WO2017070476A2 - Synthetic combinatorial aav3 capsid library - Google Patents

Synthetic combinatorial aav3 capsid library Download PDF

Info

Publication number
WO2017070476A2
WO2017070476A2 PCT/US2016/058130 US2016058130W WO2017070476A2 WO 2017070476 A2 WO2017070476 A2 WO 2017070476A2 US 2016058130 W US2016058130 W US 2016058130W WO 2017070476 A2 WO2017070476 A2 WO 2017070476A2
Authority
WO
WIPO (PCT)
Prior art keywords
group
independently selected
amino acid
aav
acid residue
Prior art date
Application number
PCT/US2016/058130
Other languages
French (fr)
Other versions
WO2017070476A3 (en
Inventor
Sergei Zolotukhin
Damien MARSIC
Original Assignee
University Of Florida Research Foundation, Inc.
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
Application filed by University Of Florida Research Foundation, Inc. filed Critical University Of Florida Research Foundation, Inc.
Priority to US15/769,615 priority Critical patent/US20200181644A1/en
Publication of WO2017070476A2 publication Critical patent/WO2017070476A2/en
Publication of WO2017070476A3 publication Critical patent/WO2017070476A3/en

Links

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
    • 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
    • C12N7/00Viruses; Bacteriophages; Compositions thereof; Preparation or purification thereof
    • CCHEMISTRY; METALLURGY
    • C40COMBINATORIAL TECHNOLOGY
    • C40BCOMBINATORIAL CHEMISTRY; LIBRARIES, e.g. CHEMICAL LIBRARIES
    • C40B40/00Libraries per se, e.g. arrays, mixtures
    • C40B40/04Libraries containing only organic compounds
    • C40B40/06Libraries containing nucleotides or polynucleotides, or derivatives thereof
    • C40B40/08Libraries containing RNA or DNA which encodes proteins, e.g. gene libraries
    • 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
    • C12N2750/14141Use 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
    • 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
    • C12N2810/00Vectors comprising a targeting moiety
    • C12N2810/50Vectors comprising as targeting moiety peptide derived from defined protein
    • C12N2810/60Vectors comprising as targeting moiety peptide derived from defined protein from viruses
    • C12N2810/6027Vectors comprising as targeting moiety peptide derived from defined protein from viruses ssDNA viruses

