Recombinant adeno-associated virus vectors

Information

  • Patent Grant
  • 11981914
  • Patent Number
    11,981,914
  • Date Filed
    Monday, September 25, 2023
    8 months ago
  • Date Issued
    Tuesday, May 14, 2024
    20 days ago
Abstract
AAV capsid proteins comprising a modification in the amino acid sequence and virus vectors comprising the modified AAV capsid protein are described. Also described are methods of administering the virus vectors and virus capsids to a cell or to a subject in vivo.
Description
DESCRIPTION OF THE TEXT FILE SUBMITTED ELECTRONICALLY

The instant application contains a Sequence Listing, which has been submitted via Patent Center and is hereby incorporated by reference in its entirety. Said .xml copy, created on Oct. 16, 2023 is named GSB-01302, and is 881,333 bytes in size.


TECHNICAL FIELD

This application relates to adeno-associated virus (AAV) vectors comprising recombinant capsid proteins. In some embodiments, the recombinant AAV vectors evade neutralizing antibodies without decreased transduction efficiency.


BACKGROUND

Host-derived pre-existing antibodies generated upon natural encounter of AAV or recombinant AAV vectors prevent first time as well as repeat administration of AAV vectors as vaccines and/or for gene therapy. Serological studies reveal a high prevalence of antibodies in the human population worldwide with about 67% of people having antibodies against AAV1, 72% against AAV2, and about 40% against AAV5 through AAV9.


In gene therapy, certain clinical scenarios involving gene silencing or tissue degeneration may require multiple AAV vector administrations to sustain long term expression of the transgene. Accordingly, there is a need in the art for recombinant AAV vectors which evade antibody recognition. Such vectors will help a) expand the eligible cohort of subjects suitable for AAV-based gene therapy and b) allow multiple, repeat administrations of AAV-based gene therapy vectors.


BRIEF SUMMARY

Provided herein are recombinant AAV vectors which evade antibody recognition and/or selectively target tissues of the CNS.


In some embodiments, an adeno-associated virus (AAV) vector comprises (i) a recombinant capsid protein and (ii) a cargo nucleic acid encapsidated by the capsid protein, wherein the capsid protein comprises a peptide having the sequence of any one of SEQ ID NO: 12-20.


In some embodiments, an adeno-associated virus (AAV) vector comprises (i) a mutant AAV9 capsid protein and (ii) a cargo nucleic acid encapsidated by the capsid protein, wherein the capsid protein comprises a peptide having the sequence X1-X2-X3-X4-X5-X6-X7-X8 (SEQ ID NO: 158) at amino acids 451-458 of the native AAV9 capsid protein sequence, wherein the peptide does not occur in the native AAV9 capsid protein sequence.


In some embodiments, an adeno-associated virus (AAV) vector comprises (i) a mutant AAV9 capsid protein and (ii) a cargo nucleic acid encapsidated by the capsid protein, wherein the capsid protein comprises a peptide having the sequence X1-X2-X3-X4-X5-X6-X7-X8 (SEQ ID NO: 158) at amino acids 587-594 of the native AAV9 capsid protein sequence, wherein the peptide does not occur in the native AAV9 capsid protein sequence.


In some embodiments, the an adeno-associated virus (AAV) vector comprises (i) a recombinant capsid protein and (ii) a cargo nucleic acid encapsidated by the capsid protein, wherein the capsid protein comprises an amino acid sequence that is at least 95%, at least 96%, at least 97%, at least 98%, or at least 99% identical to any one of SEQ ID NO: 165-187.


Also provided herein are nucleic acid sequences encoding a recombinant capsid protein, and expression vectors comprising the same.


Also provided are cells comprising a nucleic acid, an expression vector, an AAV vector, or an AAV capsid described herein.


Also provided are pharmaceutical compositions comprising a nucleic acid, an expression vector, an AAV vector, an AAV capsid, or a cell described herein.


Also provided are methods for treating a subject in need thereof comprising administering to the subject a therapeutically effective amount of an AAV vector described herein.


Also provided are in vitro method of introducing a nucleic acid molecule into a cell, the methods comprising contacting the cell with an AAV vector described herein.


Also provided is an AAV vector described herein for use as a medicament.


Also provided is an AAV vector described herein for use in a method of treatment of a subject in need thereof.


Also provided is an AAV vector described herein for use in a method of treating or preventing a disease or disorder of the CNS in a subject in need thereof.


These and other embodiments are described in more detail below.





BRIEF DESCRIPTION OF THE DRAWINGS


FIG. 1A-1C. Bubble plots showing analysis of library diversity, directed evolution and enrichment of novel antigenic footprints. Parental (FIG. 1A) and evolved libraries from a first round (FIG. 1B) and a second round (FIG. 1C) of evolution were subjected to high-throughput sequencing using the Illumina MiSeq platform. Following analysis with a custom Perl script, enriched amino acid sequences were plotted. Each bubble represents a distinct capsid amino acid sequence with the radius of the bubble proportional to the number of reads for that variant in the respective library. The y-axis represents the percentage of total reads from the sequencing run. Data are spread along the x-axis for ease of visualization. The percent reduction in unique clones (96.5%) directly demonstrates that numerous “un-fit” sequences were removed after a first and second round of evolution. Dominant isolates were selected for further analysis.



FIG. 2. Volumetric yield of AAV vectors comprising AAV capsid variants STRD.101 and STRD.102, as compared to wildtype AAV9. Bars represent mean+/−95% confidence interval.



FIG. 3. Infectivity values of AAV-STRD.101 and wildtype AAV9 determined using a standard TCID50 assay. Data are graphed as the natural log of the number of particles required to generate an infectious unit (P:I Ratio). Error bars represent standard deviation.



FIG. 4A-4D. Transduction of U87 cells (FIG. 4A), N2A cells (FIG. 4B), Sy5Y cells (FIG. 4C), and U2OS cells (FIG. 4D) by recombinant AAV vectors comprising the STRD.101 capsid and packaging a luciferase transgene, as compared to wildtype AAV9 vectors similarly packaging a luciferase sequence. Error bars represent standard error.



FIG. 5. Representative fluorescent microscopy images showing tdTomato expression in coronal vibratome sections 24 hours post-fixation with 4% PFA. Each section is 25 μm thick. Top panel shows images obtained using a 4× objective lens with native tdTomato fluorescence. The bottom panel shows images obtained using a 10× objective lens with native tdTomato fluorescence.



FIG. 6. Representative immunohistochemistry images showing tdTomato expression in coronal vibratome sections 24 hours post-fixation with 4% PFA. Each section is 25 μm thick.



FIG. 7. Representative fluorescent microscopy images showing TdTomato expression in vibratrome liver sections 24 hours post-fixation with 4% PFA. Each section is 25 μm in thick. Panels show native tdTomato fluorescence with DAPI counterstain.



FIG. 8. Representative fluorescent microscopy images showing TdTomato expression in vibratrome heart sections 24 hours post-fixation with 4% PFA. Each section is 50 μm in thick. Panels show native tdTomato fluorescence with DAPI counterstain.



FIG. 9. Biodistribution of recombinant AAVs in non-human primates. Horizontal line shows limit of detection.





DETAILED DESCRIPTION

Unless otherwise defined, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this disclosure belongs. The terminology used in the detailed description herein is for the purpose of describing particular embodiments only and is not intended to be limiting.


All publications, patent applications, patents, GenBank or other accession numbers and other references mentioned herein are incorporated by reference in their entirety for all purposes.


The designation of amino acid positions in the AAV capsid proteins in the disclosure and the appended claims is with respect to VP1 capsid subunit numbering. It will be understood by those skilled in the art that the modifications described herein if inserted into the AAV cap gene may result in modifications in the VP1, VP2 and/or VP3 capsid subunits. Alternatively, the capsid subunits can be expressed independently to achieve modification in only one or two of the capsid subunits (VP1, VP2, VP3, VP1+VP2, VP1+VP3, or VP2+VP3).


Definitions

The following terms are used in the description herein and the appended claims.


The singular forms “a,” “an” and “the” are intended to include the plural forms as well, unless the context clearly indicates otherwise.


Furthermore, the term “about” as used herein when referring to a measurable value such as an amount of the length of a polynucleotide or polypeptide sequence, dose, time, temperature, and the like, is meant to encompass variations of ±20%, 10%, 5%, 1%, +0.5%, or even ±0.1% of the specified amount.


Also as used herein, “and/or” refers to and encompasses any and all possible combinations of one or more of the associated listed items, as well as the lack of combinations when interpreted in the alternative (“or”).


Unless the context indicates otherwise, it is specifically intended that the various features described herein can be used in any combination. Moreover, in some embodiments, any feature or combination of features set forth herein can be excluded or omitted. To illustrate further, if, for example, the specification indicates that a particular amino acid can be selected from A, G, I, L and/or V, this language also indicates that the amino acid can be selected from any subset of these amino acid(s) for example A, G, I or L; A, G, I or V; A or G; only L; etc., as if each such subcombination is expressly set forth herein. Moreover, such language also indicates that one or more of the specified amino acids can be disclaimed. For example, in some embodiments the amino acid is not A, G or I; is not A; is not G or V; etc., as if each such possible disclaimer is expressly set forth herein.


As used herein, the terms “reduce,” “reduces,” “reduction” and similar terms mean a decrease of at least about 10%, about 15%, about 20%, about 25%, about 35%, about 50%, about 75%, about 80%, about 85%, about 90%, about 95%, about 97% or more.


As used herein, the terms “increase,” “improve,” “enhance,” “enhances,” “enhancement” and similar terms indicate an increase of at least about 10%, about 15%, about 20%, about 25%, about 50%, about 75%, about 100%, about 150%, about 200%, about 300%, about 400%, about 500% or more.


The term “parvovirus” as used herein encompasses the family Parvoviridae, including autonomously replicating parvoviruses and dependoviruses. The autonomous parvoviruses include members of the genera Protoparvovirus, Erythroparvovirus, Bocaparvovirus, and Densovirus subfamily. Exemplary autonomous parvoviruses include, but are not limited to, minute virus of mouse, bovine parvovirus, canine parvovirus, chicken parvovirus, feline panleukopenia virus, feline parvovirus, goose parvovirus, H1 parvovirus, muscovy duck parvovirus, B19 virus, and any other autonomous parvovirus now known or later discovered. Other autonomous parvoviruses are known to those skilled in the art. See, e.g., BERNARD N. FIELDS et al, VIROLOGY, volume 2, chapter 69 (4th ed., Lippincott-Raven Publishers; Cotmore et al. Archives of Virology DOI 10.1007/s00705-013-1914-1).


As used herein, the term “adeno-associated virus” (AAV), includes but is not limited to, AAV type 1, AAV type 2, AAV type 3 (including types 3A and 3B), AAV type 4, AAV type 5, AAV type 6, AAV type 7, AAV type 8, AAV type 9, AAV type 10, AAV type 11, AAV type 12, AAV type 13, AAV type rh32.33, AAV type rh8, AAV type rh10, AAV type rh74, AAV type hu.68, avian AAV, bovine AAV, canine AAV, equine AAV, ovine AAV, snake AAV, bearded dragon AAV, AAV2i8, AAV2g9, AAV-LK03, AAV7m8, AAV Anc80, AAV PHP.B, and any other AAV now known or later discovered. See, e.g., BERNARD N. FIELDS et al., VIROLOGY, volume 2, chapter 69 (4th ed., Lippincott-Raven Publishers). A number of AAV serotypes and clades have been identified (see, e.g., Gao et al, (2004) J. Virology 78:6381-6388; Moris et al, (2004) Virology 33-:375-383; and Table 2). Exemplary AAV capsid sequences for AAV1-9, AAVrh.10 and AAV11 are provided in SEQ ID NO: 1-11.


As used herein, the term “chimeric AAV” refers to an AAV comprising a capsid protein with regions, domains, individual amino acids that are derived from two or more different serotypes of AAV. In some embodiments, a chimeric AAV comprises a capsid protein comprised of a first region that is derived from a first AAV serotype and a second region that is derived from a second AAV serotype. In some embodiments, a chimeric AAV comprises a capsid protein comprised of a first region that is derived from a first AAV serotype, a second region that is derived from a second AAV serotype, and a third region that is derived from a third AAV serotype. In some embodiments, the chimeric AAV may comprise regions, domains, individual amino acids derived from AAV1, AAV2, AAV3, AAV4, AAV5, AAV6, AAV7, AAV8, AAV9, AAV10, AAV11, and/or AAV12. For example, the chimeric AAV may include regions, domains, and/or individual amino acids from a first and a second AAV serotype as shown below (Table 1), wherein AAVX+Y indicates a chimeric AAV including sequences derived from AAVX and AAVY:









TABLE 1





Chimeric AAVs



















Second AAV Serotype

















AAV1
AAV2
AAV3
AAV4
AAV5
AAV6
AAV7





First
AAV1
x
AAV1 + 2
AAV1 + 3
AAV1 + 4
AAV1 + 5
AAV1 + 6
AAV1 + 7


AAV
AAV2
AAV2 + 1
x
AAV2 + 3
AAV2 + 4
AAV2 + 5
AAV2 + 6
AAV2 + 7


Sertoype
AAV3
AAV3 + 1
AAV3 + 2
x
AAV3 + 4
AAV3 + 5
AAV3 + 6
AAV3 + 7



AAV4
AAV4 + 1
AAV4 + 2
AAV4 + 3
x
AAV4 + 5
AAV4 + 6
AAV4 + 7



AAV5
AAV5 + 1
AAV5 + 2
AAV5 + 3
AAV5 + 4
x
AAV5 + 6
AAV5 + 7



AAV6
AAV6 + 1
AAV6 + 2
IAAV6 + 3
AAV6 + 4
AAV6 + 5
x
AAV6 + 7



AAV7
AAV7 + 1
AAV7 + 2
AAV7 + 3
AAV7 + 4
AAV7 + 5
AAV7 + 6
x



AAV8
AAV8 + 1
AAV8 + 2
AAV8 + 3
AAV8 + 4
AAV8 + 5
AAV8 + 6
AAV8 + 7



AAV9
AAV9 + 1
AAV9 + 2
AAV9 + 3
AAV9 + 4
AAV9 + 5
AAV9 + 6
AAV9 + 7



AAV10
AAV10 + 1
AAV10 + 2
AAV10 + 3
AAV10 + 4
AAV10 + 5
AAV10 + 6
AAV10 + 7



AAV11
AAV11 + 1
AAV11 + 2
AAV11 + 3
AAV11 + 4
AAV11 + 5
AAV11 + 6
AAV11 + 7



AAV12
AAV12 + 1
AAV12 + 2
AAV12 + 3
AAV12 + 4
AAV12 + 5
AAV12 + 6
AAV12 + 7
















Second AAV Serotype

















AAV8
AAV9
AAV10
AAV11
AAV12






First
AAV1
AAV1 + 8
AAV1 + 9
AAV1 + 10
AAV1 + 11
AAV1 + 12



AAV
AAV2
AAV2 + 8
AAV2 + 9
AAV2 + 10
AAV2 + 11
AAV2 + 12



Sertoype
AAV3
AAV3 + 8
AAV3 + 9
AAV3 + 10
AAV3 + 11
AAV3 + 12




AAV4
AAV4 + 8
AAV4 + 9
AAV4 + 10
AAV4 + 11
AAV4 + 12




AAV5
AAV5 + 8
AAV5 + 9
AAV5 + 10
AAV5 + 11
AAV5 + 12




AAV6
AAV6 + 8
AAV6 + 9
AAV6 + 10
AAV6 + 11
AAV6 + 12




AAV7
AAV7 + 8
AAV7 + 9
AAV7 + 10
AAV7 + 11
AAV7 + 12




AAV8
x
AAV8 + 9
AAV8 + 10
AAV8 + 11
AAV8 + 12




AAV9
AAV9 + 8
x
AAV9 + 10
AAV9 + 11
AAV9 + 12




AAV10
AAV10 + 8
AAV10 + 9
x
AAV10 + 11
AAV10 + 12




AAV11
AAV11 + 8
AAV11 + 9
AAV11 + 10
x
AAV11 + 12




AAV12
AAV12 + 8
AAV12 + 9
AAV12 + 10
AAV12 + 11
x









By including individual amino acids or regions from multiple AAV serotypes in one capsid protein, capsid proteins that have multiple desired properties that are separately derived from the multiple AAV serotypes may be obtained.


