US20180135076A1 - Adeno-associated virus vector - Google Patents

Adeno-associated virus vector Download PDF

Info

Publication number
US20180135076A1
US20180135076A1 US15/570,687 US201515570687A US2018135076A1 US 20180135076 A1 US20180135076 A1 US 20180135076A1 US 201515570687 A US201515570687 A US 201515570687A US 2018135076 A1 US2018135076 A1 US 2018135076A1
Authority
US
United States
Prior art keywords
capsid protein
variant
aav
sequence
amino acid
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US15/570,687
Other languages
English (en)
Inventor
Ralph Michael Linden
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Kings College London
Icahn School of Medicine at Mount Sinai
Original Assignee
Kings College London
Icahn School of Medicine at Mount Sinai
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=50490674&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=US20180135076(A1) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Application filed by Kings College London, Icahn School of Medicine at Mount Sinai filed Critical Kings College London
Priority to US15/570,687 priority Critical patent/US20180135076A1/en
Assigned to ICAHN SCHOOL OF MEDICINE AT MOUNT SINAI, KING'S COLLEGE LONDON reassignment ICAHN SCHOOL OF MEDICINE AT MOUNT SINAI ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: LINDEN, RALPH MICHAEL
Publication of US20180135076A1 publication Critical patent/US20180135076A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/85Vectors or expression systems specially adapted for eukaryotic hosts for animal cells
    • C12N15/86Viral vectors
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
    • A61K48/0008Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy characterised by an aspect of the 'non-active' part of the composition delivered, e.g. wherein such 'non-active' part is not delivered simultaneously with the 'active' part of the composition
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/16Drugs for disorders of the alimentary tract or the digestive system for liver or gallbladder disorders, e.g. hepatoprotective agents, cholagogues, litholytics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • A61P27/06Antiglaucoma agents or miotics
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/005Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from viruses
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/85Vectors or expression systems specially adapted for eukaryotic hosts for animal cells
    • C12N15/86Viral vectors
    • C12N15/861Adenoviral vectors
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2750/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssDNA viruses
    • C12N2750/00011Details
    • C12N2750/14011Parvoviridae
    • C12N2750/14111Dependovirus, e.g. adenoassociated viruses
    • C12N2750/14122New viral proteins or individual genes, new structural or functional aspects of known viral proteins or genes
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2750/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssDNA viruses
    • C12N2750/00011Details
    • C12N2750/14011Parvoviridae
    • C12N2750/14111Dependovirus, e.g. adenoassociated viruses
    • C12N2750/14132Use of virus as therapeutic agent, other than vaccine, e.g. as cytolytic agent
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2750/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssDNA viruses
    • C12N2750/00011Details
    • C12N2750/14011Parvoviridae
    • C12N2750/14111Dependovirus, e.g. adenoassociated viruses
    • C12N2750/14141Use of virus, viral particle or viral elements as a vector
    • C12N2750/14142Use of virus, viral particle or viral elements as a vector virus or viral particle as vehicle, e.g. encapsulating small organic molecule
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2750/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssDNA viruses
    • C12N2750/00011Details
    • C12N2750/14011Parvoviridae
    • C12N2750/14111Dependovirus, e.g. adenoassociated viruses
    • C12N2750/14141Use of virus, viral particle or viral elements as a vector
    • C12N2750/14143Use of virus, viral particle or viral elements as a vector viral genome or elements thereof as genetic vector

