, operably linked to a promoter element that is either inducible or constitutively active in the target cell.
- the viral vector is a replication-defective adenovirus (as described above).
- a viral vector containing a nucleic acid encoding a mda-7 nucleic acid or a M4 nucleic acid in expressible form, operably linked to a suitable promoter element may be administered to a population of target cells at a multiplicity of infection (MOI) ranging from 10- 100 MOI.
- MOI multiplicity of infection
- the amount of a viral vector administered to a subject may be 1 X 10 9 pfu to 1 X 10 12 pfu.
- a mda-7 nucleic acid or a M4- nucleic acid preferably operably linked to a promoter element, comprised in a vector or otherwise, may be introduced into a cell ex vivo and then the cell may be introduced into a subject.
- a nucleic acid encoding mda-7 may be introduced into a cell of a subject ex vivo and then the cell containing the nucleic acid may be optionally propagated and then (with its progeny) introduced into the subject.
- a MDA-7 protein or a M4 protein may be used in protein/peptide therapy of a subject in need of such treatment.
- the MDA-7 protein or M4 protein may be prepared by chemical synthesis or recombinant DNA techniques, purified by methods known in the art, and then administered to a subject in need of such treatment.
- MDA-7 protein or M4 protein may be comprised, for example, in solution, in suspension, and/or in a carrier particle such as microparticles, liposomes, or other protein-stabilizing formulations known in the art
- formulations of MDA-7 protein or M4 protein may stabilized by addition of zinc and/or protamine stabilizers as in the case of certain types of insulin formulations.
- a MDA-7 protein or a M4 protein may be linked covalently or non-covalently, to a carrier protein which is preferably non-immunogenic.
- a MDA-7 protein or a M4 protein is administered in an amount which achieves a local concentration in the range of 18 to 50 ng per microliter.
- a subject may be administered a range of 50- 100 mg of MDA-7 protein or M4 protein per kilogram.
- the dose range may be between 100-2500 mg/day of MDA-7 protein or M4 protein.
- Any of the foregoing molecules may be administered by direct instillation, injection (intravenous, subcutaneous, intramuscular, intraarterial, intrathecal, intrahepatic, intratumoral), or any other method or route known in the art.
- the present invention relates to a method for improving the therapeutic response of a subject suffering from a disorder to chemotherapy, where the disorder in the subject had previously been resistant to chemotherapy and/or where the mdr-1 gene and/or MDR-I protein are shown to be over-expressed, comprising administering, to the subject, an effective amount of a mda-7 nucleic acid or a MDA-7 protein or a M4 nucleic acid or a M4 protein, in conjunction with one or more chemotherapeutic agent.
- the subject may be a human or a non-human subject, but preferably is a human subject.
- the subject may be suffering from a disorder which is a malignancy or a non-malignant disorder.
- malignant disorders include, but are not limited to, any malignancy that exhibits multi-drug resistance, and that preferably has been demonstrated to over-express mdr-1 gene and/or MDR-I protein (also referred to as "P-glycoprotein” or "P-gp” herein).
- Such malignancies may include, but not be limited to, breast cancer, ovarian cancer, lung cancer (e.g., adenocarcinoma, small- cell carcinoma, non-small cell carcinoma, mesothelioma), colorectal cancer, renal carcinoma, chronic lymphocytic leukemia, acute lymphocytic leukemia, Hodgkin's lymphoma, non-Hodgkin's lymphoma, neuroblastoma, glioblastoma, prostate cancer, pancreatic cancer, gastric cancer, squamous cell carcinoma (e.g., oral squamous cell carcinoma and squamous cell carcinoma of the head and neck), melanoma, hepatic adenocarcinoma, bladder cancer, testicular cancer, and rhabdosarcoma.
- lung cancer e.g., adenocarcinoma, small- cell carcinoma, non-small cell carcinoma, mesothelioma
- subjects for treatment according to the invention include any subject suffering from a non-malignant disease, where the response of the disease to therapy is negatively impacted by multi-drug resistance, where the subject has preferably been demonstrated to over express mdr-1 gene and/or MDR-1 protein.
- Such subjects may include, but not be limited to, subjects suffering from a viral infection, such as, but not limited to, human immunodeficiency infection, hepatitis B virus infection, Epstein-Barr virus infection, Herpes simplex virus infection, human papilloma virus infection, or hepatitis C infection.
- the subject may be suffering from an autoimmune disease, such as, but not limited to, rheumatoid arthritis, juvenile rheumatoid arthritis, system lupus erythematosis, Sjogren's syndrome, scleroderma, polymyositis, or diabetes mellitus.
- an autoimmune disease such as, but not limited to, rheumatoid arthritis, juvenile rheumatoid arthritis, system lupus erythematosis, Sjogren's syndrome, scleroderma, polymyositis, or diabetes mellitus.
- the subject may be suffering from multiple sclerosis.
- an improved therapeutic response may be characterized by, for example and not by way of limitation, one or more of the following: higher rate of survival, longer life expectancy, longer period to relapse, increased comfort, decreased tumor burden, decreased pain, decreased rate or incidence of metastasis.
- an improved therapeutic response may be characterized by, for example and not by way of limitation, one or more of the following: increased survival, longer life expectancy, decreased viral titer, increased CD4+ T cell count, decreased incidence of secondary infection, decreased incidence of associated carcinoma.
- an improved therapeutic response may be characterized by, for example and not by way of limitation, decreased serologic markers of inflammation such as C-reactive protein and erythrocyte sedimentation rate, decreased clinical inflammation, (for arthritis) increased range of motion, decreased pain, longer disease-free interval, decreased need for pain medication or insulin therapy, increased life expectancy, decreased workplace absenteeism.
- an improved therapeutic response may be characterized by, for example and not by way of limitation, decreased incidence of relapse, longer relapse-free intervals, shorter duration of relapse, decreased severity of symptoms at relapse, decreased lesions by imaging studies such as MRI, and improved nerve conduction studies.
- “Resistance to chemotherapy” means that the subject had been administered one or more chemotherapeutic agent which did not achieve a satisfactory therapeutic response (examples of types of therapeutic responses being set forth above).
- “Over-expression of mdr-1 gene or protein” means an increase in the level of mdr-1 gene transcription and/or MDR-I protein levels.
- a sample of cells afflicted by the disorder may be collected from the subject and then tested for levels of mdr-1 mRNA and/or MDR-1 protein, using techniques known in the art, such as PCR analysis, Northern Blot, Western blot, immunohistochemistry, etc.
- the sequence of the human MDR-1 gene and protein are known.
- the Genbank accession numbers for mdr-1 nucleic acid and MDR-1 protein are M 14758 and P08183 respectively.
- "Over-expressing" means an increase relative to a normal control cell, preferably by at least about 10, at least about 20, at least about 30, at least about 40, at least about 50, at least about 60, at least about 70, at least about 80, or at least about 90 percent.
- a subject need not be tested for over-expression of Mdr-1 molecules if they have proved to be clinically refractory to treatment. Conversely, a patient need not have been demonstrated to be refractory to treatment if over-expression of mdr-1 gene or MDR-1 protein is demonstrated.
- a mda-7 nucleic acid or a MD A-7 protein or a M4 nucleic acid or a M4 protein may be performed as set forth in the section above.
- Concurrent administration of one or more chemotherapeutic agent means that the Mda-7 molecule and the one or more chemotherapeutic agent are administered close enough in time to each other for the clinical effect of the Mda-7 molecule may be exerted on the chemotherapeutic agent.
- the Mda-7 molecule (mda- 7 nucleic acid, MDA-7 protein, M4 nucleic acid or M4 protein) may be administered simultaneously with, prior to, or after the administration of the one or more chemotherapeutic agent (preferably prior to administration of the one or more chemotherapeutic agent).
- a mda-7 nucleic acid or a M4 nucleic acid is administered by gene therapy, the time period between administering it and the chemotherapeutic agent may be quite long, and one administration of gene therapy may suffice for a plurality of subsequent chemotherapeutic treatments. If MDA-7 protein or M4 protein is administered, the interval to administration of the one or more chemotherapeutic agent would be shorter in view of degradation of the protein.
- Non-limiting specific examples of intervals between administration of an Mda-7 molecule and one or more therapeutic agent include up to -5 years ("-" means prior administration of the Mda-7 molecule), up to -3 years, up to -2 years, up to -1 year, up to -12 months, up to -11 months, up to -10 months, up to -9 months, up to -8 months, up to -7 months, up to -6 months, up to -3 months, up to -1 month, up to - 2 weeks, up to -1 week, up to -6 days, up to -5 days, up to -4 days, up to -3 days, up to -2 days, up to -1 day, up to -12 hours, up to -8 hours, up to -4 hours, up to -2 hours, up to -1 hours, 0 hours, up to +1 hour (“+” means that Mda-7 molecules is administered after chemotherapeutic agent (s)), up to +2 hours, up to +4 hours, up to +8 hours, up to +12 hours, up to
- the one or more chemotherapeutic agent selected is related to the nature of the disorder being treated.
