WO2024186275A1 - Novel compositions of adeno-associated viruses engineered for enhanced tissue transduction and specificity - Google Patents
Novel compositions of adeno-associated viruses engineered for enhanced tissue transduction and specificity Download PDFInfo
- Publication number
- WO2024186275A1 WO2024186275A1 PCT/SG2024/050144 SG2024050144W WO2024186275A1 WO 2024186275 A1 WO2024186275 A1 WO 2024186275A1 SG 2024050144 W SG2024050144 W SG 2024050144W WO 2024186275 A1 WO2024186275 A1 WO 2024186275A1
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- modified
- aav
- sequence
- mer peptide
- seq
- Prior art date
Links
- 241000702421 Dependoparvovirus Species 0.000 title claims abstract description 39
- 239000000203 mixture Substances 0.000 title claims abstract description 10
- 238000010361 transduction Methods 0.000 title abstract description 11
- 230000026683 transduction Effects 0.000 title abstract description 11
- 108090000765 processed proteins & peptides Proteins 0.000 claims abstract description 126
- 239000002773 nucleotide Substances 0.000 claims abstract description 83
- 125000003729 nucleotide group Chemical group 0.000 claims abstract description 83
- 238000004519 manufacturing process Methods 0.000 claims abstract description 8
- 230000010415 tropism Effects 0.000 claims description 57
- 210000004027 cell Anatomy 0.000 claims description 40
- 238000003780 insertion Methods 0.000 claims description 37
- 230000037431 insertion Effects 0.000 claims description 37
- 210000004185 liver Anatomy 0.000 claims description 36
- 210000004556 brain Anatomy 0.000 claims description 23
- 210000003205 muscle Anatomy 0.000 claims description 20
- 230000001225 therapeutic effect Effects 0.000 claims description 8
- 150000007523 nucleic acids Chemical class 0.000 claims description 7
- 210000002889 endothelial cell Anatomy 0.000 claims description 6
- 230000007423 decrease Effects 0.000 claims description 5
- 102000039446 nucleic acids Human genes 0.000 claims description 5
- 108020004707 nucleic acids Proteins 0.000 claims description 5
- 210000001130 astrocyte Anatomy 0.000 claims description 3
- 210000003494 hepatocyte Anatomy 0.000 claims description 3
- 210000002569 neuron Anatomy 0.000 claims description 3
- 239000000546 pharmaceutical excipient Substances 0.000 claims description 3
- 239000000427 antigen Substances 0.000 claims description 2
- 108091007433 antigens Proteins 0.000 claims description 2
- 102000036639 antigens Human genes 0.000 claims description 2
- 210000003289 regulatory T cell Anatomy 0.000 claims description 2
- 210000000234 capsid Anatomy 0.000 abstract description 27
- 241000700605 Viruses Species 0.000 description 73
- 208000002267 Anti-neutrophil cytoplasmic antibody-associated vasculitis Diseases 0.000 description 38
- 210000001519 tissue Anatomy 0.000 description 37
- 210000000056 organ Anatomy 0.000 description 26
- 150000001413 amino acids Chemical group 0.000 description 24
- 102000004196 processed proteins & peptides Human genes 0.000 description 24
- 235000001014 amino acid Nutrition 0.000 description 21
- 229940024606 amino acid Drugs 0.000 description 21
- 229920001184 polypeptide Polymers 0.000 description 17
- 108091033319 polynucleotide Proteins 0.000 description 13
- 102000040430 polynucleotide Human genes 0.000 description 13
- 239000002157 polynucleotide Substances 0.000 description 13
- 239000013612 plasmid Substances 0.000 description 11
- 108020004705 Codon Proteins 0.000 description 9
- 230000004048 modification Effects 0.000 description 9
- 238000012986 modification Methods 0.000 description 9
- 108090000623 proteins and genes Proteins 0.000 description 9
- 230000008685 targeting Effects 0.000 description 9
- 230000003612 virological effect Effects 0.000 description 9
- 210000003169 central nervous system Anatomy 0.000 description 8
- 241001465754 Metazoa Species 0.000 description 7
- UYTPUPDQBNUYGX-UHFFFAOYSA-N guanine Chemical compound O=C1NC(N)=NC2=C1N=CN2 UYTPUPDQBNUYGX-UHFFFAOYSA-N 0.000 description 6
- 230000035772 mutation Effects 0.000 description 6
- 239000000523 sample Substances 0.000 description 6
- RWQNBRDOKXIBIV-UHFFFAOYSA-N thymine Chemical compound CC1=CNC(=O)NC1=O RWQNBRDOKXIBIV-UHFFFAOYSA-N 0.000 description 6
- 108020004414 DNA Proteins 0.000 description 5
- 108091028043 Nucleic acid sequence Proteins 0.000 description 5
- 230000008859 change Effects 0.000 description 5
- 238000000034 method Methods 0.000 description 5
- 238000003752 polymerase chain reaction Methods 0.000 description 5
- 238000002560 therapeutic procedure Methods 0.000 description 5
- 108010067902 Peptide Library Proteins 0.000 description 4
- 230000004075 alteration Effects 0.000 description 4
- 239000012634 fragment Substances 0.000 description 4
- 230000028993 immune response Effects 0.000 description 4
- 238000002703 mutagenesis Methods 0.000 description 4
- 231100000350 mutagenesis Toxicity 0.000 description 4
- 230000009467 reduction Effects 0.000 description 4
- 210000002027 skeletal muscle Anatomy 0.000 description 4
- 108091093088 Amplicon Proteins 0.000 description 3
- 101710132601 Capsid protein Proteins 0.000 description 3
- 101710197658 Capsid protein VP1 Proteins 0.000 description 3
- ROHFNLRQFUQHCH-YFKPBYRVSA-N L-leucine Chemical compound CC(C)C[C@H](N)C(O)=O ROHFNLRQFUQHCH-YFKPBYRVSA-N 0.000 description 3
- 101710084218 Master replication protein Proteins 0.000 description 3
- 101710112078 Para-Rep C2 Proteins 0.000 description 3
- 101710118046 RNA-directed RNA polymerase Proteins 0.000 description 3
- 101710108545 Viral protein 1 Proteins 0.000 description 3
- 238000004458 analytical method Methods 0.000 description 3
- 230000014509 gene expression Effects 0.000 description 3
- 208000015181 infectious disease Diseases 0.000 description 3
- 108020004999 messenger RNA Proteins 0.000 description 3
- 238000010606 normalization Methods 0.000 description 3
- 235000018102 proteins Nutrition 0.000 description 3
- 102000004169 proteins and genes Human genes 0.000 description 3
- 229940113082 thymine Drugs 0.000 description 3
- 231100000419 toxicity Toxicity 0.000 description 3
- 230000001988 toxicity Effects 0.000 description 3
- 108091032973 (ribonucleotides)n+m Proteins 0.000 description 2
- 238000011740 C57BL/6 mouse Methods 0.000 description 2
- 238000002965 ELISA Methods 0.000 description 2
- WHUUTDBJXJRKMK-UHFFFAOYSA-N Glutamic acid Natural products OC(=O)C(N)CCC(O)=O WHUUTDBJXJRKMK-UHFFFAOYSA-N 0.000 description 2
- DHMQDGOQFOQNFH-UHFFFAOYSA-N Glycine Chemical compound NCC(O)=O DHMQDGOQFOQNFH-UHFFFAOYSA-N 0.000 description 2
- QNAYBMKLOCPYGJ-REOHCLBHSA-N L-alanine Chemical compound C[C@H](N)C(O)=O QNAYBMKLOCPYGJ-REOHCLBHSA-N 0.000 description 2
- DCXYFEDJOCDNAF-REOHCLBHSA-N L-asparagine Chemical compound OC(=O)[C@@H](N)CC(N)=O DCXYFEDJOCDNAF-REOHCLBHSA-N 0.000 description 2
- CKLJMWTZIZZHCS-REOHCLBHSA-N L-aspartic acid Chemical compound OC(=O)[C@@H](N)CC(O)=O CKLJMWTZIZZHCS-REOHCLBHSA-N 0.000 description 2
- COLNVLDHVKWLRT-QMMMGPOBSA-N L-phenylalanine Chemical compound OC(=O)[C@@H](N)CC1=CC=CC=C1 COLNVLDHVKWLRT-QMMMGPOBSA-N 0.000 description 2
- OUYCCCASQSFEME-QMMMGPOBSA-N L-tyrosine Chemical compound OC(=O)[C@@H](N)CC1=CC=C(O)C=C1 OUYCCCASQSFEME-QMMMGPOBSA-N 0.000 description 2
- ROHFNLRQFUQHCH-UHFFFAOYSA-N Leucine Natural products CC(C)CC(N)C(O)=O ROHFNLRQFUQHCH-UHFFFAOYSA-N 0.000 description 2
- KDXKERNSBIXSRK-UHFFFAOYSA-N Lysine Natural products NCCCCC(N)C(O)=O KDXKERNSBIXSRK-UHFFFAOYSA-N 0.000 description 2
- 241000699666 Mus <mouse, genus> Species 0.000 description 2
- FAPWRFPIFSIZLT-UHFFFAOYSA-M Sodium chloride Chemical compound [Na+].[Cl-] FAPWRFPIFSIZLT-UHFFFAOYSA-M 0.000 description 2
- 101150024766 VP1 gene Proteins 0.000 description 2
- 101150036700 VP3 gene Proteins 0.000 description 2
- 235000004279 alanine Nutrition 0.000 description 2
- 238000003556 assay Methods 0.000 description 2
- 238000010276 construction Methods 0.000 description 2
- OPTASPLRGRRNAP-UHFFFAOYSA-N cytosine Chemical compound NC=1C=CNC(=O)N=1 OPTASPLRGRRNAP-UHFFFAOYSA-N 0.000 description 2
- 230000003247 decreasing effect Effects 0.000 description 2
- 238000012217 deletion Methods 0.000 description 2
- 230000037430 deletion Effects 0.000 description 2
- 238000010586 diagram Methods 0.000 description 2
- 238000001943 fluorescence-activated cell sorting Methods 0.000 description 2
- 238000001727 in vivo Methods 0.000 description 2
- 230000006698 induction Effects 0.000 description 2
- 238000002347 injection Methods 0.000 description 2
- 239000007924 injection Substances 0.000 description 2
- 125000000468 ketone group Chemical group 0.000 description 2
- 230000009437 off-target effect Effects 0.000 description 2
- 238000004806 packaging method and process Methods 0.000 description 2
- 239000002245 particle Substances 0.000 description 2
- 238000011160 research Methods 0.000 description 2
- 238000012216 screening Methods 0.000 description 2
- 238000006467 substitution reaction Methods 0.000 description 2
- 238000001890 transfection Methods 0.000 description 2
- 229930024421 Adenine Natural products 0.000 description 1
- GFFGJBXGBJISGV-UHFFFAOYSA-N Adenine Chemical compound NC1=NC=NC2=C1N=CN2 GFFGJBXGBJISGV-UHFFFAOYSA-N 0.000 description 1
- 239000004475 Arginine Substances 0.000 description 1
- DCXYFEDJOCDNAF-UHFFFAOYSA-N Asparagine Natural products OC(=O)C(N)CC(N)=O DCXYFEDJOCDNAF-UHFFFAOYSA-N 0.000 description 1
- 241000894006 Bacteria Species 0.000 description 1
- 238000010455 CRISPR delivery Methods 0.000 description 1
- 108090000565 Capsid Proteins Proteins 0.000 description 1
- 102100023321 Ceruloplasmin Human genes 0.000 description 1
- 108091026890 Coding region Proteins 0.000 description 1
- 241000701022 Cytomegalovirus Species 0.000 description 1
- 102100034343 Integrase Human genes 0.000 description 1
- AGPKZVBTJJNPAG-WHFBIAKZSA-N L-isoleucine Chemical compound CC[C@H](C)[C@H](N)C(O)=O AGPKZVBTJJNPAG-WHFBIAKZSA-N 0.000 description 1
- FFEARJCKVFRZRR-BYPYZUCNSA-N L-methionine Chemical compound CSCC[C@H](N)C(O)=O FFEARJCKVFRZRR-BYPYZUCNSA-N 0.000 description 1
- QIVBCDIJIAJPQS-VIFPVBQESA-N L-tryptophane Chemical compound C1=CC=C2C(C[C@H](N)C(O)=O)=CNC2=C1 QIVBCDIJIAJPQS-VIFPVBQESA-N 0.000 description 1
- 239000004472 Lysine Substances 0.000 description 1
- 241000699670 Mus sp. Species 0.000 description 1
- 108091092724 Noncoding DNA Proteins 0.000 description 1
- ONIBWKKTOPOVIA-UHFFFAOYSA-N Proline Natural products OC(=O)C1CCCN1 ONIBWKKTOPOVIA-UHFFFAOYSA-N 0.000 description 1
- 241000125945 Protoparvovirus Species 0.000 description 1
- 108010092799 RNA-directed DNA polymerase Proteins 0.000 description 1
- 108700008625 Reporter Genes Proteins 0.000 description 1
- MTCFGRXMJLQNBG-UHFFFAOYSA-N Serine Natural products OCC(N)C(O)=O MTCFGRXMJLQNBG-UHFFFAOYSA-N 0.000 description 1
- 108010034546 Serratia marcescens nuclease Proteins 0.000 description 1
- 108091081024 Start codon Proteins 0.000 description 1
- AYFVYJQAPQTCCC-UHFFFAOYSA-N Threonine Natural products CC(O)C(N)C(O)=O AYFVYJQAPQTCCC-UHFFFAOYSA-N 0.000 description 1
- 239000004473 Threonine Substances 0.000 description 1
- 239000007983 Tris buffer Substances 0.000 description 1
- QIVBCDIJIAJPQS-UHFFFAOYSA-N Tryptophan Natural products C1=CC=C2C(CC(N)C(O)=O)=CNC2=C1 QIVBCDIJIAJPQS-UHFFFAOYSA-N 0.000 description 1