Definitions

  • the present invention relates to the field of recombinant viral vectors.
  • the invention relates to recombinant viral vectors which are suitable for the delivery of therapeutic genes in vivo.
  • adeno-associated virus remains one of the most promising vectors for the delivery of therapeutic genes.
  • a significant number of preclinical and clinical studies have firmly established that this approach is suitable for the development of gene-based drugs that can reach market approval.
  • AAV2 As a vector for gene therapy in the 1980s much progress has been made in optimizing this platform for a variety of applications and target tissues. Among those developments, possibly the most consequential has been the discovery of a wide variety of serotypes of which ten to twelve are now commonly explored. Among the most prominent characteristics of these various serotypes are their respective relative tissue tropism and—in some cases—the ability of neuronal retrograde transport. Of these serotypes, AAV1-10 are broadly used for pre-clinical and clinical purposes.
  • a newer platform has been developed that involves processes that allow for the targeting and de-targeting of specific tissues and cell sub-types in patients.
  • the core technology of these approaches is based on trial and error evaluation of existing AAV variants (serotypes) and in vivo selection of randomly introduced AAV capsid mutants. Together, these two promising approaches provide tens—if not hundreds of potential vectors with different transduction behaviour.
  • AAV serotypes The most interesting aspect of AAV serotypes is their ability to efficiently transduce specific tissues in animal models and in man. To date, comprehensive molecular understanding of the underlying mechanisms for the tissue tropism has yet to be put forward and it is thus generally assumed that the available tissue-specific receptors for each serotype play a central role in the efficient transduction by the various serotypes.
  • AAV vectors which have improved properties in terms of in vivo transgene expression and tissue specificity.
  • such vectors have the potential to provide greatly enhanced benefits for gene delivery to various target tissues in humans.
  • the present invention provides a recombinant adeno-associated virus (AAV) vector comprising: (a) a variant AAV2 capsid protein, wherein the variant AAV2 capsid protein comprises at least four amino acid substitutions with respect to a wild type AAV2 capsid protein; wherein the at least four amino acid substitutions are present at the following positions in an AAV2 capsid protein sequence: 457, 492, 499 and 533; and (b) a heterologous nucleic acid comprising a nucleotide sequence encoding a gene product.
  • AAV adeno-associated virus
  • the variant AAV capsid protein comprises a sequence of SEQ ID NO:2, or a sequence having at least 95% sequence identity thereto.
  • the wild type AAV capsid protein comprises a sequence of SEQ ID NO:1.
  • the variant AAV2 capsid protein comprises one or more of the following residues: M457, A492, D499 and Y533. In a preferred embodiment, the variant AAV2 capsid protein comprises one or more of the following amino acid substitutions with respect to a wild type AAV2 capsid protein: Q457M, S492A, E499D and F533Y.
  • the variant AAV2 capsid protein further comprises one or more amino acid substitutions with respect to the wild type AAV capsid protein at the following positions in the AAV2 capsid protein sequence: 125, 151, 162 and 205.
  • the variant AAV2 capsid protein comprises one or more of one or more of the following residues: I125, A151, S162 and S205.
  • the variant AAV2 capsid protein comprises one or more of the following amino acid substitutions with respect to a wild type AAV2 capsid protein: V125I, V151A, A162S and T205S.
  • the variant AAV2 capsid protein further comprises one or more amino acid substitutions with respect to the wild type AAV capsid protein at the following positions in the AAV2 capsid protein sequence: 585 and 588.
  • the variant AAV2 capsid protein comprises one or more of one or more of the following residues: S585 and T588.
  • the variant AAV2 capsid protein comprises one or more of the following amino acid substitutions with respect to a wild type AAV2 capsid protein: R585S and R588T.
  • the variant AAV2 capsid protein further comprises one or more amino acid substitutions with respect to the wild type AAV capsid protein at the following positions in the AAV2 capsid protein sequence: 546, 548 and 593.
  • the variant AAV2 capsid protein comprises one or more of one or more of the following residues: D546, G548, and S593.
  • the variant AAV2 capsid protein comprises one or more of the following amino acid substitutions with respect to a wild type AAV2 capsid protein: G546D, E548G and A593S.
  • the variant AAV2 capsid protein comprises the residue N312, i.e. the residue which is present in the wild type AAV2 capsid protein at position 312. In this embodiment, the variant AAV2 capsid protein is not mutated at position 312 compared to the wild type AAV2 capsid protein sequence.
  • the present invention provides a recombinant adeno-associated virus (AAV) vector comprising: (a) a variant AAV8 capsid protein, wherein the variant AAV8 capsid protein comprises an amino acid substitution with respect to a wild type AAV8 capsid protein at position 315 in an AAV8 capsid protein sequence; and (b) a heterologous nucleic acid comprising a nucleotide sequence encoding a gene product.
  • AAV adeno-associated virus
  • the variant AAV capsid protein comprises a sequence having at least 95% sequence identity to SEQ ID NO:6.
  • the wild type AAV capsid protein comprises a sequence of SEQ ID NO:6.
  • the variant AAV8 capsid protein comprises the amino acid substitution S315N with respect to a wild type AAV8 capsid protein.
  • the AAV8 capsid protein sequence comprises one or more amino acid substitution present at one or more of the following positions: 125, 151, 163, 206, 460, 495, 502, 536, 549, 551, 588, 591 and/or 596.
  • the variant AAV8 capsid protein comprises one or more of the following amino acid substitutions with respect to a wild type AAV8 capsid protein: (a) V125I, Q151A, K163S, A206S, T460M, T495A, N502D, F536Y, N549D, A551G, Q588S and/or G596S; and/or (b) T591R.
  • the present invention provides a recombinant adeno-associated virus (AAV) vector comprising: (a) a variant AAV3B capsid protein, wherein the variant AAV3B capsid protein comprises an amino acid substitution with respect to a wild type AAV3B capsid protein at position 312 in an AAV3B capsid protein sequence; and (b) a heterologous nucleic acid comprising a nucleotide sequence encoding a gene product.
  • AAV adeno-associated virus
  • the variant AAV3B capsid protein comprises a sequence having at least 95% sequence identity to SEQ ID NO:11.
  • the wild type AAV capsid protein comprises a sequence of SEQ ID NO:11.
  • the variant AAV3B capsid protein comprises the amino acid substitution S312N with respect to a wild type AAV3B capsid protein.
  • the present invention provides a recombinant adeno-associated virus (AAV) vector comprising (a) a variant AAV-LK03 capsid protein, wherein the variant AAV-LK03 capsid protein comprises an amino acid substitution at position 312 with respect to a AAV-LK03 capsid protein sequence as defined in SEQ ID NO:12; and (b) a heterologous nucleic acid comprising a nucleotide sequence encoding a gene product.
  • AAV adeno-associated virus
  • the variant AAV-LK03 capsid protein comprises a sequence having at least 95% sequence identity to SEQ ID NO:12.
  • the present invention provides a recombinant adeno-associated virus (AAV) vector comprising: (a) a variant AAV capsid protein, wherein the variant AAV capsid protein comprises at least one amino acid substitution with respect to a wild type AAV capsid protein at a position corresponding to one or more of the following positions in an AAV2 capsid protein sequence: 125, 151, 162, 205, 312, 457, 492, 499, 533, 546, 548, 585, 588 and/or 593; and (b) a heterologous nucleic acid comprising a nucleotide sequence encoding a gene product.
  • AAV adeno-associated virus
  • the at least one amino acid substitution is present at one or more of the following positions in an AAV2 capsid protein sequence: 125, 151, 162, 205, 312, 457, 492, 499, 533, 546, 548, 585, 588 and/or 593; or at one or more corresponding positions in an alternative AAV capsid protein sequence.
  • the vector comprises a variant AAV2 capsid protein.
  • the variant AAV capsid protein comprises a sequence of SEQ ID NO:2, or a sequence having at least 95% sequence identity thereto.
  • the wild type AAV capsid protein is from AAV2.
  • the wild type AAV capsid protein comprises a sequence of SEQ ID NO:1.
  • the variant AAV2 capsid protein comprises one or more of the following residues: I125, A151, S162, S205, S312, M457, A492, D499, Y533, D546, G548, S585, T588 and/or S593.
  • the variant AAV2 capsid protein comprises one or more of the following amino acid substitutions with respect to a wild type AAV2 capsid protein: V125I, V151A, A162S, T205S, N312S, Q457M, S492A, E499D, F533Y, G546D, E548G, R585S, R588T and/or A593S.
  • the variant AAV capsid protein is from AAV1, AAV5, AAV6, AAV8, AAV9 or AAV10.
  • the vector comprises a variant AAV1 capsid protein.
  • the variant AAV capsid protein comprises a sequence having at least 95% sequence identity to SEQ ID NO:3.
  • the wild type AAV capsid protein is from AAV1.
  • the wild type AAV capsid protein comprises a sequence of SEQ ID NO:3.
  • At least one amino acid substitution is present at one or more of the following positions in the AAV1 capsid protein sequence: 125, 151, 162, 205, 313, 458, 493, 500, 534, 547, 549, 586, 589 and/or 594.
  • the variant AAV1 capsid protein comprises one or more of the following amino acid substitutions with respect to a wild type AAV1 capsid protein: V125I, Q151A, T162S, N313S, N458M, K493A, N500D, F534Y, S547D, and/or G594S.
  • the variant AAV1 capsid protein comprises one or more of the following amino acid substitutions with respect to a wild type AAV1 capsid protein: S205T, G549E, S586R and/or T589R.
  • the vector comprises a variant AAV5 capsid protein.
  • the variant AAV capsid protein comprises a sequence having at least 95% sequence identity to SEQ ID NO:4.
  • the wild type AAV capsid protein is from AAV5.
  • the wild type AAV capsid protein comprises a sequence of SEQ ID NO:4.
  • At least one amino acid substitution is present at one or more of the following positions in the AAV5 capsid protein sequence: 124, 150, 153, 195, 303, 444, 479, 486, 520, 533, 537, 575, 578 and/or 583.
  • the variant AAV5 capsid protein comprises one or more of the following amino acid substitutions with respect to a wild type AAV5 capsid protein: V124I, K150A, K153S, A195S, R303S, T444M, S479A, V486D, T520Y, P533D, and/or G583S.
  • the variant AAV5 capsid protein comprises one or more of the following amino acid substitutions with respect to a wild type AAV5 capsid protein: G537E, S575R and/or T578R.
  • the vector comprises a variant AAV6 capsid protein.
  • the variant AAV capsid protein comprises a sequence having at least 95% sequence identity to SEQ ID NO:5.
  • the wild type AAV capsid protein is from AAV6.
  • the wild type AAV capsid protein comprises a sequence of SEQ ID NO:5.
  • At least one amino acid substitution is present at one or more of the following positions in the AAV6 capsid protein sequence: 125, 151, 162, 205, 313, 458, 493, 500, 534, 547, 549, 586, 589 and/or 594.
  • the variant AAV6 capsid protein comprises one or more of the following amino acid substitutions with respect to a wild type AAV6 capsid protein: V125I, Q151A, T162D, N313S, N458M, K493A, N500D, F534Y, S547D, and/or G594S.
  • the variant AAV6 capsid protein comprises one or more of the following amino acid substitutions with respect to a wild type AAV6 capsid protein: S205T, G549E, S586R and/or T589R.
  • the vector comprises a variant AAV8 capsid protein.
  • the variant AAV capsid protein comprises a sequence having at least 95% sequence identity to SEQ ID NO:6.
  • the wild type AAV capsid protein is from AAV8.
  • the wild type AAV capsid protein comprises a sequence of SEQ ID NO:6.
  • At least one amino acid substitution is present at one or more of the following positions in the AAV8 capsid protein sequence: 125, 151, 163, 206, 315, 460, 495, 502, 536, 549, 551, 588, 591 and/or 596.
  • the variant AAV8 capsid protein comprises one or more of the following amino acid substitutions with respect to a wild type AAV8 capsid protein: V125I, Q151A, K163S, A206S, T460M, T495A, N502D, F536Y, N549D, A551G, Q588S and/or G596S.
  • the variant AAV8 capsid protein comprises one or more of the following amino acid substitutions with respect to a wild type AAV8 capsid protein: S315N and/or T591R.
  • the vector comprises a variant AAV9 capsid protein.
  • the variant AAV capsid protein comprises a sequence having at least 95% sequence identity to SEQ ID NO:7.
  • the wild type AAV capsid protein is from AAV9.
  • the wild type AAV capsid protein comprises a sequence of SEQ ID NO:7.
  • At least one amino acid substitution is present at one or more of the following positions in the AAV9 capsid protein sequence: 125, 151, 162, 205, 314, 458, 493, 500, 534, 547, 549, 586, 589 and/or 594.
  • the variant AAV9 capsid protein comprises one or more of the following amino acid substitutions with respect to a wild type AAV9 capsid protein: L125I, Q151A, N314S, Q458M, V493A, E500D, F534Y, G547D, A589T and/or G594S.
  • the variant AAV9 capsid protein comprises one or more of the following amino acid substitutions with respect to a wild type AAV9 capsid protein: S162A, S205T, G549E and/or S586R.
  • the vector comprises a variant AAV10 capsid protein.
  • the variant AAV capsid protein comprises a sequence having at least 95% sequence identity to SEQ ID NO:8.
  • the wild type AAV capsid protein is from AAV10.
  • the wild type AAV capsid protein comprises a sequence of SEQ ID NO:8.
  • the variant AAV10 capsid protein comprises one or more of the following amino acid substitutions with respect to a wild type AAV10 capsid protein: V125I, Q151A, K163S, A206S, N315S, T460M, L495A, N502D, F536Y, G549D, Q588S, A591T and/or G596S.
  • the variant AAV10 capsid protein comprises the following amino acid substitution with respect to a wild type AAV10 capsid protein: G551E.
  • the recombinant AAV vector exhibits increased transduction of a neuronal or retinal tissue compared to an AAV vector comprising a corresponding wild type AAV capsid protein.
  • the recombinant AAV vector exhibits increased transduction of liver tissue compared to a corresponding wild type AAV capsid protein.
  • the gene product comprises an interfering RNA or an aptamer.
  • the gene product comprises a polypeptide.
  • the gene product comprises a neuroprotective polypeptide, an anti-angiogenic polypeptide, or a polypeptide that enhances function of a neuronal or retinal cell.
  • the gene product comprises glial derived neurotrophic factor, fibroblast growth factor, nerve growth factor, brain derived neurotrophic factor, rhodopsin, retinoschisin, RPE65 or peripherin.
  • the present invention provides a pharmaceutical composition
  • a pharmaceutical composition comprising: (a) a recombinant AAV vector as defined above; and (b) a pharmaceutically acceptable excipient.
  • the present invention provides a method for delivering a gene product to a tissue in a subject, the method comprising administering to the subject a recombinant AAV vector or pharmaceutical composition as defined above.
  • the tissue is selected from blood, bone marrow, muscle tissue, neuronal tissue, retinal tissue, pancreatic tissue, liver tissue, kidney tissue, lung tissue, intestinal tissue or heart tissue.
  • the tissue is neuronal, retinal or liver tissue.
  • the present invention provides a method for treating a disorder in a subject, the method comprising administering to the subject a recombinant AAV vector or pharmaceutical composition as defined above.
  • the disorder is a neurological, ocular or hepatic disorder.
  • the present invention provides a recombinant AAV vector or pharmaceutical composition as defined above, for use in treating a disorder in a subject.
  • the disorder is a neurological, ocular or hepatic disorder.
  • the neurological disorder is a neurodegenerative disease.
  • the ocular disorder is glaucoma, retinitis pigmentosa, macular degeneration, retinoschisis or diabetic retinopathy.
  • the present invention provides an isolated variant AAV capsid protein, wherein the variant AAV capsid protein comprises at least one amino acid substitution with respect to a wild type AAV capsid protein; wherein the at least one amino acid substitution is present at one or more of the following positions in an AAV2 capsid protein sequence: 125, 151, 162, 205, 312, 457, 492, 499, 533, 546, 548, 585, 588 and/or 593; or at one or more corresponding positions in an alternative AAV capsid protein sequence.
  • the present invention provides an isolated nucleic acid comprising a nucleotide sequence that encodes a variant AAV capsid protein as defined above.
  • the present invention provides an isolated host cell comprising a nucleic acid as defined above.
  • FIG. 1 shows the amino acid sequence of wild-type adeno-associated virus 2 capsid protein VP1 (SEQ ID NO:1; NCBI Reference Sequence: NC_001401). Residues V125, V151, A162, T205, N312, Q457, S492, E499, F533, G546, E548, R585, R588 and A593 are highlighted.
  • FIG. 2 shows the amino acid sequence of true-type adeno-associated virus 2 (ttAAV2) capsid protein VP1 (SEQ ID NO:2). Residues I125, A151, S162, S205, S312, M457, A492, D499, Y533, D546, G548, S585, T588, S593 differ compared to wild-type AAV2 VP1 (SEQ ID NO:1) and are highlighted.
  • tAAV2 true-type adeno-associated virus 2
  • FIG. 3 shows the amino acid sequence of wild-type adeno-associated virus 1 capsid protein VP1 (SEQ ID NO:3; NCBI Reference Sequence: NC_002077). Highlighted residues: S205 (aligns with S205 in ttAAV2 (SEQ ID NO:2))-G549 (aligns with G548 in ttAAV2)-S586 (aligns with S585 in ttAAV2)-T589 (aligns with T588 in ttAAV2).
  • FIG. 4 shows the amino acid sequence of wild-type adeno-associated virus 5 capsid protein VP1 (SEQ ID NO:4; NCBI Reference Sequence: AF085716). Highlighted residues: G537 (aligns with G548 in ttAAV2)-S575 (aligns with S585 in ttAAV2)-T578 (aligns with T588 in ttAAV2).
  • FIG. 5 shows the amino acid sequence of wild-type adeno-associated virus 6 capsid protein VP1 (SEQ ID NO:5; NCBI Reference Sequence: AF028704). Highlighted residues: S205 (aligns with S205 in ttAAV2)-G549 (aligns with G548 in ttAAV2)-S586 (aligns with S585 in ttAAV2)-T589 (aligns with T588 in ttAAV2).
  • FIG. 6 shows the amino acid sequence of wild-type adeno-associated virus 8 capsid protein VP1 (SEQ ID NO:6; NCBI Reference Sequence: NC_006261). Highlighted residues: S315 (aligns with S312 in ttAAV2)-T591 (aligns with T588 in ttAAV2).
  • FIG. 7 shows the amino acid sequence of wild-type adeno-associated virus 9 capsid protein VP1 (SEQ ID NO:7; NCBI Reference Sequence: AY530579). Highlighted residues: S162 (aligns with S162 in ttAAV2)-S205 (aligns with S205 in ttAAV2)-G549 (aligns with G548 in ttAAV2)-S586 (aligns with S585 in ttAAV2).
  • FIG. 8 shows the amino acid sequence of wild-type adeno-associated virus 10 capsid protein VP1 (SEQ ID NO:8). Highlighted residue: G551 (aligns with G548 in ttAAV2).
  • FIG. 9 shows an alignment of AAV capsid protein VP1 amino acid sequences.
  • FIG. 10 The plasmid used to produce AAV2 vectors was the packaging plasmid pDG. Above: pDG with the wild-type AAV2 genes. Below: pDG-ttAAV2 with the true-type AAV2 genes, highlighted are the two key mutations in the heparan binding domains at positions 585 and 588. MMTV: promoter driving AAV rep expression, E2a, E4ORF6 and VA are the genes expressing adenovirus helper factors.
  • FIG. 11 Quantification of viral titres of rAAV2 true-type (TT) and wild-type (WT) for in vivo injections by SDS-PAGE showing Krypton staining for separated proteins, and scanned using an infrared-fluorescence scanner (Odyssey Imaging systems).
  • A 10 ⁇ lof AAV2 virus particles, and 62.5 ng-500 ng of BSA were separated on a 12% separating gel containing SDS and stained with Krypton Protein Stain. The image was converted to grayscale.
  • the capsid gene proteins VP1, VP2, VP 3 are labelled on the left.
  • B Table showing titres from qPCR (vector genome [vg/ml]) and SDS-Page (capsid titre [capsid/ml]).
  • FIG. 12 A Representative examples of rat brain sections stained with a GFP-specific antibody are shown. The vector was injected into the striatum as shown by the arrow. B. representative example of an injection into the substantia nigra is shown.
  • FIG. 13 GFP transduction of the eye using ttAAV2 and wtAAV2 is shown.
  • FIG. 14 Transduction of mouse brains after neonatal vector injection. i.v., intra-venous vector administration; i.c., intra-cranial injection; AAV-2, wtAAV2; AAV-TT, ttAAV2.
  • FIG. 15 Three-dimensional representation of the AAV2 capsid. The highlighted residues correspond to the amino acid changes between ttAAV2 and wild-type particles, grouped by colour depending on their position.
  • FIG. 16 Representation of a threefold spike on the AAV2 capsid.
  • the highlighted residues correspond to the amino acid changes between True-type and Wild-type particles.
  • the heparin binding site residues are highlighted in green.
  • FIG. 17 Representation of the internal side of the AAV2 capsid.
  • the highlighted residues in light-blue correspond to the single amino acid change in ttAAV2 that is located on the internal side of the capsid.
  • FIG. 18 Representation of a threefold spike on the AAV2 capsid.
  • the residues highlighted in beige correspond to two amino acid changes in the True-type vector that are spatially close and located in the groove between two threefold-proximal peaks on the AAV capsid.
  • FIG. 19 Representation of a threefold spike on the AAV2 capsid.
  • the residue highlighted in brown corresponds to a single isolated amino acid change (S593) in the True-type vector that is located in the groove between threefold-proximal peaks
  • FIG. 20 Representation of a threefold spike on the AAV2 capsid. The four amino acids highlighted in pink are involved in receptor binding and closely situated on the threefold spikes.
  • FIG. 21 Three-dimensional representation of an alignment between VP1 capid monomer from AAV2 (light blue) and VP1 monomer from AAV1 (orange).
  • the highlighted residues in the middle-left of the picture correspond to G549 in AAV1 (orange spheres) and E548 in AAV2 (cyan sphere).
  • the highlighted residues in the top-right of the picture correspond to S586 and T589 in AAV1 (orange spheres) and R585 and R588 in AAV2 (cyan sphere).
  • FIG. 22 Three-dimensional representation of an alignment between VP1 capsid monomer from AAV2 (light blue) and VP1 monomer from AAV5 (purple).
  • the highlighted residues in the middle of the picture correspond to G537 in AAV5 (purple spheres) and E548 in AAV2 (cyan sphere).
  • the highlighted residues in the top-right of the picture correspond to S575 and T578 in AAV5 (purple spheres) and R585 and R588 in AAV2 (cyan sphere).
  • FIG. 23 Three-dimensional representation of an alignment between VP1 capsid monomer from AAV2 (light blue) and VP1 monomer from AAV6 (yellow).
  • the highlighted residues in the bottom of the picture correspond to G549 in AAV6 (orange spheres) and E548 in AAV2 (cyan sphere).
  • the highlighted residues in the top-right of the picture correspond to S586 and T589 in AAV6 (orange spheres) and R585 and R588 in AAV2 (cyan sphere).
  • FIG. 24 Three-dimensional representation of an alignment between VP1 capsid monomer from AAV2 (light blue) and VP1 monomer from AAV8 (pink).
  • the highlighted residues in the top-left of the picture correspond to S315 in AAV8 (red spheres) and N312 in AAV2 (cyan sphere).
  • the highlighted residues in the bottom-right of the picture correspond to T591 in AAV8 (red spheres) and R588 in AAV2 (cyan sphere).
  • FIG. 25 Three-dimensional representation of an alignment between VP1 capsid monomer from AAV2 (light blue) and VP1 monomer from AAV9 (green).
  • the highlighted residues in the middle of the picture correspond to G549 in AAV9 (yellow spheres) and E548 in AAV2 (cyan sphere).
  • the highlighted residues in the bottom-left of the picture correspond to S586 in AAV9 (yellow spheres) and R585 in AAV2 (cyan sphere).
  • FIG. 26 Analysis of rAAV2 TT and WT expression in the parafascicularis nucleus after striatal injection in rat brain.
  • A Representative images of rat brain sections showing the rostral side on the left and the caudal side on the right. The site of injection in the striatum is indicated, and the area of projection in the hypothalamus observed in B and C is shown (parafascicularis nucleus, pf).
  • B and C High magnification images of the GFP expression detected in the parafascicularis nucleus (pf) after striatal injection of rAAV2 WT (B) or TT (C).
  • FIG. 27 Overview of intracranial injections of rAAV2 TT and WT in neonatal mice. Representative examples of neonate brain sections stained with a GFP-specific antibody are shown. 5 ⁇ 10 10 vg of rAAV2 TT (top) or rAAV2 WT (middle) were injected into the lateral ventricle of neonatal mouse brains. An uninjected brain from a neonatal mouse, stained simultaneously, is represented as a negative control (NT, non transduced).
  • NT negative control
  • FIG. 28 High magnification pictures of neonatal mouse brain sections after intracranial injections of rAAV2 TT or WT. Neonate brain sections stained with a GFP-specific antibody are shown. 5 ⁇ 10 10 vg of rAAV2 TT (left panels) or rAAV2 WT (right panels) were injected into the lateral ventricle of neonatal mouse brains. S1BF: barrel field primary somatosensory cortex.
  • FIG. 29 Overview of brain transduction after systemic injection of rAAV2 TT and WT in neonatal mice. Representative examples of neonate brain sections stained with a GFP-specific antibody are shown. 2 ⁇ 10 11 vg of rAAV2 TT (top) or rAAV2 WT (bottom) were injected into the jugular veins of neonatal mice.
  • FIG. 30 High magnification pictures of neonatal mouse brain sections after systemic injections of rAAV2 TT or WT. Neonate brain sections stained with a GFP-specific antibody are shown. 2 ⁇ 10 11 vg of rAAV2 TT (left panels) or rAAV2 WT (right panels) were injected into the jugular veins of neonatal mice. S1BF: barrel field primary somatosensory cortex.
  • FIG. 31 High magnification pictures of neonatal mouse tissue sections after systemic injections of rAAV2 TT or WT. 2 ⁇ 10 11 vg of rAAV2 TT or rAAV2 WT were injected into the jugular veins of neonatal mice. Uninjected mouse organs were used as negative controls.
  • FIG. 32 High magnification images of adult rat brain sections after striatal injections of rAAV2 TT, WT and HBnull. Representative examples of rat brain sections stained with a GFP-specific antibody are shown. 3.5 ⁇ 10 9 vg of rAAV2 WT (left), TT (right) or AAV2-HBnull (middle) were injected into the striatum of adult rat brains and representative pictures were taken in the thalamus or in the substantia nigra (SN).
  • FIG. 33 Overview of intracranial injections of the full AAV-TT compared with various TT mutants in neonatal mice. Representative examples of neonate brain sections stained with a GFP-specific antibody are shown. 5 ⁇ 10 10 vg of rAAV2 TT, TT-S312N, TT-S593A or TT-D546G/G548E (TT-DG) were injected into the lateral ventricle of neonatal mouse brains. An uninjected brain from a neonatal mouse, stained simultaneously, is represented as a negative control (NT).
  • NT negative control
  • FIG. 34 High magnification pictures of neonatal mouse brain sections after intracranial injections of various TT mutant vectors. Neonate brain sections stained with a GFP-specific antibody are shown. 5 ⁇ 10 10 vg of vectors were injected into the lateral ventricle of neonatal mouse brains.
  • TT-DG TT-D546G/G548E.
  • FIG. 35 Overview of neonatal mice intracranial injections of the full AAV-TT compared with the TT-S312N mutant and the potential final TT vector containing 10 mutations.
  • Representative examples of neonate brain sections stained with a GFP-specific antibody are shown.
  • 5 ⁇ 10 09 vg of rAAV2 TT, TT-S312N, TT or TT-S312N-D546G/G548E-S593A (TT-S312N-DG-S593A) were injected into the lateral ventricle of neonatal mouse brains.
  • An uninjected brain from a neonatal mouse, stained simultaneously, is represented as a negative control (NT).
  • FIG. 36 High magnification pictures of neonatal mouse brain sections after intracranial injections of various TT mutant vectors. Neonate brain sections stained with a GFP-specific antibody are shown. 5 ⁇ 10 09 vg of vectors were injected into the lateral ventricle of neonatal mouse brains.
  • FIG. 38 Amino acid sequence of the VP1 capsid protein of AAV3B.
  • the highlighted residues represent the residues that are identical to the ones in AAV-tt at corresponding positions.
  • the internal serine residue at position 312 is underlined.
  • FIG. 39 Amino acid sequence of the VP1 capsid protein of AAV-LK03.
  • SEQ ID NO:1 is the amino acid sequence of wild-type adeno-associated virus 2 capsid protein VP1 (see FIG. 1 ).
  • SEQ ID NO:2 is the amino acid sequence of true-type adeno-associated virus 2 (ttAAV2) capsid protein (see FIG. 2 ).
  • SEQ ID NO:3 is the amino acid sequence of wild-type adeno-associated virus 1 capsid protein VP1 (see FIG. 3 ).
  • SEQ ID NO:4 is the amino acid sequence of wild-type adeno-associated virus 5 capsid protein VP1 (see FIG. 4 ).
  • SEQ ID NO:5 is the amino acid sequence of wild-type adeno-associated virus 6 capsid protein VP1 (see FIG. 5 ).
  • SEQ ID NO:6 is the amino acid sequence of wild-type adeno-associated virus 8 capsid protein VP1 (see FIG. 6 ).
  • SEQ ID NO:7 is the amino acid sequence of wild-type adeno-associated virus 9 capsid protein VP1 (see FIG. 7 ).
  • SEQ ID NO:8 is the amino acid sequence of wild-type adeno-associated virus 10 Upenn capsid protein VP1 (see FIG. 8 ).