- the one or more chemotherapeutic agent may be, for example and not by way of limitation, an alkylating agent such as busulfan, cisplatin, carboplatin, chlorambucil, cyclophosphamide, ifosfamide, dacarbazine, mechlorethamine (nitrogen mustard), melphalan, and temozolomide; a nitrosourea such as streptozocin, carmustine, and lomustine, an antimetabolite such as 5-fluorouracil, capecitabine, 6-mercaptopurine, methotrexate, gemcitabine, cytarabine, fludarabine, and pemetrexed; an anthracyline-like drug such as daunorubicin, doxorubicin, epirubicin, idarubicin, and mitoxantrone; a topoisomerase I inhibitor such as topotecan and irinotecan;
- an alkylating agent such
- the one or more chemotherapeutic agent may be, for example and not by way of limitation, a reverse transcriptase inhibitor, including nucleoside reverse transcriptase inhibitors such as 3TC (lamivudine), abacavir, AZT (zidovudine), d4T (stavudine), ddC (zalcitabine), ddl (didanosine) and FTC (emtricitabine) and non-nucleoside reverse transcriptase inhibitors such as efavirenz, delavirdine, neviripine, etravirine, rilpirivine ; a protease inhibitor such as saquinivir, nelfinivir, ritonivir, indinivir, amprenavir, lopinavir, atazanavir, fosamprenavir, or tipranavir; a fusion inhibitor such enfuvirtide as 3TC (lamivudine), a
- the one or more chemotherapeutic agent may be, for example and not by way of limitation, aciclovir, cidofovir, docosanol, famiciclovir, fomivirsen, foscarnet, ganciclovir, idoxuridine, penciclovir, trifluridine, tromantidine, valaciclovir, valganciclovir, or vidarabine, or combinations thereof.
- the one or more chemotherapeutic agent may be, for example and not by way of limitation, adefovir or lamivudine, or a combination thereof.
- a chemotherapeutic agent may be, for example and not by way of limitation, ribivirin.
- the one or more chemotherapeutic agent may be glatiramir or novantrone or a combination thereof.
- the present invention provides for a method of treating a subject suffering from a cancer found to be resistant to chemotherapy and/or where the mdr-1 gene and/or MDR-I protein are shown to be over-expressed comprising administering, to the subject, an effective amount of a Mda-7 molecule, for example mda-7 nucleic acid, MDA-7 protein, M4 nucleic acid or M4 protein.
- a Mda-7 molecule for example mda-7 nucleic acid, MDA-7 protein, M4 nucleic acid or M4 protein.
- Mda-7 melanoma differentiation associated gene -7
- MDRI multidrug resistance
- the present invention provides for a method of treating a subject suffering from a cancer, comprising (i) determining that cancer cells of the subject over express the mdr-1 gene and/or MDR-I protein; and (ii) administering, to the subject, an effective amount of an Mda-7 molecule (as described above).
- the present invention provides for a method of evaluating the likelihood that a subject, suffering from a cancer, would benefit from therapy with a Mda-7 molecule, comprising determining whether or not cancer cells in the subject over-express the mdr-1 gene or MDR-I protein (Mdr-1 molecules), wherein over-expression of mdr-1 gene and/or MDR-I protein is consistent with a greater likelihood that the subject would benefit from
- FIGURE IA depicts the results of Western blot analysis of endogenous MDR- 1 ("P-gp") levels in S W620/WT and S W620/DOX cells, and shows that the SW620/DOX cell line over expressed P-gp.
- FIGURE IB depicts the effect of different doses of Dox on cell viability in SW620/WT and S W620/DOX cells. Cells were cultured in the presence of different concentrations of
- FIGURE 1C particularly addresses the ability of Dox to induce apoptosis in SW620/WT and SW620/Dox cells, based on the levels of Annexin V-
- FIGURE 1C shows that the level of Annexin V binding as a result of Dox treatment was greater in SW620/WT cells, and the effect was dose-dependent.
- SW620/DOC cells was explored. Experiments were performed in which SW620 and SW620/Dox cells were infected with either (empty vector) Ad.vec or Ad.mda7 (25 pfu/cell) and then treated with the indicated concentrations of Dox for 48 h. The percentages of apoptotic cells were determined by Annexin V binding assay and flow cytometry. As shown in FIGURE 2A, the S W620/DOX cells infected with Ad.vec remained relatively resistant to Dox, but SW620/DOX cells infected with Ad.mda7 exhibited a sensitivity to Dox that approached that of S W620/WT.
- Ad.mda7 When cells were infected with either Ad.vec or Ad.mda7 (25 pfu/cell), treated with Dox for 24 h., and then total proteins were extracted and subjected to Western blot analysis, Ad.mda7 infection was found to down regulate P-gp expression in SW620/Dox cells (FIGURE 2B).
- FIGURE 2C the intracellular accumulation of doxorubicin, determined after cells were infected with either Ad.vec or Ad.mda7 (25 pfu/cell) for 24 h, incubated with 2 or 4 ⁇ M of Dox for 60 min., washed twice with ice-cold PBS, resuspended in 400 ⁇ l PBS and then analyzed by flow cytometry, was increased in Ad.mda7-infected SW620/Dox cells.
- FIGURE 2A-C demonstrates that Ad.mda7 infection induces MDR-I reversal in SW620/Dox cells.
- FIGURE 3 The ability of Ad.mda7 infection to increase Dox accumulation in SW620/DOX cells is further supported by FIGURE 3.
- Cells were incubated with 10 ⁇ M doxorubicin at 37 0 C for 30 min (substrate loading phase) and then washed twice with PBS. Cells were then infected with either Ad.vec or Ad.mda7 at the indicated MOI. At the defined time points, cells were harvested, centrifuged and washed in ice- cold PBS. Cell pellets were then resuspended in 400 ⁇ l PBS and used immediately for flow cytometric analysis for intracellular doxorubicin retention.
- SW620 and SW620/Dox cells were infected with either Ad.vec or Ad.mda7 at MOIs of either 25 or 50 pfu/cell, and then either viability was determined by MTT assay 5 days after infection or apoptosis induction was evaluated by staining infected cells after 48 h with APC-labeled Annexin V followed immediately by flow cytometry analysis. The percentage of early and late apoptotic cells (only Annexin V stained) was calculated using FlowJo Version 6.31 software.
- FIGURE 4A-B Ad.mda7 infection inhibited cell growth and induced apoptosis preferentially in P-gp-expressing mdr-1 variant SW620/Dox cells.
- FIGURE 4C Western blot analysis showed that MDA- 7/IL-24 protein expression was increased in S W620/DOX relative to S W620/WT.
- SW620/WT cells were transiently transfected with either empty vector (pCDNA3.1) or MDR, infected with either Ad.vec or Ad.mda7, and then apoptosis induction was evaluated by Annexin V binding assay 48 h after infection. The percentage of early and late apoptotic cells (only Annexin V stained) was calculated using FlowJo Version 6.31 software. As shown in FIGURE 5 A. over- expression of MDR was observed to induce increased sensitivity to Ad.mda7 infection in SW620 cells.
- FIGURE 5B Western blot analysis performed on cell lysates harvested at 24 h after infection indicated that over-expression of mdr- 1 /MDR-1 was associated with enhanced expression of MDA-7/IL-24 protein.
- SW620/WT cells were transiently transfected either with empty vector (pCDNA3.1) or mdr-1, infected with either Ad.vec or Ad.mda7 and, 24 hours later, 1 x 10 5 cells were replated in 0.4% agar on 0.8% base agar and then, after two weeks, colonies >50 cells were counted under a dissection microscope, it was found that ectopic expression of mdr-l/MDR-1 in SW620 cells significantly inhibited colony formation in soft agar (FIGURE 5C).
- SW620/DOX cells were transiently transfected with either control siRNA or mdr-1 siRNA using lipofectamine prior to infection with either Ad.vec or Ad.mda7. Forty-eight hours later, apoptosis was measured by Annexin V binding assay using APC-conjugated Annexin V by flow cytometry. The percentage of early and late apoptotic cells (only Annexin V stained) was calculated using FlowJo Version 6.31 software. As shown in FIGURE 6, down regulation of mdr-1 partially protected SW620/DOX cells from Ad.mda7-induced apoptosis.
- FIGURE 7 shows confocal images of SW620/WT and SW620/DOX cells immunofluorescently stained with anti-k-Ras antibody. Note extensive plasma-membrane localization in the SW620/WT cell images (typical pattern) and surface distribution in the SW620/DOX cell images.
- mda-7/IL-24 when administered by means of a replication-incompetent adenovirus, Ad.mda-7, at a multiplicity of infection of 25-50 pfu/cell, significantly increased the sensitivity of SW620/DOX cells to doxorubicin.
- Ad.mda-7 at a multiplicity of infection of 25-50 pfu/cell, significantly increased the sensitivity of SW620/DOX cells to doxorubicin.
- the cytotoxic effect of doxorubicin in the SW620/WT sensitive cell line was not significantly altered by Ad.mda-7 infection.
- Annexin V binding assays revealed that Ad.mda-7 infection effectively reversed resistance to doxorubicin-induced apoptosis in SW620/DOX cells.
- infection of SW620/DOX cells with Ad.mda-7 increased intracellular accumulation of doxorubicin and significantly decreased P-gp expression.
- mda-7/IL-24 is a potent MDR reversal agent, preferentially causing apoptosis in P-gp over expressing MDR cells, suggesting significant clinical implications for the use of mda-7/IL-24 in treating neoplasms that have failed chemotherapy mediated by the P-gp multidrug resistance mechanism.
- Various references are cited herein, the contents of which are hereby incorporated by reference in their entireties.
- SEQ IDNO:1 nucleotides 275 to 895 ofGenBankAccessionNo. U16261 atgaattttc aacagaggct gcaaagcctg tggactttag ccagaccctt ctgccctcct 60
- caagctcagg ataacatcac gagtgcccgg ctgctgcagc aggaggttct gcagaacgtc 300
- SEQ ID NO:2 amino acid sequence of complete wild-type MDA-7 protein (GenBank Accession No. U 16261)
- SEQ ID NO: 3 nucleic acid sequence encoding the portion of wild-type MDA-7 lacking the signal sequence atgcagggcc aagaattcca ctttgggccc tgccaagtga agggggttgt tccccagaa 60
- SEQ ID NO:4 amino acid sequence of wild-type MDA-7 protein lacking the signal sequence
- SEQ ID NO:5 amino acid sequence of M4; residues 104 to 206 of SEQ ID NO:2.