The genomic sequences of various serotypes of AAV and the autonomous parvoviruses, as well as the sequences of the native terminal repeats (TRs), Rep proteins, and capsid subunits are known in the art. Such sequences may be found in the literature or in public databases such as GenBank. See, e.g., GenBank Accession Numbers NC_002077, NC_001401, NC_001729, NC_001863, NC_001829, NC_001862, NC_000883, NC_001701, NC_001510, NC_006152, NC_006261, AF063497, U89790, AF043303, AF028705, AF028704, J02275, J01901, J02275, X01457, AF288061, AH009962, AY028226, AY028223, NC_001358, NC_001540, AF513851, AF513852, AY530579; the disclosures of which are incorporated by reference herein for teaching parvovirus and AAV nucleic acid and amino acid sequences. See also, e.g., Srivistava et al., (1983) J. Virology 45:555; Chiorini et al, (1998) J Virology 71:6823; Chiorini et al., (1999) J. Virology 73: 1309; Bantel-Schaal et al., (1999) J Virology 73:939; Xiao et al, (1999) J Virology 73:3994; Muramatsu et al., (1996) Virology 221:208; Shade et al, (1986) J. Virol. 58:921; Gao et al, (2002) Proc. Nat. Acad. Sci. USA 99:11854; Moris et al, (2004) Virology 33:375-383; international patent publications WO 00/28061, WO 99/61601, WO 98/11244; and U.S. Pat. No. 6,156,303; the disclosures of which are incorporated by reference herein for teaching parvovirus and AAV nucleic acid and amino acid sequences. See also Table 2. The capsid structures of autonomous parvoviruses and AAV are described in more detail in BERNARD N. FIELDS et al., VIROLOGY, volume 2, chapters 69 & 70 (4th ed., Lippincott-Raven Publishers). See also, description of the crystal structure of AAV2 (Xie et al., (2002) Proc. Nat. Acad. Sci. 99: 10405-10), AAV9 (DiMattia et al., (2012) J. Virol. 86:6947-6958), AAV8 (Nam et al, (2007) J. Virol. 81: 12260-12271), AAV6 (Ng et al., (2010) J. Virol. 84:12945-12957), AAV5 (Govindasamy et al. (2013) J. Virol. 87, 11187-11199), AAV4 (Govindasamy et al. (2006) J. Virol. 80:11556-11570), AAV3B (Lerch et al., (2010) Virology 403:26-36), BPV (Kailasan et al., (2015) J. Virol. 89:2603-2614) and CPV (Xie et al, (1996) J. Mol. Biol. 6:497-520 and Tsao et al, (1991) Science 251:1456-64).









TABLE 2







AAV Serotypes and Clades









GenBank Accession Number





Complete Genomes



Adeno-associated virus 1
NC_002077, AF063497


Adeno-associated virus 2
NC_001401


Adeno-associated virus 3
NC_001729


Adeno-associated virus 3B
NC_001863


Adeno-associated virus 4
NC_001829


Adeno-associated virus 5
Y18065, AF085716


Adeno-associated virus 6
NC_001862


Avian AAV ATCC VR-865
AY186198, AY629583, NC_004828


Avian AAV strain DA-1
NC_006263, AY629583


Bovine AAV
NC_005889, AY388617, AAR26465


AAV11
AAT46339, AY631966


AAV12
ABI16639, DQ813647


Clade A



AAV1
NC_002077, AF063497


AAV6
NC_001862


Hu.48
AY530611


Hu 43
AY530606


Hu 44
AY530607


Hu 46
AY530609


Clade B



Hu. 19
AY530584


Hu. 20
AY530586


Hu 23
AY530589


Hu22
AY530588


Hu24
AY530590


Hu21
AY530587


Hu27
AY530592


Hu28
AY530593


Hu 29
AY530594


Hu63
AY530624


Hu64
AY530625


Hu13
AY530578


Hu56
AY530618


Hu57
AY530619


Hu49
AY530612


Hu58
AY530620


Hu34
AY530598


Hu35
AY530599


AAV2
NC_001401


Hu45
AY530608


Hu47
AY530610


Hu51
AY530613


Hu52
AY530614


Hu T41
AY695378


Hu S17
AY695376


Hu T88
AY695375


Hu T71
AY695374


Hu T70
AY695373


Hu T40
AY695372


Hu T32
AY695371


Hu T17
AY695370


Hu LG15
AY695377


Clade C



Hu9
AY530629


Hu10
AY530576


Hu11
AY530577


Hu53
AY530615


Hu55
AY530617


Hu54
AY530616


Hu7
AY530628


Hu18
AY530583


Hu15
AY530580


Hu16
AY530581


Hu25
AY530591


Hu60
AY530622


Ch5
AY243021


Hu3
AY530595


Hu1
AY530575


Hu4
AY530602


Hu2
AY530585


Hu61
AY530623


Clade D



Rh62
AY530573


Rh48
AY530561


Rh54
AY530567


Rh55
AY530568


Cy2
AY243020


AAV7
AF513851


Rh35
AY243000


Rh37
AY242998


Rh36
AY242999


Cy6
AY243016


Cy4
AY243018


Cy3
AY243019


Cy5
AY243017


Rh13
AY243013


Clade E



Rh38
AY530558


Hu66
AY530626


Hu42
AY530605


Hu67
AY530627


Hu40
AY530603


Hu41
AY530604


Hu37
AY530600


Rh40
AY530559


Rh2
AY243007


Bb1
AY243023


Bb2
AY243022


Rh10
AY243015


Hu17
AY530582


Hu6
AY530621


Rh25
AY530557


Pi2
AY530554


Pi1
AY530553


Pi3
AY530555


Rh57
AY530569


Rh50
AY530563


Rh49
AY530562


Hu39
AY530601


Rh58
AY530570


Rh61
AY530572


Rh52
AY530565


Rh53
AY530566


Rh51
AY530564


Rh64
AY530574


Rh43
AY530560


AAV8
AF513852


Rh8
AY242997


Rh1
AY530556


Clade F



Hu14 (AAV9)
AY530579


Hu31
AY530596


Hu32
AY530597


HSC1
MI332400.1


HSC2
MI332401.1


HSC3
MI332402.1


HSC4
MI332403.1


HSC5
MI332405.1


HSC6
MI332404.1


HSC7
MI332407.1


HSC8
MI332408.1


HSC9
MI332409.1


HSC11
MI332406.1


HSC12
MI332410.1


HSC13
MI332411.1


HSC14
MI332412.1


HSC15
MI332413.1


HSC16
MI332414.1


HSC17
MI332415.1


Hu68



Clonal Isolate



AAV5
Y18065, AF085716


AAV3
NC_001729


AAV3B
NC_001863


AAV4
NC_001829


Rh34
AY243001


Rh33
AY243002


Rh32
AY243003


Others



Rh74



Bearded Dragon AAV



Snake AAV
NC_006148.1









The term “self-complimentary AAV” or “scAAV” refers to a recombinant AAV vector which forms a dimeric inverted repeat DNA molecule that spontaneously anneals, resulting in earlier and more robust transgene expression compared with conventional single-strand (ss) AAV genomes. See, e.g., McCarty, D. M., et al., Gene Therapy 8, 1248-1254 (2001). Unlike conventional ssAAV, scAAV can bypass second-strand synthesis, the rate-limiting step for gene expression. Moreover, double-stranded scAAV is less prone to DNA degradation after viral transduction, thereby increasing the number of copies of stable episomes. Notably, scAAV can typically only hold a genome that is about 2.4 kb, half the size of a conventional AAV vector. In some embodiments, the AAV vectors described herein are self-complementary AAVs.


As used herein, the term “peptide” refers to a short amino acid sequence. The term peptide may be used to refer to portion or region of an AAV capsid amino acid sequence. The peptide may be a peptide that naturally occurs in a native AAV capsid, or a peptide that does not naturally occur in a native AAV capsid. Naturally occurring AAV peptides in an AAV capsid may be substituted by non-naturally occurring peptides. For example, a non-naturally occurring peptide may be substituted into an AAV capsid to provide a modified capsid, such that the naturally-occurring peptide is replaced by the non-naturally occurring peptide.


The term “tropism” as used herein refers to preferential entry of the virus into certain cells or tissues, optionally followed by expression (e.g., transcription and, optionally, translation) of a sequence(s) carried by the viral genome in the cell, e.g., for a recombinant virus, expression of a transgene of interest.


As used here, “systemic tropism” and “systemic transduction” (and equivalent terms) indicate that the virus capsid or virus vector as described herein exhibits tropism for or transduces, respectively, tissues throughout the body (e.g., brain, lung, skeletal muscle, heart, liver, kidney and/or pancreas). In some embodiments, systemic transduction of muscle tissues (e.g., skeletal muscle, diaphragm and cardiac muscle) is achieved. In some embodiments, systemic transduction of skeletal muscle tissues is achieved. For example, in some embodiments, essentially all skeletal muscles throughout the body are transduced (although the efficiency of transduction may vary by muscle type). In some embodiments, systemic transduction of limb muscles, cardiac muscle and diaphragm muscle is achieved. Optionally, the virus capsid or virus vector is administered via a systemic route (e.g., systemic route such as intravenously, intra-articularly or intra-lymphatically).


Alternatively, in some embodiments, the capsid or virus vector is delivered locally (e.g., to the footpad, intramuscularly, intradermally, subcutaneously, topically). In some embodiments, the capsid or virus vector is delivered locally to a tissue of the central nervous system (CNS), such as the brain or the spinal cord. In some embodiments, the capsid or virus vector is administered by intrathecal, intracerebral or intracerebroventricular injection.


Unless indicated otherwise, “efficient transduction” or “efficient tropism,” or similar terms, can be determined by reference to a suitable control (e.g., at least about 50%, about 60%, about 70%, about 80%, about 85%, about 90%, about 95% or more of the transduction or tropism, respectively, of the control). In some embodiments, the virus vector (e.g., the AVV vector) efficiently transduces or has efficient tropism for skeletal muscle, cardiac muscle, diaphragm muscle, pancreas (including β-islet cells), spleen, the gastrointestinal tract (e.g., epithelium and/or smooth muscle), cells of the central nervous system, lung, joint cells, and/or kidney. Suitable controls will depend on a variety of factors including the desired tropism profile. For example, AAV8 and AAV9 are highly efficient in transducing skeletal muscle, cardiac muscle and diaphragm muscle, but have the disadvantage of also transducing liver with high efficiency. Thus, viral vectors can be identified that demonstrate the efficient transduction of skeletal, cardiac and/or diaphragm muscle of AAV8 or AAV9, but with a much lower transduction efficiency for liver. Further, because the tropism profile of interest may reflect tropism toward multiple target tissues, it will be appreciated that a suitable vector may represent some tradeoffs. To illustrate, a virus vector may be less efficient than AAV8 or AAV9 in transducing skeletal muscle, cardiac muscle and/or diaphragm muscle, but because of low level transduction of liver, may nonetheless be very desirable.


Similarly, it can be determined if a virus “does not efficiently transduce” or “does not have efficient tropism” for a target tissue, or similar terms, by reference to a suitable control. In some embodiments, the virus vector does not efficiently transduce (i.e., does not have efficient tropism) for liver, kidney, gonads and/or germ cells. In some embodiments, undesirable transduction of tissue(s) (e.g., liver) is about 20% or less, about 10% or less, about 5% or less, about 1% or less, about 0.1% or less of the level of transduction of the desired target tissue(s) (e.g., skeletal muscle, diaphragm muscle, cardiac muscle and/or cells of the central nervous system).


As used herein in connection with an AAV vector (or a capsid protein or peptide thereof), the terms “selectively binds,” “selective binding” and similar terms, refer to binding of the AAV vector (or a capsid protein or peptide thereof) to a target in a manner dependent upon the presence of a particular molecular structure. In some embodiments, selective binding refers to binding of the AAV predominantly to a specific target, without substantial or significant binding to other targets. In some embodiments, an AAV vector (or capsid protein or peptide thereof) specifically binds to a receptor in a cell or tissue of interest, but does not exhibit substantial or significant binding to other receptors.


A “polynucleotide” is a sequence of nucleotide bases, and may be RNA, DNA or DNA-RNA hybrid sequences (including both naturally occurring and non-naturally occurring nucleotide). In some embodiments, a polynucleotide is either a single or double stranded DNA sequence.


As used herein, an “isolated” polynucleotide (e.g., an “isolated DNA” or an “isolated RNA”) means a polynucleotide at least partially separated from at least some of the other components of the naturally occurring organism or virus, for example, the cell or viral structural components or other polypeptides or nucleic acids commonly found associated with the polynucleotide. In some embodiments an “isolated” nucleotide is enriched by at least about 10-fold, about 100-fold, about 1000-fold, about 10,000-fold or more as compared with the starting material.


Likewise, an “isolated” polypeptide means a polypeptide that is at least partially separated from at least some of the other components of the naturally occurring organism or virus, for example, the cell or viral structural components or other polypeptides or nucleic acids commonly found associated with the polypeptide. In some embodiments an “isolated” polypeptide is enriched by at least about 10-fold, about 100-fold, about 1000-fold, about 10,000-fold or more as compared with the starting material.


As used herein, by “isolate” or “purify” (or grammatical equivalents) a virus vector, it is meant that the virus vector is at least partially separated from at least some of the other components in the starting material. In some embodiments an “isolated” or “purified” virus vector is enriched by at least about 10-fold, about 100-fold, about 1000-fold, about 10,000-fold or more as compared with the starting material.