Definitions

  • Adeno-associated virus is a single-stranded DNA virus belonging to the Parvoviridae family (Muzyczka and Berns, 2001).
  • AAV-derived vectors are promising tools for human gene therapy applications because of their absence of pathogenicity, low immunogenicity, episomal localization and stable transgene expression.
  • significant limitations to the clinical use of AAV are its promiscuity and its susceptibility to neutralization by human antibodies (Jeune et al, 2013). Both of these limitations are determined by nature of the amino acid residues exposed at the surface of the capsid. Therefore, major efforts aiming at developing useful and effective gene therapy vectors have been devoted to obtaining and studying capsid variants (Wu et al, 2006).
  • the first approach was to study naturally occurring AAV isolates. So far, 13 serotypes have been formally characterized and hundreds of variant isolates have been sequenced. Additional capsid variation has been investigated through the generation of mosaics (viral particles made of capsid proteins from more than one serotype) (Hauck et al, 2003; Stachler and Bartlett, 2006; Gigout et al, 2005), chimeras (capsid proteins with domains from various origins) (Shen et al, 2007), and various substitutional or insertional mutants (Wu et al, 2000). However, the most significant advances are expected to result from directed evolution approaches through the development of capsid libraries.
  • Random display peptide libraries (Govindasamy et al, 2006) are limited to an insertion at one particular capsid location.
  • Libraries generated using error-prone PCR contain a very small fraction of gene variants encoding proteins that can fold properly and assemble into a functional capsid, due to the randomness of the mutations.
  • DNA shuffling and staggered extension processes are more efficient because they recombine naturally-occurring parental sequences and therefore are more likely to generate actual capsid variants. However, they can only recombine blocks of DNA as opposed to single nucleotide positions, which results in sequence bias (parental polymorphisms will tend to cluster together instead of being randomly distributed).
  • An embodiment of a non-naturally occurring nucleic acid of these teachings includes (a) a first nucleotide sequence encoding at least one AAV Rep protein from serotype 3; (b) a second nucleotide sequence encoding at least one AAV Cap protein differing from wildtype serotype 3 at least at one nucleotide position; and (c) a first AAV terminal repeat from serotype 3 and a second AAV terminal repeat from serotype 3, where the first and second nucleotide sequences are interposed between the first and second AAV terminal repeat.
  • An aspect of an embodiment of the non-naturally occurring nucleic acid of these teachings further includes a third nucleotide sequence encoding at least one molecule providing helper function.
  • the third nucleotide sequence can be a polynucleotide from an adenovirus or a herpes virus, preferably adenovirus.
  • An embodiment of a vector library of these teachings includes at least a first vector and a second vector, and each vector includes (a) a first nucleotide sequence encoding at least one AAV Rep protein from serotype 3; (b) a second nucleotide sequence encoding at least one AAV Cap protein differing from wildtype serotype 3 at least at one nucleotide position; and (c) a first AAV terminal repeat from serotype 3 and a second AAV terminal repeat from serotype 3, where the first and second nucleotide sequences are interposed between the first and second AAV terminal repeat, and the second vector differs from the first vector by at least one nucleotide.
  • An aspect of an embodiment of the vector library of these teachings includes the vector library being incorporated into at least one host cell.
  • suitable host cells include HEK293 embryonic kidney cells, HeLa cells, Cos cells, U87 cells, KB cells, HepG2 cells and Vero cells, preferably HEK293 embryonic kidney cells.
  • An aspect of an embodiment of the vector library of these teachings further includes a third nucleotide sequence encoding at least one molecule providing helper function.
  • the third nucleotide sequence can be a polynucleotide from an adenovirus or a herpes virus, preferably adenovirus.
  • An embodiment of an AAV virion of these teachings includes (a) a first nucleotide sequence encoding at least one AAV Rep protein from serotype 3; (b) a second nucleotide sequence encoding at least one AAV Cap protein differing from wildtype serotype 3 at least at one nucleotide position; and (c) a first AAV terminal repeat from serotype 3 and a second AAV terminal repeat from serotype 3, where the first and second nucleotide sequences are interposed between the first and second AAV terminal repeat.
  • An aspect of an embodiment of the AAV virion of these teachings includes the AAV virion being incorporated into at least one host cell.
  • suitable host cells are mammalian cells including human host cells, including, for example blood cells, stem cells, hematopoietic cells, CD34 ' cells, liver cells, cancer cells, vascular cells, pancreatic cells, neural cells, ocular or retinal cells, epithelial or endothelial cells, dendritic cells, fibroblasts, or any other cell of mammalian origin, including, without limitation, hepatic
  • liver cells i.e., liver cells
  • lung cells cardiac cells, pancreatic cells, intestinal cells, diaphragmatic cells, renal (i.e., kidney) cells, neural cells, blood cells, bone marrow cells, or any one or more selected tissues of a mammal for which viral-based gene therapy is contemplated.
  • the host cells are liver cells.
  • An aspect of an embodiment of the AAV virion of these teachings further includes a third nucleotide sequence encoding at least one molecule providing helper function.
  • the third nucleotide sequence can be a polynucleotide from an adenovirus or a herpes virus, preferably adenovirus.
  • non-naturally occurring nucleic acid and the vector library and the AAV virion of these teachings include the second nucleotide sequence encoding an AAV Cap protein that differs from wildtype serotype 3 at least at one amino position.
  • the at least one differing amino acid position is preferably in a variable region (VR), and can be in VR-I, VR-IV, VR-V, VR-VI, VR-VII, VR-VIII and combinations thereof.
  • Non-naturally occurring nucleic acid and the vector library and the AAV virion of these teachings include VR-I encoding amino acid sequence X1X2GAX3 where Xi is independently Q, N, K, T, S, R, H, P, D, E, A or G; X2 is independently S, T or A; and X3 is independently S or T.
  • Non-naturally occurring nucleic acid and the vector library and the AAV virion of these teachings include VR-IV encoding amino acid sequence X4X5X6X7GTX8X9X10X11X12LX13 where X 4 is independently G or S; X5 is independently T or N; X 6 is independently T, P or A; X7 is independently S or G; Xs is independently T or A; X9 is independently N, T, S, D, A or G; X10 is independently Q, H, P, L, K, N, T, M or I; X11 is independently S, Q, H, R, K or N; X12 is independently R, K, N, T, S, Q, H, P, E, D, A or G; and Xi3 is independently L, K, T, R, M, Q, P, E, A, G or V.
  • Non-naturally occurring nucleic acid and the vector library and the AAV virion of these teachings include VR-V encoding amino acid sequence X14X15X16X17X18 NNSX19FPWX20AASX21 where Xi4 is independently K or T; X15 is independently T, I, A or V; Xi6 is independently A, P, H, D, S or Y; X17 is independently N, T, S, D, A or G; Xis is independently D, E, G, Q, H or R; X19 is independently N, K, E or D; X20 is independently T or P; and X21 is independently K or T.
  • Non-naturally occurring nucleic acid and the vector library and the AAV virion of these teachings include VR-VI encoding amino acid sequence X22DDX23X24X25 where X22 is independently K, R, E or G; X23 is independently E, T, K, N, A or D; X24 is independently E, D or G; and X25 is independently K or R.
  • Non-naturally occurring nucleic acid and the vector library and the AAV virion of these teachings include VR-VII encoding amino acid sequence X26X27X28X29X30X31X32X33X34X35X36X37 where X26 is independently E or Q; X27 is independently G, N, S or D; X28 is independently T, S, G or A; X29 is independently T, K, N, R, S, E, D, A or G; X30 is independently A, K, T, R, E or G; X31 is independently S, N, T, D, A or G; X32 is independently N or D; X33 is independently A, T, I or V; X34 is independently E, A or D; X35 is independently L, N, I, D, V, Y or F; X36 is independently D, E or G; and X37 is independently N, K, R, S, Q, H, E, D or G.
  • Non-naturally occurring nucleic acid and the vector library and the AAV virion of these teachings include VR-VIII encoding amino acid sequence X38X39X40X41X42PTX43X44X45VX46 where X38 is independently S, K, N, T, R, E, D, A or G; X39 is independently S, K, N, T, R, Q, H, P, E, D, A or G; X 4 o is independently N, Q, H, R, K or S; X 4 i is independently T, N, S, D, A or G; X42 is independently A, K, N, T, R, S, Q, H, P, E, D or G; X43 is independently T, N, I, D, A, V, Y, S or F; X 4 4 is independently G, K, N, T, R, S, Q, H, P, E, D or A; X45 is independently T, K, N, R, M, I, E, D
  • nucleic acid and the vector library and the AAV virion of these teachings include the second nucleotide sequence encoding an AAV Cap protein having the sequence:
  • nucleic acid and the vector library and the AAV virion of these teachings include the second nucleotide encoding variants of an AAV Cap protein as listed in Table 4 (sequences numbered 2-86).
  • An embodiment of an AAV virion of these teachings includes (a) a first nucleotide sequence encoding at least one therapeutic molecule; (b) a second nucleotide sequence comprising a regulatory sequence; (c) a third nucleotide sequence comprising a first AAV terminal repeat from serotype 3; (d) a fourth nucleotide sequence comprising a second AAV terminal repeat from serotype 3; and (e) a capsid comprising at least one AAV Cap protein that differs from wildtype serotype 3 at least at one amino acid position.
  • the first nucleotide sequence is operably linked to the second nucleotide sequence and the first and second nucleotide sequences are interposed between the first and second AAV terminal repeat to form a transgene, and the resulting transgene is packaged within the capsid.
  • suitable regulatory sequences include promoters and enhancers, preferably a tissue specific promoeter.
  • suitable therapeutic molecules include polypeptides, peptides, antibody, antigen binding fragment, ribozyme, peptide nucleic acid, siRNA, RNAi, antisense oligonucleotide, antisense polynucleotide, and any combination thereof, preferably a polypeptide, a peptide or an RNA.
  • An embodiment of a method of treating a disease of these teachings includes administering an effective amount of an AAV virion of these teachings.
  • Such an AAV virion includes includes (a) a first nucleotide sequence encoding at least one therapeutic molecule; (b) a second nucleotide sequence comprising a regulatory sequence; (c) a third nucleotide sequence comprising a first AAV terminal repeat from serotype 3; (d) a fourth nucleotide sequence comprising a second AAV terminal repeat from serotype 3; and (e) a capsid comprising at least one AAV Cap protein that differs from wildtype serotype 3 at least at one amino acid position.
  • the first nucleotide sequence is operably linked to the second nucleotide sequence and the first and second nucleotide sequences are interposed between the first and second AAV terminal repeat to form a transgene, and the resulting transgene is packaged within the capsid.
  • suitable regulatory sequences include promoters and enhancers, preferably a tissue specific promoeter.
  • suitable therapeutic molecules include polypeptides, peptides, antibody, antigen binding fragment, ribozyme, peptide nucleic acid, siRNA, RNAi, antisense oligonucleotide, antisense polynucleotide, and any combination thereof, preferably a polypeptide, a peptide or an RNA.
  • An embodiment of a method of selecting tissue-specific or cell-specific variants of an AAV virion includes (a) introducing a plurality of AAV virions into target tissues or cells; (b) allowing sufficient time to elapse to propagate additional virions; and (c) isolating the virions.
  • Such an AAV virion includes (a) a first nucleotide sequence encoding at least one AAV Rep protein from serotype 3; (b) a second nucleotide sequence encoding at least one AAV Cap protein differing from wildtype serotype 3 at least at one nucleotide position; and (c) a first AAV terminal repeat from serotype 3 and a second AAV terminal repeat from serotype 3, where the first and second nucleotide sequences are interposed between the first and second AAV terminal repeat. Steps (a) - (c) can be repeated one or more times to enrich for a tissue- specific or cell-specific variant. Such enriched variants exhibit a higher target tropism for the target tissues or cells as compared to AAV serotype 3.
  • Figure 1 shows the wildtype (WT) nucleotide sequence (bottom rows) and corresponding WT amino acids (top rows, bold font) of AAV3B capsid gene and capsid protein, respectively.
  • WT wildtype nucleotide sequence
  • A3CL AAV serotype 3 capsid library
  • Figure 2 shows the nucleotide sequence of the synthetic fragment A3CL as designed.
  • the degenerate nucleotide positions (in IUPAC code) are underlined. The overlap stretches of the synthetic DNA and the plasmid vector backbone are highlighted.
  • Figure 3 shows the amino acid sequence of AAV3B VP1. Degenerate positions are labeled by X and underlined.
  • Figure 4 shows the amino acid sequences of the A3CL VRs encompassing WT AAV3B
  • VP1 capsid residues 259-600 WT sequences are shown in black, degenerate residues - in italics. Not modified conservative residues between VRs are not shown. VRs borders are indicated by vertical lines.
  • Figure 5 is a flowchart illustrating design and construction of AAV3B (A3CL) combinatorial capsid libraries ABC and D.
  • Figures 6-9 are photographs of agarose gels showing products of PCR reactions as per Example 2.
  • the present invention also provides improved rAAV-based genetic constructs that encode one or more therapeutic agents useful in the preparation of medicaments for the prevention, treatment, and/or amelioration of one or more diseases, disorders or dysfunctions resulting from a deficiency in one or more cellular components.
  • the invention provides libraries of rAAV-based genetic constructs encoding one or more selected molecules of interest, such as, for example, one or more diagnostic or therapeutic agents (including, e.g., proteins, polypeptides, peptides, antibodies, antigen binding fragments, siRNAs, RNAis, antisense oligo- and poly-nucleotides, ribozymes, and variants and/or active fragments thereof), for use in the diagnosis, prevention, treatment, and/or amelioration of symptoms of mammalian diseases, disorders, dysfunctions, deficiencies, defects, trauma, injury, and such like.
  • diagnostic or therapeutic agents including, e.g., proteins, polypeptides, peptides, antibodies, antigen binding fragments, siRNAs, RNAis, antisense oligo- and poly-nucleotides, ribozymes, and variants and/or active fragments thereof.
  • the present invention also provides infectious rAAV virions, as well as nucleic acid molecules and rAAV vectors that encode the novel AAV vectors described herein, as well as nucleic acids encoding one or more selected diagnostic and/or therapeutic agents for delivery to a selected population of mammalian cells.
  • the novel rAAV vectors, express constructs, and infectious virions and viral particles comprising them as disclosed herein preferably have an improved efficiency in transducing one or more of a variety of cells, tissues and organs of interest, when compared to wild-type, unmodified, expression constructs, and to the corresponding rAAV vectors and virions comprising them.
  • the improved rAAV vectors provided herein may transduce one or more selected host cells at higher-efficiencies (and often much higher efficiencies) than conventional, wild type (i.e., "unmodified") rAAV vectors.
  • vectors prepared as described herein may be of different AAV serotypes, and the mutation of one or more of the sequences described herein may result in improved viral vectors, which are capable of higher-efficiency transduction than that of the corresponding, non- substituted vectors from which the mutants were prepared.
  • next-generation rAAV viral vectors may dramatically reduce the number of viral particles needed for a conventional gene therapy regimen.
  • the rAAV vectors prepared as described herein may be more stable, less immunogenic, and/or can be produced at much lower cost, or in a higher titer, than an equivalent wild type viral vector prepared in conventional fashion.
  • native amino acids normally present in the sequence of a viral capsid protein may be substituted by one or more non-native amino acids, including, a substitution of one or more amino acids not normally present at a particular residue in the corresponding wild-type protein.
  • the invention also provides isolated and purified polynucleotides that encode one or more of the disclosed viral vectors as described herein, as well as polynucleotides that encode such vectors.
  • the vector constructs of the present invention further include at least promoter capable of expressing the nucleic acid segment in a suitable host cell comprising the vector.
  • the transduction efficiency of a mutated rAAV vector will be higher than that of the corresponding, unmodified, wild-type vector, and as such, will preferably possess a transduction efficiency in a mammalian cell that is at least 2-fold, at least about 4-fold, at least about 6-fold, at least about 8— fold, at least about 10-fold, or at least about 12-fold or higher in a selected mammalian host cell than that of a virion that comprises a corresponding, unmodified, rAAV vector.
  • the transduction efficiency of the rAAV vectors provided herein will be at least about 15-fold higher, at least about 20-fold higher, at least about 25-fold higher, at least about 30-fold higher, or at least about 40, 45, or 50-fold or more greater than that of a virion that comprises a corresponding, wild-type vectors.
  • the present invention also concerns rAAV vectors, wherein the nucleic acid segment further comprises a promoter, an enhancer, a post-transcriptional regulatory sequence, a polyadenylation signal, or any combination thereof, operably linked to the nucleic acid segment that encodes the selected polynucleotide of interest.
  • the promoter is a heterologous promoter, a tissue-specific promoter, a cell-specific promoter, a constitutive promoter, an inducible promoter, or any combination thereof.
  • nucleic acid segments cloned into one or more of the novel rAAV expression vectors described herein will preferably express or encode one or more polypeptides, peptides, ribozymes, peptide nucleic acids, siRNAs, RNAis, antisense oligonucleotides, antisense polynucleotides, antibodies, antigen binding fragments, or any combination thereof.
  • the therapeutic agents useful in the invention may include one or more agonists, antagonists, anti-apoptosis factors, inhibitors, receptors, cytokines, cytotoxins, erythropoietic agents, glycoproteins, growth factors, growth factor receptors, hormones, hormone receptors, interferons, interleukins, interleukin receptors, nerve growth factors, neuroactive peptides, neuroactive peptide receptors, proteases, protease inhibitors, protein decarboxylases, protein kinases, protein kinase inhibitors, enzymes, receptor binding proteins, transport proteins or one or more inhibitors thereof, serotonin receptors, or one or more uptake inhibitors thereof, serpins, serpin receptors, tumor suppressors, diagnostic molecules, chemotherapeutic agents, cytotoxins, or any combination thereof.
  • the invention further provides populations and pluralities of such rAAV vectors as prepared herein, as well as virions, infectious viral particles, and mammalian host cells that include one or more nucleic acid segments encoding them.
  • the mammalian host cells will be human host cells, including, for example blood cells, stem cells, hematopoietic cells, CD34 ' cells, liver cells, cancer cells, vascular cells, pancreatic cells, neural cells, ocular or retinal cells, epithelial or endothelial cells, dendritic cells, fibroblasts, or any other cell of mammalian origin, including, without limitation, hepatic (i.e., liver) cells, lung cells, cardiac cells, pancreatic cells, intestinal cells, diaphragmatic cells, renal (i.e., kidney) cells, neural cells, blood cells, bone marrow cells, retinal cells or any one or more selected tissues of a mammal for which viral-based gene therapy is contemplated.
  • human host cells including, for example blood cells, stem cells, hematopoietic cells, CD34 ' cells, liver cells, cancer cells, vascular cells, pancreatic cells, neural cells, ocular or retinal cells, epithelial or endotheli
  • compositions that include one or more of the proteins nucleic acid segments viral vectors, host cells, or viral particles of the present invention together with one or more pharmaceutically-acceptable buffers, diluents, or excipients.
  • Such compositions may be included in one or more diagnostic or therapeutic kits, for diagnosing, preventing, treating or ameliorating one or more symptoms of a mammalian disease, injury, disorder, trauma or dysfunction.
  • the invention further includes a method for providing a mammal in need thereof with a diagnostically- or therapeutically-effective amount of a selected biological molecule, the method comprising providing to a cell, tissue or organ of a mammal in need thereof, an amount of an rAAV vector; and for a time effective to provide the mammal with a diagnostically- or a therapeutically-effective amount of the selected biological molecule.
  • the invention further provides a method for diagnosing, preventing, treating, or ameliorating at least one or more symptoms of a disease, a disorder, a dysfunction, an injury, an abnormal condition, or trauma in a mammal.
  • the method includes at least the step of administering to a mammal in need thereof one or more of the disclosed rAAV vectors, in an amount and for a time sufficient to diagnose, prevent, treat or ameliorate the one or more symptoms of the disease, disorder, dysfunction, injury, abnormal condition, or trauma in the mammal.
  • the invention also provides a method of transducing a population of mammalian cells.
  • the method includes at least the step of introducing into one or more cells of the population, a composition that comprises an effective amount of one or more of the rAAV vectors disclosed herein.
  • the invention also provides isolated nucleic acid segments that encode one or more of the mutant viral capsid proteins as described herein, and provides recombinant vectors, virus particles, infectious virions, and isolated host cells that comprise one or more of the improved vector sequences described and tested herein.
  • compositions as well as therapeutic and/or diagnostic kits that include one or more of the disclosed AAV compositions, formulated with one or more additional ingredients, or prepared with one or more instructions for their use.
  • the invention also demonstrates methods for making, as well as methods of using the disclosed improved rAAV vectors in a variety of ways, including, for example, ex situ, in vitro and in vivo applications, methodologies, diagnostic procedures, and/or gene therapy regimens. Because many of the improved vectors described herein are also resistant to proteasomal degradation, they possess significantly increased transduction efficiencies in vivo making them particularly well suited for viral vector-based human gene therapy regimens, and in particular, for delivering one or more genetic constructs to selected mammalian cells in vivo and/or in vitro.
  • the invention provides compositions comprising AAV vectors, virions, viral particles, and pharmaceutical formulations thereof, useful in methods for delivering genetic material encoding one or more beneficial or therapeutic product(s) to mammalian cells and tissues.
  • compositions and methods of the invention provide a significant advancement in the art through their use in the treatment, prevention, and/or amelioration of symptoms of one or more mammalian diseases. It is contemplated that human gene therapy will particularly benefit from the present teachings by providing new and improved viral vector constructs for use in the treatment of a number of diverse diseases, disorders, and dysfunctions.
  • the invention concerns libraries of rAAV vector mutants that demonstrate improved properties useful in the delivery of one or more therapeutic agents to selected mammalian cells, and particularly for use in the prevention, treatment, and/or amelioration of one or more disorders in a mammal into which the vector construct may be introduced.
  • the rAAV vectors of the present invention may optionally further include one or more enhancer sequences that are each operably linked to the nucleic acid segment.
  • enhancer sequences include, but are not limited to, one or more selected from the group consisting of a CMV enhancer, a synthetic enhancer, a liver-specific enhancer, an vascular- specific enhancer, a brain-specific enhancer, a neural cell-specific enhancer, a lung-specific enhancer, a muscle-specific enhancer, a kidney-specific enhancer, a pancreas-specific enhancer, retinal-specific enhancer and an islet cell-specific enhancer.
  • promoters useful in the practice of the invention include, without limitation, one or more heterologous, tissue-specific, constitutive or inducible promoters, including, for example, but not limited to, a promoter selected from the group consisting of a CMV promoter, a I3-actin promoter, an insulin promoter, an enolase promoter, a BDNF promoter, an NGF promoter, an EGF promoter, a growth factor promoter, an axon-specific promoter, a dendrite- specific promoter, a brain-specific promoter, a hippocampal-specific promoter, a kidney- specific promoter, a retinal-specific promoter, an elafin promoter, a cytokine promoter, an interferon promoter, a growth factor promoter, an ai-antitrypsin promoter, a brain cell-specific promoter, a neural cell-specific promoter, a central nervous system cell-specific promoter, a peripheral nervous system cell-specific promoter
  • the vector-encoding nucleic acid segments may also further include one or more post- transcriptional regulatory sequences or one or more polyadenylation signals, including, for example, but not limited to, a woodchuck hepatitis virus post-transcription regulatory element, a polyadenylation signal sequence, or any combination thereof.
  • Exemplary diagnostic or therapeutic agents deliverable to host cells by the present vector constructs include, but are not limited to, an agent selected from the group consisting of a polypeptide, a peptide, an antibody, an antigen binding fragment, a ribozyme, a peptide nucleic acid, a siRNA, an RNAi, an antisense oligonucleotide, an antisense polynucleotide, and any combination thereof.
  • the rAAV vectors obtained by the disclosed methods will preferably encode at least one diagnostic or therapeutic protein or polypeptide selected from the group consisting of a molecular marker, an adrenergic agonist, an anti-apoptosis factor, an apoptosis inhibitor, a cytokine receptor, a cytokine, a cytotoxin, an erythropoietic agent, a glutamic acid decarboxylase, a glycoprotein, a growth factor, a growth factor receptor, a hormone, a hormone receptor, an interferon, an interleukin, an interleukin receptor, a kinase, a kinase inhibitor, a nerve growth factor, a netrin, a neuroactive peptide, a neuroactive peptide receptor, a neurogenic factor, a neurogenic factor receptor, a neuropilin, a neurotrophic factor, a neurotrophin, a neurotrophin receptor, an N-methyl-
  • the rAAV vectors of the present invention may include one or more nucleic acid segments that encode a polypeptide selected from the group consisting of BD F, CNTF, CSF, EGF, FGF, G-SCF, GM-CSF, gonadotropin, IFN, IFG-1, M-CSF, NGF, PDGF, PEDF, TGF, TGF-B2, TNF, VEGF, prolactin, somatotropin, XIAP1, IL-1, IL-2, IL-3, IL-4, IL-5, IL-6, IL-7, IL-8, IL-9, IL-10, IL-10(I87A), viral IL-10, IL-11, IL-12, IL-13, IL-14, IL-15, IL-16, IL-17, IL-18, and any combination thereof.
  • the invention concerns genetically-modified, improved- transduction-efficiency rAAV vectors that include at least a first nucleic acid segment that encodes one or more therapeutic agents that alter, inhibit, reduce, prevent, eliminate, or impair the activity of one or more endogenous biological processes in the cell.
  • therapeutic agents may be those that selectively inhibit or reduce the effects of one or more metabolic processes, dysfunctions, disorders, or diseases.
  • the defect may be caused by injury or trauma to the mammal for which treatment is desired.
  • the defect may be caused the over-expression of an endogenous biological compound, while in other embodiments still; the defect may be caused by the under-expression or even lack of one or more endogenous biological compounds.
  • the genetically-modified rAAV vectors and expression systems of the present invention may also further optionally include a second distinct nucleic acid segment that comprises, consists essentially of, or consists of, one or more enhancers, one or more regulatory elements, one or more transcriptional elements, or any combination thereof, that alter, improve, regulate, and/or affect the transcription of the nucleotide sequence of interest expressed by the modified rAAV vectors.
  • the rAAV vectors of the present invention may further include a second nucleic acid segment that comprises, consists essentially of, or consists of, a CMV enhancer, a synthetic enhancer, a cell-specific enhancer, a tissue-specific enhancer, or any combination thereof.
  • the second nucleic acid segment may also further comprise, consist essentially of, or consist of, one or more intron sequences, one or more post-transcriptional regulatory elements, or any combination thereof.
  • the improved vectors and expression systems of the present invention may also optionally further include a polynucleotide that comprises, consists essentially of, or consists of, one or more polylinkers, restriction sites, and/or multiple cloning region(s) to facilitate insertion (cloning) of one or more selected genetic elements, genes of interest, or therapeutic or diagnostic constructs into the rAAV vector at a selected site within the vector.
  • a polynucleotide that comprises, consists essentially of, or consists of, one or more polylinkers, restriction sites, and/or multiple cloning region(s) to facilitate insertion (cloning) of one or more selected genetic elements, genes of interest, or therapeutic or diagnostic constructs into the rAAV vector at a selected site within the vector.
  • the exogenous polynucleotide(s) that may be delivered into suitable host cells by the improved, capsid-modified, rAAV vectors disclosed herein are preferably of mammalian origin, with polynucleotides encoding one or more polypeptides or peptides of human, non-human primate, porcine, bovine, ovine, feline, canine, equine, epine, caprine, or lupine origin being particularly preferred.
  • exogenous polynucleotide(s) that may be delivered into host cells by the disclosed capsid-modified viral vectors may, in certain embodiments, encode one or more proteins, one or more polypeptides, one or more peptides, one or more enzymes, or one or more antibodies (or antigen-binding fragments thereof), or alternatively, may express one or more siRNAs, ribozymes, antisense oligonucleotides, PNA molecules, or any combination thereof.
  • two or more different molecules may be produced from a single rAAV expression system, or alternatively, a selected host cell may be transfected with two or more unique rAAV expression systems, each of which may comprise one or more distinct polynucleotides that encode a therapeutic agent.
  • the invention also provides rAAV vector mutants that are comprised within an infectious adeno-associated viral particle or a virion, as well as pluralities of such virions or infectious particles.
  • Such vectors and virions may be comprised within one or more diluents, buffers, physiological solutions or pharmaceutical vehicles, or formulated for administration to a mammal in one or more diagnostic, therapeutic, and/or prophylactic regimens.
  • the vectors, virus particles, virions, and pluralities thereof of the present invention may also be provided in excipient formulations that are acceptable for veterinary administration to selected livestock, exotics, domesticated animals, and companion animals (including pets and such like), as well as to non-human primates, zoological or otherwise captive specimens, and such like.
  • the invention also concerns host cells that comprise at least one of the disclosed rAAV expression vectors, or one or more virus particles or virions that comprise such an expression vector.
  • host cells are particularly mammalian host cells, with human host cells being particularly highly preferred, and may be either isolated, in cell or tissue culture. In the case of genetically modified animal models, the transformed host cells may even be comprised within the body of a non-human animal itself.
  • the creation of recombinant non-human host cells, and/or isolated recombinant human host cells that comprise one or more of the disclosed rAAV vectors is also contemplated to be useful for a variety of diagnostic, and laboratory protocols, including, for example, means for the production of large-scale quantities of the rAAV vectors described herein.
  • virus production methods are particularly contemplated to be an improvement over existing methodologies including in particular, those that require very high titers of the viral stocks in order to be useful as a gene therapy tool.
  • the inventors contemplate that one very significant advantage of the present methods will be the ability to utilize lower titers of viral particles in mammalian transduction protocols, yet still retain transfection rates at a suitable level.
  • compositions comprising one or more of the disclosed rAAV vectors, expression systems, infectious AAV particles, or host cells also form part of the present invention, and particularly those compositions that further comprise at least a first pharmaceutically- acceptable excipient for use in therapy, and for use in the manufacture of medicaments for the treatment of one or more mammalian diseases, disorders, dysfunctions, or trauma.
  • Such pharmaceutical compositions may optionally further comprise one or more diluents, buffers, liposomes, a lipid, a lipid complex; or the tyrosine-modified rAAV vectors may be comprised within a microsphere or a nanoparticle.
  • compositions suitable for intramuscular, intravenous, or direct injection into an organ or tissue or a plurality of cells or tissues of a human or other mammal are particularly preferred, however, the compositions disclosed herein may also find utility in administration to discreet areas of the mammalian body, including for example, formulations that are suitable for direct injection into one or more organs, tissues, or cell types in the body.
  • Such injection sites include, but are not limited to, the brain, a joint or joint capsule, a synovium or subsynovium tissue, tendons, ligaments, cartilages, bone, peri-articular muscle or an articular space of a mammalian joint, as well as direct administration to an organ such as the heart, liver, lung, pancreas, intestine, brain, bladder, kidney, or other site within the patient's body, including, for example , introduction of the viral vectors via intraabdominal, intrathorascic, intravascular, or intracerebroventricular delivery.
  • aspects of the invention concern recombinant adeno-associated virus virion particles, compositions, and host cells that comprise, consist essentially of, or consist of, one or more of the rAAV vectors disclosed herein, such as for example pharmaceutical formulations of the vectors intended for administration to a mammal through suitable means, such as, by intramuscular, intravenous, intra-articular, or direct injection to one or more cells, tissues, or organs of a selected mammal.
  • compositions may be formulated with pharmaceutically-acceptable excipients as described hereinbelow, and may comprise one or more liposomes, lipids, lipid complexes, microspheres or nanoparticle formulations to facilitate administration to the selected organs, tissues, and cells for which therapy is desired.
  • Kits comprising one or more of the disclosed rAAV vectors (as well as one or more virions, viral particles, transformed host cells or pharmaceutical compositions comprising such vectors); and instructions for using such kits in one or more therapeutic, diagnostic, and/or prophylactic clinical embodiments are also provided by the present invention.
  • kits may further comprise one or more reagents, restriction enzymes, peptides, therapeutics, pharmaceutical compounds, or means for delivery of the composition(s) to host cells, or to an animal (e.g., syringes, injectables, and the like).
  • kits include those for treating, preventing, or ameliorating the symptoms of a disease, deficiency, dysfunction, and/or injury, or may include components for the large-scale production of the viral vectors themselves, such as for commercial sale, or for use by others, including e.g., virologists, medical professionals, and the like.
  • Another important aspect of the present invention concerns methods of use of the disclosed rAAV vectors, virions, expression systems, compositions, and host cells described herein in the preparation of medicaments for diagnosing, preventing, treating or ameliorating at least one or more symptoms of a disease, a dysfunction, a disorder, an abnormal condition, a deficiency, injury, or trauma in an animal, and in particular, in a vertebrate mammal.
  • Such methods generally involve administration to a mammal in need thereof, one or more of the disclosed vectors, virions, viral particles, host cells, compositions, or pluralities thereof, in an amount and for a time sufficient to diagnose, prevent, treat, or lessen one or more symptoms of such a disease, dysfunction, disorder, abnormal condition, deficiency, injury, or trauma in the affected animal.
  • the methods may also encompass prophylactic treatment of animals suspected of having such conditions, or administration of such compositions to those animals at risk for developing such conditions either following diagnosis, or prior to the onset of symptoms.
  • the exogenous polynucleotide will preferably encode one or more proteins, polypeptides, peptides, nbozymes, or antisense oligonucleotides, or a combination of these.
  • the exogenous polynucleotide may encode two or more such molecules, or a plurality of such molecules as may be desired.
  • two or more different molecules may be produced from a single rAAV expression system, or alternatively, a selected host cell may be transfected with two or more unique rAAV expression systems, each of which will provide unique heterologous polynucleotides encoding at least two different such molecules.
  • compositions comprising one or more of the disclosed rAAV vectors, expression systems, infectious AAV particles, host cells also form part of the present invention, and particularly those compositions that further comprise at least a first pharmaceutically- acceptable excipient for use in the manufacture of medicaments and methods involving therapeutic administration of such rAAV vectors.
  • Such pharmaceutical compositions may optionally further comprise liposomes, a lipid, a lipid complex; or the rAAV vectors may be comprised within a microsphere or a nanoparticle.
  • Pharmaceutical formulations suitable for intramuscular, intravenous, or direct injection into an organ or tissue of a human are particularly preferred.