- 101150093578 VP2 gene Proteins 0.000 description 1
- 239000002253 acid Substances 0.000 description 1
- 229960000643 adenine Drugs 0.000 description 1
- ODKSFYDXXFIFQN-UHFFFAOYSA-N arginine Natural products OC(=O)C(N)CCCNC(N)=N ODKSFYDXXFIFQN-UHFFFAOYSA-N 0.000 description 1
- 235000009582 asparagine Nutrition 0.000 description 1
- 229960001230 asparagine Drugs 0.000 description 1
- 235000003704 aspartic acid Nutrition 0.000 description 1
- 230000003416 augmentation Effects 0.000 description 1
- 239000011324 bead Substances 0.000 description 1
- 230000008901 benefit Effects 0.000 description 1
- OQFSQFPPLPISGP-UHFFFAOYSA-N beta-carboxyaspartic acid Natural products OC(=O)C(N)C(C(O)=O)C(O)=O OQFSQFPPLPISGP-UHFFFAOYSA-N 0.000 description 1
- 230000033228 biological regulation Effects 0.000 description 1
- 230000037396 body weight Effects 0.000 description 1
- 238000010804 cDNA synthesis Methods 0.000 description 1
- 238000004364 calculation method Methods 0.000 description 1
- 230000015556 catabolic process Effects 0.000 description 1
- 239000006285 cell suspension Substances 0.000 description 1
- 238000010367 cloning Methods 0.000 description 1
- 230000000052 comparative effect Effects 0.000 description 1
- 239000002299 complementary DNA Substances 0.000 description 1
- 239000007859 condensation product Substances 0.000 description 1
- 210000004748 cultured cell Anatomy 0.000 description 1
- 235000018417 cysteine Nutrition 0.000 description 1
- XUJNEKJLAYXESH-UHFFFAOYSA-N cysteine Natural products SCC(N)C(O)=O XUJNEKJLAYXESH-UHFFFAOYSA-N 0.000 description 1
- 229940104302 cytosine Drugs 0.000 description 1
- 238000006731 degradation reaction Methods 0.000 description 1
- 239000003814 drug Substances 0.000 description 1
- 230000000694 effects Effects 0.000 description 1
- 238000000605 extraction Methods 0.000 description 1
- 210000001508 eye Anatomy 0.000 description 1
- 230000037433 frameshift Effects 0.000 description 1
- 230000008014 freezing Effects 0.000 description 1
- 238000007710 freezing Methods 0.000 description 1
- 238000001415 gene therapy Methods 0.000 description 1
- 235000013922 glutamic acid Nutrition 0.000 description 1
- 239000004220 glutamic acid Substances 0.000 description 1
- ZDXPYRJPNDTMRX-UHFFFAOYSA-N glutamine Natural products OC(=O)C(N)CCC(N)=O ZDXPYRJPNDTMRX-UHFFFAOYSA-N 0.000 description 1
- 239000001963 growth medium Substances 0.000 description 1
- 210000002216 heart Anatomy 0.000 description 1
- 238000012165 high-throughput sequencing Methods 0.000 description 1
- HNDVDQJCIGZPNO-UHFFFAOYSA-N histidine Natural products OC(=O)C(N)CC1=CN=CN1 HNDVDQJCIGZPNO-UHFFFAOYSA-N 0.000 description 1
- 230000005745 host immune response Effects 0.000 description 1
- 230000006872 improvement Effects 0.000 description 1
- 238000011534 incubation Methods 0.000 description 1
- 230000002458 infectious effect Effects 0.000 description 1
- 238000001802 infusion Methods 0.000 description 1
- 210000000936 intestine Anatomy 0.000 description 1
- AGPKZVBTJJNPAG-UHFFFAOYSA-N isoleucine Natural products CCC(C)C(N)C(O)=O AGPKZVBTJJNPAG-UHFFFAOYSA-N 0.000 description 1
- 229960000310 isoleucine Drugs 0.000 description 1
- 210000003734 kidney Anatomy 0.000 description 1
- 210000004072 lung Anatomy 0.000 description 1
- 239000006166 lysate Substances 0.000 description 1
- 239000012139 lysis buffer Substances 0.000 description 1
- 239000000463 material Substances 0.000 description 1
- 239000002609 medium Substances 0.000 description 1
- 229930182817 methionine Natural products 0.000 description 1
- 230000000956 myotropic effect Effects 0.000 description 1
- 238000007481 next generation sequencing Methods 0.000 description 1
- 210000000496 pancreas Anatomy 0.000 description 1
- 230000001717 pathogenic effect Effects 0.000 description 1
- 239000008194 pharmaceutical composition Substances 0.000 description 1
- COLNVLDHVKWLRT-UHFFFAOYSA-N phenylalanine Natural products OC(=O)C(N)CC1=CC=CC=C1 COLNVLDHVKWLRT-UHFFFAOYSA-N 0.000 description 1
- 229920001983 poloxamer Polymers 0.000 description 1
- 230000008569 process Effects 0.000 description 1
- 238000000746 purification Methods 0.000 description 1
- 238000011002 quantification Methods 0.000 description 1
- 238000004445 quantitative analysis Methods 0.000 description 1
- 230000010076 replication Effects 0.000 description 1
- 238000012163 sequencing technique Methods 0.000 description 1
- 238000002741 site-directed mutagenesis Methods 0.000 description 1
- 239000011780 sodium chloride Substances 0.000 description 1
- 241000894007 species Species 0.000 description 1
- 239000006228 supernatant Substances 0.000 description 1
- 239000000725 suspension Substances 0.000 description 1
- 210000001258 synovial membrane Anatomy 0.000 description 1
- 230000009885 systemic effect Effects 0.000 description 1
- 238000010257 thawing Methods 0.000 description 1
- LENZDBCJOHFCAS-UHFFFAOYSA-N tris Chemical compound OCC(N)(CO)CO LENZDBCJOHFCAS-UHFFFAOYSA-N 0.000 description 1
- OUYCCCASQSFEME-UHFFFAOYSA-N tyrosine Natural products OC(=O)C(N)CC1=CC=C(O)C=C1 OUYCCCASQSFEME-UHFFFAOYSA-N 0.000 description 1
- 210000003462 vein Anatomy 0.000 description 1
- 210000005253 yeast cell Anatomy 0.000 description 1
Classifications
-
- 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
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K35/00—Medicinal preparations containing materials or reaction products thereof with undetermined constitution
- A61K35/66—Microorganisms or materials therefrom
- A61K35/76—Viruses; Subviral particles; Bacteriophages
- A61K35/761—Adenovirus
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N15/00—Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
- C12N15/09—Recombinant DNA-technology
- C12N15/63—Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
- C12N15/79—Vectors or expression systems specially adapted for eukaryotic hosts
- C12N15/85—Vectors or expression systems specially adapted for eukaryotic hosts for animal cells
- C12N15/86—Viral vectors
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- 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
-
- 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
- C12N2750/14145—Special targeting system for viral vectors
Definitions
- the invention is in the field of biotechnology.
- the invention relates to production of engineered adeno-associated viruses More specifically, the invention provides the use of engineered adeno-associated viruses for enhanced tissue transduction and specificity and compositions thereof.
- Adeno-associated virus (AAV) vector-based therapeutics are rapidly advancing to the clinic.
- Systemic vector infusion represents the ideal delivery route in many gene therapy applications, especially when local administration is not feasible
- AAV capsids are based on naturally occurring serotypes, which are sequestrated in the liver and have reduced therapeutic efficacy in organs such as in the central nervous system (CNS) or in skeletal muscles. Consequently, higher vector doses are required, resulting in off-target toxicities.
- CNS central nervous system
- AAV capsids with organ-specific tropism at low vector doses.
- a modified adeno-associated virus serotype DJ comprising a nucleotide sequence encoding a 7-mer peptide inserted between the 1767th and 1768th nucleotides of SEQ ID NO: 399, wherein the modified AAV-DJ has a range of at least 50% to 60%, 60% to 65%, 65% to 70%, 70% to 75%, 75% to 80%, 80% to 85%, 85% to 90%, 90% to 95%, or 95% to 99% sequence identity with the sequence of SEQ ID NO: 399 outside the nucleotide sequence encoding the 7-mer peptide
- a modified adeno-associated virus serotype 2 comprising a nucleotide sequence encoding a 7-mer peptide inserted between the 1764th and 1765th nucleotides of SEQ ID NO: 401, wherein the modified AAV2 has a range of at least 50% to 60%, 60% to 65%, 65% to 70%, 70% to 75%, 75% to 80%, 80% to 85%, 85% to 90%, 90% to 95%, or 95% to 99% sequence identity with the sequence of SEQ ID NO:
- a modified adeno-associated virus serotype 5 comprising a nucleotide sequence encoding a 7-mer peptide inserted between the 1722th and 1723th nucleotides of SEQ ID NO: 402, wherein the modified AAV5 has a range of at least 50% to 60%, 60% to 65%, 65% to 70%, 70% to 75%, 75% to 80%, 80% to 85%, 85% to 90%, 90% to 95%, or 95% to 99% sequence identity with the sequence of SEQ ID NO:
- a modified adeno-associated virus serotype 6 comprising a nucleotide sequence encoding a 7-mer peptide inserted between the 1770th and 1771th nucleotides of SEQ ID NO: 403, wherein the modified AAV6 has a range of at least 50% to 60%, 60% to 65%, 65% to 70%, 70% to 75%, 75% to 80%, 80% to 85%, 85% to 90%, 90% to 95%, or 95% to 99% sequence identity with the sequence of SEQ ID NO:
- a modified adeno-associated virus serotype 6 comprising a nucleotide sequence encoding a 7-mer peptide inserted between the 1764th and 1765th nucleotides of SEQ ID NO: 403, wherein the modified AAV6 has a range of at least 50% to 60%, 60% to 65%, 65% to 70%, 70% to 75%, 75% to 80%, 80% to 85%, 85% to 90%, 90% to 95%, or 95% to 99% sequence identity with the sequence of SEQ ID NO:
- a modified adeno-associated virus serotype 9 comprising a nucleotide sequence encoding a 7-mer peptide inserted between the 1764th and 1765th nucleotides of SEQ ID NO: 404, wherein the modified AAV9 has a range of at least 50% to 60%, 60% to 65%, 65% to 70%, 70% to 75%, 75% to 80%, 80% to 85%, 85% to 90%, 90% to 95%, or 95% to 99% sequence identity with the sequence of SEQ ID NO:
- composition comprising any modified AAV as described herein, and a pharmaceutically acceptable excipient.
- nucleic acid encoding any modified AAV as described herein.
- AAV or “adeno-associated virus” refers to a non-pathogenic, small viruses of the Parvovirus family which infect humans.
- AAV includes, but is not limited to, naturally occurring and engineered AAV subtypes or serotypes, for example, AAV type 1 (AAV-1), AAV type 2 (AAV-2), AAV type 3 (AAV-3), AAV type 4 (AAV-4), AAV type 5 (AAV-5), AAV type 6 (AAV-6), AAV type 7 (AAV-7), AAV type 8 (AAV-8) and AAV- DJ
- capsid refers to the protein shell of a virus particle.
- the capsid of a virus is composed of one or more capsid units or subunits.
- the capsid of an AAV is composed of VP1, VP2 and/or VP3 subunits.
- the capsid of a virus may have several functional purposes.
- the capsid of a virus may protect the viral genome from degradation, assist the virus in evading host immune response, and/or determine viral tropism to different cell or tissue types.
- the term “identity” in the context of a polynucleotide or polypeptide sequence refers to the overall relatedness or homology between polymeric molecules, e.g., between polynucleotide molecules (e g., DNA molecules and/or RNA molecules) and/or between polypeptide molecules. Calculation of the percent identity of two polynucleotide sequences, for example, can be performed by aligning the two sequences for optimal comparison purposes (e g , gaps can be introduced in one or both of a first and a second nucleic acid sequences for optimal alignment and non-identical sequences can be disregarded for comparison purposes) The nucleotides at corresponding nucleotide positions are then compared.