- SEQ IDNO:6 portionofSEQ IDNO:1 encoding M4 atggagagct gttaccttgt ccacaccctg ctggagttct acttgaaaac tgttttcaaa 60
Landscapes
- Health & Medical Sciences (AREA)
- Life Sciences & Earth Sciences (AREA)
- Engineering & Computer Science (AREA)
- Genetics & Genomics (AREA)
- Chemical & Material Sciences (AREA)
- Bioinformatics & Cheminformatics (AREA)
- Biotechnology (AREA)
- Zoology (AREA)
- General Health & Medical Sciences (AREA)
- Animal Behavior & Ethology (AREA)
- Epidemiology (AREA)
- Pharmacology & Pharmacy (AREA)
- Public Health (AREA)
- Veterinary Medicine (AREA)
- Molecular Biology (AREA)
- Organic Chemistry (AREA)
- Biomedical Technology (AREA)
- Medicinal Chemistry (AREA)
- General Engineering & Computer Science (AREA)
- Wood Science & Technology (AREA)
- Virology (AREA)
- Microbiology (AREA)
- Plant Pathology (AREA)
- Biophysics (AREA)
- Biochemistry (AREA)
- Physics & Mathematics (AREA)
- Gastroenterology & Hepatology (AREA)
- Immunology (AREA)
- Proteomics, Peptides & Aminoacids (AREA)
- Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
- Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
Abstract
The present invention relates to the use of melanoma differentiation associated gene-7, its encoded protein, and related molecules (Collectively, 'Mda-7 Molecules') for improving the response of a subject to chemotherapy and the particular benefit of Mda-7 treatment in a subject resistant to chemotherapy. It is based, at least in part, on the discovery that mda-7 expression increases the response of hitherto resistant cells to chemotherapeutic agent, as well as on the discovery that mda-7 expression is itself enhanced in the context of over- expression of the multidrug resistance gene (mdr-l).
Description
INCREASED EFFECTIVENESS OF MDA-7 WITH MDR-I OVER-EXPRESSION
GRANT INFORMATION
This invention was made with government support under Grant Nos. POl CA104177, ROl CA35675 and ROl CA097318 awarded by the National Institutes of Health/National Cancer Institutes. The government has certain rights in the invention.
PRIORITY BENEFIT This application claims the benefit of priority to United States
Provisional Application Serial No. 60/894,068, filed March 9, 2007, the contents of which is hereby incorporated by reference in its entirety.
1. INTRODUCTION
The present invention relates to the use of melanoma differentiation associated gene-7, its encoded protein, and related molecules ( Collectively, "Mda-7 Molecules") for improving the response of a subject to chemotherapy and the particular benefit of Mda-7 treatment in a subject resistant to chemotherapy. It is based, at least in part, on the discovery that mda-7 expression increases the response of hitherto resistant cells to chemotherapeutic agent, as well as on the discovery that mda-7 expression is itself enhanced in the context of over- expression of the multidrug resistance gene (mdr-l).
2. BACKGROUND OF THE INVENTION
2.1 MDA-7
Melanoma differentiation associated gene-7 (mda-7) (Jiang et al., 1995, Oncogene 11 :2477-2486) is a secreted cytokine belonging to the interleukin (IL)-IO family designated as IL-24 (Pestka et al., 2004, Annu Rev Immunol 22:929- 979). Multiple independent studies demonstrate that delivery of mda-7/IL-24 by a replication incompetent adenovirus, Ad.mda-7, or as a GST-MD A-7 fusion protein selectively kills diverse cancer cells. In contrast to its harmful effects on tumor cells, mda-7/IL-24 does not induce toxicity in normal endothelial and epithelial cells, fibroblasts, melanocytes and astrocytes (Ekmekcioglu et al., 2001, Int J Cancer 94(l):54-59; Ellerhorst et al., 2002, J CHn Oncol 20(4):1069-1074; Fisher, 2005, Cancer Res 65(22):10128-10138; Fisher et al., 2003, Cancer Biol Ther 2 (4 Suppl l):S23-37; Gupta et al., 2006, Pharmacol Ther 111 : 596-628; Huang et al., 2001, Oncogene 20(48):7051-7063; Jiang et al., 1996, Proc Natl Acad Sci U S A
93(17):9160-9165; Lebedeva et al., 2005, MoI Ther 11(1):4-18; Lebedeva et al., 2002, Oncogene 21(5):708-718; Madireddi et al., 2000, Adv Exp Med Biol 465:239-261; Mhashilkar et al., 2001, MoI Med 7(4):271-282; Saeki et al., 2000, Gene Ther 7(23):2051-2057; Saeki et al., 2002, Oncogene 21(29):4558-4566; Sarkar et al., 2006, Anti-Inflammatory & Anti- Allergy Agents in Medicinal Chemistry 5: 259-274; Sarkar et al., 2002, Biotechniques Suppl:30-39; Sarkar et al., 2002, Proc Natl Acad Sci U S A 99(15):10054-10059; Sauane et al., 2003, Cytokine Growth Factor Rev 14(1):35-51; Su et al., 2001, Proc Natl Acad Sci U S A 98(18):10332-10337; Su et al., 1998, Proc Natl Acad Sci U S A 95(24): 14400-14405). In addition, mda-7/IL-24 possesses potent anti-angiogenic, immunostimulatory and bystander activities (Fisher,
2005, Cancer Res 65(22):10128-10138; Fisher et al., 2003, Cancer Biol Ther 2 (4 Suppl l):S23-37; Gupta et al., 2006, Pharmacol Ther 111 : 596-628; Lebedeva et al., 2005, MoI Ther 11(1):4-18). The sum of these attributes makes mda-7/IL-24 a significant candidate for cancer gene therapy (Fisher, 2005, Cancer Res 65(22): 10128- 10138).
Indeed, Ad.mda-7 has been successfully used for a Phase I clinical trial for advanced carcinomas and melanomas and has shown promising results in tumor growth inhibition and induction of cancer apoptosis (Cunningham et al., 2005, MoI Ther 11(1):149-159 ; Fisher, 2005, Cancer Res 65(22):10128-10138; Fisher et al., 2003, Cancer Biol Ther 2 (4 Suppl l):S23-37, ; Lebedeva et al., 2005, MoI Ther
11(1):4-18; Tong et al., 2005, MoI Ther 1 l(l):160-172). Previous studies demonstrate that Ad.mda-7 induces growth suppression and apoptosis in histologically diverse cancer cells containing single or multiple genetic defects, including alterations in p53, pl6/INK4a and/or Rb (Emdad et al., 2006, J Cell Physiol 208(2):298-306; Huang et al., 2001, Oncogene 20(48):7051-7063; Jiang et al., 1996, Proc Natl Acad Sci U S A 93(17):9160-9165; Lebedeva et al., 2002, Oncogene 21(5):708-718; Mhashilkar et al., 2001, MoI Med 7(4):271-282; Saeki et al., 2000, Gene Ther 7(23):2051-2057; Su et al., 2001, Proc Natl Acad Sci U S A 98(18):10332-10337; Su et al., 1998, Proc Natl Acad Sci U S A 95(24):14400-14405). Moreover, Ad.mda-7 is equally effective in inducing apoptosis in breast and lung carcinoma and melanoma cells containing wtp53, mutp53 or which are null for p53 expression (Lebedeva et al., 2002, Lebedeva et al., 2002, Oncogene 21(5):708-718; Madireddi et al., 2000, Adv Exp Med Biol 465:239-261; Saeki et al., 2000, Gene Ther 7(23):2051-2057; Saeki et al., 2002, Oncogene 21(29):4558-4566; Su et al., 1998, Proc Natl Acad Sci U S A 95(24): 14400-14405).
A fragment of MDA-7, retaining amino acid residues 104 to 206 of the wild-type sequence, has been found to retain the cancer-specific growth suppressive and apoptosis-inducing properties of the full-length protein (Gupta et al., 2006, Cancer Res. 66(16^:8182-8191).
2.2 MDR-I
The mdr-1 gene encodes a multi-drug resistant transporter protein, MDR-1/P-glycoprotein (also referred to herein as "P-gp"), a 170 kDa glycosylated membrane protein which is a member of the ATP-binding cassette superfamily of membrane transporters (Meijer et al., 1999, J. Clin. Pathol. 52:450-454; Sakeada et al., 2002, Biol. Pharma Cull. 25:1391-1400). MDR-1/P-glycoprotein acts as a molecular pump which can expel chemotherapeutic agents from tumor cells. The phenomenon of multi-drug resistance arises in a wide variety of cancers, and creates serious obstacles to successful cancer treatment (Mignona et al., 2006, BMC Cancer, 6:293). Mdr-1 over-expression has been linked to a worse clinical prognosis which is both dependent as well as independent of multi-drug resistance (Mignona et al., 2006, BMC Cancer 6:293; Hille et al., 2006, Anticancer Drugs 17(9 V.1041 -1044: Matthews et al., 2006, Leuk. Lymphoma 470i}:2308-2313).
In addition to its obstructive role in cancer therapy, over-expression of mdr-1 gene and protein also interferes with chemotherapy of diseases other than cancer. MDR-1 is believed to function in the transport and metabolism of antiretroviral agents, and variants of MDR-1 have been associated with different levels of therapeutic response in human immunodeficiency virus infection (Saitoh et al., 2005, AIDS Jj9{4}:371-380). Hepatitis B virus has been shown to transactivate mdr-1 gene expression (Doong et al., 1998, J. Hepatol. 29(6): 872-878). Further, Mdr-
1 over-expression has been associated with various autoimmune diseases (Levy et al., 2002, Br. J. Haematol. 118(3):836-838: Tsujimura et al., 2004, Genes Cells 9(121:1265-1273).