  • compositions including one or more of the disclosed rAAV vectors comprised within a kit for diagnosing, preventing, treating or ameliorating one or more symptoms of a mammalian disease, injury, disorder, trauma or dysfunction.
  • kits may be useful in the diagnosis, prophylaxis, and/or therapy or a human disease, and may be particularly useful in the treatment, prevention, and/or amelioration of one or more symptoms of wet age-related macular degeneration, dry age-related macular degeneration, glaucoma, retinitis pigmentosa, diabetic retinopathy, orphan ophthalmological diseases, cancer, diabetes, autoimmune disease, kidney disease, cardiovascular disease, pancreatic disease, intestinal disease, liver disease, neurological disease, neuromuscular disorder, neuromotor deficit, neuroskeletal impairment, neurological disability, neurosensory dysfunction, stroke, ischemia, a 1-antitiypsin (AAT) deficiency, Batten's disease, Alzheimer's disease, sickle cell disease, f3- thalassamia, Huntington's disease, Parkinson's disease, skeletal disease, trauma, pulmonary disease in a human.
  • AAT 1-antitiypsin
  • the invention also provides for the use of a composition disclosed herein in the manufacture of a medicament for treating, preventing or ameliorating the symptoms of a disease, disorder, dysfunction, injury or trauma, including, but not limited to, the treatment, prevention, and/or prophylaxis of a disease, disorder or dysfunction, and/or the amelioration of one or more symptoms of such a disease, disorder or dysfunction.
  • the invention also provides a method for treating or ameliorating the symptoms of such a disease, injury, disorder, or dysfunction in a mammal.
  • Such methods generally involve at least the step of administering to a mammal in need thereof, one or more of the rAAV vectors as disclosed herein, in an amount and for a time sufficient to treat or ameliorate the symptoms of such a disease, injury, disorder, or dysfunction in the mammal.
  • Such treatment regimens are particularly contemplated in human therapy, via administration of one or more compositions either intramuscularly, intravenously, subcutaneously, intrathecally, intraperitoneally, or by direct injection into an organ or a tissue of the mammal under care.
  • the invention also provides a method for providing to a mammal in need thereof, a therapeutically-effective amount of an rAAV composition of the present invention, in an amount, and for a time effective to provide the patient with a therapeutically-effective amount of the desired therapeutic agent(s) encoded by one or more nucleic acid segments comprised within the rAAV vector.
  • exemplary therapeutic agents include, but are not limited to, a polypeptide, a peptide, an antibody, an antigen-binding fragment, a ribozyme, a peptide nucleic acid, an siRNA, an RNAi, an antisense oligonucleotide, an antisense polynucleotide, or a combination thereof.
  • the genetic constructs of the present invention may be prepared in a variety of compositions, and may also be formulated in appropriate pharmaceutical vehicles for administration to human or animal subjects.
  • the invention also provides compositions comprising one or more of the disclosed rAAV vectors, expression systems, virions, viral particles, mammalian cells, or combinations thereof.
  • the present invention provides pharmaceutical formulations of one or more rAAV vectors disclosed herein for administration to a cell or an animal, either alone or in combination with one or more other modalities of therapy, and in particular, for therapy of human cells, tissues, and diseases affecting man.
  • Formulation of pharmaceutically- acceptable excipients and carrier solutions is well-known to those of skill in the art, as is the development of suitable dosing and treatment regimens for using the particular compositions described herein in a variety of treatment regimens, including e.g., oral, parenteral, intravenous, intranasal, intra-articular, intramuscular administration and formulation.
  • VRs I through IX variant regions I-IX; also known as "loops"
  • VRs are localized at the surface of the assembled capsid and are assumed to be responsible for the capsid interaction with cell surface receptors and other host factors. Because of their location, VRs are also predicted to be less critical for capsid assembly. Therefore, the guiding principle of the library's design was to modify only surface VRs while keeping the backbone sequence unchanged to maintain the integrity of the assembling scaffold.
  • the library was built in three steps: first, VR parent sub-libraries were prepared each containing mutations in only one VR (B: VR-IV, C: VR-VII, D: VR-VIII) or a subset of VRs (A: VR-I + VR-V + VR-VI), then, structurally compatible sequences were combined to generate master libraries (A + B + C: VRs I, IV, V, VI, VII) and (D: VR-VIII), and finally the master libraries were packaged. See Examples, Example 2 and Figure 5. Methods for generating and assembling DNA fragments for the library are disclosed in WO2015/048534 and US 7220577, both of which are incorporated herein in their entirety.
  • the master libraries can be used to select virions having capsids containing degenerate or otherwise modified Cap protein (i.e., Cap protein that differs from wildtype serotype 3 at least at one amino acid position) that are targeted to particular tissue or cell types.
  • virions made according to the invention include those that exhibit a new tropism, e.g., those capable of infecting cells normally non-permissive to AAV infection in general or at least non-permissive to AAV3 infection, as well as those that exhibit an increased or decreased ability to infect a particular cell or tissue type.
  • virions made according to the invention include those that lack the ability to infect cells normally permissive to AAV infection in general or at least normally permissive to AAV3 infection.
  • a packaged master library is introduced into a target cell.
  • the target cell is also infected with a helper virus (e.g. Ad).
  • Ad helper virus
  • the target cell is cultured under conditions that allow for the production of virions, resulting in a population of virions that are harvested from the target cell. This population of virions has been selected for having a tropism for that target cell.
  • cells in different flasks or dishes can be simultaneously infected with WT AAV3 or rAAV using the same conditions as used for the library. After a suitable time post-infection, cells can be harvested, washed and the virions purified using a suitable purification method (See Gao et al., Hum. Gene Ther. 9:2353-62, 1998; U.S. Pat. No. 6, 146,874; and Zolotukhin et al., Gene Ther. 6:973-85, 1999).
  • AAV and helper virions (e.g., Ad) from each infection can be tittered, by real-time PCT for example, and the AAV virions can then be further propagated, resulting in a stock of selected virions.
  • nucleic acid from the virions is isolated and the sequence of the nucleotide sequence encoding the at least one AAV Cap protein is determined.
  • Virions made and selected according to the invention that can specifically target diseased cells or tissues over non-diseased cells or tissues are particularly useful.
  • tissue- or cell-specific virions can be selected using an in vivo approach.
  • mice or other suitable host
  • a suitable amount viral preparation e.g., 1 x 10 10 to 1 x 10 11 vg in the case of mice
  • More than one round of selection can be performed by injecting original master library for the first round and target-enriched libraries in subsequent rounds.
  • Hosts are euthanized after an incubation period (3 to 4 days for mice), and episomal DNA is purified from the target cells or tissue and used as a template to amplify capsid DNA sequences.
  • Target-enriched libraries can then be generated, purified and quantified.
  • amplified capsid DNA can be inserted into an appropriate vector for cloning and random clones can be analyzed by sequencing.
  • carrier is intended to include any solvent(s), dispersion medium, coating(s), diluent(s), buffer(s), isotonic agent(s), solution(s), suspension(s), colloid(s), inert(s) or such like, or a combination thereof, that is pharmaceutically acceptable for administration to the relevant animal.
  • one or more delivery vehicles for chemical compounds in general, and chemotherapeutics in particular is well known to those of ordinary skill in the pharmaceutical arts. Except insofar as any conventional media or agent is incompatible with the active ingredient, its use in the diagnostic, prophylactic, and therapeutic compositions is contemplated.
  • One or more supplementary active ingredient(s) may also be incorporated into, or administered in association with, one or more of the disclosed chemotherapeutic compositions.
  • chimeric rcAAV refers to a replication-competent AAV- derived nucleic acid containing at least one nucleotide sequence that 1) encodes an AAV protein and 2) differs from the corresponding native nucleotide sequence in one or more bases.
  • DNA segment refers to a DNA molecule that has been isolated free of total genomic DNA of a particular species. Therefore, a DNA segment obtained from a biological sample using one of the compositions disclosed herein refers to one or more DNA segments that have been isolated away from, or purified free from, total genomic DNA of the particular species from which they are obtained. Included within the term “DNA segment,” are DNA segments and smaller fragments of such segments, as well as recombinant vectors, including, for example, plasmids, cosmids, phage, viruses, and the like.
  • an effective amount would be understood by those of ordinary skill in the art to provide a therapeutic, prophylactic, or otherwise beneficial effect against the organism, its infection, or the symptoms of the organism or its infection, or any combination thereof
  • expression control sequence refers to any genetic element (e.g., polynucleotide sequence) that can exert a regulatory effect on the replication or expression (transcription or translation) of another genetic element.
  • Common expression control sequences include promoters, polyadenylation (polyA) signals, transcription termination sequences, upstream regulatory domains, origins of replication, internal ribosome entry sites (IRES), enhancers, and the like.
  • a "tissue specific expression control sequence” is one that exerts a regulatory effect on the replication or expression (transcription or translation) of another genetic element in only one type of tissue or a small subset of tissues.
  • helper function is meant as a functional activity performed by a nucleic acid or polypeptide that is derived from a virus such as Adenovirus (Ad) or herpesvirus and that facilitates AAV replication in a host cell.
  • Ad Adenovirus
  • herpesvirus a virus that facilitates AAV replication in a host cell.
  • heterologous is defined in relation to a predetermined referenced gene sequence.
  • a heterologous promoter is defined as a promoter which does not naturally occur adjacent to the referenced structural gene, but which is positioned by laboratory manipulation.
  • a heterologous gene or nucleic acid segment is defined as a gene or segment that does not naturally occur adjacent to the referenced promoter and/or enhancer elements.
  • the term “homology” refers to a degree of complementarity between two or more polynucleotide or polypeptide sequences.
  • the word “identity” may substitute for the word “homology” when a first nucleic acid or amino acid sequence has the exact same primary sequence as a second nucleic acid or amino acid sequence.
  • Sequence homology and sequence identity can be determined by analyzing two or more sequences using algorithms and computer programs known in the art. Such methods may be used to assess whether a given sequence is identical or homologous to another selected sequence.
  • homologous means, when referring to polynucleotides, sequences that have the same essential nucleotide sequence, despite arising from different origins. Typically, homologous nucleic acid sequences are derived from closely related genes or organisms possessing one or more substantially similar genomic sequences. By contrast, an "analogous" polynucleotide is one that shares the same function with a polynucleotide from a different species or organism, but may have a significantly different primary nucleotide sequence that encodes one or more proteins or polypeptides that accomplish similar functions or possess similar biological activity. Analogous polynucleotides may often be derived from two or more organisms that are not closely related (e.g., either genetically or phylogenetically).
  • nucleic acid or polypeptide sequences refer to two or more sequences or subsequences that are the same or have a specified percentage of amino acid residues or nucleotides that are the same, when compared and aligned for maximum correspondence, as measured using one of the sequence comparison algorithms described below (or other algorithms available to persons of ordinary skill) or by visual inspection.
  • the phrase "in need of treatment” refers to a judgment made by a caregiver such as a physician or veterinarian that a patient requires (or will benefit in one or more ways) from treatment. Such judgment may made based on a variety of factors that are in the realm of a caregiver's expertise, and may include the knowledge that the patient is ill as the result of a disease state that is treatable by one or more compound or pharmaceutical compositions such as those set forth herein.
  • isolated or “biologically pure” refer to material that is substantially, or essentially, free from components that normally accompany the material as it is found in its native state.
  • isolated polynucleotides in accordance with the invention preferably do not contain materials normally associated with those polynucleotides in their natural, or in situ, environment.
  • kit may be used to describe variations of the portable, self- contained enclosure that includes at least one set of components to conduct one or more of the diagnostic or therapeutic methods of the invention.
  • Link refers to any method known in the art for functionally connecting one or more proteins, peptides, nucleic acids, or polynucleotides, including, without limitation, recombinant fusion, covalent bonding, disulfide bonding, ionic bonding, hydrogen bonding, electrostatic bonding, and the like.
  • a vector library refers to a collection of elements that differ from one another in at least one aspect.
  • a vector library is a collection of at least two vectors that differ from one another by at least one nucleotide.
  • a "virion library” is a collection of at least two virions that differ from one another by at least one nucleotide or at least one capsid protein.
  • the term "master library” or “combined library” refers to a pool of rAAV virions composed of chimeric rcAAV vectors encapsidated in cognate chimeric capsids (e.g., capsids containing a degenerate or otherwise modified Cap protein).
  • the term “parent sub-library” refers to a pool of rAAV virions composed of chimeric rcAAV vectors encapsidated in cognate chimeric capsids (e.g., capsids containing degenerate or otherwise modified Cap protein). More than one parent sub-library can be combined to create a master library or combined library.
  • nucleic acid molecule or polypeptide When referring to a nucleic acid molecule or polypeptide, the term “native” refers to a naturally-occurring (e.g., a WT) nucleic acid or polypeptide.
  • naturally occurring refers to the fact that an object can be found in nature.
  • a polypeptide or polynucleotide sequence that is present in an organism (including viruses) that can be isolated from a source in nature and which has not been intentionally modified by the hand of man in a laboratory is naturally- occurring.
  • laboratory strains of rodents that may have been selectively bred according to classical genetics are considered naturally occurring animals.
  • nucleic acid means a chain of two or more nucleotides such as RNA (ribonucleic acid) and DNA (deoxyribonucleic acid).
  • RNA ribonucleic acid
  • DNA deoxyribonucleic acid
  • G or C (S) Amino, i.e. A or C (M), Keto, i.e. G or T (K), not A, i.e. G or C or T (B), not G, i.e. A or C or T (H), not C, i.e. A or G or T (D) and not T, i. e. A or G or C (V).
  • cap nucleic acid means a nucleic acid that encodes a Cap protein.
  • examples of cap nucleic acids include "wild-type” (WT) Cap-encoding nucleic acid sequences from AAV serotypes 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13; a native form cap cDNA; a nucleic acid having sequences from which a cap cDNA can be transcribed; and/or allelic variants and homologs of the foregoing.
  • WT wild-type Cap-encoding nucleic acid sequences from AAV serotypes 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13
  • a native form cap cDNA a nucleic acid having sequences from which a cap cDNA can be transcribed
  • allelic variants and homologs of the foregoing are allelic variants and homologs of the foregoing.
  • VR amino acid stretches of capsid protein that do not have a high degree of homology between AAV variants. These amino acid stretches are commonly designated as VRs I through IX (also known as “loops"). VRs are localized at the surface of the assembled capsid and interact with host cell surface receptors and other host factors.
  • the term “patient” also interchangeably referred to as “host” or “subject” refers to any host that can receive one or more of the pharmaceutical compositions disclosed herein.
  • the subject is a vertebrate animal, which is intended to denote any animal species (and preferably, a mammalian species such as a human being).
  • a "patient” refers to any animal host including without limitation any mammalian host.
  • the term refers to any mammalian host, the latter including but not limited to, human and non-human primates, bovines, canines, caprines, cavines, corvines, epines, equines, felines, hircines, lapines, leporines, lupines, murines, ovines, porcines, ranines, racines, vulpines, and the like, including livestock, zoological specimens, exotics, as well as companion animals, pets, and any animal under the care of a veterinary practitioner.
  • pharmaceutically-acceptable refers to molecular entities and compositions that preferably do not produce an allergic or similar untoward reaction when administered to a mammal, and in particular, when administered to a human.
  • pharmaceutically acceptable salt refers to a salt that preferably retains the desired biological activity of the parent compound and does not impart any undesired toxicological effects.
  • salts include, without limitation, acid addition salts formed with inorganic acids (e.g., hydrochloric acid, hydrobromic acid, sulfuric acid, phosphoric acid, nitric acid, and the like); and salts formed with organic acids including, without limitation, acetic acid, oxalic acid, tartaric acid, succinic acid, maleic acid, fumaric acid, gluconic acid, citric acid, malic acid, ascorbic acid, benzoic acid, tannic acid, pamoic (embonic) acid, alginic acid, naphthoic acid, polyglutamic acid, naphthalenesulfonic acids, naphthalenedisulfonic acids, polygalacturonic acid; salts with polyvalent metal cations such as zinc, calcium, bismuth, barium, magnesium, aluminum, copper, cobalt, nickel, cadmium, and the like; salts formed with an organic cation formed from ⁇ , ⁇ '-dibenzylethylened
  • acids include, but are not limited to, hydrochloric, hydrobromic, sulfuric, nitric, perchloric, fumaric, maleic, phosphoric, glycollic, lactic, salicyclic, succinic, toluene-p- sulfonic, tartaric, acetic, citric, methanesulfonic, formic, benzoic, malonic, naphthalene-2- sulfonic, trifluoroacetic and benzenesulfonic acids.
  • Salts derived from appropriate bases include, but are not limited to, alkalis such as sodium and ammonia.
  • Plasmids and vectors of the present invention may include one or more genetic elements as described herein arranged such that an inserted coding sequence can be transcribed and translated in a suitable expression cells.
  • the plasmid or vector may include one or more nucleic acid segments, genes, promoters, enhancers, activators, multiple cloning regions, or any combination thereof, including segments that are obtained from or derived from one or more natural and/or artificial sources.
  • polypeptide is intended to encompass a singular “polypeptide” as well as plural “polypeptides,” and includes any chain or chains of two or more amino acids.
  • terms including, but not limited to “peptide,” “dipeptide,” “tripeptide,” “protein,” “enzyme,” “amino acid chain,” and “contiguous amino acid sequence” are all encompassed within the definition of a “polypeptide,” and the term “polypeptide” can be used instead of, or interchangeably with, any of these terms.
  • polypeptides that have undergone one or more post-translational modification(s), including for example, but not limited to, glycosylation, acetylation, phosphorylation, amidation, derivatization, proteolytic cleavage, post-translation processing, or modification by inclusion of one or more non-naturally occurring amino acids.
  • post-translational modification(s) including for example, but not limited to, glycosylation, acetylation, phosphorylation, amidation, derivatization, proteolytic cleavage, post-translation processing, or modification by inclusion of one or more non-naturally occurring amino acids.
  • Conventional nomenclature exists in the art for polynucleotide and polypeptide structures.
  • one-letter and three-letter abbreviations are widely employed to describe amino acids: Alanine (A; Ala), Arginine (R; Arg), Asparagine (N; Asn), Aspartic Acid (D; Asp), Cysteine (C; Cys), Glutamine (Q; Gin), Glutamic Acid (E; Glu), Glycine (G; Gly), Histidine (H; His), Isoleucine (I; He), Leucine (L; Leu), Methionine (M; Met), Phenylalanine (F; Phe), Proline (P; Pro), Serine (S; Ser), Threonine (T; Thr), Tryptophan (W; Trp), Tyrosine (Y; Tyr), Valine (V; Val), and Lysine (K; Lys).
  • the terms "prevent,” “preventing,” “prevention,” “suppress,” “suppressing,” and “suppression” as used herein refer to administering a compound either alone or as contained in a pharmaceutical composition prior to the onset of clinical symptoms of a disease state so as to prevent any symptom, aspect or characteristic of the disease state. Such preventing and suppressing need not be absolute to be deemed medically useful.
  • promoter refers to a region or regions of a nucleic acid sequence that regulates transcription.
  • Protein is used herein interchangeably with “peptide” and “polypeptide,” and includes both peptides and polypeptides produced synthetically, recombinantly, or in vitro and peptides and polypeptides expressed in vivo after nucleic acid sequences are administered into a host animal or human subject.
  • polypeptide is preferably intended to refer to any amino acid chain length, including those of short peptides from two to about 20 amino acid residues in length, oligopeptides from about 10 to about 100 amino acid residues in length, and longer polypeptides including those of about 100 or more amino acid residues in length.
  • polypeptides and proteins of the present invention also include polypeptides and proteins that are or have been post-translationally modified, and include any sugar or other derivative(s) or conjugate(s) added to the backbone amino acid chain.
  • proliferative typed is meant a nucleic acid or genome derived from a first AAV serotype that is encapsidated (packaged) into an AAV capsid containing at least one AAV Cap protein of a second serotype differing from the first serotype.
  • rcAAV vector refers to a replication-competent AAV- derived nucleic acid capable of DNA replication in a cell without any additional AAV genes or gene products.
  • recombinant indicates that the material (e.g., a polynucleotide or a polypeptide) has been artificially or synthetically (non-naturally) altered by human intervention. The alteration can be performed on the material within or removed from, its natural environment or state. Specifically, e.g., a promoter sequence is "recombinant” when it is produced by the expression of a nucleic acid segment engineered by the hand of man.
  • a "recombinant nucleic acid” is one that is made by recombining nucleic acids, e.g., during cloning, DNA shuffling or other procedures, or by chemical or other mutagenesis
  • a "recombinant polypeptide” or “recombinant protein” is a polypeptide or protein which is produced by expression of a recombinant nucleic acid
  • a "recombinant virus,” e.g., a recombinant AAV virus is produced by the expression of a recombinant nucleic acid.
  • regulatory element refers to a region or regions of a nucleic acid sequence that regulates transcription.
  • exemplary regulatory elements include, but are not limited to, enhancers, post-transcriptional elements, transcriptional control sequences, and such like.
  • RNA segment refers to an RNA molecule that has been isolated free of total cellular RNA of a particular species. Therefore, RNA segments can refer to one or more RNA segments (either of native or synthetic origin) that have been isolated away from, or purified free from, other RNAs. Included within the term “RNA segment,” are RNA segments and smaller fragments of such segments.
  • substantially corresponds to denote a characteristic of a nucleic acid or an amino acid sequence, wherein a selected nucleic acid or amino acid sequence has at least about 70 or about 75 percent sequence identity as compared to a selected reference nucleic acid or amino acid sequence. More typically, the selected sequence and the reference sequence will have at least about 76, 77, 78, 79, 80, 81, 82, 83, 84 or even 85 percent sequence identity, and more preferably, at least about 86, 87, 88, 89, 90, 91, 92, 93, 94, or 95 percent sequence identity. More preferably still, highly homologous sequences often share greater than at least about 96, 97, 98, or 99 percent sequence identity between the selected sequence and the reference sequence to which it was compared.
  • the percentage of sequence identity may be calculated over the entire length of the sequences to be compared, or may be calculated by excluding small deletions or additions which total less than about 25 percent or so of the chosen reference sequence.
  • the reference sequence may be a subset of a larger sequence, such as a portion of a gene or flanking sequence, or a repetitive portion of a chromosome.
  • the reference sequence will typically comprise at least about 18-25 nucleotides, more typically at least about 26 to 35 nucleotides, and even more typically at least about 40, 50, 60, 70, 80, 90, or even 100 or so nucleotides.
  • the extent of percent identity between the two sequences will be at least about 80%, preferably at least about 85%, and more preferably about 90% or 95% or higher, as readily determined by one or more of the sequence comparison algorithms well-known to those of skill in the art, such as e.g., the FASTA program analysis described by Pearson and Lipman (1988).
  • structural gene is intended to generally describe a polynucleotide, such as a gene, that is expressed to produce an encoded peptide, polypeptide, protein, ribozyme, catalytic RNA molecule, or antisense molecule.
  • subject describes an organism, including a mammal such as a human primate, to which treatment with one or more of the disclosed compositions may be provided.
  • Mammalian species that may benefit from the disclosed treatment methods include, without limitation, humans, non-human primates such as apes; chimpanzees; monkeys, and orangutans, domesticated animals, including dogs and cats, as well as livestock such as horses, cattle, pigs, sheep, and goats, or other mammalian species including, without limitation, mice, rats, guinea pigs, rabbits, hamsters, and the like.
  • Transcriptional regulatory element refers to a polynucleotide sequence that activates transcription alone or in combination with one or more other nucleic acid sequences.
  • a transcriptional regulatory element may include, for example, one or more promoters, one or more response elements, one or more negative regulatory elements, one or more enhancers, or any combination thereof
  • transcription factor recognition site and a “transcription factor binding site” refer to a polynucleotide sequence(s) or sequence motif(s) that are identified as being sites for the sequence-specific interaction of one or more transcription factors, frequently taking the form of direct protein-DNA binding.
  • transcription factor binding sites can be identified by DNA footprinting, gel mobility shift assays, and the like, and/or can be predicted based on known consensus sequence motifs, or by other methods known to one of ordinary skill in the relevant molecular biological and virology arts.
  • Transcriptional unit refers to a polynucleotide sequence that comprises at least a first structural gene operably linked to at least a first cis-acting promoter sequence and optionally linked operably to one or more other cis-acting nucleic acid sequences necessary for efficient transcription of the structural gene sequences, and at least a first distal regulatory element as may be required for the appropriate tissue-specific and developmental transcription of the structural gene sequence operably positioned under the control of the promoter and/or enhancer elements, as well as any additional cis sequences that are necessary for efficient transcription and translation (e.g., polyadenylation site(s), mRNA stability controlling sequence(s), etc.
  • transformed cell is intended to mean a host cell whose nucleic acid complement has been altered by the introduction of one or more exogenous polynucleotides into that cell.
  • transformation is intended to generally describe a process of introducing an exogenous polynucleotide sequence (e.g., a viral vector, a plasmid, or a recombinant DNA or RNA molecule) into a host cell or protoplast in which the exogenous polynucleotide is incorporated into at least a first chromosome or is capable of autonomous replication within the transformed host cell.
  • an exogenous polynucleotide sequence e.g., a viral vector, a plasmid, or a recombinant DNA or RNA molecule
  • Transfection, electroporation, and "naked" nucleic acid uptake all represent examples of techniques used to transform a host cell with one or more polynucleotides.
  • the terms “treat,” “treating,” and “treatment” refer to the administration of one or more compounds (either alone or as contained in one or more pharmaceutical compositions) after the onset of clinical symptoms of a disease state so as to reduce, or eliminate any symptom, aspect or characteristic of the disease state. Such treating need not be absolute to be deemed medically useful.
  • the terms “treatment,” “treat,” “treated,” or “treating” may refer to therapy, or to the amelioration or the reduction, in the extent or severity of disease, of one or more symptom thereof, whether before or after its development afflicts a patient.
  • the term "vector” refers to a nucleic acid molecule capable of transporting another nucleic acid to which it has been linked, e.g., a plasmid.
  • a vector is an episome, i.e., a nucleic acid capable of extra-chromosomal replication.
  • Preferred vectors are those capable of autonomous replication and/or expression of nucleic acids to which they are linked.
  • An "rAAV vector” is a recombinant AAV-derived nucleic acid containing at least one terminal repeat (TR) sequence.
  • the use of "virion” is meant to describe a virus particle that contains a nucleic acid and a protein coat (capsid).
  • An "rAAV virion” is a virion that includes nucleic acid sequences and/or proteins derived from a rAAV vector.
  • the term "tropism" refers to the cells and/or tissues of a host which support growth of a particular serotype of AAV. Some serotypes may have a broad tissue tropism and can infect many types of cells and tissues. Other serotypes may infect primarily a single tissue or cell type.
  • sequence essentially as set forth in SEQ ID NO:X means that the sequence substantially corresponds to a portion of SEQ ID NO:X and has relatively few nucleotides (or amino acids in the case of polypeptide sequences) that are not identical to, or a biologically functional equivalent of, the nucleotides (or amino acids) of SEQ ID NO:X.
  • biologically functional equivalent is well understood in the art, and is further defined in detail herein.
  • sequences that have about 85% to about 90%; or more preferably, about 91%) to about 95%; or even more preferably, about 96% to about 99%; of nucleotides that are identical or functionally equivalent to one or more of the nucleotide sequences provided herein are particularly contemplated to be useful in the practice of the invention.
  • Suitable standard hybridization conditions for the present invention include, for example, hybridization in 50% formamide, 5x Denhardt's solution, 5x SSC, 25 mM sodium phosphate, 0.1% SDS and 100 ⁇ g/ml of denatured salmon sperm DNA at 42°C for 16 h followed by 1 hr sequential washes with O. lx SSC, 0.1% SDS solution at 60°C to remove the desired amount of background signal.
  • Lower stringency hybridization conditions for the present invention include, for example, hybridization in 35% formamide, 5x Denhardt's solution, 5x SSC, 25 mM sodium phosphate, 0.1% SDS and 100 ⁇ g/ml denatured salmon sperm DNA or E. coli DNA at 42°C for 16 h followed by sequential washes with 0.8x SSC, 0.1% SDS at 55°C.
  • Those of skill in the art will recognize that conditions can be readily adjusted to obtain the desired level of stringency.
  • nucleic acid segments that are complementary, essentially complementary, and/or substantially complementary to at least one or more of the specific nucleotide sequences specifically set forth herein.
  • Nucleic acid sequences that are “complementary” are those that are capable of base-pairing according to the standard Watson-Crick complementarity rules.
  • complementary sequences means nucleic acid sequences that are substantially complementary, as may be assessed by the same nucleotide comparison set forth above, or as defined as being capable of hybridizing to one or more of the specific nucleic acid segments disclosed herein under relatively stringent conditions such as those described immediately above.
  • the probes and primers of the present invention may be of any length.
  • an algorithm defining all probes or primers contained within a given sequence can be proposed:
  • n to n + y where n is an integer from 1 to the last number of the sequence and y is the length of the probe or primer minus one, where n + y does not exceed the last number of the sequence.
  • the collection of probes or primers correspond to bases 1 to 25, bases 2 to 26, bases 3 to 27, bases 4 to 28, and so on over the entire length of the sequence.
  • exemplary primer or probe sequence include, without limitation, sequences corresponding to bases 1 to 35, bases 2 to 36, bases 3 to 37, bases 4 to 38, and so on over the entire length of the sequence.
  • probes or primers may correspond to the nucleotides from the first basepair to bp 40, from the second bp of the sequence to bp 41, from the third bp to bp 42, and so forth
  • probes or primers may correspond to a nucleotide sequence extending from bp 1 to bp 50, from bp 2 to bp 51, from bp 3 to bp 52, from bp 4 to bp 53, and so forth.
  • nucleic acid segments of the present invention in combination with an appropriate detectable marker (i.e., a "label,”), such as in the case of employing labeled polynucleotide probes in determining the presence of a given target sequence in a hybridization assay.
  • an appropriate detectable marker i.e., a "label”
  • a wide variety of appropriate indicator compounds and compositions are known in the art for labeling oligonucleotide probes, including, without limitation, fluorescent, radioactive, enzymatic or other ligands, such as avidin/biotin, etc., which are capable of being detected in a suitable assay.
  • an enzyme tag such as urease, alkaline phosphatase or peroxidase
  • colorimetric, chromogenic, or fluorigenic indicator substrates are known that can be employed to provide a method for detecting the sample that is visible to the human eye, or by analytical methods such as scintigraphy, fluorimetry, spectrophotometry, and the like, to identify specific hybridization with samples containing one or more complementary or substantially complementary nucleic acid sequences.
  • multiplexing assays where two or more labeled probes are detected either simultaneously or sequentially, it may be desirable to label a first oligonucleotide probe with a first label having a first detection property or parameter (for example, an emission and/or excitation spectral maximum), which also labeled a second oligonucleotide probe with a second label having a second detection property or parameter that is different (i.e., discreet or discernable from the first label.
  • first detection property or parameter for example, an emission and/or excitation spectral maximum
  • polynucleotides, nucleic acid segments, nucleic acid sequences, and the like include, but are not limited to, DNAs (including and not limited to genomic or extragenomic DNAs), genes, peptide nucleic acids (PNAs) RNAs (including, but not limited to, rRNAs, mRNAs and tRNAs), nucleosides, and suitable nucleic acid segments either obtained from natural sources, chemically synthesized, modified, or otherwise prepared or synthesized in whole or in part by the hand of man.
  • DNAs including and not limited to genomic or extragenomic DNAs
  • genes include peptide nucleic acids (PNAs) RNAs (including, but not limited to, rRNAs, mRNAs and tRNAs), nucleosides, and suitable nucleic acid segments either obtained from natural sources, chemically synthesized, modified, or otherwise prepared or synthesized in whole or in part by the hand of man.
  • PNAs peptide nucleic acids
  • Step 1 Sub-libraries assembly.
  • the respective PCR fragments were eluted from the agarose gel, mixed at equimolar ratios as indicated above for sub-libraries A, B, C, and D, and subjected to 15 cycles of overlap extension (OE) without primers, followed by 20 cycles of PCR using A3CL-F forward and A3CL-R reverse primers.
  • OE overlap extension
  • the resulting fragments of 1140 bp for each of the A (I+V+VI), B (IV), C (VII), or D (VIII) sub-libraries were purified on agarose gel and eluted in small volume H20.
  • Step 2 Pre-selecting structurally compatible parent viral libraries.
  • the respective PCR fragments were gel-purified and used as the templates in the OE/PCR to derive two PCR fragments, each of 1140 bp: A+B+C (VR-I, IV, V, VI, VII) and D (VR-VIII).
  • Step 3 Packaging master libraries.
  • Table 1 Theoretical (calculated) complexities of A3CL for individual VRs and combinations of VRs. The VRs and VRs combinations constructed as sub-libraries are shown in bold font.
  • A3CL-A2 F AACAACAACAGTRASTTTCCTTGGMCAGCGGCCAGCAMATATCATCTCAATG
  • A3CL-A3R GATTGCCGTGCATAGGGAAAAATYTSYCSKYATCGTCCYYGTGACTGGCCATAGCTGG
  • A3CL-R CATCCGTGTGAGGAATCTTTGC
  • Lucigen eompetent ceils were prepared from 4 L LB, resuspended in 8.5 mi H a O final volume.
  • the cell density (10 ⁇ in 3 mi H 2 0 ⁇ was A - 0.79.
  • Combined DNA (147.5 ⁇ ) was mixed with the whole volume of competent ceils and aliquoted (385 pj/a!iquot, ⁇ 10 ng plasmid DNA 50 ⁇ competent ceils) into eiectroporation cuvettes ⁇ total of -20, with outside tali electrodes) and zapped at 2.9 KV.
  • Competent ceils were prepared from 4 L L8 (grown to A 6 3 ⁇ 4 ⁇ 0,6 ⁇ and resuspended in a final volume 8 ml H a O.
  • Loop I primers A3CL-F + VR-IJV-R, template A, size 644 bp
  • the assay was split into 2x25 ⁇ assays supplemented with A3CL-F, and A3CL-R primers, DNTPs, and fresh Q5, total volume 50 ⁇ each.
  • ABC fragment was eluted in 50 ⁇ , concentration 60 ng/ ⁇