- the percent identity between the two sequences is a function of the number of identical positions shared by the sequences, taking into account the number of gaps, and the length of each gap, which needs to be introduced for optimal alignment of the two sequences.
- the comparison of sequences and determination of percent identity between two sequences can be accomplished using a mathematical algorithm.
- the term “library” refers to a mixture of heterogeneous polypeptides or polynucleotides.
- a library is composed of members that have similar polypeptide or polynucleotide sequences. Sequence differences between library members are responsible for the diversity of the library.
- the library can take the form of a mixture of polypeptides or polynucleotides, or can be in the form of organisms or cells, for example yeast cells and the like, that are transformed with a library of polynucleotides.
- modified virus and “virus variant” are interchangeable terms referring to an engineered or recombinant virus comprising an alteration, that is, “modification”, in the sequence of a polypeptide or a polynucleotide encoding a polypeptide compared to that of a reference sequence.
- the reference sequence may be a genomic sequence or a subsection thereof of a virus, or the amino acid sequence encoded by the genomic sequence or a subsection thereof of a virus.
- the reference sequence may be a sequence from a wild-type or native virus, or it may be the sequence from a different modified virus.
- a wild-type virus refers to a virus with phenotype or genotype that predominates in nature.
- Modified AAV or “variant AAV” refers to an AAV that has a change in its genomic sequence or a subsection thereof, or in the polypeptide encoded by its genomic sequence or a subsection thereof, compared to a reference AAV.
- the alteration or modification of a sequence may refer to an insertion, deletion, or substitution of one or more residues in the sequence.
- a modified AAV may be an AAV that has one or more nucleotides inserted into its genomic sequence or a subsection thereof compared to a reference AAV or has one or more amino acids inserted into the polypeptide encoded by its genomic sequence or a subsection thereof.
- peptide refers to the condensation product of two or more amino acids, wherein the length of the polypeptide is less than or equal to 50 amino acids long, e.g., 5, 6, 7, 8, 9, 10, 11-15, 15-20, 20-25, 25-30, 30-35, 35-40, 40-45 or 45-50 amino acids long.
- targeting and “tropism” are interchangeable terms that refer to the ability of a virus to infect different cell types to produce a successful infection.
- a successful infection may be defined as when the virus gains entry to the host cell, when the virus has begun replication of its genome in the host cell, when intact progeny virus is detectable within the host cell, when the virus progeny exits the host cell, or any combination thereof.
- a virus with tropism to the CNS or a CNS-targeting virus indicates that the virus is capable of infecting cells that are from or derived from the CNS and/or that the virus is capable of infecting cells that are from or derived from the CNS at a higher specificity than other cell types.
- a virus with tropism to the muscle or a muscle-targeting virus refers to a virus that is capable of successfully infecting cells that are from or derived from skeletal muscle and/or that the virus is capable of infecting cells that are from or derived from muscle at a higher specificity than other cell types.
- a virus that does not have tropism or has reduced tropism to a cell type would not be capable or have a reduced capability of infecting said cell type.
- a virus with reduced liver tropism or that is liver de-targeting indicates that the virus is less capable or unable to successfully infect cells that are from or derived from the liver. It would be understood by those skilled in the art that “tropism” may also mean “specificity to”.
- virus tropism to the CNS means that a virus has specificity for the CNS. Therefore, as used herein, the terms “tropism” and “specificity” are interchangeable and mean the ability of a virus to successfully infect a selected cell type. Further, a virus may also be defined as “specific targeting” or having “specific tropism”, to indicate that it is specific for a selected tissue type. For example, a virus with specific tropism to the brain is one that specifically infects the brain.
- FIG. 1 shows a schematic map of the construction of the AAV-DJ library.
- the top diagram shows the AAV-DJ library plasmid, where the grey lines represent the insertion site of the peptide library (between N589 and R590), and the bottom diagram shows the full-length Rep2 plasmid.
- ITR inverted terminal repeat.
- Figure 2 shows the next-gen sequencing (NGS)-driven evolution of the AAV-DJ peptide library.
- NGS next-gen sequencing
- Figures 2A, 2B and 2C show distribution of the length of the inserted sequences in the AAV-DJ capsid DNA sequenced from the liver, brain, and muscle respectively.
- the X-axis represents the length of the nucleotide sequence inserted, and the Y-axis represents the probability density of each length of nucleotide sequences.
- Figure 3 shows a table of values of the fold-change difference detected by NGS of selected AAV-DJ variants normalized to wild-type AAV-DJ, leading to identification of AAV- DJ variants with enhanced brain, muscle transduction efficiency with complete liver detargeting at low vector doses.
- the labels on the left denote the AAV-DJ variants, corresponding to the peptides listed in Table 3.
- the values in the table represent the fold-change of each variant in the liver, brain, and muscle, compared to wild-type AAV-DJ.
- the scale bar on the right represents the fold-change of the modified AAV genome sequenced from the respective organ versus the wild-type.
- the present disclosure provides for modified AAV s with altered tropism to various cell types, tissue types and/or organs compared to a reference AAV.
- the reference AAV may be a wild-type AAV or unmodified AAV, and it may be of the same or different serotype as the modified AAV.
- a modified virus or virus variant may be modified so that its tropism or targeting to one or more selected cell types, tissues and/or organs is altered.
- a wild-type virus may be modified to possess enhanced tropism, reduced tropism, or no change in tropism towards one or more selected organs, tissues and/or cell types.
- the reduction in tropism is to the extent that tropism is abolished, that is, the virus is no longer capable of infecting the one or more selected organs, tissues and/or cell types.
- the organ may be any organ from a human or an animal.
- the organ may be the CNS, ear, eye, heart, intestine, kidney, joint/synovium, lung, liver, pancreas, or skeletal muscle.
- the tissues may be primary or clinical isolates from an organ, or it may be a cultured tissue originating from an organ.
- the cells may be primary or clinical isolates from an organ or tissue, or it may be a cultured cells originating from an organ or tissue.
- An AAV with altered tropism may be obtained for example by introducing one or more mutations into the viral genome or sections thereof, or within a specific gene.
- an AAV may be modified by introducing mutations into one or more genes encoding the capsid protein. Mutations may be introduced by any method known by those skilled in the art, for example, site-directed mutagenesis or site-saturation mutagenesis, and may include insertion or deletion of nucleotides (frame-shift mutations), replacement/substitution of nucleotides, or combinations thereof.
- site-saturation mutagenesis may be used to substitute a selected site in a polypeptide with all possible amino acids through the use of degenerate codons in the nucleotide sequence encoding the polypeptide.
- the degenerate codon is NNK, where K stands for keto nucleotides such as guanine and thymine, and N is for any nucleotide.
- K stands for keto nucleotides such as guanine and thymine
- N is for any nucleotide.
- a modification of the polynucleotide sequence encoding a gene may result in the modification of the polypeptide encoded by the polynucleotide sequence.
- mutations in the context of the present invention may refer to mutations of a viral polynucleotide and/or viral polypeptide sequence.
- a wild-type or reference AAV is modified by insertion of a polynucleotide sequence encoding a peptide to a desired site in the nucleotide sequence of the reference or wild-type AAV.
- the reference or wild-type AAV may include but is not limited to the serotypes AAV-2, AAV-5, AAV-6, AAV-9, and AAV-DJ (Table 4).
- modified AAV will have a nucleotide sequence identical to that of the reference or wild-type AAV (i.e., the unmodified AAV) apart from, that is, “outside”, the nucleotide sequence encoding the inserted peptide
- the desired site at which the polynucleotide sequence is inserted may be located within a coding or non-coding region of the viral genome.
- the wild-type or reference AAV may be modified by insertion of one or more amino acids or a peptide into a polypeptide sequence that encodes for a protein or proteins forming the capsid or part of the virus.
- the peptide that is inserted to alter tropism of a wild-type or reference virus may vary in length.
- peptides may be 5-15 amino acids in length.
- the peptide may be 5, 6, 7, 8, 9, 10, 11 , 12, 13, 14 or 15 amino acids in length.
- the inserted peptide may be a 5-mer, 6-mer, 7-mer, 8-mer, 9-mer or 10-mer peptide.
- the peptide is a 7-mer peptide.
- the amino acid sequence of the 7-mer peptide may comprise any permutation of 7 amino acids.
- the amino acids are the 20 standard amino acids. Therefore, provided herein are modified AAVs comprising a 7-mer peptide sequence, wherein the peptide sequence is any permutation of the 20 standard amino acids. It would be understood by those skilled in the art which amino acids are the 20 standard amino acids.
- peptides listed in the present disclosure are non-limiting examples of 7-mer peptides with the property of conferring altered tropism to an AAV when inserted to the viral capsid.
- modified AAVs comprising a nucleotide sequence encoding a 7- mer peptide inserted between two nucleotides of a reference or wild-type AAV capsid gene.
- the two nucleotides may be within the sequence of a VP1, VP2 and/or VP3 gene. In one example, the two nucleotides are within the sequence of a VP3 gene. It would be understood by those skilled in the art that the VP1 gene encodes for the full-length capsid, while VP2 and VP3 genes encode for subunits of the capsid. Those skilled in the art will also recognize that the nucleotide sequences of the VP2 and VP3 genes are subsumed within the VP1 nucleotide sequence.
- a modified adeno-associated virus serotype DJ comprising a nucleotide sequence encoding a 7-mer peptide inserted between the 1767th and 1768th nucleotides of SEQ ID NO: 399, wherein the modified AAV-DJ has a range of at least 50% to 60%, 60% to 65%, 65% to 70%, 70% to 75%, 75% to 80%, 80% to 85%, 85% to 90%, 90% to 95%, or 95% to 99% sequence identity with the sequence of SEQ ID NO: 399 outside the nucleotide sequence encoding the 7-mer peptide.
- AAV- DJ modified adeno-associated virus serotype DJ
- a modified adeno-associated virus serotype 2 comprising a nucleotide sequence encoding a 7-mer peptide inserted between the 1764th and 1765th nucleotides of SEQ ID NO: 401, wherein the modified AAV2 has a range of at least 50% to 60%, 60% to 65%, 65% to 70%, 70% to 75%, 75% to 80%, 80% to 85%, 85% to 90%, 90% to 95%, or 95% to 99% sequence identity with the sequence of SEQ ID NO:
- a modified adeno-associated virus serotype 5 comprising a nucleotide sequence encoding a 7-mer peptide inserted between the 1722th and 1723th nucleotides of SEQ ID NO: 402, wherein the modified AAV5 has a range of at least 50% to 60%, 60% to 65%, 65% to 70%, 70% to 75%, 75% to 80%, 80% to 85%, 85% to 90%, 90% to 95%, or 95% to 99% sequence identity with the sequence of SEQ ID NO:
- a modified adeno-associated virus serotype 6 comprising a nucleotide sequence encoding a 7-mer peptide inserted between the 1770th and 1771th nucleotides of SEQ ID NO: 403, wherein the modified AAV6 has a range of at least 50% to 60%, 60% to 65%, 65% to 70%, 70% to 75%, 75% to 80%, 80% to 85%, 85% to 90%, 90% to 95%, or 95% to 99% sequence identity with the sequence of SEQ ID NO:
- a modified adeno-associated virus serotype 6 comprising a nucleotide sequence encoding a 7-mer peptide inserted between the 1764th and 1765th nucleotides of SEQ ID NO: 403, wherein the modified AAV6 has a range of at least 50% to 60%, 60% to 65%, 65% to 70%, 70% to 75%, 75% to 80%, 80% to 85%, 85% to 90%, 90% to 95%, or 95% to 99% sequence identity with the sequence of SEQ ID NO:
- a modified adeno-associated virus serotype 9 comprising a nucleotide sequence encoding a 7-mer peptide inserted between the 1764th and 1765th nucleotides of SEQ ID NO: 404, wherein the modified AAV9 has a range of at least 50% to 60%, 60% to 65%, 65% to 70%, 70% to 75%, 75% to 80%, 80% to 85%, 85% to 90%, 90% to 95%, or 95% to 99% sequence identity with the sequence of SEQ ID NO:
- a virus may have at least 50% to 60%, 60% to 65%, 65% to 70%, 70% to 75%, 75% to 80%, 80% to 85%, 85% to 90%, 90% to 95%, or 95% to 99% sequence identity to a reference virus.
- the comparison may be between the AAV of the same or different serotypes.
- an AAV-DJ may have at least 50% sequence identity with a reference AAV-2, AAV-5, AAV-6, AAV-9, or AAV-DJ virus. In one example, an AAV-DJ has at least 57% sequence identity with a reference AAV-5.