A “therapeutic” polypeptide or protein is one that can alleviate, reduce, prevent, delay and/or stabilize symptoms that result from an absence or defect in a protein in a cell or subject and/or is a polypeptide that otherwise confers a benefit to a subject, e.g., anti-cancer effects or improvement in transplant survivability.


By the terms “treat,” “treating” or “treatment of” (and grammatical variations thereof) it is meant that the severity of the subject's condition is reduced, at least partially improved or stabilized and/or that some alleviation, mitigation, decrease or stabilization in at least one clinical symptom is achieved and/or there is a delay in the progression of the disease or disorder.


The terms “prevent,” “preventing” and “prevention” (and grammatical variations thereof) refer to prevention and/or delay of the onset of a disease, disorder and/or a clinical symptom(s) in a subject and/or a reduction in the severity of the onset of the disease, disorder and/or clinical symptom(s) relative to what would occur in the absence of the compositions and/or methods described herein. The prevention can be complete, e.g., the total absence of the disease, disorder and/or clinical symptom(s). The prevention can also be partial, such that the occurrence of the disease, disorder and/or clinical symptom(s) in the subject and/or the severity of onset is less than what would occur in the absence of the compositions and/or methods described herein.


“Therapeutically effective amount” as used herein refers to an amount that, when administered to a subject for treating a disease, or at least one of the clinical symptoms of a disease, is sufficient to affect such treatment of the disease or symptom thereof. The “therapeutically effective amount” may vary depending, for example, on the disease and/or symptoms of the disease, severity of the disease and/or symptoms of the disease or disorder, the age, weight, and/or health of the patient to be treated, and the judgment of the prescribing physician. An appropriate amount in any given instance may be ascertained by those skilled in the art or capable of determination by routine experimentation.


As used herein, the terms “virus vector,” “vector” or “gene delivery vector” refer to a virus (e.g., AAV) particle that functions as a nucleic acid delivery vehicle, and which comprises the vector genome (e.g., viral DNA [vDNA]) packaged within a virion. Alternatively, in some contexts, the term “vector” may be used to refer to the vector genome/vDNA alone. In some embodiments, the vector genome (e.g., the AAV vector genome) may be comprised in a “cargo nucleic acid.” In some embodiments, the vector genome is self-complementary (i.e., double stranded). In some embodiments, the vector genome is not self-complimentary (i.e., single stranded).


A “rAAV vector genome” or “rAAV genome” is an AAV genome (i.e., vDNA) that comprises one or more heterologous nucleic acid sequences. rAAV vectors generally require only the inverted terminal repeat(s) (ITR(s)) in cis to generate virus. All other viral sequences are dispensable and may be supplied in trans (Muzyczka, (1992) Curr. Topics Microbiol. Immunol. 158:97). Typically, the rAAV vector genome will only retain the one or two ITR sequences so as to maximize the size of the transgene that can be efficiently packaged by the vector. The structural and non-structural protein coding sequences may be provided in trans (e.g., from a vector, such as a plasmid, or by stably integrating the sequences into a packaging cell). In some embodiments, the rAAV vector genome comprises at least one ITR sequence (e.g., AAV ITR sequence), optionally two ITRs (e.g., two AAV ITRs), which typically will be at the 5′ and 3′ ends of the vector genome (i.e., the 5′ ITR and the 3′ ITR) and flank the heterologous nucleic acid, but need not be contiguous thereto.


The term “inverted terminal repeat” or “ITR” includes any viral terminal repeat or synthetic sequence that forms a hairpin structure and functions as an inverted terminal repeat (i.e., mediates the desired functions such as replication, virus packaging, integration and/or provirus rescue, and the like). The ITR can be an AAV ITR or a non-AAV ITR. For example, a non-AAV ITR sequence such as those of other parvoviruses (e.g., canine parvovirus (CPV), mouse parvovirus (MVM), human parvovirus B-19) or any other suitable virus sequence (e.g., the SV40 hairpin that serves as the origin of SV40 replication) can be used as an ITR, which can further be modified by truncation, substitution, deletion, insertion and/or addition. Further, the ITR can be partially or completely synthetic, such as the “double-D sequence” as described in U.S. Pat. No. 5,478,745.


An “AAV inverted terminal repeat” or “AAV ITR” may be from any AAV, including but not limited to serotypes 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13 or any other AAV now known or later discovered (see, e.g., Table 2). An AAV inverted terminal repeat need not have the native terminal repeat sequence (e.g., a native AAV ITR sequence may be altered by insertion, deletion, truncation and/or missense mutations), as long as the terminal repeat mediates the desired functions, e.g., replication, virus packaging, integration, and/or provirus rescue, and the like.


The virus vectors described herein can further be “targeted” virus vectors (e.g., having a directed tropism) and/or “hybrid” virus vectors (i.e., in which the viral ITRs and viral capsid are from different viruses) as described in international patent publication WO00/28004 and Chao et al, (2000) Molecular Therapy 2:619. In some embodiments, the virus vectors are targeted to a cell and/or tissue of the CNS.


The virus vectors described herein can further be duplexed virus particles as described in international patent publication WO 01/92551 (the disclosure of which is incorporated herein by reference in its entirety). Thus, in some embodiments, double stranded (duplex) genomes can be packaged into the virus capsids described herein. Further, the viral capsid or genomic elements can contain other modifications, including insertions, deletions and/or substitutions.


As used herein, the term “amino acid” encompasses any naturally occurring amino acid, modified forms thereof, and synthetic amino acids. Naturally occurring, levorotatory (L-) amino acids are shown in Table 3.









TABLE 3







Amino acid residues and abbreviations.









Abbreviation









Amino Acid Residue
Three-Letter Code
One-Letter Code





Alanine
Ala
A


Arginine
Arg
R


Asparagine
Asn
N


Aspartic acid (Aspartate)
Asp
D


Cysteine
Cys
C


Glutamine
Gln
Q


Glutamic acid (Glutamate)
Glu
E


Glycine
Gly
G


Histidine
His
H


Isoleucine
Ile
I


Leucine
Leu
L


Lysine
Lys
K


Methionine
Met
M


Phenylalanine
Phe
F


Proline
Pro
P


Serine
Ser
S


Threonine
Thr
T


Tryptophan
Trp
W


Tyrosine
Tyr
Y


Valine
Val
V









Alternatively, the amino acid can be a modified amino acid residue (nonlimiting examples are shown in Table 4) and/or can be an amino acid that is modified by post-translation modification (e.g., acetylation, amidation, formylation, hydroxylation, methylation, phosphorylation or sulfatation). Methods of chemically modifying amino acids are known in the art (see, e.g., Greg T. Hermanson, Bioconjugate Techniques, 1st edition, Academic Press, 1996).









TABLE 4







Modified Amino Acid Residues










Modified Amino Acid Residue
Abbreviation










Amino Acid Residue Derivatives










2-Aminoadipic acid
Aad



3-Aminoadipic acid
bAad



beta-Alanine, beta-Aminoproprionic acid
bAla



2-Aminobutyric acid
Abu



4-Aminobutyric acid, Piperidinic acid
4Abu



6-Aminocaproic acid
Acp



2-Aminoheptanoic acid
Ahe



2-Aminoisobutyric acid
Aib



3-Aminoisobutyric acid
bAib



2-Aminopimelic acid
Apm



t-butylalanine
t-BuA



Citrulline
Cit



Cyclohexylalanine
Cha



2,4-Diaminobutyric acid
Dbu



Desmosine
Des



2,21-Diaminopimelic acid
Dpm



2,3-Diaminoproprionic acid
Dpr



N-Ethylglycine
EtGly



N-Ethylasparagine
EtAsn



Homoarginine
hArg



Homocysteine
hCys



Homoserine
hSer



Hydroxylysine
Hyl



Allo-Hydroxylysine
aHyl



3-Hydroxyproline
3Hyp



4-Hydroxyproline
4Hyp



Isodesmosine
Ide



allo-Isoleucine
alle



Methionine sulfoxide
MSO



N-Methylglycine, sarcosine
MeGly



N-Methyl isoleucine
Melle



6-N-Methyllysine
MeLys



N-Methylvaline
MeVal



2-Naphthylalanine
2-Nal



Norvaline
Nva



Norleucine
Nle



Ornithine
Orn



4-Chlorophenylalanine
Phe(4-C1)



2-Fluorophenylalanine
Phe(2-F)



3-Fluorophenylalanine
Phe(3-F)



4-Fluorophenylalanine
Phe(4-F)



Phenylglycine
Phg



Beta-2-thienylalanine
Thi









Further, the non-naturally occurring amino acid can be an “unnatural” amino acid (as described by Wang et al., Annu Rev Biophys Biomol Struct. 35-225-49 (2006)). These unnatural amino acids can advantageously be used to chemically link molecules of interest to the AAV capsid protein.


Modified AAV Capsid Proteins and AAV Vectors Comprising the Same


AAV Vectors


Additionally provided herein are adeno-associated virus (AAV) vectors comprising (i) a recombinant capsid protein and (ii) a cargo nucleic acid encapsidated by the capsid protein. In some embodiments, the recombinant capsid proteins (VP1, VP2 and/or VP3) may comprise a peptide in their amino acid sequence that does not occur in any native AAV capsid sequence. The inventors have demonstrated that capsid proteins comprising the peptides described herein can confer one or more desirable properties to virus vectors including, without limitation, the ability to evade neutralizing antibodies. Thus, AAV vectors described herein address the limitations associated with conventional AAV vectors.


Accordingly, in some embodiments, the present disclosure provides adeno-associated virus (AAV) vectors comprising (i) a recombinant capsid protein and (ii) a cargo nucleic acid encapsidated by the capsid protein; wherein the capsid protein comprises a peptide having the sequence of any one of SEQ ID NO: 12-20. In some embodiments, the cargo nucleic acid comprises 5′ and 3′ AAV inverted terminal repeats. In some embodiments, the cargo nucleic acid comprises a transgene. In some embodiments, the cargo nucleic acid is double stranded. In some embodiments, the cargo nucleic acid is single stranded. In some embodiments, the transgene encodes a therapeutic protein or RNA. In some embodiments, the recombinant capsid protein has at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99% sequence identity to the native sequence of the AAV1, AAV2, AAV3, AAV4, AAV5, AAV6, AAV7, AAV8, AAV9, AAV10, AAV11, AAV12, AAVrh.8, AAVrh.10, AAVrh32.33, AAVrh74, bovine AAV or avian AAV capsid. In some embodiments, the recombinant capsid protein has at least 90% sequence identity to the native sequence of the AAV9 capsid.


In some embodiments, the peptide is located at the amino acid positions corresponding to amino acids 451-458 of the native AAV9 capsid, or the equivalent amino acid residues in AAV1, AAV2, AAV3, AAV4, AAV5, AAV6, AAV7, AAV8, AAV10, AAV11, AAV12, AAVrh.8, AAVrh.10, AAVrh32.33, AAVrh74, bovine AAV or avian AAV, and the peptide is selected from any one of SEQ ID NO: 12-18. In some embodiments, the peptide is located at the amino acid positions corresponding to amino acids 587-594 of the native AAV9 capsid, or the equivalent amino acid residues in AAV1, AAV2, AAV3, AAV4, AAV5, AAV6, AAV7, AAV8, AAV10, AAV11, AAV12, AAVrh.8, AAVrh.10, AAVrh32.33, AAVrh74, bovine AAV or avian AAV, and the peptide is selected from SEQ ID NO: 19 or 20.


In some embodiments, a recombinant capsid protein comprises a) a first peptide having a sequence of any one of SEQ ID NO: 12-18; and b) a second peptide having a sequence of any one of SEQ ID NO: 19-20. In some embodiments, the first peptide is at amino acid positions 451-458, and the second peptide is at amino acids 587-594, wherein the amino acid numbering is based on the native AAV9 capsid, or the equivalent amino acid residues in AAV1, AAV2, AAV3, AAV4, AAV5, AAV6, AAV7, AAV8, AAV10, AAV11, AAV12, AAVrh.8, AAVrh.10, AAVrh32.33, AAVrh74, bovine AAV or avian AAV.


In some embodiments, the peptide inhibits binding of at least one antibody to the capsid protein. In some embodiments, the peptide inhibits neutralization of infectivity of the AAV vector by the antibody.


In some embodiments, the peptide selectively binds to a receptor expressed on the surface of a cell in the central nervous system (CNS). In some embodiments, the cell is in the premotor cortex, the thalamus, the cerebellar cortex, the dentate nucleus, the spinal cord, or the dorsal root ganglion. In some embodiments, the peptide selectively binds to a receptor expressed on the surface of a cell in the heart.


In some embodiments, an adeno-associated virus (AAV) vector comprises (i) a mutant AAV9 capsid protein and (ii) a cargo nucleic acid encapsidated by the capsid protein, wherein the capsid protein comprises a peptide having the sequence X1-X2-X3—X4-X5-X6-X7-X8 (SEQ ID NO: 158) at amino acids 451-458 of the native AAV9 capsid protein sequence, wherein the peptide does not occur in the native AAV9 capsid protein sequence. In some embodiments, X1 is not I, X2 is not N, X3 is not G, X4 is not S, X5 is not G, X6 is not Q, X7 is not N, and/or X8 is not Q. In some embodiments, X1 is S, F, Q, G, K, or R. In some embodiments, X2 is C, G, R, D, T, or Q. In some embodiments, X3 is Q, V, G, Y, R, F, or D. In some embodiments, X4 is P, Q, A, or R. In some embodiments, X5 is T, N, A, P, or I. In some embodiments, X6 is V, Q, A, or I. In some embodiments, X7 is M, P, R, Q, or N. In some embodiments, X8 is N, L, F, E, H, or A. In some embodiments, X1 is S, X2 is C, X3 is Q, X4 is P, X5 is T, X6 is V, X7 is M, and X8 is N. In some embodiments, X1 is F, X2 is G, X3 is V, X4 is P, X5 is N, X6 is Q, X7 is P, and X8 is L. In some embodiments, X1 is Q, X2 is R, X3 is G, X4 is Q, X5 is A, X6 is A, X7 is P, and X8 is F. In some embodiments, X1 is G, X2 is D, X3 is Y, X4 is A, X5 is P, X6 is I, X7 is R, and X8 is E. In some embodiments, X1 is K, X2 is T, X3 is R, X4 is R, X5 is I, X6 is V, X7 is Q, and X8 is H. In some embodiments, X1 is F, X2 is G, X3 is F, X4 is P, X5 is N, X6 is Q, X7 is P, and X8 is L. In some embodiments, X1 is R, X2 is Q, X3 is D, X4 is Q, X5 is P, X6 is I, X7 is N, and X8 is A.