Abstract

Compositions and methods for producing modified AAV Cap genes and combinatorial libraries of chimeric AAV vectors and virions in an AAV serotype 3 background. Selecting for modified AAV3 virions displaying cell- or tissue-specific tropisms differing from WT AAV3. Using the synthetic combinatorial AAV3 capsid libraries for introducing into a selected target host cells one or more nucleic acid molecules useful in diagnostic and/or therapeutic gene-therapy regimens.

Description

SYNTHETIC COMBINATORIAL AAV3 CAPSID LIBRARY
BACKGROUND OF THE INVENTION
Adeno-associated virus (AAV) is a single-stranded DNA virus belonging to the Parvoviridae family (Muzyczka and Berns, 2001). AAV-derived vectors are promising tools for human gene therapy applications because of their absence of pathogenicity, low immunogenicity, episomal localization and stable transgene expression. However, significant limitations to the clinical use of AAV are its promiscuity and its susceptibility to neutralization by human antibodies (Jeune et al, 2013). Both of these limitations are determined by nature of the amino acid residues exposed at the surface of the capsid. Therefore, major efforts aiming at developing useful and effective gene therapy vectors have been devoted to obtaining and studying capsid variants (Wu et al, 2006). The first approach was to study naturally occurring AAV isolates. So far, 13 serotypes have been formally characterized and hundreds of variant isolates have been sequenced. Additional capsid variation has been investigated through the generation of mosaics (viral particles made of capsid proteins from more than one serotype) (Hauck et al, 2003; Stachler and Bartlett, 2006; Gigout et al, 2005), chimeras (capsid proteins with domains from various origins) (Shen et al, 2007), and various substitutional or insertional mutants (Wu et al, 2000). However, the most significant advances are expected to result from directed evolution approaches through the development of capsid libraries.
The various strategies to generate capsid libraries that have been developed so far all suffer from sequence bias or limited diversity. Random display peptide libraries (Govindasamy et al, 2006) are limited to an insertion at one particular capsid location. Libraries generated using error-prone PCR contain a very small fraction of gene variants encoding proteins that can fold properly and assemble into a functional capsid, due to the randomness of the mutations. DNA shuffling and staggered extension processes are more efficient because they recombine naturally-occurring parental sequences and therefore are more likely to generate actual capsid variants. However, they can only recombine blocks of DNA as opposed to single nucleotide positions, which results in sequence bias (parental polymorphisms will tend to cluster together instead of being randomly distributed). SUMMARY OF THE INVENTION
An embodiment of a non-naturally occurring nucleic acid of these teachings includes (a) a first nucleotide sequence encoding at least one AAV Rep protein from serotype 3; (b) a second nucleotide sequence encoding at least one AAV Cap protein differing from wildtype serotype 3 at least at one nucleotide position; and (c) a first AAV terminal repeat from serotype 3 and a second AAV terminal repeat from serotype 3, where the first and second nucleotide sequences are interposed between the first and second AAV terminal repeat.
An aspect of an embodiment of the non-naturally occurring nucleic acid of these teachings further includes a third nucleotide sequence encoding at least one molecule providing helper function. The third nucleotide sequence can be a polynucleotide from an adenovirus or a herpes virus, preferably adenovirus.
An embodiment of a vector library of these teachings includes at least a first vector and a second vector, and each vector includes (a) a first nucleotide sequence encoding at least one AAV Rep protein from serotype 3; (b) a second nucleotide sequence encoding at least one AAV Cap protein differing from wildtype serotype 3 at least at one nucleotide position; and (c) a first AAV terminal repeat from serotype 3 and a second AAV terminal repeat from serotype 3, where the first and second nucleotide sequences are interposed between the first and second AAV terminal repeat, and the second vector differs from the first vector by at least one nucleotide.
An aspect of an embodiment of the vector library of these teachings includes the vector library being incorporated into at least one host cell. Examples of suitable host cells include HEK293 embryonic kidney cells, HeLa cells, Cos cells, U87 cells, KB cells, HepG2 cells and Vero cells, preferably HEK293 embryonic kidney cells.
An aspect of an embodiment of the vector library of these teachings further includes a third nucleotide sequence encoding at least one molecule providing helper function. The third nucleotide sequence can be a polynucleotide from an adenovirus or a herpes virus, preferably adenovirus.
An embodiment of an AAV virion of these teachings includes (a) a first nucleotide sequence encoding at least one AAV Rep protein from serotype 3; (b) a second nucleotide sequence encoding at least one AAV Cap protein differing from wildtype serotype 3 at least at one nucleotide position; and (c) a first AAV terminal repeat from serotype 3 and a second AAV terminal repeat from serotype 3, where the first and second nucleotide sequences are interposed between the first and second AAV terminal repeat.
An aspect of an embodiment of the AAV virion of these teachings includes the AAV virion being incorporated into at least one host cell. Examples of suitable host cells are mammalian cells including human host cells, including, for example blood cells, stem cells, hematopoietic cells, CD34 ' cells, liver cells, cancer cells, vascular cells, pancreatic cells, neural cells, ocular or retinal cells, epithelial or endothelial cells, dendritic cells, fibroblasts, or any other cell of mammalian origin, including, without limitation, hepatic
(i.e., liver) cells, lung cells, cardiac cells, pancreatic cells, intestinal cells, diaphragmatic cells, renal (i.e., kidney) cells, neural cells, blood cells, bone marrow cells, or any one or more selected tissues of a mammal for which viral-based gene therapy is contemplated. Preferably, the host cells are liver cells.
An aspect of an embodiment of the AAV virion of these teachings further includes a third nucleotide sequence encoding at least one molecule providing helper function. The third nucleotide sequence can be a polynucleotide from an adenovirus or a herpes virus, preferably adenovirus.
Certain embodiments of the non-naturally occurring nucleic acid and the vector library and the AAV virion of these teachings include the second nucleotide having the sequence:
T G C C C AC T TACAACAAC C AT C T C TACAAG CAAAT C T C C AG C WMD C AG GAG C T AS C AC GAC AC C AC T AC T T T G G C T AC AG C AC CCCT TGGGGG TAT T T TGACT T TAACAGAT TCCACTGCCACT TCTCACCACGTGACTGGCAGCGACTCAT T AAC AC AC T G G G GAT T C C G G C C C AG AAC T C AG C T T C AG C T C T T C AC AT C C AG T T AGAG G G G T C AC G C AGAAC GAT G G C AC GAC GAC T AT T G C C AT AC C T T AC C AG C AC G G T T CAAGTGT T TACGGACTCGGAGTATCAGCTCCCGTACGTGCTCGGGTCGGCGCACCAAGGC TGTCTCCCGCCGT T TCCAGCGGACGTCT TCATGGTCCCTCAGTATGGATACCTCACCCTG AACAACGGAAGTCAAGCGGTGGGACGCTCATCCT T T TACTGCCTGGAGTACT TCCCT TCG C AGAT G C T AG GAC T G GAAAT AC T T C C AT T C AG C TAT AC C T T C GAG GAT G T AC C T T T T C AC AG C AG C T AC G C T C AC AG C C AGAG T T T G GAT C G C T T GAT GAAT CCTCT TAT T GAT C AG TATCTGTACTACCTGAACAGAACGCAARGCAMCVCNRGCGGAACARCCRVCMHSMRSWS CTGVNGT T TAGCCAGGCTGGGCCTCAGTCTATGTCT T TGCAGGCCAGAAAT TGGCTACCT GGGCCCTGC T AC C G G C AC AGAGAC T T T C AMAR Y C BMC RVC S R S AC AC AC AG T RAS T T TCCT TGGMCAGCGGCCAGCAMATATCATCTCAATGGCCGCGACTCGCTGGTGAATCCA GGACCAGCTATGGCCAGTCACRRGGACGATRMS GRSARAT T T T TCCCTATGCACGGCAAT C TAATAT T T GGCAAAS AARRCRS CRVS RVARVC RAT R Y C GM S DWCGRS VRS GTAAT GAT T AC G GAT GAAGAAGAGAT T C G T AC C AC C AAT C C T G T G G C AAC AGAG C AG T AT G GAAC T G T G GCAAATAACT TGCAGRVSWSMRSRVCWS CCCACGDHTWSRNS GTCVMS CATCAGGGG GCCT TACCTGGCATGGTGTGGCAAGATCGT .
Certain embodiments of the non-naturally occurring nucleic acid and the vector library and the AAV virion of these teachings include the second nucleotide sequence encoding an AAV Cap protein that differs from wildtype serotype 3 at least at one amino position. The at least one differing amino acid position is preferably in a variable region (VR), and can be in VR-I, VR-IV, VR-V, VR-VI, VR-VII, VR-VIII and combinations thereof.
Certain aspects of the non-naturally occurring nucleic acid and the vector library and the AAV virion of these teachings include VR-I encoding amino acid sequence X1X2GAX3 where Xi is independently Q, N, K, T, S, R, H, P, D, E, A or G; X2 is independently S, T or A; and X3 is independently S or T.
Certain aspects of the non-naturally occurring nucleic acid and the vector library and the AAV virion of these teachings include VR-IV encoding amino acid sequence X4X5X6X7GTX8X9X10X11X12LX13 where X4 is independently G or S; X5 is independently T or N; X6 is independently T, P or A; X7 is independently S or G; Xs is independently T or A; X9 is independently N, T, S, D, A or G; X10 is independently Q, H, P, L, K, N, T, M or I; X11 is independently S, Q, H, R, K or N; X12 is independently R, K, N, T, S, Q, H, P, E, D, A or G; and Xi3 is independently L, K, T, R, M, Q, P, E, A, G or V.
Certain aspects of the non-naturally occurring nucleic acid and the vector library and the AAV virion of these teachings include VR-V encoding amino acid sequence X14X15X16X17X18 NNSX19FPWX20AASX21 where Xi4 is independently K or T; X15 is independently T, I, A or V; Xi6 is independently A, P, H, D, S or Y; X17 is independently N, T, S, D, A or G; Xis is independently D, E, G, Q, H or R; X19 is independently N, K, E or D; X20 is independently T or P; and X21 is independently K or T.
Certain aspects of the non-naturally occurring nucleic acid and the vector library and the AAV virion of these teachings include VR-VI encoding amino acid sequence X22DDX23X24X25 where X22 is independently K, R, E or G; X23 is independently E, T, K, N, A or D; X24 is independently E, D or G; and X25 is independently K or R.
Certain aspects of the non-naturally occurring nucleic acid and the vector library and the AAV virion of these teachings include VR-VII encoding amino acid sequence X26X27X28X29X30X31X32X33X34X35X36X37 where X26 is independently E or Q; X27 is independently G, N, S or D; X28 is independently T, S, G or A; X29 is independently T, K, N, R, S, E, D, A or G; X30 is independently A, K, T, R, E or G; X31 is independently S, N, T, D, A or G; X32 is independently N or D; X33 is independently A, T, I or V; X34 is independently E, A or D; X35 is independently L, N, I, D, V, Y or F; X36 is independently D, E or G; and X37 is independently N, K, R, S, Q, H, E, D or G.
Certain aspects of the non-naturally occurring nucleic acid and the vector library and the AAV virion of these teachings include VR-VIII encoding amino acid sequence X38X39X40X41X42PTX43X44X45VX46 where X38 is independently S, K, N, T, R, E, D, A or G; X39 is independently S, K, N, T, R, Q, H, P, E, D, A or G; X4o is independently N, Q, H, R, K or S; X4i is independently T, N, S, D, A or G; X42 is independently A, K, N, T, R, S, Q, H, P, E, D or G; X43 is independently T, N, I, D, A, V, Y, S or F; X44 is independently G, K, N, T, R, S, Q, H, P, E, D or A; X45 is independently T, K, N, R, S, M, I, E, D, A, G, or V; and X4e is independently N, T, K, P, Q, H, A, E or D.
Certain aspects of the non-naturally occurring nucleic acid and the vector library and the AAV virion of these teachings include the second nucleotide sequence encoding an AAV Cap protein having the sequence:
MAADGYLPDWLEDNLSEGIREWWALKPGVPQPKANQQHQDNRRGLVLPGYKYLGPGNGLD KGEPVNEADAAALEHDKAYDQQLKAGDNPYLKYNHADAEFQERLQEDTSFGGNLGRAVFQ AKKRILEPLGLVEEAAKTAPGKKGAVDQSPQEPDSSSGVGKSGKQPARKRLNFGQTGDSE SVPDPQPLGEPPAAPTSLGSNTMASGGGAPMADNNEGADGVGNSSGNWHCDSQWLGDRVI TTSTRTWALPTYNNHLYKQISSXXGAXNDNHYFGYSTPWGYFDFNRFHCHFSPRDWQRLI NNNWGFRPKKLSFKLFNIQVRGVTQNDGTTTIANNLTSTVQVFTDSEYQLPYVLGSAHQG CLPPFPADVFMVPQYGYLTLNNGSQAVGRSSFYCLEYFPSQMLRTGNNFQFSYTFEDVPF HSSYAHSQSLDRLMNPLIDQYLYYLNRTQXXXXGTXXXXXLXFSQAGPQSMSLQARNWLP GPCYRQQRLSXXXXXNNNSXFPWXAASXYHLNGRDSLVNPGPAMASHXDDXXXFFPMHGN LIFGKXXXXXXXXXXXXVMITDEEEIRTTNPVATEQYGTVANNLQXXXXXPTXXXVXHQG ALPGMVWQDRDVYLQGPIWA.
Certain aspects of the non-naturally occurring nucleic acid and the vector library and the AAV virion of these teachings include the second nucleotide encoding variants of an AAV Cap protein as listed in Table 4 (sequences numbered 2-86).
An embodiment of an AAV virion of these teachings includes (a) a first nucleotide sequence encoding at least one therapeutic molecule; (b) a second nucleotide sequence comprising a regulatory sequence; (c) a third nucleotide sequence comprising a first AAV terminal repeat from serotype 3; (d) a fourth nucleotide sequence comprising a second AAV terminal repeat from serotype 3; and (e) a capsid comprising at least one AAV Cap protein that differs from wildtype serotype 3 at least at one amino acid position. The first nucleotide sequence is operably linked to the second nucleotide sequence and the first and second nucleotide sequences are interposed between the first and second AAV terminal repeat to form a transgene, and the resulting transgene is packaged within the capsid. Examples of suitable regulatory sequences include promoters and enhancers, preferably a tissue specific promoeter. Examples of suitable therapeutic molecules include polypeptides, peptides, antibody, antigen binding fragment, ribozyme, peptide nucleic acid, siRNA, RNAi, antisense oligonucleotide, antisense polynucleotide, and any combination thereof, preferably a polypeptide, a peptide or an RNA.
An embodiment of a method of treating a disease of these teachings includes administering an effective amount of an AAV virion of these teachings. Such an AAV virion includes includes (a) a first nucleotide sequence encoding at least one therapeutic molecule; (b) a second nucleotide sequence comprising a regulatory sequence; (c) a third nucleotide sequence comprising a first AAV terminal repeat from serotype 3; (d) a fourth nucleotide sequence comprising a second AAV terminal repeat from serotype 3; and (e) a capsid comprising at least one AAV Cap protein that differs from wildtype serotype 3 at least at one amino acid position. The first nucleotide sequence is operably linked to the second nucleotide sequence and the first and second nucleotide sequences are interposed between the first and second AAV terminal repeat to form a transgene, and the resulting transgene is packaged within the capsid. Examples of suitable regulatory sequences include promoters and enhancers, preferably a tissue specific promoeter. Examples of suitable therapeutic molecules include polypeptides, peptides, antibody, antigen binding fragment, ribozyme, peptide nucleic acid, siRNA, RNAi, antisense oligonucleotide, antisense polynucleotide, and any combination thereof, preferably a polypeptide, a peptide or an RNA.
An embodiment of a method of selecting tissue-specific or cell-specific variants of an AAV virion includes (a) introducing a plurality of AAV virions into target tissues or cells; (b) allowing sufficient time to elapse to propagate additional virions; and (c) isolating the virions. Such an AAV virion includes (a) a first nucleotide sequence encoding at least one AAV Rep protein from serotype 3; (b) a second nucleotide sequence encoding at least one AAV Cap protein differing from wildtype serotype 3 at least at one nucleotide position; and (c) a first AAV terminal repeat from serotype 3 and a second AAV terminal repeat from serotype 3, where the first and second nucleotide sequences are interposed between the first and second AAV terminal repeat. Steps (a) - (c) can be repeated one or more times to enrich for a tissue- specific or cell-specific variant. Such enriched variants exhibit a higher target tropism for the target tissues or cells as compared to AAV serotype 3.
For promoting an understanding of the principles of the invention, reference will now be made to the embodiments, or examples, illustrated in the drawings and specific language will be used to describe the same. It will, nevertheless be understood that no limitation of the scope of the invention is thereby intended. Any alterations and further modifications in the described embodiments, and any further applications of the principles of the invention as described herein are contemplated as would normally occur to one of ordinary skill in the art to which the invention relates.
The following drawings form part of the present specification and are included to demonstrate certain aspects of the present invention. The application contains at least one drawing that is executed in color. Copies of this patent or patent application publication with color drawing(s) will be provided by the Patent and Trademark Office upon request and payment of the necessary fee. The invention may be better understood by reference to the following description taken in conjunction with the accompanying drawings, in which like reference numerals identify like elements.
BRIEF DESCRIPTION OF THE DRAWINGS
Figure 1 shows the wildtype (WT) nucleotide sequence (bottom rows) and corresponding WT amino acids (top rows, bold font) of AAV3B capsid gene and capsid protein, respectively. Degenerate positions within each variable region (VR) diversified in AAV serotype 3 capsid library (A3CL) are highlighted. The degenerate nucleotide positions (in IUPAC code) encoded by synthetic oligonucleotides are shown in italics below the WT sequence.
Figure 2 shows the nucleotide sequence of the synthetic fragment A3CL as designed.
The degenerate nucleotide positions (in IUPAC code) are underlined. The overlap stretches of the synthetic DNA and the plasmid vector backbone are highlighted.
Figure 3 shows the amino acid sequence of AAV3B VP1. Degenerate positions are labeled by X and underlined.
Figure 4 shows the amino acid sequences of the A3CL VRs encompassing WT AAV3B
VP1 capsid residues 259-600. WT sequences are shown in black, degenerate residues - in italics. Not modified conservative residues between VRs are not shown. VRs borders are indicated by vertical lines.
Figure 5 is a flowchart illustrating design and construction of AAV3B (A3CL) combinatorial capsid libraries ABC and D.
Figures 6-9 are photographs of agarose gels showing products of PCR reactions as per Example 2.
DETAILED DESCRIPTION OF THE INVENTION
Illustrative embodiments of the invention are described below. In the interest of clarity, not all features of an actual implementation are described in this specification. It will of course be appreciated that in the development of any such actual embodiment, numerous implementation-specific decisions must be made to achieve the developers' specific goals, such as compliance with system-related and business-related constraints, which will vary from one implementation to another. Moreover, it will be appreciated that such a development effort might be complex and time-consuming, but would be a routine undertaking for those of ordinary skill in the art having the benefit of this disclosure.
The present invention also provides improved rAAV-based genetic constructs that encode one or more therapeutic agents useful in the preparation of medicaments for the prevention, treatment, and/or amelioration of one or more diseases, disorders or dysfunctions resulting from a deficiency in one or more cellular components. In particular, the invention provides libraries of rAAV-based genetic constructs encoding one or more selected molecules of interest, such as, for example, one or more diagnostic or therapeutic agents (including, e.g., proteins, polypeptides, peptides, antibodies, antigen binding fragments, siRNAs, RNAis, antisense oligo- and poly-nucleotides, ribozymes, and variants and/or active fragments thereof), for use in the diagnosis, prevention, treatment, and/or amelioration of symptoms of mammalian diseases, disorders, dysfunctions, deficiencies, defects, trauma, injury, and such like.
The present invention also provides infectious rAAV virions, as well as nucleic acid molecules and rAAV vectors that encode the novel AAV vectors described herein, as well as nucleic acids encoding one or more selected diagnostic and/or therapeutic agents for delivery to a selected population of mammalian cells.
Preferably, the novel rAAV vectors, express constructs, and infectious virions and viral particles comprising them as disclosed herein preferably have an improved efficiency in transducing one or more of a variety of cells, tissues and organs of interest, when compared to wild-type, unmodified, expression constructs, and to the corresponding rAAV vectors and virions comprising them.
The improved rAAV vectors provided herein may transduce one or more selected host cells at higher-efficiencies (and often much higher efficiencies) than conventional, wild type (i.e., "unmodified") rAAV vectors. Likewise, vectors prepared as described herein may be of different AAV serotypes, and the mutation of one or more of the sequences described herein may result in improved viral vectors, which are capable of higher-efficiency transduction than that of the corresponding, non- substituted vectors from which the mutants were prepared.
The development of next-generation rAAV viral vectors may dramatically reduce the number of viral particles needed for a conventional gene therapy regimen. In addition to having improved transduction efficiencies for various mammalian cells, the rAAV vectors prepared as described herein may be more stable, less immunogenic, and/or can be produced at much lower cost, or in a higher titer, than an equivalent wild type viral vector prepared in conventional fashion.
In the practice of the invention, native amino acids normally present in the sequence of a viral capsid protein, may be substituted by one or more non-native amino acids, including, a substitution of one or more amino acids not normally present at a particular residue in the corresponding wild-type protein. The invention also provides isolated and purified polynucleotides that encode one or more of the disclosed viral vectors as described herein, as well as polynucleotides that encode such vectors. Preferably, the vector constructs of the present invention further include at least promoter capable of expressing the nucleic acid segment in a suitable host cell comprising the vector.
In the practice of the invention, the transduction efficiency of a mutated rAAV vector will be higher than that of the corresponding, unmodified, wild-type vector, and as such, will preferably possess a transduction efficiency in a mammalian cell that is at least 2-fold, at least about 4-fold, at least about 6-fold, at least about 8— fold, at least about 10-fold, or at least about 12-fold or higher in a selected mammalian host cell than that of a virion that comprises a corresponding, unmodified, rAAV vector. In certain embodiments, the transduction efficiency of the rAAV vectors provided herein will be at least about 15-fold higher, at least about 20-fold higher, at least about 25-fold higher, at least about 30-fold higher, or at least about 40, 45, or 50-fold or more greater than that of a virion that comprises a corresponding, wild-type vectors.
The present invention also concerns rAAV vectors, wherein the nucleic acid segment further comprises a promoter, an enhancer, a post-transcriptional regulatory sequence, a polyadenylation signal, or any combination thereof, operably linked to the nucleic acid segment that encodes the selected polynucleotide of interest. Preferably, the promoter is a heterologous promoter, a tissue-specific promoter, a cell-specific promoter, a constitutive promoter, an inducible promoter, or any combination thereof. In certain embodiments, nucleic acid segments cloned into one or more of the novel rAAV expression vectors described herein will preferably express or encode one or more polypeptides, peptides, ribozymes, peptide nucleic acids, siRNAs, RNAis, antisense oligonucleotides, antisense polynucleotides, antibodies, antigen binding fragments, or any combination thereof.
As noted herein, the therapeutic agents useful in the invention may include one or more agonists, antagonists, anti-apoptosis factors, inhibitors, receptors, cytokines, cytotoxins, erythropoietic agents, glycoproteins, growth factors, growth factor receptors, hormones, hormone receptors, interferons, interleukins, interleukin receptors, nerve growth factors, neuroactive peptides, neuroactive peptide receptors, proteases, protease inhibitors, protein decarboxylases, protein kinases, protein kinase inhibitors, enzymes, receptor binding proteins, transport proteins or one or more inhibitors thereof, serotonin receptors, or one or more uptake inhibitors thereof, serpins, serpin receptors, tumor suppressors, diagnostic molecules, chemotherapeutic agents, cytotoxins, or any combination thereof.
The invention further provides populations and pluralities of such rAAV vectors as prepared herein, as well as virions, infectious viral particles, and mammalian host cells that include one or more nucleic acid segments encoding them.
Preferably, the mammalian host cells will be human host cells, including, for example blood cells, stem cells, hematopoietic cells, CD34 ' cells, liver cells, cancer cells, vascular cells, pancreatic cells, neural cells, ocular or retinal cells, epithelial or endothelial cells, dendritic cells, fibroblasts, or any other cell of mammalian origin, including, without limitation, hepatic (i.e., liver) cells, lung cells, cardiac cells, pancreatic cells, intestinal cells, diaphragmatic cells, renal (i.e., kidney) cells, neural cells, blood cells, bone marrow cells, retinal cells or any one or more selected tissues of a mammal for which viral-based gene therapy is contemplated.
The invention further provides composition and formulations that include one or more of the proteins nucleic acid segments viral vectors, host cells, or viral particles of the present invention together with one or more pharmaceutically-acceptable buffers, diluents, or excipients. Such compositions may be included in one or more diagnostic or therapeutic kits, for diagnosing, preventing, treating or ameliorating one or more symptoms of a mammalian disease, injury, disorder, trauma or dysfunction.
The invention further includes a method for providing a mammal in need thereof with a diagnostically- or therapeutically-effective amount of a selected biological molecule, the method comprising providing to a cell, tissue or organ of a mammal in need thereof, an amount of an rAAV vector; and for a time effective to provide the mammal with a diagnostically- or a therapeutically-effective amount of the selected biological molecule.
The invention further provides a method for diagnosing, preventing, treating, or ameliorating at least one or more symptoms of a disease, a disorder, a dysfunction, an injury, an abnormal condition, or trauma in a mammal. In an overall and general sense, the method includes at least the step of administering to a mammal in need thereof one or more of the disclosed rAAV vectors, in an amount and for a time sufficient to diagnose, prevent, treat or ameliorate the one or more symptoms of the disease, disorder, dysfunction, injury, abnormal condition, or trauma in the mammal. The invention also provides a method of transducing a population of mammalian cells. In an overall and general sense, the method includes at least the step of introducing into one or more cells of the population, a composition that comprises an effective amount of one or more of the rAAV vectors disclosed herein.
In a further embodiment, the invention also provides isolated nucleic acid segments that encode one or more of the mutant viral capsid proteins as described herein, and provides recombinant vectors, virus particles, infectious virions, and isolated host cells that comprise one or more of the improved vector sequences described and tested herein.
Additionally, the present invention provides compositions, as well as therapeutic and/or diagnostic kits that include one or more of the disclosed AAV compositions, formulated with one or more additional ingredients, or prepared with one or more instructions for their use.
The invention also demonstrates methods for making, as well as methods of using the disclosed improved rAAV vectors in a variety of ways, including, for example, ex situ, in vitro and in vivo applications, methodologies, diagnostic procedures, and/or gene therapy regimens. Because many of the improved vectors described herein are also resistant to proteasomal degradation, they possess significantly increased transduction efficiencies in vivo making them particularly well suited for viral vector-based human gene therapy regimens, and in particular, for delivering one or more genetic constructs to selected mammalian cells in vivo and/or in vitro.
In one aspect, the invention provides compositions comprising AAV vectors, virions, viral particles, and pharmaceutical formulations thereof, useful in methods for delivering genetic material encoding one or more beneficial or therapeutic product(s) to mammalian cells and tissues. In particular, the compositions and methods of the invention provide a significant advancement in the art through their use in the treatment, prevention, and/or amelioration of symptoms of one or more mammalian diseases. It is contemplated that human gene therapy will particularly benefit from the present teachings by providing new and improved viral vector constructs for use in the treatment of a number of diverse diseases, disorders, and dysfunctions.
In another aspect, the invention concerns libraries of rAAV vector mutants that demonstrate improved properties useful in the delivery of one or more therapeutic agents to selected mammalian cells, and particularly for use in the prevention, treatment, and/or amelioration of one or more disorders in a mammal into which the vector construct may be introduced.
The rAAV vectors of the present invention may optionally further include one or more enhancer sequences that are each operably linked to the nucleic acid segment. Exemplary enhancer sequences include, but are not limited to, one or more selected from the group consisting of a CMV enhancer, a synthetic enhancer, a liver-specific enhancer, an vascular- specific enhancer, a brain-specific enhancer, a neural cell-specific enhancer, a lung-specific enhancer, a muscle-specific enhancer, a kidney-specific enhancer, a pancreas-specific enhancer, retinal-specific enhancer and an islet cell-specific enhancer.
Exemplary promoters useful in the practice of the invention include, without limitation, one or more heterologous, tissue-specific, constitutive or inducible promoters, including, for example, but not limited to, a promoter selected from the group consisting of a CMV promoter, a I3-actin promoter, an insulin promoter, an enolase promoter, a BDNF promoter, an NGF promoter, an EGF promoter, a growth factor promoter, an axon-specific promoter, a dendrite- specific promoter, a brain-specific promoter, a hippocampal-specific promoter, a kidney- specific promoter, a retinal-specific promoter, an elafin promoter, a cytokine promoter, an interferon promoter, a growth factor promoter, an ai-antitrypsin promoter, a brain cell-specific promoter, a neural cell-specific promoter, a central nervous system cell-specific promoter, a peripheral nervous system cell-specific promoter, an interleukin promoter, a serpin promoter, a hybrid CMV promoter, a hybrid I3-actin promoter, an EF 1 promoter, a Ul a promoter, a Ulb promoter, a Tet-inducible promoter, a VP1 6-LexA promoter, or any combination thereof. In exemplary embodiments, the promoter may include a mammalian or avian I3-actin promoter.
The vector-encoding nucleic acid segments may also further include one or more post- transcriptional regulatory sequences or one or more polyadenylation signals, including, for example, but not limited to, a woodchuck hepatitis virus post-transcription regulatory element, a polyadenylation signal sequence, or any combination thereof.
Exemplary diagnostic or therapeutic agents deliverable to host cells by the present vector constructs include, but are not limited to, an agent selected from the group consisting of a polypeptide, a peptide, an antibody, an antigen binding fragment, a ribozyme, a peptide nucleic acid, a siRNA, an RNAi, an antisense oligonucleotide, an antisense polynucleotide, and any combination thereof. In exemplary embodiments, the rAAV vectors obtained by the disclosed methods will preferably encode at least one diagnostic or therapeutic protein or polypeptide selected from the group consisting of a molecular marker, an adrenergic agonist, an anti-apoptosis factor, an apoptosis inhibitor, a cytokine receptor, a cytokine, a cytotoxin, an erythropoietic agent, a glutamic acid decarboxylase, a glycoprotein, a growth factor, a growth factor receptor, a hormone, a hormone receptor, an interferon, an interleukin, an interleukin receptor, a kinase, a kinase inhibitor, a nerve growth factor, a netrin, a neuroactive peptide, a neuroactive peptide receptor, a neurogenic factor, a neurogenic factor receptor, a neuropilin, a neurotrophic factor, a neurotrophin, a neurotrophin receptor, an N-methyl-D-aspartate antagonist, a plexin, a protease, a protease inhibitor, a protein decarboxylase, a protein kinase, a protein kinsase inhibitor, a proteolytic protein, a proteolytic protein inhibitor, a semaphorin,, a semaphorin receptor, a serotonin transport protein, a serotonin uptake inhibitor, a serotonin receptor, a serpin, a serpin receptor, a tumor suppressor, and any combination thereof.
In certain applications, the rAAV vectors of the present invention may include one or more nucleic acid segments that encode a polypeptide selected from the group consisting of BD F, CNTF, CSF, EGF, FGF, G-SCF, GM-CSF, gonadotropin, IFN, IFG-1, M-CSF, NGF, PDGF, PEDF, TGF, TGF-B2, TNF, VEGF, prolactin, somatotropin, XIAP1, IL-1, IL-2, IL-3, IL-4, IL-5, IL-6, IL-7, IL-8, IL-9, IL-10, IL-10(I87A), viral IL-10, IL-11, IL-12, IL-13, IL-14, IL-15, IL-16, IL-17, IL-18, and any combination thereof.