  • SEQ ID NO:9 is the amino acid sequence of wild-type adeno-associated virus 10 japanese capsid protein VP1 (see FIG. 9 ).
  • SEQ ID NO:10 is the consensus amino acid sequence for adeno-associated viruses shown in FIG. 9 .
  • SEQ ID NO:11 is the amino acid sequence of wild-type adeno-associated virus 3B capsid protein VP1 (see FIG. 38 ).
  • SEQ ID NO:12 is the amino acid sequence of adeno-associated virus LK-03 capsid protein VP1 (see FIG. 39 ).
  • the present invention relates to a recombinant adeno-associated virus (AAV) vector.
  • the rAAV vector typically comprises a variant capsid protein which differs compared to a wild-type AAV capsid protein.
  • the variant capsid protein may advantageously confer enhanced infectivity of the vector in brain and/or eye, making the vector particularly suited to delivery of therapeutic agents by gene therapy into these tissues.
  • rAAV recombinant adeno-associated virus
  • AAV is an abbreviation for adeno-associated virus, and may be used to refer to the virus itself or derivatives thereof. The term covers all subtypes and both naturally occurring and recombinant forms, except where required otherwise.
  • rAAV refers to recombinant adeno-associated virus, also referred to as a recombinant AAV vector (or “rAAV vector”).
  • AAV includes, 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), AAV type 9 (AAV-9), AAV type 10 (AAV-10, including AAVrh10), AAV type 12 (AAV-12), avian AAV, bovine AAV, canine AAV, equine AAV, primate AAV, non-primate AAV, and ovine AAV.
  • Primarymate AAV refers to AAV that infect primates
  • non-primate AAV refers to AAV that infect non-primate mammals
  • bovine AAV refers to AAV that infect bovine mammals, and so on.
  • rAAV vector refers to an AAV vector comprising a polynucleotide sequence not of AAV origin (i.e., a polynucleotide heterologous to AAV), typically a sequence of interest for the genetic transformation of a cell.
  • the heterologous polynucleotide may be flanked by at least one, and sometimesby two, AAV inverted terminal repeat sequences (ITRs).
  • ITRs AAV inverted terminal repeat sequences
  • the term rAAV vector encompasses both rAAV vector particles and rAAV vector plasmids.
  • An rAAV vector may either be single-stranded (ssAAV) or self-complementary (scAAV).
  • An “AAV virus” or “AAV viral particle” or “rAAV vector particle” refers to a viral particle composed of at least one AAV capsid protein (typically by all of the capsid proteins of a wild-type AAV) and an encapsidated polynucleotide rAAV vector. If the particle comprises a heterologous polynucleotide (i.e. a polynucleotide other than a wild-type AAV genome such as a transgene to be delivered to a mammalian cell), it is typically referred to as an “rAAV vector particle” or simply an “rAAV vector”. Thus, production of rAAV particle necessarily includes production of rAAV vector, as such a vector is contained within an rAAV particle.
  • Recombinant means that the vector, polynucleotide, polypeptide or cell is the product of various combinations of cloning, restriction or ligation steps (e.g. relating to a polynucleotide or polypeptide comprised therein), and/or other procedures that result in a construct that is distinct from a product found in nature.
  • a recombinant virus or vector is a viral particle comprising a recombinant polynucleotide. The terms respectively include replicates of the original polynucleotide construct and progeny of the original virus construct.
  • the rAAV vectors described herein comprise a variant AAV capsid protein.
  • variant it is meant that the AAV capsid protein differs from a corresponding wild type AAV capsid protein of the same serotype.
  • the variant AAV capsid protein may comprise one or more amino acid substitutions with respect to the corresponding wild type AAV capsid protein.
  • corresponding refers to a capsid protein of the same serotype, i.e.
  • a variant AAV1 capsid protein comprises one or more amino acid substitutions with respect to the corresponding wild type AAV1 capsid protein
  • a variant AAV2 capsid protein comprises one or more amino acid substitutions with respect to the corresponding wild type AAV2 capsid protein, and so on.
  • the variant AAV capsid protein may comprise, for example, 1 to 50, 1 to 30, 1 to 20 or 1 to 15 amino acid substitutions with respect to the wild type AAV capsid protein.
  • the variant AAV capsid protein comprises 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13 or 14 amino acid substitutions with respect to the corresponding wild type AAV capsid protein.
  • the variant AAV capsid protein retains at least 70%, at least 80%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98% or at least 99% sequence identity to the wild type capsid protein.
  • the variant AAV capsid protein comprises at least one amino acid substitution with respect to a wild type AAV capsid protein at a position corresponding to one or more of the following positions in an AAV2 capsid protein sequence: 125, 151, 162, 205, 312, 457, 492, 499, 533, 546, 548, 585, 588 and/or 593.
  • “corresponding” refers to a position in any AAV capsid protein sequence (e.g. in an AAV2 protein sequence or a non-AAV2 capsid protein sequence) which corresponds to one of the above positions in AAV2 capsid protein.
  • the at least one amino acid substitution is present at one or more of the following positions in an AAV2 capsid protein sequence: 125, 151, 162, 205, 312, 457, 492, 499, 533, 546, 548, 585, 588 and/or 593; or at one or more corresponding positions in an alternative AAV capsid protein sequence.
  • AAV capsid proteins include VP1, VP2 and VP3.
  • the capsid protein comprises AAV capsid protein VP1.
  • polynucleotide refers to a polymeric form of nucleotides of any length, including deoxyribonucleotides or ribonucleotides, or analogs thereof.
  • a polynucleotide may comprise modified nucleotides, such as methylated nucleotides and nucleotide analogs, and may be interrupted by non-nucleotide components. If present, modifications to the nucleotide structure may be imparted before or after assembly of the polymer.
  • polynucleotide refers interchangeably to double- and single-stranded molecules. Unless otherwise specified or required, any embodiment of the invention described herein that is a polynucleotide encompasses both the double-stranded form and each of two complementary single-stranded forms known or predicted to make up the double-stranded form.
  • polypeptide refers to polymers of amino acids of any length.
  • the terms also encompass an amino acid polymer that has been modified; for example, disulfide bond formation, glycosylation, lipidation, phosphorylation, or conjugation with a labeling component.
  • Polypeptides such as anti-angiogenic polypeptides, neuroprotective polypeptides, and the like, when discussed in the context of delivering a gene product to a mammalian subject, and compositions therefor, refer to the respective intact polypeptide, or any fragment or genetically engineered derivative thereof, which retains the desired biochemical function of the intact protein.
  • references to nucleic acids encoding anti-angiogenic polypeptides, nucleic acids encoding neuroprotective polypeptides, and other such nucleic acids for use in delivery of a gene product to a mammalian subject include polynucleotides encoding the intact polypeptide or any fragment or genetically engineered derivative possessing the desired biochemical function.
  • a polynucleotide or polypeptide has a certain percent “sequence identity” to another polynucleotide or polypeptide, meaning that, when aligned, that percentage of bases or amino acids are the same when comparing the two sequences. Sequence similarity can be determined in a number of different manners. To determine sequence identity, sequences can be aligned using the methods and computer programs, including BLAST, available over the world wide web at ncbi.nlm.nih.gov/BLAST/. Another alignment algorithm is FASTA, available in the Genetics Computing Group (GCG) package, from Madison, Wis., USA, a wholly owned subsidiary of Oxford Molecular Group, Inc. Other techniques for alignment are described in Methods in Enzymology, vol.
  • GCG Genetics Computing Group
  • the program has default parameters determined by the sequences inputted to be compared.
  • the sequence identity is determined using the default parameters determined by the program. This program is available also from Genetics Computing Group (GCG) package, from Madison, Wis., USA.
  • GCG Genetics Computing Group
  • FastDB is described in Current Methods in Sequence Comparison and Analysis, Macromolecule Sequencing and Synthesis, Selected Methods and Applications, pp. 127-149, 1988, Alan R. Liss, Inc. Percent sequence identity is calculated by FastDB based upon the following parameters:
  • the vector comprises a variant AAV2 capsid protein.
  • the variant AAV2 capsid protein comprises at least one amino acid substitution at one or more of the following positions in an AAV2 capsid protein sequence: 125, 151, 162, 205, 312, 457, 492, 499, 533, 546, 548, 585, 588 and/or 593.
  • Wild type AAV2 capsid protein VP1 is known, and is shown in FIG. 1 (SEQ ID NO:1). Wild type AAV2 capsid protein sequences are also available from database accession no.s: NC-001401; UniProt P03135; NCBI Reference Sequence: YP_680426.1; GenBank: AAC03780.1.
  • the variant AAV2 capsid protein has at least 70%, at least 80%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98% or at least 99% sequence identity to SEQ ID NO:1.
  • the variant AAV2 capsid protein comprises a sequence of SEQ ID NO:2, or a sequence having at least 70%, at least 80%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98% or at least 99% sequence identity thereto.
  • the variant AAV2 capsid protein comprises one or more of the following residues: I125, A151, S162, S205, S312, M457, A492, D499, Y533, D546, G548, S585, T588 and/or S593.
  • the variant AAV2 capsid protein comprises one or more of the following amino acid substitutions with respect to a wild type AAV2 capsid protein: V125I, V151A, A162S, T205S, N312S, Q457M, S492A, E499D, F533Y, G546D, E548G, R585S, R588T and/or A593S.
  • the variant AAV2 capsid protein may comprise any combination of the above amino acid substitutions. Therefore in particular embodiments, the variant AAV2 capsid protein comprises 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13 or 14 amino acid substitutions selected from the list above. In one embodiment, the variant AAV2 capsid protein comprises all 14 amino acid substitutions disclosed above, e.g. the variant AAV2 capsid protein comprises a sequence of SEQ ID NO:2 (i.e. ttAAV2 or AAV2-TT as referred to herein).
  • the variant AAV2 capsid protein may comprise a sub-set of the above 14 mutations.
  • the variant AAV2 capsid protein may comprise the following residues, which are divided below into functional groups:
  • the variant AAV2 capsid protein comprises four or more mutations at the positions mentioned above which may be associated with receptor binding, i.e. residues 457, 492, 499 and 533.
  • residues 457, 492, 499 and 533 may be associated with receptor binding, i.e. residues 457, 492, 499 and 533.
  • the variant AAV2 capsid protein comprises the following residues M457, A492, D499 and Y533.
  • the variant AAV2 capsid protein is not mutated with respect to the wild type AAV2 capsid protein at position 312, e.g. the variant AAV2 capsid protein comprises the residue N312 (which is present in the wild type AAV2 capsid protein).
  • the variant AAV2 capsid protein may comprise 1 to 13 of the specific mutations mentioned above, but not the mutation N312S.
  • the variant AAV capsid protein is from an alternative AAV serotype, i.e. an AAV serotype other than AAV2.
  • the variant AAV capsid protein may be derived from an AAV1, AAV3B, AAV-LK03, AAV5, AAV6, AAV8, AAV9 or AAV10 (e.g. AAVrh10) capsid protein.
  • the variant AAV capsid protein comprises at least one amino acid substitution at one or more positions corresponding to those described above with respect to AAV2.
  • the variant AAV capsid protein comprises at least one amino acid substitution at a position in an alternative (i.e. non-AAV2) AAV capsid protein sequence which corresponds to positions 125, 151, 162, 205, 312, 457, 492, 499, 533, 546, 548, 585, 588 and/or 593 in an AAV2 capsid protein sequence.
  • positions in alternative AAV capsid protein sequences which correspond in three-dimensional space to positions 125, 151, 162, 205, 312, 457, 492, 499, 533, 546, 548, 585, 588 and/or 593 in an AAV2 capsid protein.
  • Methods for three-dimensional modelling and alignment of protein structures are well known in the art, and can be used to identify such corresponding positions in non-AAV2 capsid protein sequences.
  • Exemplary 3D alignments of AAV2 capsid protein sequences with capsid protein sequences of alternative AAV serotypes are shown in FIGS. 21 to 25 and discussed below.
  • a skilled person can perform similar 3D alignments with capsid proteins from further serotypes, e.g. AAV2, AAV3, AAV7, AAV10 and AAV12), and identify positions in such sequences which correspond with to the positions defined above in AAV2.
  • the vector comprises a variant AAV1 capsid protein.
  • the variant AAV1 capsid protein comprises at least one amino acid substitution at one or more of the following positions in the AAV1 capsid protein sequence: 125, 151, 162, 205, 313, 458, 493, 500, 534, 547, 549, 586, 589 and/or 594. These positions in AAV1 capsid protein VP1 correspond to those disclosed above in relation to AAV2.
  • wild type AAV1 capsid protein VP1 The sequence of wild type AAV1 capsid protein VP1 is known, and is shown in FIG. 3 (SEQ ID NO:3).
  • a wild type AAV1 capsid protein sequences is also available from database accession no.: NC-002077.
  • the variant AAV1 capsid protein has at least 70%, at least 80%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98% or at least 99% sequence identity to SEQ ID NO:3.
  • Wild type AAV1 capsid protein VP1 already contains the following residues at positions which correspond to amino acid residues which are present in the variant AAV2 capsid protein disclosed above (SEQ ID NO:2, ttAAV2), but not wild type AAV2 (SEQ ID NO:1): S205 (aligns with S205 in ttAAV2); G549 (aligns with G548 in ttAAV2); S586 (aligns with S585 in ttAAV2); and T589 (aligns with T588 in ttAAV2).
  • the variant AAV1 capsid protein comprises one or more of the following amino acid substitutions with respect to a wild type AAV1 capsid protein: V125I, Q151A, T162S, N313S, N458M, K493A, N500D, F534Y, S547D, and/or G594S.
  • a variant AAV1 capsid protein may share one or more functional properties with the variant AAV2 capsid protein (SEQ ID NO:2, ttAAV2), e.g. may confer increased infectivity and/or transduction of neuronal of retinal tissue compared to wild type AAV1 capsid protein.
  • the variant AAV1 capsid protein comprises one or more amino acid substitutions which correspond to reversions of mutations present in ttAAV2 back to the wild type AAV2 sequence.
  • the variant AAV1 capsid protein may comprise one or more of the following substitutions: S205T, G549E, S586R and/or T589R.
  • such a variant AAV1 capsid protein may share one or more functional properties with the wild type AAV2 capsid protein (SEQ ID NO:1), e.g. may confer reduced infectivity and/or transduction of neuronal of retinal tissue compared to wild type AAV1 capsid protein.
  • the vector comprises a variant AAV5 capsid protein.
  • the variant AAV5 capsid protein comprises at least one amino acid substitution at one or more of the following positions in the AAV5 capsid protein sequence: 124, 150, 153, 195, 303, 444, 479, 486, 520, 533, 537, 575, 578 and/or 583. These positions in AAV5 capsid protein VP1 correspond to those disclosed above in relation to AAV2.
  • a wild type AAV5 capsid protein VP1 is known, and is shown in FIG. 4 (SEQ ID NO:4).
  • a wild type AAV5 capsid protein sequences is also available from database accession no.: AF085716.
  • the variant AAV5 capsid protein has at least 70%, at least 80%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98% or at least 99% sequence identity to SEQ ID NO:4.
  • Wild type AAV5 capsid protein VP1 already contains the following residues at positions which correspond to amino acid residues which are present in the variant AAV2 capsid protein disclosed above (SEQ ID NO:2, ttAAV2), but not wild type AAV2 (SEQ ID NO:1): G537 (aligns with G548 in ttAAV2); S575 (aligns with S585 in ttAAV2); T578 (aligns with T588 in ttAAV2).
  • the variant AAV5 capsid protein comprises one or more of the following amino acid substitutions with respect to a wild type AAV5 capsid protein: V124I, K150A, K1535, A195S, R3035, T444M, S479A, V486D, T520Y, P533D, and/or G583S.
  • a variant AAV5 capsid protein may share one or more functional properties with the variant AAV2 capsid protein (SEQ ID NO:2, ttAAV2), e.g. may confer increased infectivity and/or transduction of neuronal of retinal tissue compared to wild type AAV5 capsid protein.
  • the variant AAV5 capsid protein comprises one or more amino acid substitutions which correspond to reversions of mutations present in ttAAV2 back to the wild type AAV2 sequence.
  • the variant AAV5 capsid protein may comprise one or more of the following substitutions: G537E, S575R and/or T578R.
  • a variant AAV5 capsid protein may share one or more functional properties with the wild type AAV2 capsid protein (SEQ ID NO:1), e.g. may confer reduced infectivity and/or transduction of neuronal of retinal tissue compared to wild type AAV5 capsid protein.
  • the vector comprises a variant AAV6 capsid protein.
  • the variant AAV6 capsid protein comprises at least one amino acid substitution at one or more of the following positions in the AAV6 capsid protein sequence: 125, 151, 162, 205, 313, 458, 493, 500, 534, 547, 549, 586, 589 and/or 594. These positions in AAV6 capsid protein VP1 correspond to those disclosed above in relation to AAV2.
  • wild type AAV6 capsid protein VP1 The sequence of wild type AAV6 capsid protein VP1 is known, and is shown in FIG. 5 (SEQ ID NO:5).
  • a wild type AAV6 capsid protein sequences is also available from database accession no.: AF028704.
  • the variant AAV6 capsid protein has at least 70%, at least 80%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98% or at least 99% sequence identity to SEQ ID NO:5.
  • Wild type AAV6 capsid protein VP1 already contains the following residues at positions which correspond to amino acid residues which are present in the variant AAV2 capsid protein disclosed above (SEQ ID NO:2, ttAAV2), but not wild type AAV2 (SEQ ID NO:1): S205 (aligns with S205 in ttAAV2); G549 (aligns with G548 in ttAAV2); S586 (aligns with S585 in ttAAV2); T589 (aligns with T588 in ttAAV2).
  • the variant AAV6 capsid protein comprises one or more of the following amino acid substitutions with respect to a wild type AAV6 capsid protein: V125I, Q151A, T162S, N313S, N458M, K493A, N500D, F534Y, S547D, and/or G594S.
  • a variant AAV6 capsid protein may share one or more functional properties with the variant AAV2 capsid protein (SEQ ID NO:2, ttAAV2), e.g. may confer increased infectivity and/or transduction of neuronal of retinal tissue compared to wild type AAV6 capsid protein.
  • the variant AAV6 capsid protein comprises one or more amino acid substitutions which correspond to reversions of mutations present in ttAAV2 back to the wild type AAV2 sequence.
  • the variant AAV6 capsid protein may comprise one or more of the following substitutions: S205T, G549E, S586R and/or T589R.
  • such a variant AAV6 capsid protein may share one or more functional properties with the wild type AAV2 capsid protein (SEQ ID NO:1), e.g. may confer reduced infectivity and/or transduction of neuronal of retinal tissue compared to wild type AAV6 capsid protein.
  • the vector comprises a variant AAV8 capsid protein.
  • the variant AAV8 capsid protein comprises at least one amino acid substitution at one or more of the following positions in the AAV8 capsid protein sequence: 125, 151, 163, 206, 315, 460, 495, 502, 536, 549, 551, 588, 591 and/or 596. These positions in AAV8 capsid protein VP1 correspond to those disclosed above in relation to AAV2.
  • wild type AAV8 capsid protein VP1 The sequence of wild type AAV8 capsid protein VP1 is known, and is shown in FIG. 6 (SEQ ID NO:6).
  • a wild type AAV8 capsid protein sequences is also available from database accession no.: NC_006261.
  • the variant AAV8 capsid protein has at least 70%, at least 80%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98% or at least 99% sequence identity to SEQ ID NO:6.
  • Wild type AAV8 capsid protein VP1 already contains the following residues at positions which correspond to amino acid residues which are present in the variant AAV2 capsid protein disclosed above (SEQ ID NO:2, ttAAV2), but not wild type AAV2 (SEQ ID NO:1): S315 (aligns with S312 in ttAAV2); T591 (aligns with T588 in ttAAV2).
  • the variant AAV8 capsid protein comprises one or more of the following amino acid substitutions with respect to a wild type AAV8 capsid protein: V125I, Q151A, K163S, A206S, T460M, T495A, N502D, F536Y, N549D, A551G, Q588S and/or G596S.
  • a variant AAV8 capsid protein may share one or more functional properties with the variant AAV2 capsid protein (SEQ ID NO:2, ttAAV2), e.g. may confer increased infectivity and/or transduction of neuronal of retinal tissue compared to wild type AAV8 capsid protein.
  • the variant AAV8 capsid protein comprises one or more amino acid substitutions which correspond to reversions of mutations present in ttAAV2 back to the wild type AAV2 sequence.
  • the variant AAV8 capsid protein may comprise one or more of the following substitutions: S315N and/or T591R.
  • such a variant AAV8 capsid protein may share one or more functional properties with the wild type AAV2 capsid protein (SEQ ID NO:1), e.g. may confer reduced infectivity and/or transduction of neuronal of retinal tissue compared to wild type AAV8 capsid protein.
  • the variant AAV8 capsid protein comprises an amino acid substitution with respect to a wild type AAV8 capsid protein at position 315 in an AAV8 capsid protein sequence.
  • the variant AAV8 capsid protein may comprise the residue N315.
  • the variant AAV8 capsid protein comprises the amino acid substitution S315N with respect to a wild type AAV8 capsid protein.
  • the vector comprises a variant AAV9 capsid protein.
  • the variant AAV9 capsid protein comprises at least one amino acid substitution at one or more of the following positions in the AAV9 capsid protein sequence: 125, 151, 162, 205, 314, 458, 493, 500, 534, 547, 549, 586, 589 and/or 594. These positions in AAV9 capsid protein VP1 correspond to those disclosed above in relation to AAV2.
  • wild type AAV9 capsid protein VP1 The sequence of wild type AAV9 capsid protein VP1 is known, and is shown in FIG. 7 (SEQ ID NO:7).
  • a wild type AAV9 capsid protein sequences is also available from database accession no.: AY530579.
  • the variant AAV9 capsid protein has at least 70%, at least 80%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98% or at least 99% sequence identity to SEQ ID NO:7.
  • Wild type AAV9 capsid protein VP1 already contains the following residues at positions which correspond to amino acid residues which are present in the variant AAV2 capsid protein disclosed above (SEQ ID NO:2, ttAAV2), but not wild type AAV2 (SEQ ID NO:1): S162 (aligns with S162 in ttAAV2); S205 (aligns with S205 in ttAAV2); G549 (aligns with G548 in ttAAV2); S586 (aligns with S585 in ttAAV2).
  • the variant AAV9 capsid protein comprises one or more of the following amino acid substitutions with respect to a wild type AAV9 capsid protein: L125S, Q151A, N314S, Q458M, V493A, E500D, F534Y, G547D, A589T and/or G594S.
  • a variant AAV9 capsid protein may share one or more functional properties with the variant AAV2 capsid protein (SEQ ID NO:2, ttAAV2), e.g. may confer increased infectivity and/or transduction of neuronal of retinal tissue compared to wild type AAV9 capsid protein.
  • the variant AAV9 capsid protein comprises one or more amino acid substitutions which correspond to reversions of mutations present in ttAAV2 back to the wild type AAV2 sequence.
  • the variant AAV9 capsid protein may comprise one or more of the following substitutions: S162A, S205T, G549E and/or S586R.
  • SEQ ID NO:1 the wild type AAV2 capsid protein (SEQ ID NO:1), e.g. may confer reduced infectivity and/or transduction of neuronal of retinal tissue compared to wild type AAV9 capsid protein.
  • the vector comprises a variant AAV10 capsid protein.
  • AAV10 includes AAVrh10.
  • the variant AAV10 (e.g. AAVrh10) capsid protein comprises at least one amino acid substitution at one or more of the following positions in the AAV10 capsid protein sequence: 125, 151, 163, 206, 315, 460, 495, 502, 536, 549, 551, 588, 591 and/or 596. These positions in AAV10 capsid protein VP1 correspond to those disclosed above in relation to AAV2.
  • the sequence of wild type AAV10 capsid protein VP1 is known, and is shown in FIG. 8 (SEQ ID NO:8).
  • the variant AAV10 capsid protein has at least 70%, at least 80%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98% or at least 99% sequence identity to SEQ ID NO:8.
  • Wild type AAV10 capsid protein VP1 already contains the following residue at a position which corresponds to an amino acid residue which is present in the variant AAV2 capsid protein disclosed above (SEQ ID NO:2, ttAAV2), but not wild type AAV2 (SEQ ID NO:1): G551 (aligns with G548 in ttAAV2).
  • the variant AAV10 capsid protein comprises one or more of the following amino acid substitutions with respect to a wild type AAV10 capsid protein: V125I, Q151A, K163S, A206S, N315S, T460M, L495A, N502D, F536Y, G549D, Q588S, A591T and/or G596S.
  • a variant AAV10 capsid protein may share one or more functional properties with the variant AAV2 capsid protein (SEQ ID NO:2, ttAAV2), e.g. may confer increased infectivity and/or transduction of neuronal of retinal tissue compared to wild type AAV10 capsid protein.
  • the variant AAV10 capsid protein comprises an amino acid substitution which corresponds to a reversion of a mutations present in ttAAV2 back to the wild type AAV2 sequence.
  • the variant AAV10 capsid protein may comprise the following substitution: G551E.
  • such a variant AAV10 capsid protein may share one or more functional properties with the wild type AAV2 capsid protein (SEQ ID NO:1), e.g. may confer reduced infectivity and/or transduction of neuronal of retinal tissue compared to wild type AAV10 capsid protein.
  • the vector comprises a variant AAV3B capsid protein.
  • the variant AAV3B capsid protein may comprise an amino acid substitution with respect to a wild type AAV3B capsid protein at position 312.
  • the variant AAV3B capsid protein may comprise the residue N312.
  • the variant AAV8 capsid protein comprises the amino acid substitution S312N with respect to a wild type AAV8 capsid protein.
  • the variant AAV3B capsid protein may comprise one or more additional mutations at positions which correspond to those disclosed above in relation to AAV2.
  • a wild type AAV3B capsid protein VP1 is known, and is shown in FIG. 38 (SEQ ID NO:11).
  • a wild type AAV3B capsid protein sequence is also available from NCBI database accession no. AF028705.
  • the variant AAV3B capsid protein has at least 70%, at least 80%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98% or at least 99% sequence identity to SEQ ID NO:11.
  • the vector comprises a variant AAV-LK03 capsid protein.
  • the variant AAV-LK03 capsid protein may comprise an amino acid substitution at position 312 with respect to a AAV-LK03 capsid protein sequence as defined in SEQ ID NO:12.
  • the variant AAV-LK03 capsid protein may comprise the residue N312.
  • the variant AAV-LK03 capsid protein comprises the amino acid substitution S312N with respect to a AAV-LK03 capsid protein sequence as defined in SEQ ID NO:12.
  • the variant AAV-LK03 capsid protein may comprise one or more additional mutations at positions which correspond to those disclosed above in relation to AAV2.
  • a AAV-LK03 capsid protein sequence is also disclosed in WO 2013/029030 as sequence number 31 therein.
  • the variant AAV-LK03 capsid protein has at least 70%, at least 80%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98% or at least 99% sequence identity to SEQ ID NO:12.
  • the rAAV further comprises a heterologous nucleic acid comprising a nucleotide sequence encoding a gene product.
  • a “gene” refers to a polynucleotide containing at least one open reading frame that is capable of encoding a particular protein after being transcribed and translated.
  • a “gene product” is a molecule resulting from expression of a particular gene. Gene products include, e.g., a polypeptide, an aptamer, an interfering RNA, an mRNA, and the like.
  • Heterologous means derived from a genotypically distinct entity from that of the rest of the entity to which it is being compared.
  • a polynucleotide introduced by genetic engineering techniques into a plasmid or vector derived from a different species is a heterologous polynucleotide.
  • a promoter removed from its native coding sequence and operatively linked to a coding sequence with which it is not naturally found linked is a heterologous promoter.
  • an rAAV that includes a heterologous nucleic acid encoding a heterologous gene product is an rAAV that includes a nucleic acid not normally included in a naturally-occurring, wild-type AAV, and the encoded heterologous gene product is a gene product not normally encoded by a naturally-occurring, wild-type AAV.
  • the gene product is an interfering RNA. In some embodiments, the gene product is an aptamer. In some embodiments, the gene product is a polypeptide.
  • RNAi interfering RNA
  • suitable RNAi include RNAi that decrease the level of an apoptotic or angiogenic factor in a cell.
  • an RNAi can be an shRNA or siRNA that reduces the level of a gene product that induces or promotes apoptosis in a cell.
  • Genes whose gene products induce or promote apoptosis are referred to herein as “pro-apoptotic genes” and the products of those genes (mRNA; protein) are referred to as “pro-apoptotic gene products.”