- modified AAV2 as described herein, wherein the modified AAV2 has at least 57% sequence identity with the sequence of SEQ ID NO: 401 outside the 7-mer peptide sequence.
- modified AAV5 as described herein, wherein the modified AAV5 has at least 57% sequence identity with the sequence of SEQ ID NO: 402 outside the 7-mer peptide sequence.
- a modified AAV6 comprising a sequence corresponding to a 7-mer peptide inserted between the 1770th and 1771th nucleotides of SEQ ID NO: 403, wherein the modified AAV6 has at least 57% sequence identity with the sequence of SEQ ID NO: 403 outside the 7-mer peptide sequence.
- a modified AAV6 comprising a sequence corresponding to a 7-mer peptide inserted between the 1764th and 1765th nucleotides of SEQ ID NO: 403, wherein the modified AAV 6 has at least 57% sequence identity with the sequence of SEQ ID NO: 403 outside the 7-mer peptide sequence.
- modified AAV9 as described herein, wherein the modified AAV9 has at least 57% sequence identity with the sequence of SEQ ID NO: 404 outside the 7-mer peptide sequence.
- the inserted peptides of the present invention may comprise a sequence as set forth in Table 1 and confers enhanced liver, brain, and muscle tropism to a modified AAV compared to a reference or wild-type AAV.
- the targeting peptide may comprise a sequence as set forth in Table 2 and confers enhanced brain and muscle tropism with reduced liver tropism to a modified AAV compared to a wild-type AAV.
- the targeting peptide may comprise a sequence as set forth in Table 3 and confers enhanced brain and muscle tropism with liver de-targeting to a modified AAV compared to a wild-type AAV
- the modified virus or virus variant of the present invention may have increased or decreased specificity to one or more selected cell types compared to a reference virus.
- the increase or decrease in specificity may refer to the increased ability of the virus to bind to and/or infect said cell type.
- the increase or decrease in specificity of a virus to a cell type may be compared to that of a reference cell type may be measured by quantitative methods known to those skilled in the art.
- the degree of enhancement or reduction in tropism of a modified AAV relative to a reference AAV, such as a wild-type AAV, may be calculated by comparing the infectivity of the variant relative to the infectivity of the reference virus in a selected tissue sample. Those skilled in the art will be aware of the methods of measuring infectivity of viruses.
- infectivity of a virus may be measured by plaque assay, enzyme-linked immunosorbent assay (ELSA), transduction assay using reporter genes, polymerase chain reaction (PCR), high-throughput sequencing or any combination thereof.
- ELSA enzyme-linked immunosorbent assay
- PCR polymerase chain reaction
- NGS next-generation sequencing
- the comparing of the infectivity or tropism of an AAV variant relative to the infectivity of a reference or original AAV, wherein infectivity is measured by NGS may be done by dividing the number of reads of the AAV variant in a selected tissue sample, for example the brain, muscle, or liver, by the number of reads of the reference or original virus in the tissue sample to obtain the degree fold-change in infectivity or tropism of the AAV variant relative to the reference or original virus.
- the AAV variant has fold-change of > 1 relative to the reference or original AAV in a tissue type, it has increased or improved tropism for that tissue type compared to the reference or original virus.
- Table 1 AAV-DJ variants with enhanced brain, muscle, and liver tropism
- the modified AAV comprises a 7-mer peptide sequence selected from the sequences set forth in Table 2, and the insertion increases brain, and muscle tropism, and decreases liver tropism compared to a wild-type AAV without a 7-mer peptide sequence insertion.
- the modified AAV comprises a 7-mer peptide sequence selected from the sequences set forth in Table 3, and wherein the insertion increases brain, and muscle tropism, and is liver de-targeting.
- the modified AAV comprises a 7-mer peptide sequence selected from the group consisting of SEQ ID NOs: 22, 24, 32, 85, 106, 198, 216, 232, 251, 293, 312, 320, 321, 224, 346 or 376.
- the single letter symbol has the following description: A for adenine; C for cytosine; G for guanine; T for thymine; K for keto nucleotides such as guanine and thymine; and N is for any nucleotide (which is not a gap).
- the use of the NNK codon in site-saturation mutagenesis allows for coverage of codons for all 20 standard amino acids, including degenerate codons. “Degenerate codons” refers to codons differing in one or two nucleotides but coding for the same amino acid.
- the amino acid Alanine may be encoded by the codons GCT, GCC, GCA or GCG and the amino acid Leucine may be encoded by the codons CTT, CTC, CTA, CTG, TTA and TTG.
- the single letter symbol has the following description: G (Gly) for Glycine; A (Ala) for Alanine; L (Leu) for Leucine; M (Met) for Methionine; F (Phe) for Phenylalanine; W (Trp) for Tryptophan; K (Lys) for Lysine; Q (Gin) for Glutamine; E (Glu) for Glutamic Acid; S (Ser) for Serine; P (Pro) for Proline, V (Vai) for Valine, I (He) for Isoleucine; C (Cys) for Cysteine; Y (Tyr) for Tyrosine; H (His) for His
- modified AAVs provided herein may possess advantageous characteristics when used for research and/or therapeutic applications as a result of their enhanced tropism to an organ, tissue and/or cell type.
- modified AAVs with enhanced tropism may be used at lower vector doses resulting reduced off-target toxicities.
- Modified AAVs with enhanced tropism may also allow for the regulation of anti-capsid immune responses, and may be used for therapeutic purposes, such as CRISPR delivery and gene augmentation therapy. Such targeting may enhance the specificity of AAV s to the desired cell types and improve the overall therapeutic effect.
- liver sinusoidal endothelial cells (LSECs) and hepatocytes and other cell types in the liver could be targeted, whereas in the case of brain targeting, neurons and astrocytes and other cell types in the brain could be targeted.
- the LSEC targeting can reduce the induction of the immune responses to AAV capsid.
- the modified AAVs provided herein may also possess advantageous characteristics when used for research and/or therapeutic applications as a result of their reduced tropism to an organ, tissue and/or cell type.
- the modified AAVs with reduced tropism could be used in therapy at lower vector doses because they are not sequestrated in the liver, leading to reduced off-target toxicities.
- AAV dosage in therapy may be represented as the amount of vector genomes or viral genomes administered to a subject.
- the dose may be specified as the amount of vector genomes per body weight (vg/kg) or amount of vector genomes per volume (vg/ml or vg/L) or vector genomes per animal (vg/animal).
- the therapeutically effective vector dose may be a range.
- the effective dose per animal may be as low as 1 x 10 9 to as high as 1 x IO 17 vg, depending on the route and site of administration.
- the modified AAV as described herein wherein the 7-mer peptide sequence is selected from the sequences set forth in Table 1 or Table 7, and wherein the insertion increases specificity to cells selected from the group consisting of: liver sinusoidal endothelial cells, or hepatocytes, compared to a wild-type AAV without a 7-mer peptide sequence insertion.
- the modified AAV as described herein wherein the 7-mer peptide sequence is selected from the sequences set forth in Table 1, Table 2 or Table 3, and wherein the insertion increases specificity to cells selected from the group consisting of: neurons, or astrocytes, compared to a wild-type AAV without a 7-mer peptide sequence insertion.
- the modified AAV as described herein wherein the 7-mer peptide sequence is selected from the sequences set forth in Table 7, and wherein the insertion increases production of antigen-specific regulatory T cells compared to a wild-type AAV without a 7-mer peptide sequence insertion.
- insertion of the 7-mer peptide sequence may reduce the induction of immune responses to the modified AAV capsid compared to a wild-type AAV without a 7-mer peptide sequence insertion.
- insertion of the 7-mer peptide sequence may regulate anti-capsid immune response compared to a wild-type AAV without a 7-mer peptide sequence insertion.
- the modified AAV as described herein wherein the 7-mer peptide sequence is selected from the sequences set forth in Table 1, Table 2 or Table 3, and wherein the insertion improves therapeutic effect compared to a wild-type AAV without a 7-mer peptide sequence insertion.
- the improvement in therapeutic effect may be reduction of off-target effects or the reduction of vector dose required for therapy.
- Off-target effects refer to effects in organs or tissues other than in the intended organ or tissue when an AAV is used in therapy.
- composition comprising one or more of the modified AAV described herein.
- the composition is a pharmaceutical composition comprising one or more of the modified AAV described herein and a pharmaceutically acceptable excipient.
- nucleic acid encoding the modified AAV as described herein.
- the modified AAV described herein may be produced using several methods known in the art.
- the modified AAV may be produced via the use of a virus packaging cell, such as the HEK-293T cell line
- a virus packaging cell such as the HEK-293T cell line
- one or more nucleic acid molecules comprising a nucleic acid sequence encoding the virus would be introduced into the packaging cell, which would then produce intact, infectious virus particles.
- the invention illustratively described herein may suitably be practiced in the absence of any element or elements, limitation or limitations, not specifically disclosed herein.
- the terms “comprising”, “including”, “containing”, etc. shall be read expansively and without limitation.
- Example 1 Construction of the NNK-based 7-mer peptide insertion library and vector production.
- AAV-DJ-based capsid libraries were generated by the insertion of random 7-mer peptide between amino acids 589 and 590.
- the capsid gene is placed under the control of a ubiquitous promoter (cytomegalovirus “CMV” promoter) in tandem with Rep2 P40 promoter (starting at nucleotide #1700).
- CMV cytomegalovirus
- Rep2 P40 promoter starting at nucleotide #1700.
- the P40 sequence is required for capsid mRNA expression and splicing during AAV production.
- the sequence is itself flanked by inverted terminal repeats (ITR) so each variant will encapsulate its own capsid sequence ( Figure 1).
- a 453-bp gBlock fragment of the wtAAV-DJ capsid was synthesized and used as a template to introduce the random 7-mer peptide library and overhangs for subcloning.
- PCR was performed for 15 cycles using Neb Q5 polymerase.
- the CMV promoter was amplified by PCR to introduce the Swal restriction site and overhangs.
- the resulting amplicons were purified via gel-extraction.
- the remaining fragments were synthesized as gBlocks.
- the different fragments were then assembled by Gibson assembly following manufacturer’s instructions into the recipient ITR plasmid previously digested with SphI and Mfel.
- the gBlocks were synthesized by IDT.
- Rep2 The full-length and unmodified Rep2 (Rep2-ACap, Figure 1) was provided in trans by a separate plasmid during vector production.
- the diversity of the plasmid library was determined by the number of clones growing from a representative aliquot of transformed chemically competent DH5a bacteria. Library plasmids were harvested and purified using a maxi prep plasmid purification kit.
- Virus production was performed by triple transfection in HEK293T cells using 15 pg of helper plasmid, 10 pg of Rep2-Acap plasmid and 1 pg of library DNA plasmid per T175 flask. 24 hours later, the medium was changed to a fresh culture medium containing 2%FBS. Three days after transfection, cells were collected and resuspended in lysis buffer (Tris HC1 pH
- the 40% fraction was extracted and dialyzed with lx PBS (pH 7.2) with 0.001% pluronic acid, using Amicon® Ultra-15 (100 kDa MWCO) filters. Primers and probes localizing to within the AAV2 ITR were used for the quantification of the viral titer by digital droplet PCR (ddPCR).
- Example 2 in vivo library screening in mice
- the altered tropism of the AAV variants compared to that of the reference AAV, that is, a wild-type AAV was assessed by calculating transduction fold-change of the variant relative to the wild-type virus for a selected tissue type.
- NF normalisation factor
- Transduction fold-change (FC) Normalised AAV variant count /AAV wt count in original virus pool.
- the counts, normalized counts and transduction fold change in each selected tissue for each variant relative to wildtype AAV can be found in Tables 5 and 6. In said tables, a count of indicates that the variant was not detected in that tissue sample.
- the amino acids “N” and “R” at the beginning and end of each sequence respectively denote N589 and R590 of the wild-type AAV-DJ capsid, between which the 7-mer peptide is inserted.
- Example 3 selection of peptides conferring tropism to liver sinusoidal endothelial cells (LSECs)
Landscapes
- Health & Medical Sciences (AREA)
- Life Sciences & Earth Sciences (AREA)
- Chemical & Material Sciences (AREA)
- Virology (AREA)
- General Health & Medical Sciences (AREA)
- Organic Chemistry (AREA)
- Medicinal Chemistry (AREA)
- Gastroenterology & Hepatology (AREA)
- Biochemistry (AREA)
- Animal Behavior & Ethology (AREA)
- Microbiology (AREA)
- Public Health (AREA)
- Veterinary Medicine (AREA)
- Mycology (AREA)
- Epidemiology (AREA)
- Biophysics (AREA)
- Genetics & Genomics (AREA)
- Molecular Biology (AREA)
- Proteomics, Peptides & Aminoacids (AREA)
- Pharmacology & Pharmacy (AREA)
- Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
- Micro-Organisms Or Cultivation Processes Thereof (AREA)
Abstract
The invention relates to the production of engineered adeno-associated viruses. More specifically, the invention provides for modified adeno-associated viruses comprising a nucleotide sequence encoding a 7-mer peptide inserted in the capsid. Further provided is the use of engineered adeno-associated viruses for enhanced tissue transduction and specificity, and compositions thereof.