In some embodiments, an adeno-associated virus (AAV) vector comprises (i) a mutant AAV9 capsid protein and (ii) a cargo nucleic acid encapsidated by the capsid protein, wherein the capsid protein comprises a peptide having the sequence X1-X2-X3—X4-X5-X6-X7-X8 (SEQ ID NO: 158) at amino acids 587-594 of the native AAV9 capsid protein sequence, wherein the peptide does not occur in the native AAV9 capsid protein sequence. In some embodiments, X1 is not A, X2 is not Q, X3 is not A, X4 is not Q, X5 is not A, X6 is not Q, X7 is not T, and/or X8 is not G. In some embodiments, X1 is S. In some embodiments, X2 is K or T. In some embodiments, X3 is V. In some embodiments, X4 is E or D. In some embodiments, X5 is S. In some embodiments, X6 is W or I. In some embodiments, X7 is T or A. In some embodiments, X8 is E or I. In some embodiments, X1 is S, X2 is K, X3 is V, X4 is E, X5 is S, X6 is W, X7 is T, and X8 is E. In some embodiments, X1 is S, X2 is T, X3 is V, X4 is D, X5 is S, X6 is I, X7 is A, and X8 is I.


In some embodiments, an adeno-associated virus (AAV) vector comprises (i) a recombinant capsid protein and (ii) a cargo nucleic acid encapsidated by the capsid protein, wherein the capsid protein comprises an amino acid sequence that is at least 95%, at least 96%, at least 97%, at least 98%, or at least 99% identical to any one of SEQ ID NO: 165-187. In some embodiments, the capsid protein comprises the amino acid sequence of any one of SEQ ID NO: 165-187. In some embodiments, the capsid protein comprises the amino acid sequence of SEQ ID NO: 175. In some embodiments, the capsid protein comprises the amino acid sequence of SEQ ID NO: 180.


In some embodiments, an AAV vector selectively delivers the cargo nucleic acid to a cell or tissue of the central nervous system. In some embodiments, the tissue of the central nervous system is the premotor cortex, the thalamus, the cerebellar cortex, the dentate nucleus, the spinal cord, or the dorsal root ganglion. In some embodiments, the AAV vector delivers the cargo nucleic acid to the brain, but does not deliver the AAV vector to the heart. In some embodiments, the AAV vector delivers the cargo nucleic acid to the brain and to the heart. In some embodiments, delivery of the cargo nucleic acid is greater to the brain than to the heart. In some embodiments, delivery of the cargo nucleic acid is approximately equal in the brain and in the heart.


AAV Capsid Proteins


In some embodiments, the disclosure provides an adeno-associated virus (AAV) capsid protein comprising one or more amino acid modifications (e.g., substitutions and/or deletions) compared to a native AAV capsid protein, wherein the one or more modifications modify one or more antigenic sites on the AAV capsid protein. The modification of the one or more antigenic sites results in inhibition of binding by an antibody to the one or more antigenic sites and/or inhibition of neutralization of infectivity of a virus particle comprising the AAV capsid protein. The one or more amino acid modifications (e.g., substitutions and/or deletions) can be in one or more antigenic footprints identified by peptide epitope mapping and/or cryo-electron microscopy studies of AAV-antibody complexes containing AAV capsid proteins. In some embodiments, the one or more antigenic sites are common antigenic motifs or CAMs as described in WO 2017/058892, which is incorporated herein by reference in its entirety. In some embodiments, the antigenic sites are in a variable region (VR) of the AAV capsid protein, such as VR-I, VR-II, VR-III, VR-IV, VR-V, VR-VI, VR-VII, VR-VIII, VR-IX. In some embodiments, one or more antigenic sites is in the HI loop of the AAV capsid protein.


In some embodiments, an AAV1, AAV2, AAV3, AAV4, AAV5, AAV6, AAV7, AAV8, AAV9, AAVrh8, AAVrh10, AAV10, AAV11, AAV12, AAVrh32.22, bovine AAV, or Avian AAV capsid protein comprises an amino acid modification (e.g., a substitution or deletion) in one or more of the regions identified in Table 5, below.









TABLE 5





Exemplary antigenic or other regions on various AAV capsids that may be partially or fully


substituted/replaced. Respective VP1 numbering of residues in the native AAV capsid


sequence is shown.
























AAV1

AAV2

AAV3

AAV4

AAV5



Sequence

Sequence

Sequence

Sequence

Sequence



(amino
SEQ
(amino
SEQ
(amino
SEQ
(amino
SEQ
(amino
SEQ


acid
ID
acid
ID
acid
ID
acid
ID
acid
ID


numbers)
NO
numbers)
NO
numbers)
NO
numbers)
NO
numbers)
NO





SASTGAS
2591
SQSGAS
2601
SQSGAS
2611
RLGESLQS
2621
EIKSGSVDGS
2631


(262-268)

(262-267)

(262-267)

(253-260)

(249-258)






VFMIPQYGYL
2592
VFMVPQYGYL
2602
VFMVPQYGYL
2612
VFMVPQYGYC
2622
VFTLPQYGYA
2632


(370-379) 

(369-378)

(369-378)

(360-369)

(360-369






NQSGSAQNK
2593
TPSGTTTQS
2603
TTSGTTNQS
2613
GTTLNAGTA
2623
STNNTGGVQ
2633


(451-459)

(450-458)

(451-459)

(445-453)

(440-448)






SV 
2594
RD 
2604
SL 
2614
SN 
2624
AN 
2634


(472-473)

(471-472)

(472-473)

(466-467

(458-459)






KTDNNNSN
2595
SADNNNSE
2605
ANDNNNSN
2615
ANQNYKIPATGS
2625
SGVNRAS
2635


(493-500)

(492-499)

(493-500)

(487-498)

(479-485)






KDDEDKF
2596
KDDEEKF
2606
KDDEEKF
2616
GPADSKF
2626
LQGSNTY
2636


(528-534)

(527-533)

(528-534)

(527-533

(515-521)






SAGASN
2597
GSEKTN
2607
GTTASN
2617
QNGNTA
2627
ANPGTTAT 
2637


(547-552)

(546-551)

(547-552)

(545-560)

(534-541)






STDPATGDVH
2598
NRQAATADVN
2608
NTAPTTGTVN
2618
SNLPTVDRLT
2628
TTAPATGTYN 
2638


(588-597)

(587-596)

(588-597)

(583-595)

(577-586)






AN 
2599
VN 
2609
VN 
2619
NS 
2629
QF 
2639


(709-710)

(708-709)

(709-710

(707-708)

(697-698






DNNGLYT
2600
DTNGVYS
2610
DTNGVYS
2620
DAAGKYT
2630
DSTGEYR
2640


(716-722) 

(715-721)

(716-722)

(714-720)

(704-710)





AAV6

AAV7

AAV8

AAV9

AAVrh8



(amino
SEQ
(amino
SEQ
(amino
SEQ
(amino
SEQ
(amino
SEQ


acid
ID
acid
ID
acid
ID
acid
ID
acid
ID


numbers
NO
numbers)
NO
numbers)
NO
numbers
NO
numbers)
NO





SASTGAS
2641
SETAGST
2651
NGTSGGAT 
2661
NSTSGGSS 
2671
NGTSGGST 
2681


(262-268)

(263-269)

(263-270)

(262-269)

(262-269)






VFMIPQYGYL
2642
VFMIPQYGYL
2652
VFMIPQYGYL
2662
VFMIPQYGYL
2672
VFMVPQYGYL 
2682


(370-379

(371-380)

(372-381)

(371-380)

(371-380)






NQSGSAQNK
2643
NPGGTAGNR 
2653
TTGGTANTQ
2663
INGSGQNQQ
2673
QTTGTGGTQ
2683


(451-459)

(453-461)

(453-461)

(451-459)

(451-459)






SV
2644
AN 
2654
AN 
2664
AV 
2674
AN 
2684


(472-473)

(474-475)

(474-475)

(472-473)

(472-473)






KTDNNNSN 
2645
LDQNNNSN
2655
TGQNNNSN
2665
VTQNNNSE
2675
TNQNNNSN
2685


(493-500)

(495-502)

(495-502)

(493-500)

(493-500)






KDDKDKF
2646
KDDEDRF
2656
KDDEERF 
2666
KEGEDRF 
2676
KDDDDRF
2686


(528-534)

(530-536)

(530-536)

(528-534)

(528-534)






SAGASN
2647
GATNKT
2657
NAARDN
2667
GTGRDN
2677
GAGNDG
2687


(547-552)

(549-554)

(549-554)

(547-552)

(547-552)






STDPATGDVH
2648
NTAAQTQVVN
2658
NTAPQIGTVNS
2668
QAQAQTGWVQ
2678
NTQAQTGLVH
2688


(588-897)

(589-598)

(590-600)

(588-597)

(588-597)






AN 
2649
TG 
2659
TS
2669
NN
2679
TN 
2689


(709-710)

(710-711)

(711-712)

(709-710)

(709-710)






DNNGLYT
2650
DSQGVYS 
2660
NTEGVYS 
2670
NTEGVYS 
2680
NTEGVYS
2690


(716-722)

(717-723)

(718-724)

(716-722)

(716-722)



AAVrh10

AAV10

AAV11

AAV12





(amino
SEQ
(amino
SEQ
(amino
SEQ
(amino
SEQ
AAVrh32.33
SEQ


acid
ID
acid
ID
acid
ID
acid
ID
(amino acid
ID


numbers)
NO
numbers)
NO
numbers)
NO
numbers
NO
numbers)
NO





NGTSGGST 
2691
NGTSGGST
2701
RLGTTSSS
2711
RIGTTANS 
2721
RLGTTSNS
2731


(263-270

(263-270

(253-260)

(262-269

(253-260)






VFMIPQYGYL
2692
VFMIPQYGYL
2702
VFMVPQYGYC
2712
VFMVPQYGYC
2722
VFMVPQYGYC
2732


(372-381)

(372-381)

(360-369)

(369-378)

(360-369)






STGGTAGTQ 
2693
STGGTQGTQ
2703
GETLNQGNA
2713
GNSLNQGTA
2723
GETLNQGNA 
2733


(453-461)

(453-461)

(444-452)

(453-461)

(444-452)






SA 
2694
SA 
2704
AF 
2714
AY 
2724
AF 
2734


(474-475)

(474-475)

(465-466)

(474-475

(465-466)






LSQNNNSN 
2695
LSQNNNSN 
2705
ASQNYKIPASGG
2715
ANQNYKIPASGG
2725
ASQNYKIPASGG
2735


(495-502)

(495-502)

(486-497)

(495-506)

(486-497






KDDEERF
2696
KDDEERF
2706
GPSDGDF 
2716
GAGDSDF
2726
GPSDGDF
2736


(530-536)

(530-536)

(526-532)

(535-541)

(526-532)






GAGKDN
2697
GAGRDN
2707
VTGNTT
2717
PSGNTT
2727
VTGNTT
2737


(549-554)

(549-554)

(544-549)

(553-558)

(544-549)






NAAPIVGAVN 
2698
NTGPIVGNVN 
2708
TTAPITGNVT 
2718
TTAPHIANLD
2728
TTAPITGNVT
2738


(590-599)

(590-599

(585-594)

(594-503)

(585-594)






TN
2699
TN 
2709
SS 
2719
NS 
2729
SS 
2739


(711-712

(711-712)

(706-707)

(715-716)

(706-707)






NTDGTYS
2700
NTEGTYS
2710
DTTGKYT
2720
DNAGNYH
2730
DTTGKYT
2740


(718-724)

(718-724)

(713-719)

(722-728)

(713-719)













Bovine AAV (amino acid numbers)
SEQ ID NO
Avian AAV (amino acid numbers)
SEQ ID NO





RLGSSNAS (255-262)
2741
RIQGPSGG (265-272)
2751





VFMVPQYGYC (362-371)
2742
IYTIPQYGYC (375-384) 
2752





GGTLNQGNS (447-455)
2743
VSQAGSSGR (454-462)
2753





SG (468-469)
2744
AA (475-476)
2754





ASQNYKIPQGRN (489-500)
2745
ASNITKNNVFSV (496-507)
2755





ANDATDF (529-535)
2746
FSGEPDR (533-539)
2756





ITGNTT (547-552)
2747
VYDQTTAT (552-559)
2757





TTVPTVDDVD (588-597)
2748
VTPGTRAAVN (595-604)
2758





DS (709-710)
2749
AD (716-717)
2759





DNAGAYK (716-722)
2750
SDTGSYS (723-729)
2760









In some embodiments, the amino acid substitution replaces any eight amino acids in an AAV capsid protein from any one of the following serotypes: AAV1, AAV2, AAV3, AAV4, AAV5, AAV6, AAV7, AAV8, AAV9, AAVrh8, AAVrh10, AAV10, AAV11, AAV12, AAVrh32.22, bovine AAV, or Avian AAV. For example, the amino acid substitution may replace the following amino acids (VP1 numbering): 355-362, 363-370, 371-378, 379-386, 387-394, 395-402, 403-410, 411-418, 419-426, 427-434, 435-442, 443-450, 451-458, 459-466, 467-474, 475-482, 483-490, 491-498, 499-506, 507-514, 515-522, 523-530, 531-538, 539-546, 547-554, 555-562, 563-570, 571-578, 579-586, 587-594, 595-602, 603-610, 611-618, 619-626, 627-634, 635-642, 643-650, 651-658, 659-666, 667-674, 675-682, 683-690, 691-698, 699-706, 707-714, 715-722 in any of the above-listed AAV serotypes.


In some embodiments, the amino acid substitution is selected from any one of SEQ ID NO: 19-20. In some embodiments, the amino acid substitution has at least 95%, at least 96%, at least 97%, at least 98%, or at least 99% sequence homology with any one of SEQ ID NO: 12-18. In some embodiments, the substitution is at the amino acids corresponding to amino acids 587-594 of the wildtype AAV9 capsid. In some embodiments, the substitution is at the amino acids corresponding to amino acids 587-594 of the wildtype AAV1 capsid. In some embodiments, the substitution is at the amino acids corresponding to amino acids 587-594 of the wildtype AAV6 capsid. In some embodiments, the substitution is at the amino acids corresponding to amino acids 589-596 of the wildtype AAV8 capsid. In some embodiments, the substitution is at the amino acids corresponding to amino acids 587-594 of the wildtype AAVrh8 capsid. In some embodiments, the substitution is at the amino acids corresponding to amino acids 589-596 of the wildtype AAVrh10 capsid.


In some embodiments, the amino acid substitution is selected from any one of SEQ ID NO: 18-20. In some embodiments, the amino acid substitution has at least 95%, at least 96%, at least 97%, at least 98%, or at least 99% sequence homology with any one of SEQ ID NO: 18-20. In some embodiments, the substitution is at the amino acids corresponding to amino acids 451-458 of the wildtype AAV9 capsid.


In some embodiments, an amino acid deletion comprises a deletion of at least one, at least two, at least three, at least four, at least five, at least six, at least seven, at least eight, at least nine, or at least ten amino acids compared to the wildtype capsid.


In some embodiments, an AAV capsid comprises one or more amino acid substitutions and one or more amino acid deletions. In some embodiments, a capsid comprises at least one amino acid substitution and at least one amino acid deletion. In some embodiments, a capsid comprises at least one amino acid substitution and at least one amino acid deletion, wherein the at least one amino acid substitution and the at least one amino acid deletion are immediately adjacent to one another in the capsid amino acid sequence.