  • Pro-apoptotic gene products include, e.g., Bax, Bid, Bak, and Bad gene products. See, e.g., U.S. Pat. No. 7,846,730.
  • Interfering RNAs could also be against an angiogenic product, for example VEGF (e.g., Cand5; see, e.g., U.S. Patent Publication No. 2011/0143400; U.S. Patent Publication No. 2008/0188437; and Reich et al. (2003) Mol. Vis. 9:210), VEGFR1 (e.g., Sirna-027; see, e.g., Kaiser et al. (2010) Am. J. Ophthalmol. 150:33; and Shen et al. (2006) Gene Ther. 13:225), or VEGFR2 (Kou et al. (2005) Biochem. 44: 15064). See also, U.S. Pat. Nos. 6,649,596, 6,399,586, 5,661,135, 5,639,872, and 5,639,736; and U.S. Pat. Nos. 7,947,659 and 7,919,473.
  • VEGF e.g., Cand5; see,
  • a “small interfering” or “short interfering RNA” or siRNA is a RNA duplex of nucleotides that is targeted to a gene interest (a “target gene”).
  • An “RNA duplex” refers to the structure formed by the complementary pairing between two regions of a RNA molecule.
  • siRNA is “targeted” to a gene in that the nucleotide sequence of the duplex portion of the siRNA is complementary to a nucleotide sequence of the targeted gene.
  • the length of the duplex of siRNAs is less than 30 nucleotides.
  • the duplex can be 29, 28, 27, 26, 25, 24, 23, 22, 21, 20, 19, 18, 17, 16, 15, 14, 13, 12, 11 or 10 nucleotides in length.
  • the length of the duplex is 19-25 nucleotides in length.
  • the RNA duplex portion of the siRNA can be part of a hairpin structure.
  • the hairpin structure may contain a loop portion positioned between the two sequences that form the duplex.
  • the loop can vary in length. In some embodiments the loop is 5, 6, 7, 8, 9, 10, 11, 12 or 13 nucleotides in length.
  • the hairpin structure can also contain 3′ or 5′ overhang portions. In some embodiments, the overhang is a 3′ or a 5′ overhang 0, 1, 2, 3, 4 or 5 nucleotides in length.
  • shRNA is a polynucleotide construct that can be made to express an interfering RNA such as siRNA.
  • exemplary aptamers of interest include an aptamer against vascular endothelial growth factor (VEGF).
  • VEGF vascular endothelial growth factor
  • a PDGF-specific aptamer e.g., E10030; see, e.g., Ni and Hui (2009) Ophthalmologica 223:401; and Akiyama et al. (2006) J. Cell Physiol. 207:407).
  • the gene product is a therapeutic protein.
  • a “therapeutic” peptide or protein is a peptide or protein that may alleviate or reduce symptoms that result from an absence or defect in a protein in a cell or subject.
  • a “therapeutic” peptide or protein is one that otherwise confers a benefit to a subject, e.g., anti-degenerative effects.
  • the polypeptide is generally a polypeptide that enhances function of a cell, for example a cell present in neuronal, retinal or liver tissue, e.g., a hepatocyte, a neuron, a glial cell, a rod or cone photoreceptor cell, a retinal ganglion cell, a Muller cell, a bipolar cell, an amacrine cell, a horizontal cell, or a retinal pigmented epithelial cell.
  • a cell present in neuronal, retinal or liver tissue e.g., a hepatocyte, a neuron, a glial cell, a rod or cone photoreceptor cell, a retinal ganglion cell, a Muller cell, a bipolar cell, an amacrine cell, a horizontal cell, or a retinal pigmented epithelial cell.
  • Exemplary polypeptides include neuroprotective polypeptides (e.g., GDNF, CNTF, NT4, NGF, and NTN); anti-angiogenic polypeptides (e.g., a soluble vascular endothelial growth factor (VEGF) receptor; a VEGF-binding antibody; a VEGF-binding antibody fragment (e.g., a single chain anti-VEGF antibody); endostatin; tumstatin; angiostatin; a soluble Fit polypeptide (Lai et al. (2005) Mol. Ther. 12:659); an Fc fusion protein comprising a soluble Fit polypeptide (see, e.g., Pechan et al. (2009) Gene Ther.
  • VEGF vascular endothelial growth factor
  • PEDF pigment epithelium-derived factor
  • TMP-3 tissue inhibitor of metalloproteinases-3
  • opsin e.g., a rhodopsin
  • anti- apoptotic polypeptides e.g., Bcl-2, Bcl-X1; and the like.
  • Suitable polypeptides include, but are not limited to, glial derived neurotrophic factor (GDNF); fibroblast growth factor 2; neurturin (NTN); ciliary neurotrophic factor (CNTF); nerve growth factor (NGF); neurotrophin-4 (NT4); brain derived neurotrophic factor (BDNF); epidermal growth factor; rhodopsin; X-linked inhibitor of apoptosis; and Sonic hedgehog.
  • GDNF glial derived neurotrophic factor
  • NTN fibroblast growth factor 2
  • CNTF ciliary neurotrophic factor
  • NTF nerve growth factor
  • NT4 neurotrophin-4
  • BDNF brain derived neurotrophic factor
  • epidermal growth factor rhodopsin
  • X-linked inhibitor of apoptosis and Sonic hedgehog.
  • Exemplary polypeptides for gene deliver to the liver include, for example, PBGD (porphobilinogen deaminase) IDUA (iduronidase) Fah (fumarylacetoacetate hydrolyase) A1AT (alpha(1)-antitrypsin), 1A1(hUGT1A1) (uridine disphoshate glucuronyltransferase), HCCS1 (hepatocellular carcinoma suppressor 1), CD (cytosine deaminase), SOCS3 (suppressor of cytokine signaling 3), TNF (tumor necrosis factor), thymidine kinase, IL-24 (interleukin-24), IL-12 (interleukin-12), and TRAIL (tumor necrosis factor-related apoptosis-inducing ligand).
  • a nucleotide sequence encoding a gene product of interest is operably linked to a constitutive promoter. In other embodiments, a nucleotide sequence encoding a gene product of interest is operably linked to an inducible promoter. In some instances, a nucleotide sequence encoding a gene product of interest is operably linked to a tissue specific or cell type specific regulatory element.
  • a nucleotide sequence encoding a gene product of interest is operably linked to a hepatocyte-specific, neuron-specific or photoreceptor-specific regulatory element (e.g., a photoreceptor-specific promoter), e.g., a regulatory element that confers selective expression of the operably linked gene in a neuron or photoreceptor cell.
  • a photoreceptor-specific regulatory element include, e.g., a rhodopsin promoter; a rhodopsin kinase promoter (Young et al. (2003) Ophthalmol. Vis. Sci. 44:4076); a beta phosphodiesterase gene promoter (Nicoud et al.
  • Suitable neuronal-specific promoters include neuron-specific enolase (NSE) promoter, Andersen et al. Cell. Mol. Neurobiol., 13:503-15 (1993; neurofilament light-chain gene promoter, Piccioli et al., Proc. Natl. Acad. Sci.
  • hepatocyte-specific promoters include an albumin promoter (Heard et al., Mol Cell Biol 1987; 7: 2425) or an alpha 1-antitrypsin promoter (Hafenrichter et al. Blood 1994; 84, 3394-404).
  • control element or “control sequence” is a nucleotide sequence involved in an interaction of molecules that contributes to the functional regulation of a polynucleotide, including replication, duplication, transcription, splicing, translation, or degradation of the polynucleotide. The regulation may affect the frequency, speed, or specificity of the process, and may be enhancing or inhibitory in nature.
  • Control elements known in the art include, for example, transcriptional regulatory sequences such as promoters and enhancers.
  • a promoter is a DNA region capable under certain conditions of binding RNA polymerase and initiating transcription of a coding region usually located downstream (in the 3′ direction) from the promoter.
  • “Operatively linked” or “operably linked” refers to a juxtaposition of genetic elements, wherein the elements are in a relationship permitting them to operate in the expected manner. For instance, a promoter is operatively linked to a coding region if the promoter helps initiate transcription of the coding sequence. There may be intervening residues between the promoter and coding region so long as this functional relationship is maintained.
  • promoter can refer to a DNA sequence that is located adjacent to a DNA sequence that encodes a recombinant product.
  • a promoter is preferably linked operatively to an adjacent DNA sequence.
  • a promoter typically increases an amount of recombinant product expressed from a DNA sequence as compared to an amount of the expressed recombinant product when no promoter exists.
  • a promoter from one organism can be utilized to enhance recombinant product expression from a DNA sequence that originates from another organism.
  • a vertebrate promoter may be used for the expression of jellyfish GFP in vertebrates.
  • one promoter element can increase an amount of recombinant products expressed for multiple DNA sequences attached in tandem. Hence, one promoter element can enhance the expression of one or more recombinant products.
  • Multiple promoter elements are well-known to persons of ordinary skill in the art.
  • Enhancer elements can refer to a DNA sequence that is located adjacent to the DNA sequence that encodes a recombinant product.
  • Enhancer elements are typically located upstream of a promoter element or can be located downstream of or within a coding DNA sequence (e.g., a DNA sequence transcribed or translated into a recombinant product or products).
  • a coding DNA sequence e.g., a DNA sequence transcribed or translated into a recombinant product or products.
  • an enhancer element can be located 100 base pairs, 200 base pairs, or 300 or more base pairs upstream or downstream of a DNA sequence that encodes recombinant product.
  • Enhancer elements can increase an amount of recombinant product expressed from a DNA sequence above increased expression afforded by a promoter element. Multiple enhancer elements are readily available to persons of ordinary skill in the art.
  • the present disclosure provides a pharmaceutical composition
  • a pharmaceutical composition comprising: a) a rAAV vector, as described above; and b) a pharmaceutically acceptable carrier, diluent, excipient, or buffer.
  • the pharmaceutically acceptable carrier, diluent, excipient, or buffer is suitable for use in a human.
  • excipients include any pharmaceutical agent that can be administered without undue toxicity.
  • Pharmaceutically acceptable excipients include, but are not limited to, liquids such as water, saline, glycerol and ethanol.
  • Pharmaceutically acceptable salts can be included therein, for example, mineral acid salts such as hydrochlorides, hydrobromides, phosphates, sulfates, and the like; and the salts of organic acids such as acetates, propionates, malonates, benzoates, and the like. Additionally, auxiliary substances, such as wetting or emulsifying agents, pH buffering substances, and the like, may be present in such vehicles.
  • mineral acid salts such as hydrochlorides, hydrobromides, phosphates, sulfates, and the like
  • organic acids such as acetates, propionates, malonates, benzoates, and the like
  • auxiliary substances such as wetting or emulsifying agents, pH buffering substances, and the like, may be present in such vehicles.
  • the present invention provides a pharmaceutical composition
  • a pharmaceutical composition comprising a rAAV vector as described above in a pharmaceutically-acceptable carrier or other medicinal agents, pharmaceutical agents, carriers, adjuvants, diluents, etc.
  • the carrier will typically be a liquid.
  • the carrier may be either solid or liquid, such as sterile, pyrogen-free water or sterile pyrogen-free phosphate-buffered saline solution.
  • the carrier will be respirable, and will preferably be in solid or liquid particulate form.
  • water that contains the additives usual for injection solutions, such as stabilizing agents, salts or saline, and/or buffers.
  • pharmaceutically acceptable it is meant a material that is not biologically or otherwise undesirable, e.g., the material may be administered to a subject without causing any undesirable biological effects.
  • a pharmaceutical composition may be used, for example, in transfection of a cell ex vivo or in administering a viral particle or cell directly to a subject.
  • the methods of the present invention provide a means for delivering heterologous nucleic acid sequences into a host tissue or cell, including both dividing and non-dividing cells.
  • the vectors and other reagents, methods and pharmaceutical formulations of the present invention are additionally useful in a method of administering a protein or peptide to a subject in need thereof, as a method of treatment or otherwise.
  • the protein or peptide may thus be produced in vivo in the subject.
  • the subject may be in need of the protein or peptide because the subject has a deficiency of the protein or peptide, or because the production of the protein or peptide in the subject may impart some therapeutic effect, as a method of treatment or otherwise, and as explained further below.
  • treatment refers to obtaining a desired pharmacologic and/or physiologic effect.
  • the effect may be prophylactic in terms of completely or partially preventing a disease or symptom thereof and/or may be therapeutic in terms of a partial or complete cure for a disease and/or adverse effect attributable to the disease.
  • Treatment covers any treatment of a disease in a mammal, particularly in a human, and includes: (a) preventing the disease from occurring in a subject which may be predisposed to the disease or at risk of acquiring the disease but has not yet been diagnosed as having it; (b) inhibiting the disease, i.e., arresting its development; and (c) relieving the disease, i.e., causing regression of the disease.
  • the present invention may be employed to deliver any foreign nucleic acid with a biological effect to treat or ameliorate the symptoms associated with any disorder related to gene expression in any organ, tissue or cell, especially those associated with e.g. the liver, brain or eye.
  • Illustrative disease states include, but are not limited to: lysosomal storage disease, acute intermittent porphyria, ornithine transcarbamylase deficiency, alpha(1)-antitrypsin deficiency, acute liver failure, Pompe disease, Tyrosinemia, Crigler-Najjar syndrome, hepatitis, cirrhosis, hepatocellular carcinoma, AIDS, Alzheimer's disease, Parkinson's disease, Huntington's disease, amyotrophic lateral sclerosis, epilepsy, and other neurological disorders, cancer (e.g. brain cancer), retinal degenerative diseases and other diseases of the eye.
  • cancer e.g. brain cancer
  • retinal degenerative diseases and other diseases of the eye e.g. brain cancer
  • Gene transfer has substantial potential use in understanding and providing therapy for disease states.
  • diseases in which defective genes are known and have been cloned. In some cases, the function of these cloned genes is known.
  • the above disease states fall into two classes: deficiency states, usually of enzymes, which are generally inherited in a recessive manner, and unbalanced states, at least sometimes involving regulatory or structural proteins, which are inherited in a dominant manner.
  • gene transfer could be used to bring a normal gene into affected tissues for replacement therapy, as well as to create animal models for the disease using antisense mutations.
  • gene transfer could be used to create a disease state in a model system, which could then be used in efforts to counteract the disease state.
  • the methods of the present invention permit the treatment of genetic diseases.
  • a disease state is treated by partially or wholly remedying the deficiency or imbalance that causes the disease or makes it more severe.
  • the use of site-specific integration of nucleic sequences to cause mutations or to correct defects is also possible.
  • the present invention provides a method of delivering a gene product to a tissue or cell (e.g. a hepatic, neuronal or retinal tissue or cell) in a subject, the method comprising administering to the subject a rAAV vector as described above.
  • the gene product can be a polypeptide or an interfering RNA (e.g., an shRNA, an siRNA, and the like), or an aptamer, e.g. as described above.
  • the cell may, for example, be a blood cell, stem cell, bone marrow (e.g.
  • tissue may, for example, be selected from blood, bone marrow, muscle tissue (e.g. skeletal muscle, cardiac muscle or smooth muscle including vascular smooth muscle), central or peripheral nervous system tissue (e.g. brain, neuronal tissue or retinal tissue), pancreatic tissue, liver tissue, kidney tissue, lung tissue, intestinal tissue or heart tissue.
  • muscle tissue e.g. skeletal muscle, cardiac muscle or smooth muscle including vascular smooth muscle
  • central or peripheral nervous system tissue e.g. brain, neuronal tissue or retinal tissue
  • pancreatic tissue liver tissue, kidney tissue, lung tissue, intestinal tissue or heart tissue.
  • Delivering a gene product to a retinal cell can provide for treatment of a retinal disease.
  • the retinal cell can be a photoreceptor, a retinal ganglion cell, a Muller cell, a bipolar cell, an amacrine cell, a horizontal cell, or a retinal pigmented epithelial cell.
  • the retinal cell is a photoreceptor cell, e.g., a rod or cone cell.
  • delivering a gene product to a neuronal tissue or cell can provide for treatment of a neurological disorder.
  • the gene product may be delivered to various cell types present in neuronal tissue, e.g. neurons or glial cells (e.g. astrocytes, oligodendrocytes and so on). Delivering a gene product to the liver may provide treatment for a hepatic disorder.
  • the gene product may be delivered to, for example, hepatocytes.
  • the present disclosure provides a method of treating a disease (e.g. a hepatic, neurological or ocular disease), the method comprising administering to an individual in need thereof an effective amount of a rAAV vector as described above.
  • a disease e.g. a hepatic, neurological or ocular disease
  • a subject rAAV vector can be administered via intracranial injection, intracerebral injection, intraocular injection, by intravitreal injection, retinal injection, sub-retinal injection, intravenous injection or by any other convenient mode or route of administration.
  • exemplary modes of administration include oral, rectal, transmucosal, topical, transdermal, inhalation, parenteral (e.g., intravenous, subcutaneous, intradermal, intramuscular, and intraarticular) administration, and the like, as well as direct tissue or organ injection, alternatively, intrathecal, direct intramuscular, intraventricular, intravenous, intraperitoneal, intranasal, or intraocular injections.
  • injectables can be prepared in conventional forms, either as liquid solutions or suspensions, solid forms suitable for solution or suspensions in liquid prior to injection, or as emulsions.
  • Recombinant virus vectors are preferably administered to the subject in an amount that is sufficient to result in infection (or transduction) and expression of the heterologous nucleic acid sequence in cells (e.g. liver, neuronal or retinal cells) of the subject.
  • the target cells are hepatocytes, neural cells (including cells of the central and peripheral nervous systems, in particular, brain cells) or retinal cells.
  • the retinal cell is a photoreceptor cell (e.g., rods and/or cones).
  • the retinal cell is an RGC cell.
  • the retinal cell is an RPE cell.
  • retinal cells may include amacrine cells, bipolar cells, and horizontal cells.
  • the vector is administered in a therapeutically effective amount.
  • a “therapeutically-effective” amount as used herein is an amount of that is sufficient to alleviate (e.g., mitigate, decrease, reduce) at least one of the symptoms associated with a disease state.
  • a “therapeutically-effective” amount is an amount that is sufficient to provide some improvement in the condition of the subject.
  • a “therapeutically effective amount” will fall in a relatively broad range that can be determined through experimentation and/or clinical trials. For example, for in vivo injection, a therapeutically effective dose will be on the order of from about 10 6 to about 10 15 of rAAV virions, e.g., from about 10 8 to 10 12 rAAV virions.
  • an effective amount of rAAV virions to be delivered to cells will be on the order of from about 10 8 to about 10 13 of the rAAV virions.
  • Other effective dosages can be readily established by one of ordinary skill in the art through routine trials establishing dose response curves.
  • more than one administration may be employed to achieve the desired level of gene expression over a period of various intervals, e.g., daily, weekly, monthly, yearly, etc.
  • Neurological diseases which may be treated include any disease associated with the brain or CNS, including psychiatric diseases. Diseases of the brain fall into two general categories: (a) pathologic processes such as infections, trauma and neoplasm; and (b) diseases unique to the nervous system which include diseases of myelin and degeneration of neurons. Disease from either category may be treated.
  • the neurological disease may be selected from neurodegenerative diseases such as Alzheimer's Disease, Parkinson's Disease, amyotrophic lateral sclerosis (ALS), spinal muscular atrophy and cerebella degeneration; schizophrenia; epilepsy; ischemia-related disease and stroke; demyelinating diseases such as multiple sclerosis, perivenous encephalitis, leukodystrophies such as metachromatic leukodystrophy due to deficiency of arylsulfatase A, Krabbe's disease due to deficiency of galactocerebroside beta-galactosidase, adrenoleukodystrophy and adrenomyeloneuropathy; post-viral diseases such as progressive multifocal leukoencephalopathy, acute disseminated encephalomyelitis, acute necrotizing hemorrhagic leukoencephalitis; mitochondrial encephalomyopathies; neurological cancers, such as primary brain tumors including glioma, meningi
  • Ocular diseases that can be treated using a subject method include, but are not limited to, acute macular neuroretinopathy; Behcet's disease; choroidal neovascularization; diabetic uveitis; histoplasmosis; macular degeneration, such as acute macular degeneration, non-exudative age related macular degeneration and exudative age related macular degeneration; edema, such as macular edema, cystoid macular edema and diabetic macular edema; multifocal choroiditis; ocular trauma which affects a posterior ocular site or location; ocular tumors; retinal disorders, such as central retinal vein occlusion, diabetic retinopathy (including proliferative diabetic retinopathy), proliferative vitreoretinopathy (PVR), retinal arterial occlusive disease, retinal detachment, uveitic retinal disease; sympathetic opthalmia; Vogt Koyanagi-Ha
  • lysosomal storage diseases e.g. acute intermittent porphyria, ornithine transcarbamylase deficiency, Wilson's disease, mucopolysaccharidoses (e.g. MPS type I or MPS type VI), Sly syndrome, Pompe disease, tyrosinemia, alpha(1)-antitrypsin deficiency, Crigler-Najjar syndrome; hepatitis A, B or C; liver cirrhosis; liver cancer, e.g. hepatocellular carcinoma; or acute liver failure.
  • lysosomal storage diseases e.g. acute intermittent porphyria, ornithine transcarbamylase deficiency, Wilson's disease, mucopolysaccharidoses (e.g. MPS type I or MPS type VI), Sly syndrome, Pompe disease, tyrosinemia, alpha(1)-antitrypsin deficiency, Crigler-Najjar syndrome; hepatitis A, B or C
  • Suitable subjects include both avians and mammals, with mammals being preferred.
  • avian as used herein includes, but is not limited to, chickens, ducks, geese, quail, turkeys and pheasants.
  • mamal as used herein includes, but is not limited to, humans, bovines, ovines, caprines, equines, felines, canines, lagomorphs, etc. Human subjects are the most preferred. Human subjects include fetal, neonatal, infant, juvenile and adult subjects.
  • the rAAV vectors disclosed herein exhibit increased transduction of a tissue (e.g. hepatic, neuronal and/or retinal tissues), e.g. compared to a corresponding AAV vector (from the same serotype) comprising a wild type AAV capsid protein.
  • a tissue e.g. hepatic, neuronal and/or retinal tissues
  • the rAAV vector may exhibit at least 10%, 50%, 100%, 500% or 1000% increased infectivity, compared to the infectivity by an AAV virion comprising the corresponding wild type AAV capsid protein.
  • the rAAV vectors disclosed herein may selectively or specifically infect a tissue (e.g. hepatic, neuronal or retinal tissues), e.g. show increased transduction of hepatic, neuronal or retinal cells compared to other cell types.
  • the rAAV vector may exhibit at least 10%, 50%, 100%, 500% or 1000% increased infectivity of a particular cell type (e.g. hepatic, neuronal or retinal cells), compared to another cell type (e.g. non-hepatic, non-neuronal and/or non-retinal cells).
  • the rAAV vector may selectively infect hepatocytes, neurons and/or photoreceptor cells compared to cells outside the liver, brain and/or eye.
  • the recombinant AAV vector exhibits increased transduction of a neuronal or retinal tissue, e.g. where the vector is used to treat a neurological or ocular disorder
  • the vector preferably comprises a variant AAV2 capsid protein.
  • the recombinant AAV vector exhibits increased transduction of liver tissue, e.g. where the vector is used to treat a hepatic disorder, the vector preferably comprises a variant AAV3B, AAV-LK03 or AAV8 capsid protein.
  • the present disclosure provides an isolated nucleic acid comprising a nucleotide sequence that encodes a variant adeno-associated virus (AAV) capsid protein as described above.
  • the isolated nucleic acid can be comprised in an AAV vector, e.g., a recombinant AAV vector.
  • a recombinant AAV vector comprising such a variant AAV capsid protein-encoding sequence can be used to generate a recombinant AAV virion (i.e. a recombinant AAV vector particle).
  • a recombinant AAV vector particle i.e. a recombinant AAV vector particle.
  • the present disclosure provides a recombinant AAV vector that, when introduced into a suitable cell, can provide for production of a recombinant AAV virion.
  • the present invention further provides host cells, e.g., isolated (genetically modified) host cells, comprising a subject nucleic acid.
  • a subject host cell can be an isolated cell, e.g., a cell in in vitro culture.
  • a subject host cell is useful for producing a subject rAAV virion, as described below. Where a subject host cell is used to produce a subject rAAV virion, it is referred to as a “packaging cell.”
  • a subject host cell is stably genetically modified with a subject nucleic acid.
  • a subject host cell is transiently genetically modified with a subject nucleic acid.
  • a subject nucleic acid is introduced stably or transiently into a host cell, using established techniques, including, but not limited to, electroporation, calcium phosphate precipitation, liposome-mediated transfection, and the like.
  • a subject nucleic acid will generally further include a selectable marker, e.g., any of several well-known selectable markers such as neomycin resistance, and the like.
  • a subject host cell is generated by introducing a subject nucleic acid into any of a variety of cells, e.g., mammalian cells, including, e.g., murine cells, and primate cells (e.g., human cells).
  • Suitable mammalian cells include, but are not limited to, primary cells and cell lines, where suitable cell lines include, but are not limited to, 293 cells, COS cells, HeLa cells, Vero cells, 3T3 mouse fibroblasts, C3H10T1/2 fibroblasts, CHO cells, and the like.
  • suitable host cells include, e.g., HeLa cells (e.g., American Type Culture Collection (ATCC) No. CCL-2), CHO cells (e.g., ATCC Nos.
  • CRL9618, CCL61, CRL9096 293 cells (e.g., ATCC No. CRL-1573), Vero cells, NIH 3T3 cells (e.g., ATCC No. CRL-1658), Huh-7 cells, BHK cells (e.g., ATCC No. CCL10), PC12 cells (ATCC No. CRL1721), COS cells, COS-7 cells (ATCC No. CRL1651), RATI cells, mouse L cells (ATCC No. CCLI.3), human embryonic kidney (HEK) cells (ATCC No. CRL1573), HLHepG2 cells, and the like.
  • a subject host cell can also be made using a baculovirus to infect insect cells such as Sf9 cells, which produce AAV (see, e.g., U.S. Pat. No. 7,271,002; U.S. patent application Ser. No. 12/297,958).
  • a subject genetically modified host cell includes, in addition to a nucleic acid comprising a nucleotide sequence encoding a variant AAV capsid protein, as described above, a nucleic acid that comprises a nucleotide sequence encoding one or more AAV rep proteins.
  • a subject host cell further comprises an rAAV vector.
  • An rAAV virion can be generated using a subject host cell. Methods of generating an rAAV virion are described in, e.g., U.S. Patent Publication No. 2005/0053922 and U.S. Patent Publication No. 2009/0202490.
  • AAV assembly associated protein
  • helper virus for AAV refers to a virus that allows AAV (e.g. wild-type AAV) to be replicated and packaged by a mammalian cell.
  • helper viruses for AAV are known in the art, including adenoviruses, herpesviruses and poxviruses such as vaccinia.
  • the adenoviruses encompass a number of different subgroups, although Adenovirus type 5 of subgroup C is most commonly used.
  • Numerous adenoviruses of human, non-human mammalian and avian origin are known and available from depositories such as the ATCC.
  • Viruses of the herpes family include, for example, herpes simplex viruses (HSV) and Epstein-Barr viruses (EBV), as well as cytomegaloviruses (CMV) and pseudorabies viruses (PRV); which are also available from depositories such as ATCC.
  • HSV herpes simplex viruses
  • EBV Epstein-Barr viruses
  • CMV cytomegaloviruses
  • PRV pseudorabies viruses
  • Helper virus function(s) refers to function(s) encoded in a helper virus genome which allow AAV replication and packaging (in conjunction with other requirements for replication and packaging described herein).
  • helper virus function may be provided in a number of ways, including by providing helper virus or providing, for example, polynucleotide sequences encoding the requisite function(s) to a producer cell in trans.
  • a plasmid or other expression vector comprising nucleotide sequences encoding one or more adenoviral proteins is transfected into a producer cell along with an rAAV vector.
  • infectious virus or viral particle is one that comprises a competently assembled viral capsid and is capable of delivering a polynucleotide component into a cell for which the viral species is tropic. The term does not necessarily imply any replication capacity of the virus.