3. SUMMARY OF THE INVENTION
The present invention relates to a method for improving the therapeutic response of a subject to chemotherapy, where the subject had previously been resistant to chemotherapy and/or where the mdr-1 gene and/or MDR-I protein are shown to be over-expressed, comprising administering, to the subject, an effective amount of a Mda-7 molecule (mda-7 nucleic acid, MDA-7 protein, M4 nucleic acid or M4 protein, defined below) in conjunction with one or more chemotherapeutic agent. It is based, at least in part, on the discovery that expression of mda-7 in drug-resistant cancer cells resulted in increased accumulation of, and sensitivity to, the drug.
In addition, in alternative embodiments, the present invention provides for a method of treating a subject suffering from a cancer found to be resistant to chemotherapy and/or where the mdr-1 gene and/or MDR-I protein are shown to be over-expressed comprising administering, to the subject, an effective amount of a Mda-7 molecule. This aspect is based, at least in part, on the discovery that the anticancer effect of the melanoma differentiation associated gene -7 (mda-7) was increased in the context of over-expression of the multidrug resistance (MDR-I) gene.
4. BRIEF DESCRIPTION OF THE FIGURES FIGURE IA-C. A. Western blot analysis of endogenous P-gp levels in SW620/WT and SW620/DOX cells. B. Effect of different doses of doxorubicin (Dox) on cell growth in SW620/WT and SW620/DOX cells. Graphs show the average ± SD
of results of 3 independent experiments. C. Effect of Dox on apoptosis induction in SW620/WT and SW620/DOX cells by Annexin V binding assay. Results are the average ± SD from replicate studies.
FIGURE 2A-C. A. SW620/WT (also referred to herein as "SW620") and SW620/DOX cells were infected with either Ad.vec or Ad.mda7 (25 pfu/cell) and then treated with the indicated concentrations of Dox for 48 h. Percentage of apoptotic cells were determined by Annexin V binding assay and flow cytometry. B. Cells were infected with either Ad.vec or Ad.mda7 (25 pfu/cell) and then treated with the indicated concentrations of Dox for 24 h. Total proteins were extracted and Western blot analysis was performed with the indicated antibodies. C. Effect of Ad.mda7 on the accumulation of Dox in SW620/WT and SW620/DOX cells. Upper panel, representative flow cytometric image of Dox accumulation. Lower panel, graphical presentation of Dox accumulation. Values are the average ± SD of three independent experiments. FIGURE 3 A-B. Effect of Ad.mda7 on the efflux of Dox from
SW620/WT and SW620/DOX cells. A. Representative flow cytometric image of Dox retention. B. Graphical presentation of Dox retention. Values are the average ± SD of three independent experiments.
FIGURE 4A-C. A. SW620/WT and SW620/DOX cells were infected with either Ad.vec or Ad.mda7 with the indicated MOI and cell viability was determined by MTT assay 5 days after infection. Values are the average ± SD of three independent experiments. B. Effect of Ad.mda7 infection on apoptosis induction in SW620/WT and SW620/DOX cells by Annexin V binding assay. C. MDA-7/IL-24 protein expression in SW620/WT and SW620/DOX cells following infection with Ad.mda-7 determined by Western blot analysis.
FIGURE 5A-D. A. SW620/WT cells were transiently transfected with either empty vector (pCDNA3.1) or MDR and then infected with either Ad.vec or Ad.mda7 at the indicated MOI. Apoptosis induction was evaluated by Annexin V binding assay 48 h after infection. B. Effect of MDR over-expression on MDA-7/IL- 24 protein expression. Western blot Analysis was performed with the cell lysates harvested at 24 h after infection. C. Colony formation in soft agar as a function of Ad.vec or Ad.mda-7 in the presence or absence of MDR. Data represents mean ± SD of triplicate plates in three independent experiments. D. Graph shows cloning efficiency. FIGURE 6. Effect of down-regulation of MDR (by siRNA) on
Ad.mda7-induced apoptosis in SW620/DOX cells.
FIGURE 7A-B. K-ras localization in (A) SW620/WT and (B) SW620/DOX cells. Confocal images of SW620/WT and SW620/DOX cells immunofluorescently stained with anti-k-Ras antibody.
5. DETAILED DESCRIPTION OF THE INVENTION For clarity of presentation, and not by way of limitation, the detailed description of the invention is divided into the following subsections:
(i) Mda-7 molecules; (ii) methods of administering Mda-7 molecules;
(iii) use of Mda-7 to improve response to chemotherapy; and (iv) use of Mda-7 therapy in MDRl -expressing subjects.
5.1 MDA-7 MOLECULES
This application uses a convention whereby a nucleic acid is referred to by lower case letters (e.g., mda-7 or mdr-1), a protein is referred to by capital letters (e.g., MDA-7 or MDR-I) and both are generically referred to by an initial capital letter followed by lower case (e.g. Mda-7 or Mdr-1).
A "mda-7 nucleic acid" as defined herein is a nucleic acid which may be:
(i) a nucleic acid having a nucleotide sequence comprising SEQ ID NO:1 (nucleotides 275 to 895 of GenBank Accession No. U 16261; Jiang et al., 1995, Oncogene 11 -.2477-2486), which encodes wild-type human MDA-7 protein;
(ii) a nucleic acid which encodes an MDA-7 protein having an amino acid sequence comprising SEQ ID NO:2 (GenBank Accession No. Q13007; Jiang et al., 1995, Oncogene 11 :2477-2486; the human wild-type MDA-7 amino acid sequence); (iii) a nucleic acid having a nucleotide sequence comprising SEQ ID NO:3, which encodes a version of wild-type human MDA-7 protein lacking the signal sequence;
(iv) a nucleic acid which encodes a protein (lacking a signal peptide) having an amino acid sequence comprising SEQ ID NO:4 (the human wild-type MDA-7 amino acid sequence lacking the signal peptide);
(v) a nucleic acid having a nucleotide sequence which is at least 90 percent or at least 95 percent homologous to SEQ ID NO: 1 ;
(vi) a nucleic acid having a nucleotide sequence which is at least 90 percent or at least 95 percent homologous to SEQ ID NO:3;
(vii) a nucleic acid which encodes a protein having an amino acid sequence which is at least 90 percent or at least 95 percent homologous to SEQ ID NO:2; or (viii) a nucleic acid which encodes a protein having an amino acid sequence which is at least 90 percent or at least 95 percent homologous to SEQ ID NO:4 (where homology may be determined, for example, using standard software such as BLAST or FASTA).
A MDA-7 protein, as defined herein is a protein which may be: (i) a protein having an amino acid sequence comprising SEQ ID NO:2 (the human wild-type MDA-7 amino acid sequence); (ii) a protein (lacking a signal peptide) having an amino acid sequence comprising SEQ ID NO: 4 (the human wild-type MDA-7 amino acid sequence lacking the signal peptide);
(iii) a protein having an amino acid sequence which is at least 90 percent or at least 95 percent or at least 98 percent homologous to SEQ ID NO:2; or (iv) _ a protein having an amino acid sequence which is at least 90 percent or at least 95 percent or at least 98 percent homologous to SEQ ID NO:4 (where homology may be determined, for example, using standard software such as BLAST or FASTA).
In further non-limiting embodiments of the invention, as an alternative to using an mda-7 nucleic acid or a MDA-7 protein as defined above, the M4 fragment of wild-type MDA-7 may be used, having the sequence of residues 104 to 206 of SEQ ID NO:2, which is identified herein as SEQ ID NO:5, or a protein which is at least 90 percent, at least 95 percent, or at least 98 percent homologous thereto (all of which are defined herein to be "M4 proteins"). A nucleic acid encoding a M4
protein is referred to as a "M4 nucleic acid" herein, and includes, but is not limited to, a nucleic acid, the sequence of which is SEQ ID NO:6.
"Mda-7 molecules" is defined herein as the class of molecules including mda-7 nucleic acids, MDA-7 proteins, M4 nucleic acids, and M4 proteins. The present invention provides for mda-7 nucleic acids or M4 nucleic acids operably linked to a promoter element and optionally comprised in an expression vector. For example, the mda-7 nucleic acid or M4 nucleic acid may be operably linked to a suitable promoter element, such as, but not limited to, the cytomegalovirus immediate early (CMV) promoter, the Rous sarcoma virus (RSV) long terminal repeat promoter, the human elongation factor 1 α promoter, the human ubiquitin c promoter, etc. It may be desirable, in certain embodiments of the invention, to use an inducible promoter. Non-limiting examples of inducible promoters include the murine mammary tumor virus promoter (inducible with dexamethasone), commercially-available tetracycline-responsive or ecdysone- responsive promoters, etc. It may also be desirable to utilize a promoter which is selectively active in the cancer cell to be treated, for example the PEG-3 gene promoter (U.S. No. 6,472,520). Examples of tissue- and cancer cell-specific promoters are well known to those of ordinary skill in the art.
Other elements that may be included in an mda-7 or M4 nucleic acid - bearing vector include transcription start sites, stop sites, polyadenylation sites, ribosomal binding sites, etc.