In some embodiments, the capsid proteins are modified to produce an AAV capsid that, when present in an AAV virus particle or AAV virus vector, has a phenotype of selectively targeting the CNS (e.g., the brain, the spinal cord). In some embodiments, the capsid proteins are modified to produce an AAV capsid that, when present in an AAV virus particle or AAV virus vector, has a phenotype of evading neutralizing antibodies. The AAV virus particle or vector can also have a phenotype of enhanced or maintained transduction efficiency in addition to the phenotype of evading neutralizing antibodies and/or targeting the CNS.


In some embodiments, the one or more substitutions can introduce one or more sequences from a capsid protein of a first AAV serotype into the capsid protein of a second AAV serotype that is different from the first AAV serotype.


The base AAV capsid protein to which modifications are added can be a capsid protein of an AAV serotype selected from AAV1, AAV2, AAV3, AAV3B, AAV4, AAV5, AAV6, AAV7, AAV8, AAV9, AAV10, AAV11, AAV12, AAVrh.8, AAVrh.10, AAVrh.32.33, AAVrh74, bovine AAV, avian AAV or any other AAV now known or later identified. In some embodiments, the base AAV capsid protein is of the AAV9 serotype. In some embodiments, the base AAV capsid protein is chimeric. In some embodiments, the base AAV capsid protein is an AAV8/9 chimera.


Several examples of a modified AAV capsid protein are provided herein. In the following examples, the capsid protein can comprise the specific substitutions described and, in some embodiments, can comprise fewer or more substitutions than those described. As used herein, “substitution” may refer to a single amino acid substitution, or a substitution of more than one contiguous amino acid. For example in some embodiments, a capsid protein can comprise at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, etc., single amino acid substitutions. In some embodiments, a capsid protein can comprise one or more substitutions of multiple contiguous amino acids, such as one or more substitutions of 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, or 12 contiguous amino acids.


Furthermore, in some embodiments described herein wherein an amino acid residue is substituted by any amino acid residue other than the amino acid residue present in the wildtype or native amino acid sequence, the any other amino acid residue can be any natural or non-natural amino acid residue known in the art (see, e.g., Tables 2 and 3). In some embodiments, the substitution can be a conservative substitution and in some embodiments, the substitution can be a nonconservative substitution. In some embodiments, an AAV capsid protein comprises one or more amino acid substitutions, wherein the amino acid substitutions are each individually selected from SEQ ID NO: 12-18 as shown in Table 6.1.









TABLE 6.1







AMINO ACID SUBSTITUTIONS










Amino Acid Substitution
SEQ ID NO.







SCQPTVMN
12







FGVPNQPL
13







QRGQAAPF
14







GDYAPIRE
15







KTRRIVQH
16







FGFPNQPL
17







RQDQPINA
18










In some embodiments, an AAV capsid protein comprises one or more amino acid substitutions, wherein the amino acid substitutions are each selected from SEQ ID NO: 19-20 as shown in Table 6.2.









TABLE 6.2







AMINO ACID SUBSTITUTIONS










Amino Acid Substitution
SEQ ID NO.







SKVESWTE
19







STVDSIAI
20










In some embodiments, an AAV capsid protein may comprise a first substitution selected from the sequences listed in Table 6.1 and a second substitution selected from the sequences listed in Table 6.2. In some embodiments, an AAV capsid protein may comprise a first substitution, a second substitution as shown in Tables 6.3 and 6.4.









TABLE 6.3







COMBINATIONS OF AMINO ACID SUBSTITUTIONS










First Substitution
Second Substitution



(SEQ ID NO)
(SEQ ID NO)







12, 13 , 14 , 15 , 16, 17, or 18
19 or 20

















TABLE 6.4







COMBINATIONS OF AMINO ACID SUBSTITUTIONS










First Substitution
Second Substitution



(SEQ ID NO)
(SEQ ID NO)






12
19



12
20



13
19



13
20



14
19



14
20



15
19



15
20



16
19



16
20



17
19



17
20



18
19



18
20









In some embodiments, an AAV capsid protein comprises an amino acid modification (e.g., substitution and/or deletion), wherein the amino acid modification modifies one or more surface-exposed regions, such as an antigenic region, on the AAV capsid protein.


In some embodiments, an AAV capsid protein comprises one or more amino acid substitutions, wherein at least one of the amino acid substitutions comprises one of SEQ ID NOs: 19-20. In some embodiments, the substitution replaces the amino acids corresponding to amino acids 587-594 of the wildtype AAV9 capsid.


In some embodiments, an AAV capsid protein comprises one or more amino acid substitutions, wherein at least one of the amino acid substitutions comprises one of SEQ ID NOs: 12-18. In some embodiments, the substitution replaces the amino acids corresponding to amino acids 451-458 of the wildtype AAV9 capsid.


In some embodiments, an AAV capsid protein comprises a substitution comprising a sequence of eight amino acids (X1—X2-X3-X4-X5-X6-X7-X8) (SEQ ID NO: 158) that does not occur in the native capsid protein sequence. In some embodiments, X1 is not I, X2 is not N, X3 is not G, X4 is not S, X5 is not G, X6 is not Q, X7 is not N, and/or X8 is not Q. In some embodiments, X1 is S, F, Q, G, K, or R. In some embodiments, X2 is C, G, R, D, T, or Q. In some embodiments, X3 is Q, V, G, Y, R, F, or D. In some embodiments, X4 is P, Q, A, or R. In some embodiments, X5 is T, N, A, P, or I. In some embodiments, X6 is V, Q, A, or I. In some embodiments, X7 is M, P, R, Q, or N. In some embodiments, X8 is N, L, F, E, H, or A. In some embodiments, X1 is S, X2 is C, X3 is Q, X4 is P, X5 is T, X6 is V, X7 is M, and X8 is N. In some embodiments, X1 is F, X2 is G, X3 is V, X4 is P, X5 is N, X6 is Q, X7 is P, and X8 is L. In some embodiments, X1 is Q, X2 is R, X3 is G, X4 is Q, X5 is A, X6 is A, X7 is P, and X8 is F. In some embodiments, X1 is G, X2 is D, X3 is Y, X4 is A, X5 is P, X6 is I, X7 is R, and X8 is E. In some embodiments, X1 is K, X2 is T, X3 is R, X4 is R, X5 is I, X6 is V, X7 is Q, and X8 is H. In some embodiments, X1 is F, X2 is G, X3 is F, X4 is P, X5 is N, X6 is Q, X7 is P, and X8 is L. In some embodiments, X1 is R, X2 is Q, X3 is D, X4 is Q, X5 is P, X6 is I, X7 is N, and X8 is A.


In some embodiments, X1 is not A, X2 is not Q, X3 is not A, X4 is not Q, X5 is not A, X6 is not Q, X7 is not T, and/or X8 is not G. In some embodiments, X1 is S. In some embodiments, X2 is K or T. In some embodiments, X3 is V. In some embodiments, X4 is E or D. In some embodiments, X5 is S. In some embodiments, X6 is W or I. In some embodiments, X7 is T or A. In some embodiments, X8 is E or I. In some embodiments, X1 is S, X2 is K, X3 is V, X4 is E, X5 is S, X6 is W, X7 is T, and X8 is E. In some embodiments, X1 is S, X2 is T, X3 is V, X4 is D, X5 is S, X6 is I, X7 is A, and X8 is I.


In some embodiments, an AAV capsid protein comprises one or more amino acid deletions, wherein the amino acid deletion comprises a deletion of at least six or at least eight amino acids compared to the wildtype AAV capsid. In some embodiments, an AAV capsid protein comprises a deletion of eight consecutive amino acids compared to the native capsid protein sequence. In some embodiments, an AAV capsid protein comprises a deletion of six consecutive amino acids compared to the native capsid protein sequence.


In some embodiments, an AAV capsid protein comprises the sequence LSKTQTLK (SEQ ID NO: 1374) or the sequence LSKTDPQTLK (SEQ ID NO: 1375). In some embodiments, the AAV capsid protein comprising SEQ ID NO: 1374 or 1375 is of a serotype selected from AAV1, AAV2, AAV3, AAV4, AAV5, AAV6, AAV7, AAV8, AAV9, AAV10, AAV11, AAV12, AAVrh8, AAVrh10, AAVrh32.33, AAVrh74, Avian AAV and Bovine AAV.


In some embodiments, an AAV capsid protein comprises a first substitution comprising a sequence selected from SEQ ID NO: 12-18; and a second substitution comprising a sequence selected from SEQ ID NO: 19-20.


In some embodiments, an AAV capsid protein comprises an amino acid deletion and a substitution, wherein the substitution comprises a sequence selected from SEQ ID NO: 12-20.


In some embodiments, a recombinant capsid protein has a sequence that is at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99% or 100% identical to SEQ ID NO: 9 (AAV9) and comprises one or more of the following amino acid substitutions: 1451S, 1451F, 1451Q, 1451G, 1451K, 1451R, N452C, N452G, N452R, N452D, N452T, N452Q, G453Q, G453V, G453Y, G453R, G453F, G453D, S454P, S454Q, S454A, S454R, G455T, G455N, G455A, G455P, G4551, Q456V, Q456A, Q4561, N457M, N457P, N457R, N457Q, Q458N, Q458L, Q458F, Q458E, Q458H, Q458A, A587S, Q588K, Q588T, A589V, Q590E, Q590D, A591 S, Q592W, Q5921, T593A, G594E, G5941.


Any of the AAV capsids described herein may further comprise a modification (e.g., a substitution or a deletion) in the HI loop. The HI loop is a prominent domain on the AAV capsid surface, between β strands βH and βI, that extends from each viral protein (VP) subunit overlapping the neighboring fivefold VP. In some embodiments, an AAV capsid comprises one, two, three, four, five, six, seven, or eight amino acid substitutions in the HI loop. In some embodiments, the AAV capsid comprises one or more of the following substitutions in the HI loop: P661R, T662S, Q666G, S667D, wherein the numbering corresponds to the wildtype AAV8 capsid (SEQ ID NO: 8). In some embodiments, the AAV capsid comprises one or more of the following substitutions in the HI loop: P659R, T660S, A661T, K664G, wherein the numbering corresponds to the wildtype AAV9 capsid (SEQ ID NO: 9).


In some embodiments, an AAV capsid protein comprises one, two, three, or four amino acid substitutions, wherein each substitution modifies a different antigenic site on the AAV capsid protein, and wherein at least one of the amino acid substitutions modifies the HI loop of the capsid protein.


In some embodiments, an AAV capsid protein comprises a first, a second, a third, and a fourth amino acid substitution. In some embodiments, at least one of the substitutions modifies the HI Loop of the capsid protein. In some embodiments, the AAV capsid comprises one or more of the following substitutions in the HI loop: P661R, T662S, Q666G, S667D, wherein the numbering corresponds to the wildtype AAV8 capsid (SEQ ID NO: 8); or P659R, T660S, A661T, K664G, wherein the numbering corresponds to the wildtype AAV9 capsid (SEQ ID NO: 9). In some embodiments, an AAV capsid protein comprises the amino acid sequence of any one of SEQ ID NO: 185-187. In some embodiments, an AAV capsid protein comprises an amino acid sequence sharing at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99% sequence identity to any one of SEQ ID NO: 165-187.


Also provided herein is a nucleotide sequence, or an expression vector comprising the same that encodes one or more of the AAV capsid proteins described herein. The nucleotide sequence may be a DNA sequence or an RNA sequence. In some embodiments, cell comprises one or more nucleotide sequences or expression vectors described herein.


In some embodiments, an AAV capsid comprises an AAV capsid protein as described herein. Further provided herein is a viral vector comprising an AAV capsid as well as a composition comprising the AAV capsid protein, AAV capsid and/or viral vector in a pharmaceutically acceptable carrier.


In some embodiments, modification of one or more antigenic sites results in inhibition of binding by an antibody to the one or more antigenic sites. In some embodiments, modification of the one or more antigenic sites results in inhibition of neutralization of infectivity of a virus particle comprising the AAV capsid protein.


As described herein, the nucleic acid and amino acid sequences of the capsid proteins from a number of AAV are known in the art. Thus, the amino acids “corresponding” to amino acid positions of the native AAV capsid protein can be readily determined for any other AAV (e.g., by using sequence alignments).


The modified capsid proteins can be produced by modifying the capsid protein of any AAV now known or later discovered. Further, the base AAV capsid protein that is to be modified can be a naturally occurring AAV capsid protein (e.g., an AAV2, AAV3a or 3b, AAV4, AAV5, AAV6, AAV7, AAV8, AAV9, AAV10 or AAV11 capsid protein or any of the AAV shown in Table 2) but is not so limited. Those skilled in the art will understand that a variety of manipulations to the AAV capsid proteins are known in the art and the disclosure is not limited to modifications of naturally occurring AAV capsid proteins. For example, the capsid protein to be modified may already have alterations as compared with naturally occurring AAV (e.g., is derived from a naturally occurring AAV capsid protein, e.g., AAV2, AAV3a, AAV3b, AAV4, AAV5, AAV6, AAV7, AAV8, AAV9, AAV10, AAV11, AAV12 or any other AAV now known or later discovered). In some embodiments, the capsid protein may be a chimeric capsid protein. In some embodiments, the capsid protein may be an engineered AAV, such as AAV2i8, AAV2g9, AAV-LK03, AAV7m8, AAV Anc80, AAV PHP.B.


Thus, in some embodiments, the AAV capsid protein to be modified can be derived from a naturally occurring AAV but further comprises one or more foreign sequences (e.g., that are exogenous to the native virus) that are inserted and/or substituted into the capsid protein and/or has been altered by deletion of one or more amino acids.


Accordingly, when referring herein to a specific AAV capsid protein (e.g., an AAV2, AAV3, AAV4, AAV5, AAV6, AAV7, AAV8, AAV9, AAV10 or AAV11 capsid protein or a capsid protein from any of the AAV shown in Table 2, etc.), it is intended to encompass the native capsid protein as well as capsid proteins that have alterations other than the modifications described herein. Such alterations include substitutions, insertions and/or deletions. In some embodiments, the capsid protein comprises 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19 or 20, less than 20, less than 30, less than 40, less than 50, less than 60, or less than 70 amino acids inserted therein (other than the insertions described herein) as compared with the native AAV capsid protein sequence. In some embodiments, the capsid protein comprises 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19 or 20, less than 20, less than 30, less than 40, less than 50, less than 60, or less than 70 amino acid substitutions (other than the amino acid substitutions described herein) as compared with the native AAV capsid protein sequence, in some embodiments, the capsid protein comprises a deletion of 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19 or 20, less than 20, less than 30, less than 40, less than 50, less than 60, or less than 70 amino acids as compared with the native AAV capsid protein sequence.