  • Assays for counting infectious viral particles are described elsewhere in this disclosure and in the art.
  • Viral infectivity can be expressed as the ratio of infectious viral particles to total viral particles. Methods of determining the ratio of infectious viral particle to total viral particle are known in the art. See, e.g., Grainger et al. (2005) Mol. Ther. 11:S337 (describing a TCID50 infectious titer assay); and Zolotukhin et al. (1999) Gene Ther. 6:973.
  • a “replication-competent” virus refers to a phenotypically wild-type virus that is infectious, and is also capable of being replicated in an infected cell (i.e. in the presence of a helper virus or helper virus functions).
  • replication competence generally requires the presence of functional AAV packaging genes.
  • rAAV vectors as described herein are replication-incompetent in mammalian cells (especially in human cells) by virtue of the lack of one or more AAV packaging genes.
  • rAAV vector preparations as described herein are those which contain few if any replication competent AAV (rcAAV, also referred to as RCA) (e.g., less than about 1 rcAAV per 10 2 rAAV particles, less than about 1 rcAAV per 10 4 rAAV particles, less than about 1 rcAAV per 10 8 rAAV particles, less than about 1 rcAAV per 10 12 rAAV particles, or no rcAAV).
  • rcAAV also referred to as RCA
  • an “isolated” nucleic acid, vector, virus, virion, host cell, or other substance refers to a preparation of the substance devoid of at least some of the other components that may also be present where the substance or a similar substance naturally occurs or is initially prepared from.
  • an isolated substance may be prepared by using a purification technique to enrich it from a source mixture. Enrichment can be measured on an absolute basis, such as weight per volume of solution, or it can be measured in relation to a second, potentially interfering substance present in the source mixture. Increasing enrichments of the embodiments of this disclosure are increasingly more isolated.
  • An isolated nucleic acid, vector, virus, host cell, or other substance is in some embodiments purified, e.g., from about 80% to about 90% pure, at least about 90% pure, at least about 95% pure, at least about 98% pure, or at least about 99%, or more, pure.
  • ttAAV2 a novel AAV2 vector designated ttAAV2 (as in true-type).
  • novel vector was tested in a number of animal models (rats, mice and neonatal mice) in order to evaluate whether ttAAV2 behaved differently as compared to the tissue culture adapted (wild type) AAV2.
  • the inventors demonstrated that ttAAV2 has advantages for gene delivery over AAV2, and is particularly useful for in vivo transduction of brain or eye tissues with heterologous sequences.
  • the capsid gene of wtAAV2 was taken from our producer plasmid pDG ( FIG. 10 ). This plasmid contains wtAAV2 rep and cap genes. Subfragments of the capsid gene (pDG nucleotides A:3257-3759, B:4025-4555, C:4797-5287 and D:5149-5425, respectively) were subcloned into pBS for subsequent mutagenesis. Four mutations were introduced into fragment A via site-directed mutagenesis resulting in a construct that encodes for amino acid (AA) changes V125I, V151A, A162S and T205S. Fragment B was mutated to encode the single AA change, N312S.
  • AA amino acid
  • Fragment C was mutated to encode the AA exchanges Q457M, S492A, E499D, F533Y, G546D, E548G, R585S, R588T, and A593S.
  • A-C were re-cloned into pDG, resulting in a producer plasmid (pDG-ttAAV2) that would support the production of a recombinant virus that is encapsidated by ttAAV2 capsid.
  • Vector production was established following the standard protocols employing co-transfection of rAAV plasmids with pDG, which provides both the Ad helper functions as well as the AAV rep and cap genes.
  • pDG which provides both the Ad helper functions as well as the AAV rep and cap genes.
  • a variety of rAAV plasmids were used to generate recombinant plasmids.
  • pTR-UF11 (CAG-GFP) was used as the rAAV plasmid. 8 ⁇ 10 8 293 cells were seeded per cell factory (CF10). 14-18 hours later, the cells were transfected with pDG or pDG-rrAAV2 and prAAV (e.g. pTR-UF11) using the CaPO 4 co-precipitation method. After 72 hours the cells were harvested and resuspended in lysis buffer (20 mMTris-HCl, pH8, 150 mM NaCl, 0.5% deoxycholate). The cell pellets were lysed by four cycles of freeze and thaw to release the virus, where each cycle consists of 30 minutes at ⁇ 80° C. followed by 30 minutes at 37° C. After the last thaw the lysate was treated with benzonase at a concentration of 50 U/ml and incubated for 30 min at 37° C. The recombinant virus was purified using gravity flow columns.
  • the crude lysate was clarified by centrifugation at 4000 g for 15 minutes and applied to the pre-formed iodixanol step gradient.
  • the viral fraction was then collected and re-buffered into Lactate Ringer's solution as well as concentrated using Amicon centrifugation filters.
  • the crude extract contained 4.5 ⁇ 10 12 particles; the collected viral fractions contained 1.5 ⁇ 10 12 particles. At this point the purification method recovered ca. 33% of the virus present in the crude extract.
  • the rAAV2 vector is produced as follows. To produce rAAV2 virions, 5 ⁇ 10 8 293T cells were seeded per cell factory (CF10). 14-18 hours later, the cells were double transfected with the GFP-containing vector PD10-pST2-CMV-GFP, and either the pDG or the pDG capsid mutant (pDG-ttAAV2) to produce AAV2-CMV-GFP wild-type or true-type vectors, respectively. The double transfections were realised using PEI-max from Polysciences at a ratio of 3.5 ml of PEI per mg of DNA. The cells were harvested after 72 hours of incubation at 37° C.
  • the cell pellets were then lysed by 4 cycles of freeze and thaw to release the virus, where each cycle consists of 30 minutes at ⁇ 80° C. followed by 30 minutes at 37° C. After the last thaw the lysate was treated with benzonase at a concentration of 150 U/ml and incubated at 37° for 30 minutes. The lysate was then spun at 2000 rpm for 20 minutes to clarify the lysate. The supernatant was filtered using a 0.22 ⁇ m cellulose acetate filter and the recombinant AAV2 virus preparations were purified by FPLC using the AKTApurifier chromatography system (GE Healthcare) and an AVB sepharose affinity column (bfr.
  • the ttAAV2 vector was tested in vivo in order to assess the bioactivity of the modified virus in such a context.
  • Samples of AAV2-CMV-GFP WT and TT viruses were prepared for injections into a number of in vivo models. For this purpose we concentrated the viruses, as only limited volumes of vectors can be injected in vivo. We then performed a qPCR and SDS-PAGE to assess the new titres of the concentrated vectors ( FIG. 11 ).
  • AAV2-CMV-GFP TT at 1.33 ⁇ 10 12 viral genomes/ml
  • AAV2-CMV-GFP WT at 1.25 ⁇ 10 12 viral genomes/ml.
  • capsid titres were as follow: AAV2-CMV-GFP TT at 8.89 ⁇ 10 12 capsids/ml and AAV2-CMV-GFP WT at 7.83 ⁇ 10 12 capsids/ml.
  • the titres differ between the genome copies and the capsid copies as the SDS-PAGE also shows empty capsids, which are normally generated during recombinant AAV vectors production, hence the capsid titre is higher than the viral genome titre obtained from the qPCR.
  • the rAAV2 TT and WT viruses were injected in the substantia nigra or in the striatum of wild-type rats, with 3 rats being injected per condition, at a dose of 2 ⁇ 10 9 vg or 3.5 ⁇ 10 9 vg per injection. After 28 days brains were dissected and tissue sections were prepared for immunofluorescence analysis. The primary data are shown in FIG. 12 and FIG. 26 .
  • both the rAAV2 TT and WT viruses were able to transduce neuronal and glial cells from each injection site, albeit with varying efficiencies.
  • the TT vector transduced brain tissues more efficiently and spread more from the site of injection than the WT vector.
  • the TT vector was able to travel from the transduced cell bodies at the site of injection to the hypothalamus by active transport along the neuron projections, highlighting a strong ability for retrograde transport. This retrograde transport ability has been lost in the tissue-culture adapted WT rAAV2 vector as no transduced cells could be observed in the same area (see FIG. 26 ).
  • ttAAV2 and wt-AAV2 from the same batch as was used for our rat brain studies was injected into adult mouse eyes at a dose of 2 ⁇ 10 9 vg per eye.
  • each mouse received an injection of rAAV2 TT in one eye and an injection of rAAV2 WT in the contra-lateral eye.
  • Three different routes of intra-ocular injections were analysed: intra-cameral, intra-vitreal and sub-retinal. The animals were harvested and GFP expression was assessed by immunofluorescence after 6 weeks. The results are shown in FIG. 13 .
  • both ttAAV2 and wtAAV2 GFP vectors were injected into mouse neonates. Two routes of injections were tested, intra-venous injection and intra-cranial injections. After 4 weeks, the animals were sacrificed and all tissues were harvested from all mice. We have analysed the brain, which after harvesting was visualised by direct fluorescence of the organ on a fluorescence microscope. The results are shown in FIG. 14 . The results of intracranial and systemic injections are discussed in more detail below.
  • AAV2 TT displays enhanced transduction of mouse brain tissues and higher spread after intracranial injection as compared to the AAV2 WT vector.
  • the TT vector performed better both in terms of level of expression and of number of cells transduced.
  • AAV2 TT and WT seem to have the same cell type affinity, each displaying transduction of neuronal as well as glial cells, suggesting that the differences observed are differences in efficiency rather than in cell-type specificity ( FIG. 28 ).
  • Taken together these data indicate that ttAAV2 shows much enhanced transduction of mouse brain tissues after i.c.
  • Intrajugular injections of 2 ⁇ 10 11 vg of either vector were done in P1 neonates.
  • the animals were sacrificed 4 weeks post-injection and various organs were harvested and assessed for GFP transduction by immunohistochemistry using an anti-GFP antibody (brain, liver, heart, muscle, lungs, spleen and kidney). Results of the brains staining are shown in FIG. 29 and high magnification pictures are presented in FIG. 30 .
  • AAV-TT vector doesn't seem to have a strong affinity for other organs but instead displays specificity mainly for neuronal tissues. This observation could prove beneficial for the treatment of neuronal genetic disorders by intravenous injections of AAV as it ensures that the vector will not transduce non-target peripheral organs but mainly only the brain via this injection route.
  • ttAAV2 has extraordinary transduction characteristics in eye and brain tissues, displaying specificity for neuronal tissues almost exclusively.
  • the three-dimensional positions of these mutations in the AAV2 capsid protein are known and are shown in FIGS. 15 to 20 . Corresponding positions can be identified in the AAV capsid proteins from other serotypes (see below).
  • the ttAAV2 capsid can be mutated in order to reverse individual chosen amino acids (or groups of amino acids, e.g. based on groups 1 to 6 discussed above) to their corresponding sequence in the wild-type AAV2 capsid. Each mutant vector can then be analysed using methods as described in the examples above.
  • the various mutant vectors, expressing GFP can be submitted to a first screen in an animal model by intracranial (IC) injection in CD1 neonatal mice.
  • the GFP signal obtained in the injected brains from each mutant vector is then observed by fluorescence microscopy and compared to that obtained from the original GFP-expressing wtAAV2 and ttAAV2 vectors.
  • the injected brains are then further sectioned and analysed by immunohistochemistry.
  • these selected mutant vectors are submitted to a second screen by adult rat intracranial injections. Additionally, relevant mutant combinations are analysed by intravenous (IV) injections into neonatal mice in order to evaluate the biodistribution of the vectors. Selected organs (heart, lung, liver, spleen, kidney, muscle) are then processed for immunohistochemistry and evaluation of GFP expression.
  • IV intravenous
  • the AAV2 TT capsid was mutated step by step in order to reverse the chosen amino acids to their corresponding sequence in the wild-type AAV2 capsid. This strategy enabled us to discern more specifically the contribution of each of the 14 amino acid mutations towards the phenotype of AAV2 TT and to define a minimal true-type vector, containing only the necessary mutations.
  • the 14 TT-specific residues into groups based on their positions on the AAV capsid and their associated potential contributions to the transduction profile.
  • the various AAV-TT mutants were screened by intracranial injections in neonatal mouse brains or in adult rat brains in order to observe whether the reversion of some TT-specific residues to the WT equivalents was associated with a loss of phenotype, thereby identifying the important amino acid changes amongst the 14 TT residues.
  • HBS Heparin Binding Site
  • residues 585 and 588 are responsible for heparin binding of the AAV2 WT capsid.
  • AAV-TT these are replaced and we assumed that this replacement support the spread of the virus in heparin sulphate proteoglycan-rich brain tissue.
  • These two residues are situated on the three-fold proximal spikes of the AAV2 capsid structure (see FIG. 16 ).
  • AAV2-TT, AAV2-WT and AAV2-HBnull vectors were injected in the substantia nigra or in the striatum of wild-type rats at a dose of 3.5 ⁇ 10 9 vg per injection. After 28 days brains were dissected and tissue sections were prepared for immunohistochemistry analysis of GFP expression ( FIG. 32 ).
  • the S312 residue is the only TT-specific change that is located on the internal side of the AAV2 capsid. Our assumption was that this internal residue might play a role in capsid-DNA interactions, thereby potentially contributing to either virus stability, genome packaging or genome release during infection.
  • the D and G residues at positions 546 and 548, respectively, are located in the groove between the proximal 3-fold peaks.
  • a single isolated serine, S593, is situated in the groove between threefold-proximal spikes.
  • the mutation S312N was engineered on the AAV2-TT capsid to create the TT-S312N mutant.
  • the mutations D546G and G548E were engineered on the AAV2-TT capsid to generate the AAV-TT-DG mutant.
  • the mutation S593A was engineered on the AAV2-TT capsid to create the AAV-TT-S593A mutant.
  • TT-S312N internal mutant transduces mouse brains with more efficiency than the full TT vector as it leads to more GFP expression overall in all the injected brains.
  • the minimal TT vector TT-S312N-D546G/G548E-S593A
  • the variability between animals was very high, with only one animal out of five performing better than the animals transduced with TT-S312N. This high variability led us to consider this provisional minimal TT vector with caution, especially since the immunohistochemistry analyses also showed that the TT-S312N variant performed better than the TT-S312N-DG-S593A.
  • TT-S312N variant as our most preferred AAV TT vector, which is composed of the following 13 amino acid mutations compared to the wild-type AAV2: V125I, V151A, A162S, T205S, Q457M, S492A, E499D, F533Y, G546D, E548G, R585S, R588T, and A593S.
  • these studies illustrate the individual function and contribution of a number TT-specific residues.
  • four amino acids closely situated on the threefold spike of the capsid are likely involved in receptor binding.
  • these residues are reverted in the AAV-TT back to the AAV2 WT corresponding residues.
  • the mutations M457Q, A492S, D499E and Y533F may be engineered on the AAV-TT capsid and this mutant vector analysed as previously described, in order to illustrate the role of these residues.
  • four amino acids situated in VP1/VP2 primary sequence which are likely to be involved in trafficking of the virus, may be reverted in the AAV-TT back to AAV2 WT corresponding residues (I125V, A151V, S162A, S205T) and analysed similarly.
  • ttAAV2-specific amino acid changes in the context of other, non-AAV2 serotypes can also be determined.
  • the capsid amino acid sequences of the main adeno-associated viruses can be aligned with the one from ttAAV2, e.g. as shown in FIG. 9 .
  • AAV adeno-associated viruses
  • the various mutant vectors, expressing GFP are then submitted to a first screen by intracranial (IC) injection in CD1 neonatal mice.
  • the GFP signal obtained in the injected brains is then compared to that obtained from the appropriate GFP-expressing serotype controls.
  • the diminution or increase of GFP expression when mutating the identified amino acids into their corresponding AAV2 residues demonstrates the importance of these particular residues at these specific positions.
  • the injected brains are then further sectioned and analyzed by immunohistochemistry.
  • chosen mutant serotypes are analyzed by intravenous (IV) injections into neonatal mice in order to evaluate the biodistribution of the vectors and compare it to the original, non-mutated counterparts.
  • the relevant amino acids identified in the ttAAV2 are inserted as key mutations into the other prominent serotypes at the relevant positions.
  • the insertion of ttAAV2-specific residues in other AAV subtypes enables us to improve the transduction and biodistribution profiles of each serotype.
  • a comparative analysis of the capsid amino acid sequences of existing adeno-associated serotypes with the one from ttAAV2 first enabled us to identify ttAAV2-specific residues that are conserved in other serotypes (see FIG. 9 ). These residues consist of S162, S205, S312, G548, S585 and T588. Each non-AAV2 serotype contains one or a combination of several of these residues at a corresponding amino acid position in its sequence.
  • each of these residues in the various serotypes are converted into the corresponding wild-type AAV2 amino-acid(s) and the transduction efficiency of these new mutants is tested.
  • AAV1 contains the residues S205, G549, S586 and T589 which correspond to the following residues in ttAAV2: S205, G548, S585 and T588.
  • the VP1 monomer from AAV1 was aligned three-dimensionally with VP1 from AAV2 we could verify that the corresponding residues in wtAAV2, namely T205, E548, R585 and R588, are at perfectly matching positions on the 3D structure ( FIG. 21 ).
  • the following mutations are made in AAV1 capsid sequence: S205T, G549E, S586R, T589R.
  • AAV5 contains the residues G537, S575 and T578 which correspond to the following residues in ttAAV2: G548, S585 and T588.
  • the R585 and R588 residues in AAV2 are at matching positions with S575 and T578 in AAV5 on the 3D structure.
  • the residue E548 in AAV2 did not perfectly match the residue G537 in AAV5 according to the three-dimensional structure ( FIG. 22 ), we still decided to include it in the study as both residues are relatively spatially close. Therefore in particular embodiments the following mutations are made in AAV5 capsid sequence: G537E, S575R, T578R.
  • AAV6 contains the residues S205, G549, S586 and T589 which correspond to the following residues in ttAAV2: S205, G548, S585 and T588.
  • the corresponding residues in wtAAV2, namely T205, E548, R585 and R588 ( FIG. 23 ) are at perfectly matching positions on the VP1 3D structures. Therefore in particular embodiments the following mutations are made in AAV6 capsid sequence: S205T, G549E, S586R, T589R.
  • AAV8 contains the residues S315 and T591 which correspond to the following residues in ttAAV2: S312 and T588.
  • the corresponding residues in wtAAV2, namely N312 and R588, are at perfectly matching positions on the VP1 3D structures ( FIG. 24 ). Therefore in particular embodiments the following mutations are made in AAV8 capsid sequence: S315N, T591R.
  • the improved transduction efficiency imparted by the S312N mutation in TT AAV2 may be transferred to the AAV8 serotype by applying the amino acid change S315N.
  • AAV8-S315N vector plasmid was used to produce recombinant AAV8-S315N vectors expressing an ITR-containing CMV-GFP transgene by double transfection of 293T cells.
  • the vector was then purified from the cell lysate and from the harvested culture supernatant by FPLC affinity chromatography, using an AVB sepharose resin.
  • the capsid titer and vector genome titer were assessed by SDS-PAGE and qPCR, respectively.
  • the mutant AAV8-S315N vector, expressing GFP is screened by systemic injections in CD1 neonatal mice.
  • a titer-matched AAV8 vector is used as a control.
  • GFP expression obtained in various organs after intra-jugular injection of 2 ⁇ 10 11 vg is then analysed, primarily focusing on the liver where AAV8 has previously shown some strong transduction efficiency.
  • AAV9 contains the residues S162, S205, G549 and S586 which correspond to the following residues in ttAAV2: S162, S205, G548 and S585.
  • the corresponding residues in AAV2, namely A162, T205, E548 and R585, are at perfectly matching positions on the VP1 3D structures ( FIG. 25 ). Therefore in particular embodiments the following mutations are made in AAV8 capsid sequence: S162A, S205T, G549E, and S586R.
  • AAVrh10 contains the residue G551 which corresponds with G548 with ttAAV2. Considering how conserved this residue and position appears among the various serotypes, we assume that G551 in AAVrh10 will align with E548 in wtAAV2 three-dimensionally. Therefore in one embodiment the following mutation is made in AAVrh10 capsid sequence: G551E.
  • the AAV3B serotype also contains an internal serine at position 312 after aligning the capsid protein VP1 sequence with the one from AAV-TT (see AAV3B capsid sequence in FIG. 38 ).
  • the newly described LK03 AAV vector a chimeric capsid composed of five different parental AAV capsids engineered by M. A. Kay by DNA-shuffling, also contains the residue S312 in the internal side of the capsid (see Lisowski et al., Selection and evaluation of clinically relevant AAV variants in a xenograft liver model, Nature 506, 382-386 (2014)).
  • the capsid sequence of AAV-LK03 is disclosed in WO 2013/029030 and shown in FIG. 39 .
  • the AAV3B and the AAV-LK03 vectors are mutated by applying the amino-acid change S312N in both vectors. These new mutants, and their corresponding AAV control serotypes, are also tested by intra-jugular injections in neonatal mice. The GFP expression obtained in various harvested organs is then analysed.
  • the key amino acids identified during the ttAAV2 characterization are inserted into the other prominent serotypes at the relevant positions.
  • the newly engineered vectors are then tested using the appropriate non-mutated serotypes as controls. This validates the importance of individual amino acids at specific positions on AAV capsids, independently of the serotype.
  • AAV1, AAV5 and AAV6 naturally contain the same amino acid residue at positions in their capsid protein sequences corresponding to G548, S585 and T588 in ttAAV2. Therefore in further embodiments, the capsid proteins in these serotypes are mutated at matching positions to include the other residues S312, D546, and S593 present in ttAAV2.
  • AAV8, which already contains the same amino acid residue as in ttAAV2 at positions corresponding to S312 and T588 in ttAAV2, is further mutated to contain residues corresponding to S585, D546, G548 and S593 in ttAAV2.
  • AAV9 that already contains corresponding residues to G548 and S585 in ttAAV2, is mutated to include residues corresponding to T588, S312, D546 and S593 in ttAAV2.
  • AAV10 which already contains a residue corresponding to G548, is further modified to also contain residues corresponding to S585, T588, S312, G548 and S593.
  • AAV1, and AAV6 already naturally contain the residue S205. Therefore in further embodiments these serotypes are mutated at positions corresponding to the residues I125, A151, 5162, M457, A492, D499 and Y533 in ttAAV2.
  • AAV9 that already contains the residues S162 and S205, is further mutated to contain residues corresponding to I125, A151, M457, A492, D499 and Y533 in ttAAV2.
  • AAV5, 8 and 10 are modified to display residues corresponding to I125, A151, S162, S205, M457, A492, D499 and Y533 in ttAAV2.
  • variant non-AAV2 vectors can be constructed by mutating any of the residues shown in Table 1 for these serotypes.
  • the residues shown in italics are residues which are already present in ttAAV2 at a corresponding position.
  • the non-AAV2 serotypes are mutated at one or more the residues shown in non-italic script. In this ways, the advantageous properties shown by ttAAV2 can be transferred into alternative AAV serotypes.
  • the invention also relates additional aspects, as defined in the following summary paragraphs:
  • a recombinant adeno-associated virus (AAV) vector comprising;
  • variant AAV capsid protein comprises at least one amino acid substitution with respect to a wild type AAV capsid protein; wherein the at least one amino acid substitution is present at a position corresponding to one or more of the following positions in an AAV2 capsid protein sequence: 125, 151, 162, 205, 312, 457, 492, 499, 533, 546, 548, 585, 588 and/or 593; and
  • a heterologous nucleic acid comprising a nucleotide sequence encoding a gene product.
  • a recombinant AAV vector according to paragraph 5 wherein (i) the vector comprises a variant AAV1 capsid protein, (ii) the variant AAV capsid protein comprises a sequence having at least 95% sequence identity to SEQ ID NO:3; (iii) the wild type AAV capsid protein is from AAV1; and/or (iv) the wild type AAV capsid protein comprises a sequence of SEQ ID NO:3;
  • AAV1 capsid protein sequence 125, 151, 162, 205, 313, 458, 493, 500, 534, 547, 549, 586, 589 and/or 594.
  • variant AAV1 capsid protein comprises one or more of the following amino acid substitutions with respect to a wild type AAV1 capsid protein:
  • At least one amino acid substitution is present at one or more of the following positions in the AAV5 capsid protein sequence: 124, 150, 153, 195, 303, 444, 479, 486, 520, 533, 537, 575, 578 and/or 583.
  • variant AAV5 capsid protein comprises one or more of the following amino acid substitutions with respect to a wild type AAV5 capsid protein:
  • a recombinant AAV vector according to paragraph 5 wherein (i) the vector comprises a variant AAV6 capsid protein, (ii) the variant AAV capsid protein comprises a sequence having at least 95% sequence identity to SEQ ID NO:5; (iii) the wild type AAV capsid protein is from AAV6; and/or (iv) the wild type AAV capsid protein comprises a sequence of SEQ ID NO:5;
  • AAV6 capsid protein sequence 125, 151, 162, 205, 313, 458, 493, 500, 534, 547, 549, 586, 589 and/or 594.
  • variant AAV6 capsid protein comprises one or more of the following amino acid substitutions with respect to a wild type AAV6 capsid protein:
  • At least one amino acid substitution is present at one or more of the following positions in the AAV8 capsid protein sequence: 125, 151, 163, 206, 315, 460, 495, 502, 536, 549, 551, 588, 591 and/or 596.
  • AAV9 capsid protein sequence 125, 151, 162, 205, 314, 458, 493, 500, 534, 547, 549, 586, 589 and/or 594.
  • variant AAV9 capsid protein comprises one or more of the following amino acid substitutions with respect to a wild type AAV9 capsid protein:
  • At least one amino acid substitution is present at one or more of the following positions in the AAV10 capsid protein sequence: 125, 151, 163, 206, 315, 460, 495, 502, 536, 549, 551, 588, 591 and/or 596.
  • variant AAVrh10 capsid protein comprises one or more of the following amino acid substitutions with respect to a wild type AAVrh10 capsid protein:
  • a recombinant AAV vector according to any preceding paragraph wherein the recombinant AAV vector exhibits increased transduction of a neuronal or retinal tissue compared to an AAV vector comprising a corresponding wild type AAV capsid protein.
  • a pharmaceutical composition comprising:
  • a method for delivering a gene product to a neuronal or retinal tissue in a subject comprising administering to the subject a recombinant AAV vector or pharmaceutical composition according to any preceding paragraph.
  • a method for treating a neurological or ocular disorder comprising administering to the subject a recombinant AAV vector or pharmaceutical composition according to any preceding paragraph.
  • AAV capsid protein comprising at least one amino acid substitution with respect to a wild type AAV capsid protein; wherein the at least one amino acid substitution is present at one or more of the following positions in an AAV2 capsid protein sequence: 125, 151, 162, 205, 312, 457, 492, 499, 533, 546, 548, 585, 588 and/or 593; or at one or more corresponding positions in an alternative AAV capsid protein sequence.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Organic Chemistry (AREA)
  • Genetics & Genomics (AREA)
  • General Health & Medical Sciences (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Medicinal Chemistry (AREA)
  • Animal Behavior & Ethology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Biotechnology (AREA)
  • Molecular Biology (AREA)
  • Biomedical Technology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Zoology (AREA)
  • General Engineering & Computer Science (AREA)
  • Wood Science & Technology (AREA)
  • Ophthalmology & Optometry (AREA)
  • Virology (AREA)
  • Biophysics (AREA)
  • Biochemistry (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Plant Pathology (AREA)
  • Microbiology (AREA)
  • Physics & Mathematics (AREA)
  • Epidemiology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Neurology (AREA)
  • Neurosurgery (AREA)
  • Medicines Containing Material From Animals Or Micro-Organisms (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Peptides Or Proteins (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Medicinal Preparation (AREA)
US15/570,687 2014-02-17 2015-02-17 Adeno-associated virus vector Abandoned US20180135076A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US15/570,687 US20180135076A1 (en) 2014-02-17 2015-02-17 Adeno-associated virus vector