In another embodiment, the invention concerns genetically-modified, improved- transduction-efficiency rAAV vectors that include at least a first nucleic acid segment that encodes one or more therapeutic agents that alter, inhibit, reduce, prevent, eliminate, or impair the activity of one or more endogenous biological processes in the cell. In particular embodiments, such therapeutic agents may be those that selectively inhibit or reduce the effects of one or more metabolic processes, dysfunctions, disorders, or diseases. In certain embodiments, the defect may be caused by injury or trauma to the mammal for which treatment is desired. In other embodiments, the defect may be caused the over-expression of an endogenous biological compound, while in other embodiments still; the defect may be caused by the under-expression or even lack of one or more endogenous biological compounds.
The genetically-modified rAAV vectors and expression systems of the present invention may also further optionally include a second distinct nucleic acid segment that comprises, consists essentially of, or consists of, one or more enhancers, one or more regulatory elements, one or more transcriptional elements, or any combination thereof, that alter, improve, regulate, and/or affect the transcription of the nucleotide sequence of interest expressed by the modified rAAV vectors.
For example, the rAAV vectors of the present invention may further include a second nucleic acid segment that comprises, consists essentially of, or consists of, a CMV enhancer, a synthetic enhancer, a cell-specific enhancer, a tissue-specific enhancer, or any combination thereof. The second nucleic acid segment may also further comprise, consist essentially of, or consist of, one or more intron sequences, one or more post-transcriptional regulatory elements, or any combination thereof.
The improved vectors and expression systems of the present invention may also optionally further include a polynucleotide that comprises, consists essentially of, or consists of, one or more polylinkers, restriction sites, and/or multiple cloning region(s) to facilitate insertion (cloning) of one or more selected genetic elements, genes of interest, or therapeutic or diagnostic constructs into the rAAV vector at a selected site within the vector.
In further aspects of the present invention, the exogenous polynucleotide(s) that may be delivered into suitable host cells by the improved, capsid-modified, rAAV vectors disclosed herein are preferably of mammalian origin, with polynucleotides encoding one or more polypeptides or peptides of human, non-human primate, porcine, bovine, ovine, feline, canine, equine, epine, caprine, or lupine origin being particularly preferred.
The exogenous polynucleotide(s) that may be delivered into host cells by the disclosed capsid-modified viral vectors may, in certain embodiments, encode one or more proteins, one or more polypeptides, one or more peptides, one or more enzymes, or one or more antibodies (or antigen-binding fragments thereof), or alternatively, may express one or more siRNAs, ribozymes, antisense oligonucleotides, PNA molecules, or any combination thereof. When combinational gene therapies are desired, two or more different molecules may be produced from a single rAAV expression system, or alternatively, a selected host cell may be transfected with two or more unique rAAV expression systems, each of which may comprise one or more distinct polynucleotides that encode a therapeutic agent.
In other embodiments, the invention also provides rAAV vector mutants that are comprised within an infectious adeno-associated viral particle or a virion, as well as pluralities of such virions or infectious particles. Such vectors and virions may be comprised within one or more diluents, buffers, physiological solutions or pharmaceutical vehicles, or formulated for administration to a mammal in one or more diagnostic, therapeutic, and/or prophylactic regimens. The vectors, virus particles, virions, and pluralities thereof of the present invention may also be provided in excipient formulations that are acceptable for veterinary administration to selected livestock, exotics, domesticated animals, and companion animals (including pets and such like), as well as to non-human primates, zoological or otherwise captive specimens, and such like.
The invention also concerns host cells that comprise at least one of the disclosed rAAV expression vectors, or one or more virus particles or virions that comprise such an expression vector. Such host cells are particularly mammalian host cells, with human host cells being particularly highly preferred, and may be either isolated, in cell or tissue culture. In the case of genetically modified animal models, the transformed host cells may even be comprised within the body of a non-human animal itself.
In certain embodiments, the creation of recombinant non-human host cells, and/or isolated recombinant human host cells that comprise one or more of the disclosed rAAV vectors is also contemplated to be useful for a variety of diagnostic, and laboratory protocols, including, for example, means for the production of large-scale quantities of the rAAV vectors described herein. Such virus production methods are particularly contemplated to be an improvement over existing methodologies including in particular, those that require very high titers of the viral stocks in order to be useful as a gene therapy tool. The inventors contemplate that one very significant advantage of the present methods will be the ability to utilize lower titers of viral particles in mammalian transduction protocols, yet still retain transfection rates at a suitable level.
Compositions comprising one or more of the disclosed rAAV vectors, expression systems, infectious AAV particles, or host cells also form part of the present invention, and particularly those compositions that further comprise at least a first pharmaceutically- acceptable excipient for use in therapy, and for use in the manufacture of medicaments for the treatment of one or more mammalian diseases, disorders, dysfunctions, or trauma. Such pharmaceutical compositions may optionally further comprise one or more diluents, buffers, liposomes, a lipid, a lipid complex; or the tyrosine-modified rAAV vectors may be comprised within a microsphere or a nanoparticle.
Pharmaceutical formulations suitable for intramuscular, intravenous, or direct injection into an organ or tissue or a plurality of cells or tissues of a human or other mammal are particularly preferred, however, the compositions disclosed herein may also find utility in administration to discreet areas of the mammalian body, including for example, formulations that are suitable for direct injection into one or more organs, tissues, or cell types in the body. Such injection sites include, but are not limited to, the brain, a joint or joint capsule, a synovium or subsynovium tissue, tendons, ligaments, cartilages, bone, peri-articular muscle or an articular space of a mammalian joint, as well as direct administration to an organ such as the heart, liver, lung, pancreas, intestine, brain, bladder, kidney, or other site within the patient's body, including, for example , introduction of the viral vectors via intraabdominal, intrathorascic, intravascular, or intracerebroventricular delivery.
Other aspects of the invention concern recombinant adeno-associated virus virion particles, compositions, and host cells that comprise, consist essentially of, or consist of, one or more of the rAAV vectors disclosed herein, such as for example pharmaceutical formulations of the vectors intended for administration to a mammal through suitable means, such as, by intramuscular, intravenous, intra-articular, or direct injection to one or more cells, tissues, or organs of a selected mammal. Typically, such compositions may be formulated with pharmaceutically-acceptable excipients as described hereinbelow, and may comprise one or more liposomes, lipids, lipid complexes, microspheres or nanoparticle formulations to facilitate administration to the selected organs, tissues, and cells for which therapy is desired.
Kits comprising one or more of the disclosed rAAV vectors (as well as one or more virions, viral particles, transformed host cells or pharmaceutical compositions comprising such vectors); and instructions for using such kits in one or more therapeutic, diagnostic, and/or prophylactic clinical embodiments are also provided by the present invention. Such kits may further comprise one or more reagents, restriction enzymes, peptides, therapeutics, pharmaceutical compounds, or means for delivery of the composition(s) to host cells, or to an animal (e.g., syringes, injectables, and the like). Exemplary kits include those for treating, preventing, or ameliorating the symptoms of a disease, deficiency, dysfunction, and/or injury, or may include components for the large-scale production of the viral vectors themselves, such as for commercial sale, or for use by others, including e.g., virologists, medical professionals, and the like.
Another important aspect of the present invention concerns methods of use of the disclosed rAAV vectors, virions, expression systems, compositions, and host cells described herein in the preparation of medicaments for diagnosing, preventing, treating or ameliorating at least one or more symptoms of a disease, a dysfunction, a disorder, an abnormal condition, a deficiency, injury, or trauma in an animal, and in particular, in a vertebrate mammal. Such methods generally involve administration to a mammal in need thereof, one or more of the disclosed vectors, virions, viral particles, host cells, compositions, or pluralities thereof, in an amount and for a time sufficient to diagnose, prevent, treat, or lessen one or more symptoms of such a disease, dysfunction, disorder, abnormal condition, deficiency, injury, or trauma in the affected animal. The methods may also encompass prophylactic treatment of animals suspected of having such conditions, or administration of such compositions to those animals at risk for developing such conditions either following diagnosis, or prior to the onset of symptoms.
As described above, the exogenous polynucleotide will preferably encode one or more proteins, polypeptides, peptides, nbozymes, or antisense oligonucleotides, or a combination of these. In fact, the exogenous polynucleotide may encode two or more such molecules, or a plurality of such molecules as may be desired. When combinational gene therapies are desired, two or more different molecules may be produced from a single rAAV expression system, or alternatively, a selected host cell may be transfected with two or more unique rAAV expression systems, each of which will provide unique heterologous polynucleotides encoding at least two different such molecules.
Compositions comprising one or more of the disclosed rAAV vectors, expression systems, infectious AAV particles, host cells also form part of the present invention, and particularly those compositions that further comprise at least a first pharmaceutically- acceptable excipient for use in the manufacture of medicaments and methods involving therapeutic administration of such rAAV vectors. Such pharmaceutical compositions may optionally further comprise liposomes, a lipid, a lipid complex; or the rAAV vectors may be comprised within a microsphere or a nanoparticle. Pharmaceutical formulations suitable for intramuscular, intravenous, or direct injection into an organ or tissue of a human are particularly preferred. USE OF RAAV VECTORS IN PROPHYLAXIS, DIAGNOSIS, OR THERAPY The present invention provides compositions including one or more of the disclosed rAAV vectors comprised within a kit for diagnosing, preventing, treating or ameliorating one or more symptoms of a mammalian disease, injury, disorder, trauma or dysfunction. Such kits may be useful in the diagnosis, prophylaxis, and/or therapy or a human disease, and may be particularly useful in the treatment, prevention, and/or amelioration of one or more symptoms of wet age-related macular degeneration, dry age-related macular degeneration, glaucoma, retinitis pigmentosa, diabetic retinopathy, orphan ophthalmological diseases, cancer, diabetes, autoimmune disease, kidney disease, cardiovascular disease, pancreatic disease, intestinal disease, liver disease, neurological disease, neuromuscular disorder, neuromotor deficit, neuroskeletal impairment, neurological disability, neurosensory dysfunction, stroke, ischemia, a 1-antitiypsin (AAT) deficiency, Batten's disease, Alzheimer's disease, sickle cell disease, f3- thalassamia, Huntington's disease, Parkinson's disease, skeletal disease, trauma, pulmonary disease in a human.
The invention also provides for the use of a composition disclosed herein in the manufacture of a medicament for treating, preventing or ameliorating the symptoms of a disease, disorder, dysfunction, injury or trauma, including, but not limited to, the treatment, prevention, and/or prophylaxis of a disease, disorder or dysfunction, and/or the amelioration of one or more symptoms of such a disease, disorder or dysfunction.
The invention also provides a method for treating or ameliorating the symptoms of such a disease, injury, disorder, or dysfunction in a mammal. Such methods generally involve at least the step of administering to a mammal in need thereof, one or more of the rAAV vectors as disclosed herein, in an amount and for a time sufficient to treat or ameliorate the symptoms of such a disease, injury, disorder, or dysfunction in the mammal. Such treatment regimens are particularly contemplated in human therapy, via administration of one or more compositions either intramuscularly, intravenously, subcutaneously, intrathecally, intraperitoneally, or by direct injection into an organ or a tissue of the mammal under care.
The invention also provides a method for providing to a mammal in need thereof, a therapeutically-effective amount of an rAAV composition of the present invention, in an amount, and for a time effective to provide the patient with a therapeutically-effective amount of the desired therapeutic agent(s) encoded by one or more nucleic acid segments comprised within the rAAV vector. Exemplary therapeutic agents include, but are not limited to, a polypeptide, a peptide, an antibody, an antigen-binding fragment, a ribozyme, a peptide nucleic acid, an siRNA, an RNAi, an antisense oligonucleotide, an antisense polynucleotide, or a combination thereof.
PHARMACEUTICAL COMPOSITIONS
The genetic constructs of the present invention may be prepared in a variety of compositions, and may also be formulated in appropriate pharmaceutical vehicles for administration to human or animal subjects.
The invention also provides compositions comprising one or more of the disclosed rAAV vectors, expression systems, virions, viral particles, mammalian cells, or combinations thereof. In certain embodiments, the present invention provides pharmaceutical formulations of one or more rAAV vectors disclosed herein for administration to a cell or an animal, either alone or in combination with one or more other modalities of therapy, and in particular, for therapy of human cells, tissues, and diseases affecting man. Formulation of pharmaceutically- acceptable excipients and carrier solutions is well-known to those of skill in the art, as is the development of suitable dosing and treatment regimens for using the particular compositions described herein in a variety of treatment regimens, including e.g., oral, parenteral, intravenous, intranasal, intra-articular, intramuscular administration and formulation.
LIBRARY DESIGN AND CONSTRUCTION
Comparison of the AAV VP3 structure among various serotypes has revealed highly homologous sequences interspersed with more evolutionary divergent areas. These amino acid stretches are commonly designated as VRs I through IX (variable regions I-IX; also known as "loops"). VRs are localized at the surface of the assembled capsid and are assumed to be responsible for the capsid interaction with cell surface receptors and other host factors. Because of their location, VRs are also predicted to be less critical for capsid assembly. Therefore, the guiding principle of the library's design was to modify only surface VRs while keeping the backbone sequence unchanged to maintain the integrity of the assembling scaffold. All candidate positions for mutagenesis, in the AAV3 background, were selected from the alignment of known variants, which can be evaluated on a three dimensional model of the AAV3 capsid. The amino acid diversity of VR-I, VR-IV, VR-V, VR-VI, VR-VII and VR-VIII is shown in Figure 4. AAV3 wildtype VR-II, VR-III and VR-IX and non-variable regions of VP3 were incorporated in the plasmid library.
The library was built in three steps: first, VR parent sub-libraries were prepared each containing mutations in only one VR (B: VR-IV, C: VR-VII, D: VR-VIII) or a subset of VRs (A: VR-I + VR-V + VR-VI), then, structurally compatible sequences were combined to generate master libraries (A + B + C: VRs I, IV, V, VI, VII) and (D: VR-VIII), and finally the master libraries were packaged. See Examples, Example 2 and Figure 5. Methods for generating and assembling DNA fragments for the library are disclosed in WO2015/048534 and US 7220577, both of which are incorporated herein in their entirety.
TISSUE-SPECIFIC OR CELL-SPECIFIC VIRIONS
The master libraries can be used to select virions having capsids containing degenerate or otherwise modified Cap protein (i.e., Cap protein that differs from wildtype serotype 3 at least at one amino acid position) that are targeted to particular tissue or cell types. For example, virions made according to the invention include those that exhibit a new tropism, e.g., those capable of infecting cells normally non-permissive to AAV infection in general or at least non-permissive to AAV3 infection, as well as those that exhibit an increased or decreased ability to infect a particular cell or tissue type. As another example, virions made according to the invention include those that lack the ability to infect cells normally permissive to AAV infection in general or at least normally permissive to AAV3 infection. To select for virions having a particular cell- or tissue-specific tropism, a packaged master library is introduced into a target cell. Preferably, the target cell is also infected with a helper virus (e.g. Ad). The target cell is cultured under conditions that allow for the production of virions, resulting in a population of virions that are harvested from the target cell. This population of virions has been selected for having a tropism for that target cell.
As controls in a typical experiment in which virions having a particular tropism are selected, cells in different flasks or dishes can be simultaneously infected with WT AAV3 or rAAV using the same conditions as used for the library. After a suitable time post-infection, cells can be harvested, washed and the virions purified using a suitable purification method (See Gao et al., Hum. Gene Ther. 9:2353-62, 1998; U.S. Pat. No. 6, 146,874; and Zolotukhin et al., Gene Ther. 6:973-85, 1999). AAV and helper virions (e.g., Ad) from each infection can be tittered, by real-time PCT for example, and the AAV virions can then be further propagated, resulting in a stock of selected virions.
Once the selected population of virions having a desired tropism is isolated, nucleic acid from the virions is isolated and the sequence of the nucleotide sequence encoding the at least one AAV Cap protein is determined. Virions made and selected according to the invention that can specifically target diseased cells or tissues over non-diseased cells or tissues are particularly useful.
Alternatively tissue- or cell-specific virions can be selected using an in vivo approach. For example, mice (or other suitable host) can be injected with a suitable amount viral preparation (e.g., 1 x 1010 to 1 x 1011 vg in the case of mice) via the tail vein. More than one round of selection can be performed by injecting original master library for the first round and target-enriched libraries in subsequent rounds. Hosts are euthanized after an incubation period (3 to 4 days for mice), and episomal DNA is purified from the target cells or tissue and used as a template to amplify capsid DNA sequences. Target-enriched libraries can then be generated, purified and quantified. After several rounds of selection, amplified capsid DNA can be inserted into an appropriate vector for cloning and random clones can be analyzed by sequencing.
EXEMPLARY DEFINITIONS
Unless otherwise defined, all technical terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs. Commonly understood definitions of molecular biology terms can be found in Rieger et al., (1991); Lewin (1994). Commonly understood definitions of virology terms can be found in Granoff and Webster (1999) and Tidona and Darai (2002). Commonly understood definitions of microbiology can be found in Singleton and Sainsbury (2002).
In accordance with long standing patent law convention, the words "a" and "an" when used in this application, including the claims, denotes "one or more."
The terms "about" and "approximately" as used herein, are interchangeable, and should generally be understood to refer to a range of numbers around a given number, as well as to all numbers in a recited range of numbers (e.g., "about 5 to 15" means "about 5 to about 15" unless otherwise stated). Moreover, all numerical ranges herein should be understood to include each whole integer within the range. As used herein, the term "carrier" is intended to include any solvent(s), dispersion medium, coating(s), diluent(s), buffer(s), isotonic agent(s), solution(s), suspension(s), colloid(s), inert(s) or such like, or a combination thereof, that is pharmaceutically acceptable for administration to the relevant animal. The use of one or more delivery vehicles for chemical compounds in general, and chemotherapeutics in particular, is well known to those of ordinary skill in the pharmaceutical arts. Except insofar as any conventional media or agent is incompatible with the active ingredient, its use in the diagnostic, prophylactic, and therapeutic compositions is contemplated. One or more supplementary active ingredient(s) may also be incorporated into, or administered in association with, one or more of the disclosed chemotherapeutic compositions.
As used herein, the term "chimeric rcAAV" refers to a replication-competent AAV- derived nucleic acid containing at least one nucleotide sequence that 1) encodes an AAV protein and 2) differs from the corresponding native nucleotide sequence in one or more bases.
As used herein, the term "DNA segment" refers to a DNA molecule that has been isolated free of total genomic DNA of a particular species. Therefore, a DNA segment obtained from a biological sample using one of the compositions disclosed herein refers to one or more DNA segments that have been isolated away from, or purified free from, total genomic DNA of the particular species from which they are obtained. Included within the term "DNA segment," are DNA segments and smaller fragments of such segments, as well as recombinant vectors, including, for example, plasmids, cosmids, phage, viruses, and the like.
The term "e.g.," as used herein, is used merely by way of example, without limitation intended, and should not be construed as referring only those items explicitly enumerated in the specification.
As used herein, "an effective amount" would be understood by those of ordinary skill in the art to provide a therapeutic, prophylactic, or otherwise beneficial effect against the organism, its infection, or the symptoms of the organism or its infection, or any combination thereof
The phrase "expression control sequence" refers to any genetic element (e.g., polynucleotide sequence) that can exert a regulatory effect on the replication or expression (transcription or translation) of another genetic element. Common expression control sequences include promoters, polyadenylation (polyA) signals, transcription termination sequences, upstream regulatory domains, origins of replication, internal ribosome entry sites (IRES), enhancers, and the like. A "tissue specific expression control sequence" is one that exerts a regulatory effect on the replication or expression (transcription or translation) of another genetic element in only one type of tissue or a small subset of tissues.
The phrase "helper function" is meant as a functional activity performed by a nucleic acid or polypeptide that is derived from a virus such as Adenovirus (Ad) or herpesvirus and that facilitates AAV replication in a host cell.
As used herein, a "heterologous" is defined in relation to a predetermined referenced gene sequence. For example, with respect to a structural gene sequence, a heterologous promoter is defined as a promoter which does not naturally occur adjacent to the referenced structural gene, but which is positioned by laboratory manipulation. Likewise, a heterologous gene or nucleic acid segment is defined as a gene or segment that does not naturally occur adjacent to the referenced promoter and/or enhancer elements.
As used herein, the term "homology" refers to a degree of complementarity between two or more polynucleotide or polypeptide sequences. The word "identity" may substitute for the word "homology" when a first nucleic acid or amino acid sequence has the exact same primary sequence as a second nucleic acid or amino acid sequence. Sequence homology and sequence identity can be determined by analyzing two or more sequences using algorithms and computer programs known in the art. Such methods may be used to assess whether a given sequence is identical or homologous to another selected sequence.
As used herein, "homologous" means, when referring to polynucleotides, sequences that have the same essential nucleotide sequence, despite arising from different origins. Typically, homologous nucleic acid sequences are derived from closely related genes or organisms possessing one or more substantially similar genomic sequences. By contrast, an "analogous" polynucleotide is one that shares the same function with a polynucleotide from a different species or organism, but may have a significantly different primary nucleotide sequence that encodes one or more proteins or polypeptides that accomplish similar functions or possess similar biological activity. Analogous polynucleotides may often be derived from two or more organisms that are not closely related (e.g., either genetically or phylogenetically).
The terms "identical" or percent "identity," in the context of two or more nucleic acid or polypeptide sequences, refer to two or more sequences or subsequences that are the same or have a specified percentage of amino acid residues or nucleotides that are the same, when compared and aligned for maximum correspondence, as measured using one of the sequence comparison algorithms described below (or other algorithms available to persons of ordinary skill) or by visual inspection.
As used herein, the phrase "in need of treatment" refers to a judgment made by a caregiver such as a physician or veterinarian that a patient requires (or will benefit in one or more ways) from treatment. Such judgment may made based on a variety of factors that are in the realm of a caregiver's expertise, and may include the knowledge that the patient is ill as the result of a disease state that is treatable by one or more compound or pharmaceutical compositions such as those set forth herein.
The phrases "isolated" or "biologically pure" refer to material that is substantially, or essentially, free from components that normally accompany the material as it is found in its native state. Thus, isolated polynucleotides in accordance with the invention preferably do not contain materials normally associated with those polynucleotides in their natural, or in situ, environment.
As used herein, the term "kit" may be used to describe variations of the portable, self- contained enclosure that includes at least one set of components to conduct one or more of the diagnostic or therapeutic methods of the invention.
"Link" or "join" refers to any method known in the art for functionally connecting one or more proteins, peptides, nucleic acids, or polynucleotides, including, without limitation, recombinant fusion, covalent bonding, disulfide bonding, ionic bonding, hydrogen bonding, electrostatic bonding, and the like.
The term "library" refers to a collection of elements that differ from one another in at least one aspect. For example, a vector library is a collection of at least two vectors that differ from one another by at least one nucleotide. As another example, a "virion library" is a collection of at least two virions that differ from one another by at least one nucleotide or at least one capsid protein.
As used herein, the term "master library" or "combined library" refers to a pool of rAAV virions composed of chimeric rcAAV vectors encapsidated in cognate chimeric capsids (e.g., capsids containing a degenerate or otherwise modified Cap protein). As used herein, the term "parent sub-library" refers to a pool of rAAV virions composed of chimeric rcAAV vectors encapsidated in cognate chimeric capsids (e.g., capsids containing degenerate or otherwise modified Cap protein). More than one parent sub-library can be combined to create a master library or combined library.
When referring to a nucleic acid molecule or polypeptide, the term "native" refers to a naturally-occurring (e.g., a WT) nucleic acid or polypeptide.
The term "naturally occurring" as used herein as applied to an object refers to the fact that an object can be found in nature. For example, a polypeptide or polynucleotide sequence that is present in an organism (including viruses) that can be isolated from a source in nature and which has not been intentionally modified by the hand of man in a laboratory is naturally- occurring. As used herein, laboratory strains of rodents that may have been selectively bred according to classical genetics are considered naturally occurring animals.
As used herein, the phrase "nucleic acid" means a chain of two or more nucleotides such as RNA (ribonucleic acid) and DNA (deoxyribonucleic acid). Conventional nomenclature exists in the art for polynucleotide and polypeptide structures. For example, one-letter abbreviations are widely employed to describe nucleotides: Adenine (A), Guanine (G), Cytosine (C), Thymine (T), Uracil (U), Purine, i.e. A or G (R), Pyrimidine, i.e. C or T (Y), any nucleotide (N), Weak, i.e. A or T (W), Strong, i.e. G or C (S), Amino, i.e. A or C (M), Keto, i.e. G or T (K), not A, i.e. G or C or T (B), not G, i.e. A or C or T (H), not C, i.e. A or G or T (D) and not T, i. e. A or G or C (V).
The phrases "cap nucleic acid," "cap gene," and "capsid gene" as used herein mean a nucleic acid that encodes a Cap protein. Examples of cap nucleic acids include "wild-type" (WT) Cap-encoding nucleic acid sequences from AAV serotypes 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13; a native form cap cDNA; a nucleic acid having sequences from which a cap cDNA can be transcribed; and/or allelic variants and homologs of the foregoing.
"VR", "VRs", "variable region" or "variable regions" refer to amino acid stretches of capsid protein that do not have a high degree of homology between AAV variants. These amino acid stretches are commonly designated as VRs I through IX (also known as "loops"). VRs are localized at the surface of the assembled capsid and interact with host cell surface receptors and other host factors. As used herein, the term "patient" (also interchangeably referred to as "host" or "subject") refers to any host that can receive one or more of the pharmaceutical compositions disclosed herein. Preferably, the subject is a vertebrate animal, which is intended to denote any animal species (and preferably, a mammalian species such as a human being). In certain embodiments, a "patient" refers to any animal host including without limitation any mammalian host. Preferably, the term refers to any mammalian host, the latter including but not limited to, human and non-human primates, bovines, canines, caprines, cavines, corvines, epines, equines, felines, hircines, lapines, leporines, lupines, murines, ovines, porcines, ranines, racines, vulpines, and the like, including livestock, zoological specimens, exotics, as well as companion animals, pets, and any animal under the care of a veterinary practitioner.
The phrase "pharmaceutically-acceptable" refers to molecular entities and compositions that preferably do not produce an allergic or similar untoward reaction when administered to a mammal, and in particular, when administered to a human. As used herein, "pharmaceutically acceptable salt" refers to a salt that preferably retains the desired biological activity of the parent compound and does not impart any undesired toxicological effects. Examples of such salts include, without limitation, acid addition salts formed with inorganic acids (e.g., hydrochloric acid, hydrobromic acid, sulfuric acid, phosphoric acid, nitric acid, and the like); and salts formed with organic acids including, without limitation, acetic acid, oxalic acid, tartaric acid, succinic acid, maleic acid, fumaric acid, gluconic acid, citric acid, malic acid, ascorbic acid, benzoic acid, tannic acid, pamoic (embonic) acid, alginic acid, naphthoic acid, polyglutamic acid, naphthalenesulfonic acids, naphthalenedisulfonic acids, polygalacturonic acid; salts with polyvalent metal cations such as zinc, calcium, bismuth, barium, magnesium, aluminum, copper, cobalt, nickel, cadmium, and the like; salts formed with an organic cation formed from Ν,Ν'-dibenzylethylenediamine or ethylenediamine; and combinations thereof The term "pharmaceutically acceptable salt" as used herein refers to a compound of the present disclosure derived from pharmaceutically acceptable bases, inorganic or organic acids. Examples of suitable acids include, but are not limited to, hydrochloric, hydrobromic, sulfuric, nitric, perchloric, fumaric, maleic, phosphoric, glycollic, lactic, salicyclic, succinic, toluene-p- sulfonic, tartaric, acetic, citric, methanesulfonic, formic, benzoic, malonic, naphthalene-2- sulfonic, trifluoroacetic and benzenesulfonic acids. Salts derived from appropriate bases include, but are not limited to, alkalis such as sodium and ammonia. As used herein, the term "plasmid" or "vector" refers to a genetic construct that is composed of genetic material (i.e., nucleic acids). Typically, a plasmid or a vector contains an origin of replication that is functional in bacterial host cells, e.g., Escherichia coli, and selectable markers for detecting bacterial host cells including the plasmid. Plasmids and vectors of the present invention may include one or more genetic elements as described herein arranged such that an inserted coding sequence can be transcribed and translated in a suitable expression cells. In addition, the plasmid or vector may include one or more nucleic acid segments, genes, promoters, enhancers, activators, multiple cloning regions, or any combination thereof, including segments that are obtained from or derived from one or more natural and/or artificial sources.
