WO1997039776A9 - Gene therapy involving concurrent and repeated administration of adenoviruses and immunosuppressive agents - Google Patents
Gene therapy involving concurrent and repeated administration of adenoviruses and immunosuppressive agentsInfo
- Publication number
- WO1997039776A9 WO1997039776A9 PCT/US1997/006199 US9706199W WO9739776A9 WO 1997039776 A9 WO1997039776 A9 WO 1997039776A9 US 9706199 W US9706199 W US 9706199W WO 9739776 A9 WO9739776 A9 WO 9739776A9
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- vector
- administration
- mice
- cells
- adenoviral
- Prior art date
Links
- 238000001415 gene therapy Methods 0.000 title claims abstract description 22
- 241000701161 unidentified adenovirus Species 0.000 title description 54
- 239000003018 immunosuppressive agent Substances 0.000 title description 42
- 229940125721 immunosuppressive agent Drugs 0.000 title description 37
- 239000013598 vector Substances 0.000 claims abstract description 241
- 229960002706 gusperimus Drugs 0.000 claims abstract description 70
- IDINUJSAMVOPCM-INIZCTEOSA-N n-[(1s)-2-[4-(3-aminopropylamino)butylamino]-1-hydroxy-2-oxoethyl]-7-(diaminomethylideneamino)heptanamide Chemical compound NCCCNCCCCNC(=O)[C@H](O)NC(=O)CCCCCCN=C(N)N IDINUJSAMVOPCM-INIZCTEOSA-N 0.000 claims abstract description 70
- CMSMOCZEIVJLDB-UHFFFAOYSA-N Cyclophosphamide Chemical compound ClCCN(CCCl)P1(=O)NCCCO1 CMSMOCZEIVJLDB-UHFFFAOYSA-N 0.000 claims abstract description 60
- 229960004397 cyclophosphamide Drugs 0.000 claims abstract description 60
- 108091028043 Nucleic acid sequence Proteins 0.000 claims abstract description 29
- 238000011282 treatment Methods 0.000 claims abstract description 24
- 239000003814 drug Substances 0.000 claims abstract description 17
- 229940124597 therapeutic agent Drugs 0.000 claims abstract description 14
- 238000000034 method Methods 0.000 claims abstract description 11
- 230000028993 immune response Effects 0.000 claims description 11
- 230000001225 therapeutic effect Effects 0.000 claims description 11
- 230000014509 gene expression Effects 0.000 abstract description 33
- 241000699670 Mus sp. Species 0.000 description 112
- 210000004027 cell Anatomy 0.000 description 77
- 108090000623 proteins and genes Proteins 0.000 description 64
- 230000001506 immunosuppresive effect Effects 0.000 description 41
- 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 40
- 206010062016 Immunosuppression Diseases 0.000 description 39
- 229960003957 dexamethasone Drugs 0.000 description 39
- 229960000027 human factor ix Drugs 0.000 description 37
- 239000007924 injection Substances 0.000 description 34
- 238000002347 injection Methods 0.000 description 34
- 241000700159 Rattus Species 0.000 description 32
- 230000003472 neutralizing effect Effects 0.000 description 24
- 210000004072 lung Anatomy 0.000 description 23
- 239000013612 plasmid Substances 0.000 description 23
- 230000004044 response Effects 0.000 description 23
- 239000002245 particle Substances 0.000 description 20
- 108020004414 DNA Proteins 0.000 description 19
- 239000012634 fragment Substances 0.000 description 19
- 102100022641 Coagulation factor IX Human genes 0.000 description 16
- 230000003263 anti-adenoviral effect Effects 0.000 description 16
- 108010076282 Factor IX Proteins 0.000 description 15
- 229960004222 factor ix Drugs 0.000 description 15
- 238000012546 transfer Methods 0.000 description 15
- 238000002965 ELISA Methods 0.000 description 14
- 108700019146 Transgenes Proteins 0.000 description 14
- 241001135569 Human adenovirus 5 Species 0.000 description 13
- 230000005875 antibody response Effects 0.000 description 13
- 210000001151 cytotoxic T lymphocyte Anatomy 0.000 description 13
- 208000015181 infectious disease Diseases 0.000 description 13
- 230000036470 plasma concentration Effects 0.000 description 12
- 241000699666 Mus <mouse, genus> Species 0.000 description 11
- 239000012981 Hank's balanced salt solution Substances 0.000 description 10
- 102000036639 antigens Human genes 0.000 description 9
- 108091007433 antigens Proteins 0.000 description 9
- 230000001419 dependent effect Effects 0.000 description 9
- 102000004169 proteins and genes Human genes 0.000 description 9
- 150000003431 steroids Chemical class 0.000 description 9
- 230000001629 suppression Effects 0.000 description 9
- 241000700605 Viruses Species 0.000 description 8
- 230000003321 amplification Effects 0.000 description 8
- 239000000427 antigen Substances 0.000 description 8
- 239000003795 chemical substances by application Substances 0.000 description 8
- 230000001404 mediated effect Effects 0.000 description 8
- 238000003199 nucleic acid amplification method Methods 0.000 description 8
- 101000911390 Homo sapiens Coagulation factor VIII Proteins 0.000 description 7
- 241000714474 Rous sarcoma virus Species 0.000 description 7
- 238000010276 construction Methods 0.000 description 7
- 230000006801 homologous recombination Effects 0.000 description 7
- 238000002744 homologous recombination Methods 0.000 description 7
- 102000057593 human F8 Human genes 0.000 description 7
- 229960000900 human factor viii Drugs 0.000 description 7
- 238000001727 in vivo Methods 0.000 description 7
- 238000001990 intravenous administration Methods 0.000 description 7
- 210000004698 lymphocyte Anatomy 0.000 description 7
- 230000002685 pulmonary effect Effects 0.000 description 7
- 210000003462 vein Anatomy 0.000 description 7
- 238000011740 C57BL/6 mouse Methods 0.000 description 6
- 108010054218 Factor VIII Proteins 0.000 description 6
- 102000001690 Factor VIII Human genes 0.000 description 6
- 108090000331 Firefly luciferases Proteins 0.000 description 6
- 230000024932 T cell mediated immunity Effects 0.000 description 6
- 238000003556 assay Methods 0.000 description 6
- 210000003719 b-lymphocyte Anatomy 0.000 description 6
- 210000000601 blood cell Anatomy 0.000 description 6
- 208000037265 diseases, disorders, signs and symptoms Diseases 0.000 description 6
- 230000000694 effects Effects 0.000 description 6
- 230000028996 humoral immune response Effects 0.000 description 6
- 210000000987 immune system Anatomy 0.000 description 6
- 230000002458 infectious effect Effects 0.000 description 6
- 210000005229 liver cell Anatomy 0.000 description 6
- 238000004519 manufacturing process Methods 0.000 description 6
- 239000000047 product Substances 0.000 description 6
- 210000004988 splenocyte Anatomy 0.000 description 6
- 238000002560 therapeutic procedure Methods 0.000 description 6
- 230000001052 transient effect Effects 0.000 description 6
- PMATZTZNYRCHOR-CGLBZJNRSA-N Cyclosporin A Chemical compound CC[C@@H]1NC(=O)[C@H]([C@H](O)[C@H](C)C\C=C\C)N(C)C(=O)[C@H](C(C)C)N(C)C(=O)[C@H](CC(C)C)N(C)C(=O)[C@H](CC(C)C)N(C)C(=O)[C@@H](C)NC(=O)[C@H](C)NC(=O)[C@H](CC(C)C)N(C)C(=O)[C@H](C(C)C)NC(=O)[C@H](CC(C)C)N(C)C(=O)CN(C)C1=O PMATZTZNYRCHOR-CGLBZJNRSA-N 0.000 description 5
- 229930105110 Cyclosporin A Natural products 0.000 description 5
- 108010036949 Cyclosporine Proteins 0.000 description 5
- 108010079245 Cystic Fibrosis Transmembrane Conductance Regulator Proteins 0.000 description 5
- 102000012605 Cystic Fibrosis Transmembrane Conductance Regulator Human genes 0.000 description 5
- 208000017604 Hodgkin disease Diseases 0.000 description 5
- 208000010747 Hodgkins lymphoma Diseases 0.000 description 5
- -1 Interferon-of Proteins 0.000 description 5
- 241001465754 Metazoa Species 0.000 description 5
- 210000001744 T-lymphocyte Anatomy 0.000 description 5
- 108060008682 Tumor Necrosis Factor Proteins 0.000 description 5
- 229960001265 ciclosporin Drugs 0.000 description 5
- 238000010367 cloning Methods 0.000 description 5
- 239000002299 complementary DNA Substances 0.000 description 5
- 230000007812 deficiency Effects 0.000 description 5
- 238000010790 dilution Methods 0.000 description 5
- 239000012895 dilution Substances 0.000 description 5
- 239000012636 effector Substances 0.000 description 5
- 229960000301 factor viii Drugs 0.000 description 5
- 239000003102 growth factor Substances 0.000 description 5
- 230000008348 humoral response Effects 0.000 description 5
- 229960003444 immunosuppressant agent Drugs 0.000 description 5
- 230000028709 inflammatory response Effects 0.000 description 5
- 230000010076 replication Effects 0.000 description 5
- 108091008146 restriction endonucleases Proteins 0.000 description 5
- 210000002966 serum Anatomy 0.000 description 5
- 230000029812 viral genome replication Effects 0.000 description 5
- 230000003612 virological effect Effects 0.000 description 5
- 102000004127 Cytokines Human genes 0.000 description 4
- 108090000695 Cytokines Proteins 0.000 description 4
- 208000021519 Hodgkin lymphoma Diseases 0.000 description 4
- 206010061218 Inflammation Diseases 0.000 description 4
- 108010063738 Interleukins Proteins 0.000 description 4
- 102000015696 Interleukins Human genes 0.000 description 4
- 102000007981 Ornithine carbamoyltransferase Human genes 0.000 description 4
- 101710198224 Ornithine carbamoyltransferase, mitochondrial Proteins 0.000 description 4
- 241000144282 Sigmodon Species 0.000 description 4
- 241000144290 Sigmodon hispidus Species 0.000 description 4
- 108020004440 Thymidine kinase Proteins 0.000 description 4
- 230000000692 anti-sense effect Effects 0.000 description 4
- 230000000903 blocking effect Effects 0.000 description 4
- 230000001413 cellular effect Effects 0.000 description 4
- 238000006243 chemical reaction Methods 0.000 description 4
- HVYWMOMLDIMFJA-DPAQBDIFSA-N cholesterol Chemical compound C1C=C2C[C@@H](O)CC[C@]2(C)[C@@H]2[C@@H]1[C@@H]1CC[C@H]([C@H](C)CCCC(C)C)[C@@]1(C)CC2 HVYWMOMLDIMFJA-DPAQBDIFSA-N 0.000 description 4
- 230000000875 corresponding effect Effects 0.000 description 4
- 201000010099 disease Diseases 0.000 description 4
- 210000002950 fibroblast Anatomy 0.000 description 4
- 239000001963 growth medium Substances 0.000 description 4
- 230000005745 host immune response Effects 0.000 description 4
- 230000002757 inflammatory effect Effects 0.000 description 4
- 230000004054 inflammatory process Effects 0.000 description 4
- 229940047122 interleukins Drugs 0.000 description 4
- 230000003389 potentiating effect Effects 0.000 description 4
- 239000000243 solution Substances 0.000 description 4
- 210000000952 spleen Anatomy 0.000 description 4
- 238000002054 transplantation Methods 0.000 description 4
- 239000002753 trypsin inhibitor Substances 0.000 description 4
- 102000003390 tumor necrosis factor Human genes 0.000 description 4
- 101150029409 CFTR gene Proteins 0.000 description 3
- 108091026890 Coding region Proteins 0.000 description 3
- 241000701022 Cytomegalovirus Species 0.000 description 3
- 102000003972 Fibroblast growth factor 7 Human genes 0.000 description 3
- 108090000385 Fibroblast growth factor 7 Proteins 0.000 description 3
- 208000031220 Hemophilia Diseases 0.000 description 3
- 208000009292 Hemophilia A Diseases 0.000 description 3
- 241000282412 Homo Species 0.000 description 3
- 108060003951 Immunoglobulin Proteins 0.000 description 3
- 108010001831 LDL receptors Proteins 0.000 description 3
- 108060001084 Luciferase Proteins 0.000 description 3
- FQISKWAFAHGMGT-SGJOWKDISA-M Methylprednisolone sodium succinate Chemical compound [Na+].C([C@@]12C)=CC(=O)C=C1[C@@H](C)C[C@@H]1[C@@H]2[C@@H](O)C[C@]2(C)[C@@](O)(C(=O)COC(=O)CCC([O-])=O)CC[C@H]21 FQISKWAFAHGMGT-SGJOWKDISA-M 0.000 description 3
- 229920001213 Polysorbate 20 Polymers 0.000 description 3
- 108091008874 T cell receptors Proteins 0.000 description 3
- 102000016266 T-Cell Antigen Receptors Human genes 0.000 description 3
- 108091036066 Three prime untranslated region Proteins 0.000 description 3
- 229940121363 anti-inflammatory agent Drugs 0.000 description 3
- 239000002260 anti-inflammatory agent Substances 0.000 description 3
- 230000030741 antigen processing and presentation Effects 0.000 description 3
- 239000013599 cloning vector Substances 0.000 description 3
- 150000001875 compounds Chemical class 0.000 description 3
- 230000003247 decreasing effect Effects 0.000 description 3
- 238000012217 deletion Methods 0.000 description 3
- 230000037430 deletion Effects 0.000 description 3
- 229940079593 drug Drugs 0.000 description 3
- 210000002443 helper t lymphocyte Anatomy 0.000 description 3
- 208000006454 hepatitis Diseases 0.000 description 3
- 231100000283 hepatitis Toxicity 0.000 description 3
- 210000003494 hepatocyte Anatomy 0.000 description 3
- 102000018358 immunoglobulin Human genes 0.000 description 3
- 230000016784 immunoglobulin production Effects 0.000 description 3
- 230000000415 inactivating effect Effects 0.000 description 3
- 229940047124 interferons Drugs 0.000 description 3
- 239000003446 ligand Substances 0.000 description 3
- 210000004185 liver Anatomy 0.000 description 3
- 230000007774 longterm Effects 0.000 description 3
- 239000000463 material Substances 0.000 description 3
- 229960004584 methylprednisolone Drugs 0.000 description 3
- 210000000056 organ Anatomy 0.000 description 3
- 239000013600 plasmid vector Substances 0.000 description 3
- 239000000256 polyoxyethylene sorbitan monolaurate Substances 0.000 description 3
- 235000010486 polyoxyethylene sorbitan monolaurate Nutrition 0.000 description 3
- 108090000765 processed proteins & peptides Proteins 0.000 description 3
- 102000005962 receptors Human genes 0.000 description 3
- 108020003175 receptors Proteins 0.000 description 3
- 230000002829 reductive effect Effects 0.000 description 3
- 230000002459 sustained effect Effects 0.000 description 3
- 239000003981 vehicle Substances 0.000 description 3
- 101710169336 5'-deoxyadenosine deaminase Proteins 0.000 description 2
- 102000055025 Adenosine deaminases Human genes 0.000 description 2
- 101710081722 Antitrypsin Proteins 0.000 description 2
- 102000015081 Blood Coagulation Factors Human genes 0.000 description 2
- 108010039209 Blood Coagulation Factors Proteins 0.000 description 2
- 102000053602 DNA Human genes 0.000 description 2
- 102000016928 DNA-directed DNA polymerase Human genes 0.000 description 2
- 108010014303 DNA-directed DNA polymerase Proteins 0.000 description 2
- KCXVZYZYPLLWCC-UHFFFAOYSA-N EDTA Chemical compound OC(=O)CN(CC(O)=O)CCN(CC(O)=O)CC(O)=O KCXVZYZYPLLWCC-UHFFFAOYSA-N 0.000 description 2
- 101710104441 FK506-binding protein 1 Proteins 0.000 description 2
- 101710132880 FK506-binding protein 1A Proteins 0.000 description 2
- 101710132879 FK506-binding protein 1B Proteins 0.000 description 2
- 108010087819 Fc receptors Proteins 0.000 description 2
- 102000009109 Fc receptors Human genes 0.000 description 2
- 108700028146 Genetic Enhancer Elements Proteins 0.000 description 2
- 102000012215 HSC70 Heat-Shock Proteins Human genes 0.000 description 2
- 108010036652 HSC70 Heat-Shock Proteins Proteins 0.000 description 2
- WZUVPPKBWHMQCE-UHFFFAOYSA-N Haematoxylin Chemical compound C12=CC(O)=C(O)C=C2CC2(O)C1C1=CC=C(O)C(O)=C1OC2 WZUVPPKBWHMQCE-UHFFFAOYSA-N 0.000 description 2
- 102000002812 Heat-Shock Proteins Human genes 0.000 description 2
- 108010004889 Heat-Shock Proteins Proteins 0.000 description 2
- 206010019786 Hepatitis non-A non-B Diseases 0.000 description 2
- 102000008070 Interferon-gamma Human genes 0.000 description 2
- 108010074328 Interferon-gamma Proteins 0.000 description 2
- 102000014150 Interferons Human genes 0.000 description 2
- 108010050904 Interferons Proteins 0.000 description 2
- 108010002352 Interleukin-1 Proteins 0.000 description 2
- 108010002350 Interleukin-2 Proteins 0.000 description 2
- 102000000588 Interleukin-2 Human genes 0.000 description 2
- 102000007330 LDL Lipoproteins Human genes 0.000 description 2
- 108010007622 LDL Lipoproteins Proteins 0.000 description 2
- 102000000853 LDL receptors Human genes 0.000 description 2
- 241000283973 Oryctolagus cuniculus Species 0.000 description 2
- 102100026408 Peptidyl-prolyl cis-trans isomerase FKBP2 Human genes 0.000 description 2