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
US201461940639P 2014-02-17 2014-02-17
GB1403684.2 2014-03-03
GBGB1403684.2A GB201403684D0 (en) 2014-03-03 2014-03-03 Vector
US15/570,687 US20180135076A1 (en) 2014-02-17 2015-02-17 Adeno-associated virus vector
PCT/EP2015/053335 WO2015121501A1 (fr) 2014-02-17 2015-02-17 Vecteur viral adéno-associé

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2015/053335 A-371-Of-International WO2015121501A1 (fr) 2014-02-17 2015-02-17 Vecteur viral adéno-associé

Related Child Applications (2)

Application Number Title Priority Date Filing Date
US17/072,676 Continuation US11802293B2 (en) 2014-02-17 2020-10-16 Adeno-associated virus vector
US17/466,237 Continuation US20220106612A1 (en) 2014-02-17 2021-09-03 Adeno-associated virus vector

Publications (1)

Publication Number Publication Date
US20180135076A1 true US20180135076A1 (en) 2018-05-17

Family

ID=50490674

Family Applications (4)

Application Number Title Priority Date Filing Date
US15/570,687 Abandoned US20180135076A1 (en) 2014-02-17 2015-02-17 Adeno-associated virus vector
US17/072,676 Active 2035-12-03 US11802293B2 (en) 2014-02-17 2020-10-16 Adeno-associated virus vector
US17/466,237 Pending US20220106612A1 (en) 2014-02-17 2021-09-03 Adeno-associated virus vector
US18/487,398 Pending US20240124894A1 (en) 2014-02-17 2023-10-16 Adeno-associated virus vector

Family Applications After (3)

Application Number Title Priority Date Filing Date
US17/072,676 Active 2035-12-03 US11802293B2 (en) 2014-02-17 2020-10-16 Adeno-associated virus vector
US17/466,237 Pending US20220106612A1 (en) 2014-02-17 2021-09-03 Adeno-associated virus vector
US18/487,398 Pending US20240124894A1 (en) 2014-02-17 2023-10-16 Adeno-associated virus vector

Country Status (20)

Country Link
US (4) US20180135076A1 (fr)
EP (2) EP3597760A3 (fr)
JP (4) JP6602788B2 (fr)
KR (1) KR102082032B1 (fr)
CN (1) CN106062200B (fr)
AU (3) AU2015216847B2 (fr)
BR (1) BR112016018598A2 (fr)
CA (2) CA2939612C (fr)
DK (1) DK3108000T3 (fr)
ES (1) ES2749235T3 (fr)
GB (1) GB201403684D0 (fr)
HU (1) HUE045801T2 (fr)
IL (2) IL247332B (fr)
MX (1) MX2016010649A (fr)
PL (1) PL3108000T3 (fr)
PT (1) PT3108000T (fr)
RU (2) RU2021102290A (fr)
SG (1) SG11201606365TA (fr)
SI (1) SI3108000T1 (fr)
WO (1) WO2015121501A1 (fr)