In some embodiments, the AAV capsid protein has an amino acid sequence that is at least about 90%, about 95%, about 97%, about 98% or about 99% similar or identical to a native AAV capsid protein sequence.


Methods of determining sequence similarity or identity between two or more amino acid sequences are known in the art. Sequence similarity or identity may be determined using standard techniques, including, but not limited to, the local sequence identity algorithm of Smith & Waterman, Adv. Appl. Math. 2, 482 (1981), by the sequence identity alignment algorithm of Needleman & Wunsch, J Mol. Biol. 48,443 (1970), by the search for similarity method of Pearson & Lipman, Proc. Natl. Acad. Sci. USA 85, 2444 (1988), by computerized implementations of these algorithms (GAP, BESTFIT, FASTA, and TFASTA in the Wisconsin Genetics Software Package, Genetics Computer Group, 575 Science Drive, Madison, WI), the Best Fit sequence program described by Devereux et al., Nucl. Acid Res. 12, 387-395 (1984), or by inspection.


Another suitable algorithm is the BLAST algorithm, described in Altschul et al., J Mol. Biol. 215, 403-410, (1990) and Karlin et al., Proc. Natl. Acad. Sci. USA 90, 5873-5787 (1993). A particularly useful BLAST program is the WU-BLAST-2 program which was obtained from Altschul et al., Methods in Enzymology, 266, 460-480 (1996); http://blast.wustl/edu/blast/README.html. WU-BLAST-2 uses several search parameters, which are optionally set to the default values. The parameters are dynamic values and are established by the program itself depending upon the composition of the particular sequence and composition of the particular database against which the sequence of interest is being searched; however, the values may be adjusted to increase sensitivity.


Further, an additional useful algorithm is gapped BLAST as reported by Altschul et al, (1997) Nucleic Acids Res. 25, 3389-3402.


In some embodiments, a virus capsid comprises a modified AAV capsid protein as described herein. In some embodiments, the virus capsid is a parvovirus capsid, which may further be an autonomous parvovirus capsid or a dependovirus capsid. Optionally, the virus capsid is an AAV capsid. In some embodiments, the AAV capsid is an AAV1, AAV2, AAV3a, AAV3b, AAV4, AAV5, AAV6, AAV7, AAV8, AAV9, AAV10, AAV11, AAV12, AAVrh8, AAVrh10, AAVrh32.33, bovine AAV capsid, avian AAV capsid or any other AAV now known or later identified. A nonlimiting list of AAV serotypes is shown in Table 2. An AAV capsid can be any AAV serotype listed in Table 2 or derived from any of the foregoing by one or more insertions, substitutions and/or deletions. Molecules that can be packaged by the modified virus capsid and transferred into a cell include cargo nucleic acids (e.g., heterologous DNA or RNA), polypeptides, small organic molecules, metals, or combinations of the same.


Heterologous molecules are defined as those that are not naturally found in an AAV infection, e.g., those not encoded by a wild-type AAV genome. Further, therapeutically useful molecules can be associated with the outside of the chimeric virus capsid for transfer of the molecules into host target cells. Such associated molecules can include DNA, RNA, small organic molecules, metals, carbohydrates, lipids and/or polypeptides. In some embodiments the therapeutically useful molecule is covalently linked (i.e., conjugated or chemically coupled) to the capsid proteins. Methods of covalently linking molecules are known by those skilled in the art.


The modified virus capsids also find use in raising antibodies against the novel capsid structures. As a further alternative, an exogenous amino acid sequence may be inserted into the modified virus capsid for antigen presentation to a cell, e.g., for administration to a subject to produce an immune response to the exogenous amino acid sequence.


In some embodiments, the virus capsids can be administered to block certain cellular sites prior to and/or concurrently with (e.g., within minutes or hours of each other) administration of a virus vector delivering a nucleic acid encoding a polypeptide or functional RNA of interest. For example, the inventive capsids can be delivered to block cellular receptors on liver cells and a delivery vector can be administered subsequently or concurrently, which may reduce transduction of liver cells, and enhance transduction of other targets (e.g., skeletal, cardiac and/or diaphragm muscle).


According to some embodiments, modified virus capsids can be administered to a subject prior to and/or concurrently with a modified virus vector as described herein. Further, the disclosure provides compositions and pharmaceutical formulations comprising the inventive modified virus capsids; optionally, the composition also comprises a modified virus vector as described herein.


In some embodiments, a nucleic acid (optionally, an isolated nucleic acid) encodes the modified virus capsids and capsid proteins described herein. Further provided are vectors comprising the nucleic acids, and cells (in vivo or in culture) comprising the nucleic acids and/or vectors described herein. As one example, a virus vector may comprise: (a) a modified AAV capsid as described herein; and (b) a nucleic acid comprising at least one terminal repeat sequence, wherein the nucleic acid is encapsidated by the AAV capsid.


Other suitable vectors include without limitation viral vectors (e.g., adenovirus, AAV, herpesvirus, vaccinia, poxviruses, baculovirus, lentivirus, coronavirus, and the like), plasmids, phage, YACs, BACs, and the like. Such nucleic acids, vectors and cells can be used, for example, as reagents (e.g., helper packaging constructs or packaging cells) for the production of modified virus capsids or virus vectors as described herein.


Virus capsids described herein can be produced using any method known in the art, e.g., by using a baculovirus system (Brown et al., (1994) Virology 198:477-488).


The modifications to the AAV capsid protein as described herein are “selective” modifications. This approach is in contrast to previous work with whole subunit or large domain swaps between AAV serotypes (see, e.g., international patent publication WO 00/28004 and Hauck et al., (2003) J. Virology 77:2768-2774). In some embodiments, a “selective” modification results in the insertion and/or substitution and/or deletion of less than or equal to about 20, 18, 15, 12, 10, 9, 8, 7, 6, 5, 4 or 3 contiguous amino acids.


The modified capsid proteins and capsids described herein can further comprise any other modification, now known or later identified. For example, the AAV capsid proteins and virus capsids can be chimeric in that they can comprise all or a portion of a capsid subunit from another virus, optionally another parvovirus or AAV, e.g., as described in international patent publication WO 00/28004.


In some embodiments, the virus capsid can be a targeted virus capsid, comprising a targeting sequence (e.g., substituted or inserted in the viral capsid) that directs the virus capsid to interact with cell-surface molecules present on desired target tissue(s) (see, e.g., International patent publication WO 00/28004 and Hauck et al., (2003) J. Virology 77:2768-2774); Shi et al., Human Gene Therapy 17:353-361 (2006) [describing insertion of the integrin receptor binding motif RGD at positions 520 and/or 584 of the AAV capsid subunit]; and U.S. Pat. No. 7,314,912 [describing insertion of the PI peptide containing an RGD motif following amino acid positions 447, 534, 573 and 587 of the AAV2 capsid subunit]). Other positions within the AAV capsid subunit that tolerate insertions are known in the art (e.g., positions 449 and 588 described by Grifman et al., Molecular Therapy 3:964-975 (2001)).


For example, a virus capsid as described herein may have relatively inefficient tropism toward certain target tissues of interest (e.g., liver, skeletal muscle, heart, diaphragm muscle, kidney, brain, stomach, intestines, skin, endothelial cells, and/or lungs). A targeting sequence can advantageously be incorporated into these low-transduction vectors to thereby confer to the virus capsid a desired tropism and, optionally, selective tropism for particular tissue(s). AAV capsid proteins, capsids and vectors comprising targeting sequences are described, for example in international patent publication WO 00/28004. As another example, one or more non-naturally occurring amino acids as described by Wang et al., Annu Rev Biophys Biomol Struct. 35:225-49 (2006)) can be incorporated into an AAV capsid subunit as described herein at an orthogonal site as a means of redirecting a low-transduction vector to desired target tissue(s). These unnatural amino acids can advantageously be used to chemically link molecules of interest to the AAV capsid protein including without limitation: glycans (mannose—dendritic cell targeting); RGD, bombesin or a neuropeptide for targeted delivery to specific cancer cell types; RNA aptamers or peptides selected from phage display targeted to specific cell surface receptors such as growth factor receptors, integrins, and the like.


In some embodiments, the targeting sequence may be a virus capsid sequence (e.g., an autonomous parvovirus capsid sequence, AAV capsid sequence, or any other viral capsid sequence) that directs infection to a particular cell type(s).


As another nonlimiting example, a heparin or heparan sulfate binding domain (e.g., the respiratory syncytial virus heparin binding domain) may be inserted or substituted into a capsid subunit that does not typically bind HS receptors (e.g., AAV4, AAV5) to confer heparin and/or heparan sulfate binding to the resulting mutant.


B19 infects primary erythroid progenitor cells using globoside as its receptor (Brown et al, (1993) Science 262: 114). The structure of B19 has been determined to 8 Å resolution (Agbandje-McKenna et al, (1994) Virology 203: 106). The region of the B19 capsid that binds to globoside has been mapped between amino acids 399-406 (Chapman et al, (1993) Virology 194:419), a looped out region between β-barrel structures E and F (Chipman et al, (1996) Proc. Nat. Acad. Sci. USA 93:7502). Accordingly, the globoside receptor binding domain of the B19 capsid may be substituted into an AAV capsid protein to target a virus capsid or virus vector comprising the same to erythroid cells.


In some embodiments, the exogenous targeting sequence may be any amino acid sequence encoding a peptide that alters the tropism of a virus capsid or virus vector comprising the modified AAV capsid protein. In some embodiments, the targeting peptide or protein may be naturally occurring or, alternately, completely or partially synthetic. Exemplary targeting sequences include ligands and other peptides that bind to cell surface receptors and glycoproteins, such as ROD peptide sequences, bradykinin, hormones, peptide growth factors (e.g., epidermal growth factor, nerve growth factor, fibroblast growth factor, platelet-derived growth factor, insulin-like growth factors I and II, etc.), cytokines, melanocyte stimulating hormone (e.g., α, β or γ), neuropeptides and endorphins, and the like, and fragments thereof that retain the ability to target cells to their cognate receptors. Other illustrative peptides and proteins include substance P, keratinocyte growth factor, neuropeptide Y, gastrin releasing peptide, interleukin 2, hen egg white lysozyme, erythropoietin, gonadolibcrin, corticostatin, β-endorphin, leu-enkephalin, rimorphin, alpha-neo-enkephalin, angiotensin, pneumadin, vasoactive intestinal peptide, neurotensin, motilin, and fragments thereof as described above. As yet a further alternative, the binding domain from a toxin (e.g., tetanus toxin or snake toxins, such as alpha-bungarotoxin, and the like) can be substituted into the capsid protein as a targeting sequence. In some embodiments, the AAV capsid protein can be modified by substitution of a “nonclassical” import/export signal peptide (e.g., fibroblast growth factor-1 and -2, interleukin 1, HIV-1 Tat protein, herpes virus VP22 protein, and the like) as described by Cleves (Current Biology 7:R318 (1997)) into the AAV capsid protein. Also encompassed are peptide motifs that direct uptake by specific cells, e.g., a FVFLP (SEQ ID NO: 22) peptide motif triggers uptake by liver cells.


Phage display techniques, as well as other techniques known in the art, may be used to identify peptides that recognize any cell type of interest.


The targeting sequence may encode any peptide that targets to a cell surface binding site, including receptors (e.g., protein, carbohydrate, glycoprotein or proteoglycan). Examples of cell surface binding sites include, but are not limited to, heparan sulfate, chondroitin sulfate, and other glycosaminoglycans, sialic acid moieties found on mucins, glycoproteins, and gangliosides, MHC 1 glycoproteins, carbohydrate components found on membrane glycoproteins, including, mannose, N-acetyl-galactosamine, N-acetyl-glucosamine, fucose, galactose, and the like.


In some embodiments, a heparan sulfate (HS) or heparin binding domain is substituted into the virus capsid (for example, in an AAV capsid that otherwise does not bind to HS or heparin). It is known in the art that HS/heparin binding is mediated by a “basic patch” that is rich in arginines and/or lysines. In some embodiments, a sequence following the motif BXXB (SEQ ID NO: 23), where “B” is a basic residue and X is neutral and/or hydrophobic residue can be employed. As a nonlimiting example, BXXB can be RGNR (SEQ ID NO: 24). As another nonlimiting example, BXXB is substituted for amino acid positions 262 through 265 in the native AAV2 capsid protein or at the corresponding position(s) in the capsid protein of another AAV serotype.


Table 7 shows other non-limiting examples of suitable targeting sequences.









TABLE 7







TARGETING SEQUENCES










SEQ ID



Sequence
NO
Reference












NSVRDL(G/S)
25
Muller et al., Nature Biotechnology 21: 1040-1046




(2003)





PRSVTVP
26
Muller et al., Nature Biotechnology 21: 1040-1046




(2003)





NSVSSX(S/A)
27
Muller et al., Nature Biotechnology 21: 1040-1046




(2003)





NGR, NGRAHA
28
Grifman et al., Molecular Therapy 3: 964-975 (2001)





QPEHSST
29
Work et al., Molecular Therapy 13: 683-693 (2006)





VNTANST
30
Work et al., Molecular Therapy 13: 683-693 (2006)





HGPMQS
31
Work et al., Molecular Therapy 13: 683-693 (2006)





PHKPPLA
32
Work et al., Molecular Therapy 13: 683-693 (2006)





IKNNEMW
33
Work et al., Molecular Therapy 13: 683-693 (2006)





RNLDTPM
34
Work et al., Molecular Therapy 13: 683-693 (2006)





VDSHRQS
35
Work et al., Molecular Therapy 13: 683-693 (2006)





YDSKTKT
36
Work et al., Molecular Therapy 13: 683-693 (2006)





SQLPHQK
37
Work et al., Molecular Therapy 13: 683-693 (2006)





STMQQNT
38
Work et al., Molecular Therapy 13: 683-693 (2006)





TERYMTQ
39
Work et al., Molecular Therapy 13: 683-693 (2006)





QPEHSST
40
Work et al., Molecular Therapy 13: 683-693 (2006)





DASLSTS
41
Work et al., Molecular Therapy 13: 683-693 (2006)





DLPNKT
42
Work et al., Molecular Therapy 13: 683-693 (2006)





DLTAARL
43
Work et al., Molecular Therapy 13: 683-693 (2006)





EPHQFNY
44
Work et al., Molecular Therapy 13: 683-693 (2006)





EPQSNHT
45
Work et al., Molecular Therapy 13: 683-693 (2006)





MSSWPSQ
46
Work et al., Molecular Therapy 13: 683-693 (2006)





NPKHNAT
47
Work et al., Molecular Therapy 13: 683-693 (2006)





PDGMRTT
48
Work et al., Molecular Therapy 13: 683-693 (2006)





PNNNKTT
49
Work et al., Molecular Therapy 13: 683-693 (2006)





QSTTHDS
50
Work et al., Molecular Therapy 13: 683-693 (2006)





TGSKQKQ
51
Work et al., Molecular Therapy 13: 683-693 (2006)





SLKHQAL
52
Work et al., Molecular Therapy 13: 683-693 (2006)