- 108010041520 Pulmonary Surfactant-Associated Proteins Proteins 0.000 description 2
- 239000006146 Roswell Park Memorial Institute medium Substances 0.000 description 2
- 101100379247 Salmo trutta apoa1 gene Proteins 0.000 description 2
- 230000005867 T cell response Effects 0.000 description 2
- 101710137500 T7 RNA polymerase Proteins 0.000 description 2
- 102000003978 Tissue Plasminogen Activator Human genes 0.000 description 2
- 108090000373 Tissue Plasminogen Activator Proteins 0.000 description 2
- 102000003990 Urokinase-type plasminogen activator Human genes 0.000 description 2
- 108090000435 Urokinase-type plasminogen activator Proteins 0.000 description 2
- 238000004458 analytical method Methods 0.000 description 2
- 230000001475 anti-trypsic effect Effects 0.000 description 2
- 239000002246 antineoplastic agent Substances 0.000 description 2
- 239000003963 antioxidant agent Substances 0.000 description 2
- 230000015572 biosynthetic process Effects 0.000 description 2
- 210000004369 blood Anatomy 0.000 description 2
- 239000008280 blood Substances 0.000 description 2
- 239000003114 blood coagulation factor Substances 0.000 description 2
- PHEZJEYUWHETKO-UHFFFAOYSA-N brequinar Chemical compound N1=C2C=CC(F)=CC2=C(C(O)=O)C(C)=C1C(C=C1)=CC=C1C1=CC=CC=C1F PHEZJEYUWHETKO-UHFFFAOYSA-N 0.000 description 2
- 229950010231 brequinar Drugs 0.000 description 2
- AIYUHDOJVYHVIT-UHFFFAOYSA-M caesium chloride Chemical compound [Cl-].[Cs+] AIYUHDOJVYHVIT-UHFFFAOYSA-M 0.000 description 2
- 230000036755 cellular response Effects 0.000 description 2
- 235000012000 cholesterol Nutrition 0.000 description 2
- 238000012411 cloning technique Methods 0.000 description 2
- 230000002596 correlated effect Effects 0.000 description 2
- 229930182912 cyclosporin Natural products 0.000 description 2
- 230000007547 defect Effects 0.000 description 2
- 230000004069 differentiation Effects 0.000 description 2
- 208000035475 disorder Diseases 0.000 description 2
- 210000002889 endothelial cell Anatomy 0.000 description 2
- 210000003527 eukaryotic cell Anatomy 0.000 description 2
- 238000011156 evaluation Methods 0.000 description 2
- 238000002474 experimental method Methods 0.000 description 2
- 208000009429 hemophilia B Diseases 0.000 description 2
- 208000002672 hepatitis B Diseases 0.000 description 2
- 230000002962 histologic effect Effects 0.000 description 2
- JYGXADMDTFJGBT-VWUMJDOOSA-N hydrocortisone Chemical compound O=C1CC[C@]2(C)[C@H]3[C@@H](O)C[C@](C)([C@@](CC4)(O)C(=O)CO)[C@@H]4[C@@H]3CCC2=C1 JYGXADMDTFJGBT-VWUMJDOOSA-N 0.000 description 2
- 230000001861 immunosuppressant effect Effects 0.000 description 2
- 230000001939 inductive effect Effects 0.000 description 2
- 238000013383 initial experiment Methods 0.000 description 2
- 229960003130 interferon gamma Drugs 0.000 description 2
- 238000007912 intraperitoneal administration Methods 0.000 description 2
- VHOGYURTWQBHIL-UHFFFAOYSA-N leflunomide Chemical compound O1N=CC(C(=O)NC=2C=CC(=CC=2)C(F)(F)F)=C1C VHOGYURTWQBHIL-UHFFFAOYSA-N 0.000 description 2
- 229960000681 leflunomide Drugs 0.000 description 2
- 239000007788 liquid Substances 0.000 description 2
- 210000005265 lung cell Anatomy 0.000 description 2
- 210000002540 macrophage Anatomy 0.000 description 2
- 239000003550 marker Substances 0.000 description 2
- 231100000682 maximum tolerated dose Toxicity 0.000 description 2
- 230000007246 mechanism Effects 0.000 description 2
- 229960001810 meprednisone Drugs 0.000 description 2
- PIDANAQULIKBQS-RNUIGHNZSA-N meprednisone Chemical compound C1CC2=CC(=O)C=C[C@]2(C)[C@@H]2[C@@H]1[C@@H]1C[C@H](C)[C@@](C(=O)CO)(O)[C@@]1(C)CC2=O PIDANAQULIKBQS-RNUIGHNZSA-N 0.000 description 2
- 239000003607 modifier Substances 0.000 description 2
- 210000005170 neoplastic cell Anatomy 0.000 description 2
- 239000002773 nucleotide Substances 0.000 description 2
- 125000003729 nucleotide group Chemical group 0.000 description 2
- 239000008188 pellet Substances 0.000 description 2
- 238000001556 precipitation Methods 0.000 description 2
- 238000002360 preparation method Methods 0.000 description 2
- 230000002265 prevention Effects 0.000 description 2
- 102000004196 processed proteins & peptides Human genes 0.000 description 2
- 108010043277 recombinant soluble CD4 Proteins 0.000 description 2
- 210000001533 respiratory mucosa Anatomy 0.000 description 2
- 230000035945 sensitivity Effects 0.000 description 2
- 238000012453 sprague-dawley rat model Methods 0.000 description 2
- 238000010561 standard procedure Methods 0.000 description 2
- 210000000130 stem cell Anatomy 0.000 description 2
- 239000000758 substrate Substances 0.000 description 2
- 229960000187 tissue plasminogen activator Drugs 0.000 description 2
- 210000003171 tumor-infiltrating lymphocyte Anatomy 0.000 description 2
- 229960001134 von willebrand factor Drugs 0.000 description 2
- XLYOFNOQVPJJNP-UHFFFAOYSA-N water Substances O XLYOFNOQVPJJNP-UHFFFAOYSA-N 0.000 description 2
- DIGQNXIGRZPYDK-WKSCXVIASA-N (2R)-6-amino-2-[[2-[[(2S)-2-[[2-[[(2R)-2-[[(2S)-2-[[(2R,3S)-2-[[2-[[(2S)-2-[[2-[[(2S)-2-[[(2S)-2-[[(2R)-2-[[(2S,3S)-2-[[(2R)-2-[[(2S)-2-[[(2S)-2-[[(2S)-2-[[2-[[(2S)-2-[[(2R)-2-[[2-[[2-[[2-[(2-amino-1-hydroxyethylidene)amino]-3-carboxy-1-hydroxypropylidene]amino]-1-hydroxy-3-sulfanylpropylidene]amino]-1-hydroxyethylidene]amino]-1-hydroxy-3-sulfanylpropylidene]amino]-1,3-dihydroxypropylidene]amino]-1-hydroxyethylidene]amino]-1-hydroxypropylidene]amino]-1,3-dihydroxypropylidene]amino]-1,3-dihydroxypropylidene]amino]-1-hydroxy-3-sulfanylpropylidene]amino]-1,3-dihydroxybutylidene]amino]-1-hydroxy-3-sulfanylpropylidene]amino]-1-hydroxypropylidene]amino]-1,3-dihydroxypropylidene]amino]-1-hydroxyethylidene]amino]-1,5-dihydroxy-5-iminopentylidene]amino]-1-hydroxy-3-sulfanylpropylidene]amino]-1,3-dihydroxybutylidene]amino]-1-hydroxy-3-sulfanylpropylidene]amino]-1,3-dihydroxypropylidene]amino]-1-hydroxyethylidene]amino]-1-hydroxy-3-sulfanylpropylidene]amino]-1-hydroxyethylidene]amino]hexanoic acid Chemical compound C[C@@H]([C@@H](C(=N[C@@H](CS)C(=N[C@@H](C)C(=N[C@@H](CO)C(=NCC(=N[C@@H](CCC(=N)O)C(=NC(CS)C(=N[C@H]([C@H](C)O)C(=N[C@H](CS)C(=N[C@H](CO)C(=NCC(=N[C@H](CS)C(=NCC(=N[C@H](CCCCN)C(=O)O)O)O)O)O)O)O)O)O)O)O)O)O)O)N=C([C@H](CS)N=C([C@H](CO)N=C([C@H](CO)N=C([C@H](C)N=C(CN=C([C@H](CO)N=C([C@H](CS)N=C(CN=C(C(CS)N=C(C(CC(=O)O)N=C(CN)O)O)O)O)O)O)O)O)O)O)O)O DIGQNXIGRZPYDK-WKSCXVIASA-N 0.000 description 1
- MZOFCQQQCNRIBI-VMXHOPILSA-N (3s)-4-[[(2s)-1-[[(2s)-1-[[(1s)-1-carboxy-2-hydroxyethyl]amino]-4-methyl-1-oxopentan-2-yl]amino]-5-(diaminomethylideneamino)-1-oxopentan-2-yl]amino]-3-[[2-[[(2s)-2,6-diaminohexanoyl]amino]acetyl]amino]-4-oxobutanoic acid Chemical compound OC[C@@H](C(O)=O)NC(=O)[C@H](CC(C)C)NC(=O)[C@H](CCCN=C(N)N)NC(=O)[C@H](CC(O)=O)NC(=O)CNC(=O)[C@@H](N)CCCCN MZOFCQQQCNRIBI-VMXHOPILSA-N 0.000 description 1
- 108091032973 (ribonucleotides)n+m Proteins 0.000 description 1
- YRNWIFYIFSBPAU-UHFFFAOYSA-N 4-[4-(dimethylamino)phenyl]-n,n-dimethylaniline Chemical compound C1=CC(N(C)C)=CC=C1C1=CC=C(N(C)C)C=C1 YRNWIFYIFSBPAU-UHFFFAOYSA-N 0.000 description 1
- WNWVKZTYMQWFHE-UHFFFAOYSA-N 4-ethylmorpholine Chemical compound [CH2]CN1CCOCC1 WNWVKZTYMQWFHE-UHFFFAOYSA-N 0.000 description 1
- 208000030507 AIDS Diseases 0.000 description 1
- 102100036664 Adenosine deaminase Human genes 0.000 description 1
- 206010001258 Adenoviral infections Diseases 0.000 description 1
- 229920001817 Agar Polymers 0.000 description 1
- 102000009027 Albumins Human genes 0.000 description 1
- 108010088751 Albumins Proteins 0.000 description 1
- KHOITXIGCFIULA-UHFFFAOYSA-N Alophen Chemical compound C1=CC(OC(=O)C)=CC=C1C(C=1N=CC=CC=1)C1=CC=C(OC(C)=O)C=C1 KHOITXIGCFIULA-UHFFFAOYSA-N 0.000 description 1
- 208000024827 Alzheimer disease Diseases 0.000 description 1
- 102000008076 Angiogenic Proteins Human genes 0.000 description 1
- 108010074415 Angiogenic Proteins Proteins 0.000 description 1
- 101710145634 Antigen 1 Proteins 0.000 description 1
- 102000018655 Apolipoproteins C Human genes 0.000 description 1
- 108010027070 Apolipoproteins C Proteins 0.000 description 1
- 102000013918 Apolipoproteins E Human genes 0.000 description 1
- 108010025628 Apolipoproteins E Proteins 0.000 description 1
- 208000014644 Brain disease Diseases 0.000 description 1
- 108010021064 CTLA-4 Antigen Proteins 0.000 description 1
- 102000008203 CTLA-4 Antigen Human genes 0.000 description 1
- 229940045513 CTLA4 antagonist Drugs 0.000 description 1
- 241000282472 Canis lupus familiaris Species 0.000 description 1
- 241000283707 Capra Species 0.000 description 1
- 102000014914 Carrier Proteins Human genes 0.000 description 1
- 102100035882 Catalase Human genes 0.000 description 1
- 108010053835 Catalase Proteins 0.000 description 1
- 208000017667 Chronic Disease Diseases 0.000 description 1
- 206010010099 Combined immunodeficiency Diseases 0.000 description 1
- 208000035473 Communicable disease Diseases 0.000 description 1
- 206010010144 Completed suicide Diseases 0.000 description 1
- 206010010356 Congenital anomaly Diseases 0.000 description 1
- 108010036941 Cyclosporins Proteins 0.000 description 1
- 201000003883 Cystic fibrosis Diseases 0.000 description 1
- 102100039498 Cytotoxic T-lymphocyte protein 4 Human genes 0.000 description 1
- 230000004544 DNA amplification Effects 0.000 description 1
- 238000012286 ELISA Assay Methods 0.000 description 1
- 238000008157 ELISA kit Methods 0.000 description 1
- 206010014561 Emphysema Diseases 0.000 description 1
- 229920001917 Ficoll Polymers 0.000 description 1
- 108010093031 Galactosidases Proteins 0.000 description 1
- 208000015872 Gaucher disease Diseases 0.000 description 1
- SXRSQZLOMIGNAQ-UHFFFAOYSA-N Glutaraldehyde Chemical compound O=CCCCC=O SXRSQZLOMIGNAQ-UHFFFAOYSA-N 0.000 description 1
- 108010063907 Glutathione Reductase Proteins 0.000 description 1
- 102100036442 Glutathione reductase, mitochondrial Human genes 0.000 description 1
- 108010017213 Granulocyte-Macrophage Colony-Stimulating Factor Proteins 0.000 description 1
- 102100039620 Granulocyte-macrophage colony-stimulating factor Human genes 0.000 description 1
- 206010019799 Hepatitis viral Diseases 0.000 description 1
- 208000009889 Herpes Simplex Diseases 0.000 description 1
- 102000007625 Hirudins Human genes 0.000 description 1
- 108010007267 Hirudins Proteins 0.000 description 1
- 101000907783 Homo sapiens Cystic fibrosis transmembrane conductance regulator Proteins 0.000 description 1
- 101000889276 Homo sapiens Cytotoxic T-lymphocyte protein 4 Proteins 0.000 description 1
- 206010020575 Hyperammonaemia Diseases 0.000 description 1
- 108090001061 Insulin Proteins 0.000 description 1
- 102100022339 Integrin alpha-L Human genes 0.000 description 1
- 102000003996 Interferon-beta Human genes 0.000 description 1
- 108090000467 Interferon-beta Proteins 0.000 description 1
- 108010065805 Interleukin-12 Proteins 0.000 description 1
- 102000013462 Interleukin-12 Human genes 0.000 description 1
- 108090001005 Interleukin-6 Proteins 0.000 description 1
- 108090001007 Interleukin-8 Proteins 0.000 description 1
- 108091026898 Leader sequence (mRNA) Proteins 0.000 description 1
- 208000019693 Lung disease Diseases 0.000 description 1
- 206010058467 Lung neoplasm malignant Diseases 0.000 description 1
- 108010064548 Lymphocyte Function-Associated Antigen-1 Proteins 0.000 description 1
- 102000008072 Lymphokines Human genes 0.000 description 1
- 108010074338 Lymphokines Proteins 0.000 description 1
- 241000701076 Macacine alphaherpesvirus 1 Species 0.000 description 1
- 102000003792 Metallothionein Human genes 0.000 description 1
- 108090000157 Metallothionein Proteins 0.000 description 1
- NTIZESTWPVYFNL-UHFFFAOYSA-N Methyl isobutyl ketone Chemical compound CC(C)CC(C)=O NTIZESTWPVYFNL-UHFFFAOYSA-N 0.000 description 1
- 241000713333 Mouse mammary tumor virus Species 0.000 description 1
- 101000930477 Mus musculus Albumin Proteins 0.000 description 1
- 206010028980 Neoplasm Diseases 0.000 description 1
- 102000008299 Nitric Oxide Synthase Human genes 0.000 description 1
- 108010021487 Nitric Oxide Synthase Proteins 0.000 description 1
- 238000012408 PCR amplification Methods 0.000 description 1
- 229930040373 Paraformaldehyde Natural products 0.000 description 1
- 102000003992 Peroxidases Human genes 0.000 description 1
- 201000011252 Phenylketonuria Diseases 0.000 description 1
- 102000000528 Pulmonary Surfactant-Associated Proteins Human genes 0.000 description 1
- 239000012979 RPMI medium Substances 0.000 description 1
- 238000012300 Sequence Analysis Methods 0.000 description 1
- 241000700584 Simplexvirus Species 0.000 description 1
- FAPWRFPIFSIZLT-UHFFFAOYSA-M Sodium chloride Chemical compound [Na+].[Cl-] FAPWRFPIFSIZLT-UHFFFAOYSA-M 0.000 description 1
- 235000007235 Spartium junceum Nutrition 0.000 description 1
- 240000005622 Spartium junceum Species 0.000 description 1
- 102000019197 Superoxide Dismutase Human genes 0.000 description 1
- 108010012715 Superoxide dismutase Proteins 0.000 description 1
- 102100036011 T-cell surface glycoprotein CD4 Human genes 0.000 description 1
- 208000002903 Thalassemia Diseases 0.000 description 1
- GLNADSQYFUSGOU-GPTZEZBUSA-J Trypan blue Chemical compound [Na+].[Na+].[Na+].[Na+].C1=C(S([O-])(=O)=O)C=C2C=C(S([O-])(=O)=O)C(/N=N/C3=CC=C(C=C3C)C=3C=C(C(=CC=3)\N=N\C=3C(=CC4=CC(=CC(N)=C4C=3O)S([O-])(=O)=O)S([O-])(=O)=O)C)=C(O)C2=C1N GLNADSQYFUSGOU-GPTZEZBUSA-J 0.000 description 1
- 102000004142 Trypsin Human genes 0.000 description 1
- 108090000631 Trypsin Proteins 0.000 description 1
- 102000000852 Tumor Necrosis Factor-alpha Human genes 0.000 description 1
- 101900150902 Varicella-zoster virus Thymidine kinase Proteins 0.000 description 1
- 108700005077 Viral Genes Proteins 0.000 description 1
- 208000036142 Viral infection Diseases 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
- 230000009471 action Effects 0.000 description 1
- 201000009628 adenosine deaminase deficiency Diseases 0.000 description 1
- 230000001780 adrenocortical effect Effects 0.000 description 1
- 238000012382 advanced drug delivery Methods 0.000 description 1
- 239000008272 agar Substances 0.000 description 1
- 210000001552 airway epithelial cell Anatomy 0.000 description 1
- 102000015395 alpha 1-Antitrypsin Human genes 0.000 description 1
- 108010050122 alpha 1-Antitrypsin Proteins 0.000 description 1
- 229940024142 alpha 1-antitrypsin Drugs 0.000 description 1
- 230000004075 alteration Effects 0.000 description 1
- 210000002821 alveolar epithelial cell Anatomy 0.000 description 1
- AVKUERGKIZMTKX-NJBDSQKTSA-N ampicillin Chemical compound C1([C@@H](N)C(=O)N[C@H]2[C@H]3SC([C@@H](N3C2=O)C(O)=O)(C)C)=CC=CC=C1 AVKUERGKIZMTKX-NJBDSQKTSA-N 0.000 description 1
- 229960000723 ampicillin Drugs 0.000 description 1
- 230000003110 anti-inflammatory effect Effects 0.000 description 1
- 230000000781 anti-lymphocytic effect Effects 0.000 description 1
- 208000036556 autosomal recessive T cell-negative B cell-negative NK cell-negative due to adenosine deaminase deficiency severe combined immunodeficiency Diseases 0.000 description 1
- 229960002170 azathioprine Drugs 0.000 description 1
- LMEKQMALGUDUQG-UHFFFAOYSA-N azathioprine Chemical compound CN1C=NC([N+]([O-])=O)=C1SC1=NC=NC2=C1NC=N2 LMEKQMALGUDUQG-UHFFFAOYSA-N 0.000 description 1
- 230000001580 bacterial effect Effects 0.000 description 1
- 108010028263 bacteriophage T3 RNA polymerase Proteins 0.000 description 1
- 239000011324 bead Substances 0.000 description 1
- 102000005936 beta-Galactosidase Human genes 0.000 description 1
- 108010005774 beta-Galactosidase Proteins 0.000 description 1
- 108091008324 binding proteins Proteins 0.000 description 1
- 239000002981 blocking agent Substances 0.000 description 1
- 210000002798 bone marrow cell Anatomy 0.000 description 1
- 210000004958 brain cell Anatomy 0.000 description 1
- 239000000872 buffer Substances 0.000 description 1
- 239000001506 calcium phosphate Substances 0.000 description 1
- 229910000389 calcium phosphate Inorganic materials 0.000 description 1