Cited By (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20180057839A1 (en) * 2014-11-26 2018-03-01 The Regents Of The University Of California Therapeutic compositions comprising transcription factors and methods of making and using the same
US10745447B2 (en) 2015-09-28 2020-08-18 The University Of North Carolina At Chapel Hill Methods and compositions for antibody-evading virus vectors
WO2020219656A1 (fr) * 2019-04-26 2020-10-29 The University Of North Carolina At Chapel Hill Procédés et compositions de double liaison glycane de vecteurs de vaa2.5
WO2021030764A1 (fr) * 2019-08-14 2021-02-18 University Of Florida Research Foundation, Incorporated Variants capsidiques d'aav pour thérapie génique
CN112961219A (zh) * 2021-02-24 2021-06-15 中国农业科学院生物技术研究所 重组腺相关病毒、其突变体及其构建方法和应用
WO2022093769A1 (fr) * 2020-10-28 2022-05-05 The University Of North Carolina At Chapel Hill Procédés et compositions de double liaison glycane de vecteurs de vaa2.5
US11639509B2 (en) 2020-10-28 2023-05-02 The University Of North Carolina At Chapel Hill Methods and compositions for dual glycan binding AAV2.5 vector
CN116041443A (zh) * 2022-12-30 2023-05-02 广州派真生物技术有限公司 腺相关病毒突变体及其应用
US11802293B2 (en) 2014-02-17 2023-10-31 King's College London Adeno-associated virus vector
US11905523B2 (en) 2019-10-17 2024-02-20 Ginkgo Bioworks, Inc. Adeno-associated viral vectors for treatment of Niemann-Pick Disease type-C
US11976096B2 (en) 2018-04-03 2024-05-07 Ginkgo Bioworks, Inc. Antibody-evading virus vectors
US11981914B2 (en) 2019-03-21 2024-05-14 Ginkgo Bioworks, Inc. Recombinant adeno-associated virus vectors

Families Citing this family (143)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9217155B2 (en) 2008-05-28 2015-12-22 University Of Massachusetts Isolation of novel AAV'S and uses thereof
US8734809B2 (en) 2009-05-28 2014-05-27 University Of Massachusetts AAV's and uses thereof
WO2011133874A1 (fr) 2010-04-23 2011-10-27 University Of Massachusetts Constructions d'expression à cistrons multiples
EP3540055A1 (fr) 2010-04-23 2019-09-18 University of Massachusetts Vecteurs aav de ciblage du système nerveux central et leurs procédés d'utilisation
EP3003391B1 (fr) 2013-05-31 2021-09-22 The Regents of The University of California Variants de virus adéno-associés et leurs méthodes d'utilisation
CA2930549A1 (fr) 2013-09-26 2015-04-02 University Of Florida Research Foundation, Inc. Bibliotheque de capsides aav combinatoire synthetique pour la therapie genique ciblee
WO2015127128A2 (fr) 2014-02-19 2015-08-27 University Of Massachusetts Vaa recombinants ayant des propriétés de transcytose utiles
ES2856403T3 (es) 2014-03-18 2021-09-27 Univ Massachusetts Composiciones a base de rAAV y procedimientos para el tratamiento de la esclerosis lateral amiotrofica
US10975391B2 (en) 2014-04-25 2021-04-13 University Of Massachusetts Recombinant AAV vectors useful for reducing immunity against transgene products
US10689653B2 (en) 2014-06-03 2020-06-23 University Of Massachusetts Compositions and methods for modulating dysferlin expression
WO2015191508A1 (fr) 2014-06-09 2015-12-17 Voyager Therapeutics, Inc. Capsides chimériques
EP3200830B1 (fr) 2014-10-03 2020-09-09 University of Massachusetts Vecteurs aav identifiés au moyen de banques à efficacité élevée
WO2016054554A1 (fr) 2014-10-03 2016-04-07 University Of Massachusetts Virus adéno-associés greffés au moyen de peptides de ciblage hétérologues
WO2016065001A1 (fr) 2014-10-21 2016-04-28 University Of Massachusetts Variants de vaa recombinants et leurs utilisations
SG11201703148TA (en) 2014-11-05 2017-05-30 Voyager Therapeutics Inc Aadc polynucleotides for the treatment of parkinson's disease
CN114717264A (zh) 2014-11-14 2022-07-08 沃雅戈治疗公司 治疗肌萎缩性侧索硬化(als)的组合物和方法
KR102584655B1 (ko) 2014-11-14 2023-10-06 보이저 테라퓨틱스, 인크. 조절성 폴리뉴클레오티드
EP3230441A4 (fr) 2014-12-12 2018-10-03 Voyager Therapeutics, Inc. Compositions et méthodes pour la production de scaav
EP3256170B1 (fr) 2015-02-13 2020-09-23 University of Massachusetts Compositions et procédés pour l'administration transitoire de nucléases
US11046955B2 (en) 2015-04-24 2021-06-29 University Of Massachusetts Modified AAV constructs and uses thereof
GB201508026D0 (en) 2015-05-11 2015-06-24 Ucl Business Plc Capsid
US11426469B2 (en) 2015-10-22 2022-08-30 University Of Massachusetts Prostate-targeting adeno-associated virus serotype vectors
EP3364997B1 (fr) 2015-10-22 2024-01-17 University of Massachusetts Aspartoacylase en thérapie génique dans le traitement de la maladie de canavan
GB201519086D0 (en) * 2015-10-28 2015-12-09 Syncona Partners Llp Gene Therapy
US20180230489A1 (en) 2015-10-28 2018-08-16 Voyager Therapeutics, Inc. Regulatable expression using adeno-associated virus (aav)
PE20231949A1 (es) * 2015-10-30 2023-12-05 Spark Therapeutics Inc VARIANTES DEL FACTOR VIII REDUCIDO CON CpG, COMPOSICIONES Y METODOS Y USOS PARA EL TRATAMIENTO DE TRASTORNOS DE LA HEMOSTASIA
RU2743792C2 (ru) 2015-11-05 2021-02-26 Бамбу Терапьютикс, Инк. Модифицированные гены атаксии фридрейха и векторы для генной терапии
WO2017096162A1 (fr) 2015-12-02 2017-06-08 Voyager Therapeutics, Inc. Dosages pour la détection d'anticorps neutralisants de vaa
US11499165B2 (en) * 2015-12-11 2022-11-15 California Institute Of Technology Targeting peptides for directing adeno-associated viruses (AAVs)
CA3008280A1 (fr) * 2015-12-14 2017-06-22 The Trustees Of The University Of Pennsylvania Vecteurs viraux adeno-associes a utiliser dans le traitement de l'amyotrophie spinale
EP3411059A4 (fr) 2016-02-02 2019-10-16 University Of Massachusetts Procédé pour améliorer l'efficacité de l'administration au système nerveux central d'un gène aav par voie systémique
WO2017139643A1 (fr) 2016-02-12 2017-08-17 University Of Massachusetts Agents thérapeutiques à base de micro-arn anti-angiogéniques pour l'inhibition de la néovascularisation cornéenne
WO2017147477A1 (fr) * 2016-02-26 2017-08-31 University Of Florida Research Foundation, Inc. Mutants d'héparine aav qui présentent une transduction significativement améliorée dans l'œil et le cerveau
EP3440210A4 (fr) 2016-04-05 2019-11-27 University of Massachusetts Compositions et procédés pour l'inhibition sélective de l'expression d'une protéine de type grainhead
CN107287238B (zh) * 2016-04-11 2020-10-16 厦门继景生物技术有限责任公司 一种基因载体及其用于治疗雷柏氏先天性黑矇2型疾病的基因治疗药物
WO2017180936A1 (fr) 2016-04-15 2017-10-19 The Trustees Of The University Of Pennsylvania Composition pour le traitement de la dégénérescence maculaire liée a l'âge exsudative
US11413356B2 (en) 2016-04-15 2022-08-16 University Of Massachusetts Methods and compositions for treating metabolic imbalance
WO2017189964A2 (fr) 2016-04-29 2017-11-02 Voyager Therapeutics, Inc. Compositions pour le traitement de maladies
WO2017189959A1 (fr) 2016-04-29 2017-11-02 Voyager Therapeutics, Inc. Compositions pour le traitement de maladies
US11951121B2 (en) 2016-05-18 2024-04-09 Voyager Therapeutics, Inc. Compositions and methods for treating Huntington's disease
BR112018073384A2 (pt) 2016-05-18 2019-03-06 Voyager Therapeutics, Inc. polinucleotídeos moduladores
MA44546B1 (fr) * 2016-06-15 2021-03-31 Univ California Virus adéno-associés variants et procédés d'utilisation
US11882815B2 (en) 2016-06-15 2024-01-30 University Of Massachusetts Recombinant adeno-associated viruses for delivering gene editing molecules to embryonic cells
CA2971303A1 (fr) 2016-06-21 2017-12-21 Bamboo Therapeutics, Inc. Genes de mini-dystrophine optimises et cassettes d'expression et leur utilisation
CA3029119A1 (fr) 2016-06-29 2018-01-04 Crispr Therapeutics Ag Materiels et methodes de traitement de l'ataxie de friedreich et d'autres troubles associes
WO2018002812A1 (fr) 2016-06-29 2018-01-04 Crispr Therapeutics Ag Matériels et méthodes de traitement de la dystrophie myotonique de type 1 (dm1) et d'autres troubles associés
US11174469B2 (en) 2016-06-29 2021-11-16 Crispr Therapeutics Ag Materials and methods for treatment of Amyotrophic Lateral Sclerosis (ALS) and other related disorders
WO2018007980A1 (fr) 2016-07-06 2018-01-11 Crispr Therapeutics Ag Matériaux et méthodes de traitement de troubles liés à la douleur
WO2018007976A1 (fr) 2016-07-06 2018-01-11 Crispr Therapeutics Ag Matériaux et procédés de traitement de troubles liés à la douleur
US11458211B2 (en) 2016-07-12 2022-10-04 The University Of Manchester Gene therapy
CA3029833A1 (fr) 2016-07-29 2018-02-01 The Regents Of The University Of California Virions de virus adeno-associe a variant de capside et methodes d'utilisation associees
US20190262373A1 (en) * 2016-08-16 2019-08-29 The University Of North Carolina At Chapel Hill Methods and compositions for targeted gene transfer
WO2018044933A1 (fr) 2016-08-30 2018-03-08 The Regents Of The University Of California Procédés de ciblage et d'administration biomédicaux, et dispositifs et systèmes pour la mise en œuvre de ceux-ci
US10457940B2 (en) 2016-09-22 2019-10-29 University Of Massachusetts AAV treatment of Huntington's disease
WO2018064624A1 (fr) * 2016-09-29 2018-04-05 University Of Florida Research Foundation, Incorporated Variantes aavrh.10 à capacités d'échappement d'anticorps hôtes et propriétés de ciblage de tissu modifiées
CA3040483A1 (fr) 2016-10-13 2018-04-19 University Of Massachusetts Conceptions de capsides de vaa
US11192925B2 (en) 2016-10-19 2021-12-07 Adverum Biotechnologies, Inc. Modified AAV capsids and uses thereof
EP3585883A4 (fr) * 2017-02-21 2021-04-14 University of Florida Research Foundation, Incorporated Protéines des capsides aav modifiées et leurs utilisations
EP3585900B1 (fr) 2017-02-22 2022-12-21 CRISPR Therapeutics AG Matériels et méthodes de traitement d'ataxia spinocerebellaire de type 2 (sca2) et d'autres maladies ou de troubles liés au gene du protéine ataxia spinocerebellaire de type 2 (sca2)
US11407997B2 (en) 2017-02-22 2022-08-09 Crispr Therapeutics Ag Materials and methods for treatment of primary hyperoxaluria type 1 (PH1) and other alanine-glyoxylate aminotransferase (AGXT) gene related conditions or disorders
WO2018154387A1 (fr) 2017-02-22 2018-08-30 Crispr Therapeutics Ag Compositions et méthodes pour l'édition génétique
WO2018154439A1 (fr) 2017-02-22 2018-08-30 Crispr Therapeutics Ag Matériaux et procédés pour le traitement de l'ataxie spinocérébelleuse de type 1 (sca1) et d'autres états ou troubles liés au gène de l'ataxie spinocérébelleuse de type 1 (atxn1)
US20200040061A1 (en) 2017-02-22 2020-02-06 Crispr Therapeutics Ag Materials and methods for treatment of early onset parkinson's disease (park1) and other synuclein, alpha (snca) gene related conditions or disorders
KR20190139951A (ko) 2017-04-14 2019-12-18 리젠엑스바이오 인크. 인간 뉴런 또는 신경교 세포에 의해 생성된 재조합 인간 이두로네이트-2 설파타제(ids)를 이용한 뮤코다당증 ii형의 치료
CN110913866A (zh) 2017-05-05 2020-03-24 沃雅戈治疗公司 治疗肌萎缩性侧索硬化(als)的组合物和方法
MX2019013172A (es) 2017-05-05 2020-09-07 Voyager Therapeutics Inc Composiciones y metodos para tratar la enfermedad de huntington.
AU2018264996A1 (en) 2017-05-09 2019-12-05 University Of Massachusetts Methods of treating Amyotrophic Lateral Sclerosis (ALS)
WO2018209154A1 (fr) 2017-05-10 2018-11-15 Massachusetts Eye And Ear Infirmary Procédés et compositions pour modifier la dépendance à une protéine activant l'assemblage (app) de virus
CN110831611A (zh) * 2017-05-10 2020-02-21 马萨诸塞眼科耳科诊所 用于修饰病毒的组装活化蛋白(aap)依赖性的方法和组合物
JOP20190269A1 (ar) 2017-06-15 2019-11-20 Voyager Therapeutics Inc بولي نوكليوتيدات aadc لعلاج مرض باركنسون
BR112019019015A2 (pt) * 2017-06-30 2020-04-14 Univ California vírions de vírus adeno-associado com capsídeos variantes e seus métodos de uso
CA3070087A1 (fr) 2017-07-17 2019-01-24 Voyager Therapeutics, Inc. Systeme de guide de trajectoire d'appareillage en reseau
EP3662060A2 (fr) 2017-08-03 2020-06-10 Voyager Therapeutics, Inc. Compositions et procedes de délivrance d'aav
CN111163811A (zh) * 2017-08-25 2020-05-15 奥维德医疗公司 重组腺相关载体
CA3059995A1 (fr) 2017-08-28 2019-03-07 The Regents Of The University Of California Variants de capside de virus adeno-associe et leurs procedes d'utilisation
JP7397488B2 (ja) 2017-09-22 2023-12-13 ユニバーシティ オブ マサチューセッツ Sod1二重発現ベクターおよびその使用
EP3697908A1 (fr) 2017-10-16 2020-08-26 Voyager Therapeutics, Inc. Traitement de la sclérose latérale amyotrophique (sla)
CN111479924B (zh) 2017-10-16 2024-06-14 沃雅戈治疗公司 肌萎缩性侧索硬化症(als)的治疗
JP2021501196A (ja) * 2017-10-18 2021-01-14 リジェネックスバイオ インコーポレイテッド 完全ヒト翻訳後修飾抗体による治療剤
JP7402163B2 (ja) 2017-12-21 2023-12-20 クリスパー セラピューティクス アーゲー 2a型アッシャー症候群の処置のための材料および方法
EP3728595A1 (fr) 2017-12-21 2020-10-28 CRISPR Therapeutics AG Substances et méthodes pour le traitement du syndrome d'usher de type 2a et/ou de la rétinite pigmentaire autosomique récessive (arrp) non syndromique
JP2021519612A (ja) * 2018-03-29 2021-08-12 アスクレピオス バイオファーマシューティカル, インコーポレイテッド 中和を回避する肝臓向性組換えaav6ベクター
CN108660159B (zh) * 2018-04-12 2020-10-30 四川大学 重组蝙蝠腺相关病毒载体及其用途
KR102051393B1 (ko) 2018-05-04 2019-12-04 (주)지뉴인텍 아데노부속바이러스 기반 유전자 전달용 재조합 벡터 및 이를 이용한 암질환 예방 또는 치료용 조성물
EP3813845A4 (fr) * 2018-05-15 2022-10-26 President And Fellows Of Harvard College Vecteurs viraux présentant des propriétés d'insertion de gènes améliorées
WO2019222329A1 (fr) 2018-05-15 2019-11-21 Voyager Therapeutics, Inc. Compositions et procédés pour l'administration de vaa
WO2019222328A1 (fr) 2018-05-15 2019-11-21 Voyager Therapeutics, Inc. Compositions et méthodes pour le traitement de la maladie de parkinson
AU2019315445A1 (en) * 2018-07-31 2021-03-18 Cornell University Gene therapy methods to control organ function
CN113383010A (zh) 2018-09-28 2021-09-10 沃雅戈治疗公司 具有经工程化改造的启动子的共济蛋白表达构建体及其使用方法
US20210371470A1 (en) 2018-10-12 2021-12-02 Voyager Therapeutics, Inc. Compositions and methods for delivery of aav
UY38407A (es) 2018-10-15 2020-05-29 Novartis Ag Anticuerpos estabilizadores de trem2
MX2021008542A (es) 2019-01-18 2021-11-12 Voyager Therapeutics Inc Métodos y sistemas para la producción de partículas de aav.
EP3788826B1 (fr) 2019-03-28 2022-06-08 Ofinno, LLC Gestion de radiomessagerie centrale
US20220280655A1 (en) * 2019-04-23 2022-09-08 Institut National de la Santé et de la Recherche Médicale New Adeno-Associated Virus (AAV) Variants and Uses Thereof for Gene Therapy
US20240124889A1 (en) 2019-05-07 2024-04-18 Voyager Therapeutics, Inc. Compositions and methods for the vectored augmentation of protein destruction, expression and/or regulation
EP3990030A1 (fr) 2019-06-27 2022-05-04 Pfizer Inc. Procédés de traitement de la dystrophie musculaire de duchenne à l'aide d'une thérapie génique basée sur l'aav-mini-dystrophine
US20200407729A1 (en) 2019-06-28 2020-12-31 Crispr Therapeutics Ag Materials and methods for controlling gene editing
US20220372512A1 (en) * 2019-07-04 2022-11-24 Children's Medical Research Institute Methods and aav vectors for in vivo transduction
US20220265853A1 (en) * 2019-07-12 2022-08-25 Gene Therapy Research Institution Co., Ltd. Adeno-associated virus virion for gene transfer to human liver
JP2022541520A (ja) * 2019-07-15 2022-09-26 メイラグティーエックス ユーケー アイアイ リミティド 関節炎疾患の治療のための修飾aavキャプシドタンパク質
US20220380413A1 (en) * 2019-08-29 2022-12-01 The Board Of Trustees Of The Leland Stanford Junior University Adeno-Associated Viral Vectors for Crossing the Human Blood Brain Barrier
US20220333133A1 (en) 2019-09-03 2022-10-20 Voyager Therapeutics, Inc. Vectorized editing of nucleic acids to correct overt mutations
WO2021073568A1 (fr) * 2019-10-16 2021-04-22 Wuxi Apptec (Shanghai) Co., Ltd. Nouveau variant d'aav
TW202142552A (zh) * 2020-01-29 2021-11-16 美商健臻公司 用於眼部基因療法的經修飾的腺相關病毒衣殼蛋白及其使用方法
WO2021211753A1 (fr) 2020-04-15 2021-10-21 Voyager Therapeutics, Inc. Composés de liaison à la protéine tau
MX2022014256A (es) 2020-05-13 2023-01-11 Voyager Therapeutics Inc Redirección del tropismo de las cápsides de aav.
WO2021247995A2 (fr) 2020-06-04 2021-12-09 Voyager Therapeutics, Inc. Compositions et méthodes de traitement de la douleur neuropathique
US20230285596A1 (en) 2020-07-27 2023-09-14 Voyager Therapeutics, Inc Compositions and methods for the treatment of niemann-pick type c1 disease
BR112023001456A2 (pt) 2020-07-27 2023-04-11 Voyager Therapeutics Inc Composições e métodos para o tratamento de distúrbios neurológicos relacionados à deficiência de beta glicosilceramidase
CN111825772B (zh) * 2020-07-30 2023-10-20 中国科学院精密测量科学与技术创新研究院 具有变异衣壳蛋白的腺相关病毒及其应用
CA3181024A1 (fr) 2020-08-06 2022-02-10 Fundacion Para La Investigacion Medica Aplicada Particules virales a utiliser dans le traitement de tauopathies de type maladie d'alzheimer par therapie genique
WO2022034130A1 (fr) 2020-08-12 2022-02-17 UCB Biopharma SRL Thérapie génique utilisant des constructions d'acide nucléique comprenant des séquences promotrices de protéine 2 de liaison cpg-méthyl (mecp2)
CN112194706B (zh) * 2020-09-30 2022-03-08 广州派真生物技术有限公司 腺相关病毒突变体及其应用
KR20230082614A (ko) * 2020-10-07 2023-06-08 리젠엑스바이오 인크. 유전자 요법을 안구 전달하기 위한 아데노-연관 바이러스
PE20230987A1 (es) 2020-10-09 2023-06-21 UCB Biopharma SRL Construcciones de acido nucleico, vectores virales y particulas virales
AU2021375404A1 (en) 2020-11-03 2023-06-08 Pfizer Inc. Methods for purification of aav vectors by anion exchange chromatography
CA3201533A1 (fr) 2020-11-11 2022-05-19 European Molecular Biology Laboratory Particules virales modifiees destinees a la therapie genique
US20240052322A1 (en) 2020-12-15 2024-02-15 Pfizer Inc. HILIC UPLC-MS Method For Separating and Analyzing Intact Adeno-Associated Virus Capsid Proteins
AU2021404929A1 (en) 2020-12-21 2023-07-06 Pfizer Inc. Methods and systems for improved cell transfection
IL303959A (en) 2020-12-23 2023-08-01 Pfizer Methods for purification of AAV vectors by affinity chromatography
BR112023016075A2 (pt) 2021-02-10 2023-11-21 Regenxbio Inc Método para tratar hepatoesplenomegalia, método de distribuição de um raav que codifica hids, método para tratar um sujeito humano, método de tratamento de um sujeito humano, método para determinar a eficácia do tratamento, método para identificar um sujeito como tendo mps ii
GB202101958D0 (en) 2021-02-12 2021-03-31 Ucl Business Ltd Gene therapy for dopamine transporter deficiency syndrome
CN115044614B (zh) * 2021-03-09 2023-10-20 上海目镜生物医药科技有限公司 一种用于基因靶向与表达的aav-8型血清型的改造型载体及其构建方法及应用
WO2022208342A1 (fr) 2021-04-01 2022-10-06 Pfizer Inc. Compositions pharmaceutiques contenant un vecteur viral adéno-associé
EP4329820A1 (fr) 2021-04-26 2024-03-06 Alexion Pharma International Operations Limited Capsides de vecteur de virus adéno-associé présentant un tropisme tissulaire amélioré
CN117377771A (zh) 2021-05-12 2024-01-09 特莱索恩基金会 载体系统
WO2022269466A1 (fr) 2021-06-22 2022-12-29 Pfizer Inc. Production de vecteur de virus adéno-associé dans des cellules d'insectes
WO2023283962A1 (fr) 2021-07-16 2023-01-19 Huigene Therapeutics Co., Ltd. Capside aav modifiée pour thérapie génique et méthodes associées
WO2023069987A1 (fr) 2021-10-20 2023-04-27 University Of Rochester Traitement de régénération de référence croisée de perte de matière blanche liée à l'âge à une application associée
AU2022378524A1 (en) 2021-10-28 2024-05-02 UCB Biopharma SRL Nucleic acid constructs, viral vectors and viral particles
CA3236365A1 (fr) 2021-11-02 2023-05-11 University Of Rochester Remyelinisation mediee par tcf7l2 dans le cerveau
WO2023114816A1 (fr) 2021-12-14 2023-06-22 Neurogene, Inc. Constructions du galc optimisées recombinantes et méthodes de traitement de troubles associés au galc
CN116444626A (zh) * 2021-12-28 2023-07-18 成都弘基生物科技有限公司 经过修饰的aav衣壳蛋白及其用途
GB202201242D0 (en) 2022-01-31 2022-03-16 Univ Edinburgh Recombinant optimized mecp2 cassettes and methods for treating rett syndrome and related disorders
WO2023196818A1 (fr) 2022-04-04 2023-10-12 The Regents Of The University Of California Compositions et procédés de complémentation génétique
GB202206336D0 (en) 2022-04-29 2022-06-15 Univ Edinburgh Recombinant therapeutic FMR1 constructs and methods of treating fragile X syndrome and related disorders
WO2023214346A1 (fr) 2022-05-06 2023-11-09 Novartis Ag Nouveaux polypeptides de fusion vp2 d'aav recombinants
WO2024003687A1 (fr) 2022-06-28 2024-01-04 Pfizer Inc. Acides nucléiques codant pour l'alpha-glucosidase acide (gaa) et vecteurs pour thérapie génique
WO2024011112A1 (fr) * 2022-07-06 2024-01-11 Voyager Therapeutics, Inc. Variants de capside d'aav et leurs utilisations
WO2024015877A2 (fr) * 2022-07-12 2024-01-18 University Of Florida Research Foundation, Incorporated Nouveaux variants de capside aav3b à tropisme hépatocytaire amélioré
KR20240014846A (ko) * 2022-07-26 2024-02-02 숙명여자대학교산학협력단 Aav2-anks1a 재조합 바이러스를 포함하는 알츠하이머 질병 치료용 조성물
WO2024038365A1 (fr) 2022-08-16 2024-02-22 Pfizer Inc. Méthodes de purification de vecteurs de vaa par chromatographie d'échange d'anions
GB202216168D0 (en) 2022-10-31 2022-12-14 UCB Biopharma SRL Route of administration