SPIDGEQ
53
Work et al., Molecular Therapy 13: 683-693 (2006)





WIFPWIQL
54
Hajitou et al., TCM 16: 80-88 (2006)





CDCRGDCFC
55
Hajitou et al., TCM 16: 80-88 (2006)





CNGRC
56
Hajitou et al., TCM 16: 80-88 (2006)





CPRECES
57
Hajitou et al., TCM 16: 80-88 (2006)





CTTHWGFTLC
58
Hajitou et al., TCM 16: 80-88 (2006)





CGRRAGGSC
59
Hajitou et al., TCM 16: 80-88 (2006)





CKGGRAKDC
60
Hajitou et al., TCM 16: 80-88 (2006)





CVPELGHEC
61
Hajitou et al., TCM 16: 80-88 (2006)





CRRETAWAK
62
Koivunen et al., J. Nucl. Med. 40: 883-888 (1999)





VSWFSHRYSPFAVS
63
Koivunen et al., J. Nucl. Med. 40: 883-888 (1999)





GYRDGYAGPILYN
64
Koivunen et al., J. Nucl. Med. 40: 883-888 (1999)





XXXY*XXX
65
Koivunen et al., J. Nucl. Med. 40: 883-888 (1999)





Y*E/MNW
66
Koivunen et al., J. Nucl. Med. 40: 883-888 (1999)





RPLPPLP
67
Koivunen et al., J. Nucl. Med. 40: 883-888 (1999)





APPLPPR
68
Koivunen et al., J. Nucl. Med. 40: 883-888 (1999)





DVFYPYPYASGS
69
Koivunen et al., J. Nucl. Med. 40: 883-888 (1999)





MYWYPY
70
Koivunen et al., J. Nucl. Med. 40: 883-888 (1999)





DITWDQLWDLMK
71
Koivunen et al., J. Nucl. Med. 40: 883-888 (1999)





CWDD(G/L)WLC
72
Koivunen et al., J. Nucl. Med. 40: 883-888 (1999)





EWCEYLGGYLRCYA
73
Koivunen et al., J. Nucl. Med. 40: 883-888 (1999)





YXCXXGPXTWXCXP
74
Koivunen et al., J. Nucl. Med. 40: 883-888 (1999)





IEGPTLRQWLAARA
75
Koivunen et al., J. Nucl. Med. 40: 883-888 (1999)





LWXX(Y/W/F/H)
76
Koivunen et al., J. Nucl. Med. 40: 883-888 (1999)





XFXXYLW
77
Koivunen et al., J. Nucl. Med. 40: 883-888 (1999)





RWGLCD
78
Koivunen et al., J. Nucl. Med. 40: 883-888 (1999)





MSRPACPPNDKYE
79
Koivunen et al., J. Nucl. Med. 40: 883-888 (1999)





CLRSGRGC
80
Koivunen et al., J. Nucl. Med. 40: 883-888 (1999)





CHWMFSPWC
81
Koivunen et al., J. Nucl. Med. 40: 883-888 (1999)





WXXF
82
Koivunen et al., J. Nucl. Med. 40: 883-888 (1999)





CSSRLDAC
83
Koivunen et al., J. Nucl. Med. 40: 883-888 (1999)





CLPVASC
84
Koivunen et al., J. Nucl. Med. 40: 883-888 (1999)





CGFECVRQCPERC
85
Koivunen et al., J. Nucl. Med. 40: 883-888 (1999)





CVALCREACGEGC
86
Koivunen et al., J. Nucl. Med. 40: 883-888 (1999)





SWCEPGWCR
87
Koivunen et al., J. Nucl. Med. 40: 883-888 (1999)





YSGWGW
88
Koivunen et al., J. Nucl. Med. 40: 883-888 (1999)





GLSGGRS
89
Koivunen et al., J. Nucl. Med. 40: 883-888 (1999)





LMLPRAD
90
Koivunen et al., J. Nucl. Med. 40: 883-888 (1999)





CSCFRDVCC
91
Koivunen et al., J. Nucl. Med. 40: 883-888 (1999)





CRDVVSVIC
92
Koivunen et al., J. Nucl. Med. 40: 883-888 (1999)





CNGRC
93
Koivunen et al., J. Nucl. Med. 40: 883-888 (1999)





MARSGL
94
Newton & Deutscher, Phage Peptide Display in




Handbook of Experimental Pharmacology, pages




145-163, Springer-Verlag, Berlin (2008)





MARAKE
95
Newton & Deutscher, Phage Peptide Display in




Handbook of Experimental Pharmacology, pages




145-163, Springer-Verlag, Berlin (2008)





MSRTMS
96
Newton & Deutscher, Phage Peptide Display in




Handbook of Experimental Pharmacology, pages




145-163, Springer-Verlag, Berlin (2008)





KCCYSL
97
Newton & Deutscher, Phage Peptide Display in




Handbook of Experimental Pharmacology, pages




145-163, Springer-Verlag, Berlin (2008)





MYWGDSHWLQYWYE
98
Newton & Deutscher, Phage Peptide Display in




Handbook of Experimental Pharmacology, pages




145-163, Springer-Verlag, Berlin (2008)





MQLPLAT
99
Newton & Deutscher, Phage Peptide Display in




Handbook of Experimental Pharmacology, pages




145-163, Springer-Verlag, Berlin (2008)





EWLS
100
Newton & Deutscher, Phage Peptide Display in




Handbook of Experimental Pharmacology, pages




145-163, Springer-Verlag, Berlin (2008)





SNEW
101
Newton & Deutscher, Phage Peptide Display in




Handbook of Experimental Pharmacology, pages




145-163, Springer-Verlag, Berlin (2008)





TNYL
102
Newton & Deutscher, Phage Peptide Display in




Handbook of Experimental Pharmacology, pages




145-163, Springer-Verlag, Berlin (2008)





WIFPWIQL
103
Newton & Deutscher, Phage Peptide Display in




Handbook of Experimental Pharmacology, pages




145-163, Springer-Verlag, Berlin (2008)





WDLAWMFRLPVG
104
Newton & Deutscher, Phage Peptide Display in




Handbook of Experimental Pharmacology, pages




145-163, Springer-Verlag, Berlin (2008)





CTVALPGGYVRVC
105
Newton & Deutscher, Phage Peptide Display in




Handbook of Experimental Pharmacology, pages




145-163, Springer-Verlag, Berlin (2008)





CVPELGHEC
106
Newton & Deutscher, Phage Peptide Display in




Handbook of Experimental Pharmacology, pages




145-163, Springer-Verlag, Berlin (2008)





CGRRAGGSC
107
Newton & Deutscher, Phage Peptide Display in




Handbook of Experimental Pharmacology, pages




145-163, Springer-Verlag, Berlin (2008)





CVAYCIEHHCWTC
108
Newton & Deutscher, Phage Peptide Display in




Handbook of Experimental Pharmacology, pages




145-163, Springer-Verlag, Berlin (2008)





CVFAHNYDYLVC
109
Newton & Deutscher, Phage Peptide Display in




Handbook of Experimental Pharmacology, pages




145-163, Springer-Verlag, Berlin (2008)





CVFTSNYAFC
110
Newton & Deutscher, Phage Peptide Display in




Handbook of Experimental Pharmacology, pages




145-163, Springer-Verlag, Berlin (2008)





VHSPNKK
111
Newton & Deutscher, Phage Peptide Display in




Handbook of Experimental Pharmacology, pages




145-163, Springer-Verlag, Berlin (2008)





CDCRGDCFC
112
Newton & Deutscher, Phage Peptide Display in




Handbook of Experimental Pharmacology, pages




145-163, Springer-Verlag, Berlin (2008)





CRGDGWC
113
Newton & Deutscher, Phage Peptide Display in




Handbook of Experimental Pharmacology, pages




145-163, Springer-Verlag, Berlin (2008)





XRGCDX
114
Newton & Deutscher, Phage Peptide Display in




Handbook of Experimental Pharmacology, pages




145-163, Springer-Verlag, Berlin (2008)





PXX(S/T)
115
Newton & Deutscher, Phage Peptide Display in




Handbook of Experimental Pharmacology, pages




145-163, Springer-Verlag, Berlin (2008)





CTTHWGFTLC
116
Newton & Deutscher, Phage Peptide Display in




Handbook of Experimental Pharmacology, pages




145-163, Springer-Verlag, Berlin (2008)





SGKGPRQITAL
117
Newton & Deutscher, Phage Peptide Display in




Handbook of Experimental Pharmacology, pages




145-163, Springer-Verlag, Berlin (2008)





A(A/Q)(N/A)(L/Y)
118
Newton & Deutscher, Phage Peptide Display in


(T/V/M/R)(R/K)

Handbook of Experimental Pharmacology, pages




145-163, Springer-Verlag, Berlin (2008)





VYMSPF
119
Newton & Deutscher, Phage Peptide Display in




Handbook of Experimental Pharmacology, pages




145-163, Springer-Verlag, Berlin (2008)





MQLPLAT
120
Newton & Deutscher, Phage Peptide Display in




Handbook of Experimental Pharmacology, pages




145-163, Springer-Verlag, Berlin (2008)





ATWLPPR
121
Newton & Deutscher, Phage Peptide Display in




Handbook of Experimental Pharmacology, pages




145-163, Springer-Verlag, Berlin (2008)





HTMYYHHYQHHL
122
Newton & Deutscher, Phage Peptide Display in




Handbook of Experimental Pharmacology, pages




145-163, Springer-Verlag, Berlin (2008)





SEVGCRAGPLQWLCEKYFG
123
Newton & Deutscher, Phage Peptide Display in




Handbook of Experimental Pharmacology, pages




145-163, Springer-Verlag, Berlin (2008)





CGLLPVGRPDRNVWRWLC
124
Newton & Deutscher, Phage Peptide Display in




Handbook of Experimental Pharmacology, pages




145-163, Springer-Verlag, Berlin (2008)





CKGQCDRFKGLPWEC
125
Newton & Deutscher, Phage Peptide Display in




Handbook of Experimental Pharmacology, pages




145-163, Springer-Verlag, Berlin (2008)





SGRSA
126
Newton & Deutscher, Phage Peptide Display in




Handbook of Experimental Pharmacology, pages




145-163, Springer-Verlag, Berlin (2008)





WGFP
127
Newton & Deutscher, Phage Peptide Display in




Handbook of Experimental Pharmacology, pages




145-163, Springer-Verlag, Berlin (2008)





LWXXAr
128
Newton & Deutscher, Phage Peptide Display in




Handbook of Experimental Pharmacology, pages




145-163, Springer-Verlag, Berlin (2008)





XFXXYLW
129
Newton & Deutscher, Phage Peptide Display in




Handbook of Experimental Pharmacology, pages




145-163, Springer-Verlag, Berlin (2008)





AEPMPHSLNFSQYLWYT
130
Newton & Deutscher, Phage Peptide Display in




Handbook of Experimental Pharmacology, pages




145-163, Springer-Verlag, Berlin (2008)





WAY(W/F)SP
131
Newton & Deutscher, Phage Peptide Display in




Handbook of Experimental Pharmacology, pages




145-163, Springer-Verlag, Berlin (2008)





IELLQAR
132
Newton & Deutscher, Phage Peptide Display in




Handbook of Experimental Pharmacology, pages




145-163, Springer-Verlag, Berlin (2008)





DITWDQLWDLMK
133
Newton & Deutscher, Phage Peptide Display in




Handbook of Experimental Pharmacology, pages




145-163, Springer-Verlag, Berlin (2008)





AYTKCSRQWRTCMTTH
134
Newton & Deutscher, Phage Peptide Display in




Handbook of Experimental Pharmacology, pages




145-163, Springer-Verlag, Berlin (2008)





PQNSKIPGPTFLDPH
135
Newton & Deutscher, Phage Peptide Display in




Handbook of Experimental Pharmacology, pages




145-163, Springer-Verlag, Berlin (2008)





SMEPALPDWWWKMFK
136
Newton & Deutscher, Phage Peptide Display in




Handbook of Experimental Pharmacology, pages




145-163, Springer-Verlag, Berlin (2008)





ANTPCGPYTHDCPVKR
137
Newton & Deutscher, Phage Peptide Display in




Handbook of Experimental Pharmacology, pages




145-163, Springer-Verlag, Berlin (2008)





TACHQHVRMVRP
138
Newton & Deutscher, Phage Peptide Display in




Handbook of Experimental Pharmacology, pages




145-163, Springer-Verlag, Berlin (2008)





VPWMEPAYQRFL
139
Newton & Deutscher, Phage Peptide Display in




Handbook of Experimental Pharmacology, pages




145-163, Springer-Verlag, Berlin (2008)





DPRATPGS
140
Newton & Deutscher, Phage Peptide Display in




Handbook of Experimental Pharmacology, pages




145-163, Springer-Verlag, Berlin (2008)





FRPNRAQDYNTN
141
Newton & Deutscher, Phage Peptide Display in




Handbook of Experimental Pharmacology, pages




145-163, Springer-Verlag, Berlin (2008)





CTKNSYLMC
142
Newton & Deutscher, Phage Peptide Display in




Handbook of Experimental Pharmacology, pages




145-163, Springer-Verlag, Berlin (2008)





C(R/Q)L/RT(G/N)XX
143
Newton & Deutscher, Phage Peptide Display in


G(A/V)GC

Handbook of Experimental Pharmacology, pages




145-163, Springer-Verlag, Berlin (2008)





CPIEDRPMC
144
Newton & Deutscher, Phage Peptide Display in




Handbook of Experimental Pharmacology, pages




145-163, Springer-Verlag, Berlin (2008)





HEWSYLAPYPWF
145
Newton & Deutscher, Phage Peptide Display in




Handbook of Experimental Pharmacology, pages




145-163, Springer-Verlag, Berlin (2008)





MCPKHPLGC
146
Newton & Deutscher, Phage Peptide Display in




Handbook of Experimental Pharmacology, pages




145-163, Springer-Verlag, Berlin (2008)





RMWPSSTVNLSAGRR
147
Newton & Deutscher, Phage Peptide Display in




Handbook of Experimental Pharmacology, pages




145-163, Springer-Verlag, Berlin (2008)





SAKTAVSQRVWLPSHRGGEP
148
Newton & Deutscher, Phage Peptide Display in




Handbook of Experimental Pharmacology, pages




145-163, Springer-Verlag, Berlin (2008)





KSREHVNNSACPSKRITAAL
149
Newton & Deutscher, Phage Peptide Display in




Handbook of Experimental Pharmacology, pages




145-163, Springer-Verlag, Berlin (2008)





EGFR
150
Newton & Deutscher, Phage Peptide Display in




Handbook of Experimental Pharmacology, pages




145-163, Springer-Verlag, Berlin (2008)





AGLGVR
151
Newton & Deutscher, Phage Peptide Display in




Handbook of Experimental Pharmacology, pages




145-163, Springer-Verlag, Berlin (2008)





GTRQGHTMRLGVSDG
152
Newton & Deutscher, Phage Peptide Display in




Handbook of Experimental Pharmacology, pages




145-163, Springer-Verlag, Berlin (2008)





IAGLATPGWSHWLAL
153
Newton & Deutscher, Phage Peptide Display in




Handbook of Experimental Pharmacology, pages




145-163, Springer-Verlag, Berlin (2008)


SMSIARL
154
Newton & Deutscher, Phage Peptide Display in







Handbook of Experimental Pharmacology, pages




145-163, Springer-Verlag, Berlin (2008)





HTFEPGV
155
Newton & Deutscher, Phage Peptide Display in




Handbook of Experimental Pharmacology, pages




145-163, Springer-Verlag, Berlin (2008)





NTSLKRISNKR1RRK
156
Newton & Deutscher, Phage Peptide Display in




Handbook of Experimental Pharmacology, pages




145-163, Springer-Verlag, Berlin (2008)





LRIKRKRRKRKKTRK
157
Newton & Deutscher, Phage Peptide Display in




Handbook of Experimental Pharmacology, pages




145-163, Springer-Verlag, Berlin (2008)





Y* is phospho-Tyr






In some embodiments, the targeting sequence may be a peptide that can be used for chemical coupling (e.g., can comprise arginine and/or lysine residues that can be chemically coupled through their R groups) to another molecule that targets entry into a cell.