Description
NOVEL COMPOSITIONS OF ADENO-ASSOCIATED VIRUSES ENGINEERED
FOR ENHANCED TISSUE TRANSDUCTION AND SPECIFICITY
CROSS-REFERENCE TO RELATED APPLICATIONS
[0001] This application claims the benefit of priority of Singapore application No. 10202300637W, filed 9 March 2023, the contents of it being hereby incorporated by reference in its entirety for all purposes.
TECHNICAL FIELD
[0002] The invention is in the field of biotechnology. In particular, the invention relates to production of engineered adeno-associated viruses More specifically, the invention provides the use of engineered adeno-associated viruses for enhanced tissue transduction and specificity and compositions thereof.
BACKGROUND OF THE INVENTION
[0003] Adeno-associated virus (AAV) vector-based therapeutics are rapidly advancing to the clinic. Systemic vector infusion represents the ideal delivery route in many gene therapy applications, especially when local administration is not feasible Currently, most AAV capsids are based on naturally occurring serotypes, which are sequestrated in the liver and have reduced therapeutic efficacy in organs such as in the central nervous system (CNS) or in skeletal muscles. Consequently, higher vector doses are required, resulting in off-target toxicities. Thus, there is a need for AAV capsids with organ-specific tropism at low vector doses.
SUMMARY
[0004] Tn one aspect, provided herein is a modified adeno-associated virus serotype DJ (AAV- DJ), comprising a nucleotide sequence encoding a 7-mer peptide inserted between the 1767th and 1768th nucleotides of SEQ ID NO: 399, wherein the modified AAV-DJ has a range of at least 50% to 60%, 60% to 65%, 65% to 70%, 70% to 75%, 75% to 80%, 80% to 85%, 85% to 90%, 90% to 95%, or 95% to 99% sequence identity with the sequence of SEQ ID NO: 399 outside the nucleotide sequence encoding the 7-mer peptide
[0005] In another aspect, provided herein is a modified adeno-associated virus serotype 2 (AAV2), comprising a nucleotide sequence encoding a 7-mer peptide inserted between the 1764th and 1765th nucleotides of SEQ ID NO: 401, wherein the modified AAV2 has a range of at least 50% to 60%, 60% to 65%, 65% to 70%, 70% to 75%, 75% to 80%, 80% to 85%,
85% to 90%, 90% to 95%, or 95% to 99% sequence identity with the sequence of SEQ ID NO:
401 outside the nucleotide sequence encoding the 7-mer peptide.
[0006] In another aspect, provided herein is a modified adeno-associated virus serotype 5 (AAV5), comprising a nucleotide sequence encoding a 7-mer peptide inserted between the 1722th and 1723th nucleotides of SEQ ID NO: 402, wherein the modified AAV5 has a range of at least 50% to 60%, 60% to 65%, 65% to 70%, 70% to 75%, 75% to 80%, 80% to 85%, 85% to 90%, 90% to 95%, or 95% to 99% sequence identity with the sequence of SEQ ID NO:
402 outside the nucleotide sequence encoding 7-mer peptide.
[0007] In another aspect, provided herein is a modified adeno-associated virus serotype 6 (AAV6), comprising a nucleotide sequence encoding a 7-mer peptide inserted between the 1770th and 1771th nucleotides of SEQ ID NO: 403, wherein the modified AAV6 has a range of at least 50% to 60%, 60% to 65%, 65% to 70%, 70% to 75%, 75% to 80%, 80% to 85%, 85% to 90%, 90% to 95%, or 95% to 99% sequence identity with the sequence of SEQ ID NO:
403 outside the nucleotide sequence encoding 7-mer peptide.
[0008] In another aspect, provided herein is a modified adeno-associated virus serotype 6 (AAV6), comprising a nucleotide sequence encoding a 7-mer peptide inserted between the 1764th and 1765th nucleotides of SEQ ID NO: 403, wherein the modified AAV6 has a range of at least 50% to 60%, 60% to 65%, 65% to 70%, 70% to 75%, 75% to 80%, 80% to 85%, 85% to 90%, 90% to 95%, or 95% to 99% sequence identity with the sequence of SEQ ID NO:
403 outside the nucleotide sequence encoding the 7-mer peptide.
[0009] In yet another aspect, provided herein is a modified adeno-associated virus serotype 9 (AAV9), comprising a nucleotide sequence encoding a 7-mer peptide inserted between the 1764th and 1765th nucleotides of SEQ ID NO: 404, wherein the modified AAV9 has a range of at least 50% to 60%, 60% to 65%, 65% to 70%, 70% to 75%, 75% to 80%, 80% to 85%, 85% to 90%, 90% to 95%, or 95% to 99% sequence identity with the sequence of SEQ ID NO:
404 outside the nucleotide sequence encoding the 7-mer peptide.
[0010] In another aspect, provided herein is a composition comprising any modified AAV as described herein, and a pharmaceutically acceptable excipient.
[0011] In another aspect, provided herein is a nucleic acid encoding any modified AAV as described herein.
DEFINITIONS
[0012] As used herein, the term “AAV” or “adeno-associated virus” refers to a non-pathogenic, small viruses of the Parvovirus family which infect humans. The term “AAV” includes, but is
not limited to, naturally occurring and engineered AAV subtypes or serotypes, for example, AAV type 1 (AAV-1), AAV type 2 (AAV-2), AAV type 3 (AAV-3), AAV type 4 (AAV-4), AAV type 5 (AAV-5), AAV type 6 (AAV-6), AAV type 7 (AAV-7), AAV type 8 (AAV-8) and AAV- DJ
[0013] As used herein, the term “capsid” refers to the protein shell of a virus particle. The capsid of a virus is composed of one or more capsid units or subunits. For example, the capsid of an AAV is composed of VP1, VP2 and/or VP3 subunits. The capsid of a virus may have several functional purposes. For example, the capsid of a virus may protect the viral genome from degradation, assist the virus in evading host immune response, and/or determine viral tropism to different cell or tissue types.
[0014] As used herein, the term “identity” in the context of a polynucleotide or polypeptide sequence refers to the overall relatedness or homology between polymeric molecules, e.g., between polynucleotide molecules (e g., DNA molecules and/or RNA molecules) and/or between polypeptide molecules. Calculation of the percent identity of two polynucleotide sequences, for example, can be performed by aligning the two sequences for optimal comparison purposes (e g , gaps can be introduced in one or both of a first and a second nucleic acid sequences for optimal alignment and non-identical sequences can be disregarded for comparison purposes) The nucleotides at corresponding nucleotide positions are then compared. When a position in the first sequence is occupied by the same nucleotide as the corresponding position in the second sequence, then the molecules are identical at that position. The percent identity between the two sequences is a function of the number of identical positions shared by the sequences, taking into account the number of gaps, and the length of each gap, which needs to be introduced for optimal alignment of the two sequences. The comparison of sequences and determination of percent identity between two sequences can be accomplished using a mathematical algorithm.
[0015] As used herein, the term “library” refers to a mixture of heterogeneous polypeptides or polynucleotides. A library is composed of members that have similar polypeptide or polynucleotide sequences. Sequence differences between library members are responsible for the diversity of the library. The library can take the form of a mixture of polypeptides or polynucleotides, or can be in the form of organisms or cells, for example yeast cells and the like, that are transformed with a library of polynucleotides.
[0016] As used herein, “modified virus” and “virus variant” are interchangeable terms referring to an engineered or recombinant virus comprising an alteration, that is, “modification”, in the sequence of a polypeptide or a polynucleotide encoding a polypeptide compared to that
of a reference sequence. The reference sequence may be a genomic sequence or a subsection thereof of a virus, or the amino acid sequence encoded by the genomic sequence or a subsection thereof of a virus. The reference sequence may be a sequence from a wild-type or native virus, or it may be the sequence from a different modified virus. A wild-type virus refers to a virus with phenotype or genotype that predominates in nature. “Modified AAV” or “variant AAV” refers to an AAV that has a change in its genomic sequence or a subsection thereof, or in the polypeptide encoded by its genomic sequence or a subsection thereof, compared to a reference AAV. The alteration or modification of a sequence may refer to an insertion, deletion, or substitution of one or more residues in the sequence. For example, a modified AAV may be an AAV that has one or more nucleotides inserted into its genomic sequence or a subsection thereof compared to a reference AAV or has one or more amino acids inserted into the polypeptide encoded by its genomic sequence or a subsection thereof.
[0017] As used herein, “peptide” refers to the condensation product of two or more amino acids, wherein the length of the polypeptide is less than or equal to 50 amino acids long, e.g., 5, 6, 7, 8, 9, 10, 11-15, 15-20, 20-25, 25-30, 30-35, 35-40, 40-45 or 45-50 amino acids long.
[0018] As used herein, “targeting” and “tropism” are interchangeable terms that refer to the ability of a virus to infect different cell types to produce a successful infection. A successful infection may be defined as when the virus gains entry to the host cell, when the virus has begun replication of its genome in the host cell, when intact progeny virus is detectable within the host cell, when the virus progeny exits the host cell, or any combination thereof. In one example, a virus with tropism to the CNS or a CNS-targeting virus indicates that the virus is capable of infecting cells that are from or derived from the CNS and/or that the virus is capable of infecting cells that are from or derived from the CNS at a higher specificity than other cell types. In another example, a virus with tropism to the muscle or a muscle-targeting virus refers to a virus that is capable of successfully infecting cells that are from or derived from skeletal muscle and/or that the virus is capable of infecting cells that are from or derived from muscle at a higher specificity than other cell types. Conversely, a virus that does not have tropism or has reduced tropism to a cell type would not be capable or have a reduced capability of infecting said cell type. For example, a virus with reduced liver tropism or that is liver de-targeting indicates that the virus is less capable or unable to successfully infect cells that are from or derived from the liver. It would be understood by those skilled in the art that “tropism” may also mean “specificity to”. For example, virus tropism to the CNS means that a virus has specificity for the CNS. Therefore, as used herein, the terms “tropism” and “specificity” are interchangeable and mean the ability of a virus to successfully infect a selected cell type.
Further, a virus may also be defined as “specific targeting” or having “specific tropism”, to indicate that it is specific for a selected tissue type. For example, a virus with specific tropism to the brain is one that specifically infects the brain.
BRIEF DESCRIPTION OF THE DRAWINGS
[0019] The invention will be better understood with reference to the detailed description when considered in conjunction with the non-limiting examples and the accompanying drawings, in which:
[0020] Figure 1 shows a schematic map of the construction of the AAV-DJ library. The top diagram shows the AAV-DJ library plasmid, where the grey lines represent the insertion site of the peptide library (between N589 and R590), and the bottom diagram shows the full-length Rep2 plasmid. Abbreviations: ITR, inverted terminal repeat.
[0021] Figure 2 shows the next-gen sequencing (NGS)-driven evolution of the AAV-DJ peptide library. C57BL/6 mice (n=5) were injected intravenously with 2.5 x 109 vg/mouse of AAV-DJ peptide library. 28 days later, organs were collected, rnRNA were enriched and capsid DNA variants were amplified and analyzed by NGS. Figures 2A, 2B and 2C show distribution of the length of the inserted sequences in the AAV-DJ capsid DNA sequenced from the liver, brain, and muscle respectively. The X-axis represents the length of the nucleotide sequence inserted, and the Y-axis represents the probability density of each length of nucleotide sequences.
[0022] Figure 3 shows a table of values of the fold-change difference detected by NGS of selected AAV-DJ variants normalized to wild-type AAV-DJ, leading to identification of AAV- DJ variants with enhanced brain, muscle transduction efficiency with complete liver detargeting at low vector doses. The labels on the left denote the AAV-DJ variants, corresponding to the peptides listed in Table 3. The values in the table represent the fold-change of each variant in the liver, brain, and muscle, compared to wild-type AAV-DJ. The scale bar on the right represents the fold-change of the modified AAV genome sequenced from the respective organ versus the wild-type. Lighter shades denote higher fold-change, darker shades denote lower fold-change, with black denoting no change (baseline). Low or absence of a fold-change increase in an organ indicates a decrease or absence of infectivity or tropism in the organ, and high or presence of a fold-change increase in an organ indicates an increase or presence of infectivity or tropism in the organ.
DETAILED DESCRIPTION OF THE INVENTION
[0023] The present disclosure provides for modified AAV s with altered tropism to various cell types, tissue types and/or organs compared to a reference AAV.
[0024] The reference AAV may be a wild-type AAV or unmodified AAV, and it may be of the same or different serotype as the modified AAV.