As used herein, the term "polypeptide" is intended to encompass a singular "polypeptide" as well as plural "polypeptides," and includes any chain or chains of two or more amino acids. Thus, as used herein, terms including, but not limited to "peptide," "dipeptide," "tripeptide," "protein," "enzyme," "amino acid chain," and "contiguous amino acid sequence" are all encompassed within the definition of a "polypeptide," and the term "polypeptide" can be used instead of, or interchangeably with, any of these terms. The term further includes polypeptides that have undergone one or more post-translational modification(s), including for example, but not limited to, glycosylation, acetylation, phosphorylation, amidation, derivatization, proteolytic cleavage, post-translation processing, or modification by inclusion of one or more non-naturally occurring amino acids. Conventional nomenclature exists in the art for polynucleotide and polypeptide structures. For example, one-letter and three-letter abbreviations are widely employed to describe amino acids: Alanine (A; Ala), Arginine (R; Arg), Asparagine (N; Asn), Aspartic Acid (D; Asp), Cysteine (C; Cys), Glutamine (Q; Gin), Glutamic Acid (E; Glu), Glycine (G; Gly), Histidine (H; His), Isoleucine (I; He), Leucine (L; Leu), Methionine (M; Met), Phenylalanine (F; Phe), Proline (P; Pro), Serine (S; Ser), Threonine (T; Thr), Tryptophan (W; Trp), Tyrosine (Y; Tyr), Valine (V; Val), and Lysine (K; Lys). Additional conventions include: Asn or Asp (B; Asx), Gin or Glu (Z; Glx), Leu or He (J; Xle), Selenocysteine (U; Sec), Pyrrolysine (O; Pyl) and Unknown (X; Unk). Amino acid residues described herein are preferred to be in the "L" isomeric form. However, residues in the "D" isomeric form may be substituted for any L-amino acid residue provided the desired properties of the polypeptide are retained. As used herein, the terms "prevent," "preventing," "prevention," "suppress," "suppressing," and "suppression" as used herein refer to administering a compound either alone or as contained in a pharmaceutical composition prior to the onset of clinical symptoms of a disease state so as to prevent any symptom, aspect or characteristic of the disease state. Such preventing and suppressing need not be absolute to be deemed medically useful.
The term "promoter," as used herein refers to a region or regions of a nucleic acid sequence that regulates transcription.
"Protein" is used herein interchangeably with "peptide" and "polypeptide," and includes both peptides and polypeptides produced synthetically, recombinantly, or in vitro and peptides and polypeptides expressed in vivo after nucleic acid sequences are administered into a host animal or human subject. The term "polypeptide" is preferably intended to refer to any amino acid chain length, including those of short peptides from two to about 20 amino acid residues in length, oligopeptides from about 10 to about 100 amino acid residues in length, and longer polypeptides including those of about 100 or more amino acid residues in length. Furthermore, the term is also intended to include enzymes, i.e., functional biomolecules including at least one amino acid polymer. Polypeptides and proteins of the present invention also include polypeptides and proteins that are or have been post-translationally modified, and include any sugar or other derivative(s) or conjugate(s) added to the backbone amino acid chain.
The term "pseudotyped" is meant a nucleic acid or genome derived from a first AAV serotype that is encapsidated (packaged) into an AAV capsid containing at least one AAV Cap protein of a second serotype differing from the first serotype.
As used herein, the term "rcAAV vector" refers to a replication-competent AAV- derived nucleic acid capable of DNA replication in a cell without any additional AAV genes or gene products.
The term "recombinant" indicates that the material (e.g., a polynucleotide or a polypeptide) has been artificially or synthetically (non-naturally) altered by human intervention. The alteration can be performed on the material within or removed from, its natural environment or state. Specifically, e.g., a promoter sequence is "recombinant" when it is produced by the expression of a nucleic acid segment engineered by the hand of man. For example, a "recombinant nucleic acid" is one that is made by recombining nucleic acids, e.g., during cloning, DNA shuffling or other procedures, or by chemical or other mutagenesis; a "recombinant polypeptide" or "recombinant protein" is a polypeptide or protein which is produced by expression of a recombinant nucleic acid; and a "recombinant virus," e.g., a recombinant AAV virus, is produced by the expression of a recombinant nucleic acid.
The term "regulatory element," as used herein, refers to a region or regions of a nucleic acid sequence that regulates transcription. Exemplary regulatory elements include, but are not limited to, enhancers, post-transcriptional elements, transcriptional control sequences, and such like.
The term "RNA segment" refers to an RNA molecule that has been isolated free of total cellular RNA of a particular species. Therefore, RNA segments can refer to one or more RNA segments (either of native or synthetic origin) that have been isolated away from, or purified free from, other RNAs. Included within the term "RNA segment," are RNA segments and smaller fragments of such segments.
The terms "substantially corresponds to," "substantially homologous," or "substantial identity," as used herein, denote a characteristic of a nucleic acid or an amino acid sequence, wherein a selected nucleic acid or amino acid sequence has at least about 70 or about 75 percent sequence identity as compared to a selected reference nucleic acid or amino acid sequence. More typically, the selected sequence and the reference sequence will have at least about 76, 77, 78, 79, 80, 81, 82, 83, 84 or even 85 percent sequence identity, and more preferably, at least about 86, 87, 88, 89, 90, 91, 92, 93, 94, or 95 percent sequence identity. More preferably still, highly homologous sequences often share greater than at least about 96, 97, 98, or 99 percent sequence identity between the selected sequence and the reference sequence to which it was compared.
The percentage of sequence identity may be calculated over the entire length of the sequences to be compared, or may be calculated by excluding small deletions or additions which total less than about 25 percent or so of the chosen reference sequence. The reference sequence may be a subset of a larger sequence, such as a portion of a gene or flanking sequence, or a repetitive portion of a chromosome. However, in the case of sequence homology of two or more polynucleotide sequences, the reference sequence will typically comprise at least about 18-25 nucleotides, more typically at least about 26 to 35 nucleotides, and even more typically at least about 40, 50, 60, 70, 80, 90, or even 100 or so nucleotides. When highly-homologous fragments are desired, the extent of percent identity between the two sequences will be at least about 80%, preferably at least about 85%, and more preferably about 90% or 95% or higher, as readily determined by one or more of the sequence comparison algorithms well-known to those of skill in the art, such as e.g., the FASTA program analysis described by Pearson and Lipman (1988).
As used herein, the term "structural gene" is intended to generally describe a polynucleotide, such as a gene, that is expressed to produce an encoded peptide, polypeptide, protein, ribozyme, catalytic RNA molecule, or antisense molecule.
The term "subject," as used herein, describes an organism, including a mammal such as a human primate, to which treatment with one or more of the disclosed compositions may be provided. Mammalian species that may benefit from the disclosed treatment methods include, without limitation, humans, non-human primates such as apes; chimpanzees; monkeys, and orangutans, domesticated animals, including dogs and cats, as well as livestock such as horses, cattle, pigs, sheep, and goats, or other mammalian species including, without limitation, mice, rats, guinea pigs, rabbits, hamsters, and the like.
As used herein, the terms "terminal repeat" or "TR" mean a nucleic acid sequence derived from an AAV that is required in cis for replication and packaging of AAV.
"Transcriptional regulatory element" refers to a polynucleotide sequence that activates transcription alone or in combination with one or more other nucleic acid sequences. A transcriptional regulatory element may include, for example, one or more promoters, one or more response elements, one or more negative regulatory elements, one or more enhancers, or any combination thereof
As used herein, a "transcription factor recognition site" and a "transcription factor binding site" refer to a polynucleotide sequence(s) or sequence motif(s) that are identified as being sites for the sequence-specific interaction of one or more transcription factors, frequently taking the form of direct protein-DNA binding. Typically, transcription factor binding sites can be identified by DNA footprinting, gel mobility shift assays, and the like, and/or can be predicted based on known consensus sequence motifs, or by other methods known to one of ordinary skill in the relevant molecular biological and virology arts.
"Transcriptional unit" refers to a polynucleotide sequence that comprises at least a first structural gene operably linked to at least a first cis-acting promoter sequence and optionally linked operably to one or more other cis-acting nucleic acid sequences necessary for efficient transcription of the structural gene sequences, and at least a first distal regulatory element as may be required for the appropriate tissue-specific and developmental transcription of the structural gene sequence operably positioned under the control of the promoter and/or enhancer elements, as well as any additional cis sequences that are necessary for efficient transcription and translation (e.g., polyadenylation site(s), mRNA stability controlling sequence(s), etc.
As used herein, the term "transformed cell" is intended to mean a host cell whose nucleic acid complement has been altered by the introduction of one or more exogenous polynucleotides into that cell.
As used herein, the term "transformation" is intended to generally describe a process of introducing an exogenous polynucleotide sequence (e.g., a viral vector, a plasmid, or a recombinant DNA or RNA molecule) into a host cell or protoplast in which the exogenous polynucleotide is incorporated into at least a first chromosome or is capable of autonomous replication within the transformed host cell. Transfection, electroporation, and "naked" nucleic acid uptake all represent examples of techniques used to transform a host cell with one or more polynucleotides.
As used herein, the terms "treat," "treating," and "treatment" refer to the administration of one or more compounds (either alone or as contained in one or more pharmaceutical compositions) after the onset of clinical symptoms of a disease state so as to reduce, or eliminate any symptom, aspect or characteristic of the disease state. Such treating need not be absolute to be deemed medically useful. As such, the terms "treatment," "treat," "treated," or "treating" may refer to therapy, or to the amelioration or the reduction, in the extent or severity of disease, of one or more symptom thereof, whether before or after its development afflicts a patient.
As used herein, the term "vector" refers to a nucleic acid molecule capable of transporting another nucleic acid to which it has been linked, e.g., a plasmid. One type of preferred vector is an episome, i.e., a nucleic acid capable of extra-chromosomal replication. Preferred vectors are those capable of autonomous replication and/or expression of nucleic acids to which they are linked. An "rAAV vector" is a recombinant AAV-derived nucleic acid containing at least one terminal repeat (TR) sequence. The use of "virion" is meant to describe a virus particle that contains a nucleic acid and a protein coat (capsid). An "rAAV virion" is a virion that includes nucleic acid sequences and/or proteins derived from a rAAV vector.
As used herein, the term "tropism" refers to the cells and/or tissues of a host which support growth of a particular serotype of AAV. Some serotypes may have a broad tissue tropism and can infect many types of cells and tissues. Other serotypes may infect primarily a single tissue or cell type.
The term "a sequence essentially as set forth in SEQ ID NO:X" means that the sequence substantially corresponds to a portion of SEQ ID NO:X and has relatively few nucleotides (or amino acids in the case of polypeptide sequences) that are not identical to, or a biologically functional equivalent of, the nucleotides (or amino acids) of SEQ ID NO:X. The term "biologically functional equivalent" is well understood in the art, and is further defined in detail herein. Accordingly, sequences that have about 85% to about 90%; or more preferably, about 91%) to about 95%; or even more preferably, about 96% to about 99%; of nucleotides that are identical or functionally equivalent to one or more of the nucleotide sequences provided herein are particularly contemplated to be useful in the practice of the invention.
Suitable standard hybridization conditions for the present invention include, for example, hybridization in 50% formamide, 5x Denhardt's solution, 5x SSC, 25 mM sodium phosphate, 0.1% SDS and 100 μg/ml of denatured salmon sperm DNA at 42°C for 16 h followed by 1 hr sequential washes with O. lx SSC, 0.1% SDS solution at 60°C to remove the desired amount of background signal. Lower stringency hybridization conditions for the present invention include, for example, hybridization in 35% formamide, 5x Denhardt's solution, 5x SSC, 25 mM sodium phosphate, 0.1% SDS and 100 μg/ml denatured salmon sperm DNA or E. coli DNA at 42°C for 16 h followed by sequential washes with 0.8x SSC, 0.1% SDS at 55°C. Those of skill in the art will recognize that conditions can be readily adjusted to obtain the desired level of stringency.
Naturally, the present invention also encompasses nucleic acid segments that are complementary, essentially complementary, and/or substantially complementary to at least one or more of the specific nucleotide sequences specifically set forth herein. Nucleic acid sequences that are "complementary" are those that are capable of base-pairing according to the standard Watson-Crick complementarity rules. As used herein, the term "complementary sequences" means nucleic acid sequences that are substantially complementary, as may be assessed by the same nucleotide comparison set forth above, or as defined as being capable of hybridizing to one or more of the specific nucleic acid segments disclosed herein under relatively stringent conditions such as those described immediately above.
As described above, the probes and primers of the present invention may be of any length. By assigning numeric values to a sequence, for example, the first residue is 1, the second residue is 2, etc., an algorithm defining all probes or primers contained within a given sequence can be proposed:
n to n + y, where n is an integer from 1 to the last number of the sequence and y is the length of the probe or primer minus one, where n + y does not exceed the last number of the sequence. Thus, for a 25-basepair probe or primer (i.e., a "25-mer"), the collection of probes or primers correspond to bases 1 to 25, bases 2 to 26, bases 3 to 27, bases 4 to 28, and so on over the entire length of the sequence. Similarly, for a 35-basepair probe or primer (i.e., a "35-mer), exemplary primer or probe sequence include, without limitation, sequences corresponding to bases 1 to 35, bases 2 to 36, bases 3 to 37, bases 4 to 38, and so on over the entire length of the sequence. Likewise, for 40-mers, such probes or primers may correspond to the nucleotides from the first basepair to bp 40, from the second bp of the sequence to bp 41, from the third bp to bp 42, and so forth, while for 50-mers, such probes or primers may correspond to a nucleotide sequence extending from bp 1 to bp 50, from bp 2 to bp 51, from bp 3 to bp 52, from bp 4 to bp 53, and so forth.
In certain embodiments, it will be advantageous to employ one or more nucleic acid segments of the present invention in combination with an appropriate detectable marker (i.e., a "label,"), such as in the case of employing labeled polynucleotide probes in determining the presence of a given target sequence in a hybridization assay. A wide variety of appropriate indicator compounds and compositions are known in the art for labeling oligonucleotide probes, including, without limitation, fluorescent, radioactive, enzymatic or other ligands, such as avidin/biotin, etc., which are capable of being detected in a suitable assay. In particular embodiments, one may also employ one or more fluorescent labels or an enzyme tag such as urease, alkaline phosphatase or peroxidase, instead of radioactive or other environmentally less-desirable reagents. In the case of enzyme tags, colorimetric, chromogenic, or fluorigenic indicator substrates are known that can be employed to provide a method for detecting the sample that is visible to the human eye, or by analytical methods such as scintigraphy, fluorimetry, spectrophotometry, and the like, to identify specific hybridization with samples containing one or more complementary or substantially complementary nucleic acid sequences. In the case of so-called "multiplexing" assays, where two or more labeled probes are detected either simultaneously or sequentially, it may be desirable to label a first oligonucleotide probe with a first label having a first detection property or parameter (for example, an emission and/or excitation spectral maximum), which also labeled a second oligonucleotide probe with a second label having a second detection property or parameter that is different (i.e., discreet or discernable from the first label. The use of multiplexing assays, particularly in the context of genetic amplification/detection protocols are well-known to those of ordinary skill in the molecular genetic arts.
In accordance with the present invention, polynucleotides, nucleic acid segments, nucleic acid sequences, and the like, include, but are not limited to, DNAs (including and not limited to genomic or extragenomic DNAs), genes, peptide nucleic acids (PNAs) RNAs (including, but not limited to, rRNAs, mRNAs and tRNAs), nucleosides, and suitable nucleic acid segments either obtained from natural sources, chemically synthesized, modified, or otherwise prepared or synthesized in whole or in part by the hand of man.
EXAMPLES
The following examples are included to demonstrate illustrative embodiments of the invention. It should be appreciated by those of ordinary skill in the art that the techniques disclosed in these examples represent techniques discovered to function well in the practice of the invention, and thus can be considered to constitute preferred modes for its practice. However, those of ordinary skill in the art should, in light of the present disclosure appreciate that many changes can be made in the specific embodiments which are disclosed and still obtain a like or similar result without departing from the spirit and scope of the invention.
EXAMPLE 1
Step 1 : Sub-libraries assembly.
Using pITR3-R3C3-AatII as a template, the following ten PCR reactions were conducted:
A3CL-A (VRs-I, V, VI) Primers PCR fragment size
1. A3CL-F + A3CL-A1R (before VR-I) 86 bp
2. A3CL-A1F + A3CL-A2R (VR-I to most of VR-V) 747 bp
3. A3CL-A2F + A3CL-A3R (part of VR-V to VR-VI ) 136 bp
4. A3CL-A3F + A3CL-R (after VR-VI) 281 bp
A3CL-B (VR-IV) :
Primers fragment size
5. A3CL-F + A3CL-B1R (before VR-IV) 647 bp
6. A3CL-B1F + A3CL-R (VR-IV to end) 556 bp
A3CL-C (VR-VII) :
Primers PCR fragment
7. A3CL-F + A3CL-C1R (before VR-VII) 935 bp
8. A3CL-C1F + A3CL-R (VR-VII to end) 266 bp
A3CL-D (VR-VII I) :
Primers fragment size
9. A3CL-F + A3CL-D1R (before VR-VII I) 1055 bp
10. A3CL-D1F + A3CL-R (VR-VIII to end) 147 bp
The respective PCR fragments were eluted from the agarose gel, mixed at equimolar ratios as indicated above for sub-libraries A, B, C, and D, and subjected to 15 cycles of overlap extension (OE) without primers, followed by 20 cycles of PCR using A3CL-F forward and A3CL-R reverse primers. The resulting fragments of 1140 bp for each of the A (I+V+VI), B (IV), C (VII), or D (VIII) sub-libraries were purified on agarose gel and eluted in small volume H20. Using isothermal DNA assembly protocol, the respective fragments were individually sub-cloned into gel-purified pTR3-R3C3-AatII digested with Aatll+Apal. Four plasmid libraries A, B, C, and D, incorporating the respective VRs were derived. The estimated plasmid libraries' complexities were the following: A - 4.4xl07; B - 1.7xl07; C - lxlO8; D - lxlO8.
Step 2: Pre-selecting structurally compatible parent viral libraries.
Using plasmid libraries from Step 1, viral sub-libraries A, B, C, and D were packaged, AAV virus from each preparation was purified using iodixanol density gradients, and the viral DNAs were isolated. Next, using viral DNAs as templates, the following PCR reactions were conducted:
1. VR-I, primers A3CL-F + VR-I_IV-R, template A, size 644 bp.
2. VR-IV, primers VR-I_IV-F + VR-IV_V-R, template B, size 145 bp.
3. VR-V+VI, primers VR-IV_V-F+ VR-VI_VII-R, template A, size 194 bp.
4. VR-VII, primers VR-VI_VR-VII-F + A3CL-R, template C, size 274 bp.
5. VR-VIII, primers A3CL-F and A3CL-R, template D, size 1140 bp.
The respective PCR fragments were gel-purified and used as the templates in the OE/PCR to derive two PCR fragments, each of 1140 bp: A+B+C (VR-I, IV, V, VI, VII) and D (VR-VIII).
Step 3 : Packaging master libraries.
Using isothermal DNA assembly protocol, the respective fragments were individually sub-cloned into gel -purified pTR3-R3C3-AatII digested with Aatll+Apal. The estimated plasmid library A+B+C complexity was 2.5xl07, plasmid library D complexity was 4xl07. Using these plasmid libraries, two final master viral libraries were packaged: ABC, with the titer of 5.7xl012 drp/ml, and D, with the titer of 8.7xl012 drp/ml. The assembly flowchart is shown in Figure 5.
Table 1. Theoretical (calculated) complexities of A3CL for individual VRs and combinations of VRs. The VRs and VRs combinations constructed as sub-libraries are shown in bold font.
VR Complexity
I 72
IV 2. lxlO6
V 27 , 648
VI 144
VII 4xl07
VIII 5. 44xl08
I+V 1. 99xl06
I+VI 1. 04xl04
I+V+VI 2. 87xl08
V+VI 3. 98xl06
I+IV+V+VI+VII 2. 37xl022
I+IV+V+VI+VI I+VI II 1. 29xl031 Table 2. Theoretical (calculated) complexities of constructed sub-libraries A, B, C, and D.
Sub-library VRs Complexity
A3CL-A I-V-VI 2.9xl08
A3CL-B IV 2xl06
A3CL-C VII 4xl07
A3CL-D VIII 5.4x10s
Table 3. Synthetic oligonucleotides used to assemble the AAV3B capsid library
Name Sequence
A3CL-F GGCTGGGCGACAGAGTCATC
A3CL-A1R GCTGGAGATTTGCTTGTAGAGATG
A3CL-A1F CATCTCTACAAGCAAATCTCCAGCVVMDCAGGAGCTASCAACGACAACCACTACTTTGGC A3CL-A2R CCAAGGAAASTYACTGTTGTTGTTSYSGBYGKVGRYTKTTGAAAGTCTCTGTTGCC A3CL-A2 F AACAACAACAGTRASTTTCCTTGGMCAGCGGCCAGCAMATATCATCTCAATG A3CL-A3R GATTGCCGTGCATAGGGAAAAATYTSYCSKYATCGTCCYYGTGACTGGCCATAGCTGG A3CL-A3F ATTTTTCCCTATGCACGGCAATC
A3CL-R CATCCGTGTGAGGAATCTTTGC
A3CL-B1R TTGCGTTCTGTTCAGGTAGTACAGA
A3CL-B1F CTGTACTACCTGAACAGAACGCAARGCAMCVCNRGCGGAACARCCRVCMHSMRSVVSCTG
VNGTTTAGCCAGGCTGGGCC
A3CL-C1R TTTGCCAAATATTAGATTGCC
A3CL-C1F CGGCAATCTAATATTTGGCAAASAARRCRSCRVSRVARVCRATRYCGMSDWCGRSVRS
GTAATGATTACGGATGAAGAAG
A3CL-D1R CTGCAAGTTATTTGCCACAGTTC
A3CL-D1F GAACTGTGGCAAATAACTTGCAGRVSVVSMRSRVCVVSCCCACGDHTVVSRNSGTC
VMSCATCAGGGGGCCTTACCTG VR-I_IV-F CAGTATCTGTACTACCTGAACAGAACGC
VR-I_IV-R GCGTTCTGTTCAGGTAGTACAGATACTG
VR-IV_V-F CCTGGGCCCTGCTACCGGCAACAGAG
VR-IV_V-R CTCTGTTGCCGGTAGCAGGGCCCAGG
VR-VI_VII-F CCCTATGCACGGCAATCTAATATTTGGC
VR-VI VII-R GCCAAATATTAGATTGCCGTGCATAGGG
NGS sequencing. Number of sequences processed: 1817050
Number of distinct sequences (complexity): 1708473 (0.94)
Copy number distribution:
Copy number Number of sequences
1 1603700
2 101430
3 3257
4 83
5 2
377 1
Table 4. Examples of the most representative variants within VRs IV, V, VI, and VII from the master viral library ABC as deduced from the NGS sequencing (the dots in each of sequences 1-86 below represent amino acid residues that are identical to those listed in wild type as shown below).
450 491 528 546 cn type GTTSGTTNQSRLL KTANDNNNSNFPWTAASK KDDEEK EGTTASNAELDN
1 377 0 0
2 .N. G. .. SP.. R . IYDR.. .T G. . TGR Q.. GEG. V. VGK 5 0 0
3 S .... .. G . RK A . AYGH .. .. D. .P. .T G. .. DR QDSGENDVAIGR 5 0 0
4 ..A.. ..AN.N K .. YS... G. . DDR ... DGA. V. I . R 4 0 0
5 .. P.. . AAHKT E .. SAE .. G. .AGR . DAEGGD . AIGG 4 0 0
6 S .AG. .AT.KA T . VHAH .. E. .TGR QDA. R.. VAFEE 4 0 0
7 .NP.. .. GLRG T T.D.E.. .P. .AG. Q .. DGN . IAFGE 4 0 0
8 .N... .. SKRP M T...E.. .T E. .N. R . DAKGTDT . F . R 4 0 0
9 SNA.. .. GIHQ K TAPDR.. .. E. E. . NGR QNGATADT . VER 4 0 0
10 SN. G. .. AMRE E . AP .... .. K. .T E. .TG. . S .AETDV. DGR 4 0 0
11 SNA.. .AGLQ. K . IPDQ.. .NG. QSGG. ADIDNG. 4 0 0
12 .NP.. ..APH. TIH. G.. .P. .T . DGR QDGGT.. IDI.G 4 0 0
13 .. P.. .. DLRE A . IP.... .T E. .. DR 4 0 0
14 SNP.. .A. PRT M . IDAH.. .. E. .P. .T E. .NG. QSS.TGDV.D.D 4 0 0
15 .NA.. .. DTK. T . ASGG.. .T . DD. .. SNRDD..V. R 4 0 0
16 .N. G. .. DIR. R .. HSE .. .. E. .T .N. R QD. RETDVAI . R 4 0 0
17 .NA.. . AGMRE M .A..H.. E. .. D. . SGS . DDVAIGR 4 0 0
18 SN. G. . ATPKQ Q . ASAH .. .. E. ... R .. S . RNDIANEH 4 0 0
19 SN. G. .A. IKE T .. S .... .T R. .ND. QSASKNDI . YEQ 4 0 0
20 SNAG. .. SNRE R T. SSQ.. .T R. . DDR QDAGGNDV. VGD 4 0 0
21 SN... .ATT. A K .. YGH .. G. .T.. Q. GS .N. V. VES 4 0 0 SNAG.. AATN . . IYDR.. .T R... D. .. GEKG. VDI . R 4 0 0
S.P.. ..ATKG T TAHTG.. G. . DG. .. S.. TDVAIGS 4 0 0
.N. G. .. DLR. M T.D.H.. ..E...P. .T G. . KGR . NGAKNDIAFEG 4 0 0
S .... .. TLKA Q .IP.R.. .T G. .. DR . NSKGA. T . I . E 4 0 0
.. DPKD V T.HG... .. D .T . DD. . D.A. D. V. FGR 4 0 0
S .AG. .. TIKD V . VPD... .. K E. . D. R QDSG.T.V. FGR 4 0 0
... G. .. TMRK G . VYGG .. .T E. .A. R QSSGRNDV.YGD 4 0 0
.N. G. .ASTR. T . IPDQ.. P. E. .. GR QSAEKGDI . YGR 4 0 0
.N... .. ATHT A . IHSR.. ..D...P. E. .AG. Q.A.. G. IDVEQ 4 0 0
SNPG. .. SIRG Q TIP.R.. P. .T R. .TD. Q.GG.G.TDF.H 4 0 0
S .... . AAPRG V TVYGH .. .. E .. GR ..AG... VAIEE 4 0 0
.NA.. . ATKQG M . VP . Q .. .. D . DDR QSSDKN... D. S 4 0 0
SNAG. .ATT . Q R TAPAE .. P. .T R. .ADR . SGRGD. VDFEK 4 0 0
SN. G. .AGIRA Q . VDTG .. .. D .T E. .T.. . NSARND . DIGR 4 0 0
.NA.. . AA. NG R .IP.E.. .. K .T .. G. . SSSGDD.. FGG 4 0 0
SN... . AGPQQ R .. HAQ .. E. .TG. ..AR.NDIAF.Q 4 0 0
S.P.. .. SMRT E .APAR.. .. E .T R. .AG. Q.SRENDT.F.G 4 0 0
S .AG. .. ALKG K TI . DH.. ..E...P. .T R. . K.. . DS . GA. IAD. R 4 0 0
S.P.. . ASTRT M .. H . H .. .. E .T .. D. ...E.T.VAIGG 4 0 0
. NPG . ... NQA R . IHGQ.. .. D .T R. .ND. .. SARGDVAYEK 4 0 0
SNA.. .. DTRE V TI . D... .. E .T R. .TD. Q. SAGADV. VEK 4 0 0
SNPG. ... LRE R TIHTE.. .. E .T R. . KDR Q.GGGT.V.IGS 4 0 0
... G. .A.NNT . I . SG.. .T . KGR .. AEKNDTAVG. 4 0 0
.. DKQQ M .. H . G .. .. D E. .TG. QSAEGN. VAY. G 4 0 0
..AG. .ATL.T V . I SAG .. .. D G. .NG. QNS ... DVAI . G 4 0 0
SN... . AGLRT T .ADA... ..D...P. .T G. .NG. . DASGN. V. DGR 4 0 0
SNA.. .ATP.T R .. DTH.. ..E...P. R. . NDR ..ARG.. IDVGD 4 0 0
S .A.. . ASLRA M . VP . R .. .T G. .ND. .NAR.. D.. V. R 4 0 0
..A.. . ATTKG . ISTQ.. E. .AD. Q. GETD. VDVGD 4 0 0
..A.. . AALKQ A .ADS ... .T E. .ADR Q.GETG.I.Y.G 4 0 0
.NA.. .ATT . N M .ADDR.. .T .. DR Q.AKR. DTAVEE 4 0 0
..AG. . A.MKD R T.. SE.. .. D .T E. . KD. .. ANGGDVAIGQ 4 0 0
S . PG. ..TIRD K TVST ... ..D...P. .T . DDR .. SGRN. VAVEE 4 0 0
S.P.. .A. INT R ..P.R.. .T E. .. GR QSA. KDDVDIGG 4 0 0
.N. G. . AGLQK M .. HGG .. P. .T . DG. QSSRGNDVAV. D 4 0 0
.N... .. TPRT A . IPSH.. .. E Q. SNG.. I . FGS 4 0 0
SNAG. . AGLRQ T . APAE .. .. D .T E. .AG. .. GGGA. IAVEE 4 0 0
..A.. .AAK.T V . ISTR.. .T E. ... SKNDV. VE. 4 0 0 60 .. TTR. M . IYGG.. .T E..AGR Q..ATA. V. VES 4 0 0
61 ..AG. . AGMRE A TIYTG.. .. GR .. SSTGD . DVGR 4 0 0
62 S .. G. .A. PKE R TA.. H.. .. E. .T E. .. D. Q . AGE .. VAI . G 4 0 0
63 .. GT . T R .. DTG.. G. ... R ..AGTAD.AV. G 4 0 0
64 .NAG. ... KRD TAYTR.. .. D. .T . D.. Q.. GKTD. DNGG 4 0 0
65 .NA.. .. DMKH T . ISDR.. .P. .T R. .N. R QS . RGG... I . G 4 0 0
66 SN. G. . ADLRD TIPTQ.. .. E. .NDR QSAK.NDV. V. R 4 0 0
67 S .A.. .. ATQQ V T.DSQ.. .. E. .T R. .NDR . NAEGG . V. IGQ 4 0 0
68 .NAG. .. ANKT M . I . AH .. .. E. .T R. . DG. QDSS.D.I.YGK 4 0 0
69 .NAG. .. GTKE R TI .. E.. .T . D.. . DAKRN . VDY . G 4 0 0
70 SNP.. .. GK. S K .. S . E.. .NG. . DSR. GD . DFEK 4 0 0
71 .N... . ASIRQ Q .. PDG.. .. K. R. . NGR .. S . EG. I . IEG 4 0 0
72 .N. G. ..TL.A G TAHTQ .. R. .ND. QSS . GGDTAF. G 4 0 0
73 SNP.. .. TTQ . Q .. D.... .. D. .P. .T R. . TDR .. S . GGD.. IER 4 0 0
74 SNA.. .. TMRK G TISSG.. .P. G. .N.. QDSSENDVADER 4 0 0
75 S .AG. . ATMQ . M T.DTG.. .T R. .N. R Q. GEGGDI . D. R 4 0 0
76 ..AG. . ATTRD Q T.DDH.. .. D. .NGR Q.GRGA.TAYEG 4 0 0
77 ... G. .AAM.A R T.DDG.. .. K. .P. . DGGT ... AIGD 4 0 0
78 .N... .. TNRE M . IP.... .T R. .. D. . D. GRADV. VGR 4 0 0
79 SNAG. . ADKQD V TAHSE.. E. . DDR Q.AAGGDI . VGS 4 0 0
80 .NA.. . AATHE T.HDH.. .. D. R. .A. R .. GAK. DVDFGS 4 0 0
81 SNA.. . ADTRH M T. PGE.. .. D. .P. .T G. .TG. Q. SATTDI . YGE 4 0 0
82 SN... ..A.. K Q . IH. R.. .. D. .T R. . DDR Q.AEG. DVAVGD 4 0 0
83 .NP.. .AD. RA Q . IPTG.. .. D. .T R. .T.. Q.. GG. DI . IGG 4 0 0
84 SNA.. . AGLNA K . AYTH .. .. D. .T G. . D. R . NAK . G .. AI . G 4 0 0
85 .NP.. ...LQ. M . IDDQ.. .P. .T .. D. . SGGTADVAV. K 4 0 0
86 .ASIQ. Q .. YA... .. E. .T .. D. .SAAG.DT.V.G 4 0 0
Calculated plasmid library complexity based on colony count (2.5x107) and NGS sequencing (0.94 of unique sequences) is 2.35x107. WT AAV3 contamination is 0.02%.
EXAMPLE 2 Q5 PGR:
50 μΙ: 10 μΙ 5xB Q5
0.4 μΙ 25 mM dNTPs
2.5 μΙ F
2.5 μΙ R
1 μΙ (1 ng) plTR3-R3C3-Aat!i 0.5 μΙ Q5 Po!
H20 up to 50 μ!
98°C 30 sec
98°C 10 sec
65°C 20 sec 30 cycles
72°C 30 sec
72°C 2 min
See Figure 6.
Table 5
Figure imgf000044_0001
Table 6: OE Q5
PCR
Figure imgf000045_0002
1 , Assays A, B, C, and O are assembled without primers, substituting H2Q for the primers' volumes (5 μΐ) and subjected to the following overlap extension:
&8°C 30 sec
98°C 10 sec
65°C 20 sec 5 cyoies
72°C 60 sec
72°C 2 min
2, 40 μΐ each A, 8, C, and 0 ffom Step 1 transferred to 10 μ! containing:
X5
2.5 ^ A3CL-F 12.5
2.5 ^ A3CL-R 12.5
2 μ! 5xB Q5 10
0.08 μ! dNTPs 0.4
0.1 μ! Q5 0.5
2.82 μί H20 14,1
Assays are subjected to the following PCRs:
98°C 30 sec
§8°C 10 sec
59°C 20 sec 20 cycles
72°C 60 sec
72°C 2 min
See Figure 7: Eluted in 50μ1 each A, B, C, or D; pTR3-R3C3-AatII/AatII+ApaI eluted in 75 μΐ.
Figure imgf000045_0001
(DA
40 μΙ assay: 20≠ 2xGibsen Master Mix (NEE) + A 8
2.1 3.7 14.2 A
1.S 3,7 Ι4.δ B
2.1 3,7 14.2 C
1 ,7 3.7 14 6 D Large-scale !DA for the Loop A
300 μΙ assay; 150 μΙ 2xGtbsort aster Mix
27.6 μΙ pH'R3-R3C3 Aatli Aafli-Apai cut (15 ng)
15.8 μΙ A (0.75 }.g)
106.6 μΐ HjO
Incubated 2 h. Βϋ°€, Zymo-purified, eiuted in 100 μΙ H2Os combined with 47.5 μ! of A from the pilot IDA above. Total™ 1.7 μ of vector p!asmid DNA.
Lucigen eompetent ceils were prepared from 4 L LB, resuspended in 8.5 mi HaO final volume. The cell density (10 μΙ in 3 mi H20} was A - 0.79.
Combined DNA (147.5 μΙ) was mixed with the whole volume of competent ceils and aliquoted (385 pj/a!iquot, ~ 10 ng plasmid DNA 50 μΙ competent ceils) into eiectroporation cuvettes {total of -20, with outside tali electrodes) and zapped at 2.9 KV.
Cells were transferred into 1 L LB, incubated shaking at 37X for 1 h. GarbenSciin was added up 00 g/iTi!, cell were grown at 30°C, o/n.
Total complexity from the iarge-scale SPA/transformation is 4,4x107 clones.
Repeat IDA for the Loop C
100 μί assay: 50 μί 2xGibson Master Mix
9.25 μί p!TR3~R3C3-Aat!i AatlS-Apa! cut {0.5 VQ)
Figure imgf000046_0001
Zymo, 50 μ! HjO.
Competent ceils were prepared from 4 L L8 (grown to A6¾~0,6} and resuspended in a final volume 8 ml HaO. The cell density (10 μ! in 3 mi H.O) was Asso= .46.
180 ng vector with fragment B from the pilot IDA were electroporated with 1 mi of comp. ceils, whereas 0.68 μg with fragment C ~ with 3 ml of cells.
After eiectroporation the complexity of 8 was ~1.7x 07 (-5 times over theoretical complexity), while C - 1x10a (~2,5 times over theoretical complexity).
Table 8
Figure imgf000047_0001
Q5 PCR of viral DNA
Conditions, as above, except: 50 ng viral DNA/50 μΐ assay, 20 PCR cycles 5 μΙ out of 50
1. Loop I, primers A3CL-F + VR-IJV-R, template A, size 644 bp
2. Loop IV, primers VR-IJV-F + VR-IV_V-R, template B, size 145 bp
3. Loops V+VI, primers VR-IV_V-F+ VR-VI_Vli-R, tempi. A, size 194 bp
4. Loop VII, primers VR-VM/R-VII-F + A3CL-R, template C, size 274 bp
Remaining 45 μΙ were purified using preparative gel, all four gel cutouts were pooled in one tube and purified using one column, final volume 50 μΙ H20.
See Figure 8. Overlap extension
Full-length fragment was assombSe without primers, substituBog H20 for the primers' volumes (5 μί) and subjected to the following overlap extension;
50 μΙ: 10 μΙ 5xB Q5
0.4 μΙ 25 mM dNTPs
25 μ! (out of 50 μΙ) individual overlap 4 fragments mix <p,1 )
0.5 μΙ Q5 Poi
14,1 μ! H20
98°C 30 sec
9S°C 10 sec
65°C 20 sec 15 cycles
72°C 60 sec
?2°C 2 min
After primer-less extension, the assay was split into 2x25 μΙ assays supplemented with A3CL-F, and A3CL-R primers, DNTPs, and fresh Q5, total volume 50 μΙ each.
Assays are subjected to the following PCRs:
98°C 30 sec
98°C 10 sec
59°C 20 sec 20 cycles
72°C 60 sec
72°C 2 min
ABC fragment was eluted in 50 μΙ, concentration 60 ng/ μΙ
(0,085 pmoles/μΙ).
D fragment was eluted in 50 μί, concentration 46 ng/ μΙ (0.065
pmoles/μΙ).
See Figure 9.
IDA using HE Builder Master Mix
Totai volume - 200 μΙ, plasmid 1.5 μ§ (o.348 pmoles), insert - 0.5 9 (0.7 pmoies), total DNA amount -1 ρΓηο!β/2«όμ! assay.
Reaction 60 min @ 50°C, Lucigen eiectrocompetent E.coli ceiis, 8 mi, final density 0.8 A550. Library's complexity 2.5x10'.
What is claimed is:

Claims

1. A non-naturally occurring nucleic acid comprising:
(a) a first nucleotide sequence encoding at least one AAV Rep protein from serotype
3;
(b) a second nucleotide sequence encoding at least one AAV Cap protein, wherein the second nucleotide sequence differs from wildtype serotype 3 at least at one nucleotide position; and
(c) a first AAV terminal repeat from serotype 3 and a second AAV terminal repeat from serotype 3,
wherein the first and second nucleotide sequences are interposed between the first and the second AAV terminal repeat.
2. The non-naturally occurring nucleic acid of claim 1, wherein the nucleic acid further comprises a third nucleotide sequence encoding at least one molecule providing helper function.
3. The non-naturally occurring nucleic acid of claim 2, wherein the third nucleotide sequence encoding at least one molecule providing helper function is a polynucleotide from a virus selected from the group consisting of: adenovirus and herpesvirus.
4. The non-naturally occurring nucleic acid of any one of claims 1-3, wherein the second nucleotide sequence comprises: TGCCCACTTACAACAACCATCTCTACAAGCAAATC
TCCAGCVVMDCAGGAGCTASCAACGACAACCACTACTTTGGCTACAGCACCCCTTGGGGG TATTTTGACTTTAACAGATTCCACTGCCACTTCTCACCACGTGACTGGCAGCGACTCATT AACAACAACTGGGGATTCCGGCCCAAGAAACTCAGCTTCAAGCTCTTCAACATCCAAGTT AGAGGGGTCACGCAGAACGATGGCACGACGACTATTGCCAATAACCTTACCAGCACGGTT CAAGTGTTTACGGACTCGGAGTATCAGCTCCCGTACGTGCTCGGGTCGGCGCACCAAGGC TGTCTCCCGCCGTTTCCAGCGGACGTCTTCATGGTCCCTCAGTATGGATACCTCACCCTG AACAACGGAAGTCAAGCGGTGGGACGCTCATCCTTTTACTGCCTGGAGTACTTCCCTTCG CAGATGCTAAGGACTGGAAATAACTTCCAATTCAGCTATACCTTCGAGGATGTACCTTTT CACAGCAGCTACGCTCACAGCCAGAGTTTGGATCGCTTGATGAATCCTCTTATTGATCAG TATCTGTACTACCTGAACAGAACGCAARGCAMCVCNRGCGGAACARCCRVCMHSMRSVVS CTGVNGTTTAGCCAGGCTGGGCCTCAGTCTATGTCTTTGCAGGCCAGAAATTGGCTACCT GGGCCCTGCTACCGGCAACAGAGACTTTCAAMARYCBMCRVCSRSAACAACAACAGTRAS TTTCCTTGGMCAGCGGCCAGCAMATATCATCTCAATGGCCGCGACTCGCTGGTGAATCCA GGACCAGCTATGGCCAGTCACRRGGACGATRMSGRSARATTTTTCCCTATGCACGGCAAT CTAATATTTGGCAAASAARRCRSCRVSRVARVCRA RYCGMSDWCGRSVRSGTAATGATT ACGGATGAAGAAGAGATTCGTACCACCAATCCTGTGGCAACAGAGCAGTATGGAACTGTG GCAAATAACTTGCAGRVSVVSMRSRVCVVSCCCACGDHTVVSRNSGTCVMSCATCAGGGG GCCTTACCTGGCATGGTGTGGCAAGATCG .
5. The non-naturally occurring nucleic acid of any one of claims 1-4, wherein the AAV Cap protein differs from wildtype serotype 3 at least at one amino acid position.
6. The non-naturally occurring nucleic acid of claim 5, wherein the at least one differing amino acid position is in a variable region (VR).
7. The non-naturally occurring nucleic acid of claim 6, wherein the VR is selected from the group consisting of VR-I, VR-IV, VR-V, VR-VI, VR-VII, VR-VIII and combinations thereof.
8. The non-naturally occurring nucleic acid of claim 7, wherein VR-I comprises amino acid sequence X1X2GAX3.
9. The non-naturally occurring nucleic acid of claim 8, wherein Xi is independently selected from the group consisting of Q, N, K, T, S, R, H, P, D, E, A and G; X2 is independently selected from the group consisting of S, T and A; and X3 is independently selected from the group consisting of S and T.
10. The non-naturally occurring nucleic acid of claim 7, wherein VR-IV comprises amino acid sequence X4X5X6X7GTX8X9X10X11X12LX13.
11. The non-naturally occurring nucleic acid of claim 10, wherein X4 is independently selected from the group consisting of G and S; X5 is independently selected from the group consisting of T and N; X6 is independently selected from the group consisting of T, P and A; X7 is independently selected from the group consisting of S and G; Xs is independently selected from the group consisting of T and A; X9 is independently selected from the group consisting of N, T, S, D, A and G; Xio is independently selected from the group consisting of Q, H, P, L, K, N, T, M and I; X11 is independently selected from the group consisting of S, Q, H, R, K and N; X12 is independently selected from the group consisting of R, K, N, T, S, Q, H, P, E, D, A and G; and Xi3 is independently selected from the group consisting of L, K, T, R, M, Q, P, E, A, G and V.
12. The non-naturally occurring nucleic acid of claim 7, wherein VR-V comprises amino acid sequence X14X15X16X17X18 NNSX19FPWX20AASX21.
13. The non-naturally occurring nucleic acid of claim 12, wherein X14 is independently selected from the group consisting of K and T; X15 is independently selected from the group consisting of T, I, A and V; Xi6 is independently selected from the group consisting of A, P, H, D, S and Y; X17 is independently selected from the group consisting of N, T, S, D, A and G; Xis is independently selected from the group consisting of D, E, G, Q, H and R; X19 is independently selected from the group consisting of N, K, E and D; X20 is independently selected from the group consisting of T and P; and X21 is independently selected from the group consisting of K and T.
14. The non-naturally occurring nucleic acid of claim 7, wherein VR-VI comprises amino acid sequence X22DDX23X24X25.
15. The non-naturally occurring nucleic acid of claim 14, wherein X22 is independently selected from the group consisting of K, R, E and G; X23 is independently selected from the group consisting of E, T, K, N, A and D; X24 is independently selected from the group consisting of E, D and G; and X25 is independently selected from the group consisting of K and R.
16. The non-naturally occurring nucleic acid of claim 7, wherein VR-VII comprises amino acid sequence X26X27X28X29X30X31X32X33X34X35X36X37.
17. The non-naturally occurring nucleic acid of claim 16, wherein X26 is independently selected from the group consisting of E and Q; X27 is independently selected from the group consisting of G, N, S and D; X28 is independently selected from the group consisting of T, S, G and A; X29 is independently selected from the group consisting of T, K, N, R, S, E, D, A and G; X30 is independently selected from the group consisting of A, K, T, R, E and G; X31 is independently selected from the group consisting of S, N, T, D, A and G; X32 is independently selected from the group consisting of N and D; X33 is independently selected from the group consisting of A, T, I and V; X34 is independently selected from the group consisting of E, A and D; X35 is independently selected from the group consisting of L, N, I, D, V, Y and F; X36 is independently selected from the group consisting of D, E and G; and X37 is independently selected from the group consisting of N, K, R, S, Q, H, E, D and G.
18. The non-naturally occurring nucleic acid of claim 7, wherein VR-VIII comprises amino acid sequence X38X39X40X41X42PTX43X44X45VX46.
19. The non-naturally occurring nucleic acid of claim 18, wherein X38 is independently selected from the group consisting of S, K, N, T, R, E, D, A and G; X39 is independently selected from the group consisting of S, K, N, T, R, Q, H, P, E, D, A and G; X40 is independently selected from the group consisting of N, Q, H, R, K and S; X41 is independently selected from the group consisting of T, N, S, D, A and G; X42 is independently selected from the group consisting of A, K, N, T, R, S, Q, H, P, E, D and G; X43 is independently selected from the group consisting of T, N, I, D, A, V, Y, S and F; X44 is independently selected from the group consisting of G, K, N, T, R, S, Q, H, P, E, D and A; X45 is independently selected from the group consisting of T, K, N, R, S, M, I, E, D, A, G, and V; and X46 is independently selected from the group consisting of N, T, K, P, Q, H, A, E and D.
20. The non-naturally occurring nucleic acid of claim 5, wherein the AAV Cap protein comprises: MAADGYLPDWLEDNLSEGI REWWALKPGVPQPKA QQHQDNRRGLVLPGYKYLGPGNGLD
KGEPVNEADAAALEHDKAYDQQLKAGDNPYLKYNHADAE FQERLQEDT S FGGNLGRAVFQ AKKRILEPLGLVEEAAKTAPGKKGAVDQSPQE PDSS SGVGKSGKQPARKRLNFGQTGDSE SVPDPQPLGE PPAAPT SLGSNTMASGGGAPMADNNEGADGVGNS SGNWHCDSQWLGDRVI TT STRTWALPTYNNHLYKQI SSXXGAXNDNHY FGYSTPWGY FDFNRFHCH FS PRDWQRLI NNNWGFRPKKLS FKLFNIQVRGVTQNDGTTT IA NLTSTVQVFTDSEYQLPYVLGSAHQG CLPP FPADVFMVPQYGYLTLNNGSQAVGRS S FYCLEY FPSQMLRTGN FQ FSY FEDVPF HSSYAHSQSLDRLMNPLIDQYLYYLNRTQXXXXGTXXXXXLXFSQAGPQSMSLQARNWLP GPCYRQQRLSXXXXXNNNSXFPWXAASXYHLNGRDSLVNPGPAMASHXDDXXXFFPMHGN LIFGKXXXXXXXXXXXXVMITDEEEIRTTNPVATEQYGTVA NLQXXXXXPTXXXVXHQG ALPGMVWQDRDVYLQGPIWA .
21. The non-naturally occurring nucleic acid of claim 5, wherein the AAV Cap protein comprises one of sequences 2-86 as listed in Table 4.
22. A vector library comprising at least a first vector and a second vector, each vector comprising a nucleic acid comprising:
(a) a first nucleotide sequence encoding at least one AAV Rep protein from serotype 3;
(b) a second nucleotide sequence encoding at least one AAV Cap protein, wherein the second nucleotide sequence differs from wildtype serotype 3 at least at one nucleotide position; and
(c) a first AAV terminal repeat from serotype 3 and a second AAV terminal repeat from serotype 3,
wherein the first and second nucleotide sequences are interposed between the first and the second AAV terminal repeat, and the second vector differs from the first vector by at least one nucleotide.
23. The vector library of claim 22, wherein the vector library is incorporated into at least one host cell.
24. The vector library of claim 23, wherein the at least one host cell is a HEK293 embryonic kidney cell.
25. The vector library of any one of claims 22-24, wherein the nucleic acid further comprises a third nucleotide sequence encoding at least one molecule providing helper function.
26. The vector library of claim 25, wherein the third nucleotide sequence encoding at least one molecule providing helper function is a polynucleotide from a virus selected from the group consisting of: adenovirus and herpesvirus.
27. The vector library of any one of claims 22-26, wherein the second nucleotide sequence
COmpri ses : GC CC AC TACAACAAC CA C C AC AAGC AAA c
TCCAGCVVMDCAGGAGCTASCAACGACAACCACTACTTTGGCTACAGCACCCCTTGGGGG TATTTTGACTTTAACAGATTCCACTGCCACTTCTCACCACGTGACTGGCAGCGACTCATT AACAACAACTGGGGATTCCGGCCCAAGAAACTCAGCTTCAAGCTCTTCAACATCCAAGTT AGAGGGGTCACGCAGAACGATGGCACGACGACTATTGCCAATAACCTTACCAGCACGGTT CAAGTGTTTACGGACTCGGAGTATCAGCTCCCGTACGTGCTCGGGTCGGCGCACCAAGGC TGTCTCCCGCCGTTTCCAGCGGACGTCTTCATGGTCCCTCAGTATGGATACCTCACCCTG AACAACGGAAGTCAAGCGGTGGGACGCTCATCCTTTTACTGCCTGGAGTACTTCCCTTCG CAGATGCTAAGGACTGGAAATAACTTCCAATTCAGCTATACCTTCGAGGATGTACCTTTT CACAGCAGCTACGCTCACAGCCAGAGTTTGGATCGCTTGATGAATCCTCTTATTGATCAG TATCTGTACTACCTGAACAGAACGCAARGCAMCVCNRGCGGAACARCCRVCMHSMRSVVS CTGVNGTTTAGCCAGGCTGGGCCTCAGTCTATGTCTTTGCAGGCCAGAAATTGGCTACCT GGGCCCTGCTACCGGCAACAGAGACTTTCAAMARYCBMCRVCSRSAACAACAACAGTRAS TTTCCTTGGMCAGCGGCCAGCAMATATCATCTCAATGGCCGCGACTCGCTGGTGAATCCA GGACCAGCTATGGCCAGTCACRRGGACGATRMSGRSARATTTTTCCCTATGCACGGCAAT CTAATATTTGGCAAASAARRCRSCRVSRVARVCRATRYCGMSDWCGRSVRSGTAATGATT ACGGATGAAGAAGAGATTCGTACCACCAATCCTGTGGCAACAGAGCAGTATGGAACTGTG GCAAATAACTTGCAGRVSVVSMRSRVCVVSCCCACGDHTVVSRNSGTCVMSCATCAGGGG GCCTTACCTGGCATGGTGTGGCAAGATCG .
28. The vector library of any one of claims 22-26, wherein the AAV Cap protein differs from wildtype serotype 3 at least at one amino acid position.
29. The vector library of claim 28, wherein the at least one differing amino acid position is in a variable region (VR).
30. The vector library of claim 29, wherein the VR is selected from the group consisting of VR-I, VR-IV, VR-V, VR-VI, VR-VII, VR-VIII and combinations thereof.
31. The vector library of claim 30, wherein VR-I comprises amino acid sequence
32. The vector library of claim 31, wherein Xi is independently selected from the group consisting of Q, N, K, T, S, R, H, P, D, E, A and G; X2 is independently selected from the group consisting of S, T and A; and X3 is independently selected from the group consisting of S and T.
33. The vector library of claim 30, wherein VR-IV comprises amino acid sequence
X4X5X6X7GTX8X9X10X11X12LX13.
34. The vector library of claim 33, wherein X4 is independently selected from the group consisting of G and S; X5 is independently selected from the group consisting of T and N; X6 is independently selected from the group consisting of T, P and A; X7 is independently selected from the group consisting of S and G; Xs is independently selected from the group consisting of T and A; X9 is independently selected from the group consisting of N, T, S, D, A and G; X10 is independently selected from the group consisting of Q, H, P, L, K, N, T, M and I; X11 is independently selected from the group consisting of S, Q, H, R, K and N; X12 is independently selected from the group consisting of R, K, N, T, S, Q, H, P, E, D, A and G; and X13 is independently selected from the group consisting of L, K, T, R, M, Q, P, E, A, G and V.
35. The vector library of claim 30, wherein VR-V comprises amino acid sequence
X14X15X16X17X18 NNSX19FPWX20AASX21.
36. The vector library of claim 35, wherein X14 is independently selected from the group consisting of K and T; X15 is independently selected from the group consisting of T, I, A and V; Xi6 is independently selected from the group consisting of A, P, H, D, S and Y; X17 is independently selected from the group consisting of N, T, S, D, A and G; Xis is independently selected from the group consisting of D, E, G, Q, H and R; X19 is independently selected from the group consisting of N, K, E and D; X20 is independently selected from the group consisting of T and P; and X21 is independently selected from the group consisting of K and T.
37. The vector library of claim 30, wherein VR-VI comprises amino acid sequence
Figure imgf000055_0001
38. The vector library of claim 37, wherein X22 is independently selected from the group consisting of K, R, E and G; X23 is independently selected from the group consisting of E, T, K, N, A and D; X24 is independently selected from the group consisting of E, D and G; and X25 is independently selected from the group consisting of K and R.
39. The vector library of claim 30, wherein VR-VII comprises amino acid sequence
X26X27X28X29X30X31X32X33X34X35X36X37.
40. The vector library of claim 39, wherein X26 is independently selected from the group consisting of E and Q; X27 is independently selected from the group consisting of G, N, S and D; X28 is independently selected from the group consisting of T, S, G and A; X29 is independently selected from the group consisting of T, K, N, R, S, E, D, A and G; X30 is independently selected from the group consisting of A, K, T, R, E and G; X31 is independently selected from the group consisting of S, N, T, D, A and G; X32 is independently selected from the group consisting of N and D; X33 is independently selected from the group consisting of A, T, I and V; X34 is independently selected from the group consisting of E, A and D; X35 is independently selected from the group consisting of L, N, I, D, V, Y and F; X36 is independently selected from the group consisting of D, E and G; and X37 is independently selected from the group consisting of N, K, R, S, Q, H, E, D and G.
41. The vector library of claim 30, wherein VR-VIII comprises amino acid sequence
X38X39X40X41X42PTX43X44X45VX46.
42. The vector library of claim 41, wherein X38 is independently selected from the group consisting of S, K, N, T, R, E, D, A and G; X39 is independently selected from the group consisting of S, K, N, T, R, Q, H, P, E, D, A and G; X40 is independently selected from the group consisting of N, Q, H, R, K and S; X41 is independently selected from the group consisting of T, N, S, D, A and G; X42 is independently selected from the group consisting of A, K, N, T, R, S, Q, H, P, E, D and G; X43 is independently selected from the group consisting of T, N, I, D, A, V, Y, S and F; X44 is independently selected from the group consisting of G, K, N, T, R, S, Q, H, P, E, D and A; X45 is independently selected from the group consisting of T, K, N, R, S, M, I, E, D, A, G, and V; and X46 is independently selected from the group consisting of N, T, K, P, Q, H, A, E and D.
43. The vector library of claim 28, wherein the AAV Cap protein comprises:
MAADGYLPDWLEDNLSEGIREWWALKPGVPQPKANQQHQDNRRGLVLPGYKYLGPGNGLD KGEPVNEADAAALEHDKAYDQQLKAGDNPYLKYNHADAE FQERLQEDT S FGGNLGRAVFQ AKKRILEPLGLVEEAAKTAPGKKGAVDQSPQE PDSS SGVGKSGKQPARKRLNFGQTGDSE SVPDPQPLGE PPAAPT SLGSNTMASGGGAPMADNNEGADGVGNS SGNWHCDSQWLGDRVI TT STRTWALPTYNNHLYKQI SSXXGAXNDNHY FGYSTPWGY FDFNRFHCH FS PRDWQRLI NNNWGFRPKKLS FKLFNIQVRGVTQNDGTTT IA NLTSTVQVFTDSEYQLPYVLGSAHQG CLPP FPADVFMVPQYGYLTLNNGSQAVGRS S FYCLEY FPSQMLRTGN FQ FSY FEDVPF HS SYAHSQSLDRLMNPLI DQYLYYLNRTQXXXXGTXXXXXLXFSQAGPQSMSLQARNWLP GPCYRQQRLSXXXXXNNNSXFPWXAASXYHLNGRDSLVNPGPAMASHXDDXXXFFPMHGN LI FGKXXXXXXXXXXXXVMITDEEE I RTTNPVATEQYGTVA NLQXXXXXPTXXXVXHQG ALPGMVWQDRDVYLQGPIWA .
44. The vector library of claim 28, wherein the AAV Cap protein comprises one of sequences 2-86 as listed in Table 4.
45. An AAV virion comprising a nucleic acid comprising:
(a) a first nucleotide sequence encoding at least one AAV Rep protein from serotype 3;
(b) a second nucleotide sequence encoding at least one AAV Cap protein, wherein the second nucleotide sequence differs from wildtype serotype 3 at least at one nucleotide position; and
(c) a first AAV terminal repeat from serotype 3 and a second AAV terminal repeat from serotype 3,
wherein the first and second nucleotide sequences are interposed between the first and the second AAV terminal repeat.
46. The AAV virion of claim 45, wherein the AAV virion is incorporated into at least one host cell.
47. The AAV virion of claim 46, wherein the at least one host cell is a mammalian cell.
48. The AAV virion of any one of claims 45-47, wherein the nucleic acid further comprises a third nucleotide sequence encoding at least one molecule providing helper function.
49. The AAV virion of claim 48, wherein the third nucleotide sequence encoding at least one molecule providing helper function is a polynucleotide from a virus selected from the group consisting of: adenovirus and herpesvirus.
50. The AAV virion of any one of claims 45-49, wherein the second nucleotide sequence comprises: TGCCCACTTACAACAACCATCTCTACAAGCAAATC
TCCAGCVVMDCAGGAGCTASCAACGACAACCACTACTTTGGCTACAGCACCCCTTGGGGG TATTTTGACTTTAACAGATTCCACTGCCACTTCTCACCACGTGACTGGCAGCGACTCATT AACAACAACTGGGGATTCCGGCCCAAGAAACTCAGCTTCAAGCTCTTCAACATCCAAGTT AGAGGGGTCACGCAGAACGATGGCACGACGACTATTGCCAATAACCTTACCAGCACGGTT CAAGTGTTTACGGACTCGGAGTATCAGCTCCCGTACGTGCTCGGGTCGGCGCACCAAGGC TGTCTCCCGCCGTTTCCAGCGGACGTCTTCATGGTCCCTCAGTATGGATACCTCACCCTG AACAACGGAAGTCAAGCGGTGGGACGCTCATCCTTTTACTGCCTGGAGTACTTCCCTTCG CAGATGCTAAGGACTGGAAATAACTTCCAATTCAGCTATACCTTCGAGGATGTACCTTTT CACAGCAGCTACGCTCACAGCCAGAGTTTGGATCGCTTGATGAATCCTCTTATTGATCAG TATCTGTACTACCTGAACAGAACGCAARGCAMCVCNRGCGGAACARCCRVCMHSMRSVVS CTGVNGTTTAGCCAGGCTGGGCCTCAGTCTATGTCTTTGCAGGCCAGAAATTGGCTACCT GGGCCCTGCTACCGGCAACAGAGACTTTCAAMARYCBMCRVCSRSAACAACAACAGTRAS TTTCCTTGGMCAGCGGCCAGCAMATATCATCTCAATGGCCGCGACTCGCTGGTGAATCCA GGACCAGCTATGGCCAGTCACRRGGACGATRMSGRSARATTTTTCCCTATGCACGGCAAT CTAATATTTGGCAAASAARRCRSCRVSRVARVCRA RYCGMSDWCGRSVRSGTAATGATT ACGGATGAAGAAGAGATTCGTACCACCAATCCTGTGGCAACAGAGCAGTATGGAACTGTG GCAAATAACTTGCAGRVSVVSMRSRVCVVSCCCACGDHTVVSRNSGTCVMSCATCAGGGG GCCTTACCTGGCATGGTGTGGCAAGATCG .
51. The AAV virion of claim 45, wherein the AAV Cap protein differs from wildtype serotype 3 at least at one amino acid position.
52. The AAV virion of claim 51, wherein the at least one differing amino acid position is in a variable region (VR).
53. The AAV virion of claim 52, wherein the VR is selected from the group consisting of VR-I, VR-IV, VR-V, VR-VI, VR-VII, VR-VIII and combinations thereof.
54. The AAV virion of claim 53, wherein VR-I comprises amino acid sequence X1X2GAX3.
55. The AAV virion of claim 54, wherein Xi is independently selected from the group consisting of Q, N, K, T, S, R, H, P, D, E, A and G; X2 is independently selected from the group consisting of S, T and A; and X3 is independently selected from the group consisting of S and T.
56. The AAV virion of claim 53, wherein VR-IV comprises amino acid sequence
X4X5X6X7GTX8X9X10X11X12LX13.
57. The AAV virion of claim 56, wherein X4 is independently selected from the group consisting of G and S; X5 is independently selected from the group consisting of T and N; X6 is independently selected from the group consisting of T, P and A; X7 is independently selected from the group consisting of S and G; Xs is independently selected from the group consisting of T and A; X9 is independently selected from the group consisting of N, T, S, D, A and G; X10 is independently selected from the group consisting of Q, H, P, L, K, N, T, M and I; X11 is independently selected from the group consisting of S, Q, H, R, K and N; X12 is independently selected from the group consisting of R, K, N, T, S, Q, H, P, E, D, A and G; and X13 is independently selected from the group consisting of L, K, T, R, M, Q, P, E, A, G and V.
58. The AAV virion of claim 53, wherein VR-V comprises amino acid sequence
X14X15X16X17X18 NNSX19FPWX20AASX21.
59. The AAV virion of claim 58, wherein X14 is independently selected from the group consisting of K and T; X15 is independently selected from the group consisting of T, I, A and V; Xi6 is independently selected from the group consisting of A, P, H, D, S and Y; Xn is independently selected from the group consisting of N, T, S, D, A and G; Xis is independently selected from the group consisting of D, E, G, Q, H and R; X19 is independently selected from the group consisting of N, K, E and D; X20 is independently selected from the group consisting of T and P; and X21 is independently selected from the group consisting of K and T.
60. The AAV virion of claim 53, wherein VR-VI comprises amino acid sequence
Figure imgf000060_0001
61. The AAV virion of claim 60, wherein X22 is independently selected from the group consisting of K, R, E and G; X23 is independently selected from the group consisting of E, T, K, N, A and D; X24 is independently selected from the group consisting of E, D and G; and X25 is independently selected from the group consisting of K and R.
62. The AAV virion of claim 53, wherein VR-VII comprises amino acid sequence
X26X27X28X29X30X31X32X33X34X35X36X37.
63. The AAV virion of claim 62, wherein X26 is independently selected from the group consisting of E and Q; X27 is independently selected from the group consisting of G, N, S and D; X28 is independently selected from the group consisting of T, S, G and A; X29 is independently selected from the group consisting of T, K, N, R, S, E, D, A and G; X30 is independently selected from the group consisting of A, K, T, R, E and G; X31 is independently selected from the group consisting of S, N, T, D, A and G; X32 is independently selected from the group consisting of N and D; X33 is independently selected from the group consisting of A, T, I and V; X34 is independently selected from the group consisting of E, A and D; X35 is independently selected from the group consisting of L, N, I, D, V, Y and F; X36 is independently selected from the group consisting of D, E and G; and X37 is independently selected from the group consisting of N, K, R, S, Q, H, E, D and G.
64. The AAV virion of claim 53, wherein VR-VIII comprises amino acid sequence
X38X39X40X41X42PTX43X44X45VX46.
65. The AAV virion of claim 64, wherein X38 is independently selected from the group consisting of S, K, N, T, R, E, D, A and G; X39 is independently selected from the group consisting of S, K, N, T, R, Q, H, P, E, D, A and G; X40 is independently selected from the group consisting of N, Q, H, R, K and S; X4i is independently selected from the group consisting of T, N, S, D, A and G; X42 is independently selected from the group consisting of A, K, N, T, R, S, Q, H, P, E, D and G; X43 is independently selected from the group consisting of T, N, I, D, A, V, Y, S and F; X44 is independently selected from the group consisting of G, K, N, T, R, S, Q, H, P, E, D and A; X45 is independently selected from the group consisting of T, K, N, R, S, M, I, E, D, A, G, and V; and X46 is independently selected from the group consisting of N, T, K, P, Q, H, A, E and D.
66. The AAV virion of claim 51, wherein the AAV Cap protein comprises:
MAADGYLPDWLEDNLSEGIREWWALKPGVPQPKA QQHQDNRRGLVLPGYKYLGPGNGLD KGEPVNEADAAALEHDKAYDQQLKAGDNPYLKYNHADAE FQERLQEDT S FGGNLGRAVFQ AKKRILEPLGLYEEAAKTAPGKKGAVDQSPQE PDSS SGVGKSGKQPARKRLNFGQTGDSE SVPDPQPLGE PPAAPT SLGSNTMASGGGAPMADNNEGADGVGNS SGNWHCDSQWLGDRVI TT STRTWALPTYNNHLYKQI SSXXGAXNDNHY FGYSTPWGY FDFNRFHCHFS PRDWQRLI NNNWGFRPKKLS FKLFNIQVRGVTQNDGTTT IA NLTSTVQVFTDSEYQLPYVLGSAHQG CLPP FPADVFMVPQYGYLTLNNGSQAVGRSS FYCLEY FPSQMLRTGNNFQFSYT FEDVPF HS SYAHSQSLDRLMNPLI DQYLYYLNRTQXXXXGTXXXXXLXFSQAGPQSMSLQARNWLP GPCYRQQRLSXXXXXNNNSXFPWXAASXYHLNGRDSLVNPGPAMASHXDDXXXFFPMHGN LI FGKXXXXXXXXXXXXVMITDEEE I RTTNPVATEQYGTVA NLQXXXXXPTXXXVXHQG ALPGMVWQDRDVYLQGPIWA .
67. The AAV virion of claim 51, wherein the AAV Cap protein comprises one of sequences 2-86 as listed in Table 4.
68. An AAV virion comprising:
(a) a first nucleotide sequence encoding at least one therapeutic molecule;
(b) a second nucleotide sequence comprising a regulatory sequence;
(c) a third nucleotide sequence comprising a first AAV terminal repeat from serotype 3; (d) a fourth nucleotide sequence comprising a second AAV terminal repeat from serotype 3; and
(e) a capsid comprising at least one AAV Cap protein that differs from wildtype serotype 3 at least at one amino acid position.
69. The AAV virion of claim 68, wherein the first nucleotide sequence is operably linked to the second nucleotide sequence and the first and second nucleotide sequences are interposed between the first and second AAV terminal repeat to form a transgene.
70. The AAV virion of claim 68, wherein said transgene is packaged within said capsid.
71. The AAV virion of any one of claims 68-70, wherein said second nucleotide sequence is a promoter or an enhancer.
72. The AAV virion of any one of claims 68-71, wherein the therapeutic molecule is selected from the group consisting of a polypeptide, a peptide, and an RNA.
73. A method of treating a disease comprising administering an effective amount of the AAV virion of any one of claims 68-72 to a patient in need thereof.
74. A method of selecting a tissue-specific or cell-specific variant of an AAV virion comprising:
(a) introducing AAV virions of any one of claims 45-73 into target tissues or cells;
(b) allowing sufficient time to elapse to propagate additional virions; and
(c) isolating the virions.
75. The method of claim 74, further comprising repeating steps (a) - (c) one or more times.
76. The method of claim 75, wherein the variant exhibits a higher target tropism for the target tissues or cells as compared to AAV serotype 3.
77. A modified AAV capsid protein, comprising:
a) a non-glutamine amino acid residue at one or more positions corresponding to Q263 and Q458 of wild-type AAV3 capsid protein;
b) a non-serine amino acid residue at one or more positions corresponding to S264, S267, S453, S459, S551, S586 and S587 of wild-type AAV3 capsid protein;
c) a non-glycine amino acid residue at one or more positions corresponding to G450, G547 and G594 of wild-type AAV3 capsid protein;
d) a non-threonine amino acid residue at one or more positions corresponding to T451, T452, T456, T492, T504, T548, T549, T589, T593 and T595 of wild-type AAV3 capsid protein; e) a non-asparagine amino acid residue at one or more positions corresponding to N457,
N494, N500, N552, N557, N58, and N597 of wild-type AAV3 capsid protein;
f) a non-leucine amino acid residue at one or more positions corresponding to L462 and L555 of wild-type AAV3 capsid protein;
g) a non-lysine amino acid residue at one or more positions corresponding to K491, K508, K528 and K533 of wild-type AAV3 capsid protein;
h) a non-alanine amino acid residue at one or more positions corresponding to A493, A550, A553 and A590 of wild-type AAV3 capsid protein;
i) a non-aspartic acid amino acid residue at one or more positions corresponding to D495 and D556 of wild-type AAV3 capsid protein;
j) a non-glutamic acid amino acid residue at one or more positions corresponding to
E531, E532, E546 and E554 of wild-type AAV3 capsid protein;
k) a non-arginine amino acid residue at position corresponding to R460 of wild-type AAV3 capsid protein; or
1) a combination of two or more amino acid substitutions listed in a) - k).
78. The modified AAV capsid protein of claim 77, wherein
a) the non-glutamine amino acid residue is selected from the group consisting of N, K, T, S, R, H, P, D, E, A, G, L, M and I;
b) the non-serine amino acid residue is selected from the group consisting of T, A, G, Q, H, R, K, N, D, E and P; c) the non-glycine amino acid residue is selected from the group consisting of S, N, D, K, T, R, Q, H, P, E and A;
d) the non-threonine amino acid residue is selected from the group consisting of N, P, A, I, V, P, S, G, K, R, E, D, Y, F and M;
e) the non-asparagine amino acid residue is selected from the group consisting of T, S, D,
A, G, K, E, R, Q, H and P;
f) the non-1 eucine amino acid residue is selected from the group consisting of K, T, R, M, Q, P, E, A, G, V, N, I, D, Y and F;
g) the non-lysine amino acid residue is selected from the group consisting of T, R, E and G;
h) the non-alanine amino acid residue is selected from the group consisting of P, H, D, S, Y, K, T, R, E, G, I, V, N and Q;
i) the non-aspartic acid amino acid residue is selected from the group consisting of E, G, Q, H and R;
j) the non-glutamic acid amino acid residue is selected from the group consisting of T, K,
N, A, D, G and Q; and
k) the non-arginine amino acid residue is selected from the group consisting of K, N, T, S, Q, H, P, E, D, A and G.
79. An isolated nucleic acid segment that encodes the modified AAV capsid protein of claim 77 or 78.
80. A recombinant adeno-associated viral (rAAV) vector comprising a nucleic acid segment that encodes the protein of claim 77 or 78.
81. The rAAV vector of claim 80, wherein the vector further at least comprises a nucleic acid segment that encodes a diagnostic or therapeutic molecule operably linked to a promoter capable of expressing the nucleic acid segment in a suitable host cell comprising the vector.
82. The rAAV vector of claim 80 or 81, wherein the transduction efficiency of a virion comprising the modified AAV capsid protein is about 2- to about 50-fold higher in a selected mammalian host cell than that of a virion that comprises a corresponding, unmodified, capsid protein.
83. The rAAV vector of claim 82, wherein the transduction efficiency of a virion comprising the modified AAV capsid protein is about 6- to about 40-fold higher in a selected mammalian host cell than that of a virion that comprises a corresponding, unmodified, capsid protein.
84. The rAAV vector of claim 83, wherein the transduction efficiency of a virion comprising the modified AAV capsid protein is about 8- to about 30-fold higher in a selected mammalian host cell than that of a virion that comprises a corresponding, unmodified, capsid protein.
85. The rAAV vector of any one of claims 81-84, wherein the nucleic acid segment further comprises an enhancer, a post-transcriptional regulatory sequence, a polyadenylation signal, or any combination thereof, operably linked to the nucleic acid segment.
86. The rAAV vector of any one of claims 81-85, wherein the nucleic acid segment expresses or encodes a polypeptide, a peptide, a ribozyme, a peptide nucleic acid, an siRNA, an RNAi, an antisense oligonucleotide, an antisense polynucleotide, an antibody, an antigen binding fragment, or any combination thereof.
87. The rAAV vector of any one of claims 81-86, wherein the therapeutic agent is an agonist, an antagonist, an anti-apoptosis factor, an inhibitor, a receptor, a cytokine, a cytotoxin, an erythropoietic agent, a glycoprotein, a growth factor, a growth factor receptor, a hormone, a hormone receptor, an interferon, an interleukin, an interleukin receptor, a nerve growth factor, a neuroactive peptide, a neuroactive peptide receptor, a protease, a protease inhibitor, a protein decarboxylase, a protein kinase, a protein kinase inhibitor, an enzyme, a receptor binding protein, a transport protein or an inhibitor thereof, a serotonin receptor or an uptake inhibitor thereof, a serpin, a serpin receptor, a tumor suppressor, a chemotherapeutic, or any combination thereof.
88. The rAAV vector of any one of claims 81-87, wherein the nucleic acid segment is of human, non-human primate, murine, porcine, bovine, ovine, feline, canine, equine, epine, caprine or lupine origin.
89. The rAAV vector of any one of claims 80-88, comprised within an adeno-associated viral particle or infectious rAAV virion.
90. The rAAV vector of claim 89, comprised within a virion having a serotype that is selected from the group consisting of AAV serotype 1, AAV serotype 2, AAV serotype 3, AAV serotype 4, AAV serotype 5, AAV serotype 6, AAV serotype 7, AAV serotype 8, AAV serotype 9, AAV serotype 10, AAV serotype 11, AAV serotype 12 and AAV serotype 13.
91. A plurality of rAAV vectors, virions or infectious viral particles comprising a nucleic acid segment that encodes a modified AAV3 capsid protein that comprises:
a) a non-glutamine amino acid residue at one or more positions corresponding to Q263 and Q458 of wild-type AAV3 capsid protein;
b) a non-serine amino acid residue at one or more positions corresponding to S264, S267, S453, S459, S551, S586 and S587 of wild-type AAV3 capsid protein;
c) a non-glycine amino acid residue at one or more positions corresponding to G450, G547 and G594 of wild-type AAV3 capsid protein;
d) a non-threonine amino acid residue at one or more positions corresponding to T451, T452, T456, T492, T504, T548, T549, T589, T593 and T595 of wild-type AAV3 capsid protein; e) a non-asparagine amino acid residue at one or more positions corresponding to N457, N494, N500, N552, N557, N58, and N597 of wild-type AAV3 capsid protein;
f) a non-leucine amino acid residue at one or more positions corresponding to L462 and
L555 of wild-type AAV3 capsid protein;
g) a non-lysine amino acid residue at one or more positions corresponding to K491, K508, K528 and K533 of wild-type AAV3 capsid protein;
h) a non-alanine amino acid residue at one or more positions corresponding to A493, A550, A553 and A590 of wild-type AAV3 capsid protein; i) a non-aspartic acid amino acid residue at one or more positions corresponding to D495 and D556 of wild-type AAV3 capsid protein;
j) a non-glutamic acid amino acid residue at one or more positions corresponding to E531, E532, E546 and E554 of wild-type AAV3 capsid protein;
k) a non-arginine amino acid residue at position corresponding to R460 of wild-type
AAV3 capsid protein; or
1) a combination of two or more amino acid substitutions listed in a) - k).
92. An isolated mammalian host cell comprising a modified AAV3 capsid protein that comprises:
a) a non-glutamine amino acid residue at one or more positions corresponding to Q263 and Q458 of wild-type AAV3 capsid protein;
b) a non-serine amino acid residue at one or more positions corresponding to S264, S267, S453, S459, S551, S586 and S587 of wild-type AAV3 capsid protein;
c) a non-glycine amino acid residue at one or more positions corresponding to G450,
G547 and G594 of wild-type AAV3 capsid protein;
d) a non-threonine amino acid residue at one or more positions corresponding to T451, T452, T456, T492, T504, T548, T549, T589, T593 and T595 of wild-type AAV3 capsid protein; e) a non-asparagine amino acid residue at one or more positions corresponding to N457, N494, N500, N552, N557, N58, and N597 of wild-type AAV3 capsid protein;
f) a non-leucine amino acid residue at one or more positions corresponding to L462 and L555 of wild-type AAV3 capsid protein;
g) a non-lysine amino acid residue at one or more positions corresponding to K491, K508, K528 and K533 of wild-type AAV3 capsid protein;
h) a non-alanine amino acid residue at one or more positions corresponding to A493,
A550, A553 and A590 of wild-type AAV3 capsid protein;
i) a non-aspartic acid amino acid residue at one or more positions corresponding to D495 and D556 of wild-type AAV3 capsid protein;
j) a non-glutamic acid amino acid residue at one or more positions corresponding to E531, E532, E546 and E554 of wild-type AAV3 capsid protein; k) a non-arginine amino acid residue at position corresponding to R460 of wild-type AAV3 capsid protein; or
1) a combination of two or more amino acid substitutions listed in a) - k).
93. The isolated mammalian host cell of claim 91, wherein the host cell is a stem cell, a hematopoietic cell, a blood cell, a neural cell, a retinal cell, an epithelial cell, an endothelial cell, a pancreatic cell, a cancer cell, a muscle cell, a vascular cell, a diaphragm cell, a stomach cell or a CD34+ cell.
94. A composition comprising:
I) a modified AAV3 capsid protein that comprises:
a) a non-glutamine amino acid residue at one or more positions corresponding to Q263 and Q458 of wild-type AAV3 capsid protein;
b) a non-serine amino acid residue at one or more positions corresponding to S264, S267, S453, S459, S551, S586 and S587 of wild-type AAV3 capsid protein;
c) a non-glycine amino acid residue at one or more positions corresponding to G450, G547 and G594 of wild-type AAV3 capsid protein;
d) a non-threonine amino acid residue at one or more positions corresponding to T451, T452, T456, T492, T504, T548, T549, T589, T593 and T595 of wild-type AAV3 capsid protein; e) a non-asparagine amino acid residue at one or more positions corresponding to N457,
N494, N500, N552, N557, N58, and N597 of wild-type AAV3 capsid protein;
f) a non-leucine amino acid residue at one or more positions corresponding to L462 and L555 of wild-type AAV3 capsid protein;
g) a non-lysine amino acid residue at one or more positions corresponding to K491, K508, K528 and K533 of wild-type AAV3 capsid protein;
h) a non-alanine amino acid residue at one or more positions corresponding to A493, A550, A553 and A590 of wild-type AAV3 capsid protein;
i) a non-aspartic acid amino acid residue at one or more positions corresponding to D495 and D556 of wild-type AAV3 capsid protein;
j) a non-glutamic acid amino acid residue at one or more positions corresponding to
E531, E532, E546 and E554 of wild-type AAV3 capsid protein; k) a non-arginine amino acid residue at position corresponding to R460 of wild-type AAV3 capsid protein; or
1) a combination of two or more amino acid substitutions listed in a) - k);
II) a nucleic acid segment that encodes the protein of I), or
III) a viral vector that comprises the nucleic acid segment of II); and a pharmaceutically- acceptable buffer, diluent or excipient.
95. The composition of claim 94, comprised within a kit for diagnosing, preventing, treating or ameliorating one or more symptoms of a mammalian disease, injury, disorder, trauma, or dysfunction.
96. A method for providing a mammalian in need thereof with a diagnostically- or therapeutically-effective amount of a selected biological molecule, the method comprising providing to a cell, tissue or organ of a mammal in need thereof, an amount of the rAAV vector of claim 85; and for a time effective to provide the mammal with a diagnostically- or a therapeutically-effective amount of the selected biological molecule.
97. A method for diagnosing preventing, treating or ameliorating at least one or more symptoms of a disease, a disorder, a dysfunction, an injury, an abnormal condition or trauma in a mammal, the method comprising administering to a mammal in need thereof the rAAV vector of any one of claims 81-87, in an amount and for a time sufficient to diagnose, prevent, treat or ameliorate the one or more symptoms of the disease, disorder, dysfunction, injury, abnormal condition or trauma in the mammal.
98. The method of claim 97, wherein the mammal is human.
99. A method of transducing a population of mammalian cells, comprising introducing into one or more cells of the population, a composition that comprises an effective amount of the rAAV vector of claim 80.
100. The method of claim 99, wherein the population of cells comprises CD34+ cells, stem cells, hematopoietic cells, endothelial cells, epithelial cells, blood cells, retinal cells, pancreatic cells, cancer cells, muscle cells, vascular cells, diaphragm cells, stomach cells, liver cells, lung cells, heart cells, intestinal cells, kidney cells or brain cells of a mammal.
101. A rAAV particle comprising a modified capsid protein that comprises:
a) a non-glutamine amino acid residue at one or more positions corresponding to Q263 and Q458 of wild-type AAV3 capsid protein;
b) a non-serine amino acid residue at one or more positions corresponding to S264, S267, S453, S459, S551, S586 and S587 of wild-type AAV3 capsid protein;
c) a non-glycine amino acid residue at one or more positions corresponding to G450, G547 and G594 of wild-type AAV3 capsid protein;
d) a non-threonine amino acid residue at one or more positions corresponding to T451, T452, T456, T492, T504, T548, T549, T589, T593 and T595 of wild-type AAV3 capsid protein; e) a non-asparagine amino acid residue at one or more positions corresponding to N457,
N494, N500, N552, N557, N58, and N597 of wild-type AAV3 capsid protein;
f) a non-leucine amino acid residue at one or more positions corresponding to L462 and L555 of wild-type AAV3 capsid protein;
g) a non-lysine amino acid residue at one or more positions corresponding to K491, K508, K528 and K533 of wild-type AAV3 capsid protein;
h) a non-alanine amino acid residue at one or more positions corresponding to A493, A550, A553 and A590 of wild-type AAV3 capsid protein;
i) a non-aspartic acid amino acid residue at one or more positions corresponding to D495 and D556 of wild-type AAV3 capsid protein;
j) a non-glutamic acid amino acid residue at one or more positions corresponding to
E531, E532, E546 and E554 of wild-type AAV3 capsid protein;
k) a non-arginine amino acid residue at position corresponding to R460 of wild-type AAV3 capsid protein; or
1) a combination of two or more amino acid substitutions listed in a) - k).
102. The rAAV particle of claim 101, wherein said particle further comprises at least a first nucleic acid segment that encodes a therapeutic agent operably linked to a promoter capable of expressing said segment in a host cell.
103. The rAAV particle of claim 102, further comprising an enhancer sequence operably linked to said nucleic acid segment.
104. The rAAV particle of clam 103, wherein said enhancer sequence is selected from the group consisting of a CMV enhancer, a synthetic enhancer, a retinal-specific enhancer, a liver-specific enhancer, a vascular-specific enhancer, a brain-specific enhancer, a neural cell-specific enhancer, a lung-specific enhancer, a muscle-specific enhancer, a kidney-specific enhancer, a pancreas- specific enhancer, and an islet cell-specific enhancer.
105. The rAAV particle of any one of claims 102-104, wherein said promoter is a heterologous, tissue-specific, constitutive or inducible promoter.
106. The rAAV particle of claim 105, wherein said promoter is selected from the group consisting of a CMV promoter, a β-actin promoter, an insulin promoter, an enolase promoter, a BDNF promoter, an NGF promoter, an EGF promoter, a growth factor promoter, an axon- specific promoter, a dendrite-specific promoter, a brain-specific promoter, a hippocampal- specific promoter, a kidney-specific promoter, an elafin promoter, a cytokine promoter, an interferon promoter, an alpha- 1 antitrypsin promoter, a neural cell-specific promoter, a central nervous system cell-specific promoter, a peripheral nervous system cell-specific promoter, an interleukin promoter, a serpin promoter, a retinal-specific promoter, a hybrid CMV promoter, a hybrid β-actin promoter, an EF1 promoter, a Ula promoter, a Ulb promoter, a Tet-inducible promoter and a VP16-LexA promoter.
107. The rAAV particle of claim 105, wherein said promoter is a mammalian or avian β-actin promoter.
108. The rAAV particle of any one of claims 102-107, wherein said first nucleic acid segment further comprises a post-transcriptional regulatory sequence or a polyadenylation signal.
109. The rAAV particle of any one of claims 102-107, wherein said first nucleic acid segment further comprises a woodchuck hepatitis virus post-transcription regulatory element.
110. The rAAV particle of any one of claims 102-109, wherein said therapeutic agent is selected from the group consisting of a polypeptide, a peptide, an antibody, an antigen binding fragment, a ribozyme, a peptide nucleic acid, an siRNA, an RNAi, an antisense oligonucleotide and an antisense polynucleotide.
111. The rAAV particle of any one of claims 102-110, wherein said therapeutic agent is a protein or polypeptide selected from the group consisting of an adrenergic agonist, an anti- apoptosis factor, an apoptosis inhibitor, a cytokine receptor, a cytokine, a cytotoxin, an erythropoietic agent, a glutamic acid decarboxylase, a glycoprotein, a growth factor, a growth factor receptor, a hormone, a hormone receptor, an interferon, an interleukin, an interleukin receptor, a kinase, a kinase inhibitor, a nerve growth factor, a netrin, a neuroactive peptide, a neuroactive peptide receptor, a neurogenic factor, a neurogenic factor receptor, a neuropilin, a neurotrophic factor, a neurotrophin, a neurotrophin receptor, an N-methyl-D-aspartate antagonist, a plexin, a protease, a protease inhibitor, a protein decarboxylase, a protein kinase, a protein kinase inhibitor, a proteolytic protein, a proteolytic protein inhibitor, a semaphoring, a semaphoring receptor, a serotonin transport protein, a serotonin uptake inhibitor, a serotonin receptor, a serpin, a serpin receptor and a tumor suppressor.
112. The rAAV particle according to any one of claims 102-110, wherein said therapeutic agent is a polypeptide selected from the group consisting of BDNF, CNTF, CSF, EGF, FGF, G-SCF, GM-CSF, gonadotropin, IFN, IFG-1, M-CSF, NGF, PDGF, PEDF, TGF, TGF-B2, TNF, VEGF, prolactin, somatotropin, XIAPl, IL-1, IL-2, IL-3, IL-4, IL-5, IL-6, IL-7, IL-8, IL-9, IL-10, IL- 10(I87A), viral IL-10, IL-11, IL-12, IL-13, IL-14, IL-15, IL-16, IL-17, IL-18, and any combination thereof.
113. The rAAV particle according to any one of claims 102-109, wherein said first nucleic acid segment encodes a peptide or polypeptide of human, murine, avian, porcine, bovine, ovine, feline, canine, equine, epine, caprine, lupine or primate origin.
114. The rAAV particle according to any one of claims 101-113, wherein
a) the non-glutamine amino acid residue is selected from the group consisting of N, K, T, S, R, H, P, D, E, A, G, L, M and I;
b) the non-serine amino acid residue is selected from the group consisting of T, A, G, Q,
H, R, K, N, D, E and P;
c) the non-glycine amino acid residue is selected from the group consisting of S, N, D, K,
T, R, Q, H, P, E and A;
d) the non-threonine amino acid residue is selected from the group consisting of N, P, A,
I, V, P, S, G, K, R, E, D, Y, F and M;
e) the non-asparagine amino acid residue is selected from the group consisting of T, S, D, A, G, K, E, R, Q, H and P;
f) the non-1 eucine amino acid residue is selected from the group consisting of K, T, R, M, Q, P, E, A, G, V, N, I, D, Y and F;
g) the non-lysine amino acid residue is selected from the group consisting of T, R, E and
G;
h) the non-alanine amino acid residue is selected from the group consisting of P, H, D, S,
Y, K, T, R, E, G, I, V, N and Q;
i) the non-aspartic acid amino acid residue is selected from the group consisting of E, G, Q, H and R;
j) the non-glutamic acid amino acid residue is selected from the group consisting of T, K, N, A, D, G and Q; and
k) the non-arginine amino acid residue is selected from the group consisting of K, N, T, S, Q, H, P, E, D, A and G.
115. A composition comprising the rAAV particle in accordance with any one of claims 101- 114.
116. A method for providing a mammal in need thereof with a therapeutically-effective amount of a therapeutic agent, said method comprising providing to a cell, tissue or organ of said mammal, an amount of a composition according to claim 114, for a time effective to provide said mammal with a therapeutically-effective amount of said therapeutic agent.
117. The method of claim 115, wherein said therapeutic agent is selected from the group consisting of a polypeptide, a peptide, an antibody, an antigen binding fragment, a ribozyme, a peptide nucleic acid, an siRNA, an RNAi, a PNA, an antisense oligonucleotide and an antisense polynucleotide.
PCT/US2016/058130 2015-10-22 2016-10-21 Synthetic combinatorial aav3 capsid library WO2017070476A2 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US15/769,615 US20200181644A1 (en) 2015-10-22 2016-10-21 Synthetic combinatorial aav3 capsid library