- 235000011010 calcium phosphates Nutrition 0.000 description 1
- 201000011510 cancer Diseases 0.000 description 1
- 230000015556 catabolic process Effects 0.000 description 1
- 230000010261 cell growth Effects 0.000 description 1
- 230000022534 cell killing Effects 0.000 description 1
- 230000007541 cellular toxicity Effects 0.000 description 1
- 230000008859 change Effects 0.000 description 1
- 230000001876 chaperonelike Effects 0.000 description 1
- 239000003153 chemical reaction reagent Substances 0.000 description 1
- 238000012761 co-transfection Methods 0.000 description 1
- 230000001276 controlling effect Effects 0.000 description 1
- 238000012937 correction Methods 0.000 description 1
- 239000003246 corticosteroid Substances 0.000 description 1
- 229960001334 corticosteroids Drugs 0.000 description 1
- 238000005520 cutting process Methods 0.000 description 1
- 230000006378 damage Effects 0.000 description 1
- 230000007423 decrease Effects 0.000 description 1
- 230000006735 deficit Effects 0.000 description 1
- 239000005547 deoxyribonucleotide Substances 0.000 description 1
- 125000002637 deoxyribonucleotide group Chemical class 0.000 description 1
- 238000001514 detection method Methods 0.000 description 1
- 238000011161 development Methods 0.000 description 1
- 206010012601 diabetes mellitus Diseases 0.000 description 1
- 230000029087 digestion Effects 0.000 description 1
- 238000007865 diluting Methods 0.000 description 1
- 230000003292 diminished effect Effects 0.000 description 1
- 239000003937 drug carrier Substances 0.000 description 1
- 238000012377 drug delivery Methods 0.000 description 1
- 238000004520 electroporation Methods 0.000 description 1
- 210000001842 enterocyte Anatomy 0.000 description 1
- 238000001976 enzyme digestion Methods 0.000 description 1
- YQGOJNYOYNNSMM-UHFFFAOYSA-N eosin Chemical compound [Na+].OC(=O)C1=CC=CC=C1C1=C2C=C(Br)C(=O)C(Br)=C2OC2=C(Br)C(O)=C(Br)C=C21 YQGOJNYOYNNSMM-UHFFFAOYSA-N 0.000 description 1
- 210000002919 epithelial cell Anatomy 0.000 description 1
- 230000008472 epithelial growth Effects 0.000 description 1
- 210000003743 erythrocyte Anatomy 0.000 description 1
- 239000012530 fluid Substances 0.000 description 1
- 238000001943 fluorescence-activated cell sorting Methods 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
- 210000001035 gastrointestinal tract Anatomy 0.000 description 1
- 238000001476 gene delivery Methods 0.000 description 1
- 230000002068 genetic effect Effects 0.000 description 1
- 208000037824 growth disorder Diseases 0.000 description 1
- 208000019622 heart disease Diseases 0.000 description 1
- 229940006607 hirudin Drugs 0.000 description 1
- WQPDUTSPKFMPDP-OUMQNGNKSA-N hirudin Chemical compound C([C@@H](C(=O)N[C@@H](CCC(O)=O)C(=O)N[C@@H](CCC(O)=O)C(=O)N[C@@H]([C@@H](C)CC)C(=O)N1[C@@H](CCC1)C(=O)N[C@@H](CCC(O)=O)C(=O)N[C@@H](CCC(O)=O)C(=O)N[C@@H](CC=1C=CC(OS(O)(=O)=O)=CC=1)C(=O)N[C@@H](CC(C)C)C(=O)N[C@@H](CCC(N)=O)C(O)=O)NC(=O)[C@H](CC(O)=O)NC(=O)CNC(=O)[C@H](CC(O)=O)NC(=O)[C@H](CC(N)=O)NC(=O)[C@H](CC=1NC=NC=1)NC(=O)[C@H](CO)NC(=O)[C@H](CCC(N)=O)NC(=O)[C@H]1N(CCC1)C(=O)[C@H](CCCCN)NC(=O)[C@H]1N(CCC1)C(=O)[C@@H](NC(=O)CNC(=O)[C@H](CCC(O)=O)NC(=O)CNC(=O)[C@@H](NC(=O)[C@@H](NC(=O)[C@H]1NC(=O)[C@H](CCC(N)=O)NC(=O)[C@H](CC(N)=O)NC(=O)[C@H](CCCCN)NC(=O)[C@H](CCC(O)=O)NC(=O)CNC(=O)[C@H](CC(O)=O)NC(=O)[C@H](CO)NC(=O)CNC(=O)[C@H](CC(C)C)NC(=O)[C@H]([C@@H](C)CC)NC(=O)[C@@H]2CSSC[C@@H](C(=O)N[C@@H](CCC(O)=O)C(=O)NCC(=O)N[C@@H](CO)C(=O)N[C@@H](CC(N)=O)C(=O)N[C@H](C(=O)N[C@H](C(NCC(=O)N[C@@H](CCC(N)=O)C(=O)NCC(=O)N[C@@H](CC(N)=O)C(=O)N[C@@H](CCCCN)C(=O)N2)=O)CSSC1)C(C)C)NC(=O)[C@H](CC(C)C)NC(=O)[C@H]1NC(=O)[C@H](CC(C)C)NC(=O)[C@H](CC(N)=O)NC(=O)[C@H](CCC(N)=O)NC(=O)CNC(=O)[C@H](CO)NC(=O)[C@H](CCC(O)=O)NC(=O)[C@H]([C@@H](C)O)NC(=O)[C@@H](NC(=O)[C@H](CC(O)=O)NC(=O)[C@@H](NC(=O)[C@H](CC=2C=CC(O)=CC=2)NC(=O)[C@@H](NC(=O)[C@@H](N)C(C)C)C(C)C)[C@@H](C)O)CSSC1)C(C)C)[C@@H](C)O)[C@@H](C)O)C1=CC=CC=C1 WQPDUTSPKFMPDP-OUMQNGNKSA-N 0.000 description 1
- 229940088597 hormone Drugs 0.000 description 1
- 239000005556 hormone Substances 0.000 description 1
- 102000056427 human CFTR Human genes 0.000 description 1
- 229960000890 hydrocortisone Drugs 0.000 description 1
- 239000012642 immune effector Substances 0.000 description 1
- 229940072221 immunoglobulins Drugs 0.000 description 1
- 229940121354 immunomodulator Drugs 0.000 description 1
- 238000001114 immunoprecipitation Methods 0.000 description 1
- 238000002650 immunosuppressive therapy Methods 0.000 description 1
- 210000004969 inflammatory cell Anatomy 0.000 description 1
- 230000004941 influx Effects 0.000 description 1
- 238000001802 infusion Methods 0.000 description 1
- 239000003112 inhibitor Substances 0.000 description 1
- 239000002054 inoculum Substances 0.000 description 1
- 238000003780 insertion Methods 0.000 description 1
- 230000037431 insertion Effects 0.000 description 1
- 230000003993 interaction Effects 0.000 description 1
- 229940079322 interferon Drugs 0.000 description 1
- 229960001388 interferon-beta Drugs 0.000 description 1
- 229940117681 interleukin-12 Drugs 0.000 description 1
- 238000007918 intramuscular administration Methods 0.000 description 1
- 239000007928 intraperitoneal injection Substances 0.000 description 1
- 238000005304 joining Methods 0.000 description 1
- 210000002510 keratinocyte Anatomy 0.000 description 1
- 101150066555 lacZ gene Proteins 0.000 description 1
- 208000032839 leukemia Diseases 0.000 description 1
- 210000000265 leukocyte Anatomy 0.000 description 1
- 230000000670 limiting effect Effects 0.000 description 1
- 238000011068 loading method Methods 0.000 description 1
- 201000005202 lung cancer Diseases 0.000 description 1
- 208000020816 lung neoplasm Diseases 0.000 description 1
- 239000012139 lysis buffer Substances 0.000 description 1
- 238000012423 maintenance Methods 0.000 description 1
- 239000002609 medium Substances 0.000 description 1
- MIKKOBKEXMRYFQ-WZTVWXICSA-N meglumine amidotrizoate Chemical compound C[NH2+]C[C@H](O)[C@@H](O)[C@H](O)[C@H](O)CO.CC(=O)NC1=C(I)C(NC(C)=O)=C(I)C(C([O-])=O)=C1I MIKKOBKEXMRYFQ-WZTVWXICSA-N 0.000 description 1
- 210000005033 mesothelial cell Anatomy 0.000 description 1
- 108020004999 messenger RNA Proteins 0.000 description 1
- 230000004060 metabolic process Effects 0.000 description 1
- HPNSFSBZBAHARI-UHFFFAOYSA-N micophenolic acid Natural products OC1=C(CC=C(C)CCC(O)=O)C(OC)=C(C)C2=C1C(=O)OC2 HPNSFSBZBAHARI-UHFFFAOYSA-N 0.000 description 1
- 238000000520 microinjection Methods 0.000 description 1
- 238000000386 microscopy Methods 0.000 description 1
- 238000002156 mixing Methods 0.000 description 1
- 239000000203 mixture Substances 0.000 description 1
- 229950007856 mofetil Drugs 0.000 description 1
- 230000009456 molecular mechanism Effects 0.000 description 1
- 210000001616 monocyte Anatomy 0.000 description 1
- 229940014456 mycophenolate Drugs 0.000 description 1
- RTGDFNSFWBGLEC-SYZQJQIISA-N mycophenolate mofetil Chemical compound COC1=C(C)C=2COC(=O)C=2C(O)=C1C\C=C(/C)CCC(=O)OCCN1CCOCC1 RTGDFNSFWBGLEC-SYZQJQIISA-N 0.000 description 1
- 229960004866 mycophenolate mofetil Drugs 0.000 description 1
- HPNSFSBZBAHARI-RUDMXATFSA-N mycophenolic acid Chemical compound OC1=C(C\C=C(/C)CCC(O)=O)C(OC)=C(C)C2=C1C(=O)OC2 HPNSFSBZBAHARI-RUDMXATFSA-N 0.000 description 1
- 210000003098 myoblast Anatomy 0.000 description 1
- 210000000822 natural killer cell Anatomy 0.000 description 1
- 239000013642 negative control Substances 0.000 description 1
- 230000001613 neoplastic effect Effects 0.000 description 1
- 210000003061 neural cell Anatomy 0.000 description 1
- 238000006386 neutralization reaction Methods 0.000 description 1
- 210000000440 neutrophil Anatomy 0.000 description 1
- 230000005937 nuclear translocation Effects 0.000 description 1
- 230000008520 organization Effects 0.000 description 1
- 239000012188 paraffin wax Substances 0.000 description 1
- 229920002866 paraformaldehyde Polymers 0.000 description 1
- 210000004738 parenchymal cell Anatomy 0.000 description 1
- 150000002972 pentoses Chemical group 0.000 description 1
- 230000002093 peripheral effect Effects 0.000 description 1
- 108040007629 peroxidase activity proteins Proteins 0.000 description 1
- 239000002831 pharmacologic agent Substances 0.000 description 1
- 230000000144 pharmacologic effect Effects 0.000 description 1
- 230000007505 plaque formation Effects 0.000 description 1
- 238000003752 polymerase chain reaction Methods 0.000 description 1
- 229920005862 polyol Polymers 0.000 description 1
- 150000003077 polyols Chemical class 0.000 description 1
- 229920001184 polypeptide Polymers 0.000 description 1
- 210000003240 portal vein Anatomy 0.000 description 1
- 239000002243 precursor Substances 0.000 description 1
- 229960005205 prednisolone Drugs 0.000 description 1
- OIGNJSKKLXVSLS-VWUMJDOOSA-N prednisolone Chemical compound O=C1C=C[C@]2(C)[C@H]3[C@@H](O)C[C@](C)([C@@](CC4)(O)C(=O)CO)[C@@H]4[C@@H]3CCC2=C1 OIGNJSKKLXVSLS-VWUMJDOOSA-N 0.000 description 1
- 210000001948 pro-b lymphocyte Anatomy 0.000 description 1
- 230000000644 propagated effect Effects 0.000 description 1
- 230000000069 prophylactic effect Effects 0.000 description 1
- 230000012846 protein folding Effects 0.000 description 1
- 108010030416 proteoliposomes Proteins 0.000 description 1
- 230000002797 proteolythic effect Effects 0.000 description 1
- 238000000746 purification Methods 0.000 description 1
- ZAHRKKWIAAJSAO-UHFFFAOYSA-N rapamycin Natural products COCC(O)C(=C/C(C)C(=O)CC(OC(=O)C1CCCCN1C(=O)C(=O)C2(O)OC(CC(OC)C(=CC=CC=CC(C)CC(C)C(=O)C)C)CCC2C)C(C)CC3CCC(O)C(C3)OC)C ZAHRKKWIAAJSAO-UHFFFAOYSA-N 0.000 description 1
- 238000003753 real-time PCR Methods 0.000 description 1
- 230000003252 repetitive effect Effects 0.000 description 1
- 238000009256 replacement therapy Methods 0.000 description 1
- 230000000241 respiratory effect Effects 0.000 description 1
- 230000002441 reversible effect Effects 0.000 description 1
- 238000012552 review Methods 0.000 description 1
- 150000003839 salts Chemical class 0.000 description 1
- 230000001235 sensitizing effect Effects 0.000 description 1
- 238000013207 serial dilution Methods 0.000 description 1
- 208000002491 severe combined immunodeficiency Diseases 0.000 description 1
- 230000035939 shock Effects 0.000 description 1
- 208000007056 sickle cell anemia Diseases 0.000 description 1
- 229960002930 sirolimus Drugs 0.000 description 1
- QFJCIRLUMZQUOT-HPLJOQBZSA-N sirolimus Chemical compound C1C[C@@H](O)[C@H](OC)C[C@@H]1C[C@@H](C)[C@H]1OC(=O)[C@@H]2CCCCN2C(=O)C(=O)[C@](O)(O2)[C@H](C)CC[C@H]2C[C@H](OC)/C(C)=C/C=C/C=C/[C@@H](C)C[C@@H](C)C(=O)[C@H](OC)[C@H](O)/C(C)=C/[C@@H](C)C(=O)C1 QFJCIRLUMZQUOT-HPLJOQBZSA-N 0.000 description 1
- 229910000029 sodium carbonate Inorganic materials 0.000 description 1
- 239000001509 sodium citrate Substances 0.000 description 1
- NLJMYIDDQXHKNR-UHFFFAOYSA-K sodium citrate Chemical compound O.O.[Na+].[Na+].[Na+].[O-]C(=O)CC(O)(CC([O-])=O)C([O-])=O NLJMYIDDQXHKNR-UHFFFAOYSA-K 0.000 description 1
- 239000007787 solid Substances 0.000 description 1
- 230000010473 stable expression Effects 0.000 description 1
- 230000000638 stimulation Effects 0.000 description 1
- 239000012089 stop solution Substances 0.000 description 1
- 239000006228 supernatant Substances 0.000 description 1
- 239000013589 supplement Substances 0.000 description 1
- 238000007910 systemic administration Methods 0.000 description 1
- 238000012360 testing method Methods 0.000 description 1
- 210000001519 tissue Anatomy 0.000 description 1
- 210000003014 totipotent stem cell Anatomy 0.000 description 1
- 231100000419 toxicity Toxicity 0.000 description 1
- 230000001988 toxicity Effects 0.000 description 1
- 238000010361 transduction Methods 0.000 description 1
- 230000026683 transduction Effects 0.000 description 1
- 238000001890 transfection Methods 0.000 description 1
- 230000032258 transport Effects 0.000 description 1
- 238000011269 treatment regimen Methods 0.000 description 1
- QORWJWZARLRLPR-UHFFFAOYSA-H tricalcium bis(phosphate) Chemical compound [Ca+2].[Ca+2].[Ca+2].[O-]P([O-])([O-])=O.[O-]P([O-])([O-])=O QORWJWZARLRLPR-UHFFFAOYSA-H 0.000 description 1
- 239000012588 trypsin Substances 0.000 description 1
- 229960005356 urokinase Drugs 0.000 description 1
- 210000004509 vascular smooth muscle cell Anatomy 0.000 description 1
- 230000002227 vasoactive effect Effects 0.000 description 1
- 201000001862 viral hepatitis Diseases 0.000 description 1
- 230000009385 viral infection Effects 0.000 description 1
- 239000013603 viral vector Substances 0.000 description 1
- 210000002845 virion Anatomy 0.000 description 1
- 238000005406 washing Methods 0.000 description 1
- 239000012224 working solution Substances 0.000 description 1
Definitions
- This invention relates to gene therapy comprising the use of adenoviruses as the gene delivery vehicles . More particularly, this invention relates to gene therapy involving the concurrent and repeated administration of adenoviruses and immunosuppressive agents, whereby the efficiency of the gene therapy treatment is enhanced through suppression of an immune response against the adenoviruses.
- Adenoviruses can be very efficient in gene transfer into cells in vivo, and, thus may be employed as delivery vehicles for introducing desired genes into eukaryotic cells, whereby the adenovirus delivers such genes to eukaryotic cells by binding cellular receptors.
- adenovirus gene transfer There are, however, several limitations to adenovirus gene transfer which are due in part to host responses directed at either the adenovirus vector particle, breakdown products of the vector particle, or the transduced cells. These host responses include non-specific responses and specific immune responses. The non-specific responses include inflammatory and non-inflammatory changes. An example of the latter is a change in host cell gene expression.
- Specific immune responses include various cellular responses and humoral antibody responses. Cellular responses include those mediated by T-helper lymphocytes, T- suppressor lymphocytes, cytotoxic T lymphocytes (CTL) , and natural killer cells.
- T-cell response contributes to, but is not responsible solely for, the limited duration of expression in adults from adenovirus vectors.
- the authors further show that cyclosporin A is not effective in blocking the humoral response to the vector.
- Figure 3 is a schematic of the construction of plasmid pAvS6;
- Figure 4 is a map of plasmid pAvS6;
- Figure 5 is a map of plasmid pBQ4.7 ;
- Figures 10A, 10B, and IOC depict the histologic appearance of the lung in response to AvlCf2 administration three days after vector administration;
- Figure 12 is a graph of anti-adenoviral antibody titers of lung lavage samples from rats infected with AvlCf2 and which were treated or not treated with dexamethasone;
- Figure 13 is a graph of CTL responses in rats 42 days after infection with AvlCf2 ;
- Figure 15 is a map of plasmid pAvlH9FR;
- Figure 16 is a schematic of the adenoviral vector AvlH9F2 ;
- a method of effecting a gene therapy treatment in a host comprises administering to a host (i) an adenoviral vector including at least one DNA sequence and (ii) an immunosuppressive agent.
- the course of administration of the adenoviral vector and immunosuppressive agent then is discontinued.
- Administration of the immunosuppressive agent and the adenoviral vector then is repeated at least once.
- the adenoviral vector is administered in an amount effective to produce a therapeutic effect in the host.
- the immunosuppressive agent is administered in an amount effective to prevent or suppress a humoral and/or cellular immune response to the vector and/or cells containing the vector.
- Agents which affect humoral antibody responses are generally directed at antibody producing B lymphocytes (B- cells) or at the T-cells which are responsible for inducing B-cell antibody production to high levels.
- the ITR, encapsidation signal, Rous Sarcoma Virus promoter, the adenoviral tripartite leader (TPL) sequence and linking sequences were assembled as a block using PCR amplification ( Figure 2) .
- the ITR and encapsidation signal (sequences 1-392 of Ad-dl327 [identical to sequences from Ad5, Genbank accession #M73260] incorporated herein by reference) were amplified (amplification 1) together from Ad-dl327 using primers containing NotI or Ascl restriction sites.
- Sensitizer cells were prepared by infecting cotton rat lung fibroblasts with Ad-dl327 at a multiplicity of infection of 100. The cells were incubated for 3 days, and checked for hexon expression by FACS . The cells then were washed with PBS/EDTA, contacted with trypsin, washed, spun, and resuspended in 1 ml RPMI medium. The cells then were irradiated with 13'7 Cs at 5,000 rads in order to inactivate the DNA.
- Spleens then were isolated from uninfected (control) rats and adenovirus-infected rats 42 days after infection.
- the spleens were kept in sterile HBSS and ice. 10 ml of HBSS then was injected into each spleen with a 25/27 gauge needle. The spleen was mashed, and filtered with a cell strainer into a 50 ml tube. The volume then was brought to 40 ml in RPMI plus 10% FCS. The tube was spun at 1,500 rpm for 10 minutes. Red blood cells then were lysed by adding 2.5 ml of ACK lysis buffer, and the liquid was swirled for less than 1 minute. The volume was brought up to 50 ml with RPMI-10.
- AvlH9F2 is constructed from a derivative of the adenoviral shuttle plasmid vector pAvlH9FR ( Figure 15) , which includes human Factor IX DNA, and is described in PCT application No. W094/29471, published December 22, 1994.
- Neutralizing antibody titers in 20 mice are given in Table I below. As indicated in Table I, DSG is deoxyspergualin, Cy is cyclophosphamide, and Dex is dexamethasone .
- AvlALAPH ⁇ l is an adenoviral vector which co ⁇ Ui WS tkiQ &* domain deleted human Factor VIII cDNA expressed from the mouse albumin promoter, and is described in published PCT Application No. W094/29471.
- Deoxyspergualin is an immunosuppressant currently being tested clinically in organ transplantation. It has a potent, long term effect on antigen specific B cells and has been shown to prevent effectively the production of specific antibody when co-administered with protein antigens.
- Alegre, et al . , Transplantation, Vol. 57, pgs. 1786-1794 (1994); Tepper, Ann. N. Y. Acad. Sci .. pgs. 123-132 (1993); Tufveson, et al . , Transplant . Proc .. Vol. 26, pgs. 3029-3039 (1994) .
- the mode of action of DSG is not fully understood at the molecular level .
- Plasma levels of human Factor VIII were determined by ELISA, as described in Connelly, et al . , Human Gene Therapy, Vol. 6, pgs. 185-193 (1995). The limit of sensitivity with mouse plasma samples containing B domain deleted human Factor VIII was 3 to 6 ng/ml. Mouse plasma samples were diluted 1:5 prior to the assay, therefore, the actual limit of detection was 15 to 30 ng/ml.
- Plasma levels of human Factor IX were determined by ELISA. Asserachrom IX:Ag ELISA kits were purchased from American Bioproducts Company (Parsippany, NJ) and assays were performed according to the manufacturer's instructions. The limit of sensitivity was 1.6 ng/ml.
- Mouse plasma or serum samples were heat inactivated at 55 °C for 30 minutes and then diluted in Improved Minimal Essential Medium (Biofluids, Rockville, MD) plus 2% FBS (IMEM/2%FBS) in two-fold steps beginning at 1:2.
- 55 ⁇ l of each sample were mixed with 10 ⁇ l of AvllacZ4 (containing 4 x 10 5 pfu) , incubated for 1 hour at 37 °C and applied to nearly confluent 293 cells in 96 well plates (4 x 10 4 cells per well) . After 60 minutes in the tissue culture incubator, the virus was aspirated from each well and replaced with 150 ⁇ l of IMEM/10%FBS.
- adenovirus vector AvllacZ4 to C57BL/6 mice via tail vein were administered.
- the vector inoculum ranged from 1 x 10 3 pfu to 1 x 10 8 pfu in single log increments.
- serum levels of anti-adenovirus neutralizing antibodies were determined (Fig. 18) for mice which received 1x10 s pfu or greater of vector. A minus sign indicates that none of the mice in the cohort had detectable antibody.
- C57BL/6 mice were immunosuppressed with either deoxyspergualin (DSG) , cyclophosphamide (Cy) , or dexamethasone (Dex) at the time of administration of 1 x 10 ⁇ pfu of AvllacZ4. As shown above, this dose completely prevented an effective second delivery. Mice were injected daily with 33 mg/kg of DSG, beginning one day before vector delivery and continuing for seven more days. Dexamethasone was delivered over the same time course, at a dose of 5 mg/kg. Cyclophosphamide was administered once, the day before vector delivery, at a dose of either 100 mg/kg or 300 mg/kg. Control mice received AvllacZ4 without immunosuppression, or were immunosuppressed without initial vector delivery.
- DSG deoxyspergualin
- Cy cyclophosphamide
- Dex dexamethasone
- each mouse received 1 x 10 8 pfu of AvlH9F2.
- human Factor IX plasma levels were determined by ELISA (Fig. 21) .
- Control mice which were not pre-immunized with AvlLacZ4, expressed an average of 9 ⁇ g/ml of human Factor IX.
- Other control mice which received AvllacZ4 but were not immunosuppressed, expressed no human Factor IX after AvlH9F2 administration.
- the one mouse which was immunosuppressed with 33 mg/kg DSG expressed 12 ⁇ g/ml of human Factor IX.
- DSG does not produce a general suppression of the immune system, but rather results in a selective lack of response to specific antigens presented at the time of drug treatment .
- immunosuppression with DSG over a 7 day period following vector delivery efficiently inhibited the humoral response to the vector and permitted an effective second administration.
- the initial experiment with DSG employed a high dose of 33 mg/kg, which is close to the maximum tolerated dose in mice and several fold higher than the doses used in human trials .
- lower doses of DSG were also effective in permitting repeat delivery of vector.
- a greater degree of individual variability in levels of Factor IX expre ⁇ ion was seen with reduced doses, although even at the lowest dose tested (5 mg/kg) significant Factor IX expression was obtained in 3 of 6 animals .
- Cyclophosphamide administered at a dose of 300 mg/kg the day before vector injection, was also effective in blocking the humoral response and allowed a completely effective second injection with a Factor IX adenovirus vector. Furthermore, a third injection with a Factor VIII encoding vector was also completely efficacious when the previous two vector administrations were each preceded by a single dose of cyclophosphamide. Cyclophosphamide is used clinically as an anti-cancer agent in the treatment of Hodgkins disease and other leukemias . It is also employed as an immunosuppres ⁇ ive agent in the treatment of hemophilia patients who develop inhibitors to Factor VIII protein replacement therapy. (Aledort, Am. J . Hemat .. Vol.
- mice While the dose used to successfully obtain readministration in mice is substantially higher than is generally used in humans, it remains to be established whether lower, clinically acceptable doses, might be effective in humans.
- One possibility suggested by experience in the organ transplantation setting is that combinations of immunosuppressants would yield more potent suppression of the immune system with less toxicity.
- cyclophosphamide may be effective at lower dose ⁇ when u ⁇ ed in combination with dexamethasone.
- AvlH9F2 used in this study, 1 x 10 8 pfu, yielded plasma levels of human Factor IX of 5-10 ⁇ g/ml, which is 20 to 50 times above a level that would be therapeutic in a hemophiliac.
- Vectors such as AvlH9F2, which express high levels of transgene product and which can be administered at relatively low doses, should reduce the extent of immune stimulation and the degree of immunosuppression required.
- This example describes transient suppression of the humoral immune system with a combination of DSG and cyclophosphamide.
- the data presented in this example demonstrate that these two drugs can be used in combination at clinically relevant doses to inhibit a humoral response directed against an adenoviral vector and thus allow multiple repeat administrations of vector.
- the adenoviral vector employed in this example was Av3nLacZ.
- Av3nLacZ is described in Example 2 of U.S. Patent Application Serial No. 08/458,403, entitled "Improved Adenoviral Vectors and Producer Cells," filed June 2, 1995, the contents of which are incorporated herein by reference.
- mice treated in this example and a general outline of the treatment regimen for each cohort are given in Table II below.
- mice which received a first injection of Av3nLacZ without immunosuppression received a second injection of 1 x 10 8 pfu of Av3nLacZ without immunosuppression (cohort 4) .
- All of the mice which received AvlH9F2 were bled one week after vector admini ⁇ tration and the plasma levels of human Factor IX were determined by ELISA. The results are shown in Figure 22.
- mice which received Av3nLacZ without immunosuppression all had undetectable levels of human Factor IX one week after AvlH9F2 delivery.
- the results corresponding to these mice are designated as "2 nd administration" in Figure 22.
- immunosuppre ⁇ sion with a combination of DSG and cyclophosphamide enabled a completely efficacious second administration of vector.
- mice which received two injections of Av3nLacZ were bled five weeks after the second vector administration and plasma levels of anti-adenovirus neutralizing antibodies for three mice from each cohort were determined.
- the mice which received two injections of Av3nLacZ without immunosuppression had relatively high antibody titers, while mice immunosuppressed with DSG and cyclophosphamide at the time of both " adenovira vector administrations had undetectable levels .
- mice 1 and 4-6 The twelve mice which received two injections of Av3nLacZ (cohorts 1 and 4) , plus six naive mice (cohort 6) , each received 1 x 10 8 pfu of AvlH9F2 thirty-five days after the second Av3nLacZ administration. One week later, the mice were bled and plasma levels of human Factor IX were determined by ELISA.
- Figure 22 is a graph of the results.
Abstract
A method of effecting a gene therapy treatment in a host which comprises the steps of: (a) administering to a host concurrently (i) an adenoviral vector including at least one DNA sequence encoding a therapeutic agent, (ii) deoxyspergualin, and (iii) cyclophosphamide; (b) discontinuing the administration of said adenoviral vector and said deoxyspergualin, and said cyclophosphamide; and (c) repeating the administration of the adenoviral vector deoxyspergualin, and the cyclophosphamide at least once. The repeated course of treatment of administration of an adenoviral vector, deoxyspergualin, and the cyclophosphamide provides for continued or increased expression of the at least one DNA sequence encoding the therapeutic agent.
Description
GENE THERAPY INVOLVING CONCURRENT AND REPEATED ADMINISTRATION OF ADENOVIRUSES AND IMMUNOSUPPRESSIVE AGENTS
This application is a continuation-in-part of PCT Application Serial No. PCT/US95/13253, filed with the United States Receiving Office on October 19, 1995, which is a continuatio -i -part of Application Serial No. 08/478,482, filed June 7, 1995, which is a continuation-in-part of Application Serial No. 08/325,679, filed October 19, 1994, the disclosures of which are incorporated by reference.
This invention relates to gene therapy comprising the use of adenoviruses as the gene delivery vehicles . More particularly, this invention relates to gene therapy involving the concurrent and repeated administration of adenoviruses and immunosuppressive agents, whereby the efficiency of the gene therapy treatment is enhanced through suppression of an immune response against the adenoviruses.
BACKGROUND OF THE INVENTION Adenovirus genomes are linear, double-stranded DNA molecules of approximately 36 kilobase pairs. Each extremity of the viral genome has a short sequence known as the inverted terminal repeat (or ITR) , which is necessary for viral replication. The well-characterized molecular genetics
of adenovirus render it an advantageous vector for gene transfer. Portions of the viral genome can be substituted with DNA of foreign origin. In addition, recombinant adenoviruses are structurally stable.
Adenoviruses can be very efficient in gene transfer into cells in vivo, and, thus may be employed as delivery vehicles for introducing desired genes into eukaryotic cells, whereby the adenovirus delivers such genes to eukaryotic cells by binding cellular receptors. There are, however, several limitations to adenovirus gene transfer which are due in part to host responses directed at either the adenovirus vector particle, breakdown products of the vector particle, or the transduced cells. These host responses include non-specific responses and specific immune responses. The non-specific responses include inflammatory and non-inflammatory changes. An example of the latter is a change in host cell gene expression. Specific immune responses include various cellular responses and humoral antibody responses. Cellular responses include those mediated by T-helper lymphocytes, T- suppressor lymphocytes, cytotoxic T lymphocytes (CTL) , and natural killer cells.
Despite the high efficiency of adenovirus vector mediated gene transfer, the transient nature of adenovirus vector mediated gene transfer has suggested that repeat administrations of adenovirus vectors may be necessary. Recent studies in cotton rats, however, have demonstrated that host immune responses directed towards adenoviral vectors correlate with decreased efficiency of gene transfer and expression after repeated administration. Yei et al . , Gene Therapy, 1:192-200 (1994).
Smith, et al . , Nature Genetics , Vol. 5, pgs . 397-402 (1993) discloses the administration to mice of an adenoviral
vector including a human Factor IX gene . Such administration resulted in efficient liver transduction and plasma levels of human Factor IX that would be therapeutic for hemophilia B patients. Human Factor IX levels, however, slowly declined to baseline by nine weeks after injection, and were not reestablished by a second vector injection. Smith, et al . , also found that neutralizing antibodies to adenovirus block successful repeat administration of the adenovirus .
Kozarsky, et al . , J. Biol . Chem. , Vol. 269, No. 18, pgs . 13695-13702 (May 6, 1994) discloses the infusion of an adenoviral vector including DNA encoding the LDL receptor to rabbits . Stable expression of the LDL receptor gene was found in the rabbits for 7 to 10 days, and diminished to undetectable levels within 3 weeks . The development of neutralizing antibodies to the adenovirus resulted in a second dose being completely ineffective.
Kass-Eisler, et al . , Gene Therapy, Vol. 1, pgs. 395-402 (1994) suggest that a T-cell response contributes to, but is not responsible solely for, the limited duration of expression in adults from adenovirus vectors. The authors further show that cyclosporin A is not effective in blocking the humoral response to the vector.
Fang, et al . , J. Cell. Biochem. , Supplement 21A, C6-109, pg. 363 (1995) disclose the attempted re-injection of an adenovirus vector in dogs which were treated with cyclosporin A, an immunosuppressive agent. Such attempted re-injection was unsuccessful .
Yang, et al . , Proc . Nat. Acad. Sci . , Vol. 91, pgs. 4407- 4411 (May 1994) describe recombinant adenoviruses in which the Ela and Elb regions have been deleted. Such viruses also include a transgene. When such adenoviruses are administered
-3-
SUBST1TUTE SHEET (RULE 26)
to an animal host, cells harboring the recombinant viral genome express the transgene as desired; however, low level expression of viral genes also occurs. A virus-specific cellular immune response is stimulated that leads to destruction of the genetically modified cells, thereby limiting the duration of expression of the transgene.
The aforementioned studies clearly document the need for a method of circumventing or blocking the host immune response to adenovirus vectors which prevents effective readministration of the vector, and limits the effectiveness of expression; however, they do not describe how to accomplish this .
It is therefore an object of the present invention to provide for sustained efficacy of gene transfer via repeated administration of adenoviral vectors, and for sustained expression of the transferred gene, through the suppression of an immune response against the adenoviral vectors .
BRIEF DESCRIPTION OF THE DRAWINGS
The invention now will be described with respect to the drawings, wherein:
Figure 1 is a schematic of the construction of plasmid pHR.
Figure 2 is a schematic of the construction of an expression vehicle including an adenoviral ITR, an encapsidation signal, a Rous Sarcoma Virus promoter, an adenoviral tripartite leader sequence, and linking sequences;
Figure 3 is a schematic of the construction of plasmid pAvS6;
Figure 4 is a map of plasmid pAvS6;
Figure 5 is a map of plasmid pBQ4.7 ;
Figure 6 is a map of plasmid pAvS6 - CFTR;
Figure 7 is a schematic of adenoviral vectors AvlLucl and AvlCf2;
Figure 8 is a map of plasmid pGEM-luc;
Figure 9 is a map of plasmid pAVS6-luc;
Figures 10A, 10B, and IOC depict the histologic appearance of the lung in response to AvlCf2 administration three days after vector administration;
Figures 11A and 11B are graphs showing the effect of dexamethasone administration on lung lavage cells at 3 days and 42 days after the administration of AvlCf2;
Figure 12 is a graph of anti-adenoviral antibody titers of lung lavage samples from rats infected with AvlCf2 and which were treated or not treated with dexamethasone; and
Figure 13 is a graph of CTL responses in rats 42 days after infection with AvlCf2 ;
Figure 14 is a graph of luciferase enzyme activity in rats infected with AvlCf2 and which were treated or not treated with dexamethasone, followed by infection with AvlLucl;
Figure 15 is a map of plasmid pAvlH9FR;
Figure 16 is a schematic of the adenoviral vector AvlH9F2 ;
Figure 17 is a graph of plasma human Factor IX levels in mice which were given AvlH9F2 , and were or were not given one of the immunosuppressive agents deoxyspergualin, cyclophosphamide, or dexamethasone with or without the administration of the vector AvlLacZ4 five weeks earlier;
Figure 18 is a graph of plasma Factor IX levels (ng/ml) in mice which received from lxlO3 to lxlO8 pfu of AvlLacZ4 , followed by administration of AvlH9FR five weeks later;
Figure 19 is a graph of neutralizing antibody titer in mice that were given AvlLacZ4 , and received no immunosuppression, or were treated with deoxyspergualin, cyclophosphamide, or dexamethasone;
Figure 20 is a graph of plasma Factor VIII levels in mice which were given AvlLacZ4 , and received no immunosuppression, or were given cyclophosphamide, followed by administration of AvlH9F2 with or without cyclophosphamide, followed by administration of AvlALAPHβl;
Figure 21 is a graph of plasma Factor IX levels (ng/ml) in mice which received AvlLacZ4 and 0 mg/kg, 5 mg/kg, 10 mg/kg, 20 mg/kg, or 33 mg/kg deoxyspergualin, followed by administration of AvlH9F2 ; and
Figure 22 is a graph of plasma Factor IX levels (μg/ml) in mice which received Av3nLacZ and/or AvlH9F2, and which received no immunosuppression, or received one or two courses of immunosuppression with deoxyspergualin and cyclophosphamide .
DETAILED DESCRIPTION OF THE INVENTION
In accordance with an aspect of the present invention, there is provided a method of effecting a gene therapy treatment in a host . The method comprises administering to a host (i) an adenoviral vector including at least one DNA sequence and (ii) an immunosuppressive agent. The course of administration of the adenoviral vector and immunosuppressive agent then is discontinued. Administration of the immunosuppressive agent and the adenoviral vector then is repeated at least once. The adenoviral vector is administered in an amount effective to produce a therapeutic effect in the host. The immunosuppressive agent is administered in an amount effective to prevent or suppress a humoral and/or cellular immune response to the vector and/or cells containing the vector.
The term "DNA sequence" as used herein, refers generally to a polydeoxyribonucleotide molecule and more specifically to a linear series of deoxyribonucleotides connected one to the other by phosphodiester bonds between the 3' and 5' carbons of the adjacent pentoses .
Applicants have found that, when an immunosuppressive agent is administered with the adenoviral vector, and then administration of the vector is repeated, one achieves enhanced efficacy of the repeat in vivo adenoviral-mediated gene transfer through suppression of an immune response (such as a humoral antibody response) against the adenoviral vector and/or cells transduced with the vector, and thereby achieves increased expression of the transferred genes.
The adenoviral vector which is employed may, in one embodiment , be an adenoviral vector which includes essentially the complete adenoviral genome. Shenk et al . , Curr. Top. Microbiol . Immunol . , 111 (3) :l-39 (1984).
Alternatively, the adenoviral vector may be a modified adenoviral vector in which at least a portion of the adenoviral genome has been deleted.
In another embodiment, the adenoviral vector comprises an adenoviral 5' ITR; an adenoviral 3' ITR; an adenoviral encapsidation signal; at least one DNA sequence encoding a therapeutic agent; and a promoter controlling the at least one DNA sequence encoding the therapeutic agent . The vector is free of at least the majority of adenoviral El and E3 DNA sequences, but is not free of all of the E2 and E4 DNA sequences , and DNA sequences encoding adenoviral proteins promoted by the adenoviral major late promoter.
In one embodiment, the vector also is free of at least a portion of at least one DNA sequence selected from the group consisting of the E2 and E4 DNA sequences.
In another embodiment, the vector is free of at least the majority of the adenoviral El and E3 DNA sequences, and is free of a portion of the other of the E2 and E4 DNA sequences .
In still another embodiment, the gene in the E2a region that encodes the 72 kilodalton binding protein is mutated to produce a temperature sensitive protein that is active at 32°C, the temperature at which the viral particles are produced. This temperature sensitive mutant is described in Ensinger et al . , J. Virology, 10:328-339 (1972); Van der Vliet, et al . , J. Virology, 15:348-354 (1975); and Friefeld et al., Virology, 124:380-389 (1983); Englehardt, et al . , Proc.Nat. Acad. Sci., Vol. 91, pgs. 6196-6200 (June 1994); Yang, et al . , Nature Genetics. Vol. 7, pgs 362-369 (July 1994) .
-8-
SUBSTΓΓUTE SHEET(RULE 26)
Such a vector, in a preferred embodiment, is constructed first by constructing, according to standard techniques, a shuttle plasmid which contains, beginning at the 5' end, the "critical left end elements," which include an adenoviral 5' ITR, an adenoviral encapsidation signal, and an Ela enhancer sequence; a promoter (which may be an adenoviral promoter or a foreign promoter) ; a multiple cloning site (which may be as hereinabove described) ; a poly A signal; and a DNA segment which corresponds to a segment of the adenoviral genome . The vector also may contain a tripartite leader sequence . The DNA segment corresponding to the adenoviral genome serves as a substrate for homologous recombination with a modified or mutated adenovirus, and such sequence may encompass, for example, a segment of the adenovirus 5 genome no longer than from base 3329 to base 6246 of the genome. The plasmid may also include a selectable marker and an origin of replication. The origin of replication may be a bacterial origin of replication. Representative examples of such shuttle plasmids include pAvS6, shown in Figure 4. A desired DNA sequence encoding a clotting factor may then be inserted into the multiple cloning site to produce a plasmid vector.
This construct is then used to produce an adenoviral vector. Homologous recombination is effected with a modified or mutated adenovirus in which at least the majority of the El and E3 adenoviral DNA sequences have been deleted. Such homologous recombination may be effected through co- transfection of the plasmid vector and the modified adenovirus into a helper cell line, such as 293 cells, by CaP04 precipitation. Upon such homologous recombination, a recombinant adenoviral vector is formed that includes DNA sequences derived from the shuttle plasmid between the Not I site and the homologous recombination fragment, and DNA derived from the El and E3 deleted adenovirus between the homologous recombination fragment and the 3' ITR.
In one embodiment, the homologous recombi{r!crt*£©TI rragrtiefTt overlaps with nucleotides 3329 to 6246 of the adenovirus 5 (ATCC VR-5) genome.
Through such homologous recombination, a vector is formed which includes an adenoviral 5' ITR, an adenoviral encapsidation signal; an Ela enhancer sequence; a promoter; at least one DNA sequence encoding a therapeutic agent ; a poly A signal; adenoviral DNA free of at least the majority of the El and E3 adenoviral DNA sequences; and an adenoviral 3' ITR. The vector also may include a tripartite leader sequence . This vector may then be transfected into a helper cell line, such as the 293 helper cell line (ATCC No. CRL1573) , which will include the Ela and Elb DNA sequences, which are necessary for viral replication, and to generate infectious adenoviral particles. Transfection may take place by electroporation, calcium phosphate precipitation, microinjection, or through proteoliposomes .
The vectors hereinabove described may include a multiple cloning site to facilitate the insertion of the at least one DNA sequence encoding a therapeutic agent into the cloning vector. In general, the multiple cloning site includes "rare" restriction enzyme sites; i.e., sites which are found in eukaryotic genes at a frequency of from about one in every 10,000 to about one in every 100,000 base pairs. An appropriate vector in accordance with the present invention is thus formed by cutting the cloning vector by standard techniques at appropriate restriction sites in the multiple cloning site, and then ligating the DNA sequence encoding a therapeutic agent into the cloning vector.
The adenoviral vector, as stated hereinabove, includes at least one DNA sequence encoding at least one therapeutic agent. The term "therapeutic" is used in a generic sense and
includes treating agents, prophylactic agents, and replacement agents.
DNA sequences encoding therapeutic agents which may be placed into the adenoviral vector include, but are not limited to, DNA sequences encoding tumor necrosis factor (TNF) genes, such as TNF-α; genes encoding interferons such as Interferon-of, Interferon-β, and Interferon-γ ; genes encoding interleukins such as IL-1, IL-lβ, and Interleukins 2 through 14; genes encoding GM-CSF; genes encoding adenosine deaminase, or ADA; genes encoding antioxidants such as Mn- SOD, catalase, CuZnSOD, extracellular superoxide dismutase, and glutathione reductase; genes which encode cellular growth factors, such as lymphokines, which are growth factors for lymphocytes; genes encoding growth factors such as epithelial growth factor (EGF) and keratinocyte growth factor (KGF) ; genes encoding soluble CD4; Factor VIII; Factor IX; von Willebrand's factor; T-cell receptors; the LDL receptor, ApoE, ApoC, ApoAI and other genes involved in cholesterol transport and metabolism; the alpha-1 antitrypsin (orlAT) gene, the ornithine transcarbamylase (OTC) gene, the CFTR gene, lung surfactant protein genes, the B-glucuronidase gene, the insulin gene, negative selective markers or "suicide" genes, such as viral thymidine kinase genes, such as the Herpes Simplex Virus thymidine kinase gene, the cytomegalovirus virus thymidine kinase gene, and the varicella-zoster virus thymidine kinase gene; Fc receptors for antigen-binding domains of antibodies, and antisense sequences which inhibit viral replication, such as antisense sequences which inhibit replication of hepatitis B or hepatitis non-A non-B virus; tissue plasminogen activator (tPA) ; urinary plasminogen activator (urokinase) ; hirudin; nitric oxide synthase; vasoactive peptides; and angiogenic peptides .
The DNA sequence encoding a therapeutic agent is under the control of a suitable promoter. Suitable promoters which may be employed include, but are not limited to, adenoviral promoters, such as the adenoviral major late promoter; or heterologous promoters, such as the cytomegalovirus (CMV) promoter; the Rous Sarcoma Virus (RSV) promoter; inducible promoters, such as the MMTV promoter, the metallothionein promoter; heat shock promoters; the albumin promoter; and the ApoAI promoter. Alternatively, the DNA sequence encoding a therapeutic agent may be under the control of its native promoter. It is to be understood, however, that the scope of the present invention is not to be limited to specific foreign genes or promoters .
Immunosuppressive agents which may be employed include those which prevent: (i) a humoral (antibody) response against the adenoviral vector; (ii) a cellular immune response, such as, for example, a T-cell response to cells containing the adenoviral vector; or (iii) a non-specific inflammatory responses against the vector and against cells containing the vector. By preventing a humoral and/or T-cell and/or non-specific inflammatory response against the vector, and/or cells containing the vector administration of the immunosuppressive agent permits effective re-administration of the vector in order to produce a therapeutic effect in the host. Preferably, where repeat administration of the adenoviral vector is desired, the immunosuppressive agent is an immunosuppressive agent which prevents a humoral antibody response against the adenoviral vector. Preferably, where longer duration or higher levels of expression is desired, the immunosuppressive agent is one that prevents or suppresses a cellular or non-specific inflammatory response.
Host immune responses to in vivo adenovirus vector administration vary in relation to (i) the dose of vector;
(ii) the route of administration; (iii) the level of replication (if any occurs) ; (iv) the nature of the transgene contained in the recombinant vector; (v) the genetic and physiological characteristics of the host; and (vi) the existence and level of pre-existing immune responses to previously administered adenovirus vectors.
In general, host responses are dependent on the dose of vector administered. Importantly, the magnitude of specific host responses is dependent on the route of vector administration. For example, intravenous administration will yield a higher host antibody response than that of an equivalent amount of vector given via the respiratory route .
The diverse host responses to adenovirus vectors occur due to separate inflammatory and immune effector mechanisms, although most, if not all, of these distinct molecular mechanisms are connected and significantly interdependent. Thus, for example, humoral antibody formation is very dependent on certain T-helper lymphocyte support. Also, some cell-mediated cellular toxicity is dependent on antibody formation, e.g., opsonized macrophage cell killing.
The two principal host responses affecting the duration of transgene expression are the inflammatory response and the cellular immune response.
Inflammation is one of the first host responses that occurs following vector administration. Cytokine release is very likely involved in the subsequent influx of inflammatory cells. Such cytokines likely include IL-1, IL-6, IL-8, and TNF.
The amount of inflammation seen following in vivo adenoviral vector administration increases with incresing doses of vector given. Higher doses lead to a more rapid decline in transgene expression than do smaller doses .
CTL responses directed towards the transduced cells are believed to be important in reducing the duration of transgene expression. It is believed that the CTL are directed against low level adenovirus gene expression by the cells , which induces the CTL .
The principal host response affecting the ability to administer adenovirus vectors repeatedly to a host is the humoral antibody response. It develops to adenovirus administered by a variety of routes, including oral, intravenous, intraperitoneal, and intrapulmonary . In general , the level of antibody response achieved is very dependent on the dose of vector administered. The antibody response is also dependent on the route of vector administration. Intravenous vector administration results in higher antibody levels than pulmonary administration for a given dose of vector. In contrast, wild type adenovirus elicits high antibody levels irrespective of the amount of virus given due to virus replication in vivo. The ability to repeat successfully adenovirus vector administration is inversely correlated with the level of circulating anti- adenovirus vector antibody present.
Pharmacologic modulation of host immune responses to adenovirus vectors involves the use of anti-inflammatory agents, celllular immune modifiers, and humoral antibody immune modifiers.
Anti- inflammatory agents include steroids, cyclophosphamide , and azothiophrine .
Steroids have potent anti-inflammatory properties. Applicants have shown that steroids, such as dexamethasone, given parenterally prolong the duration of transgene expression following in vivo administration of vector via the lung route . Steroids also block the function of lymphocytes . Thus, dexamethasone reduces the CTL responses observed after
pulmonary vector administration. Dexamethasone also blocks, at least in part, the host antibody response to adenovirus vector administration.
Antibodies directed at cellular components of the immune system reduce cellular immune response. For example, anti T- cell receptor antibody (such as, for example anti-CD4 and anti-CD3 antibodies) administration prolongs transgene expression. CTLA4 immunoglobulin is another example. Anti- CD4 antibody is directed against the T-helper lymphocytes and reduces their function. Other agents directed primarily at the cellular immune response include cyclosporins such as cyclosporin A; rapamycin binding protein ligandε such as FK506; and steroids such as dexamethasone.
Agents which affect humoral antibody responses are generally directed at antibody producing B lymphocytes (B- cells) or at the T-cells which are responsible for inducing B-cell antibody production to high levels.
Examples of immunosuppressive agents which prevent a humoral antibody response against the adenoviral vector include, but are not limited to, deoxyspergualin, or DSG, which has the following structure : H2NCNH (CH2) 4CH2CH2CONHCHCONH(CH2) 4NH(CH2) 3NH2
II I
NH OH
The terms "deoxyspergualin" and "DSG" as used herein, mean deoxyspergualin or DSG and derivatives or analogues thereof, such as salts of deoxyspergualin, including but not limited to, trihydrochlorides thereof, and any other analogues which have immunosuppressive activity. Such compounds are described further in U.S. Patent Nos . 4,525,299; 4,847,299; 5,162,581; and 5 , 196, 453.
In one embodiment, the immunosuppressive agent which prevents a humoral antibody response is a steroid. Steroids which may be employed include, but are not limited to, dexamethasone, and any adrenocortical hormones, such as, for example corticosteroids; hydrocortisone; prednisolone; and methylprednisolone .
In another embodiment, the immunosuppressive agent which prevents a humoral antibody response is a cyclosporin, such as, for example, cyclosporin A. Other immunosuppressive agents which prevent a humoral antibody response and which may be employed include, but are not limited to, azathioprine ; cyclophosphamide; brequinar; leflunomide; mycophenolate mofetil; anti-CD40 antibody; anti-CD40 ligand antibody; cyclophosphamine; rapamycin; anti-CD4 antibody; CTLA-4 immunoglobulin; Interleukin -12; Interferon -γ; rapamycin binding protein (FEBP) ligands, such as, for example, FK506, as described in Bierer, et al . , Proc . Nat . Acad. Sci.. Vol. 87, pgs. 9231-9235 (1990); Dumont, et al . , J . Immunol .. Vol. 144, pgs. 1418-1424 (1990); and Bierer, et al . , Science, Vol. 250, pgs. 556-559 (1990); anti-lymphocyte function antigen-1 (LFA-1) antibody; and anti-T-cell receptor antibody.
Applicants have found that, when compounds which prevent, suppress, or eliminate humoral immune responses to foreign antigens (such as, for example, deoxyspergualin, cyclophosphamide, brequinar, leflunomide, mycophenolate, mofetil, anti-CD40 antibody, or anti-CD40 ligand antibody) are administered at a short time prior to, and/or during, and/or for a short time after adenoviral vector administration, to a host, such compounds prevent the production of anti-adenoviral neutralizing antibodies in the host. The prevention of the production of such neutralizing
antibodies enables the efficient re-administration of the adenoviral vector to the host .
It is to be understood that , within the scope of the present invention, that an immunosuppressive agent may prevent more than one of the immune responses hereinabove described. It also is to be understood, however, that the scope of the present invention is not intended to be limited to any specific immunosuppressive agents.
It is also contemplated that within the scope of the present invention, a combination of immunosuppressive agents may be employed. In a preferred embodiment, a combination of deoxyspergualin and cyclophosphamide is administered to the host .
The adenoviral vector and immunosuppressive agent, in general, are administered concurrently in an amount effective to produce a therapeutic effect in the host while preventing an immune response against the vector or against cells transduced with the vector. The term "concurrently," as used herein, means that the administration of the adenoviral vector and administration of the immunosuppressive agent are begun at approximately the same time, i.e., within a brief time frame of each other, and the administration of the adenoviral vector and the administration of the immunosuppressive agent are parts of a unitary course of treatment. Thus, for example, the immunosuppressive agent is administered at approximately the same time the adenoviral vector is administered, i.e., the administration of the immunosuppressive agent is begun at a short time (for example, about 24 hours) before, or during, or at a short time (e.g., 24 hours) after the administration of the adenoviral vector. In general, the immunosuppressive agent is administered according to standard dosage schedules
established for that agent, and for a period of time whic ^Λ- general does not exceed 14 days, and preferably does not exceed 11 days, and more preferably does not exceed 8 days. Thus, long-term administration of the immunosuppressive agent is not required for enabling repeated administration of the adenovirus .
At the conclusion of the course of administration of the immunosuppressive agent, the course of administration of the adenoviral vector and immunosuppressive agent is discontinued for a period of time. The period of time between courses of administration of the adenoviral vector and the immunosuppressive agent, and the number of courses of administration of the adenoviral vector and immunosuppressive agent is dependent upon a variety of factors, including the age, weight, and sex of the patient, the disease or disorder being treated, and the severity of the disease or disorder being treated.
It is to be understood that the above course of administration of the immunosuppressive agent is repeated with each administration of the adenoviral vector.
In one embodiment, the adenoviral vector may be administered, at each administration, in an amount of from 1 plaque forming unit to about 1014 plaque forming units, preferably from about 106 plaque forming units to about 1013 plaque forming units, more preferably from about 108 to about 1010 plaque forming units per kg. The host may be a human or non-human animal host.
The adenoviral vector may be administered systemically or topically. Examples of systemic administration include, but are not limited to, intravenous administration (such as for example, portal vein injection or peripheral vein
injection) , intramuscular administration, intraperitoneal administration, intranasal administration, or encapsulated oral administration.
The immunosuppressive agent is administered in an amount effective to produce a desired immunosuppressive effect in the host. The immunosuppressive agent may be administered, at each administration, in an amount of from about 1 mg/kg to about 15 mg/kg, preferably from about 2.25 mg/kg to about 5.0 mg/kg, more preferably at about 2.5 mg/kg, when dexamethasone is employed, or at the dose equivalents for other steroids. When methylprednisolone is employed, the methylprednisolone may be administered in an amount of from about 1 mg/kg to about 33 mg/kg, preferably from about 3 mg/kg to about 7 mg/kg. When cyclophosphamide is employed, the cyclophophamide may be administered in an amount of from about 5 mg/kg to about 300 mg/kg, preferably from about 50 mg/kg to about 100 mg/kg. When methylprednisone is employed, the methylprednisone may be administered in an amount of from about 15 mg/kg to about 30 mg/kg, preferably at about 30 mg/kg.
The adenoviral vector particles and the immunosuppressive agent each may be administered in combination with a pharmaceutically acceptable carrier suitable for administration to a patient. The carrier may be a liquid carrier such as, for example, a saline solution. The adenoviral vector particles also may be administered in combination with a solid carrier, such as, for example, microcarrier beads, or a sustained drug delivery material, such as, for example, a polyol.
Cells which may be transduced by the adenoviral particles include, but are not limited to, lung, airway, or alveolar epithelial cells; primary cells, such as primary
nucleated blood cells, such as leukocytes, g'rahulocytes , monocytes, macrophages, lymphocytes (including T- lymphocytes and B-lymphocytes) , totipotent stem cells, and tumor infiltrating lymphocytes (TIL cells); bone marrow cells; endothelial cells; activated endothelial cells; epithelial cells; keratinocytes; stem cells; hepatocytes, including hepatocyte precursor cells; fibroblasts; mesenchymal cells; mesothelial cells; parenchymal cells; vascular smooth muscle cells; brain cells and other neural cells; gut enterocytes; gut stem cells; and myoblasts.