[0025] A modified virus or virus variant may be modified so that its tropism or targeting to one or more selected cell types, tissues and/or organs is altered. For example, a wild-type virus may be modified to possess enhanced tropism, reduced tropism, or no change in tropism towards one or more selected organs, tissues and/or cell types. In some examples, the reduction in tropism is to the extent that tropism is abolished, that is, the virus is no longer capable of infecting the one or more selected organs, tissues and/or cell types. The organ may be any organ from a human or an animal. For example, the organ may be the CNS, ear, eye, heart, intestine, kidney, joint/synovium, lung, liver, pancreas, or skeletal muscle. The tissues may be primary or clinical isolates from an organ, or it may be a cultured tissue originating from an organ. The cells may be primary or clinical isolates from an organ or tissue, or it may be a cultured cells originating from an organ or tissue.
[0026] An AAV with altered tropism may be obtained for example by introducing one or more mutations into the viral genome or sections thereof, or within a specific gene. For example, an AAV may be modified by introducing mutations into one or more genes encoding the capsid protein. Mutations may be introduced by any method known by those skilled in the art, for example, site-directed mutagenesis or site-saturation mutagenesis, and may include insertion or deletion of nucleotides (frame-shift mutations), replacement/substitution of nucleotides, or combinations thereof. For example, site-saturation mutagenesis may be used to substitute a selected site in a polypeptide with all possible amino acids through the use of degenerate codons in the nucleotide sequence encoding the polypeptide. In an example, the degenerate codon is NNK, where K stands for keto nucleotides such as guanine and thymine, and N is for any nucleotide. It would be understood by those skilled in the art that a modification of the polynucleotide sequence encoding a gene may result in the modification of the polypeptide encoded by the polynucleotide sequence. It would also be understood that mutations in the context of the present invention may refer to mutations of a viral polynucleotide and/or viral polypeptide sequence.
[0027] In one example, a wild-type or reference AAV is modified by insertion of a polynucleotide sequence encoding a peptide to a desired site in the nucleotide sequence of the reference or wild-type AAV. The reference or wild-type AAV may include but is not limited to
the serotypes AAV-2, AAV-5, AAV-6, AAV-9, and AAV-DJ (Table 4). It would be understood by those skilled in the art that the resulting AAV, that is, “modified AAV”, will have a nucleotide sequence identical to that of the reference or wild-type AAV (i.e., the unmodified AAV) apart from, that is, “outside”, the nucleotide sequence encoding the inserted peptide
[0028] The desired site at which the polynucleotide sequence is inserted may be located within a coding or non-coding region of the viral genome. In one example, the wild-type or reference AAV may be modified by insertion of one or more amino acids or a peptide into a polypeptide sequence that encodes for a protein or proteins forming the capsid or part of the virus.
[0029] The peptide that is inserted to alter tropism of a wild-type or reference virus may vary in length. For example, peptides may be 5-15 amino acids in length. As non-limiting examples, the peptide may be 5, 6, 7, 8, 9, 10, 11 , 12, 13, 14 or 15 amino acids in length. For example, the inserted peptide may be a 5-mer, 6-mer, 7-mer, 8-mer, 9-mer or 10-mer peptide. In one example, the peptide is a 7-mer peptide. The amino acid sequence of the 7-mer peptide may comprise any permutation of 7 amino acids. In some examples, the amino acids are the 20 standard amino acids. Therefore, provided herein are modified AAVs comprising a 7-mer peptide sequence, wherein the peptide sequence is any permutation of the 20 standard amino acids. It would be understood by those skilled in the art which amino acids are the 20 standard amino acids.
[0030] It would be understood by those skilled in the art that certain methods of modifying a polypeptide sequence, for example, site saturation mutagenesis, allows for the screening of every of the 20 standard amino acids for a selected site in a polypeptide sequence. Therefore, the peptides listed in the present disclosure are non-limiting examples of 7-mer peptides with the property of conferring altered tropism to an AAV when inserted to the viral capsid.
[0031] Provided herein are modified AAVs comprising a nucleotide sequence encoding a 7- mer peptide inserted between two nucleotides of a reference or wild-type AAV capsid gene. The two nucleotides may be within the sequence of a VP1, VP2 and/or VP3 gene. In one example, the two nucleotides are within the sequence of a VP3 gene. It would be understood by those skilled in the art that the VP1 gene encodes for the full-length capsid, while VP2 and VP3 genes encode for subunits of the capsid. Those skilled in the art will also recognize that the nucleotide sequences of the VP2 and VP3 genes are subsumed within the VP1 nucleotide sequence.
[0032] In one aspect, provided herein is a modified adeno-associated virus serotype DJ (AAV- DJ), comprising a nucleotide sequence encoding a 7-mer peptide inserted between the 1767th and 1768th nucleotides of SEQ ID NO: 399, wherein the modified AAV-DJ has a range of at
least 50% to 60%, 60% to 65%, 65% to 70%, 70% to 75%, 75% to 80%, 80% to 85%, 85% to 90%, 90% to 95%, or 95% to 99% sequence identity with the sequence of SEQ ID NO: 399 outside the nucleotide sequence encoding the 7-mer peptide.
[0033] In another aspect, provided herein is a modified adeno-associated virus serotype 2 (AAV2), comprising a nucleotide sequence encoding a 7-mer peptide inserted between the 1764th and 1765th nucleotides of SEQ ID NO: 401, wherein the modified AAV2 has a range of at least 50% to 60%, 60% to 65%, 65% to 70%, 70% to 75%, 75% to 80%, 80% to 85%, 85% to 90%, 90% to 95%, or 95% to 99% sequence identity with the sequence of SEQ ID NO:
401 outside the nucleotide sequence encoding the 7-mer peptide.
[0034] In another aspect, provided herein is a modified adeno-associated virus serotype 5 (AAV5), comprising a nucleotide sequence encoding a 7-mer peptide inserted between the 1722th and 1723th nucleotides of SEQ ID NO: 402, wherein the modified AAV5 has a range of at least 50% to 60%, 60% to 65%, 65% to 70%, 70% to 75%, 75% to 80%, 80% to 85%, 85% to 90%, 90% to 95%, or 95% to 99% sequence identity with the sequence of SEQ ID NO:
402 outside the nucleotide sequence encoding 7-mer peptide.
[0035] In another aspect, provided herein is a modified adeno-associated virus serotype 6 (AAV6), comprising a nucleotide sequence encoding a 7-mer peptide inserted between the 1770th and 1771th nucleotides of SEQ ID NO: 403, wherein the modified AAV6 has a range of at least 50% to 60%, 60% to 65%, 65% to 70%, 70% to 75%, 75% to 80%, 80% to 85%, 85% to 90%, 90% to 95%, or 95% to 99% sequence identity with the sequence of SEQ ID NO:
403 outside the nucleotide sequence encoding 7-mer peptide.
[0036] In another aspect, provided herein is a modified adeno-associated virus serotype 6 (AAV6), comprising a nucleotide sequence encoding a 7-mer peptide inserted between the 1764th and 1765th nucleotides of SEQ ID NO: 403, wherein the modified AAV6 has a range of at least 50% to 60%, 60% to 65%, 65% to 70%, 70% to 75%, 75% to 80%, 80% to 85%, 85% to 90%, 90% to 95%, or 95% to 99% sequence identity with the sequence of SEQ ID NO:
403 outside the nucleotide sequence encoding the 7-mer peptide.
[0037] In another aspect, provided herein is a modified adeno-associated virus serotype 9 (AAV9), comprising a nucleotide sequence encoding a 7-mer peptide inserted between the 1764th and 1765th nucleotides of SEQ ID NO: 404, wherein the modified AAV9 has a range of at least 50% to 60%, 60% to 65%, 65% to 70%, 70% to 75%, 75% to 80%, 80% to 85%, 85% to 90%, 90% to 95%, or 95% to 99% sequence identity with the sequence of SEQ ID NO:
404 outside the nucleotide sequence encoding the 7-mer peptide.
[0038] It would generally be understood that there may be some variation in the sequence identity of a virus, for example a modified virus, compared to a reference virus, for example a wild-type virus. For example, a virus may have at least 50% to 60%, 60% to 65%, 65% to 70%, 70% to 75%, 75% to 80%, 80% to 85%, 85% to 90%, 90% to 95%, or 95% to 99% sequence identity to a reference virus. The comparison may be between the AAV of the same or different serotypes. For example, an AAV-DJ may have at least 50% sequence identity with a reference AAV-2, AAV-5, AAV-6, AAV-9, or AAV-DJ virus. In one example, an AAV-DJ has at least 57% sequence identity with a reference AAV-5.
[0039] In one example, provided herein is a modified AAV-DJ as described herein, wherein the modified AAV-DJ has at least 57% sequence identity with the sequence of SEQ ID NO: 399 outside the 7-mer peptide sequence.
[0040] In another example, provided herein is a modified AAV2 as described herein, wherein the modified AAV2 has at least 57% sequence identity with the sequence of SEQ ID NO: 401 outside the 7-mer peptide sequence.
[0041] In another example, provided herein is a modified AAV5 as described herein, wherein the modified AAV5 has at least 57% sequence identity with the sequence of SEQ ID NO: 402 outside the 7-mer peptide sequence.
[0042] In another example, provided herein is a modified AAV6 comprising a sequence corresponding to a 7-mer peptide inserted between the 1770th and 1771th nucleotides of SEQ ID NO: 403, wherein the modified AAV6 has at least 57% sequence identity with the sequence of SEQ ID NO: 403 outside the 7-mer peptide sequence.
[0043] In yet another example, provided herein is a modified AAV6 comprising a sequence corresponding to a 7-mer peptide inserted between the 1764th and 1765th nucleotides of SEQ ID NO: 403, wherein the modified AAV 6 has at least 57% sequence identity with the sequence of SEQ ID NO: 403 outside the 7-mer peptide sequence.
[0044] In yet another example, provided herein is a modified AAV9 as described herein, wherein the modified AAV9 has at least 57% sequence identity with the sequence of SEQ ID NO: 404 outside the 7-mer peptide sequence.
[0045] In one example, the inserted peptides of the present invention may comprise a sequence as set forth in Table 1 and confers enhanced liver, brain, and muscle tropism to a modified AAV compared to a reference or wild-type AAV. In another example, the targeting peptide may comprise a sequence as set forth in Table 2 and confers enhanced brain and muscle tropism with reduced liver tropism to a modified AAV compared to a wild-type AAV. In yet another example, the targeting peptide may comprise a sequence as set forth in Table 3 and confers
enhanced brain and muscle tropism with liver de-targeting to a modified AAV compared to a wild-type AAV
[0046] The modified virus or virus variant of the present invention may have increased or decreased specificity to one or more selected cell types compared to a reference virus. The increase or decrease in specificity may refer to the increased ability of the virus to bind to and/or infect said cell type. The increase or decrease in specificity of a virus to a cell type may be compared to that of a reference cell type may be measured by quantitative methods known to those skilled in the art. The degree of enhancement or reduction in tropism of a modified AAV relative to a reference AAV, such as a wild-type AAV, may be calculated by comparing the infectivity of the variant relative to the infectivity of the reference virus in a selected tissue sample. Those skilled in the art will be aware of the methods of measuring infectivity of viruses. For example, infectivity of a virus may be measured by plaque assay, enzyme-linked immunosorbent assay (ELSA), transduction assay using reporter genes, polymerase chain reaction (PCR), high-throughput sequencing or any combination thereof. In one example, the infectivity or tropism of an AAV is assessed by next-generation sequencing (NGS).
[0047] The comparing of the infectivity or tropism of an AAV variant relative to the infectivity of a reference or original AAV, wherein infectivity is measured by NGS, may be done by dividing the number of reads of the AAV variant in a selected tissue sample, for example the brain, muscle, or liver, by the number of reads of the reference or original virus in the tissue sample to obtain the degree fold-change in infectivity or tropism of the AAV variant relative to the reference or original virus. When the AAV variant has fold-change of > 1 relative to the reference or original AAV in a tissue type, it has increased or improved tropism for that tissue type compared to the reference or original virus. When the AAV variant has fold-change of < 1 relative to the reference or original AAV in a tissue type, it has decreased or reduced tropism forthat tissue type compared to the reference or original virus. When the AAV variant has foldchange = 1 relative to the reference or original AAV in a tissue type, it has same or similar tropism for that tissue type compared to the reference or original virus. When the AAV variant has fold-change = 0 relative to the reference or original AAV in a tissue type, it is not detectable in that tissue type or de-targeting towards that tissue type compared to the reference or original virus.
Table 2: AAV-DJ Variants with Enhanced Brain and Muscle Tropism and Reduced Liver
Table 3: AAV-DJ Variants with Superior Brain and Muscle Tropism and Liver Detargeting
[0048] In one example, the modified AAV comprises a 7-mer peptide sequence selected from the sequences set forth in Table 1 , and the insertion increases brain, muscle, and liver tropism compared to a wild-type AAV without a 7-mer peptide sequence insertion.
[0049] Tn another example, the modified AAV comprises a 7-mer peptide sequence selected from the sequences set forth in Table 2, and the insertion increases brain, and muscle tropism, and decreases liver tropism compared to a wild-type AAV without a 7-mer peptide sequence insertion.
[0050] In yet another example, the modified AAV comprises a 7-mer peptide sequence selected from the sequences set forth in Table 3, and wherein the insertion increases brain, and muscle tropism, and is liver de-targeting.
[0051] In yet another example, the modified AAV comprises a 7-mer peptide sequence selected from the group consisting of SEQ ID NOs: 22, 24, 32, 85, 106, 198, 216, 232, 251, 293, 312, 320, 321, 224, 346 or 376.
[0052] In any of the DNA sequences referenced and/or described herein, the single letter symbol has the following description: A for adenine; C for cytosine; G for guanine; T for thymine; K for keto nucleotides such as guanine and thymine; and N is for any nucleotide (which is not a gap). The use of the NNK codon in site-saturation mutagenesis allows for coverage of codons for all 20 standard amino acids, including degenerate codons. “Degenerate codons” refers to codons differing in one or two nucleotides but coding for the same amino acid. For example, the amino acid Alanine may be encoded by the codons GCT, GCC, GCA or GCG and the amino acid Leucine may be encoded by the codons CTT, CTC, CTA, CTG, TTA and TTG.
[0053] In any of the amino acid sequences referenced and/or described herein, the single letter symbol has the following description: G (Gly) for Glycine; A (Ala) for Alanine; L (Leu) for Leucine; M (Met) for Methionine; F (Phe) for Phenylalanine; W (Trp) for Tryptophan; K (Lys) for Lysine; Q (Gin) for Glutamine; E (Glu) for Glutamic Acid; S (Ser) for Serine; P (Pro) for Proline, V (Vai) for Valine, I (He) for Isoleucine; C (Cys) for Cysteine; Y (Tyr) for Tyrosine; H (His) for Histidine; R (Arg) for Arginine; N (Asn) for Asparagine; D (Asp) for Aspartic Acid; and T (Thr) for Threonine.
[0054] The modified AAVs provided herein may possess advantageous characteristics when used for research and/or therapeutic applications as a result of their enhanced tropism to an organ, tissue and/or cell type. For example, modified AAVs with enhanced tropism may be used at lower vector doses resulting reduced off-target toxicities. Modified AAVs with enhanced tropism may also allow for the regulation of anti-capsid immune responses, and may be used for therapeutic purposes, such as CRISPR delivery and gene augmentation therapy. Such targeting may enhance the specificity of AAV s to the desired cell types and improve the overall therapeutic effect. In yet another example, in the case of liver targeting, liver sinusoidal endothelial cells (LSECs) and hepatocytes and other cell types in the liver could be targeted, whereas in the case of brain targeting, neurons and astrocytes and other cell types in the brain could be targeted. Additionally, the LSEC targeting can reduce the induction of the immune responses to AAV capsid. The modified AAVs provided herein may also possess advantageous characteristics when used for research and/or therapeutic applications as a result of their reduced tropism to an organ, tissue and/or cell type. For example, the modified AAVs with reduced tropism could be used in therapy at lower vector doses because they are not sequestrated in the liver, leading to reduced off-target toxicities. AAV dosage in therapy may be represented as the amount of vector genomes or viral genomes administered to a subject. For example, the dose may be specified as the amount of vector genomes per body weight (vg/kg) or amount of vector genomes per volume (vg/ml or vg/L) or vector genomes per animal (vg/animal). The therapeutically effective vector dose may be a range. For example, the effective dose per animal may be as low as 1 x 109 to as high as 1 x IO17 vg, depending on the route and site of administration.
[0055] In one example, provided herein is the modified AAV as described herein, wherein the 7-mer peptide sequence is selected from the sequences set forth in Table 1 or Table 7, and wherein the insertion increases specificity to cells selected from the group consisting of: liver sinusoidal endothelial cells, or hepatocytes, compared to a wild-type AAV without a 7-mer peptide sequence insertion.
[0056] In another example, provided herein is the modified AAV as described herein, wherein the 7-mer peptide sequence is selected from the sequences set forth in Table 1, Table 2 or Table 3, and wherein the insertion increases specificity to cells selected from the group consisting of: neurons, or astrocytes, compared to a wild-type AAV without a 7-mer peptide sequence insertion.
[0057] In another example, provided herein is the modified AAV as described herein, wherein the 7-mer peptide sequence is selected from the sequences set forth in Table 7, and wherein the insertion increases production of antigen-specific regulatory T cells compared to a wild-type AAV without a 7-mer peptide sequence insertion.
[0059] In some examples, insertion of the 7-mer peptide sequence may reduce the induction of immune responses to the modified AAV capsid compared to a wild-type AAV without a 7-mer peptide sequence insertion.
[0060] In other examples, insertion of the 7-mer peptide sequence may regulate anti-capsid immune response compared to a wild-type AAV without a 7-mer peptide sequence insertion.
[0058] In yet another example, provided herein is the modified AAV as described herein, wherein the 7-mer peptide sequence is selected from the sequences set forth in Table 1, Table 2 or Table 3, and wherein the insertion improves therapeutic effect compared to a wild-type AAV without a 7-mer peptide sequence insertion. In some examples, the improvement in therapeutic effect may be reduction of off-target effects or the reduction of vector dose required for therapy. Off-target effects refer to effects in organs or tissues other than in the intended organ or tissue when an AAV is used in therapy.
[0059] In one aspect, there is provided herein a composition comprising one or more of the modified AAV described herein. In one example, the composition is a pharmaceutical composition comprising one or more of the modified AAV described herein and a pharmaceutically acceptable excipient.
[0060] In another aspect, there is provided a nucleic acid encoding the modified AAV as described herein.
[0061] The modified AAV described herein may be produced using several methods known in the art. For example, the modified AAV may be produced via the use of a virus packaging cell, such as the HEK-293T cell line It would be understood by those skilled in the art that one or more nucleic acid molecules comprising a nucleic acid sequence encoding the virus would be introduced into the packaging cell, which would then produce intact, infectious virus particles. [0062] The invention illustratively described herein may suitably be practiced in the absence of any element or elements, limitation or limitations, not specifically disclosed herein. Thus,
for example, the terms “comprising”, “including”, “containing”, etc. shall be read expansively and without limitation. Additionally, the terms and expressions employed herein have been used as terms of description and not of limitation, and there is no intention in the use of such terms and expressions of excluding any equivalents of the features shown and described or portions thereof, but it is recognized that various modifications are possible within the scope of the invention claimed. Thus, it should be understood that although the present invention has been specifically disclosed by preferred embodiments and optional features, modification and variation of the inventions embodied therein herein disclosed may be resorted to by those skilled in the art, and that such modifications and variations are considered to be within the scope of this invention.
[0063] The invention has been described broadly and generically herein Each of the narrower species and subgeneric groupings falling within the generic disclosure also form part of the invention. This includes the generic description of the invention with a proviso or negative limitation removing any subject matter from the genus, regardless of whether or not the excised material is specifically recited herein.
[0064] Other embodiments are within the following claims and non- limiting examples In addition, where features or aspects of the invention are described in terms of Markush groups, those skilled in the art will recognize that the invention is also thereby described in terms of any individual member or subgroup of members of the Markush group.
EXPERIMENTAL SECTION
[0065] Non-limiting examples of the invention and comparative examples will be further described in greater detail by reference to specific Examples, which should not be construed as in any way limiting the scope of the invention.
[0066] Example 1: Construction of the NNK-based 7-mer peptide insertion library and vector production.
[0067] AAV-DJ-based capsid libraries were generated by the insertion of random 7-mer peptide between amino acids 589 and 590. In order to generate a transcription-competent library for mRNA-driven directed evolution, the capsid gene is placed under the control of a ubiquitous promoter (cytomegalovirus “CMV” promoter) in tandem with Rep2 P40 promoter (starting at nucleotide #1700). The P40 sequence is required for capsid mRNA expression and splicing during AAV production. The sequence is itself flanked by inverted terminal repeats (ITR) so each variant will encapsulate its own capsid sequence (Figure 1).
[0068] For cloning of capsid libraries, a 453-bp gBlock fragment of the wtAAV-DJ capsid was synthesized and used as a template to introduce the random 7-mer peptide library and overhangs for subcloning. PCR was performed for 15 cycles using Neb Q5 polymerase. The CMV promoter was amplified by PCR to introduce the Swal restriction site and overhangs. The resulting amplicons were purified via gel-extraction. The remaining fragments were synthesized as gBlocks. The different fragments were then assembled by Gibson assembly following manufacturer’s instructions into the recipient ITR plasmid previously digested with SphI and Mfel. The gBlocks were synthesized by IDT.
[0069] The full-length and unmodified Rep2 (Rep2-ACap, Figure 1) was provided in trans by a separate plasmid during vector production. The VP1 (MIL), VP2 (T138A), and VP3 (M205L, M21 IL, and M235L) were silenced and the plasmid was assembled by Gibson assembly using pRep2-Cap-DJ_MlL as a template and gBlock containing the AAV-DJ fragment with VP2/VP3 start codon mutations.
[0070] The diversity of the plasmid library was determined by the number of clones growing from a representative aliquot of transformed chemically competent DH5a bacteria. Library plasmids were harvested and purified using a maxi prep plasmid purification kit.
[0071] Virus production was performed by triple transfection in HEK293T cells using 15 pg of helper plasmid, 10 pg of Rep2-Acap plasmid and 1 pg of library DNA plasmid per T175 flask. 24 hours later, the medium was changed to a fresh culture medium containing 2%FBS. Three days after transfection, cells were collected and resuspended in lysis buffer (Tris HC1 pH
7.5 + 2 mM MgC12 + 150 mM NaCl) and lysed by three cycles of freezing/thawing. The supernatant was collected and treated with 50 U/ml of benzonase and 1 U/ml of Rnase cocktail for 30 min at 37°C to remove unpackaged nucleic acids. After incubation, the lysate was loaded on top of a discontinuous density gradient (15%, 25%, 40%, and 60%) and ultracentrifuged at 54,000 rpm, at 18 °C, for 1.5 hr, on a Type 70 Ti rotor. The 40% fraction was extracted and dialyzed with lx PBS (pH 7.2) with 0.001% pluronic acid, using Amicon® Ultra-15 (100 kDa MWCO) filters. Primers and probes localizing to within the AAV2 ITR were used for the quantification of the viral titer by digital droplet PCR (ddPCR).
[0072] Example 2: in vivo library screening in mice
[0073] AAV-DJ libraries were injected into C57BL/6 mice (male, N = 5) at a vector dose of
2.5 xlO9 vg/mouse into the lateral tail vein. Animals were euthanized 28 days later, and the organs were harvested and stored at -80°C. Total RNA was extracted from different organs using the Rneasy® Plus mini kit. The sample was then enriched for mRNA using Oligo(dT)25 magnetic beads, and cDNA synthesis was performed using SuperScript™ IV reverse
transcriptase following the manufacturer’s instructions. Capsid DNA variants were amplified from cDNA for the generation of NGS amplicons The amplicons were quantified using a fluorometer, pooled at an equimolar ratio, and sequenced on an iSeq™ sequencer. Following in vivo selection, more than 10,000 unique capsid variants were identified, with some showing superior transduction efficiency in the liver, brain, and skeletal muscle (Figure 2, A-C). Moreover, potential variants with enhanced brain-tropic or myotropic with complete liver detargeting were also identified (Figure 3).
[0074] The altered tropism of the AAV variants compared to that of the reference AAV, that is, a wild-type AAV, was assessed by calculating transduction fold-change of the variant relative to the wild-type virus for a selected tissue type. First, a normalization factor was calculated by factoring the wild-type count in the original virus pool (that is, the library) by the wild-type count in a selected tissue (Normalisation factor (NF)= AAV_WT count in original virus pool / AAV WT count in tissue). Next, a normalized variant count for each variant was obtained by multiplying each variant count in the selected tissue by the normalization factor of that tissue (Normalised AAV variant count = count AAV variant in tissue x NF Tissue). Finally, the transduction fold change of each variant was calculated by factoring the normalized variant count by the wild-type count in the original virus pool (Transduction fold-change (FC) = Normalised AAV variant count /AAV wt count in original virus pool). The counts, normalized counts and transduction fold change in each selected tissue for each variant relative to wildtype AAV can be found in Tables 5 and 6. In said tables, a count of indicates that the variant was not detected in that tissue sample. The amino acids “N” and “R” at the beginning and end of each sequence respectively denote N589 and R590 of the wild-type AAV-DJ capsid, between which the 7-mer peptide is inserted.
[0075] Example 3: selection of peptides conferring tropism to liver sinusoidal endothelial cells (LSECs)
[0076] Animals received injections of 2.5 x 10A9 vg/animal of AAV. One month after the injection of the AAV library, organs were harvested. Specifically, for this round of selection, after liver collection, a portion of the liver was sectioned off to prepare a cell suspension. From this suspension, CD146+ liver sinusoidal endothelial cells (LSECs) were isolated using fluorescence-activated cell sorting (FACS). This process led to the identification of AAV-DJ variants with improved tropism to LSECs (Table 7). Table 7. Peptides identified conferring tropism to liver sinusoidal endothelial cells (LSECs)
[0077] Equivalents
[0078] The foregoing examples are presented for the purpose of illustrating the invention and should not be construed as imposing any limitation on the scope of the invention. It will readily
be apparent that numerous modifications and alterations may be made to the specific embodiments of the invention described above and illustrated in the examples without departing from the principles underlying the invention. All such modifications and alterations are intended to be embraced by this application.
Claims
1. A modified adeno-associated virus serotype DJ (AAV-DJ), comprising a nucleotide sequence encoding a 7-mer peptide inserted between the 1767th and 1768th nucleotides of SEQ ID NO: 399, wherein the modified AAV-DJ has a range of at least 50% to 60%, 60% to 65%, 65% to 70%, 70% to 75%, 75% to 80%, 80% to 85%, 85% to 90%, 90% to 95%, or 95% to 99% sequence identity with the sequence of SEQ ID NO: 399 outside the nucleotide sequence encoding the 7-mer peptide.
2. A modified adeno-associated virus serotype 2 (AAV2), comprising a nucleotide sequence encoding a 7-mer peptide inserted between the 1764th and 1765th nucleotides of SEQ ID NO: 401, wherein the modified AAV2 has a range of at least 50% to 60%, 60% to 65%, 65% to 70%, 70% to 75%, 75% to 80%, 80% to 85%, 85% to 90%, 90% to 95%, or 95% to 99% sequence identity with the sequence of SEQ ID NO: 401 outside the nucleotide sequence encoding the 7-mer peptide.
3. A modified adeno-associated virus serotype 5 (AAV5), comprising a nucleotide sequence encoding a 7-mer peptide inserted between the 1722th and 1723th nucleotides of SEQ ID NO: 402, wherein the modified AAV5 has a range of at least 50% to 60%, 60% to 65%, 65% to 70%, 70% to 75%, 75% to 80%, 80% to 85%, 85% to 90%, 90% to 95%, or 95% to 99% sequence identity with the sequence of SEQ ID NO: 402 outside the nucleotide sequence encoding the 7-mer peptide.
4. A modified adeno-associated virus serotype 6 (AAV6), comprising a nucleotide sequence encoding a 7-mer peptide inserted between the 1770th and 1771th nucleotides of SEQ ID NO: 403, wherein the modified AAV6 has a range of at least 50% to 60%, 60% to 65%, 65% to 70%, 70% to 75%, 75% to 80%, 80% to 85%, 85% to 90%, 90% to 95%, or 95% to 99% sequence identity with the sequence of SEQ ID NO: 403 outside the nucleotide sequence encoding the 7-mer peptide.
5. A modified adeno-associated virus serotype 6 (AAV6), comprising a nucleotide sequence encoding a 7-mer peptide inserted between the 1764th and 1765th nucleotides of SEQ ID NO: 403, wherein the modified AAV6 has a range of at least 50% to 60%, 60% to 65%, 65% to 70%, 70% to 75%, 75% to 80%, 80% to 85%, 85% to 90%, 90% to 95%, or 95%
to 99% sequence identity with the sequence of SEQ ID NO: 403 outside the nucleotide sequence encoding the 7-mer peptide.
6. A modified adeno-associated virus serotype 9 (AAV9), comprising a nucleotide sequence encoding a 7-mer peptide inserted between the 1764th and 1765th nucleotides of SEQ ID NO: 404, wherein the modified AAV9 has a range of at least 50% to 60%, 60% to 65%, 65% to 70%, 70% to 75%, 75% to 80%, 80% to 85%, 85% to 90%, 90% to 95%, or 95% to 99% sequence identity with the sequence of SEQ ID NO: 404 outside the nucleotide sequence encoding the 7-mer peptide.
7. The modified A AV-DJ of claim 1 , wherein the modified A AV-DJ has at least 57% sequence identity with the sequence of SEQ ID NO: 399 outside the nucleotide sequence encoding the 7-mer peptide.
8. The modified AAV2 of claim 2, wherein the modified AAV2 has at least 57% sequence identity with the sequence of SEQ ID NO: 401 outside the nucleotide sequence encoding the 7-mer peptide.
9. The modified AAV5 of claim 3, wherein the modified AAV5 has at least 57% sequence identity with the sequence of SEQ ID NO: 402 outside the nucleotide sequence encoding the 7-mer peptide.
10. The modified AAV6 of claim 4, wherein the modified AAV6 has at least 57% sequence identity with the sequence of SEQ ID NO: 403 outside the nucleotide sequence encoding the 7-mer peptide.
11. The modified AAV6 of claim 5, wherein the modified AAV6 has at least 57% sequence identity with the sequence of SEQ ID NO: 403 outside the nucleotide sequence encoding the 7-mer peptide.
12. The modified AAV9 of claim 6, wherein the modified AAV9 has at least 57% sequence identity with the sequence of SEQ ID NO: 404 outside the nucleotide sequence encoding the 7-mer peptide.
13. The modified AAV of any one of claims 1-12, wherein the 7-mer peptide sequence is selected from the sequences set forth in Table 1 , and wherein the insertion increases brain, muscle, and liver tropism compared to a wild-type AAV without a 7-mer peptide sequence insertion.
14. The modified AAV of any one of claims 1-12, wherein the 7-mer peptide sequence is selected from the sequences set forth in Table 2, and wherein the insertion increases brain, and muscle tropism, and decreases liver tropism compared to a wild-type AAV without a 7-mer peptide sequence insertion.
15. The modified AAV of any one of claims 1 -12, wherein the 7-mer peptide sequence is selected from the sequences set forth in Table 3, and wherein the insertion increases brain, and muscle tropism, and is liver de-targeting.
16. The modified AAV-DJ of any one of claims 1-12, wherein the 7-mer peptide sequence is selected from the group consisting of SEQ ID NOs: 22, 24, 32, 85, 106, 198, 216, 232, 251, 293, 312, 320, 321, 224, 346 or 376.
17. The modified AAV of any one of claims 1-12, wherein the 7-mer peptide sequence is selected from the sequences set forth in Table 1 or Table 7, and wherein the insertion increases specificity to cells selected from the group consisting of: liver sinusoidal endothelial cells or hepatocytes, compared to a wild-type AAV without a 7-mer peptide sequence insertion.
18. The modified AAV of any one of claims 1 -12, wherein the 7-mer peptide sequence is selected from the sequences set forth in Table 1, Table 2 or Table 3, and wherein the insertion increases specificity to cells selected from the group consisting of: neurons, or astrocytes, compared to a wild-type AAV without a 7-mer peptide sequence insertion.
19. The modified AAV of any one of claims 1-12, wherein the 7-mer peptide sequence is selected from the sequences set forth in Table 7, and wherein the insertion increases production of antigen-specific regulatory T cells compared to a wild-type AAV without a 7-mer peptide sequence insertion.
20. The modified AAV of any one of claims 1-12, wherein the 7-mer peptide sequence is selected from the sequences set forth in Table 1, Table 2, or Table 3, and wherein the insertion improves therapeutic effect compared to a wild-type AAV without a 7-mer peptide sequence insertion.
21. A composition comprising the modified AAV of any one of the preceding claims, and a pharmaceutically acceptable excipient.
22. A nucleic acid encoding the modified AAV of any one of the preceding claims.
Applications Claiming Priority (2)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
SG10202300637W | 2023-03-09 | ||
SG10202300637W | 2023-03-09 |
Publications (1)
Publication Number | Publication Date |
---|---|
WO2024186275A1 true WO2024186275A1 (en) | 2024-09-12 |
Family
ID=92675619
Family Applications (1)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
PCT/SG2024/050144 WO2024186275A1 (en) | 2023-03-09 | 2024-03-11 | Novel compositions of adeno-associated viruses engineered for enhanced tissue transduction and specificity |
Country Status (1)
Country | Link |
---|---|
WO (1) | WO2024186275A1 (en) |
Citations (6)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US7588772B2 (en) * | 2006-03-30 | 2009-09-15 | Board Of Trustees Of The Leland Stamford Junior University | AAV capsid library and AAV capsid proteins |
US20170166926A1 (en) * | 2015-12-11 | 2017-06-15 | California Institute Of Technology | TARGETING PEPTIDES FOR DIRECTING ADENO-ASSOCIATED VIRUSES (AAVs) |
WO2018189244A1 (en) * | 2017-04-11 | 2018-10-18 | Ruprecht-Karls-Universität Heidelberg | Adeno-associated virus library |
WO2020193799A1 (en) * | 2019-03-28 | 2020-10-01 | Fraunhofer-Gesellschaft zur Förderung der angewandten Forschung e.V. | Mutated adeno-associated virus capsid proteins, aav particle comprising the same and liver directed aav vector gene therapy |
US20210355171A1 (en) * | 2019-01-30 | 2021-11-18 | Shanghaitech University | Adeno-associated virus having a variant capsid protein, and use thereof |
WO2022173847A2 (en) * | 2021-02-09 | 2022-08-18 | Affinia Therapeutics Inc. | Recombinant aavs with improved tropism and specificity |
-
2024
- 2024-03-11 WO PCT/SG2024/050144 patent/WO2024186275A1/en unknown
Patent Citations (6)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US7588772B2 (en) * | 2006-03-30 | 2009-09-15 | Board Of Trustees Of The Leland Stamford Junior University | AAV capsid library and AAV capsid proteins |
US20170166926A1 (en) * | 2015-12-11 | 2017-06-15 | California Institute Of Technology | TARGETING PEPTIDES FOR DIRECTING ADENO-ASSOCIATED VIRUSES (AAVs) |
WO2018189244A1 (en) * | 2017-04-11 | 2018-10-18 | Ruprecht-Karls-Universität Heidelberg | Adeno-associated virus library |
US20210355171A1 (en) * | 2019-01-30 | 2021-11-18 | Shanghaitech University | Adeno-associated virus having a variant capsid protein, and use thereof |
WO2020193799A1 (en) * | 2019-03-28 | 2020-10-01 | Fraunhofer-Gesellschaft zur Förderung der angewandten Forschung e.V. | Mutated adeno-associated virus capsid proteins, aav particle comprising the same and liver directed aav vector gene therapy |
WO2022173847A2 (en) * | 2021-02-09 | 2022-08-18 | Affinia Therapeutics Inc. | Recombinant aavs with improved tropism and specificity |
Similar Documents
Publication | Publication Date | Title |
---|---|---|
CN104937100B (en) | AAV variants | |
US8574583B2 (en) | AAV capsid library and AAV capsid proteins | |
WO2018139634A1 (en) | Mutant of adeno-associated virus (aav) capsid protein | |
CN113748122B (en) | AAV mutants with brain targeting properties | |
WO2017066764A2 (en) | Modification of n-terminal region of capsid proteins for enhanced properties of adeno-associated viruses | |
AU2019270900A1 (en) | Viral vectors exhibiting improved gene delivery properties | |
US20230049066A1 (en) | Novel aav3b variants that target human hepatocytes in the liver of humanized mice | |
KR20220158674A (en) | Isolation of AAV5 and Modified VP1 Capsid Protein | |
CN111718420B (en) | Fusion protein for gene therapy and application thereof | |
CN111718418B (en) | Fusion protein for enhancing gene editing and application thereof | |
EP4159863A1 (en) | Codon-optimized nucleic acid encoding smn1 protein | |
WO2024186275A1 (en) | Novel compositions of adeno-associated viruses engineered for enhanced tissue transduction and specificity | |
CN112011571A (en) | Gene therapy medicine for treating spinal muscular atrophy | |
RU2809389C2 (en) | Aav mutant with ability to target brain | |
HK40062349A (en) | Aav mutant having brain-targeting property | |
OA21075A (en) | Codon-optimized nucleic acid that encodes SMN1 protein, and use thereof | |
WO2024237335A1 (en) | Adeno-associated virus vector | |
WO2025023319A1 (en) | Brain-tropic aav mutant | |
HK40069854A (en) | Aav mutant having brain-targeting property | |
EA045749B1 (en) | CODON-OPTIMIZED NUCLEIC ACID ENCODING THE SMN1 PROTEIN AND ITS APPLICATION | |
WO2025006829A1 (en) | Improved adeno-associated virus vectors | |
CN117264912A (en) | Recombinant adeno-associated virus particle, recombinant adeno-associated virus vector system and application thereof | |
HK40014535A (en) | Mutant of adeno-associated virus (aav) capsid protein |
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: 24767512 Country of ref document: EP Kind code of ref document: A1 |