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201562245025P 2015-10-22 2015-10-22
US62/245,025 2015-10-22

Publications (2)

Publication Number Publication Date
WO2017070476A2 true WO2017070476A2 (en) 2017-04-27
WO2017070476A3 WO2017070476A3 (en) 2017-06-01

Family

ID=58557810

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2016/058130 WO2017070476A2 (en) 2015-10-22 2016-10-21 Synthetic combinatorial aav3 capsid library

Country Status (2)

Country Link
US (1) US20200181644A1 (en)
WO (1) WO2017070476A2 (en)

Cited By (21)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10335466B2 (en) 2014-11-05 2019-07-02 Voyager Therapeutics, Inc. AADC polynucleotides for the treatment of parkinson's disease
US10570395B2 (en) 2014-11-14 2020-02-25 Voyager Therapeutics, Inc. Modulatory polynucleotides
US10577627B2 (en) 2014-06-09 2020-03-03 Voyager Therapeutics, Inc. Chimeric capsids
US10584337B2 (en) 2016-05-18 2020-03-10 Voyager Therapeutics, Inc. Modulatory polynucleotides
US10597660B2 (en) 2014-11-14 2020-03-24 Voyager Therapeutics, Inc. Compositions and methods of treating amyotrophic lateral sclerosis (ALS)
WO2021108467A1 (en) * 2019-11-25 2021-06-03 University Of Florida Research Foundation, Incorporated Novel aav3b variants that target human hepatocytes and evade the humoral immune response
WO2021108468A1 (en) * 2019-11-25 2021-06-03 University Of Florida Research Foundation, Incorporated Novel aav3b variants that target human hepatocytes in the liver of humanized mice
US11091777B2 (en) 2013-09-26 2021-08-17 University Of Florida Research Foundation, Incorporated Synthetic combinatorial AAV capsid library for targeted gene therapy
US11299751B2 (en) 2016-04-29 2022-04-12 Voyager Therapeutics, Inc. Compositions for the treatment of disease
US11298041B2 (en) 2016-08-30 2022-04-12 The Regents Of The University Of California Methods for biomedical targeting and delivery and devices and systems for practicing the same
US11326182B2 (en) 2016-04-29 2022-05-10 Voyager Therapeutics, Inc. Compositions for the treatment of disease
US11332502B2 (en) 2017-02-21 2022-05-17 University Of Florida Research Foundation, Incorporated Modified AAV capsid proteins and uses thereof
US11434502B2 (en) 2017-10-16 2022-09-06 Voyager Therapeutics, Inc. Treatment of amyotrophic lateral sclerosis (ALS)
US11497576B2 (en) 2017-07-17 2022-11-15 Voyager Therapeutics, Inc. Trajectory array guide system
US11512327B2 (en) 2017-08-03 2022-11-29 Voyager Therapeutics, Inc. Compositions and methods for delivery of AAV
US11603542B2 (en) 2017-05-05 2023-03-14 Voyager Therapeutics, Inc. Compositions and methods of treating amyotrophic lateral sclerosis (ALS)
US11697825B2 (en) 2014-12-12 2023-07-11 Voyager Therapeutics, Inc. Compositions and methods for the production of scAAV
US11752181B2 (en) 2017-05-05 2023-09-12 Voyager Therapeutics, Inc. Compositions and methods of treating Huntington's disease
US11759506B2 (en) 2017-06-15 2023-09-19 Voyager Therapeutics, Inc. AADC polynucleotides for the treatment of Parkinson's disease
US11931375B2 (en) 2017-10-16 2024-03-19 Voyager Therapeutics, Inc. Treatment of amyotrophic lateral sclerosis (ALS)
US11951121B2 (en) 2017-05-18 2024-04-09 Voyager Therapeutics, Inc. Compositions and methods for treating Huntington's disease

Family Cites Families (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP2292779B1 (en) * 2003-09-30 2016-11-16 The Trustees Of The University Of Pennsylvania Adeno-associated virus (AAV) clades, sequences, vectors containing same, and uses thereof
EP2357010B1 (en) * 2005-04-07 2013-06-12 The Trustees of The University of Pennsylvania Method of increasing the function of an AAV vector
US9169494B2 (en) * 2010-01-12 2015-10-27 The University Of North Carolina At Chapel Hill Restrictive inverted terminal repeats for viral vectors
CN111411125A (en) * 2013-05-21 2020-07-14 佛罗里达大学研究基金会有限公司 Capsid-modified RAAV3 vector compositions and uses in gene therapy of human liver cancer
CA2930549A1 (en) * 2013-09-26 2015-04-02 University Of Florida Research Foundation, Inc. Synthetic combinatorial aav capsid library for targeted gene therapy

Cited By (27)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11091777B2 (en) 2013-09-26 2021-08-17 University Of Florida Research Foundation, Incorporated Synthetic combinatorial AAV capsid library for targeted gene therapy
US10577627B2 (en) 2014-06-09 2020-03-03 Voyager Therapeutics, Inc. Chimeric capsids
US10335466B2 (en) 2014-11-05 2019-07-02 Voyager Therapeutics, Inc. AADC polynucleotides for the treatment of parkinson's disease
US11027000B2 (en) 2014-11-05 2021-06-08 Voyager Therapeutics, Inc. AADC polynucleotides for the treatment of Parkinson's disease
US10570395B2 (en) 2014-11-14 2020-02-25 Voyager Therapeutics, Inc. Modulatory polynucleotides
US10597660B2 (en) 2014-11-14 2020-03-24 Voyager Therapeutics, Inc. Compositions and methods of treating amyotrophic lateral sclerosis (ALS)
US10920227B2 (en) 2014-11-14 2021-02-16 Voyager Therapeutics, Inc. Compositions and methods of treating amyotrophic lateral sclerosis (ALS)
US11542506B2 (en) 2014-11-14 2023-01-03 Voyager Therapeutics, Inc. Compositions and methods of treating amyotrophic lateral sclerosis (ALS)
US11198873B2 (en) 2014-11-14 2021-12-14 Voyager Therapeutics, Inc. Modulatory polynucleotides
US11697825B2 (en) 2014-12-12 2023-07-11 Voyager Therapeutics, Inc. Compositions and methods for the production of scAAV
US11299751B2 (en) 2016-04-29 2022-04-12 Voyager Therapeutics, Inc. Compositions for the treatment of disease
US11326182B2 (en) 2016-04-29 2022-05-10 Voyager Therapeutics, Inc. Compositions for the treatment of disease
US11193129B2 (en) 2016-05-18 2021-12-07 Voyager Therapeutics, Inc. Modulatory polynucleotides
US10584337B2 (en) 2016-05-18 2020-03-10 Voyager Therapeutics, Inc. Modulatory polynucleotides
US11298041B2 (en) 2016-08-30 2022-04-12 The Regents Of The University Of California Methods for biomedical targeting and delivery and devices and systems for practicing the same
US11332502B2 (en) 2017-02-21 2022-05-17 University Of Florida Research Foundation, Incorporated Modified AAV capsid proteins and uses thereof
US11767346B2 (en) 2017-02-21 2023-09-26 University Of Florida Research Foundation, Incorporated Modified AAV capsid proteins and uses thereof
US11752181B2 (en) 2017-05-05 2023-09-12 Voyager Therapeutics, Inc. Compositions and methods of treating Huntington's disease
US11603542B2 (en) 2017-05-05 2023-03-14 Voyager Therapeutics, Inc. Compositions and methods of treating amyotrophic lateral sclerosis (ALS)
US11951121B2 (en) 2017-05-18 2024-04-09 Voyager Therapeutics, Inc. Compositions and methods for treating Huntington's disease
US11759506B2 (en) 2017-06-15 2023-09-19 Voyager Therapeutics, Inc. AADC polynucleotides for the treatment of Parkinson's disease
US11497576B2 (en) 2017-07-17 2022-11-15 Voyager Therapeutics, Inc. Trajectory array guide system
US11512327B2 (en) 2017-08-03 2022-11-29 Voyager Therapeutics, Inc. Compositions and methods for delivery of AAV
US11434502B2 (en) 2017-10-16 2022-09-06 Voyager Therapeutics, Inc. Treatment of amyotrophic lateral sclerosis (ALS)
US11931375B2 (en) 2017-10-16 2024-03-19 Voyager Therapeutics, Inc. Treatment of amyotrophic lateral sclerosis (ALS)
WO2021108468A1 (en) * 2019-11-25 2021-06-03 University Of Florida Research Foundation, Incorporated Novel aav3b variants that target human hepatocytes in the liver of humanized mice
WO2021108467A1 (en) * 2019-11-25 2021-06-03 University Of Florida Research Foundation, Incorporated Novel aav3b variants that target human hepatocytes and evade the humoral immune response

Also Published As

Publication number Publication date
US20200181644A1 (en) 2020-06-11
WO2017070476A3 (en) 2017-06-01

Similar Documents

Publication Publication Date Title
WO2017070476A2 (en) Synthetic combinatorial aav3 capsid library
US11091777B2 (en) Synthetic combinatorial AAV capsid library for targeted gene therapy
AU2014324717A1 (en) Synthetic combinatorial AAV capsid library for targeted gene therapy
JP2022528416A (en) Recombinant adeno-associated virus and its use
KR102234672B1 (en) Capsid-modified, raav3 vector compositions and uses in gene therapy of human liver cancer
RU2683497C2 (en) Composition and methods for highly efficient gene transfer using aav capsid variants
CN104470945B (en) High transduction efficiency rAAV vectors, compositions, and methods of use thereof
WO2022076750A2 (en) Recombinant adeno-associated viruses for cns or muscle delivery
CN111718420B (en) Fusion protein for gene therapy and application thereof
CN116685329A (en) Nucleic acid constructs and their use for the treatment of spinal muscular atrophy
US20230049066A1 (en) Novel aav3b variants that target human hepatocytes in the liver of humanized mice
WO2024015877A2 (en) Novel aav3b capsid variants with enhanced hepatocyte tropism
WO2020187272A1 (en) Fusion protein for gene therapy and application thereof
US20230340526A1 (en) Novel aav3b variants that target hepatocytes and evade the humoral immune response
CN111718418B (en) Fusion protein for enhancing gene editing and application thereof
JP2023545384A (en) Recombinant adeno-associated virus for central nervous system or muscle delivery
CN115160410A (en) Novel muscle high-efficiency affinity adeno-associated virus serotype and related application
WO2023060269A1 (en) Recombinant adeno-associated viruses for targeted delivery
JP2024513907A (en) Artificial regulatory cassettes for muscle-specific gene expression
WO2023201277A1 (en) Recombinant adeno-associated viruses for cns tropic delivery
WO2023133593A9 (en) Aav5 capsid variants
WO2023133593A2 (en) Aav5 capsid variants
WO2023060272A2 (en) Recombinant adeno-associated viruses for cns tropic delivery
WO2023205751A1 (en) Aav capsid proteins for nucleic acid transfer

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 16858297

Country of ref document: EP

Kind code of ref document: A2

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 16858297

Country of ref document: EP

Kind code of ref document: A2