In one embodiment, the adenoviral particles may be targeted to blood cells, whereby such adenoviral vector particles infect the blood cells with a gene which directly or indirectly enhances the therapeutic effects of the blood cells. The gene carried by the blood cells can be any gene which allows the blood cells to exert a therapeutic effect that it would not ordinarily have, such as a gene encoding a clotting factor useful in the treatment of hemophilia. The gene can encode one or more products having therapeutic effects. Examples of suitable genes include those that encode the CFTR gene; cytokines such as TNF, interleukins (interleukins 1-14) , interferons ( , β, γ-interferons) , T- cell receptor proteins and Fc receptors for antigen-binding domains of antibodies, such as immunoglobulins . Other examples of suitable genes include genes encoding soluble CD4 which is used in the treatment of AIDS and genes encoding a- antitrypsin, which is useful in the treatment of emphysema caused by α-antitrypsin deficiency.
The transduced cells are useful in the treatment of a variety of diseases including but not limited to, cystic fibrosiε, adenosine deaminase deficiency, sickle cell anemia, thalassemia, hemophilia, diabetes, α-antitrypsin deficiency, brain disorders such as Alzheimer's disease,
phenylketonuria and other illnesses such as growth disorders and heart diseases, for example, those caused by alterations in the way cholesterol is metabolized and defects of the immune system.
In another embodiment, the adenoviral vector particles may transduce liver cells, and such adenoviral vector particles may include genets) encoding polypeptides or proteins which are useful in prevention and therapy of an acquired or an inherited defect in hepatocyte (liver) function. For example, they can be used to correct an inherited deficiency of the low density lipoprotein (LDL) receptor, and/or to correct an inherited deficiency of ornithine transcarbamylase (OTC) , which results in congenital hyperammonemia .
In another embodiment, the adenoviral particles may transduce liver cells, whereby the adenoviral particles include a gene encoding a therapeutic agent employed to treat acquired infectious diseases, such as diseases resulting from viral infection. For example, the infectious adenoviral particles may be employed to treat viral hepatitis, particularly hepatitis B or non-A non-B hepatitis. For example, an infectious adenoviral particle containing a gene encoding an antisense gene could be employed to infect liver cells to inhibit viral replication. In this case, the infectious adenoviral particle, which includes a vector including a structural hepatitis gene in the reverse or opposite orientation, would be introduced into liver cells, resulting in production in the infected liver cells of an anti-sense gene capable of inactivating the hepatitis virus or its RNA transcripts. Alternatively, the liver cells may be infected with an infectious adenoviral particle which includes a gene which encodes a protein, such as, for
example, a-interferon, which may confer resistance to the hepatitis virus.
The vector particles also may be employed in treating Hodgkin's lymphoma. An infectious adenoviral vector particle may be targeted to neoplastic cells of Hodgkin's lymphoma. The adenoviral vector particle also includes a negative selective marker or "suicide gene, such as the Herpes Simplex thymidine kinase gene . The adenovirus may be administered in vivo to a patient, whereby the virus infects neoplastic cells of Hodgkin's lymphoma. After the adenovirus is administered to the patient, the patient is given an interaction agent such as gancyclovir or acyclovir, whereby the neoplastic Hodgkin's lymphoma cells infected with the adenovirus are killed.
In addition, a vector may be constructed which includes the CFTR gene . The vector then may be administered to the respiratory epithelium in an effective therapeutic amount for the correction of the pulmonary deficit in patients with cystic fibrosis. In another example, vectors containing functional proteins may be delivered to the respiratory epithelium in order to correct deficiencies in such proteins . Such functional proteins include antioxidants, ot- 1- antitrypsin, CFTR, lung surfactant proteins, cytokines , and growth factors such as EGF and KGF, and may also include adenosine deaminase for treatment of severe combined immune deficiency, von Willebrand's factor for treatment of Christmas disease, and 3-glucuronidase for treatment of Gaucher's disease. Also, vectors including genes encoding anti-cancer agents or anti-inflammatory agents may be administered to lung cells of a patient for the treatment of lung cancer or inflammatory lung disease .
EXAMPLES
The invention now will be described with respect to the following examples; however, the scope of the present invention is not intended to be limited thereby.
Example 1 Construction of AylCf2 and AylLucl A. Construction of pAvS6
The adenoviral construction shuttle plasmid pAvS6 was constructed in several steps using standard cloning techniques including polymerase chain reaction based cloning techniques. First, the 2913 bp Bglll, Hindlll fragment was removed from Ad-dl327 and inserted as a blunt fragment into the Xhol site of pBluescript II KS- (Stratagene, La Jolla, CA) (Figure 1) .
Ad-dl327 is identical to adenovirus 5 except that an Xbal fragment including bases 28591 to 30474 (or map units 78.5 to 84.7) of the Adenovirus 5 genome, and which is located in the E3 region, has been deleted. The E3 deletion in Ad-dl327 is similar to the E3 deletion in Ad-dl324, which is described in Thimmappaya et al . , Cell , 31:543 (1983) . The complete Adenovirus 5 genome is registered as Genbank accession #M73260, incorporated herein by reference, and the virus is available from the American Type Culture Collection, Rockville, Maryland, U.S.A. under accession number VR-5.
Ad-dl327 was constructed by routine methods from Adenovirus 5 (Ad5) . The method is outlined briefly as follows and previously described by Jones and Shenk, Cell 13:181-188, (1978). Ad5 DNA is isolated by proteolytic digestion of the virion and partially cleaved with Xba I restriction endonuclease. The Xba I fragments are then reassembled by ligation as a mixture of fragments. This results in some ligated genomes with a sequence similar to Ad5, except excluding sequences 28591 bp to 30474 bp. This
DNA is then transfected into suitable cells ( e. g. KB cells, HeLa cells, 293 cells) and overlaid with soft agar to allow plaque formation. Individual plaques are then isolated, amplified, and screened for the absence of the 1878 bp E3 region Xba I fragment .
The orientation of this fragment was such that the Bglll site was nearest the T7 RNA polymerase site of pBluescript II KS" and the Hindlll site was nearest the T3 RNA polymerase site of pBluescript II KS" . This plasmid was designated PHR. (Figure 1) .
Second, the ITR, encapsidation signal, Rous Sarcoma Virus promoter, the adenoviral tripartite leader (TPL) sequence and linking sequences were assembled as a block using PCR amplification (Figure 2) . The ITR and encapsidation signal (sequences 1-392 of Ad-dl327 [identical to sequences from Ad5, Genbank accession #M73260] incorporated herein by reference) were amplified (amplification 1) together from Ad-dl327 using primers containing NotI or Ascl restriction sites. The Rous Sarcoma Virus LTR promoter was amplified (amplification 2) from the plasmid pRC/RSV (sequences 209 to 605; Invitrogen, San Diego, CA) using primers containing an Ascl site and an Sfil site. DNA products from amplifications 1 and 2 were joined using the "overlap" PCR method (amplification 3) (Horton et al . , BioTechniques , 8:528-535 (1990)) with only the NotI primer and the Sfil primer. Complementarity between the Ascl- containing end of each initial DNA amplification product from reactions 1 and 2 allowed joining of these two pieces during amplification. Next the TPL was amplified (amplification 4) (sequences 6049 to 9730 of Ad-dl327 [identical to similar sequences from Ad5, Genbank accession #M73260] ) from cDNA made from mRNA isolated from 293 cells (ATCC Accession No.
CRL 1573) infected for 16 hrs . with Ad-dl327 using primers containing Sfil and Xbal sites respectively. DNA fragments from amplification reactions 3 and 4 were then joined using PCR (amplification 5) with the NotI and Xbal primers, thus creating the complete gene block.
Third, the ITR-encapsidation signal-TPL fragment was then purified, cleaved with NotI and Xbal and inserted into the NotI, Xbal cleaved PHR plasmid. This plasmid was designated pAvS6A" and the orientation was such that the NotI site of the fragment was next to the T7 RNA polymerase site (Figure 3) .
Fourth, the SV40 early polyA signal was removed from SV40 DNA as an Hpal-BamHI fragment, treated with T4 DNA polymerase and inserted into the Sail site of the plasmid pAvS6a- (Figure 3) to create pAvS6 (Figures 3 and 4) .
B. Construction of AylCf2 and AylLucl
AvlCf2 (Figure 7) (Yei et al . , Gene Therapy, 1:192-200 (1994) ) , an El-deleted (1.18 map units to 9.2 map units) , E3- deleted (78.5 map units to 84.7 map units) adenoviral vector constructed first by inserting the normal human CFTR cDNA coding sequence fragment into the EcoRV site of pAvS6 so that the 5' end of the CFTR coding sequence was closest to the Adenovirus 5 tripartite leader. The CFTR cDNA was removed as a PstI fragment (nucleotides 75 to 4,725; for numbering see GenBank Accession No. M28688) from the plasmid pBQ4.7 (Figure 5) (provided by L.-C. Tsui, The Hospital for Sick Children, Toronto, Canada), and inserted as a blunt fragment. The resulting plasmid, pAvS6-CFTR (Figure 6) was linearized with Kpnl and recombined with the large (35 kb) Clal fragment of Ad-dl327 in 293 cells as described in Trapnell, Advanced Drug Delivery Reviews, 12:185-199 (1993) to form AvlCf2 (Figure 7) .
After double-plaque purification, the identify of the clonal isolates was confirmed by Southern analysis, immunoprecipitation of CFTR, as previously described. (Tolstoshev, et al . , Proceedings of the Ninth Nagoya International Symposium on Cancer Treatment, September 17-18, 1993, Nagoya, Japan (in press) .
AvlLucl (Figure 7) (Yei et al . , Gene Therapy, Vol . 1 , pgs . 192-200 (1994) ) is an adenoviral reporter vector identical in genomic organization and sequence to AvlCf2, except that it expresses the firefly luciferase gene (Genbank Accession No. M15077) .
The firefly luciferase gene was obtained from pGEM-luc (Figure 8 - Promega) . pGEM-luc was digested with StuI and Hindlll in order to splice out the firefly luciferase gene.
The firefly luciferase gene was inserted into the EcoRV site of pAvS6 so that the 5' end of the firefly luciferase coding sequence was closest to the Adenovirus 5 tripartite leader. The resulting plasmid, pAvS6-Lucl (Figure 9) was linearized with Kpnl and recombined with the large (35 kb) Clal fragment of Ad-dl327 as hereinabove described. Clonal isolates then were identified as hereinabove described.
Both viral vectors were propagated, purified by double- banding in CsCl gradients, and titered in 293 cells as described in Rosenfeld et al . , Cell , 68:143-155 (1992).
Example 2 Adenoviral-mediated gene transfer with concurrent intermittent steroid administration Cotton rats (weight approximately 150g) were divided into four groups with 9 rats in each group. AvlCf2 was administered by intranasal inhalation (Yei et al . , Human Gene Therapy, Vol. 5, pgs. 731-744 (1994)) to the lungs of cotton rats at a low dose (108 pfu) or at a high dose (1010 pfu) , either with or without coadministration of dexamethasone by intraperitoneal injection in an amount of 2 mg/kg daily, beginning 1 day prior to and continuing for 10 days after administration of the vector. A control group of rats was given PBS instead of AvlCf2 , either with or without coadministration of dexamethasone as hereinabove described.
At 3 days and at 42 days after vector administration, 3 rats from each group were evaluated for host responses to the AvlCf2 vector. Evaluations of host responses included pulmonary histopathology appearance, total lung lavage cellularity, lung lavage anti-adenovirus antibody production, and cytotoxic T-lymphocyte (CTL) response.
Evaluation of pulmonary histopathology was performed as described in Yei et al . , Human Gene Therapy, Vol . 5 , pgs 731-744 (1994). The lungs were removed, fixed in 2.5% (wt . /vol . ) paraformaldehyde , 0.25% (wt . /vol . ) glutaraldehyde at 4°C overnight, embedded in paraffin, and 6μm sections were stained with hematoxylin and eosin.
The histologic appearance of the lungs of the cotton rats in response to AvlCf2 injection, with and without daily administration of immunosuppression therapy, at three days after adenoviral infection, is shown in Figures 10A, 10B, and IOC. Figure 10a is a section of the lung of a control rat that did not receive adenovirus and instead received PBS.
Figure 10B is a section of the lung of a rat which received AvlCf2 without immunosuppression therapy. Figure IOC is a section of the lung of a rat which received AvlCf2 with daily administration of dexamethasone. All sections have been magnified 100 times. As shown in Figures 10A, 10B, and IOC, there was less pulmonary parenchymal inflammation in the rat which received adenovirus and immunosuppressive therapy as compared with the control rat and the rat infected with adenovirus, but did not receive immunosuppression therapy.
Lung lavage fluid was collected by lavaging the lung with 4.0 ml of PBS, and the total number of cells determined by counting in a hemocytometer, or the cells were evaluated in cytocentrifuge preparations for the percentage of neutrophils by light microscopy.
Figure 11A is a graph of the lung lavage cell count from rats infected with AvlCf2 three days after infection, as compared with control rats which received PBS . The control rats either received dexamethasone or did not receive dexamethasone . Figure 11B is a graph of the lung lavage cell count from rats infected with AvlCf2 at 42 days after infection, as compared with control rats which received PBS. The rats either received dexamethasone or did not receive immunosuppressant therapy.
As shown in Figure 11A, dexamethasone significantly reduced the non-specific host cellular inflammatory responses (represented by total lung lavage cellularity) at three days after vector administration, which is the peak of inflammation .
Lung lavage anti-adenovirus production was measured by an ELISA assay carried out as follows.
lOμl of AvlLacZ4 (Yei, et al . , Human Gene Therapy, Vol. 5, pgs. 731-744 (1994)) at lxlO11 pfu/ml was added to 90 μl of double distilled H20 in a 0.5 ml Eppendorf tube. The tube was irradiated with ultraviolet light for 30 minutes in order to kill the adenovirus, and protein concentration was measured with a Bio-Rad kit. 8 ml of 0.1M Na2C03 (pH 9.6) was added to the tube to provide a protein yield of 10 μg/4 ml or 125 ng of adenoviral antigen per 50 μl per well.
50μl of antigen then was added to each well of a 96-well microtiter plate (Immulon 2) ; and the plate was incubated at 37°C for 1 hour, or room temperature for 2 hours, or at 4°C overnight . The plate then was washed twice with PBS or double distilled H20.
300μl of blocking buffer (1% BSA in PBS) was added to each well, and the plate was incubated for 1 hour at room temperature. The plate then was rinsed with double distilled H20.
Blocking agent then was added to the background wells . 50μl of antibody samples (i.e., lung lavage samples prepared as hereinabove described) then were added to coated wells at serial two-fold dilutions, beginning at 1/4 and ending at 1/8192. 50 μl of negative control samples of serum from an uninfected cotton rat were added to another set of coated wells at the same serial dilutions. The plate the was incubated for 2 hours at room temperature, and 300 μl of 0.05% Tween 20/PBS then was added. The plate was incubated for 5 minutes at room temperature, emptied, and 300μl of 0.05% Tween 20/PBS again was added. The plate again was incubated at room temperature for 5 minutes, and was emptied. The plate then was washed twice with double distilled H20 or PBS.
Peroxidase-labeled goat anti-hamster IgG (lOμg/lOμϊ) was diluted with 10 ml BSA and PBS to make a working solution of 1 mg/ml concentration. (1:1,000 dilution). 100 μl of this solution then was added to each well, and the plate was incubated at room temperature for 2 hours . The plate then was washed five times with 300 μl of 0.01% Tween 20/PBS. The plate then was emptied and dried.
100 μl of tetramethyl benzidine (TMB) substrate was added to each well at room temperate, and the color was developed immediately. The blue color was monitored by reading at OD650 with an ELISA reader. The reaction was stopped when the OD6S0 was 0.5 to 0.6 by adding 100 μl of TMB stop solution to the wells. The OD450 then was read between 5 minutes and 1 hour after stopping the reaction. The antibody titer is the reciprocal of the dilution that gave an OD value of 0.1 larger than background OD. Alternatively, antibody titer also may be determined as 3 standard deviations above the OD of non-specific background. The average results, expressed in antibody titer, for the rats in each group at 3 days and 42 days after vector administration are shown in Figure 12. As shown in Figure 12 , the rats which were given dexamethasone showed a decreased antibody titer 42 days after vector administration as compared with rats that were not given dexamethasone.
CTL assays were carried out at 42 days after vector administratio .
Sensitizer cells were prepared by infecting cotton rat lung fibroblasts with Ad-dl327 at a multiplicity of infection of 100. The cells were incubated for 3 days, and checked for hexon expression by FACS . The cells then were washed with PBS/EDTA, contacted with trypsin, washed, spun, and resuspended in 1 ml RPMI medium. The cells then were
irradiated with 13'7Cs at 5,000 rads in order to inactivate the DNA.
Spleens then were isolated from uninfected (control) rats and adenovirus-infected rats 42 days after infection. The spleens were kept in sterile HBSS and ice. 10 ml of HBSS then was injected into each spleen with a 25/27 gauge needle. The spleen was mashed, and filtered with a cell strainer into a 50 ml tube. The volume then was brought to 40 ml in RPMI plus 10% FCS. The tube was spun at 1,500 rpm for 10 minutes. Red blood cells then were lysed by adding 2.5 ml of ACK lysis buffer, and the liquid was swirled for less than 1 minute. The volume was brought up to 50 ml with RPMI-10. The tube then was spun again at 1,500 rpm for 10 minutes. The cell pellet then was resuspended, and cells were counted at a 1:10 dilution. The splenocytes then were plated with the sensitizer cells at a ratio of splenocytes to sensitizer cells of 4:1 in RPMI. The splenocytes and sensitizer cells were incubated at 37°C in the presence of 20-50 units/ml of Interleukin-2. Interleukin-2 was added daily for 5 to 6 days .
Target cells were prepared by infecting 3X106 cotton rat lung fibroblasts with Ad-dl327 at a multiplicity of infection of 100 for 1 hour. Culture medium is added to the cells, and 51Cr in an amount greater than 50 μCi is added for 18 hours.
Target cells are harvested by washing the cotton rat lung fibroblasts with EDTA/PBS, followed by trypsinization. The cells then were washed, spun, resuspended in 5 ml culture medium, and counted. The cells were resuspended to 105 cells/ml and 10* cells/0.1 ml well were used for the CTL assay.
Effector cells (i.e., the combination of splenocytes and sensitizing cells (also sometimes referred to as Es cells) were spun at 1,500 rpm for 10 minutes at 4°C. The cells were resuspended in 2 ml of HBSS-10, loaded onto 7 ml Ficoll Hypaque, and spun at 1,500 rpm for 10 minutes. The top portion (4 ml) was harvested, and 5 ml of culture medium was added. This material was spun, the cell pellet was saved, and resuspended in 1 ml of culture medium. The effector cells were counted by mixing 50 μl of effector cells with 50 μl Trypan blue .
The effector cells then were added to wells containing 10* target cells, at effector: target (E:T) ratios of 3.125, 6.25, 12.5, 25, 50, and 100. The cells then were spun at 500 rpm for 5 minutes. The cells then were incubated at 37°C for 4 hours. The cells then were spun, and 100 μl of supernatant was analyzed for 51Cr release with a WALLAC gamma counter. The average results for CTL response in splenocytes taken from infected rats (with and without dexamethasone treatment) , and from two uninfected control rats, are shown in Figure 13.
As shown in Figure 13 , a lower CTL response was obtained from splenocytes obtained from infected rats that were treated with dexamethasone.
At 42 days after vector administration, the remaining three rats in each group received an intranasal pulmonary administration of AvlLucl at a dose of 2xl09 pfu. Remaining control rats, which initially received PBS, also received AvlLucl at a dose of 2xl09 pfu. Lung lavage anti-adenovirus antibody production was evaluated three days after administration of AvlLucl, according to the procedure hereinabove described, and the results are shown in Figure 12. As shown in Figure 12, the rats which initially were
given 1010 pfu of AvlCf2, and were treated witή"d~exa eT.rrasbTϊe, showed a decreased antibody titer as compared with rats that were not given dexamethasone .
The efficacy of transfer and expression of the firefly luciferase gene was evaluated three days after administration by quantifying directly intrapulmonary luciferase enzyme activity in light units (lu) by routine luminometry as described in Yei et al . , Gene Therapy, Vol . 1 , pgs 192-200 (1994 ) . The results are shown in Figure 14. As shown in Figure 14, each dot represents the luciferase enzyme activity for one rat, and the cross-bar represents the mean for each group. The efficiency of repeat adenovirus-mediated gene transfer was significantly higher in the rats which received AvlCf2 and dexamethasone than those which did not receive dexamethasone at the time of the first adenoviral administration (11,786 ± 3523 lu vs. 622 ± 192 lu, respectively) . The efficiency of gene transfer from AvlLucl also was higher in the control group which initially received PBS in conjunction with dexamethasone.
Example 3
Suppression of humoral immune response with
DSG or high dose cyclophosphamide, permitting effective repeat administration of an adenoviral vector
This example describes the intravenous administration of the adenoviral vectors AvlLacZ4 and AvlH9F2 to C57BL/6 male mice (Harlan Sprague Dawley, Indianapolis, Indiana) at 5 to
6 weeks of age at the start of the experiment . AvlLacZ4 is an adenoviral vector which includes a nuclear targeted B- galactosidase gene, (lacZ) and is described in PCT application No. W095/09654, published April 13, 1995.
AvlH9F2 is constructed from a derivative of the adenoviral shuttle plasmid vector pAvlH9FR (Figure 15) , which includes
human Factor IX DNA, and is described in PCT application No. W094/29471, published December 22, 1994.
To construct AvlH9F2, the shuttle plasmid pAvlH9FR was digested with the restriction enzyme Sfil, the DNA ends were made blunt using T4 DNA Polymerase, and the DNA molecule was recircularized by ligation. Competent DH5 cells were transformed and ampicillin-resistant clones were amplified and screened by restriction enzyme digestion of miniprep DNA. A positive clone was identified and the resulting shuttle plasmid was referred to as pAvS17H9F.
Subsequently, 293 cells were cotransfected with pAvS17H9F and the large DNA fragment of Clal digested Ad- dl327. Recombinant adenoviral vector plaques were picked, expanded, and screened for expression of Factor IX by ELISA. A positive clone was identified and amplified, thus generating the vector AvlH9F2. A schematic of the left end of the vector is shown in Figure 16. AvlH9F2 has a base pair deletion at the beginning of the tripartite leader, or TPL, which effectively changes the ATG into a CTG. The structure of the vector was verified by restriction enzyme diagnostics and by DNA sequence analysis of the region between the RSV promoter and the 3 ' untranslated region of the Factor IX cDNA.
Fifteen mice were immunosuppressed with 33 mg/kg of deoxyspergualin DSG (Nippon Kayaku Co. LTD, Tokyo, Japan) , delivered intraperitoneally (ip) , once daily, beginning the day before vector administration and continuing for a total of eight days. A vial containing 100 mg of lyophilized DSG was reconstituted with 3.8 ml of injection grade water to yield a 25 mg/ml solution, which was aliquoted and frozen at -20°C. Each day, immediately before immunosuppression, an aliquot was thawed at room temperature and 0.7 ml was mixed
with 6.3 ml of Hank's Balanced Salt Solution (HBSS) to yield a 2.5 mg/ml solution. The mice were weighed once, immediately prior to the first dose of DSG. Six mice received 1 x 108 pfu of AvllacZ4 via tail vein injection on the second day of the immunosuppression regimen and 1 x 108 pfu of AvlH9F2 five weeks later. Another six mice received only AvlH9F2, five weeks after immunosuppression. Three mice were immunosuppressed, but received no adenoviral vector.
Six mice were immunosuppressed with a low dose (100 mg/kg) cyclophosphamide (Sigma) and fifteen mice were treated with a high dose (300 mg/kg) . The animals received a single ip injection of cyclophosphamide one day before administration of adenoviral vector. All six mice which were treated with a low dose of cyclophosphamide also received 1 x 108 pfu of AvllacZ4 the day after cyclophosphamide and 1 x 108 pfu of AvlH9F2 five weeks later. Six of the mice immunosuppressed with a high dose of cyclophosphamide received a 1 x 108 pfu of AvllacZ4 the next day and 1 x 108 pfu of AvlH9F2 five weeks later. Another six did not receive AvllacZ4 but did receive AvlH9F2. Finally, three mice were immunosuppressed, but received no adenoviral vector.
Twelve mice were immunosuppressed with 5 mg/kg dexamethasone (American Reagent Laboratories, Inc., Shirley, New York) , delivered ip, once daily, beginning the day before vector administration and continuing for a total of eight days . Six of these mice receive 1 x 108 pfu of AvllacZ4 on the second day of dexamethasone treatment and 1 x 108 pfu of AvlH9F2 five weeks later. Five immunosuppressed mice received only AvlH9F2 and one mouse received no vector.
Five weeks after administration of AvllacZ4 , but prior to administration of AvlH9F2, plasma was prepared from some mice and analyzed for antiadenovirus neutralizing antibodies.
Neutralizing antibodies were detected in the plasma of mice which receive AvlLacZ4 without immunosuppression, however, mice which received vector and either DSG or high dose cyclophosphamide had no detectable neutralizing antibodies . In contrast, mice immunosuppressed with low dose cyclophosphamide or dexamethasone developed neutralizing antibodies .
Neutralizing antibody titers in 20 mice are given in Table I below. As indicated in Table I, DSG is deoxyspergualin, Cy is cyclophosphamide, and Dex is dexamethasone .
Table I
One week after administration of AvlH9F2 , plasma was prepared and analyzed by ELISA to determine the levels of human Factor IX. The results are shown in Figure 17. Mice which received AvllacZ4 without immunosuppression, then received AvlH9F2 five weeks later, expressed no human Factor IX. Mice which received neither immunosuppression nor AvllacZ4 , but were treated with AvlH9F2 , expressed an average of 9.2 μg/ml . Mice which were immunosuppressed with DSG at the time of AvllacZ4 delivery, and then received AvlH9F2, expressed an average of 6.6 μg/ml . Mice which were immunosuppressed with DSG, but received no AvllacZ4, and then treated with AvlH9F2 , had an average level of 5.2 μg/ml . Finally, mice which were treated with DSG, but received neither vector, expressed no human Factor IX.
Mice immunosuppressed with a low dose of cyclophosphamide at the time of AvllacZ4 administration, did not express human Factor IX after delivery of AvlH9F2. Mice treated with a high dose of cyclophosphamide, but not administered AvllacZ4 , expressed an average of 8 μg/ml one week after delivery of AvlH9F2. Mice immunosuppressed with
a high dose of cyclophosphamide at the time of AvllacZ4 treatment, expressed an average of 15.4 μg/ml of human Factor IX after treatment with AvlH9F2. Mice treated with cyclophosphamide, but not treated with either adenoviral vector, did not express human Factor IX.
Mice immunosuppressed with dexamethasone, but not treated with AvllacZ4 , expressed an average of 5.5 μg/ml of human Factor IX one week after administration of AvlH9F2. However, mice which received AvllacZ4 at the time of immunosuppression did not express Factor IX after delivery of AvlH9F2. Mice which were immunosuppressed, but not treated with either vector, did not express human Factor IX.
Example 4 Suppression of humoral immune response to adenoviral vectors to enable the repeat administrations thereof This example is an elaboration and expansion of the data contained in Example 3. In this example, the following materials and methods were employed.
Adenoviral vectors
AvlLacZ4 and AvlH9F2 were described in Example 3 hereinabove. AvlH9FR was made by cotransfeeting 293 cells with pAvlH9FR (Figure 15) with the large DNA fragment from Clal digested Ad dl327. Recombinant adenoviral vector plaques were picked, expanded, and screened for expression of Factor IX by ELISA. A positive clone was identified and amplified, thus generating the vector AvlH9FR. This vector, like AvlH9F2, contains a centrally truncated first intron and the complete 5 ' and 3 ' untranslated regions from the human Factor IX gene . The centrally truncated first intron and 3 ' untranslated region are essentially the same sequences described by Jallat, et al . , EMBO J . , Vol. 9, pgs. 3295-3301 (1990) .
AvlALAPHβl is an adenoviral vector which coϋUi WS tkiQ &* domain deleted human Factor VIII cDNA expressed from the mouse albumin promoter, and is described in published PCT Application No. W094/29471.
All vector stocks contained less than 1 in 106 wild-type adenovirus, as determined by quantitative PCR analysis of Ela sequences .
Immunosuppressants
Deoxyspergualin (manufactured by Nippon Kayaku Co . , Ltd., Tokyo, Japan) was a gift from Bristol-Myers-Squibb, Princeton, N.J. A 100 mg vial of deoxyspergualin was reconstituted with water to a final concentration of 25 mg/ l, aliquoted, and frozen at -70°C. Frozen stocks were thawed at room temperature and diluted with Hanks Balanced Salt Solution (HBSS) prior to injection.
Deoxyspergualin is an immunosuppressant currently being tested clinically in organ transplantation. It has a potent, long term effect on antigen specific B cells and has been shown to prevent effectively the production of specific antibody when co-administered with protein antigens. (Alegre, et al . , Transplantation, Vol. 57, pgs. 1786-1794 (1994); Tepper, Ann. N. Y. Acad. Sci .. pgs. 123-132 (1993); Tufveson, et al . , Transplant . Proc .. Vol. 26, pgs. 3029-3039 (1994) ) . The mode of action of DSG is not fully understood at the molecular level . The data suggest that it may interfere with differentiation of B and T cells and also with antigen processing. Recent studies showed that DSG inhibited K light chain expression and therefore blocked IgM expression on the surface of pre B cells. (Tepper, 1993) . In addition, the data showed that DSG inhibited nuclear translocation of NFKB (Tepper, 1993) , which could be the mechanism by which DSG inhibits differentiation of B and T cells. Finally, it
has been demonstrated that DSG binds to Hsc70, a heat shock protein. (Tepper, 1993) . Heat shock proteins are involved in protein folding, molecular chaperoning, peptide loading of MHC molecules, and antigen presentation. Therefore, binding of DSG to Hsc70 may explain the effect on antigen presentation .
Cyclophosphamide (Cy) was obtained from Sigma and Dissolved in HBSS. Dexamethasone (Dex) solution from American Regent Laboratories, Inc., Shirley, N. Y. was diluted in HBSS prior to injection. All three immunosuppressants were delivered intraperitoneally, according to the doses indicated hereinbelow.
Animal procedures
C57BL/6 mice were obtained from Harlan Sprague Dawley (Indianapolis, IN) . Adenoviral vectors were administered via tail vein injection after diluting the appropriate amount of vector stock to 0.5 ml with Hanks Balanced Salt Solution (HBSS) . At the time points indicated in the text, blood was obtained from the retroorbital plexus . For preparation of plasma samples, sodium citrate was added immediately to a final concentration of 0.38% (w/v) . To prepare sera samples, the blood was allowed to clot. Samples were centrifuged for 5 min. in an Eppendorf Microfuge after which the plasma or serum was collected, aliquoted, and frozen.
Human Factor VIII ELISA
Plasma levels of human Factor VIII were determined by ELISA, as described in Connelly, et al . , Human Gene Therapy, Vol. 6, pgs. 185-193 (1995). The limit of sensitivity with mouse plasma samples containing B domain deleted human Factor VIII was 3 to 6 ng/ml. Mouse plasma samples were diluted 1:5 prior to the assay, therefore, the actual limit of detection was 15 to 30 ng/ml.
Human Factor IX ELISA
Plasma levels of human Factor IX were determined by ELISA. Asserachrom IX:Ag ELISA kits were purchased from American Bioproducts Company (Parsippany, NJ) and assays were performed according to the manufacturer's instructions. The limit of sensitivity was 1.6 ng/ml.
Anti-adenoviral antibody assay
Mouse plasma or serum samples were heat inactivated at 55 °C for 30 minutes and then diluted in Improved Minimal Essential Medium (Biofluids, Rockville, MD) plus 2% FBS (IMEM/2%FBS) in two-fold steps beginning at 1:2. 55 μl of each sample were mixed with 10 μl of AvllacZ4 (containing 4 x 105 pfu) , incubated for 1 hour at 37 °C and applied to nearly confluent 293 cells in 96 well plates (4 x 104 cells per well) . After 60 minutes in the tissue culture incubator, the virus was aspirated from each well and replaced with 150 μl of IMEM/10%FBS. The following day, cells were fixed and stained for β-galactosidase expression, as described previously. (Smith, et al . , Nature Genetics . Vol. 5, pgs. 397-402 (1993)). In the absence of inactivating antibodies, all of the cells stained blue. The titer of inactivating antibodies for each sample was reported as the reciprocal of the highest dilution with which less than 25% of the cells stained blue .
Results
Dose dependence of the humoral immune response to adenovirus vectors
Previous studies which have examined repeat delivery of adenovirus vectors have employed relatively high doses of vector, which would be expected to maximize the strength of the immune response. (Smith, et al . , 1993; Kozarsky, et al . ,
J. Biol. Chem.. Vol. 269, pgs. 13695-13702 (1994); Kay, et al., Proc. Nat. Acad. Sci .. Vol. 91, pgs. 2353-233357 (1994);
Yei, et al . , Gene Therapy, Vol. 1, pgs. 192-200 (1994); Yang, et al . , J . Virol . , Vol. 69, pgs. 2004-2015 (1995); Dai, et al . , Proc . Nat . Acad . Sci .. Vol. 92, pgs. 1401-1405 (1995); Barr, et al., Gene Therapy. Vol. 2, pgs. 151-155 (1995)).
To determine whether the production of neutralizing antibodies and the block to repeat delivery is dependent on the dose of vector inoculated, various doses of the adenovirus vector AvllacZ4 to C57BL/6 mice via tail vein were administered. The vector inoculum ranged from 1 x 103 pfu to 1 x 108 pfu in single log increments. Thirty-four days after vector delivery, the serum levels of anti-adenovirus neutralizing antibodies were determined (Fig. 18) for mice which received 1x10s pfu or greater of vector. A minus sign indicates that none of the mice in the cohort had detectable antibody. The plus sign corresponding to mice which received lxlO8 pfu of vector indicates that three of the five mice had an antibody titer of 8, while two mice had no detectable antibody. Thus, three of five mice which received 1 x 108 pfu had a level of anti-adenovirus antibody which was sufficient to neutralize 4xl05 pfu of AvlLacZ4, while two mice had undetectable levels. None of the mice which received lower doses of vector had detectable antibodies using this relatively stringent neutralization assay.
Thirty-five days after administration of AvllacZ4 , each mouse received 2 x 10e pfu of AvlH9FR, an adenoviral vector encoding human Factor IX. One week later, the plasma levels of human Factor IX were determined by ELISA (Fig. 18) . An average of approximately 2 μg/ml of Factor IX was detected in mice which received either no AvllacZ4, or up to 1 x 105 pfu of the first vector. Factor IX was also readily detected in the mice which had received a first dose of 1 x 106 and 1 x 107 pfu, although the levels were reduced. Mice which received 1 x 108 pfu of AvllacZ4 yielded little or no human
Factor IX after administration of AvlH9FR. Thus effective gene transfer and expression can be achieved with a second vector administration, provided the initial vector dose is below a certain threshold level . The data indicate that for intravenous delivery in C57BL/6 mice, this value is between 107 and 108 pfu.
Transient immunosuppression increases the efficiency of vector re-administration.
To determine whether transient immunosuppression would allow readministration of an adenoviral vector, C57BL/6 mice were immunosuppressed with either deoxyspergualin (DSG) , cyclophosphamide (Cy) , or dexamethasone (Dex) at the time of administration of 1 x 10β pfu of AvllacZ4. As shown above, this dose completely prevented an effective second delivery. Mice were injected daily with 33 mg/kg of DSG, beginning one day before vector delivery and continuing for seven more days. Dexamethasone was delivered over the same time course, at a dose of 5 mg/kg. Cyclophosphamide was administered once, the day before vector delivery, at a dose of either 100 mg/kg or 300 mg/kg. Control mice received AvllacZ4 without immunosuppression, or were immunosuppressed without initial vector delivery.
Five weeks after vector administration, the plasma levels of anti-adenovirus neutralizing antibodies were determined (Fig. 19) . Mice immunosuppressed with DSG or 300 mg/kg of cyclophosphamide had no detectable neutralizing antibodies, but all other mice which received AvllacZ4 generated neutralizing antibodies. Thirty-five days after the first vector injection, the mice received 1 x 108 pfu of AvlH9F2. One week after AvlH9F2 injection, plasma samples were prepared and the levels of human Factor IX were determined by ELISA (Fig. 17) . Mice immunosuppressed with DSG or 300 mg/kg cyclophosphamide expressed human Factor IX
at levels which were approximately the same as levels in mice which had not been treated with the first vector. No human Factor IX was detected in the plasma of the mice which were not immunosuppressed at the time of AvllacZ4 administration. Under the conditions used in these studies, immunosuppression with dexamethasone or 100 mg/kg cyclophosphamide was not effective in permitting expression of Factor IX on repeat injection. Thus the ability to achieve transgene expression on a second vector injection correlated with the suppression of neutralizing antibodies by DSG or high dose cyclophosphamide treatment .
At the time of administration of the second vector, AvlH9F2 , half of the mice in each cohort which had been immunosuppressed with cyclophosphamide or dexamethasone were immunosuppressed again, using the same regimen as at the first vector delivery. Five weeks later, plasma levels of anti-adenovirus neutralizing antibodies were determined. The mice immunosuppressed with 300 mg/kg cyclophosphamide had no detectable neutralizing antibodies, while mice immunosuppressed with 100 mg/kg cyclophosphamide or 5 mg/kg dexamethasone had a measurable response (data not shown) . Thirty-five days after AvlH9F2 injection, 1 x 109 pfu of a Factor VIII vector, AvlALAPHβl, were administered to the mice which had been immunosuppressed with 300 mg/kg cyclophosphamide. In addition, Factor VIII vector was administered to control mice which had received AvllacZ4 and AvlH9F2 without immunosuppression and also to naive mice. One week later, plasma levels of human Factor VIII were determined by ELISA (Fig. 20) . Control mice which received only the AvlALAPHβl vector, and mice immunosuppressed with 300 mg/kg of cyclophosphamide at the time of the two prior vector injections expressed human Factor VIII. Mice which received AvllacZ4 and AvlH9F2 without imunosuppression, as
well as mice which were immunosuppressed only at the time of AvllacZ4 delivery, did not express human Factor VIII.
DSG permits effective repeat administration at a clinically re1evant dose .
The previous experiment demonstrated that a high dose of either DSG or cyclophosphamide permitted readministration of an adenoviral vector, but that clinically relevant doses of cyclophosphamide and dexamethasone were not effective . The next objective was to determine whether lower doses of DSG would be effective. The dose of DSG used in the initial experiment (33 mg/kg) is near the maximum tolerated dose in mice and is significantly higher than the 5-7 mg/kg dose used in human clinical trials for organ transplantion. (Suzuki, et al . , Ann. N.Y. Acad. Sci .. Vol. 696, pgs. 263-269 (1993); Jindal, et al . , Mt . Sinai J. Med. , Vol. 61, pgs. 51-56 (1994) ) . To determine if lower doses would also be effective in allowing vector readministration, mice were immunosuppressed with 5, 10, 20, and 33 mg/kg of DSG at the time of administration of 1 x 108 pfu of AvllacZ4. Immunosuppression was started the day before vector delivery and continued for a total of 8 days. On the day of vector delivery, DSG was given after injection of the adenovirus since it is most effective when administered after antigen. (Takahara, et al . , Transplantation, Vol. 53, pgs. 914-918 (1992) ) .
Thirty-five days later each mouse received 1 x 108 pfu of AvlH9F2. One week after AvlH9F2 injection, human Factor IX plasma levels were determined by ELISA (Fig. 21) . Control mice, which were not pre-immunized with AvlLacZ4, expressed an average of 9 μg/ml of human Factor IX. Other control mice, which received AvllacZ4 but were not immunosuppressed, expressed no human Factor IX after AvlH9F2 administration. The one mouse which was immunosuppressed with 33 mg/kg DSG
expressed 12 μg/ml of human Factor IX. Five of six mice immunosuppressed with 20 mg/kg DSG expressed an average of 3.0 μg/ml of human Factor IX, and one mouse expressed none. Mice immunosuppressed with a dose of 10 mg/kg expressed a wide range of Factor IX, extending from 30 ng/ml to 8 μg/ml. Three mice immunosuppressed with 5 mg/kg expressed no human Factor IX, while three others expressed levels ranging from 2 to 7 μg/ml . Mice which were not immunosuppressed at the time of AvllacZ4 administration expressed no human Factor IX.
Discussion The data demonstrate that multiple intravenous administrations of adenovirus vectors with resulting transgene expression can be accomplished in immune competent animals treated with a short course of immunosuppression at the time of vector delivery. This observation is significant because several recent studies have demonstrated that a humoral immune response directed against adenoviral vector prevents readministration. (Smith, et al . , 1993; Kay, et al . , 1994; Yei, et al . , 1994; Yang, et a . , 1995.) The inability to readminister vector has presented a major obstacle to the clinical utility of adenoviral vectors, since effective readministration will almost certainly be required for the clinical application of these vectors to gene therapy of chronic diseases .
The failure to obtain expression following repeat dosing in the absence of immunosuppression correlates with anti- adenovirus neutralizing antibodies. Evidence that such antibodies are sufficient to block readministration was provided by Yang et al . , 1995, who showed that passive transfer of serum from a mouse previously treated with.vector into the venous circulation of a naive mouse was able to prevent vector-mediated gene expression in the liver. The role of the immune system in preventing repeat administration
also was demonstrated by the observation that repeat administration of vector was effective in immunodeficient mice. (Yang, et al . , 1995; Dai, et al . , 1995; Barr, et al . , Gene Therapy, Vol. 2, pgs. 151-155 (1995)). This result has been confirmed by demonstrating effective administration of AvlH9F2 to εcid mice 5 weeks after delivery of 5 x 108 pfu of AvlALAPHδl (data not shown) .
Since previous reports describing humoral responses to adenovirus administration used relatively high vector doses, we evaluated the relationship between initial vector dose and the ability to achieve an effective repeat gene transfer. The results indicated that the magnitude of the immune response was dependent on the initial dose of vector and that if the dose is below a threshold level, a second administration is possible. The finding that this level was only one to two orders of magnitude below a clinically relevant vector dose further suggested that immunosuppression at the time of vector delivery would permit readministration. In addition, these results emphasize the need for potent vectors which would be efficacious at low doses .
Applicants have observed that anti-adenovirus neutralizing antibody titers are maintained for at least ten months in mice after a single administration of vector via tail vein. The long-term maintenance of titer may have been due to a low level of ongoing adenoviral backbone gene expression in transduced cells. Vectors designed to reduce or eliminate backbone gene expresεion may elicit a weaker immune reεponεe and therefore may require less immunosuppression for successful readministration.
An important property of DSG is that it does not produce a general suppression of the immune system, but rather results in a selective lack of response to specific antigens
presented at the time of drug treatment . We found that immunosuppression with DSG over a 7 day period following vector delivery efficiently inhibited the humoral response to the vector and permitted an effective second administration. The initial experiment with DSG employed a high dose of 33 mg/kg, which is close to the maximum tolerated dose in mice and several fold higher than the doses used in human trials . When administered over the same 8 day course, including 7 days post vector treatment, lower doses of DSG were also effective in permitting repeat delivery of vector. A greater degree of individual variability in levels of Factor IX expreεεion was seen with reduced doses, although even at the lowest dose tested (5 mg/kg) significant Factor IX expression was obtained in 3 of 6 animals .
Cyclophosphamide, administered at a dose of 300 mg/kg the day before vector injection, was also effective in blocking the humoral response and allowed a completely effective second injection with a Factor IX adenovirus vector. Furthermore, a third injection with a Factor VIII encoding vector was also completely efficacious when the previous two vector administrations were each preceded by a single dose of cyclophosphamide. Cyclophosphamide is used clinically as an anti-cancer agent in the treatment of Hodgkins disease and other leukemias . It is also employed as an immunosuppresεive agent in the treatment of hemophilia patients who develop inhibitors to Factor VIII protein replacement therapy. (Aledort, Am. J . Hemat .. Vol. 47, pgs. 208-217 (1994); Nilsson, et a . , N. Engl . J. Med.. Vol. 318, pgs. 947-950 (1988)). While the dose used to successfully obtain readministration in mice is substantially higher than is generally used in humans, it remains to be established whether lower, clinically acceptable doses, might be effective in humans. One possibility suggested by experience in the organ transplantation setting is that combinations of
immunosuppressants would yield more potent suppression of the immune system with less toxicity. For example, cyclophosphamide may be effective at lower doseε when uεed in combination with dexamethasone. It is also posεible that the degree of immunosuppression required will depend on the dose of vector which is needed to effect therapy. The dose of AvlH9F2 used in this study, 1 x 108 pfu, yielded plasma levels of human Factor IX of 5-10 μg/ml, which is 20 to 50 times above a level that would be therapeutic in a hemophiliac. Vectors, such as AvlH9F2, which express high levels of transgene product and which can be administered at relatively low doses, should reduce the extent of immune stimulation and the degree of immunosuppression required.
In summary, Applicants have shown that effective repetitive delivery of systemically administered adenovirus vectors can be achieved with short term immunosuppression. Importantly, this can be accomplished using pharmacologic agents which are either approved for use in humans, or are in clinical testing.
Example 5
Transient immunosuppression with a combination of
DSG and cyclophosphamide permits effective second and third administrations of an adenoviral vector.
This example describes transient suppression of the humoral immune system with a combination of DSG and cyclophosphamide. The data presented in this example demonstrate that these two drugs can be used in combination at clinically relevant doses to inhibit a humoral response directed against an adenoviral vector and thus allow multiple repeat administrations of vector.
The adenoviral vector employed in this example was Av3nLacZ. Av3nLacZ is described in Example 2 of U.S. Patent Application Serial No. 08/458,403, entitled "Improved Adenoviral Vectors and Producer Cells," filed June 2, 1995, the contents of which are incorporated herein by reference.
The cohorts of mice treated in this example and a general outline of the treatment regimen for each cohort are given in Table II below.
Table II
H c r" rπ
8
Twenty-four C57BL/6 male mice, six weeks of age, each received, via the tail vein, 1 x 108 pfu of the adenoviral vector Av3nLacZ (Table II, cohorts 1-4) . Twelve of these mice (cohorts 1 and 2) were immunosuppressed at the time of vector delivery using DSG and cyclophosphamide, as follows. The day before vector administration, the mice received a single ip injection of cyclophosphamide at a dose of 100 mg/kg. In addition, each of the twelve mice received a daily ip injection of DSG at a dose of 10 mg/kg, beginning the day before vector delivery and continuing for a total of eight days .
Five weeks after administration of Av3nLacZ, but prior to the second vector delivery, plasma was prepared from some mice and analyzed for anti-adenovirus neutralizing antibodies. Neutralizing antibodies were detected in the plasma of mice which received Av3nLacZ without immunosuppression, however, mice which received vector and a combination of DSG and cyclophosphamide had no detectable neutralizing antibodies .
Thirty-five days after administration of Av3nLacZ, six of the mice which previously received vector without immunosuppression (cohort 3) plus six of the twelve mice which were immunosuppressed (cohort 2) , plus five naive mice (cohort 5) , each received, via the tail vein, 1 x 108 pfu of AvlH9F2. The other six mice which were immunosuppressed at the time of Av3nLacZ delivery received a second injection of Av3nLacZ at a dose of 1 x 108 pfu per mouse (cohort 1) . These six mice were immunosuppressed at the time of the second vector administration in a manner identical to that used at the first vector administration. In addition, six mice which received a first injection of Av3nLacZ without immunosuppression, received a second injection of 1 x 108 pfu of Av3nLacZ without immunosuppression (cohort 4) .
All of the mice which received AvlH9F2 were bled one week after vector adminiεtration and the plasma levels of human Factor IX were determined by ELISA. The results are shown in Figure 22. The five mice which received only AvlH9F2 (No 1st vector) had human Factor IX levels of 15.7, 15.4, 14.5, 8.9, and 4.4 μg/ml (average = 11.8 μg/ml) . The mice which received Av3nLacZ without immunosuppression all had undetectable levels of human Factor IX one week after AvlH9F2 delivery. The mice which were immunosuppressed at the time of Av3nLacZ delivery had levels of 18.9, 18.0, 14.5, 7.7, 6.2, and 6.2 μg/ml (average = 11.9 μg/ml). The results corresponding to these mice are designated as "2nd administration" in Figure 22. Thus, immunosuppreεsion with a combination of DSG and cyclophosphamide enabled a completely efficacious second administration of vector.
As shown in Example 3, mice immunosuppressed with only 100 mg/kg cyclophosphamide at the time of the first vector delivery, expressed no transgene product following a second vector administration. It also was observed that immunosuppression with only 10 mg/kg DSG yielded variation in the efficacy of a second vector administration, as described in Smith et al . , Gene Therapy, in press . The observation that DSG and cyclophosphamide are completely effective when used in combination at doses which are less effective when the drugs are used separately is very significant.
The twelve mice which received two injections of Av3nLacZ (six with immunosuppression and six without) were bled five weeks after the second vector administration and plasma levels of anti-adenovirus neutralizing antibodies for three mice from each cohort were determined. The mice which received two injections of Av3nLacZ without immunosuppression had relatively high antibody titers, while mice immunosuppressed
with DSG and cyclophosphamide at the time of both"adenovira vector administrations had undetectable levels .
The twelve mice which received two injections of Av3nLacZ (cohorts 1 and 4) , plus six naive mice (cohort 6) , each received 1 x 108 pfu of AvlH9F2 thirty-five days after the second Av3nLacZ administration. One week later, the mice were bled and plasma levels of human Factor IX were determined by ELISA. Figure 22 is a graph of the results. The six mice which received only AvlH9F2 (No 1st or 2nd vector) had human Factor IX levels of 18.9, 11.8, 9.4, 8.0, 4.9, and 2.6 μg/ml (average = 9.3 μg/ml). The six mice which received two injections of Av3nLacZ without immunosuppression expressed undetectable levels of human Factor IX one week after AvlH9F2 delivery. The six mice which were immunosuppressed with DSG and cyclophosphamide at the time of both Av3nLacZ injections expressed levels of human Factor IX of 18.1, 15.4, 8.6, 1.3, 0.2, and 0 μg/ml (average - 7.3 μg/ml). The results corresponding to these mice are designated "3rd administration" in Figure 22. Thus, a third vector administration yielded transgene expression in five of six mice, provided the first two vector administrations were accompanied by transient immunosuppression with a combination of DSG and cyclophsophamide .
The disclosure of all patents, publications, (including published patent applications) , and database accession numbers, and depository accession numbers referenced in this specification are specifically incorporated herein by reference in their entirety to the same extent as if each such individual patent, publication, and database accession number, and depository accession number were specifically and individually indicated to be incorporated by reference .
It is to be understood, however, that the scope of the present invention is not to be limited to the specific embodiments described above. The invention may be practiced other than as particularly described and still be within the scope of the accompanying claims .
Claims
1. A method of effecting a gene therapy treatment in a host, comprising the steps of:
(a) administering to a host (i) an adenoviral vector including at least one DNA sequence encoding a therapeutic agent; (ii) deoxyspergualin; and (iii) cyclophosphamide ;
(b) discontinuing said administration of said adenoviral vector, said deoxyspergualin, and said cyclophosphamide; and
(c) repeating the course of administration of said adenoviral vector including at least one DNA sequence encoding a therapeutic agent, said deoxyspergualin, and said cyclophosphamide of step (a) at least once, said adenoviral vector being administered in an amount effective to produce a therapeutic effect in said host, and said deoxyspergualin and said cyclophosphamide being administered in amounts effective to prevent or suppress an immune response against said adenoviral vector in said host .
Priority Applications (1)
| Application Number | Priority Date | Filing Date | Title |
|---|---|---|---|
| AU26688/97A AU2668897A (en) | 1996-04-19 | 1997-04-15 | Gene therapy involving concurrent and repeated administration of adenoviruses and immunosuppressive agents |
Applications Claiming Priority (2)
| Application Number | Priority Date | Filing Date | Title |
|---|---|---|---|
| US63477496A | 1996-04-19 | 1996-04-19 | |
| US08/634,774 | 1996-04-19 |
Publications (2)
| Publication Number | Publication Date |
|---|---|
| WO1997039776A1 WO1997039776A1 (en) | 1997-10-30 |
| WO1997039776A9 true WO1997039776A9 (en) | 1998-01-29 |
Family
ID=24545153
Family Applications (1)
| Application Number | Title | Priority Date | Filing Date |
|---|---|---|---|
| PCT/US1997/006199 WO1997039776A1 (en) | 1996-04-19 | 1997-04-15 | Gene therapy involving concurrent and repeated administration of adenoviruses and immunosuppressive agents |
Country Status (2)
| Country | Link |
|---|---|
| AU (1) | AU2668897A (en) |
| WO (1) | WO1997039776A1 (en) |
Families Citing this family (8)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| DE19711803A1 (en) * | 1997-03-21 | 1998-09-24 | Hoechst Ag | Extension of expression of transgenic proteins by immunomodulating treatment with 15-deoxyspergualin |
| AU3883299A (en) * | 1998-05-12 | 1999-11-29 | Baylor College Of Medicine | Cancer prevention by selective delivery methods |
| US8063022B1 (en) * | 1999-06-08 | 2011-11-22 | The Children's Hospital Of Philadelphia | Methods for preventing formation of inhibitory antibodies in the setting of gene therapy |
| US20020014242A1 (en) | 2000-07-31 | 2002-02-07 | Abraham Scaria | Use of rapamycin to inhibit immune response and induce tolerance to gene therapy vector and encoded transgene products |
| SG11202012044QA (en) | 2018-06-29 | 2021-01-28 | Wuhan Neurophth Biological Tech Limited Company | Compositions and methods for treating leber's hereditary optic neuropathy |
| JP7403852B2 (en) | 2018-08-20 | 2023-12-25 | ウーハン ニューロフス バイオテクノロジー リミテッド カンパニー | Compositions and methods for treating Leber's hereditary optic neuropathy |
| CN113025633B (en) | 2019-12-09 | 2024-08-27 | 武汉纽福斯生物科技有限公司 | Nucleic acid for encoding human NADH dehydrogenase subunit 1 protein and application thereof |
| WO2024168252A2 (en) * | 2023-02-10 | 2024-08-15 | Yale University | Use of inhibitors of vegf-c signaling for inhibiting immune responses and increasing gene therapy efficacy |
-
1997
- 1997-04-15 AU AU26688/97A patent/AU2668897A/en not_active Abandoned
- 1997-04-15 WO PCT/US1997/006199 patent/WO1997039776A1/en active Application Filing
Similar Documents
| Publication | Publication Date | Title |
|---|---|---|
| EP0804076A1 (en) | Gene therapy involving concurrent and repeated administration of adenoviruses and immunosuppressive agents | |
| WO1996012406A9 (en) | Gene therapy involving concurrent and repeated administration of adenoviruses and immunosuppressive agents | |
| EP0710288B1 (en) | Adenoviral vectors for treatment of hemophilia | |
| Tao et al. | Sequestration of adenoviral vector by Kupffer cells leads to a nonlinear dose response of transduction in liver | |
| Christ et al. | Gene therapy with recombinant adenovirus vectors: evaluation of the host immune response | |
| EP0696206B1 (en) | Adenoviruses having modified fiber proteins | |
| Yang et al. | Transient subversion of CD40 ligand function diminishes immune responses to adenovirus vectors in mouse liver and lung tissues | |
| JP3565859B2 (en) | Improved adenovirus and uses thereof | |
| Brown et al. | Helper-dependent adenoviral vectors mediate therapeutic factor VIII expression for several months with minimal accompanying toxicity in a canine model of severe hemophilia A | |
| AU697022B2 (en) | Methods and compositions for administering gene therapy vectors | |
| EP0811074B1 (en) | Methods and compositions for gene therapy for the treatment of defects in lipoprotein metabolism | |
| Geest et al. | Elimination of innate immune responses and liver inflammation by PEGylation of adenoviral vectors and methylprednisolone | |
| Varnavski et al. | Preexisting immunity to adenovirus in rhesus monkeys fails to prevent vector-induced toxicity | |
| NZ330553A (en) | Recombinase-mediated generation of adenoviral vectors where all adenoviral genes of all regions E1 to E4 and L1 to L5 of the adenoviral genome are eliminated | |
| EP1007641A1 (en) | Adenoviruses having altered hexon proteins | |
| WO1997039776A9 (en) | Gene therapy involving concurrent and repeated administration of adenoviruses and immunosuppressive agents | |
| WO1997039776A1 (en) | Gene therapy involving concurrent and repeated administration of adenoviruses and immunosuppressive agents | |
| US6372208B1 (en) | Method of reducing an immune response to a recombinant virus | |
| US6251957B1 (en) | Method of reducing an immune response to a recombinant virus | |
| WO2005027711A2 (en) | Capsid-modified adenovirus vectors and methods of using the same | |
| EP1183361B1 (en) | Gene therapy of pulmonary disease | |
| US6132989A (en) | Methods and compositions for enhanced stability of non-adenoviral DNA | |
| NZ527645A (en) | Method of enhancing delivery of a therapeutic nucleic acid | |
| AU2002237910A1 (en) | Method of enhancing delivery of a therapeutic nucleic acid | |
| WO2003024384A2 (en) | Improved methods for treatment with viral vectors |