Suitable expression vectors include virus-based vectors and non- virus based DNA or RNA delivery systems. Examples of appropriate virus-based vectors include, but are not limited to, those derived from retroviruses, for example Moloney murine leukemia-virus based vectors such as LX, LNSX, LNCX or LXSN (Miller and
Rosman, 1989, Biotechniques 7:980-989); lentiviruses, for example human immunodeficiency virus ("HIV"), feline leukemia virus ("FIV") or equine infectious anemia virus (ΕIAV)-based vectors (Case et al, 1999, Proc. Natl. Acad. Sci. U.S.A. 96: 22988-2993; Curran et al, 2000, Molecular Ther. 1:31-38; Olsen, 1998, Gene Ther. 5:1481-1487; United States Patent Nos. 6,255,071 and 6,025, 192); adenoviruses (Zhang, 1999, Cancer Gene Ther. 6(2):113-138; Connelly, 1999, Curr. Opin. MoL Ther. i{5):565-572; Stratford-Perricaudet, 1990, Human Gene Ther. 1:241-256; Rosenfeld, 1991, Science 252:431-434; Wang et al, 1991, Adv. Exp. Med. Biol. 309:61-66; Jaffe et al, 1992, Nat. Gen. 1:372-378; Quantin et al, 1992, Proc. Natl. Acad. Sci. U.S.A. 89:2581-2584; Rosenfeld et al, 1992, Cell 68:143-155;
Mastrangeli et al, 1993, J. Clin. Invest. 91:225-234; Ragot et al, 1993, Nature 361:647-650; Hayaski et al, 1994, J. Biol. Chem. 269:23872-23875; Bett et al, 1994, Proc. Natl. Acad. Sci. U.S.A. 91:8802-8806), for example Ad5/CMV-based El- deleted vectors (Li et al, 1993, Human Gene Ther. 4:403-409); adeno-associated viruses, for example pSub201 -based AAV2-derived vectors (Walsh et al , 1992, Proc. Natl. Acad. Sci. U.S.A. .89:7257-7261); herpes simplex viruses, for example vectors based on HSV-I (Geller and Freese, 1990, Proc. Natl. Acad. Sci. U.S.A. 87:1149- 1153); baculoviruses, for example AcMNPV-based vectors (Boyce and Bucher, 1996, Proc. Natl. Acad. Sci. U.S.A. 93:2348-2352); SV40, for example SVluc (Strayer and Milano, 1996, Gene Ther. 2:581-587); Epstein-Barr viruses, for example EBV-based replicon vectors (Hambor et al, 1988, Proc. Natl. Acad. Sci. U.S.A. 85:4010-4014); alphaviruses, for example Semliki Forest virus- or Sindbis virus-based vectors (Polo et al, 1999, Proc. Natl. Acad. Sci. U.S.A. 96:4598-4603); vaccinia viruses, for example modified vaccinia virus (MVA)-based vectors (Sutter and Moss, 1992, Proc. Natl. Acad. Sci. U.S.A. 89:10847-10851) or any other class of viruses that can
efficiently transduce human tumor cells and that can accommodate the nucleic acid sequences required for therapeutic efficacy.
Non-limiting examples of non- virus-based delivery systems which may be used according to the invention include, but are not limited to, so-called naked nucleic acids (Wolff et al. , 1990, Science 247: 1465- 1468), nucleic acids encapsulated in liposomes (Nicolau et al, 1987, Methods in Enzymology 149:157-176), nucleic acid/lipid complexes (Legendre and Szoka, 1992, Pharmaceutical Research 9:1235- 1242), and nucleic acid/protein complexes (Wu and Wu, 1991, Biother. 3:87-95).
In specific, non-limiting embodiments of the invention, the expression vector is an El -deleted human adenovirus vector of serotype 5, although those of ordinary skill in the art would recognize that many of the different naturally-occurring human Ad serotypes or Ad vectors derived from non-human adenoviruses may substitute for human Ad 5 -derived vectors. In a preferred, specific, non-limiting embodiment, a recombinant replication-defective Ad.mda-7 virus for use as an mda-7 vector may be created in two steps as described in Su et al. , 1998, Proc. Natl. Acad. Sci. U.S.A. 95:14400-14405. Specifically, the coding region of the mda-7 gene may be cloned into a modified Ad expression vector pAd.CMV (Falck-Pedersen et al. , 1994, MoI. Pharmacol. 45:684-689). This vector contains, in order, the first 355 bp from the left end of the Ad genome, the CMV promoter, DNA encoding splice donor and acceptor sites, the coding region of the mda-7 cDNA, DNA encoding a polyA signal sequence from the β globin gene, and ~3 kbp of adenovirus sequence extending from within the ElB coding region. This arrangement allows high-level expression of the cloned sequence by the CMV promoter, and appropriate RNA processing. The recombinant virus may be created in vitro in 293 cells (Graham et al, 1977, J. Gen. Virol. 36:59-72) by homologous recombination between an mda-7-containing version
of pAd.CMV and plasmid pJM17, which contains the whole of the Ad genome cloned into a modified version of pBR322 (McGrory et al, 1988, Virology 163:614-617). pJM17 gives rise to Ad genomes in vivo, but they are too large to be packaged in mature Ad capsids. This constraint is relieved by recombination with the vector to create a packageable genome {Id.) containing the mda-7 gene. The recombinant virus is replication defective in human cells except 293 cells, which express adenovirus ElA and ElB. Following transfection of the two plasmids, infectious virus may be recovered, and the genomes may be analyzed to confirm the recombinant structure, and then virus may be plaque purified by standard procedures (Volkert and Young, 1983, Virology 125: 175- 193).
In a specific, non-limiting embodiment of the invention, the infectivity of an adenovirus vector carrying an mda-7 gene may be improved by inserting an Arg-Gly-Asp motif into the fiber know (Ad5-Delta24RGD), as described in Lamfers et al, 2002, Cancer Res. 62:5736-5742. MDA-7 or M4 protein may be produced in vivo in an organism or in an isolated cell which may be part of a cell population. For example, MDA-7 or M4 protein may be produced in a recombinant expression and then collected and purified using methods known in the art. Such proteins may be administered comprised in liposomes, nanoparticles, with a carrier, etc. (see below).
5.2 METHODS OF ADMINISTERING MDA-7 MOLECULES
In preferred, non-limiting embodiments, a mda-7 nucleic acid or a M4 nucleic acid may be operably linked to a suitable promoter and introduced into a cell in need of such treatment (a "target cell"), which, as defined herein, is a cell which exhibits multidrug resistance and may have been demonstrated to over express the
mdr-1 gene. In preferred embodiments, the mda-7 nucleic acid or the M4 nucleic acid may be contained in a viral vector, operably linked to a promoter element that is either inducible or constitutively active in the target cell. In preferred, non-limiting embodiments, the viral vector is a replication-defective adenovirus (as described above).
In a specific, non-limiting embodiment of the invention, a viral vector containing a nucleic acid encoding a mda-7 nucleic acid or a M4 nucleic acid in expressible form, operably linked to a suitable promoter element, may be administered to a population of target cells at a multiplicity of infection (MOI) ranging from 10- 100 MOI.
In another specific, non-limiting embodiment, the amount of a viral vector administered to a subject, preferably a human subject, may be 1 X 109 pfu to 1 X 1012 pfu.
In specific, non-limiting embodiments, a mda-7 nucleic acid or a M4- nucleic acid, preferably operably linked to a promoter element, comprised in a vector or otherwise, may be introduced into a cell ex vivo and then the cell may be introduced into a subject. For example, a nucleic acid encoding mda-7 may be introduced into a cell of a subject ex vivo and then the cell containing the nucleic acid may be optionally propagated and then (with its progeny) introduced into the subject. Alternatively, a MDA-7 protein or a M4 protein may be used in protein/peptide therapy of a subject in need of such treatment. As such, the MDA-7 protein or M4 protein may be prepared by chemical synthesis or recombinant DNA techniques, purified by methods known in the art, and then administered to a subject in need of such treatment. MDA-7 protein or M4 protein may be comprised, for example, in solution, in suspension, and/or in a carrier particle such as microparticles,
liposomes, or other protein-stabilizing formulations known in the art In a non- limiting specific example, formulations of MDA-7 protein or M4 protein may stabilized by addition of zinc and/or protamine stabilizers as in the case of certain types of insulin formulations. Alternatively, in specific non-limiting embodiments, a MDA-7 protein or a M4 protein may be linked covalently or non-covalently, to a carrier protein which is preferably non-immunogenic.
In preferred, non-limiting embodiments, a MDA-7 protein or a M4 protein is administered in an amount which achieves a local concentration in the range of 18 to 50 ng per microliter. For example, a subject may be administered a range of 50- 100 mg of MDA-7 protein or M4 protein per kilogram. For a human subject, the dose range may be between 100-2500 mg/day of MDA-7 protein or M4 protein.
Any of the foregoing molecules may be administered by direct instillation, injection (intravenous, subcutaneous, intramuscular, intraarterial, intrathecal, intrahepatic, intratumoral), or any other method or route known in the art.
5.3 USE OF MDA-7 TO IMPROVE RESPONSE TO CHEMOTHERAPY
The present invention relates to a method for improving the therapeutic response of a subject suffering from a disorder to chemotherapy, where the disorder in the subject had previously been resistant to chemotherapy and/or where the mdr-1 gene and/or MDR-I protein are shown to be over-expressed, comprising administering, to the subject, an effective amount of a mda-7 nucleic acid or a MDA-7 protein or a M4 nucleic acid or a M4 protein, in conjunction with one or more chemotherapeutic agent.
The subject may be a human or a non-human subject, but preferably is a human subject.
The subject may be suffering from a disorder which is a malignancy or a non-malignant disorder.
Non-limiting examples of malignant disorders include, but are not limited to, any malignancy that exhibits multi-drug resistance, and that preferably has been demonstrated to over-express mdr-1 gene and/or MDR-I protein (also referred to as "P-glycoprotein" or "P-gp" herein). Such malignancies may include, but not be limited to, breast cancer, ovarian cancer, lung cancer (e.g., adenocarcinoma, small- cell carcinoma, non-small cell carcinoma, mesothelioma), colorectal cancer, renal carcinoma, chronic lymphocytic leukemia, acute lymphocytic leukemia, Hodgkin's lymphoma, non-Hodgkin's lymphoma, neuroblastoma, glioblastoma, prostate cancer, pancreatic cancer, gastric cancer, squamous cell carcinoma (e.g., oral squamous cell carcinoma and squamous cell carcinoma of the head and neck), melanoma, hepatic adenocarcinoma, bladder cancer, testicular cancer, and rhabdosarcoma.
In alternative embodiments, subjects for treatment according to the invention include any subject suffering from a non-malignant disease, where the response of the disease to therapy is negatively impacted by multi-drug resistance, where the subject has preferably been demonstrated to over express mdr-1 gene and/or MDR-1 protein. Such subjects may include, but not be limited to, subjects suffering from a viral infection, such as, but not limited to, human immunodeficiency infection, hepatitis B virus infection, Epstein-Barr virus infection, Herpes simplex virus infection, human papilloma virus infection, or hepatitis C infection. In other embodiments, the subject may be suffering from an autoimmune disease, such as, but not limited to, rheumatoid arthritis, juvenile rheumatoid arthritis, system lupus erythematosis, Sjogren's syndrome, scleroderma, polymyositis, or diabetes mellitus.
In another non-limiting embodiment, the subject may be suffering from multiple sclerosis.
The nature of the therapeutic response to be improved depends on the nature of the disorder being treated. Where the disorder is a malignant disorder, an improved therapeutic response may be characterized by, for example and not by way of limitation, one or more of the following: higher rate of survival, longer life expectancy, longer period to relapse, increased comfort, decreased tumor burden, decreased pain, decreased rate or incidence of metastasis. Where the disorder is an infectious disorder, an improved therapeutic response may be characterized by, for example and not by way of limitation, one or more of the following: increased survival, longer life expectancy, decreased viral titer, increased CD4+ T cell count, decreased incidence of secondary infection, decreased incidence of associated carcinoma. Where the disorder is an autoimmune disorder, an improved therapeutic response may be characterized by, for example and not by way of limitation, decreased serologic markers of inflammation such as C-reactive protein and erythrocyte sedimentation rate, decreased clinical inflammation, (for arthritis) increased range of motion, decreased pain, longer disease-free interval, decreased need for pain medication or insulin therapy, increased life expectancy, decreased workplace absenteeism. Where the disorder is multiple sclerosis, an improved therapeutic response may be characterized by, for example and not by way of limitation, decreased incidence of relapse, longer relapse-free intervals, shorter duration of relapse, decreased severity of symptoms at relapse, decreased lesions by imaging studies such as MRI, and improved nerve conduction studies.
"Resistance to chemotherapy" means that the subject had been administered one or more chemotherapeutic agent which did not achieve a
satisfactory therapeutic response (examples of types of therapeutic responses being set forth above).
"Over-expression of mdr-1 gene or protein" means an increase in the level of mdr-1 gene transcription and/or MDR-I protein levels. In non-limiting example, a sample of cells afflicted by the disorder may be collected from the subject and then tested for levels of mdr-1 mRNA and/or MDR-1 protein, using techniques known in the art, such as PCR analysis, Northern Blot, Western blot, immunohistochemistry, etc. The sequence of the human MDR-1 gene and protein are known. The Genbank accession numbers for mdr-1 nucleic acid and MDR-1 protein are M 14758 and P08183 respectively. "Over-expressing" means an increase relative to a normal control cell, preferably by at least about 10, at least about 20, at least about 30, at least about 40, at least about 50, at least about 60, at least about 70, at least about 80, or at least about 90 percent.
A subject need not be tested for over-expression of Mdr-1 molecules if they have proved to be clinically refractory to treatment. Conversely, a patient need not have been demonstrated to be refractory to treatment if over-expression of mdr-1 gene or MDR-1 protein is demonstrated.
Administration of a mda-7 nucleic acid or a MD A-7 protein or a M4 nucleic acid or a M4 protein may be performed as set forth in the section above. Concurrent administration of one or more chemotherapeutic agent means that the Mda-7 molecule and the one or more chemotherapeutic agent are administered close enough in time to each other for the clinical effect of the Mda-7 molecule may be exerted on the chemotherapeutic agent. The Mda-7 molecule (mda- 7 nucleic acid, MDA-7 protein, M4 nucleic acid or M4 protein) may be administered simultaneously with, prior to, or after the administration of the one or more
chemotherapeutic agent (preferably prior to administration of the one or more chemotherapeutic agent). If a mda-7 nucleic acid or a M4 nucleic acid is administered by gene therapy, the time period between administering it and the chemotherapeutic agent may be quite long, and one administration of gene therapy may suffice for a plurality of subsequent chemotherapeutic treatments. If MDA-7 protein or M4 protein is administered, the interval to administration of the one or more chemotherapeutic agent would be shorter in view of degradation of the protein.
Non-limiting specific examples of intervals between administration of an Mda-7 molecule and one or more therapeutic agent include up to -5 years ("-" means prior administration of the Mda-7 molecule), up to -3 years, up to -2 years, up to -1 year, up to -12 months, up to -11 months, up to -10 months, up to -9 months, up to -8 months, up to -7 months, up to -6 months, up to -3 months, up to -1 month, up to - 2 weeks, up to -1 week, up to -6 days, up to -5 days, up to -4 days, up to -3 days, up to -2 days, up to -1 day, up to -12 hours, up to -8 hours, up to -4 hours, up to -2 hours, up to -1 hours, 0 hours, up to +1 hour ("+" means that Mda-7 molecules is administered after chemotherapeutic agent (s)), up to +2 hours, up to +4 hours, up to +8 hours, up to +12 hours, up to +1 day, up to +2 days, up to +3 days, up to +4 days, up to +5 days, up to +6 days, up to +1 week, up to +2 weeks, up to +1 month, up to +3 months, up to +6 months, up to +7 months, up to +8 months, up to +9 months, up to +10 months, up to +11 months, up to +1 year, up to + 3 years, and up to + 5 years.
The one or more chemotherapeutic agent selected is related to the nature of the disorder being treated.
Where the disorder is a malignancy, the one or more chemotherapeutic agent may be, for example and not by way of limitation, an alkylating agent such as busulfan, cisplatin, carboplatin, chlorambucil, cyclophosphamide, ifosfamide,
dacarbazine, mechlorethamine (nitrogen mustard), melphalan, and temozolomide; a nitrosourea such as streptozocin, carmustine, and lomustine, an antimetabolite such as 5-fluorouracil, capecitabine, 6-mercaptopurine, methotrexate, gemcitabine, cytarabine, fludarabine, and pemetrexed; an anthracyline-like drug such as daunorubicin, doxorubicin, epirubicin, idarubicin, and mitoxantrone; a topoisomerase I inhibitor such as topotecan and irinotecan; a topoisomerase II inhibitor such as etoposide and teniposide; or a mitotic inhibitor such as, for example, a taxane such as paclitaxel or docetaxel, or a vinca alkaloid such as vinblastine, vincristine, or vinorelbine, or a combination of two or more of the above. Where the disorder is human immunodeficiency virus infection, the one or more chemotherapeutic agent may be, for example and not by way of limitation, a reverse transcriptase inhibitor, including nucleoside reverse transcriptase inhibitors such as 3TC (lamivudine), abacavir, AZT (zidovudine), d4T (stavudine), ddC (zalcitabine), ddl (didanosine) and FTC (emtricitabine) and non-nucleoside reverse transcriptase inhibitors such as efavirenz, delavirdine, neviripine, etravirine, rilpirivine ; a protease inhibitor such as saquinivir, nelfinivir, ritonivir, indinivir, amprenavir, lopinavir, atazanavir, fosamprenavir, or tipranavir; a fusion inhibitor such enfuvirtide as or an integrase inhibitor such as Merck's L-000870810 or Japan Tobacco's JTK-303, or combinations thereof. Where the disorder is a herpes virus infection, the one or more chemotherapeutic agent may be, for example and not by way of limitation, aciclovir, cidofovir, docosanol, famiciclovir, fomivirsen, foscarnet, ganciclovir, idoxuridine, penciclovir, trifluridine, tromantidine, valaciclovir, valganciclovir, or vidarabine, or combinations thereof.
Where the disorder is hepatitis B infection, the one or more chemotherapeutic agent may be, for example and not by way of limitation, adefovir or lamivudine, or a combination thereof.
Where the disorder is hepatitis C infection, a chemotherapeutic agent may be, for example and not by way of limitation, ribivirin.
Where the disorder is multiple sclerosis, the one or more chemotherapeutic agent may be glatiramir or novantrone or a combination thereof.
5.4 USE OF MDA-7 THERAPY IN MDRl -OVER-EXPRESSING SUBJECTS In addition, in alternative embodiments, the present invention provides for a method of treating a subject suffering from a cancer found to be resistant to chemotherapy and/or where the mdr-1 gene and/or MDR-I protein are shown to be over-expressed comprising administering, to the subject, an effective amount of a Mda-7 molecule, for example mda-7 nucleic acid, MDA-7 protein, M4 nucleic acid or M4 protein. This aspect is based, at least in part, on the discovery that the anti-cancer effect of the melanoma differentiation associated gene -7 (mda-7) was increased in the context of over-expression of the multidrug resistance (MDRI) gene. The scope of the various terms used in this paragraph is as set forth in the preceding section. However, according to this embodiment, the Mda-7 molecule need not be administered in conjunction with one or more chemotherapeutic agent.
In one non-limiting set of embodiments, the present invention provides for a method of treating a subject suffering from a cancer, comprising (i) determining that cancer cells of the subject over express the mdr-1 gene and/or MDR-I protein; and (ii) administering, to the subject, an effective amount of an Mda-7 molecule (as described above).
In another non-limiting set of embodiments, the present invention provides for a method of evaluating the likelihood that a subject, suffering from a cancer, would benefit from therapy with a Mda-7 molecule, comprising determining whether or not cancer cells in the subject over-express the mdr-1 gene or MDR-I protein (Mdr-1 molecules), wherein over-expression of mdr-1 gene and/or MDR-I protein is consistent with a greater likelihood that the subject would benefit from
Mda-7 molecule therapy.
6. EXAMPLE
6.1 RESULTS
The experiments described in this section were carried out using two related colorectal carcinoma cell lines, SW620/WT and its doxorubicin (Dox)- resistant counterpart, SW620/DOX. Initial experiments were designed to establish a baseline phenotype for these cell lines. FIGURE IA depicts the results of Western blot analysis of endogenous MDR- 1 ("P-gp") levels in S W620/WT and S W620/DOX cells, and shows that the SW620/DOX cell line over expressed P-gp. FIGURE IB depicts the effect of different doses of Dox on cell viability in SW620/WT and S W620/DOX cells. Cells were cultured in the presence of different concentrations of
Dox (0.25, 1, 4, 16, and 64 μM) for 96 hours, and then the effects on cell growth were
determined by MTT assay. Graphs show the average ± SD of results of 3 independent experiments, and demonstrate that Dox had substantially less effect on the viability of SW620/DOX cells. FIGURE 1C particularly addresses the ability of Dox to induce apoptosis in SW620/WT and SW620/Dox cells, based on the levels of Annexin V-
binding. Cells were treated with 0.25, 1, 4, 16, and 64 μM of Dox and were stained
after 48 h with APC-labeled Annexin V and immediately analyzed by flow cytometry.
The percentage of early and late apoptotic cells (only Annexin V stained) was calculated using Flow Jo Version 6.31 software. FIGURE 1C shows that the level of Annexin V binding as a result of Dox treatment was greater in SW620/WT cells, and the effect was dose-dependent. Next, the effect of MDA-7 on the responsiveness of S W620/WT and
SW620/DOC cells was explored. Experiments were performed in which SW620 and SW620/Dox cells were infected with either (empty vector) Ad.vec or Ad.mda7 (25 pfu/cell) and then treated with the indicated concentrations of Dox for 48 h. The percentages of apoptotic cells were determined by Annexin V binding assay and flow cytometry. As shown in FIGURE 2A, the S W620/DOX cells infected with Ad.vec remained relatively resistant to Dox, but SW620/DOX cells infected with Ad.mda7 exhibited a sensitivity to Dox that approached that of S W620/WT. When cells were infected with either Ad.vec or Ad.mda7 (25 pfu/cell), treated with Dox for 24 h., and then total proteins were extracted and subjected to Western blot analysis, Ad.mda7 infection was found to down regulate P-gp expression in SW620/Dox cells (FIGURE 2B). As shown in FIGURE 2C, the intracellular accumulation of doxorubicin, determined after cells were infected with either Ad.vec or Ad.mda7 (25 pfu/cell) for 24 h, incubated with 2 or 4 μM of Dox for 60 min., washed twice with ice-cold PBS, resuspended in 400 μl PBS and then analyzed by flow cytometry, was increased in Ad.mda7-infected SW620/Dox cells. Thus, FIGURE 2A-C demonstrates that Ad.mda7 infection induces MDR-I reversal in SW620/Dox cells.
The ability of Ad.mda7 infection to increase Dox accumulation in SW620/DOX cells is further supported by FIGURE 3. Cells were incubated with 10 μM doxorubicin at 370C for 30 min (substrate loading phase) and then washed twice with PBS. Cells were then infected with either Ad.vec or Ad.mda7 at the indicated
MOI. At the defined time points, cells were harvested, centrifuged and washed in ice- cold PBS. Cell pellets were then resuspended in 400 μl PBS and used immediately for flow cytometric analysis for intracellular doxorubicin retention.
Experiments were then performed to determine the effects of Ad.mda7 infection on viability and apoptosis induction of S W620/WT and S W620/DOX cells independent of Dox. SW620 and SW620/Dox cells were infected with either Ad.vec or Ad.mda7 at MOIs of either 25 or 50 pfu/cell, and then either viability was determined by MTT assay 5 days after infection or apoptosis induction was evaluated by staining infected cells after 48 h with APC-labeled Annexin V followed immediately by flow cytometry analysis. The percentage of early and late apoptotic cells (only Annexin V stained) was calculated using FlowJo Version 6.31 software. As shown in FIGURE 4A-B, Ad.mda7 infection inhibited cell growth and induced apoptosis preferentially in P-gp-expressing mdr-1 variant SW620/Dox cells. Interestingly, as shown in FIGURE 4C, Western blot analysis showed that MDA- 7/IL-24 protein expression was increased in S W620/DOX relative to S W620/WT.
To explore the role of mdr-1 in the enhanced effectiveness of Ad.mda7 in SW620/DOX cells, SW620/WT cells were transiently transfected with either empty vector (pCDNA3.1) or MDR, infected with either Ad.vec or Ad.mda7, and then apoptosis induction was evaluated by Annexin V binding assay 48 h after infection. The percentage of early and late apoptotic cells (only Annexin V stained) was calculated using FlowJo Version 6.31 software. As shown in FIGURE 5 A. over- expression of MDR was observed to induce increased sensitivity to Ad.mda7 infection in SW620 cells. As shown in FIGURE 5B, Western blot analysis performed on cell lysates harvested at 24 h after infection indicated that over-expression of mdr- 1 /MDR-1 was associated with enhanced expression of MDA-7/IL-24 protein.
Further, when SW620/WT cells were transiently transfected either with empty vector (pCDNA3.1) or mdr-1, infected with either Ad.vec or Ad.mda7 and, 24 hours later, 1 x 105 cells were replated in 0.4% agar on 0.8% base agar and then, after two weeks, colonies >50 cells were counted under a dissection microscope, it was found that ectopic expression of mdr-l/MDR-1 in SW620 cells significantly inhibited colony formation in soft agar (FIGURE 5C).
To further explore this phenomenon, the effect of down-regulation of mdr-1 in SW620/DOX cells was evaluated. SW620/DOX cells were transiently transfected with either control siRNA or mdr-1 siRNA using lipofectamine prior to infection with either Ad.vec or Ad.mda7. Forty-eight hours later, apoptosis was measured by Annexin V binding assay using APC-conjugated Annexin V by flow cytometry. The percentage of early and late apoptotic cells (only Annexin V stained) was calculated using FlowJo Version 6.31 software. As shown in FIGURE 6, down regulation of mdr-1 partially protected SW620/DOX cells from Ad.mda7-induced apoptosis.
Finally, K-ras localization was studied in SW620/WT and SW620/DOX cells. FIGURE 7 shows confocal images of SW620/WT and SW620/DOX cells immunofluorescently stained with anti-k-Ras antibody. Note extensive plasma-membrane localization in the SW620/WT cell images (typical pattern) and surface distribution in the SW620/DOX cell images.
6.2 DISCUSSION
Inappropriate expression of the multidrug resistance (mdr-1) gene, encoding the P-glycoprotein (P-gp), is frequently implicated in resistance of cancer cells to diverse chemotherapeutic drugs. However, high toxicity of P-gp inhibitors
mandates alternative approaches for modulating mdr-1 and promoting death in drug- resistant cancer cells. In this study, the effect of melanoma differentiation-associated gene-7/interleukin-24 (mda-7/IL-24), which exhibits cancer-specific apoptosis- inducing properties, was studied in drug-sensitive (SW620/WT) and drug-resistant (SW620/DOX) colorectal carcinoma cell lines. mda-7/IL-24, when administered by means of a replication-incompetent adenovirus, Ad.mda-7, at a multiplicity of infection of 25-50 pfu/cell, significantly increased the sensitivity of SW620/DOX cells to doxorubicin. However, the cytotoxic effect of doxorubicin in the SW620/WT sensitive cell line was not significantly altered by Ad.mda-7 infection. Annexin V binding assays revealed that Ad.mda-7 infection effectively reversed resistance to doxorubicin-induced apoptosis in SW620/DOX cells. In addition, infection of SW620/DOX cells with Ad.mda-7 increased intracellular accumulation of doxorubicin and significantly decreased P-gp expression. Surprisingly, we observed that P-gp over expressing cells (SW620/DOX) displayed increased apoptosis following Ad.mda-7-infection compared to parental SW620/WT cells. Western blot analysis indicated more MDA-7/IL-24 protein in SW620/DOX than SW620/WT cells following infection with Ad.mda-7, which might explain the increased sensitivity of P-gp over expressing cells to Aά.mda-7 infection. Furthermore, over-expression of mdr-1 in SW620/WT cells increased apoptosis and MDA-7/IL-24 protein following Ad.mda-7 infection, while down modulation of mdr-1 in SW620/DOX cells by siRNA led to decreased apoptosis and MDA-7/IL-24 protein following Ad.mda-7 infection. These findings reveal that mda-7/IL-24 is a potent MDR reversal agent, preferentially causing apoptosis in P-gp over expressing MDR cells, suggesting significant clinical implications for the use of mda-7/IL-24 in treating neoplasms that have failed chemotherapy mediated by the P-gp multidrug resistance mechanism.
Various references are cited herein, the contents of which are hereby incorporated by reference in their entireties.
SEQUENCE LISTING
SEQ IDNO:1 nucleotides 275 to 895 ofGenBankAccessionNo. U16261 atgaattttc aacagaggct gcaaagcctg tggactttag ccagaccctt ctgccctcct 60
ttgctggcga cagcctctca aatgcagatg gttgtgctcc cttgcctggg ttttaccctg 120
cttctctgga gccaggtatc aggggcccag ggccaagaat tccactttgg gccctgccaa 180
gtgaaggggg ttgttcccca gaaactgtgg gaagccttct gggctgtgaa agacactatg 240
caagctcagg ataacatcac gagtgcccgg ctgctgcagc aggaggttct gcagaacgtc 300
tcggatgctg agagctgtta ccttgtccac accctgctgg agttctactt gaaaactgtt 360
ttcaaaaact accacaatag aacagttgaa gtcaggactc tgaagtcatt ctctactctg 420
gccaacaact ttgttctcat cgtgtcacaa ctgcaaccca gtcaagaaaa tgagatgttt 480
tccatcagag acagtgcaca caggcggttt ctgctattcc ggagagcatt caaacagttg 540
gacgtagaag cagctctgac caaagccctt ggggaagtgg acattcttct gacctggatg 600
cagaaattct acaagctctg a 621
SEQ ID NO:2 = amino acid sequence of complete wild-type MDA-7 protein (GenBank Accession No. U 16261)
MNFQQRLQSL WTLARPFCPP LLATASQMQM WLPCLGFTL LLWSQVSGAQ GQEFHFGPCQ 60
VKGWPQKLW EAFWAVKDTM QAQDNITSAR LLQQEVLQNV SDAESCYLVH TLLEFYLKTV 120
FKNYHNRTVE VRTLKSFSTL ANNFVLIVSQ LQPSQENEMF SIRDSAHRRF LLFRRAFKQL 180
DVEAALTKAL GEVDILLTWM QKFYKL 206
SEQ ID NO: 3 = nucleic acid sequence encoding the portion of wild-type MDA-7 lacking the signal sequence atgcagggcc aagaattcca ctttgggccc tgccaagtga agggggttgt tccccagaaa 60
ctgtgggaag ccttctgggc tgtgaaagac actatgcaag ctcaggataa catcacgagt 120
gcccggctgc tgcagcagga ggttctgcag aacgtctcgg atgctgagag ctgttacctt 180
gtccacaccc tgctggagtt ctacttgaaa actgttttca aaaactacca caatagaaca 240
gttgaagtca ggactctgaa gtcattctct actctggcca acaactttgt tctcatcgtg 300
tcacaactgc aacccagtca agaaaatgag atgttttcca tcagagacag tgcacacagg 360
cggtttctgc tattccggag agcattcaaa cagttggacg tagaagcagc tctgaccaaa 420
gcccttgggg aagtggacat tcttctgacc tggatgcaga aattctacaa gctc 474
SEQ ID NO:4 = amino acid sequence of wild-type MDA-7 protein lacking the signal sequence
MQEFHFGPCQ VKGWPQKLW EAFWAVKDTM QAQDNITSAR LLQQEVLQNV SDAESCYLVH 60
TLLEFYLKTV FKNYHNRTVE VRTLKSFSTL ANNFVLIVSQ LQPSQENEMF SIRDSAHRRF 120
LLFRRAFKQL DVEAALTKAL GEVDILLTWM QKFYKL 156
SEQ ID NO:5 = amino acid sequence of M4; residues 104 to 206 of SEQ ID NO:2.
MESCYLVHTL LEFYLKTVFK NYHNRTVEVR TLKSFSTLAN NFVLIVSQLQ PSQENEMFSI 60
RDSAHRRFLL FRRAFKQLDV EAALTKALGE VDILLTWMQK FYKL 104
SEQ IDNO:6 =portionofSEQ IDNO:1 encoding M4 atggagagct gttaccttgt ccacaccctg ctggagttct acttgaaaac tgttttcaaa 60
aactaccaca atagaacagt tgaagtcagg actctgaagt cattctctac tctggccaac 120
aactttgttc tcatcgtgtc acaactgcaa cccagtcaag aaaatgagat gttttccatc 180
agagacagtg cacacaggcg gtttctgcta ttccggagag cattcaaaca gttggacgta 240
gaagcagctc tgaccaaagc ccttggggaa gtggacattc ttctgacctg gatgcagaaa 300
ttctacaagc tctga 315
Claims
1. A method for improving the therapeutic response to chemotherapy of a subject suffering from a disorder, comprising (i) demonstrating that the mdr-1 gene and/or MDR-I protein are over-expressed; and (ii) administering, to the subject, an effective amount of a Mda-7 molecule in conjunction with one or more chemotherapeutic agent.
2. The method of claim 1 wherein the Mda-7 molecule is a mda-7 nucleic acid.
3. The method of claim 1 wherein the Mda-7 molecule is a MDA-7 protein.
4. The method of claim 1 wherein the Mda-7 molecule is a M4 nucleic acid.
5. The method of claim 1 wherein the Mda-7 molecule is a M4 protein.
6. A method of treating a subject suffering from a cancer found to be resistant to chemotherapy comprising (i) demonstrating that the mdr-1 gene and/or MDR-I protein are over-expressed and (ii) administering, to the subject, an effective amount of a Mda-7 molecule.
7. The method of claim 6 wherein the Mda-7 molecule is a mda-7 nucleic acid.
8. The method of claim 6 wherein the Mda-7 molecule is a MDA-7 protein.
9. The method of claim 6 wherein the Mda-7 molecule is a M4 nucleic acid.
10. The method of claim 6 wherein the Mda-7 molecule is a M4 protein.
Applications Claiming Priority (2)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
US89406807P | 2007-03-09 | 2007-03-09 | |
US60/894,068 | 2007-03-09 |
Publications (1)
Publication Number | Publication Date |
---|---|
WO2008112470A1 true WO2008112470A1 (en) | 2008-09-18 |
Family
ID=39759917
Family Applications (1)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
PCT/US2008/055877 WO2008112470A1 (en) | 2007-03-09 | 2008-03-05 | Increased effectiveness of mda-7 with mdr-1 over-expression |
Country Status (1)
Country | Link |
---|---|
WO (1) | WO2008112470A1 (en) |
Cited By (2)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
WO2014197535A1 (en) * | 2013-06-04 | 2014-12-11 | Virginia Commonwealth University | Recombinant cancer therapeutic cytokine |
WO2017173101A1 (en) * | 2016-03-30 | 2017-10-05 | Virginia Commonwealth University | Combination therapy for neuroblastoma using mda-7/il-24 with therapeutic agents |
Citations (2)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US20030119732A1 (en) * | 2000-11-28 | 2003-06-26 | Board Of | CDDO-compounds and combination therapies thereof |
US20060292157A1 (en) * | 2004-12-02 | 2006-12-28 | The Trustees Of Columbia University In The City Of New York | MDA-7 protein variants having antiproliferative activity |
-
2008
- 2008-03-05 WO PCT/US2008/055877 patent/WO2008112470A1/en active Application Filing
Patent Citations (2)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US20030119732A1 (en) * | 2000-11-28 | 2003-06-26 | Board Of | CDDO-compounds and combination therapies thereof |
US20060292157A1 (en) * | 2004-12-02 | 2006-12-28 | The Trustees Of Columbia University In The City Of New York | MDA-7 protein variants having antiproliferative activity |
Cited By (3)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
WO2014197535A1 (en) * | 2013-06-04 | 2014-12-11 | Virginia Commonwealth University | Recombinant cancer therapeutic cytokine |
US9951114B2 (en) | 2013-06-04 | 2018-04-24 | Virginia Commonwealth University | Recombinant cancer therapeutic cytokine |
WO2017173101A1 (en) * | 2016-03-30 | 2017-10-05 | Virginia Commonwealth University | Combination therapy for neuroblastoma using mda-7/il-24 with therapeutic agents |
Similar Documents
Publication | Publication Date | Title |
---|---|---|
JP5807236B2 (en) | Improved gene delivery system of tumor specific expression with recombined gene expression regulatory sequences | |
Roth et al. | Gene therapy for cancer: what have we done and where are we going? | |
KR100812631B1 (en) | Recombinant 4553 Adenovirus Preparation and Composition | |
US20070293453A1 (en) | Combinatorial Methods For Inducing Cancer Cell Death | |
US20080213220A1 (en) | Cancer-targeted viral vectors | |
KR20000075924A (en) | Adenovirus vectors specific for cells expressing alpha-fetoprotein and methods of use thereof | |
Willis et al. | The promise and obstacle of p53 as a cancer therapeutic agent | |
JP2020503390A (en) | Fusogenic lipid nanoparticles, and methods for making and using the same, for target cell-specific production of therapeutic proteins and for treating diseases, conditions, or disorders associated with target cells | |
AU2003206815A2 (en) | Adenoviral vectors for modulating the cellular activities associated with PODs | |
Balicki et al. | Gene therapy of human disease | |
CN1968717B (en) | Gene delivery system containing relaxin gene and pharmaceutical composition using relaxin | |
KR20210089135A (en) | Recombinant replication-competent virus comprising a coding region for glycogen synthase kinase-3 (GSK3) and a method for killing abnormal cells | |
KR101253276B1 (en) | Anti-tumor Compositions for Combined Gene Therapy | |
Cusack Jr et al. | Cancer gene therapy | |
WO2008112470A1 (en) | Increased effectiveness of mda-7 with mdr-1 over-expression | |
Palacios et al. | Mitochondrially targeted wild-type p53 induces apoptosis in a solid human tumor xenograft model | |
KR101399062B1 (en) | Anti-cancer Compositions Containing Wnt Decoy Receptor | |
Guo et al. | Recombinant adenovirus expressing a dendritic cell-targeted melanoma surface antigen for tumor-specific immunotherapy in melanoma mice model | |
Su et al. | Etoposide enhances antitumor efficacy of MDR1-driven oncolytic adenovirus through autoupregulation of the MDR1 promoter activity | |
KR101909905B1 (en) | Gene Expression System for Targeting Cancer Stem Cell and Cancer Cell | |
US20010044420A1 (en) | Combination use of gemcitabine and tumor suppressor gene therapy in the treatment of neoplasms | |
US20080293652A1 (en) | Combination of Ad-P53 and Chemotherapy for the Treatment of Tumours | |
CN102108356B (en) | Novel promoter and virus vector containing same | |
TWI391486B (en) | A novel promoter and viral vector containing the same | |
AU2002331641A1 (en) | Combinatorial methods for inducing cancer cell death |
Legal Events
Date | Code | Title | Description |
---|---|---|---|
121 | Ep: the epo has been informed by wipo that ep was designated in this application |
Ref document number: 08731415 Country of ref document: EP Kind code of ref document: A1 |
|
NENP | Non-entry into the national phase |
Ref country code: DE |
|
122 | Ep: pct application non-entry in european phase |
Ref document number: 08731415 Country of ref document: EP Kind code of ref document: A1 |