Family Cites Families (56)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6306829B1 (en) 1995-12-08 2001-10-23 Hybridon, Inc. Modified VEGF oligonucleotides for treatment of skin disorders
US5639872A (en) 1993-07-27 1997-06-17 Hybridon, Inc. Human VEGF-specific oligonucleotides
CA2265460A1 (fr) 1996-09-11 1998-03-19 The Government Of The United States Of America, Represented By The Secre Tary, Department Of Health And Human Services Vecteur de vaa4 et ses utilisations
US6156303A (en) 1997-06-11 2000-12-05 University Of Washington Adeno-associated virus (AAV) isolates and AAV vectors derived therefrom
CA2745131C (fr) 1998-05-28 2016-08-09 John A. Chiorini Vecteurs d'aav5 et leurs utilisation
PT1127150E (pt) 1998-11-05 2007-08-22 Univ Pennsylvania ''sequências de ácido nucleico do vírus adeno associado do serotipo 1, vectores e células hospedeiras que as contêm''
WO2002053703A2 (fr) 2001-01-05 2002-07-11 Children's Hospital, Inc. Vecteurs aav2 et procedes
US7749492B2 (en) 2001-01-05 2010-07-06 Nationwide Children's Hospital, Inc. AAV vectors and methods
US20030144221A1 (en) 2001-07-17 2003-07-31 Isis Pharmaceuticals Inc. Antisense modulation of BCL2-associated X protein expression
WO2003042361A2 (fr) 2001-11-09 2003-05-22 Government Of The United States Of America, Department Of Health And Human Services Production d'un virus adeno-associe dans des cellules d'insectes
HU230406B1 (hu) 2001-11-13 2016-04-28 The Trustees Of The University Of Pennsylvania Eljárás adeno-asszociált vírus(AAV)szekvenciák detektálására és/vagy azonosítására és az azzal azonosított új szekvenciák izolálására
EP3339430A1 (fr) 2001-12-17 2018-06-27 The Trustees of The University of Pennsylvania Séquences de sérotype 9 de virus adéno associé, vecteurs les contenant et leurs utilisations
AU2002360291A1 (en) 2001-12-17 2003-06-30 The Trustees Of The University Of Pennsylvania Adeno-associated virus (aav) serotype 8 sequences
ES2467156T3 (es) 2001-12-21 2014-06-12 Medigene Ag Una biblioteca de genes estructurales modificados o de partículas modificadas de cápside útiles para la identificación de clones virales con tropismo celular deseado
US8802080B2 (en) 2002-05-01 2014-08-12 University Of Florida Research Foundation, Inc. Raav expression systems for genetic modification of specific capsid proteins
US7148342B2 (en) 2002-07-24 2006-12-12 The Trustees Of The University Of Pennyslvania Compositions and methods for sirna inhibition of angiogenesis
EP1486567A1 (fr) 2003-06-11 2004-12-15 Deutsches Krebsforschungszentrum Stiftung des öffentlichen Rechts Vecteur derivé d'un virus associé aux adenovirus pour la thérapie génique
EP3235827B1 (fr) 2003-06-19 2021-01-13 Genzyme Corporation Virions aav dotés d'une immunoréactivité diminuée et leurs utilisations
US9441244B2 (en) 2003-06-30 2016-09-13 The Regents Of The University Of California Mutant adeno-associated virus virions and methods of use thereof
US9233131B2 (en) 2003-06-30 2016-01-12 The Regents Of The University Of California Mutant adeno-associated virus virions and methods of use thereof
WO2005033321A2 (fr) * 2003-09-30 2005-04-14 The Trustees Of The University Of Pennsylvania Variantes des virus associes aux adenovirus (aav), sequences, vecteurs les contenant, et leur utilisation
US7947659B2 (en) 2004-03-12 2011-05-24 Alnylam Pharmaceuticals, Inc. iRNA agents targeting VEGF
NZ555830A (en) 2004-12-15 2009-01-31 Univ North Carolina Chimeric vectors
EP2359865B1 (fr) * 2005-04-07 2013-10-02 The Trustees of The University of Pennsylvania Procédé d'amélioration de la fonction d'un vecteur AAV
US7543116B2 (en) 2006-01-30 2009-06-02 International Business Machines Corporation Data processing system, cache system and method for handling a flush operation in a data processing system having multiple coherency domains
US7588772B2 (en) 2006-03-30 2009-09-15 Board Of Trustees Of The Leland Stamford Junior University AAV capsid library and AAV capsid proteins
EP2016174A2 (fr) 2006-04-28 2009-01-21 The Trustees of the University of Pennsylvania Vecteurs aav modifiés ayant une immunogénicité de capside réduite et utilisation de ceux-ci
CN101528916B (zh) 2006-04-28 2013-09-04 宾夕法尼亚大学托管会 规模可调的aav生产方法
WO2008024998A2 (fr) 2006-08-24 2008-02-28 Virovek, Inc. Expression dans des cellules d'insecte de gènes ayant des cadres ouverts de lecture chevauchants, procédés et compositions pour cela
US7872118B2 (en) 2006-09-08 2011-01-18 Opko Ophthalmics, Llc siRNA and methods of manufacture
US9611302B2 (en) 2007-04-09 2017-04-04 University Of Florida Research Foundation, Inc. High-transduction-efficiency RAAV vectors, compositions, and methods of use
US9725485B2 (en) 2012-05-15 2017-08-08 University Of Florida Research Foundation, Inc. AAV vectors with high transduction efficiency and uses thereof for gene therapy
EP2012122A1 (fr) 2007-07-06 2009-01-07 Medigene AG Protéines structurelles de parvovirus muté
WO2009108274A2 (fr) 2008-02-26 2009-09-03 The University Of North Carolina At Chapel Hill Procédés et compositions pour virus adéno-associés (aav) présentant des mutations de la boucle hi
WO2009137006A2 (fr) 2008-04-30 2009-11-12 The University Of North Carolina At Chapel Hill Évolution dirigée et méthode d'adhérence sur plastique in vivo de vecteurs viraux
WO2010093784A2 (fr) 2009-02-11 2010-08-19 The University Of North Carolina At Chapel Hill Vecteurs viraux modifiés et procédés de fabrication et d'utilisation de ceux-ci
US10920244B2 (en) 2009-04-30 2021-02-16 The Trustees Of The University Of Pennsylvania Compositions for targeting conducting airway cells comprising adeno-associated virus constructs
US8734809B2 (en) 2009-05-28 2014-05-27 University Of Massachusetts AAV's and uses thereof
US9133479B2 (en) 2009-06-03 2015-09-15 Cedars-Sinai Medical Center Effective vector platform for gene transfer and gene therapy
US8718118B2 (en) 2009-07-22 2014-05-06 Aware, Inc. Packet detector
EP3540055A1 (fr) 2010-04-23 2019-09-18 University of Massachusetts Vecteurs aav de ciblage du système nerveux central et leurs procédés d'utilisation
CN102071206A (zh) * 2010-10-22 2011-05-25 中山大学 一种腺相关病毒衣壳蛋白基因、相应的蛋白及其应用
EP2699270B1 (fr) 2011-04-22 2017-06-21 The Regents of The University of California Virions de virus adéno-associé avec capside variant et procédés d'utilisation de ceux-ci
WO2013029030A1 (fr) 2011-08-24 2013-02-28 The Board Of Trustees Of The Leland Stanford Junior University Protéines capsidiques d'aav inédites pouvant être utilisées pour le transfert d'acides nucléiques
PT2839014T (pt) 2012-04-18 2021-03-19 Childrens Hospital Philadelphia ¿composição e métodos para transferência de genes altamente eficiente com a utilização de variantes de capsídeo de aav
EP2847337A4 (fr) 2012-05-09 2016-04-27 Univ Oregon Health & Science Plasmides et vecteurs viraux associés à un adénovirus
US10294281B2 (en) 2012-05-15 2019-05-21 University Of Florida Research Foundation, Incorporated High-transduction-efficiency rAAV vectors, compositions, and methods of use
TWI702955B (zh) 2012-05-15 2020-09-01 澳大利亞商艾佛蘭屈澳洲私營有限公司 使用腺相關病毒(aav)sflt-1治療老年性黃斑部退化(amd)
WO2014186746A1 (fr) 2013-05-16 2014-11-20 University Of Florida Research Foundation, Inc. Éléments d'arnm en épingle à cheveux et procédés de régulation de la traduction des protéines
GB201403684D0 (en) 2014-03-03 2014-04-16 King S College London Vector
ES2768763T3 (es) 2014-03-04 2020-06-23 Univ Florida Vectores rAAV mejorados y métodos para la transducción de fotorreceptores y células EPR
WO2016133917A1 (fr) 2015-02-16 2016-08-25 University Of Florida Research Foundation Compositions à base de vecteurs raav, procédés de ciblage des cellules endothéliales vasculaires et utilisation dans le traitement du diabète de type i
ES2865487T3 (es) 2015-09-28 2021-10-15 Univ North Carolina Chapel Hill Métodos y composiciones para vectores virales que evaden los anticuerpos
WO2017066764A2 (fr) 2015-10-16 2017-04-20 William Marsh Rice University Modification de la région n-terminale des protéines de capsides pour améliorer les propriétés des virus adéno-associés
WO2017189959A1 (fr) 2016-04-29 2017-11-02 Voyager Therapeutics, Inc. Compositions pour le traitement de maladies
US11458211B2 (en) 2016-07-12 2022-10-04 The University Of Manchester Gene therapy

Cited By (13)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11802293B2 (en) 2014-02-17 2023-10-31 King's College London Adeno-associated virus vector
US20180057839A1 (en) * 2014-11-26 2018-03-01 The Regents Of The University Of California Therapeutic compositions comprising transcription factors and methods of making and using the same
US10745447B2 (en) 2015-09-28 2020-08-18 The University Of North Carolina At Chapel Hill Methods and compositions for antibody-evading virus vectors
US11208438B2 (en) 2015-09-28 2021-12-28 The University Of North Carolina At Chapel Hill Methods and compositions for antibody-evading virus vectors
US11976096B2 (en) 2018-04-03 2024-05-07 Ginkgo Bioworks, Inc. Antibody-evading virus vectors
US11981914B2 (en) 2019-03-21 2024-05-14 Ginkgo Bioworks, Inc. Recombinant adeno-associated virus vectors
WO2020219656A1 (fr) * 2019-04-26 2020-10-29 The University Of North Carolina At Chapel Hill Procédés et compositions de double liaison glycane de vecteurs de vaa2.5
WO2021030764A1 (fr) * 2019-08-14 2021-02-18 University Of Florida Research Foundation, Incorporated Variants capsidiques d'aav pour thérapie génique
US11905523B2 (en) 2019-10-17 2024-02-20 Ginkgo Bioworks, Inc. Adeno-associated viral vectors for treatment of Niemann-Pick Disease type-C
US11639509B2 (en) 2020-10-28 2023-05-02 The University Of North Carolina At Chapel Hill Methods and compositions for dual glycan binding AAV2.5 vector
WO2022093769A1 (fr) * 2020-10-28 2022-05-05 The University Of North Carolina At Chapel Hill Procédés et compositions de double liaison glycane de vecteurs de vaa2.5
CN112961219A (zh) * 2021-02-24 2021-06-15 中国农业科学院生物技术研究所 重组腺相关病毒、其突变体及其构建方法和应用
CN116041443A (zh) * 2022-12-30 2023-05-02 广州派真生物技术有限公司 腺相关病毒突变体及其应用

Also Published As

Publication number Publication date
ES2749235T3 (es) 2020-03-19
KR102082032B1 (ko) 2020-02-26
JP6602788B2 (ja) 2019-11-06
PL3108000T3 (pl) 2020-05-18
KR20160130996A (ko) 2016-11-15
AU2015216847A1 (en) 2016-08-11
CA2939612C (fr) 2021-10-12
AU2023258448A1 (en) 2023-12-07
JP7309780B2 (ja) 2023-07-18
JP2021137016A (ja) 2021-09-16
RU2743382C2 (ru) 2021-02-17
HUE045801T2 (hu) 2020-01-28
SI3108000T1 (sl) 2020-02-28
CA2939612A1 (fr) 2015-08-20
PT3108000T (pt) 2019-10-25
US20220106612A1 (en) 2022-04-07
EP3108000A1 (fr) 2016-12-28
EP3108000B1 (fr) 2019-08-28
EP3597760A2 (fr) 2020-01-22
CA3126061C (fr) 2023-08-29
CN106062200A (zh) 2016-10-26
JP2017506521A (ja) 2017-03-09
AU2021202538A1 (en) 2021-05-27
IL279154A (en) 2021-01-31
US11802293B2 (en) 2023-10-31
IL247332B (en) 2021-07-29
CA3126061A1 (fr) 2015-08-20
SG11201606365TA (en) 2016-09-29
IL279154B (en) 2022-06-01
BR112016018598A2 (pt) 2017-10-17
WO2015121501A1 (fr) 2015-08-20
JP6942166B2 (ja) 2021-09-29
AU2015216847B2 (en) 2021-05-20
JP2020022476A (ja) 2020-02-13
JP2023126885A (ja) 2023-09-12
RU2016133623A (ru) 2018-03-22
CN106062200B (zh) 2020-06-23
US20210238631A1 (en) 2021-08-05
DK3108000T3 (da) 2019-10-28
US20240124894A1 (en) 2024-04-18
MX2016010649A (es) 2017-05-04
RU2016133623A3 (fr) 2018-11-16
IL247332A0 (en) 2016-09-29
AU2021202538B2 (en) 2023-08-03
GB201403684D0 (en) 2014-04-16
RU2021102290A (ru) 2021-03-01
EP3597760A3 (fr) 2020-03-11

Similar Documents

Publication Publication Date Title
US11802293B2 (en) Adeno-associated virus vector
US20220243291A1 (en) Adeno-Associated Virus Virions with Variant Capsid and Methods of Use Thereof
KR20200022372A (ko) 변이체 캡시드를 보유한 아데노-부속 바이러스 비리온 및 이의 사용 방법

Legal Events

Date Code Title Description
AS Assignment

Owner name: ICAHN SCHOOL OF MEDICINE AT MOUNT SINAI, NEW YORK

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:LINDEN, RALPH MICHAEL;REEL/FRAME:044781/0355

Effective date: 20180109

Owner name: KING'S COLLEGE LONDON, UNITED KINGDOM

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:LINDEN, RALPH MICHAEL;REEL/FRAME:044781/0355

Effective date: 20180109

STPP Information on status: patent application and granting procedure in general

Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: FINAL REJECTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: FINAL REJECTION MAILED

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION