WO2009071679A1 - Novel aav vector and uses thereof - Google Patents
Novel aav vector and uses thereof Download PDFInfo
- Publication number
- WO2009071679A1 WO2009071679A1 PCT/EP2008/066919 EP2008066919W WO2009071679A1 WO 2009071679 A1 WO2009071679 A1 WO 2009071679A1 EP 2008066919 W EP2008066919 W EP 2008066919W WO 2009071679 A1 WO2009071679 A1 WO 2009071679A1
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- aav
- vector
- gene
- gar
- cells
- Prior art date
Links
- 239000013598 vector Substances 0.000 title claims abstract description 53
- 239000013607 AAV vector Substances 0.000 claims abstract description 35
- 108090000565 Capsid Proteins Proteins 0.000 claims abstract description 21
- 102100023321 Ceruloplasmin Human genes 0.000 claims abstract description 21
- 230000014509 gene expression Effects 0.000 claims description 26
- 239000002773 nucleotide Substances 0.000 claims description 10
- 125000003729 nucleotide group Chemical group 0.000 claims description 10
- 108700019146 Transgenes Proteins 0.000 claims description 8
- 125000003275 alpha amino acid group Chemical group 0.000 claims description 6
- FWMNVWWHGCHHJJ-SKKKGAJSSA-N 4-amino-1-[(2r)-6-amino-2-[[(2r)-2-[[(2r)-2-[[(2r)-2-amino-3-phenylpropanoyl]amino]-3-phenylpropanoyl]amino]-4-methylpentanoyl]amino]hexanoyl]piperidine-4-carboxylic acid Chemical compound C([C@H](C(=O)N[C@H](CC(C)C)C(=O)N[C@H](CCCCN)C(=O)N1CCC(N)(CC1)C(O)=O)NC(=O)[C@H](N)CC=1C=CC=CC=1)C1=CC=CC=C1 FWMNVWWHGCHHJJ-SKKKGAJSSA-N 0.000 claims description 5
- 150000001413 amino acids Chemical class 0.000 claims description 5
- 239000000203 mixture Substances 0.000 claims description 5
- 230000001105 regulatory effect Effects 0.000 claims description 5
- 239000003114 blood coagulation factor Substances 0.000 claims description 3
- 101150093578 VP2 gene Proteins 0.000 claims description 2
- 238000001415 gene therapy Methods 0.000 abstract description 7
- VPZXBVLAVMBEQI-VKHMYHEASA-N Glycyl-alanine Chemical group OC(=O)[C@H](C)NC(=O)CN VPZXBVLAVMBEQI-VKHMYHEASA-N 0.000 abstract description 2
- 108700005077 Viral Genes Proteins 0.000 abstract description 2
- 210000004027 cell Anatomy 0.000 description 50
- 108090000623 proteins and genes Proteins 0.000 description 46
- 241000700605 Viruses Species 0.000 description 37
- 208000037265 diseases, disorders, signs and symptoms Diseases 0.000 description 22
- 230000006870 function Effects 0.000 description 22
- 239000005090 green fluorescent protein Substances 0.000 description 18
- 210000000234 capsid Anatomy 0.000 description 17
- 201000010099 disease Diseases 0.000 description 17
- 238000000034 method Methods 0.000 description 15
- 102000004169 proteins and genes Human genes 0.000 description 15
- 230000010076 replication Effects 0.000 description 15
- 210000002845 virion Anatomy 0.000 description 13
- 238000004806 packaging method and process Methods 0.000 description 12
- 239000002245 particle Substances 0.000 description 12
- 239000013612 plasmid Substances 0.000 description 12
- 230000001225 therapeutic effect Effects 0.000 description 12
- 238000011282 treatment Methods 0.000 description 12
- 108010043121 Green Fluorescent Proteins Proteins 0.000 description 11
- 102000004144 Green Fluorescent Proteins Human genes 0.000 description 11
- 208000009292 Hemophilia A Diseases 0.000 description 9
- 210000001151 cytotoxic T lymphocyte Anatomy 0.000 description 9
- 238000004519 manufacturing process Methods 0.000 description 9
- 238000011529 RT qPCR Methods 0.000 description 8
- 230000023555 blood coagulation Effects 0.000 description 8
- 230000004927 fusion Effects 0.000 description 8
- 241000702423 Adeno-associated virus - 2 Species 0.000 description 7
- 208000031220 Hemophilia Diseases 0.000 description 7
- 241000124008 Mammalia Species 0.000 description 7
- 108700026244 Open Reading Frames Proteins 0.000 description 7
- 108020004707 nucleic acids Proteins 0.000 description 7
- 102000039446 nucleic acids Human genes 0.000 description 7
- 150000007523 nucleic acids Chemical class 0.000 description 7
- 108090000765 processed proteins & peptides Proteins 0.000 description 7
- 102000004196 processed proteins & peptides Human genes 0.000 description 7
- 239000000523 sample Substances 0.000 description 7
- 241000701161 unidentified adenovirus Species 0.000 description 7
- 102100022641 Coagulation factor IX Human genes 0.000 description 6
- 108020004414 DNA Proteins 0.000 description 6
- 108010076282 Factor IX Proteins 0.000 description 6
- 241000699670 Mus sp. Species 0.000 description 6
- 229960004222 factor ix Drugs 0.000 description 6
- 230000003612 virological effect Effects 0.000 description 6
- 241000702421 Dependoparvovirus Species 0.000 description 5
- 108010054218 Factor VIII Proteins 0.000 description 5
- 241000699666 Mus <mouse, genus> Species 0.000 description 5
- 208000035475 disorder Diseases 0.000 description 5
- 230000002068 genetic effect Effects 0.000 description 5
- 238000001727 in vivo Methods 0.000 description 5
- 208000024891 symptom Diseases 0.000 description 5
- 238000013518 transcription Methods 0.000 description 5
- 230000035897 transcription Effects 0.000 description 5
- 101000583086 Bunodosoma granuliferum Delta-actitoxin-Bgr2b Proteins 0.000 description 4
- 102000001690 Factor VIII Human genes 0.000 description 4
- 241001465754 Metazoa Species 0.000 description 4
- 108091081024 Start codon Proteins 0.000 description 4
- 210000003169 central nervous system Anatomy 0.000 description 4
- 239000012636 effector Substances 0.000 description 4
- 229960000301 factor viii Drugs 0.000 description 4
- 210000003494 hepatocyte Anatomy 0.000 description 4
- 238000000338 in vitro Methods 0.000 description 4
- 238000002347 injection Methods 0.000 description 4
- 239000007924 injection Substances 0.000 description 4
- 229920001184 polypeptide Polymers 0.000 description 4
- 230000004044 response Effects 0.000 description 4
- 241000894007 species Species 0.000 description 4
- 210000001519 tissue Anatomy 0.000 description 4
- 238000010361 transduction Methods 0.000 description 4
- 230000026683 transduction Effects 0.000 description 4
- 239000013603 viral vector Substances 0.000 description 4
- 108020005544 Antisense RNA Proteins 0.000 description 3
- 108090000994 Catalytic RNA Proteins 0.000 description 3
- 102000053642 Catalytic RNA Human genes 0.000 description 3
- 108091026890 Coding region Proteins 0.000 description 3
- 230000004543 DNA replication Effects 0.000 description 3
- 108091028043 Nucleic acid sequence Proteins 0.000 description 3
- 108700008625 Reporter Genes Proteins 0.000 description 3
- 241000700618 Vaccinia virus Species 0.000 description 3
- 238000012412 chemical coupling Methods 0.000 description 3
- 239000003795 chemical substances by application Substances 0.000 description 3
- 230000000295 complement effect Effects 0.000 description 3
- 239000003184 complementary RNA Substances 0.000 description 3
- 230000008878 coupling Effects 0.000 description 3
- 238000010168 coupling process Methods 0.000 description 3
- 238000005859 coupling reaction Methods 0.000 description 3
- 230000000694 effects Effects 0.000 description 3
- 230000008030 elimination Effects 0.000 description 3
- 238000003379 elimination reaction Methods 0.000 description 3
- 239000003623 enhancer Substances 0.000 description 3
- 238000003780 insertion Methods 0.000 description 3
- 230000037431 insertion Effects 0.000 description 3
- 108091092562 ribozyme Proteins 0.000 description 3
- 238000001890 transfection Methods 0.000 description 3
- 238000013519 translation Methods 0.000 description 3
- 230000014616 translation Effects 0.000 description 3
- 241001529453 unidentified herpesvirus Species 0.000 description 3
- 208000003322 Coinfection Diseases 0.000 description 2
- 238000002965 ELISA Methods 0.000 description 2
- 108091029865 Exogenous DNA Proteins 0.000 description 2
- 108010023321 Factor VII Proteins 0.000 description 2
- 108010014173 Factor X Proteins 0.000 description 2
- 108010071289 Factor XIII Proteins 0.000 description 2
- 206010062016 Immunosuppression Diseases 0.000 description 2
- 102100021244 Integral membrane protein GPR180 Human genes 0.000 description 2
- 102100037850 Interferon gamma Human genes 0.000 description 2
- 108010074328 Interferon-gamma Proteins 0.000 description 2
- 102000043129 MHC class I family Human genes 0.000 description 2
- 108091054437 MHC class I family Proteins 0.000 description 2
- 101800004937 Protein C Proteins 0.000 description 2
- 241000125945 Protoparvovirus Species 0.000 description 2
- FAPWRFPIFSIZLT-UHFFFAOYSA-M Sodium chloride Chemical compound [Na+].[Cl-] FAPWRFPIFSIZLT-UHFFFAOYSA-M 0.000 description 2
- 108010067390 Viral Proteins Proteins 0.000 description 2
- 230000004913 activation Effects 0.000 description 2
- 239000012491 analyte 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
- 210000004556 brain Anatomy 0.000 description 2
- 230000002950 deficient Effects 0.000 description 2
- 210000004443 dendritic cell Anatomy 0.000 description 2
- 230000001419 dependent effect Effects 0.000 description 2
- 238000011161 development Methods 0.000 description 2
- 239000003814 drug Substances 0.000 description 2
- 238000000799 fluorescence microscopy Methods 0.000 description 2
- 238000001476 gene delivery Methods 0.000 description 2
- 102000034356 gene-regulatory proteins Human genes 0.000 description 2
- 108091006104 gene-regulatory proteins Proteins 0.000 description 2
- 230000005745 host immune response Effects 0.000 description 2
- 230000001506 immunosuppresive effect Effects 0.000 description 2
- 230000001965 increasing effect Effects 0.000 description 2
- 230000001939 inductive effect Effects 0.000 description 2
- 239000000463 material Substances 0.000 description 2
- 230000001404 mediated effect Effects 0.000 description 2
- 238000012986 modification Methods 0.000 description 2
- 210000000663 muscle cell Anatomy 0.000 description 2
- 239000013642 negative control Substances 0.000 description 2
- 210000001577 neostriatum Anatomy 0.000 description 2
- 230000010412 perfusion Effects 0.000 description 2
- 239000008363 phosphate buffer Substances 0.000 description 2
- 108091033319 polynucleotide Proteins 0.000 description 2
- 102000040430 polynucleotide Human genes 0.000 description 2
- 239000002157 polynucleotide Substances 0.000 description 2
- 239000013608 rAAV vector Substances 0.000 description 2
- 230000009467 reduction Effects 0.000 description 2
- 230000002441 reversible effect Effects 0.000 description 2
- 239000011780 sodium chloride Substances 0.000 description 2
- 210000000278 spinal cord Anatomy 0.000 description 2
- 238000006467 substitution reaction Methods 0.000 description 2
- 230000002459 sustained effect Effects 0.000 description 2
- 238000003786 synthesis reaction Methods 0.000 description 2
- 230000001052 transient effect Effects 0.000 description 2
- 210000003462 vein Anatomy 0.000 description 2
- 108091032973 (ribonucleotides)n+m Proteins 0.000 description 1
- 102000040650 (ribonucleotides)n+m Human genes 0.000 description 1
- 208000031295 Animal disease Diseases 0.000 description 1
- 108010039209 Blood Coagulation Factors Proteins 0.000 description 1
- 102000015081 Blood Coagulation Factors Human genes 0.000 description 1
- 108091003079 Bovine Serum Albumin Proteins 0.000 description 1
- 101000583080 Bunodosoma granuliferum Delta-actitoxin-Bgr2a Proteins 0.000 description 1
- 102100023804 Coagulation factor VII Human genes 0.000 description 1
- 102100026735 Coagulation factor VIII Human genes 0.000 description 1
- 206010053567 Coagulopathies Diseases 0.000 description 1
- 108020004705 Codon Proteins 0.000 description 1
- 108091028732 Concatemer Proteins 0.000 description 1
- 102000053602 DNA Human genes 0.000 description 1
- 102000003844 DNA helicases Human genes 0.000 description 1
- 108090000133 DNA helicases Proteins 0.000 description 1
- 108090000626 DNA-directed RNA polymerases Proteins 0.000 description 1
- 102000004163 DNA-directed RNA polymerases Human genes 0.000 description 1
- 102000016911 Deoxyribonucleases Human genes 0.000 description 1
- 108010053770 Deoxyribonucleases Proteins 0.000 description 1
- 239000006144 Dulbecco’s modified Eagle's medium Substances 0.000 description 1
- 108010031111 EBV-encoded nuclear antigen 1 Proteins 0.000 description 1
- 201000003542 Factor VIII deficiency Diseases 0.000 description 1
- 201000007371 Factor XIII Deficiency Diseases 0.000 description 1
- -1 Factor Xl Proteins 0.000 description 1
- 108050002220 Green fluorescent protein, GFP Proteins 0.000 description 1
- 241000282412 Homo Species 0.000 description 1
- 101000911390 Homo sapiens Coagulation factor VIII Proteins 0.000 description 1
- 241000700588 Human alphaherpesvirus 1 Species 0.000 description 1
- 108020005350 Initiator Codon Proteins 0.000 description 1
- 108020004684 Internal Ribosome Entry Sites Proteins 0.000 description 1
- 108010079364 N-glycylalanine Proteins 0.000 description 1
- 108010071690 Prealbumin Proteins 0.000 description 1
- 201000005660 Protein C Deficiency Diseases 0.000 description 1
- 238000001190 Q-PCR Methods 0.000 description 1
- 238000010240 RT-PCR analysis Methods 0.000 description 1
- 108020004511 Recombinant DNA Proteins 0.000 description 1
- 241001068295 Replication defective viruses Species 0.000 description 1
- 101800001700 Saposin-D Proteins 0.000 description 1
- 102400000827 Saposin-D Human genes 0.000 description 1
- 108050000761 Serpin Proteins 0.000 description 1
- 102000008847 Serpin Human genes 0.000 description 1
- 108020004682 Single-Stranded DNA Proteins 0.000 description 1
- 108091027967 Small hairpin RNA Proteins 0.000 description 1
- 230000005867 T cell response Effects 0.000 description 1
- 210000001744 T-lymphocyte Anatomy 0.000 description 1
- 102000009190 Transthyretin Human genes 0.000 description 1
- 241000251539 Vertebrata <Metazoa> Species 0.000 description 1
- JLCPHMBAVCMARE-UHFFFAOYSA-N [3-[[3-[[3-[[3-[[3-[[3-[[3-[[3-[[3-[[3-[[3-[[5-(2-amino-6-oxo-1H-purin-9-yl)-3-[[3-[[3-[[3-[[3-[[3-[[5-(2-amino-6-oxo-1H-purin-9-yl)-3-[[5-(2-amino-6-oxo-1H-purin-9-yl)-3-hydroxyoxolan-2-yl]methoxy-hydroxyphosphoryl]oxyoxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(5-methyl-2,4-dioxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(6-aminopurin-9-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(6-aminopurin-9-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(6-aminopurin-9-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(6-aminopurin-9-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxyoxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(5-methyl-2,4-dioxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(4-amino-2-oxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(5-methyl-2,4-dioxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(5-methyl-2,4-dioxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(6-aminopurin-9-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(6-aminopurin-9-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(4-amino-2-oxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(4-amino-2-oxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(4-amino-2-oxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(6-aminopurin-9-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(4-amino-2-oxopyrimidin-1-yl)oxolan-2-yl]methyl [5-(6-aminopurin-9-yl)-2-(hydroxymethyl)oxolan-3-yl] hydrogen phosphate Chemical class Cc1cn(C2CC(OP(O)(=O)OCC3OC(CC3OP(O)(=O)OCC3OC(CC3O)n3cnc4c3nc(N)[nH]c4=O)n3cnc4c3nc(N)[nH]c4=O)C(COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3CO)n3cnc4c(N)ncnc34)n3ccc(N)nc3=O)n3cnc4c(N)ncnc34)n3ccc(N)nc3=O)n3ccc(N)nc3=O)n3ccc(N)nc3=O)n3cnc4c(N)ncnc34)n3cnc4c(N)ncnc34)n3cc(C)c(=O)[nH]c3=O)n3cc(C)c(=O)[nH]c3=O)n3ccc(N)nc3=O)n3cc(C)c(=O)[nH]c3=O)n3cnc4c3nc(N)[nH]c4=O)n3cnc4c(N)ncnc34)n3cnc4c(N)ncnc34)n3cnc4c(N)ncnc34)n3cnc4c(N)ncnc34)O2)c(=O)[nH]c1=O JLCPHMBAVCMARE-UHFFFAOYSA-N 0.000 description 1
- 238000010171 animal model Methods 0.000 description 1
- 238000000137 annealing Methods 0.000 description 1
- 230000000692 anti-sense effect Effects 0.000 description 1
- 239000000427 antigen Substances 0.000 description 1
- 230000000890 antigenic effect Effects 0.000 description 1
- 108091007433 antigens Proteins 0.000 description 1
- 102000036639 antigens Human genes 0.000 description 1
- 210000001367 artery Anatomy 0.000 description 1
- 238000003556 assay Methods 0.000 description 1
- 210000001130 astrocyte Anatomy 0.000 description 1
- 230000037396 body weight Effects 0.000 description 1
- 210000000988 bone and bone Anatomy 0.000 description 1
- 239000000872 buffer Substances 0.000 description 1
- AIYUHDOJVYHVIT-UHFFFAOYSA-M caesium chloride Chemical compound [Cl-].[Cs+] AIYUHDOJVYHVIT-UHFFFAOYSA-M 0.000 description 1
- 210000000845 cartilage Anatomy 0.000 description 1
- 210000001159 caudate nucleus Anatomy 0.000 description 1
- 230000003915 cell function Effects 0.000 description 1
- 210000000170 cell membrane Anatomy 0.000 description 1
- 230000002490 cerebral effect Effects 0.000 description 1
- 210000000349 chromosome Anatomy 0.000 description 1
- 238000010367 cloning Methods 0.000 description 1
- 230000035602 clotting Effects 0.000 description 1
- 239000002299 complementary DNA Substances 0.000 description 1
- 238000004624 confocal microscopy Methods 0.000 description 1
- 238000007796 conventional method Methods 0.000 description 1
- 238000004132 cross linking Methods 0.000 description 1
- 230000003247 decreasing effect Effects 0.000 description 1
- 230000007812 deficiency Effects 0.000 description 1
- 238000012217 deletion Methods 0.000 description 1
- 230000037430 deletion Effects 0.000 description 1
- 239000010432 diamond Substances 0.000 description 1
- 239000012470 diluted sample Substances 0.000 description 1
- 238000010790 dilution Methods 0.000 description 1
- 239000012895 dilution Substances 0.000 description 1
- 230000006806 disease prevention Effects 0.000 description 1
- 231100000673 dose–response relationship Toxicity 0.000 description 1
- 229940079593 drug Drugs 0.000 description 1
- 238000005516 engineering process Methods 0.000 description 1
- 230000008029 eradication Effects 0.000 description 1
- 201000007386 factor VII deficiency Diseases 0.000 description 1
- 208000005376 factor X deficiency Diseases 0.000 description 1
- 229940012413 factor vii Drugs 0.000 description 1
- 229940012426 factor x Drugs 0.000 description 1
- 229940012444 factor xiii Drugs 0.000 description 1
- 239000012091 fetal bovine serum Substances 0.000 description 1
- 239000012530 fluid Substances 0.000 description 1
- 108020001507 fusion proteins Proteins 0.000 description 1
- 102000037865 fusion proteins Human genes 0.000 description 1
- 238000012246 gene addition Methods 0.000 description 1
- 238000002695 general anesthesia Methods 0.000 description 1
- VPZXBVLAVMBEQI-UHFFFAOYSA-N glycyl-DL-alpha-alanine Natural products OC(=O)C(C)NC(=O)CN VPZXBVLAVMBEQI-UHFFFAOYSA-N 0.000 description 1
- 239000001963 growth medium Substances 0.000 description 1
- 208000009429 hemophilia B Diseases 0.000 description 1
- 231100000304 hepatotoxicity Toxicity 0.000 description 1
- 208000013746 hereditary thrombophilia due to congenital protein C deficiency Diseases 0.000 description 1
- 230000028996 humoral immune response Effects 0.000 description 1
- 230000004727 humoral immunity Effects 0.000 description 1
- 230000028993 immune response Effects 0.000 description 1
- 230000003053 immunization Effects 0.000 description 1
- 230000002163 immunogen Effects 0.000 description 1
- 230000006872 improvement Effects 0.000 description 1
- 238000000099 in vitro assay Methods 0.000 description 1
- 238000005462 in vivo assay Methods 0.000 description 1
- 238000011534 incubation Methods 0.000 description 1
- 230000006698 induction Effects 0.000 description 1
- 208000015181 infectious disease Diseases 0.000 description 1
- 230000002401 inhibitory effect Effects 0.000 description 1
- 238000011221 initial treatment Methods 0.000 description 1
- 230000000977 initiatory effect Effects 0.000 description 1
- 230000010354 integration Effects 0.000 description 1
- 238000010255 intramuscular injection Methods 0.000 description 1
- 239000007927 intramuscular injection Substances 0.000 description 1
- 229940065638 intron a Drugs 0.000 description 1
- 210000005229 liver cell Anatomy 0.000 description 1
- 230000007056 liver toxicity Effects 0.000 description 1
- 230000007774 longterm Effects 0.000 description 1
- 230000002101 lytic effect Effects 0.000 description 1
- 229920002521 macromolecule Polymers 0.000 description 1
- 230000007246 mechanism Effects 0.000 description 1
- 108020004999 messenger RNA Proteins 0.000 description 1
- 238000010369 molecular cloning Methods 0.000 description 1
- 239000000178 monomer Substances 0.000 description 1
- 210000000822 natural killer cell Anatomy 0.000 description 1
- 210000004498 neuroglial cell Anatomy 0.000 description 1
- 210000000715 neuromuscular junction Anatomy 0.000 description 1
- 210000002569 neuron Anatomy 0.000 description 1
- 230000003472 neutralizing effect Effects 0.000 description 1
- 231100000252 nontoxic Toxicity 0.000 description 1
- 230000003000 nontoxic effect Effects 0.000 description 1
- 210000000056 organ Anatomy 0.000 description 1
- 238000012858 packaging process Methods 0.000 description 1
- 230000001717 pathogenic effect Effects 0.000 description 1
- 230000002085 persistent effect Effects 0.000 description 1
- 239000008194 pharmaceutical composition Substances 0.000 description 1
- 230000001766 physiological effect Effects 0.000 description 1
- 230000036470 plasma concentration Effects 0.000 description 1
- 230000008488 polyadenylation Effects 0.000 description 1
- 230000008569 process Effects 0.000 description 1
- 230000002035 prolonged effect Effects 0.000 description 1
- 229960000856 protein c Drugs 0.000 description 1
- 210000004777 protein coat Anatomy 0.000 description 1
- 238000000746 purification Methods 0.000 description 1
- 210000002637 putamen Anatomy 0.000 description 1
- 239000013646 rAAV2 vector Substances 0.000 description 1
- 238000005215 recombination Methods 0.000 description 1
- 230000006798 recombination Effects 0.000 description 1
- 230000002829 reductive effect Effects 0.000 description 1
- 238000010242 retro-orbital bleeding Methods 0.000 description 1
- 238000003757 reverse transcription PCR Methods 0.000 description 1
- 239000003001 serine protease inhibitor Substances 0.000 description 1
- 239000004055 small Interfering RNA Substances 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
- 125000006850 spacer group Chemical group 0.000 description 1
- 210000004988 splenocyte Anatomy 0.000 description 1
- 230000010473 stable expression Effects 0.000 description 1
- 230000000638 stimulation Effects 0.000 description 1
- 238000003860 storage Methods 0.000 description 1
- 239000000126 substance Substances 0.000 description 1
- 239000000758 substrate Substances 0.000 description 1
- 230000001839 systemic circulation Effects 0.000 description 1
- 238000012360 testing method Methods 0.000 description 1
- 238000002560 therapeutic procedure Methods 0.000 description 1
- 230000005030 transcription termination Effects 0.000 description 1
- 230000002463 transducing effect Effects 0.000 description 1
- 238000012546 transfer Methods 0.000 description 1
- 230000010474 transient expression Effects 0.000 description 1
- 230000032258 transport Effects 0.000 description 1
- 230000010415 tropism Effects 0.000 description 1
- 238000011144 upstream manufacturing Methods 0.000 description 1
- 230000002792 vascular Effects 0.000 description 1
- 210000005166 vasculature Anatomy 0.000 description 1
- 230000002861 ventricular Effects 0.000 description 1
Classifications
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N15/00—Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
- C12N15/09—Recombinant DNA-technology
- C12N15/63—Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
- C12N15/79—Vectors or expression systems specially adapted for eukaryotic hosts
- C12N15/85—Vectors or expression systems specially adapted for eukaryotic hosts for animal cells
- C12N15/86—Viral vectors
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K38/00—Medicinal preparations containing peptides
- A61K38/16—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
- A61K38/17—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
- A61K38/36—Blood coagulation or fibrinolysis factors
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K38/00—Medicinal preparations containing peptides
- A61K38/16—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
- A61K38/17—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
- A61K38/36—Blood coagulation or fibrinolysis factors
- A61K38/37—Factors VIII
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K38/00—Medicinal preparations containing peptides
- A61K38/16—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
- A61K38/43—Enzymes; Proenzymes; Derivatives thereof
- A61K38/46—Hydrolases (3)
- A61K38/48—Hydrolases (3) acting on peptide bonds (3.4)
- A61K38/482—Serine endopeptidases (3.4.21)
- A61K38/4846—Factor VII (3.4.21.21); Factor IX (3.4.21.22); Factor Xa (3.4.21.6); Factor XI (3.4.21.27); Factor XII (3.4.21.38)
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K38/00—Medicinal preparations containing peptides
- A61K38/16—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
- A61K38/43—Enzymes; Proenzymes; Derivatives thereof
- A61K38/46—Hydrolases (3)
- A61K38/48—Hydrolases (3) acting on peptide bonds (3.4)
- A61K38/482—Serine endopeptidases (3.4.21)
- A61K38/4866—Protein C (3.4.21.69)
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K14/00—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
- C07K14/005—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from viruses
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N9/00—Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
- C12N9/14—Hydrolases (3)
- C12N9/48—Hydrolases (3) acting on peptide bonds (3.4)
- C12N9/50—Proteinases, e.g. Endopeptidases (3.4.21-3.4.25)
- C12N9/64—Proteinases, e.g. Endopeptidases (3.4.21-3.4.25) derived from animal tissue
- C12N9/6421—Proteinases, e.g. Endopeptidases (3.4.21-3.4.25) derived from animal tissue from mammals
- C12N9/6424—Serine endopeptidases (3.4.21)
- C12N9/644—Coagulation factor IXa (3.4.21.22)
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N9/00—Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
- C12N9/14—Hydrolases (3)
- C12N9/48—Hydrolases (3) acting on peptide bonds (3.4)
- C12N9/50—Proteinases, e.g. Endopeptidases (3.4.21-3.4.25)
- C12N9/64—Proteinases, e.g. Endopeptidases (3.4.21-3.4.25) derived from animal tissue
- C12N9/6421—Proteinases, e.g. Endopeptidases (3.4.21-3.4.25) derived from animal tissue from mammals
- C12N9/6424—Serine endopeptidases (3.4.21)
- C12N9/647—Blood coagulation factors not provided for in a preceding group or according to more than one of the proceeding groups
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12Y—ENZYMES
- C12Y304/00—Hydrolases acting on peptide bonds, i.e. peptidases (3.4)
- C12Y304/21—Serine endopeptidases (3.4.21)
- C12Y304/21022—Coagulation factor IXa (3.4.21.22)
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2750/00—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssDNA viruses
- C12N2750/00011—Details
- C12N2750/14011—Parvoviridae
- C12N2750/14111—Dependovirus, e.g. adenoassociated viruses
- C12N2750/14122—New viral proteins or individual genes, new structural or functional aspects of known viral proteins or genes
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2750/00—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssDNA viruses
- C12N2750/00011—Details
- C12N2750/14011—Parvoviridae
- C12N2750/14111—Dependovirus, e.g. adenoassociated viruses
- C12N2750/14141—Use of virus, viral particle or viral elements as a vector
- C12N2750/14143—Use of virus, viral particle or viral elements as a vector viral genome or elements thereof as genetic vector
Definitions
- the present invention relates to the field of gene therapy more specifically to the field of adeno-associated viral gene vectors.
- the present invention provides a novel recombinant AAV vector comprising a chimeric capsid protein fused to a GIy-AIa repeat domain.
- Adeno-associated virus is a parvovirus belonging to the genus Dependovirus.
- the virus is presently being evaluated as an attractive gene therapy vector because it possesses several promising features not found in other viruses. Indeed, AAV can infect a wide range of host cells, including non-dividing cells and it infects also cells from different species. More importantly, AAV has not been associated with any human or animal disease and does not appear to alter the physiological properties of the host cell upon integration. Furthermore, AAV is stable at a wide range of physical and chemical conditions, which lends itself to production, storage, and transportation requirements.
- AAV Being a single-stranded DNA virus, AAV includes two large open reading frames (ORFs), known as the AAV replication (rep) and capsid (cap) regions. These ORFs encode replication and capsid gene products, respectively: replication and capsid gene products allow for the replication, assembly, and packaging of a complete AAV virion. More specifically, a family of at least four viral proteins are expressed from the AAV rep region: Rep 78, Rep 68, Rep 52, and Rep 40, all of which are named for their apparent molecular weights. The AAV cap region encodes at least three proteins: VP1 , VP2, and VP3. Said rep and capsid genes are flanked by 165-bp inverted terminal repeats (ITRs).
- ITRs inverted terminal repeats
- recombinant AAV creates a genetic cassette that can be packaged into intact virions and delivered preferentially to specific cell types. Following virus entry, nuclear trafficking, and uncoating, the released genome becomes double stranded and via intra- or intermolecular recombination of the ITRs persists as circular monomers or concatemers.
- AAV is a helper virus-dependent virus, i.e., it requires co-infection with a helper virus (e.g., adenovirus, herpesvirus, or vaccinia virus) in order to form functionally complete AAV virions.
- a helper virus e.g., adenovirus, herpesvirus, or vaccinia virus
- AAV establishes a latent state in which the viral genome inserts into a host cell chromosome or exists in an episomal form, but functional virions are not produced.
- Subsequent infection by a helper virus "rescues" the integrated genome, allowing it to be replicated and packaged into viral capsids, thereby reconstituting the functional virion.
- AAV can infect cells from different species, the helper virus must be of the same species as the host cell.
- AAV functional recombinant AAV
- a suitable host cell line is transfected with an AAV vector containing a nucleic acid.
- AAV helper functions and accessory functions are then expressed in the host cell.
- gene therapeutic uses because the patient's cells lack the rep and cap genes, as well as the adenovirus accessory function genes - the rAAV are replication defective; i.e. they cannot further replicate and package their genomes.
- AAV serotypes described which are able to infect humans.
- AAV-2 is the best characterized and has been used to successfully deliver transgenes to several cell lines, tissue types, and organs in a variety of in vitro and in vivo assays. Notwithstanding the promises of recombinant AAV vectors as a gene therapy vector there exists a severe limitation because of the immune response directed against vector and/or transgene product which significantly impedes the persistent expression of the transgenes encoding neoantigens. Long-term humoral immunity against several viral-vector systems prevails in a large part of the population, or may be induced upon the first vector administration. This may frustrate re-administration of the vector and lead to elimination of the transduced cells.
- GAR-domain a novel recombinant AAV vector which comprises a capsid protein that is coupled to a GIy-AIa repeat domain
- Said GAR-domain has been used to inhibit the T-cell mediated immune rejection of heterologous fusion proteins produced by adenoviral-transduced cells (Ossevoort M et al (2003) Gene Therapy 10, 2020).
- Figure 1 shows a real-time qPCR on the AAV-EBNA vector particles.
- DNA was extracted from the particles and subjected to a real-time qPCR (TaqMan) with vector specific primers/probe (see Example 1 ).
- AAV particles that encapsidate the corresponding vector genomes should yield a signal, as shown here for the GFP (Fig. 1A) and FIX (Fig. 1 B) vectors.
- Figure 2 represents a microscopic fluorescent image of different cell types transduced with the (stealth) AAV vector. It clearly shows that the EBNA-modified AAV vector can transduce different cell types in a dose-dependent manner consistent with increasing percentage of GFP- positive cells and increased GFP expression level/cell. This indicates that the EBNA-modified AAV vector is functional and capable of delivering genes (in casu GFP reporter gene) to different cell types.
- Figure 3 shows a microscopic fluorescent image of hepatocytes.
- Left panel negative control animals injected with PBS (phosphate buffer saline), right panel: hepatocytes transduced with the Gar-modified vector.
- PBS phosphate buffer saline
- rAAV vectors are promising delivery gene delivery systems, based on the defective and non-pathogenic parvovirus adeno-associated virus.
- Most vectors are derived from a plasmid substrate which retains only the AAV inverted terminal repeats (ITRs) flanking the transgene cassette of choice.
- ITRs inverted terminal repeats
- the deleted viral coding sequences are present on a separate template, referred to as an AAV helper of packaging plasmid.
- Generation of rAAV requires transfection of the vector and packaging constructs into adenovirus (Ad)-infected cells. Due to the lack of homology between vector and helper sequences, rAAV produced in this system is essentially free of wild-type AAV.
- the invention provides a recombinant adeno-associated vector (AAV) comprising a chimeric capsid protein which is fused to a domain of about 25-250 amino acids consisting of a repeat domain of (GIy-AIa). In a particular embodiment a domain of about 50- 200 amino acids is used. In another particular embodiment a domain of about 100-150 amino acids is used.
- said capsid protein is selected from the list consisting of VP1 and/or VP2 and/or VP3.
- said capsid protein is VP2.
- said recombinant AAV vector is derived from AAV-2.
- said recombinant adeno-associated vector comprises a chimeric capsid protein having an amino acid sequence depicted in SEQ ID NO: 2.
- said recombinant AAV vector comprises a heterologous transgene operably linked to regulatory sequences which direct its expression in a host cell.
- said heterologous transgene encodes for a blood clotting factor.
- the invention provides a pharmaceutical composition comprising a recombinant AAV vector according to the before mentioned embodiments in addition to a physiologically compatible carrier.
- the invention provides an isolated chimeric VP2 gene comprising the nucleotide sequence as depicted in SEQ ID NO: 1 which encodes a chimeric VP2 protein depicted in SEQ ID NO: 2.
- the fusion (or coupling which is an equivalent word) between the AAV capsid protein (such as for example VP2) and the GAr domain is in one embodiment a genetic fusion resulting in a chimeric capsid protein.
- Said genetic fusion can be an aminoterminal fusion with a capsid protein (such as VP2) or a carboxyterminal fusion with a capsid protein (such as VP2).
- said chimeric capsid protein is provided in trans by the AAV helper or AAV packaging construct during the recombinant AAV vector production process.
- GAr- domains are described in WO9746573 (The University of Virginia Patent Foundation) and in particular the example (pp. 8-11 ) described in the latter patent application is herein incorporated by reference.
- the capsid is a tightly packaged icosahedron of 25 nm and is composed of three different viral proteins, VP1 (90 kDa), VP2 (72 kDa), and VP3 (60 kDa). Said proteins are encoded in the same open reading frame (ORF) and share a common stop codon. They differ in their aminotermini due to alternative splicing and different initiation codons, resulting in three progressively shorter proteins.
- ORF open reading frame
- a chimeric capsid can be formed through insertion of a GAr-domain in the amino acid sequence of a particular capsid protein.
- a chimeric capsid protein proves to be non-functional in the packaging process (or capsid assembly process)
- additional wild type capsid can be provided in trans (by a specific capsid-encoding plasmid).
- Genetic coupling between the GAR domain and capsid proteins can be interrupted by a spacer encoding about 0 to about 30 amino acids.
- the coupling between a capsid protein and a GAr-domain is carried out via a chemical coupling. Methods of chemical coupling also known as cross-linking technologies are known by a person skilled in the art. In case chemical coupling with a GAr- domain is envisaged it is most conveniently carried out on a purified recombinant AAV vector.
- an “AAV vector” is meant a vector derived from any adeno-associated virus serotype isolated from any animal species, including without limitation, vectors mentioned in Gao G. et al (2005) New recombinant serotypes of AAV vectors. Curr. Gene Ther. 3:285-97.
- AAV vectors can have one or more of the AAV wild-type genes deleted in whole or part, preferably the rep and/or cap genes, but retain functional flanking ITR sequences. Functional ITR sequences are necessary for the rescue, replication and packaging of the AAV virion.
- an AAV vector is defined herein to include at least those sequences required in cis for replication and packaging
- the ITRs need not be the wild-type nucleotide sequences, and may be altered, e.g., by the insertion, deletion or substitution of nucleotides, so long as the sequences provide for functional rescue, replication and packaging.
- AAV helper or packaging functions refer to AAV-derived coding sequences which can be expressed to provide AAV gene products that, in turn, function in trans for productive AAV replication.
- AAV helper functions include both of the major AAV open reading frames (ORFs), rep and cap.
- the Rep expression products have been shown to possess many functions, including, among others: recognition, binding and nicking of the AAV origin of DNA replication; DNA helicase activity; and modulation of transcription from AAV (or other heterologous) promoters.
- the Cap expression products supply necessary packaging functions.
- AAV helper functions are used herein to complement AAV functions in trans that are missing from AAV vectors.
- AAV helper or packaging construct refers generally to a nucleic acid molecule that includes nucleotide sequences providing AAV functions deleted from an AAV vector which is to be used to produce a transducing vector for delivery of a nucleotide sequence of interest.
- AAV helper constructs are commonly used to provide transient expression of AAV rep and/or cap genes to complement missing AAV functions that are necessary for lytic AAV replication; however, helper constructs lack AAV ITRs and can neither replicate nor package themselves.
- AAV helper constructs can be in the form of a plasmid, phage, transposon, cosmid, virus, or virion. A number of AAV helper constructs and vectors that encode Rep and/or Cap expression products have been described.
- accessory functions refers to non-AAV derived viral and/or cellular functions upon which AAV is dependent for its replication.
- captures proteins and RNAs that are required in AAV replication including those moieties involved in activation of AAV gene transcription, stage specific AAV mRNA splicing, AAV DNA replication, synthesis of Cap expression products and AAV capsid assembly.
- Viral-based accessory functions can be derived from any of the known helper viruses such as adenovirus, herpesvirus (other than herpes simplex virus type-1 ) and vaccinia virus.
- accessory function vector refers generally to a nucleic acid molecule that includes nucleotide sequences providing accessory functions.
- An accessory function vector can be transfected into a suitable host cell, wherein the vector is then capable of supporting AAV virion production in the host cell.
- functional viral particles as they exist in nature, such as adenovirus, herpesvirus or vaccinia virus particles.
- accessory function vectors can be in the form of a plasmid, phage, transposon or cosmid. It has been demonstrated that the full-complement of adenovirus genes are not required for accessory helper functions.
- adenovirus mutants incapable of DNA replication and late gene synthesis have been shown to be permissive for AAV replication.
- recombinant virus is meant a virus that has been genetically altered, e.g., by the addition or insertion of a heterologous nucleic acid construct into the particle.
- AAV virus is meant a complete virus particle, such as a wild-type (wt) AAV virus particle (comprising a linear, single-stranded AAV nucleic acid genome associated with an AAV capsid protein coat).
- single-stranded AAV nucleic acid molecules of either complementary sense, e.g., "sense” or “antisense” strands can be packaged into any one AAV virus and both strands are equally functional.
- the words 'virus' and 'virion' are herein used interchangeable.
- a "recombinant AAV virus” or “rAAV virus” or “rAAV virion” or “rAAV vector” - which are all equivalent terms - is defined herein as a functional, replication-defective virus including an AAV protein shell, encapsidating a heterologous nucleotide sequence of interest which is flanked on both sides by AAV ITRs.
- a rAAV virus is produced in a suitable host cell which has had an AAV vector, AAV helper functions and accessory functions introduced therein.
- the host cell is rendered capable of encoding AAV polypeptides that are required for packaging the AAV vector (containing a recombinant nucleotide sequence of interest) into (a) functional recombinant virus particle(s) for subsequent gene delivery.
- transfection is used to refer to the uptake of foreign DNA by a cell, and a cell has been "transfected" when exogenous DNA has been introduced inside the cell membrane.
- transfection techniques are generally known in the art. Such techniques can be used to introduce one or more exogenous DNA moieties, such as an episomal or integrative vector into suitable host cells.
- the term 'transduction' refers to viral vector mediated gene transfer.
- control sequences refers collectively to promoter sequences, polyadenylation signals, transcription termination sequences, upstream regulatory domains, origins of replication, internal ribosome entry sites ("IRES"), enhancers, and the like, which collectively provide for the replication, transcription and translation of a coding sequence in a recipient cell. Not all of these control sequences need always be present so long as the selected coding sequence is capable of being replicated, transcribed and translated in an appropriate host cell.
- promoter refers to a nucleotide region comprising a DNA regulatory sequence, wherein the regulatory sequence is derived from a gene which is capable of binding RNA polymerase and initiating transcription of a downstream (3'-direction) coding sequence.
- Transcription promoters can include "inducible promoters” (where expression of a polynucleotide sequence operably linked to the promoter is induced by an analyte, cofactor, regulatory protein, etc.), “inducible promoters” (where expression of a polynucleotide sequence operably linked to the promoter is induced by an analyte, cofactor, regulatory protein, etc.), and “constitutive promoters”.
- an effective amount or “therapeutically effective amount” of a composition or agent refers to a nontoxic but sufficient amount of the composition or agent to provide the desired response.
- the exact amount required will vary from subject to subject, depending on the species, age, and general condition of the subject, the severity of the condition being treated, and the particular macromolecule of interest, mode of administration, and the like.
- An appropriate "effective” amount in any individual case may be determined by one of ordinary skill in the art using routine experimentation.
- the recombinant AAV vectors of the present invention are used for the manufacture of a medicament to treat a variety of diseases.
- the type of disease depends on the type of heterologous gene present in the recombinant AAV vector.
- "Treating" or “treatment” of a disease includes: (1 ) preventing the disease, i.e. causing the clinical symptoms of the disease not to develop in a subject that may be exposed to or predisposed to the disease but does not yet experience or display symptoms of the disease, (2) inhibiting the disease, i.e., arresting the development of the disease or its clinical symptoms, or (3) relieving the disease, i.e., causing regression of the disease or its clinical symptoms.
- the invention preferably encompasses rAAV viruses comprising at least one heterologous nucleotide sequence coding for one or more polypeptides, the rAAV viruses preferably administered to one or more cells or tissue of a mammal.
- the invention embraces the delivery of at least one heterologous nucleotide sequence encoding one or more peptides, polypeptides, or proteins, which are useful for the treatment or prevention of disease states in a mammalian subject.
- the invention also includes novel mutant viruses comprising a gene or genes coding for blood coagulation proteins, which proteins may be delivered to the cells of a mammal having hemophilia for the treatment of hemophilia.
- the invention includes: delivery of the Factor IX gene to a mammal for treatment of hemophilia B, delivery of the Factor VIII gene to a mammal for treatment of hemophilia A, delivery of the Factor VII gene for treatment of Factor VII deficiency, delivery of the Factor X gene for treatment of Factor X deficiency, delivery of the Factor Xl gene for treatment of Factor Xl deficiency, delivery of the Factor XIII gene for treatment of Factor XIII deficiency, and, delivery of the Protein C gene for treatment of Protein C deficiency.
- each of the above-recited genes to the cells of a mammal is accomplished by first generating a rAAV virus comprising the gene and then administering the rAAV virus to the mammal.
- the invention includes rAAV viruses comprising genes encoding any one of Factor IX, Factor VIII, Factor X, Factor VII, Factor Xl, Factor XIII or Protein C. Delivery of the recombinant viruses containing one or more heterologous gene sequences to a mammalian subject may be by intramuscular injection or by administration into the bloodstream of the mammalian subject.
- Administration into the bloodstream may be by injection into a vein, an artery, or any other vascular conduit the recombinant viral vectors into the bloodstream by way of isolated limb perfusion, a technique well known in the surgical arts, the method essentially enabling the artisan to isolate a limb from the systemic circulation prior to administration of the rAAV viruses.
- isolated limb perfusion technique described in U.S. Pat. No. 6,177,403 and herein incorporated by reference, can also be employed by the skilled artisan to administer the mutant viruses into the vasculature of an isolated limb to potentially enhance transduction into muscle cells or tissue.
- CNS central nervous system
- CNS central nervous system
- glial cells glial cells
- astrocytes glial cells
- CSF cereobrospinal fluid
- interstitial spaces bone, cartilage and the like.
- Recombinant AAV virions or cells transduced in vitro may be delivered directly to the CNS or brain by injection into, e.g., the ventricular region, as well as to the striatum (e.g., the caudate nucleus or putamen of the striatum), spinal cord and neuromuscular junction, or cerebellar lobule, with a needle, catheter or related device, using neurosurgical techniques known in the art, such as by stereotactic injection (see, e.g., Stein et al., J Virol 73:3424-3429, 1999; Davidson et al., PNAS 97:3428-3432, 2000; Davidson et al., Nat. Genet.
- the dose of rAAV virions required to achieve a particular "therapeutic effect,” e.g., the units of dose in vector genomes/per kilogram of body weight (vg/kg), will vary based on several factors including, but not limited to: the route of rAAV virus administration, the level of gene (or anti- sense RNA or ribozyme or shRNA) expression required to achieve a therapeutic effect, the specific disease or disorder being treated, a host immune response to the rAAV virus, a host immune response to the gene (or anti-sense RNA or ribozyme) expression product, and the stability of the gene (or anti-sense RNA or ribozyme) product.
- rAAV virus dose range to treat a patient having a particular disease or disorder based on the aforementioned factors, as well as other factors that are well known in the art.
- therapeutic effect is meant a level of expression of one or more heterologous gene sequences sufficient to alter a component of a disease (or disorder) toward a desired outcome or clinical endpoint, such that a patient's disease or disorder shows clinical improvement, often reflected by the amelioration of a clinical sign or symptom relating to the disease or disorder.
- a "therapeutic effect" for hemophilia is defined herein as an increase in the blood-clotting efficiency of a mammal afflicted with hemophilia, efficiency being determined, for example, by well known endpoints or techniques such as employing assays to measure whole blood clotting time. Reductions in either whole blood clotting time are indications of an increase in blood-clotting efficiency.
- hemophiliacs having less than 1 % of normal levels of Factor VIII or Factor IX have a whole blood clotting time of greater than 60 minutes as compared to approximately 10 minutes for non-hemophiliacs.
- pCMV-EBNA was digested with BstXI and an adapter containing a BgI Il site was cloned into this site to generate plasmid pCMV-EBNA(Bglll).
- the BgIII adapter was created by annealing the synthetic phosphorylated oligonucleotides 5'-TACG/AG/A7C7GAAG-3 (SEQ ID NO: 3) and 5'- ⁇ G,47C7CGTACTTC-3 (SEQ I D NO: 4) (The BgII I site is denoted in italics).
- the VP2 sequence was PCR amplified from plasmid pAAV-RC (Stratagene) using primers: 5'- TAAGATCTTGCTCCGGGAAAAAAGAGGCCGGTAG-3' (S E Q I D N O : 5) a n d 5 '- TAA GA TCTTTACAGATTACGAGTCAGGTAT-S' (S EQ I D NO : 6) and cloned after BgIII restriction into the corresponding BgIII site of the Bglll-restricted pCMV-EBNA(Bglll) vector. An extra T was included in the 5' primer next to the BgIII site to make sure the VP2 is in frame with the Gar-domain-containing polypeptide.
- the translation start codon ACG was deleted.
- This genetic fusion is depicted in SEQ ID NO: 1 and the corresponding amino acid sequence encoded by this fusion sequence is denoted in SEQ ID NO: 2.
- the corresponding plasmid comprising SEQ ID NO: 1 is designated as pGar-VP2.
- An AAV helper plasmid was generated in which the VP2 startcodon is mutated from ACG to ACC.
- a G-to-C substitution within the wobble position of the VP2 start codon was hereby introduced which consequently abolishes VP2 expression without altering the amino acid sequence of VP1.
- the resulting plasmid is designated as pAAV-RC ⁇ st a r t-
- pAAV-RC ⁇ s ta r t For the vector production pGar-VP2, pAAV-RC ⁇ s ta r t, pHelper and AAV vector were contransfected in HEK293 cells at a 1 :1 :2:1 w/w ratio.
- AAV vectors encoded green fluorescent protein (GFP) or a therapeutically relevant protein (e.g. clotting factor IX) (designated as AAV-GFP or AAV-FIX, see also VandenDriessche et al. (2007). J. Thromb. Hemost; 5(1 ): 16-24).
- GFP green fluorescent protein
- IX therapeutically relevant protein
- the AAV-GFP vector (SEQ ID NO: 9) expresses GFP from a CMV promoter
- the AAV-FIX vector (SEQ ID NO: 10) expresses human FIX cDNA driven from a chimeric promoter/enhancer composed of the transthyretin promoter along with a serpin enhancer.
- the AAV-FIX vector also contains a truncated FIX intron A.
- the cognate AAV2 vectors are packaged by cotransfecting pAAVRC and pHelper.
- the recombinant AAV vector comprising SEQ ID NO: 1 are then purified according to Lux K. et al (2005) J. of Virology 79(18): 1 1776, page 1 1777 - materials and methods section - here incorporated by reference or by cesium chloride purification (VandenDriessche et al. (2007). J. Thromb. Hemost.; 5(1 ):16-24).
- the determination of AAV titers is based on the same previous references and is determined by real-time quantitative PCR using vector-specific primers.
- TaqMan qPCR was performed in accordance with the manufacturer's recommendations (50 0 C for 2 min, 95°C for 1 0 min and 40 cycles of 95°C 1 5 sec, 60 0 C 1 min ), using a serially diluted plasmid corresponding to a known amount of vector as standard.
- the number of genome copies (gc) in 8.5 microliter of a 10-fold serially diluted reference samples of known quantitaties is shown in Fig. 1A & B and ranges from 10 8 to 10 2 gc/8.5 microliter.
- the qPCR profiles of the experimental samples Gar-AAV-GFP (Fig.1A) and Gar-AAV-FIX are also shown (Fig. 1 B) and did not differ from non Gar-modified AAV.
- the qPCR signal was multiplied with a dilution factor (i.e. x 100 x 1000 x 50)/(8,5 x 1 ,5) to obtain the particle titer expressed in gc/ml.
- the particle titers for Gar-AAV-GFP corresponded to 1 ,08x10 12 gc/ml and for Gar-AAV-FIX to 2,15x10 12 gc/ml. This indicates that the Gar-modified AAV is capable of packaging viral vector genomes.
- Gar-modified AAV is tested by in vitro or in vivo transduction (and compared to non-modified wt AAV) using reporter genes (e.g. green fluorescent protein, GFP) and a therapeutically relevant protein (particularly FIX), as described (VandenDriessche et al. (2007). J. Thromb. Hemost.; 5(1 ):16-24).
- reporter genes e.g. green fluorescent protein, GFP
- FIX therapeutically relevant protein
- GFP expression in transduced cells in vitro or in vivo is determined by fluorescence or confocal microscopy following transduction with Gar- modified AAV2 vectors versus unmodified wild-type AAV2 vectors as controls.
- AAV293, 293T and HeLa cellines were exposed to different doses of Gar- AAV-GFP vectors (50 ⁇ l, 30 ⁇ l, 20 ⁇ l, 10 ⁇ l, 5 ⁇ l, 2.5 ⁇ l in a total volume of 1 ml of culture medium (DMEM + 10% fetal bovine serum). Cells were kept in incubator at 37°C, 5% CO 2 and after 2 days GFP expression was determined by fluorescence microscopy.
- the results shown in Fig. 2 indicate that the Gar-AAV-GFP vector were fully functional and capable of transferring a gene (in casu the GFP reporter gene) into various different cell types leading to robust expression. This reflects the broad tropism of the Gar-modified AAV vectors.
- AAV capsids are not expressed de novo in transduced target cells , it is possible to assess the MHC class I restricted CTL response using target cells that are transfected with expression constructs encoding the cognate capsid antigens.
- CTL targets that express Gar-modified AAV capsid proteins.
- Gar-VP2 expression constructs are transfected into MHC-l-positive cells (vs. unmodified VP2 expression construct, as control). The expression of Gar-VP2 vs.
- AAV capsid-specific CTLs ("effector” cells) are generated by immunizing mice with AAV capsid expression constructs, as described previously (Sabatino et al., MoI. Ther., 12(6):1023-33., 2005). The splenocytes from immunized mice containing the "effector” CTLs are co-cultured with the "target” cells during an in vitro stimulation phase. Subsequently, IFN-gamma production by the "effector” cells is assessed following coincubation of effector and target cells at different effecto ⁇ target ratios (Ossevoort et al., Gene Ther.
- the Gar domain prevents recognition of Gar-VP2 expressing cells by VP2-specific CTLs consistent with a reduction in IFN-gamma production.
- RT-PCR analysis shows that expression of Gar-modified VP2 is more prolonged compared to unmodified VP2.
Landscapes
- Health & Medical Sciences (AREA)
- Life Sciences & Earth Sciences (AREA)
- Chemical & Material Sciences (AREA)
- Engineering & Computer Science (AREA)
- Bioinformatics & Cheminformatics (AREA)
- Genetics & Genomics (AREA)
- Organic Chemistry (AREA)
- General Health & Medical Sciences (AREA)
- Zoology (AREA)
- Medicinal Chemistry (AREA)
- Wood Science & Technology (AREA)
- Biomedical Technology (AREA)
- Proteomics, Peptides & Aminoacids (AREA)
- Gastroenterology & Hepatology (AREA)
- General Engineering & Computer Science (AREA)
- Biochemistry (AREA)
- Biotechnology (AREA)
- Pharmacology & Pharmacy (AREA)
- Epidemiology (AREA)
- Animal Behavior & Ethology (AREA)
- Public Health (AREA)
- Veterinary Medicine (AREA)
- Immunology (AREA)
- Molecular Biology (AREA)
- Hematology (AREA)
- Microbiology (AREA)
- Virology (AREA)
- Biophysics (AREA)
- Physics & Mathematics (AREA)
- Plant Pathology (AREA)
- Micro-Organisms Or Cultivation Processes Thereof (AREA)
- Medicines Containing Material From Animals Or Micro-Organisms (AREA)
Abstract
The present invention relates to the field of gene therapy more specifically to the field of adeno-associated viral gene vectors. The present invention provides a novel recombinant AAV vector comprising a chimeric capsid protein fused to a Gly-Ala repeat domain.
Description
Novel AAV vector and uses thereof
Field of the invention
The present invention relates to the field of gene therapy more specifically to the field of adeno-associated viral gene vectors. The present invention provides a novel recombinant AAV vector comprising a chimeric capsid protein fused to a GIy-AIa repeat domain.
Background to the invention
Adeno-associated virus (AAV) is a parvovirus belonging to the genus Dependovirus. The virus is presently being evaluated as an attractive gene therapy vector because it possesses several promising features not found in other viruses. Indeed, AAV can infect a wide range of host cells, including non-dividing cells and it infects also cells from different species. More importantly, AAV has not been associated with any human or animal disease and does not appear to alter the physiological properties of the host cell upon integration. Furthermore, AAV is stable at a wide range of physical and chemical conditions, which lends itself to production, storage, and transportation requirements.
Being a single-stranded DNA virus, AAV includes two large open reading frames (ORFs), known as the AAV replication (rep) and capsid (cap) regions. These ORFs encode replication and capsid gene products, respectively: replication and capsid gene products allow for the replication, assembly, and packaging of a complete AAV virion. More specifically, a family of at least four viral proteins are expressed from the AAV rep region: Rep 78, Rep 68, Rep 52, and Rep 40, all of which are named for their apparent molecular weights. The AAV cap region encodes at least three proteins: VP1 , VP2, and VP3. Said rep and capsid genes are flanked by 165-bp inverted terminal repeats (ITRs). By replacement of the endogenous sequences between the ITRs, recombinant AAV (rAAV) creates a genetic cassette that can be packaged into intact virions and delivered preferentially to specific cell types. Following virus entry, nuclear trafficking, and uncoating, the released genome becomes double stranded and via intra- or intermolecular recombination of the ITRs persists as circular monomers or concatemers.
In nature, AAV is a helper virus-dependent virus, i.e., it requires co-infection with a helper virus (e.g., adenovirus, herpesvirus, or vaccinia virus) in order to form functionally complete AAV virions. In the absence of co-infection with a helper virus, AAV establishes a latent state in which the viral genome inserts into a host cell chromosome or exists in an episomal form, but functional virions are not produced. Subsequent infection by a helper virus "rescues" the
integrated genome, allowing it to be replicated and packaged into viral capsids, thereby reconstituting the functional virion. While AAV can infect cells from different species, the helper virus must be of the same species as the host cell. To construct functional recombinant AAV (rAAV) containing a nucleic acid, a suitable host cell line is transfected with an AAV vector containing a nucleic acid. AAV helper functions and accessory functions are then expressed in the host cell. In gene therapeutic uses - because the patient's cells lack the rep and cap genes, as well as the adenovirus accessory function genes - the rAAV are replication defective; i.e. they cannot further replicate and package their genomes. There are currently a number of AAV serotypes described which are able to infect humans. AAV-2 is the best characterized and has been used to successfully deliver transgenes to several cell lines, tissue types, and organs in a variety of in vitro and in vivo assays. Notwithstanding the promises of recombinant AAV vectors as a gene therapy vector there exists a severe limitation because of the immune response directed against vector and/or transgene product which significantly impedes the persistent expression of the transgenes encoding neoantigens. Long-term humoral immunity against several viral-vector systems prevails in a large part of the population, or may be induced upon the first vector administration. This may frustrate re-administration of the vector and lead to elimination of the transduced cells. Indeed, it is observed that after an initial treatment with a given AAV serotype, anti-AAV capsid neutralizing antibodies are often made which prevent subsequent treatments by the same serotype. Although the AAV virion induces a strong humoral immune response, it cannot transduce dendritic cells (DCs) efficiently and is thought to be less immunogenic for cytotoxic T-lymphocyte (CTL) induction. However, recently the ability of rAAV to induce a CTL response resulting in the eradication of transduced hepatocytes was suggested as lying at the basis for the failure of a recent rAAV clinical trial for factor IX gene addition therapy (Mingozzi F et al (2007 Nature Medicine 13, 4, 419). In the latter trial therapeutic levels of FIX in the blood were detected from 2 to 6 weeks postinjection of rAAV2/FIX and then decreased to baseline. It should be said that the scientific data on the CTL response against rAAV transduced cells are conflicting because in a later study Li C et al (2007) Journal of Virology 81 (4), 7540 did not demonstrate in vivo elimination of rAAV2- transduced liver cells or AAV2-transduced muscle cells by AAV2 capsid-specific cytotoxic T lymphocytes. Still other studies suggest elimination of rAAV-transduced cells by the activation of NK cells (Li H et al (2007) MoI. Ther. 15:792). It is clear that the precise mechanisms of the loss of stable expression of therapeutic proteins in AAV gene therapy clinical trials are not understood. To circumvent liver toxicity and CD8+ T-cell responses (while maintaining efficacy) the use of transient immunosuppression has been proposed in future gene therapy trials. However, the use of transient immunosuppression is not without risks and therefore there exists a clear need for the development of more robust vectors as a viable alternative.
Summary of the invention
In the present invention we have developed a novel recombinant AAV vector which comprises a capsid protein that is coupled to a GIy-AIa repeat domain (GAR-domain). Said GAR-domain has been used to inhibit the T-cell mediated immune rejection of heterologous fusion proteins produced by adenoviral-transduced cells (Ossevoort M et al (2003) Gene Therapy 10, 2020).
Our novel recombinant vector surprisingly leads to a sustained production of therapeutic proteins in an animal host, as it could not have been predicted that such a large sequence would not interfere with protein expression.
Brief description of the figures
Figure 1 shows a real-time qPCR on the AAV-EBNA vector particles. DNA was extracted from the particles and subjected to a real-time qPCR (TaqMan) with vector specific primers/probe (see Example 1 ). AAV particles that encapsidate the corresponding vector genomes should yield a signal, as shown here for the GFP (Fig. 1A) and FIX (Fig. 1 B) vectors.
Figure 2 represents a microscopic fluorescent image of different cell types transduced with the (stealth) AAV vector. It clearly shows that the EBNA-modified AAV vector can transduce different cell types in a dose-dependent manner consistent with increasing percentage of GFP- positive cells and increased GFP expression level/cell. This indicates that the EBNA-modified AAV vector is functional and capable of delivering genes (in casu GFP reporter gene) to different cell types.
Figure 3 shows a microscopic fluorescent image of hepatocytes. Left panel: negative control animals injected with PBS (phosphate buffer saline), right panel: hepatocytes transduced with the Gar-modified vector.
In Figure 4, values and standard curve of human FIX in mouse plasma are shown. The standard curve is based on serially diluted known amounts of human FIX spiked in mouse plasma (diamonds on graph). The actual OD values of the mouse plasma from the recipient mouse injected with the Gar-AAV-FIX vector was plotted (circles) onto this linear standard curve.
Detailed description of the invention
The practice of the present invention employs, unless otherwise indicated, conventional methods of chemistry, biochemistry, recombinant DNA techniques and immunology, within the skill of the art. Such techniques are explained fully in the literature. See, e.g., Fundamental Virology, current edition, vol. I & Il (B. N. Fields and D. M. Knipe, eds.); Handbook of Experimental Immunology, VoIs. I-IV (D. M. Weir and C. C. Blackwell eds., Blackwell Scientific Publications); T. E. Creighton, Proteins: Structures and Molecular Properties (W. H. Freeman and Company, 1993); A. L. Lehninger, Biochemistry (Worth Publishers, Inc., current edition); Sambrook, et al., Molecular Cloning: A Laboratory Manual (current edition); Methods In Enzymology (S. Colowick and N. Kaplan eds., Academic Press, Inc.).
Recombinant adeno-associated virus (rAAV) vectors are promising delivery gene delivery systems, based on the defective and non-pathogenic parvovirus adeno-associated virus. Most vectors are derived from a plasmid substrate which retains only the AAV inverted terminal repeats (ITRs) flanking the transgene cassette of choice. The deleted viral coding sequences are present on a separate template, referred to as an AAV helper of packaging plasmid. Generation of rAAV requires transfection of the vector and packaging constructs into adenovirus (Ad)-infected cells. Due to the lack of homology between vector and helper sequences, rAAV produced in this system is essentially free of wild-type AAV.
In a first embodiment the invention provides a recombinant adeno-associated vector (AAV) comprising a chimeric capsid protein which is fused to a domain of about 25-250 amino acids consisting of a repeat domain of (GIy-AIa). In a particular embodiment a domain of about 50- 200 amino acids is used. In another particular embodiment a domain of about 100-150 amino acids is used. In a particular embodiment said capsid protein is selected from the list consisting of VP1 and/or VP2 and/or VP3. Preferably said capsid protein is VP2. In a particular embodiment said recombinant AAV vector is derived from AAV-2.
In another particular embodiment said recombinant adeno-associated vector (AAV) comprises a chimeric capsid protein having an amino acid sequence depicted in SEQ ID NO: 2. In yet another particular embodiment said recombinant AAV vector comprises a heterologous transgene operably linked to regulatory sequences which direct its expression in a host cell. In a preferred embodiment said heterologous transgene encodes for a blood clotting factor. In yet another embodiment the invention provides a pharmaceutical composition comprising a recombinant AAV vector according to the before mentioned embodiments in addition to a physiologically compatible carrier.
In yet another embodiment the invention provides an isolated chimeric VP2 gene comprising the nucleotide sequence as depicted in SEQ ID NO: 1 which encodes a chimeric VP2 protein depicted in SEQ ID NO: 2.
The fusion (or coupling which is an equivalent word) between the AAV capsid protein (such as for example VP2) and the GAr domain is in one embodiment a genetic fusion resulting in a chimeric capsid protein. Said genetic fusion can be an aminoterminal fusion with a capsid protein (such as VP2) or a carboxyterminal fusion with a capsid protein (such as VP2). In a particular embodiment said chimeric capsid protein is provided in trans by the AAV helper or AAV packaging construct during the recombinant AAV vector production process. GAr- domains are described in WO9746573 (The University of Virginia Patent Foundation) and in particular the example (pp. 8-11 ) described in the latter patent application is herein incorporated by reference.
Different strategies are commonly known in the art to incorporate peptides into the AAV capsid. The capsid is a tightly packaged icosahedron of 25 nm and is composed of three different viral proteins, VP1 (90 kDa), VP2 (72 kDa), and VP3 (60 kDa). Said proteins are encoded in the same open reading frame (ORF) and share a common stop codon. They differ in their aminotermini due to alternative splicing and different initiation codons, resulting in three progressively shorter proteins. In a particular embodiment a chimeric capsid can be formed through insertion of a GAr-domain in the amino acid sequence of a particular capsid protein. In case such a chimeric capsid protein proves to be non-functional in the packaging process (or capsid assembly process) additional wild type capsid can be provided in trans (by a specific capsid-encoding plasmid). Genetic coupling between the GAR domain and capsid proteins can be interrupted by a spacer encoding about 0 to about 30 amino acids. In another particular embodiment the coupling between a capsid protein and a GAr-domain is carried out via a chemical coupling. Methods of chemical coupling also known as cross-linking technologies are known by a person skilled in the art. In case chemical coupling with a GAr- domain is envisaged it is most conveniently carried out on a purified recombinant AAV vector.
By an "AAV vector" is meant a vector derived from any adeno-associated virus serotype isolated from any animal species, including without limitation, vectors mentioned in Gao G. et al (2005) New recombinant serotypes of AAV vectors. Curr. Gene Ther. 3:285-97. AAV vectors can have one or more of the AAV wild-type genes deleted in whole or part, preferably the rep and/or cap genes, but retain functional flanking ITR sequences. Functional ITR sequences are necessary for the rescue, replication and packaging of the AAV virion. Thus, an AAV vector is defined herein to include at least those sequences required in cis for replication and packaging
(e.g., functional ITRs) of the virus. The ITRs need not be the wild-type nucleotide sequences,
and may be altered, e.g., by the insertion, deletion or substitution of nucleotides, so long as the sequences provide for functional rescue, replication and packaging.
"AAV helper or packaging functions" refer to AAV-derived coding sequences which can be expressed to provide AAV gene products that, in turn, function in trans for productive AAV replication. Thus, AAV helper functions include both of the major AAV open reading frames (ORFs), rep and cap. The Rep expression products have been shown to possess many functions, including, among others: recognition, binding and nicking of the AAV origin of DNA replication; DNA helicase activity; and modulation of transcription from AAV (or other heterologous) promoters. The Cap expression products supply necessary packaging functions. AAV helper functions are used herein to complement AAV functions in trans that are missing from AAV vectors.
The term "AAV helper or packaging construct" refers generally to a nucleic acid molecule that includes nucleotide sequences providing AAV functions deleted from an AAV vector which is to be used to produce a transducing vector for delivery of a nucleotide sequence of interest. AAV helper constructs are commonly used to provide transient expression of AAV rep and/or cap genes to complement missing AAV functions that are necessary for lytic AAV replication; however, helper constructs lack AAV ITRs and can neither replicate nor package themselves. AAV helper constructs can be in the form of a plasmid, phage, transposon, cosmid, virus, or virion. A number of AAV helper constructs and vectors that encode Rep and/or Cap expression products have been described. See, e.g., U.S. Pat. Nos. 6,001 ,650, 5,139,941 and 6,376,237, Samulski et al. (1989) J. Virol. 63: 3822-3828; and McCarty et al. (1991 ) J. Virol. 65: 2936- 2945.
The term "accessory functions" refers to non-AAV derived viral and/or cellular functions upon which AAV is dependent for its replication. Thus, the term captures proteins and RNAs that are required in AAV replication, including those moieties involved in activation of AAV gene transcription, stage specific AAV mRNA splicing, AAV DNA replication, synthesis of Cap expression products and AAV capsid assembly. Viral-based accessory functions can be derived from any of the known helper viruses such as adenovirus, herpesvirus (other than herpes simplex virus type-1 ) and vaccinia virus.
The term "accessory function vector" refers generally to a nucleic acid molecule that includes nucleotide sequences providing accessory functions. An accessory function vector can be transfected into a suitable host cell, wherein the vector is then capable of supporting AAV virion production in the host cell. Expressly excluded from the term are functional viral particles as they exist in nature, such as adenovirus, herpesvirus or vaccinia virus particles. Thus, accessory function vectors can be in the form of a plasmid, phage, transposon or cosmid.
It has been demonstrated that the full-complement of adenovirus genes are not required for accessory helper functions. In particular, adenovirus mutants incapable of DNA replication and late gene synthesis have been shown to be permissive for AAV replication. By "recombinant virus" is meant a virus that has been genetically altered, e.g., by the addition or insertion of a heterologous nucleic acid construct into the particle. By "AAV virus" is meant a complete virus particle, such as a wild-type (wt) AAV virus particle (comprising a linear, single-stranded AAV nucleic acid genome associated with an AAV capsid protein coat). In this regard, single-stranded AAV nucleic acid molecules of either complementary sense, e.g., "sense" or "antisense" strands, can be packaged into any one AAV virus and both strands are equally functional. The words 'virus' and 'virion' are herein used interchangeable.
A "recombinant AAV virus" or "rAAV virus" or "rAAV virion" or "rAAV vector" - which are all equivalent terms - is defined herein as a functional, replication-defective virus including an AAV protein shell, encapsidating a heterologous nucleotide sequence of interest which is flanked on both sides by AAV ITRs. A rAAV virus is produced in a suitable host cell which has had an AAV vector, AAV helper functions and accessory functions introduced therein. In this manner, the host cell is rendered capable of encoding AAV polypeptides that are required for packaging the AAV vector (containing a recombinant nucleotide sequence of interest) into (a) functional recombinant virus particle(s) for subsequent gene delivery. The term "transfection" is used to refer to the uptake of foreign DNA by a cell, and a cell has been "transfected" when exogenous DNA has been introduced inside the cell membrane. A number of transfection techniques are generally known in the art. Such techniques can be used to introduce one or more exogenous DNA moieties, such as an episomal or integrative vector into suitable host cells. The term 'transduction' refers to viral vector mediated gene transfer.
The term DNA "control sequences" refers collectively to promoter sequences, polyadenylation signals, transcription termination sequences, upstream regulatory domains, origins of replication, internal ribosome entry sites ("IRES"), enhancers, and the like, which collectively provide for the replication, transcription and translation of a coding sequence in a recipient cell. Not all of these control sequences need always be present so long as the selected coding sequence is capable of being replicated, transcribed and translated in an appropriate host cell. The term "promoter" refers to a nucleotide region comprising a DNA regulatory sequence, wherein the regulatory sequence is derived from a gene which is capable of binding RNA polymerase and initiating transcription of a downstream (3'-direction) coding sequence. Transcription promoters can include "inducible promoters" (where expression of a polynucleotide sequence operably linked to the promoter is induced by an analyte, cofactor,
regulatory protein, etc.), "inducible promoters" (where expression of a polynucleotide sequence operably linked to the promoter is induced by an analyte, cofactor, regulatory protein, etc.), and "constitutive promoters".
The terms "effective amount" or "therapeutically effective amount" of a composition or agent, as provided herein, refer to a nontoxic but sufficient amount of the composition or agent to provide the desired response. The exact amount required will vary from subject to subject, depending on the species, age, and general condition of the subject, the severity of the condition being treated, and the particular macromolecule of interest, mode of administration, and the like. An appropriate "effective" amount in any individual case may be determined by one of ordinary skill in the art using routine experimentation.
In another particular embodiment the recombinant AAV vectors of the present invention are used for the manufacture of a medicament to treat a variety of diseases. The type of disease depends on the type of heterologous gene present in the recombinant AAV vector. "Treating" or "treatment" of a disease includes: (1 ) preventing the disease, i.e. causing the clinical symptoms of the disease not to develop in a subject that may be exposed to or predisposed to the disease but does not yet experience or display symptoms of the disease, (2) inhibiting the disease, i.e., arresting the development of the disease or its clinical symptoms, or (3) relieving the disease, i.e., causing regression of the disease or its clinical symptoms. The invention preferably encompasses rAAV viruses comprising at least one heterologous nucleotide sequence coding for one or more polypeptides, the rAAV viruses preferably administered to one or more cells or tissue of a mammal. Thus, the invention embraces the delivery of at least one heterologous nucleotide sequence encoding one or more peptides, polypeptides, or proteins, which are useful for the treatment or prevention of disease states in a mammalian subject. The invention also includes novel mutant viruses comprising a gene or genes coding for blood coagulation proteins, which proteins may be delivered to the cells of a mammal having hemophilia for the treatment of hemophilia. Thus, the invention includes: delivery of the Factor IX gene to a mammal for treatment of hemophilia B, delivery of the Factor VIII gene to a mammal for treatment of hemophilia A, delivery of the Factor VII gene for treatment of Factor VII deficiency, delivery of the Factor X gene for treatment of Factor X deficiency, delivery of the Factor Xl gene for treatment of Factor Xl deficiency, delivery of the Factor XIII gene for treatment of Factor XIII deficiency, and, delivery of the Protein C gene for treatment of Protein C deficiency. Delivery of each of the above-recited genes to the cells of a mammal is accomplished by first generating a rAAV virus comprising the gene and then administering the rAAV virus to the mammal. Thus, the invention includes rAAV viruses comprising genes encoding any one of Factor IX, Factor VIII, Factor X, Factor VII, Factor Xl, Factor XIII or Protein C.
Delivery of the recombinant viruses containing one or more heterologous gene sequences to a mammalian subject may be by intramuscular injection or by administration into the bloodstream of the mammalian subject. Administration into the bloodstream may be by injection into a vein, an artery, or any other vascular conduit the recombinant viral vectors into the bloodstream by way of isolated limb perfusion, a technique well known in the surgical arts, the method essentially enabling the artisan to isolate a limb from the systemic circulation prior to administration of the rAAV viruses. A variant of the isolated limb perfusion technique, described in U.S. Pat. No. 6,177,403 and herein incorporated by reference, can also be employed by the skilled artisan to administer the mutant viruses into the vasculature of an isolated limb to potentially enhance transduction into muscle cells or tissue. Moreover, for certain conditions, it may be desirable to deliver the mutant viruses to the central nervous system (CNS) of a subject. By "CNS" is meant all cells and tissue of the brain and spinal cord of a vertebrate. Thus, the term includes, but is not limited to, neuronal cells, glial cells, astrocytes, cereobrospinal fluid (CSF), interstitial spaces, bone, cartilage and the like. Recombinant AAV virions or cells transduced in vitro may be delivered directly to the CNS or brain by injection into, e.g., the ventricular region, as well as to the striatum (e.g., the caudate nucleus or putamen of the striatum), spinal cord and neuromuscular junction, or cerebellar lobule, with a needle, catheter or related device, using neurosurgical techniques known in the art, such as by stereotactic injection (see, e.g., Stein et al., J Virol 73:3424-3429, 1999; Davidson et al., PNAS 97:3428-3432, 2000; Davidson et al., Nat. Genet. 3:219-223, 1993; and Alisky and Davidson, Hum. Gene Ther. 11 :2315-2329, 2000). The dose of rAAV virions required to achieve a particular "therapeutic effect," e.g., the units of dose in vector genomes/per kilogram of body weight (vg/kg), will vary based on several factors including, but not limited to: the route of rAAV virus administration, the level of gene (or anti- sense RNA or ribozyme or shRNA) expression required to achieve a therapeutic effect, the specific disease or disorder being treated, a host immune response to the rAAV virus, a host immune response to the gene (or anti-sense RNA or ribozyme) expression product, and the stability of the gene (or anti-sense RNA or ribozyme) product. One of skill in the art can readily determine a rAAV virus dose range to treat a patient having a particular disease or disorder based on the aforementioned factors, as well as other factors that are well known in the art. Generally speaking, by "therapeutic effect" is meant a level of expression of one or more heterologous gene sequences sufficient to alter a component of a disease (or disorder) toward a desired outcome or clinical endpoint, such that a patient's disease or disorder shows clinical improvement, often reflected by the amelioration of a clinical sign or symptom relating to the disease or disorder. Using hemophilia as a specific disease example, a "therapeutic effect" for hemophilia is defined herein as an increase in the blood-clotting efficiency of a mammal
afflicted with hemophilia, efficiency being determined, for example, by well known endpoints or techniques such as employing assays to measure whole blood clotting time. Reductions in either whole blood clotting time are indications of an increase in blood-clotting efficiency. In severe cases of hemophilia, hemophiliacs having less than 1 % of normal levels of Factor VIII or Factor IX have a whole blood clotting time of greater than 60 minutes as compared to approximately 10 minutes for non-hemophiliacs. Expression of 1 % or greater of Factor VIII or Factor IX has been shown to reduce whole blood clotting time in animal models of hemophilia, so achieving a circulating Factor VIII or Factor IX plasma concentration of greater than 1 % will likely achieve the desired therapeutic effect of an increase in blood-clotting efficiency.
Examples
1 ) Production of a rAAV vector comprising a chimeric VP2 capsid-GAr fusion
A (Gly-Ala)-repeat domain (Gar-domain) comprising a contiguous 239 amino-acid sequence composed of G or A was genetically fused to the nucleotide sequence encoding the VP2- p r o t e i n . T h e p l a s m i d p C M V-EBNA (www.addgene.org/pgvec1 ?f=v&cmd=viewvecseq&from=&soid=5205&view=Draw) was obtained from Invitrogen and encodes the EBNA-1 antigen that contains the Gar-domain. pCMV-EBNA was digested with BstXI and an adapter containing a BgI Il site was cloned into this site to generate plasmid pCMV-EBNA(Bglll). The BgIII adapter was created by annealing the synthetic phosphorylated oligonucleotides 5'-TACG/AG/A7C7GAAG-3 (SEQ ID NO: 3) and 5'-ΛG,47C7CGTACTTC-3 (SEQ I D NO: 4) (The BgII I site is denoted in italics). The VP2 sequence was PCR amplified from plasmid pAAV-RC (Stratagene) using primers: 5'- TAAGATCTTGCTCCGGGAAAAAAGAGGCCGGTAG-3' (S E Q I D N O : 5) a n d 5 '- TAA GA TCTTTACAGATTACGAGTCAGGTAT-S' (S EQ I D NO : 6) and cloned after BgIII restriction into the corresponding BgIII site of the Bglll-restricted pCMV-EBNA(Bglll) vector. An extra T was included in the 5' primer next to the BgIII site to make sure the VP2 is in frame with the Gar-domain-containing polypeptide. To avoid translation from the natural VP2 start codon, the translation start codon ACG was deleted. This genetic fusion is depicted in SEQ ID NO: 1 and the corresponding amino acid sequence encoded by this fusion sequence is denoted in SEQ ID NO: 2. The corresponding plasmid comprising SEQ ID NO: 1 is designated as pGar-VP2. An AAV helper plasmid was generated in which the VP2 startcodon is mutated from ACG to ACC. A G-to-C substitution within the wobble position of the VP2 start codon was hereby introduced which consequently abolishes VP2 expression without altering the amino acid sequence of VP1. This was done by PCR cloning using plasmid pAAV-RC (Stratagene) as template, primers and methodology as described previously (Lux K. et al (2005) J. of
Virology 79(18): 1 1776, page 1 1777 - materials and methods section. Primers used were 5'- TTCCTGGTTGAGGAACCTGTTAAGACCGCTCCGGG-3' (SEQ ID NO: 7) (containing EcoNI site) and δ'-GAGGACGTACGGGAGCTGGTACTCCGAGTCAG-S' (SEQ ID NO: 8) with BsiWI restriction site. The resulting plasmid is designated as pAAV-RCΔstart- For the vector production pGar-VP2, pAAV-RCΔstart, pHelper and AAV vector were contransfected in HEK293 cells at a 1 :1 :2:1 w/w ratio. AAV vectors encoded green fluorescent protein (GFP) or a therapeutically relevant protein (e.g. clotting factor IX) (designated as AAV-GFP or AAV-FIX, see also VandenDriessche et al. (2007). J. Thromb. Hemost; 5(1 ): 16-24). The AAV-GFP vector (SEQ ID NO: 9) expresses GFP from a CMV promoter, whereas the AAV-FIX vector (SEQ ID NO: 10) expresses human FIX cDNA driven from a chimeric promoter/enhancer composed of the transthyretin promoter along with a serpin enhancer. The AAV-FIX vector also contains a truncated FIX intron A.
As controls, the cognate AAV2 vectors are packaged by cotransfecting pAAVRC and pHelper. The recombinant AAV vector comprising SEQ ID NO: 1 are then purified according to Lux K. et al (2005) J. of Virology 79(18): 1 1776, page 1 1777 - materials and methods section - here incorporated by reference or by cesium chloride purification (VandenDriessche et al. (2007). J. Thromb. Hemost.; 5(1 ):16-24). The determination of AAV titers is based on the same previous references and is determined by real-time quantitative PCR using vector-specific primers. 1 ,5 μl of AAV vector was first subjected to DNAse I treatment to remove any traces non- encapsidated DNA by 1 hour incubation at 37°C with 4 U DNAse, 10 μl DNAse buffer and topped up to 100 μl with H2O. After heat-inactivation at 99°C for 5 min to inactivate DNase, disrupt the AAV vector particles and release the single-stranded genomes, samples were diluted 50-fold and 8.5 μl of the diluted sample were assayed in triplicate by qPCR after adding 16,5 μl ABI (Perkin Elmer) Q-PCR Master Mix and TaqMan probe and primers, according to the manufacturer's instructions (i.e. UDP-qPCR Master Mix 12,5 μl; 0,8 μl of each primers (1 OmM); 0.5 μl of the FAM-TAMRA probe (1 OmM) and H2O topped up till 16,5 μl). Vector titer was performed by quantitative (q)PCR on a ABI7500 FAST (Sequencer Detector Unit from ABI). The PCR probes/primers that were used to titer the Gar-modified AAV-GFP vector (or wild-type AAV2) or are: 5'-FAM-CTT TCC AAA ATG TCG TAA CAA CTC CGC CC- 3'-TAMRA (probe) (SEQ ID NO: 1 1 ), δ'-TGGGAGTTTGTTTTGCACCAA-S' (forward) (SEQ ID NO: 12), 5'- CGCCTACCGCCCATTTG-3' (reverse) (SEQ ID NO: 13). The PCR probes/primers that was u sed to tite r th e wi l d-type AAV2 or Gar-modified AAV-F I X vecto r a re : 5'-FAM- AACCATGACATTGCCCTTCTGGAACTGG-3' TAM RA (probe) (S E Q I D N O : 1 4 ), 5 '- CACCACAACTACAATGCAGCTATTAA-3 ' (fo rwa rd ) ( S E Q I D N O : 1 5 ), 5 '-
TGCAAATAGGTGTAACGTAGCTGTT-S' (reverse) (SEQ I D NO: 16). TaqMan qPCR was performed in accordance with the manufacturer's recommendations (500C for 2 min, 95°C for 1 0 min and 40 cycles of 95°C 1 5 sec, 600 C 1 min ), using a serially diluted plasmid corresponding to a known amount of vector as standard. The number of genome copies (gc) in 8.5 microliter of a 10-fold serially diluted reference samples of known quantitaties is shown in Fig. 1A & B and ranges from 108 to 102 gc/8.5 microliter. The qPCR profiles of the experimental samples Gar-AAV-GFP (Fig.1A) and Gar-AAV-FIX are also shown (Fig. 1 B) and did not differ from non Gar-modified AAV. The qPCR signal was multiplied with a dilution factor (i.e. x 100 x 1000 x 50)/(8,5 x 1 ,5) to obtain the particle titer expressed in gc/ml. The particle titers for Gar-AAV-GFP corresponded to 1 ,08x1012 gc/ml and for Gar-AAV-FIX to 2,15x1012 gc/ml. This indicates that the Gar-modified AAV is capable of packaging viral vector genomes. 2) Effect of Gar-modification of VP2 on rAAV function
The functionality of Gar-modified AAV is tested by in vitro or in vivo transduction (and compared to non-modified wt AAV) using reporter genes (e.g. green fluorescent protein, GFP) and a therapeutically relevant protein (particularly FIX), as described (VandenDriessche et al. (2007). J. Thromb. Hemost.; 5(1 ):16-24). Briefly, GFP expression in transduced cells in vitro or in vivo is determined by fluorescence or confocal microscopy following transduction with Gar- modified AAV2 vectors versus unmodified wild-type AAV2 vectors as controls. Subconfluent (50% confluency) AAV293, 293T and HeLa cellines were exposed to different doses of Gar- AAV-GFP vectors (50 μl, 30μl, 20μl, 10μl, 5μl, 2.5μl in a total volume of 1 ml of culture medium (DMEM + 10% fetal bovine serum). Cells were kept in incubator at 37°C, 5% CO2 and after 2 days GFP expression was determined by fluorescence microscopy. The results shown in Fig. 2 indicate that the Gar-AAV-GFP vector were fully functional and capable of transferring a gene (in casu the GFP reporter gene) into various different cell types leading to robust expression. This reflects the broad tropism of the Gar-modified AAV vectors. Similarly, when the vector was injected into mice (150 μl, i.e. 1 .6x1011 vg) it was possible to readily detect GFP-positive hepatocytes, confirming the functionality of the Gar-modified vector in vivo, whereas no fluorescence was apparent in negative control animals injected with PBS (phosphate buffer saline) (Fig. 3). Most importantly, when the Gar-AAV-FIX vector (180 μl, i.e. 4x1011 vg) was administered intravenously by tail vein injection into mice, it was possible to detect robust and sustained circulating human FIX levels in the plasma of recipient mice in the therapeutic range (216 +/- 51 ng/ml human FIX) (Fig. 4, see also Table 1 ), which would be sufficient to obtain a therapeutic effect in patients suffering from hemophilia. Blood was collected by retro-orbital bleeding under general anesthesia. The presence of human FIX in plasma samples with 20% 0.1 M sodium citrate (to prevent clotting) was determined using an enzyme-linked immunosorbent assay (Asserachrome FIX ELISA, Diagnostica Stago,
Parsippany, NJ, USA) on mouse plasma. The GFP and FIX expression confirms the functionality of the Gar-modified AAV2 vectors in vivo and underscores their therapeutic potential.
Table 1. Circulating human FIX levels in plasma of recipient mice (see Fig. 4).
3) Effect of Gar-modification on recognition of rAAV by CTL
Although AAV capsids are not expressed de novo in transduced target cells , it is possible to assess the MHC class I restricted CTL response using target cells that are transfected with expression constructs encoding the cognate capsid antigens. Hence, to test whether the recognition of Gar-modified AAV vectors by CTLs is reduced, we first generate CTL targets that express Gar-modified AAV capsid proteins. To achieve this and direct presentation of antigenic peptides derived from Gar-modified VP2 (vs. unmodified VP2, as control) to MHC class I, Gar-VP2 expression constructs are transfected into MHC-l-positive cells (vs. unmodified VP2 expression construct, as control). The expression of Gar-VP2 vs. unmodified VP2 in the transfected cells ("target" cells) is tested by RT-PCR. AAV capsid-specific CTLs ("effector" cells) are generated by immunizing mice with AAV capsid expression constructs, as described previously (Sabatino et al., MoI. Ther., 12(6):1023-33., 2005). The splenocytes from immunized mice containing the "effector" CTLs are co-cultured with the "target" cells during an in vitro stimulation phase. Subsequently, IFN-gamma production by the "effector" cells is assessed following coincubation of effector and target cells at different effectoπtarget ratios (Ossevoort et al., Gene Ther. 2003; Sabatino et al. MoI Ther 2005). The Gar domain prevents recognition of Gar-VP2 expressing cells by VP2-specific CTLs consistent with a reduction in IFN-gamma production. RT-PCR analysis shows that expression of Gar-modified VP2 is more prolonged compared to unmodified VP2.
Claims
1. A recombinant adeno-associated vector (AAV) comprising a chimeric capsid protein which is fused to a domain of about 25-250 amino acids consisting of a repeat domain of (GIy-AIa).
2. An AAV vector according to claim 1 wherein said capsid protein is selected from the list consisting of VP1 and/or VP2 and/or VP3.
3. An AAV vector according to claims 1 and 2 wherein said AAV is AAV-2.
4. A recombinant adeno-associated vector (AAV) comprising a chimeric capsid protein having an amino acid sequence depicted in SEQ ID NO: 2.
5. An AAV vector according to claims 1-4 further comprising a transgene operably linked to regulatory sequences which direct its expression in a host cell.
6. An AAV vector according to claim 5 wherein said transgene encodes for a blood clotting factor.
7. A composition comprising an AW vector according to claims 1-6 and a physiologically compatible carrier.
8. An isolated chimeric VP2 gene comprising the nucleotide sequence as depicted in SEQ ID NO: 1 which encodes a chimeric VP2 protein depicted in SEQ ID NO: 2.
Applications Claiming Priority (2)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
US566807P | 2007-12-05 | 2007-12-05 | |
US61/005,668 | 2007-12-05 |
Publications (1)
Publication Number | Publication Date |
---|---|
WO2009071679A1 true WO2009071679A1 (en) | 2009-06-11 |
Family
ID=40385509
Family Applications (1)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
PCT/EP2008/066919 WO2009071679A1 (en) | 2007-12-05 | 2008-12-05 | Novel aav vector and uses thereof |
Country Status (1)
Country | Link |
---|---|
WO (1) | WO2009071679A1 (en) |
Cited By (3)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
WO2018178067A1 (en) | 2017-03-27 | 2018-10-04 | Vrije Universiteit Brussel | Diaphragm-specific nucleic acid regulatory elements and methods and use thereof |
US11419950B2 (en) * | 2012-10-26 | 2022-08-23 | Vrije Universiteit Brussel | Vectors for liver-directed gene therapy of hemophilia and methods and use thereof |
WO2023006890A1 (en) | 2021-07-28 | 2023-02-02 | Vrije Universiteit Brussel | Improving the efficacy of muscle-targeted gene therapy |
-
2008
- 2008-12-05 WO PCT/EP2008/066919 patent/WO2009071679A1/en active Application Filing
Non-Patent Citations (4)
Title |
---|
OSSEVOORT M ET AL: "Characterization of an immuno 'stealth' derivative of the herpes simplex virus thymidine-kinase gene", CANCER GENE THERAPY, NORWALK, CT, US, vol. 13, no. 6, 1 June 2006 (2006-06-01), pages 584 - 591, XP002504134, ISSN: 0929-1903 * |
OSSEVOORT M ET AL: "Creation of immune 'stealth' genes for gene therapy through fusion with the Gly-Ala repeat of EBNA-1.", GENE THERAPY, vol. 10, no. 24, November 2003 (2003-11-01), pages 2020 - 2028, XP002517964, ISSN: 0969-7128 * |
SHARIPO<A> A ET AL: "cis-Inhibition of proteasomal degradation by viral repeats: impact of length and amino acid composition", FEBS LETTERS, ELSEVIER, AMSTERDAM, NL, vol. 499, no. 1-2, 15 June 2001 (2001-06-15), pages 137 - 142, XP004247003, ISSN: 0014-5793 * |
ZALDUMBIDE A ET AL: "How not to be seen: immune-evasion strategies in gene therapy", GENE THERAPY, MACMILLAN PRESS LTD., BASINGSTOKE, GB (PUBLISHED ONLINE 29..11.2007), vol. 15, no. 4, 29 November 2007 (2007-11-29), pages 239 - 246, XP002504133, ISSN: 0969-7128 * |
Cited By (3)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US11419950B2 (en) * | 2012-10-26 | 2022-08-23 | Vrije Universiteit Brussel | Vectors for liver-directed gene therapy of hemophilia and methods and use thereof |
WO2018178067A1 (en) | 2017-03-27 | 2018-10-04 | Vrije Universiteit Brussel | Diaphragm-specific nucleic acid regulatory elements and methods and use thereof |
WO2023006890A1 (en) | 2021-07-28 | 2023-02-02 | Vrije Universiteit Brussel | Improving the efficacy of muscle-targeted gene therapy |
Similar Documents
Publication | Publication Date | Title |
---|---|---|
AU2020200041B2 (en) | Capsid-modified, rAAV3 vector compositions and uses in gene therapy of human liver cancer | |
US20240226203A9 (en) | Compositions and methods of treating huntington's disease | |
US11344608B2 (en) | Factor IX gene therapy | |
KR102373765B1 (en) | Capsid-free aav vectors, compositions, and methods for vector production and gene delivery | |
EP3393522B1 (en) | Improved hybrid dual recombinant aav vector systems for gene therapy | |
US11999965B2 (en) | Bocaparvovirus small noncoding RNA and uses thereof | |
TW201706412A (en) | Fabry disease gene therapy | |
KR20160026841A (en) | Vectors comprising stuffer/filler polynucleotide sequences and methods of use | |
CN111718947B (en) | Adeno-associated virus vector for treating type IIIA or IIIB mucopolysaccharidosis and use thereof | |
He et al. | AAV for gene therapy in ocular diseases: progress and prospects | |
WO2021113634A1 (en) | Transgene cassettes designed to express a human mecp2 gene | |
US11077208B2 (en) | Wilson's disease gene therapy | |
WO2009071679A1 (en) | Novel aav vector and uses thereof | |
CN117377771A (en) | carrier system | |
EP4410988A1 (en) | An aav2-vector variant for targeted transfer of genes | |
US20240076691A1 (en) | Codon-optimized nucleic acid encoding the fix protein | |
WO2024255792A1 (en) | Human mecp2 promoter and use thereof | |
US20130317093A1 (en) | Methods of treating alzheimer's disease with apo a-1 milano | |
CA3220037A1 (en) | Recombinant adeno-associated viruses for lesch-nyhan disorders and uses thereof | |
CN119143858A (en) | KCC2 truncated body or coding sequence thereof and application thereof | |
OA21369A (en) | Codon-optimized nucleic acid encoding the fix protein. | |
WO2024059823A1 (en) | Self-complementary AAV vectors carrying dominant negative RhoA and methods of use to treat ocular diseases | |
CN118019757A (en) | KCNV2 Gene therapy | |
US20160237141A1 (en) | Methods of treating alzheimer's disease with apo a-1 milano | |
BR112017027956B1 (en) | RECOMBINANT ADENOASSOCIATED VIRUS VECTORS, THEIR USE AND PRODUCTION METHOD, AND PHARMACEUTICAL COMPOSITION |
Legal Events
Date | Code | Title | Description |
---|---|---|---|
121 | Ep: the epo has been informed by wipo that ep was designated in this application |
Ref document number: 08856017 Country of ref document: EP Kind code of ref document: A1 |
|
NENP | Non-entry into the national phase |
Ref country code: DE |
|
122 | Ep: pct application non-entry in european phase |
Ref document number: 08856017 Country of ref document: EP Kind code of ref document: A1 |