In some embodiments, the AAV capsid protein or virus capsid can comprise a mutation as described in WO 2006/066066. For example, the capsid protein can comprise a selective amino acid substitution at amino acid position 263, 705, 708 and/or 716 of the native AAV2 capsid protein or a corresponding change(s) in a capsid protein from another AAV serotype.


Additionally, or alternatively, in some embodiments, the capsid protein, virus capsid or vector comprises a selective amino acid insertion directly following amino acid position 264 of the AAV2 capsid protein or a corresponding change in the capsid protein from other AAV. By “directly following amino acid position X” it is intended that the insertion immediately follows the indicated amino acid position (for example, “following amino acid position 264” indicates a point insertion at position 265 or a larger insertion, e.g., from positions 265 to 268, etc.).


Furthermore, in some embodiments, the capsid protein, virus capsid or vector can comprise amino acid modifications such as described in PCT Publication No. WO 2010/093784 (e.g., 2i8) and/or in PCT Publication No. WO 2014/144229 (e.g., dual glycan).


In some embodiments, the capsid protein, virus capsid or vector can have equivalent or enhanced transduction efficiency relative to the transduction efficiency of the AAV serotype from which the capsid protein, virus capsid or vector originated. In some embodiments, the capsid protein, virus capsid or vector can have reduced transduction efficiency relative to the transduction efficiency of the AAV serotype from which the capsid protein, virus capsid or vector originated. In some embodiments, the capsid protein, virus capsid or vector can have equivalent or enhanced tropism relative to the tropism of the AAV serotype from which the capsid protein, virus capsid or vector originated. In some embodiments, the capsid protein, virus capsid or vector can have an altered or different tropism relative to the tropism of the AAV serotype from which the capsid protein, virus capsid or vector originated. In some embodiments, the capsid protein, virus capsid or vector can have or be engineered to have tropism for brain tissue. In some embodiments, the capsid protein, virus capsid or vector can have or be engineered to have tropism for liver tissue.


The AAV vectors described herein can be used to deliver a heterologous nucleic acid to a cell or subject. For example, the modified vector can be used to treat a lysosomal storage disorder such as a mucopolysaccharidosis disorder (e.g., Sly syndrome [3-glucuronidase], Hurler Syndrome [alpha-L-iduronidase], Scheie Syndrome [alpha-L-iduronidase], Hurler-Scheie Syndrome [alpha-L-iduronidase], Hunter's Syndrome [iduronate sulfatase], Sanfilippo Syndrome (A [heparan sulfamidase], B [N-acetylglucosaminidase], C [acetyl-CoA:alpha-glucosaminide acetyltransferase], D [N-acetylglucosamine 6-sulfatase]), Morquio Syndrome (A [galactose-6-sulfate sulfatase], B [3-galactosidase]), Maroteaux-Lamy Syndrome [N-acetylgalactosamine-4-sulfatase], etc.), Fabry disease (a-galactosidase), Gaucher's disease (glucocerebrosidase), or a glycogen storage disorder (e.g., Pompe disease; lysosomal acid alpha-glucosidase) as described herein.


Those skilled in the art will appreciate that for some AAV capsid proteins the corresponding modification will be an insertion and/or a substitution, depending on whether the corresponding amino acid positions are partially or completely present in the virus or, alternatively, are completely absent.


In some embodiments, virus vectors comprise the modified capsid proteins and capsids described herein. In some embodiments, the virus vector is a parvovirus vector (e.g., comprising a parvovirus capsid and/or vector genome), for example, an AAV vector (e.g., comprising an AAV capsid and/or vector genome). In some embodiments, the virus vector comprises a modified AAV capsid comprising a modified capsid as described herein and a vector genome.


For example, in some embodiments, the virus vector comprises: (a) a modified virus capsid (e.g., a modified AAV capsid) comprising a modified capsid protein described herein; and (b) a nucleic acid comprising a terminal repeat sequence (e.g., an AAV TR), wherein the nucleic acid comprising the terminal repeat sequence is encapsidated by the modified virus capsid. The nucleic acid can optionally comprise two terminal repeats (e.g., two AAV TRs).


In some embodiments, the virus vector is a recombinant virus vector comprising a heterologous nucleic acid encoding a polypeptide or functional RNA of interest. Recombinant virus vectors are described in more detail below.


In some embodiments, the virus vectors (i) have reduced transduction of liver as compared with the level of transduction by a virus vector without the modified capsid protein; (ii) exhibit enhanced systemic transduction by the virus vector in an animal subject as compared with the level observed by a virus vector without the modified capsid protein; (iii) demonstrate enhanced movement across endothelial cells as compared with the level of movement by a virus vector without the modified capsid protein, and/or (iv) exhibit a selective enhancement in transduction of muscle tissue (e.g., skeletal muscle, cardiac muscle and/or diaphragm muscle), (v) exhibit a selective enhancement in transduction of liver tissue, and/or (vi) reduced transduction of brain tissues (e.g., neurons) as compared with the level of transduction by a virus vector without the modified capsid protein. In some embodiments, the virus vector has systemic transduction toward liver.


It will be understood by those skilled in the art that the modified capsid proteins, virus capsids and virus vectors described herein exclude those capsid proteins, capsids and virus vectors that have the indicated amino acids at the specified positions in their native state (i.e., are not mutants).


Methods of Producing Virus Vectors


Also provided herein are methods of producing virus vectors. In some embodiments, a method of producing an AAV vector that evades neutralizing antibodies, comprises: a) identifying contact amino acid residues that form a three dimensional antigenic footprint on an AAV capsid protein; b) generating a library of AAV capsid proteins comprising amino acid substitutions of the contact amino acid residues identified in (a); c) producing AAV particles comprising capsid proteins from the library of AAV capsid proteins of (b); d) contacting the AAV particles of (c) with cells under conditions whereby infection and replication can occur; e) selecting AAV particles that can complete at least one infectious cycle and replicate to titers similar to control AAV particles: 1) contacting the AAV particles selected in (e) with neutralizing antibodies and cells under conditions whereby infection and replication can occur; and g) selecting AAV particles that are not neutralized by the neutralizing antibodies of (f). Nonlimiting examples of methods for identifying contact amino acid residues include peptide epitope mapping and/or cryo-electron microscopy.


Resolution and identification of the antibody contact residues within the three dimensional antigenic footprint allows for their subsequent modification through random, rational and/or degenerate mutagenesis to generate antibody-evading AAV capsids that can be identified through further selection and/or screening.


Thus, in some embodiments, a method of producing an AAV vector that evades neutralizing antibodies comprises: a) identifying contact amino acid residues that form a three dimensional antigenic footprint on an AAV capsid protein; b) generating AAV capsid proteins comprising amino acid substitutions of the contact amino acid residues identified in (a) by random, rational and/or degenerate mutagenesis; c) producing AAV particles comprising capsid proteins from the AAV capsid proteins of (b); d) contacting the AAV particles of (c) with cells under conditions whereby infection and replication can occur; e) selecting AAV particles that can complete at least one infectious cycle and replicate to titers similar to control AAV particles; f) contacting the AAV particles selected in (e) with neutralizing antibodies and cells under conditions whereby infection and replication can occur; and g) selecting AAV particles that are not neutralized by the neutralizing antibodies of (f).


Nonlimiting examples of methods for identifying contact amino acid residues include peptide epitope mapping and/or cryo-electron microscopy. Methods of generating AAV capsid proteins comprising amino acid substitutions of contact amino acid residues by random, rational and/or degenerate mutagenesis are known in the art.


This comprehensive approach presents a platform technology that can be applied to modifying any AAV capsid. Application of this platform technology yields AAV antigenic variants derived from the original AAV capsid template without loss of transduction efficiency. As one advantage and benefit, application of this technology will expand the cohort of patients eligible for gene therapy with AAV vectors.


In some embodiments, a method of producing a virus vector comprises providing to a cell: (a) a nucleic acid template comprising at least one TR sequence (e.g., AAV TR sequence), and (b) AAV sequences sufficient for replication of the nucleic acid template and encapsidation into AAV capsids (e.g., AAV rep sequences and AAV cap sequences encoding the AAV capsids). Optionally, the nucleic acid template further comprises at least one heterologous nucleic acid sequence. In some embodiments, the nucleic acid template comprises two AAV ITR sequences, which are located 5′ and 3′ to the heterologous nucleic acid sequence (if present), although they need not be directly contiguous thereto.


The nucleic acid template and AAV rep and cap sequences are provided under conditions such that virus vector comprising the nucleic acid template packaged within the AAV capsid is produced in the cell. The method can further comprise the step of collecting the virus vector from the cell. The virus vector can be collected from the medium and/or by lysing the cells.


The cell can be a cell that is permissive for AAV viral replication. Any suitable cell known in the art may be employed. In some embodiments, the cell is a mammalian cell. As another option, the cell can be a trans-complementing packaging cell line that provides functions deleted from a replication-defective helper virus, e.g., 293 cells or other E1a trans-complementing cells.


The AAV replication and capsid sequences may be provided by any method known in the art. Current protocols typically express the AAV rep/cap genes on a single plasmid. The AAV replication and packaging sequences need not be provided together, although it may be convenient to do so. The AAV rep and/or cap sequences may be provided by any viral or non-viral vector. For example, the rep/cap sequences may be provided by a hybrid adenovirus or herpesvirus vector (e.g., inserted into the E1a or E3 regions of a deleted adenovirus vector). EBV vectors may also be employed to express the AAV cap and rep genes. One advantage of this method is that EBV vectors are episomal, yet will maintain a high copy number throughout successive cell divisions (i.e., are stably integrated into the cell as extra-chromosomal elements, designated as an “EBV based nuclear episome,” see Margolski, (1992) Curr. Top. Microbiol. Immun. 158:67).


As a further alternative, the rep/cap sequences may be stably incorporated into a cell.


Typically the AAV rep/cap sequences will not be flanked by the TRs, to prevent rescue and/or packaging of these sequences.


The nucleic acid template can be provided to the cell using any method known in the art. For example, the template can be supplied by a non-viral (e.g., plasmid) or viral vector. In some embodiments, the nucleic acid template is supplied by a herpesvirus or adenovirus vector (e.g., inserted into the E1a or E3 regions of a deleted adenovirus). As another illustration, Palombo et al., (1998) J. Virology 72:5025, describes a baculovirus vector carrying a reporter gene flanked by the AAV TRs. EBV vectors may also be employed to deliver the template, as described above with respect to the rep/cap genes.


In some embodiments, the nucleic acid template is provided by a replicating rAAV virus. In some embodiments, an AAV provirus comprising the nucleic acid template is stably integrated into the chromosome of the cell.


To enhance virus titers, helper virus functions (e.g., adenovirus or herpesvirus) that promote a productive AAV infection can be provided to the cell. Helper virus sequences necessary for AAV replication are known in the art. Typically, these sequences will be provided by a helper adenovirus or herpesvirus vector. Alternatively, the adenovirus or herpesvirus sequences can be provided by another non-viral or viral vector, e.g., as a noninfectious adenovirus miniplasmid that carries all of the helper genes that promote efficient AAV production as described by Ferrari et al., (1997) Nature Med. 3: 1295, and U.S. Pat. Nos. 6,040,183 and 6,093,570.


Further, the helper virus functions may be provided by a packaging cell with the helper sequences embedded in the chromosome or maintained as a stable extrachromosomal element. Generally, the helper virus sequences cannot be packaged into AAV virions, e.g., are not flanked by TRs.


Those skilled in the art will appreciate that it may be advantageous to provide the AAV replication and capsid sequences and the helper virus sequences (e.g., adenovirus sequences) on a single helper construct. This helper construct may be a non-viral or viral construct. As one nonlimiting illustration, the helper construct can be a hybrid adenovirus or hybrid herpesvirus comprising the AAV rep/cap genes.


In some embodiments, the AAV rep/cap sequences and the adenovirus helper sequences are supplied by a single adenovirus helper vector. This vector further can further comprise the nucleic acid template. The AAV rep/cap sequences and/or the rAAV template can be inserted into a deleted region (e.g., the Ela or E3 regions) of the adenovirus.


In some embodiments, the AAV rep/cap sequences and the adenovirus helper sequences are supplied by a single adenovirus helper vector. According to this embodiment, the rAAV template can be provided as a plasmid template.


In some embodiments, the AAV rep/cap sequences and adenovirus helper sequences are provided by a single adenovirus helper vector, and the rAAV template is integrated into the cell as a provirus. Alternatively, the rAAV template is provided by an EBV vector that is maintained within the cell as an extrachromosomal element (e.g., as an EBV based nuclear episome).


In some embodiments, the AAV rep/cap sequences and adenovirus helper sequences are provided by a single adenovirus helper. The rAAV template can be provided as a separate replicating viral vector. For example, the rAAV template can be provided by a rAAV particle or a second recombinant adenovirus particle.


According to the foregoing methods, the hybrid adenovirus vector typically comprises the adenovirus 5′ and 3′ cis sequences sufficient for adenovirus replication and packaging (i.e., the adenovirus terminal repeats and PAC sequence). The AAV rep/cap sequences and, if present, the rAAV template are embedded in the adenovirus backbone and are flanked by the 5′ and 3′ cis sequences, so that these sequences may be packaged into adenovirus capsids. As described above, the adenovirus helper sequences and the AAV rep/cap sequences are generally not flanked by TRs so that these sequences are not packaged into the AAV virions. Zhang et al., ((2001) Gene Ther. 18:704-12) describe a chimeric helper comprising both adenovirus and the AAV rep and cap genes.


Herpesvirus may also be used as a helper virus in AAV packaging methods. Hybrid herpesviruses encoding the AAV Rep protein(s) may advantageously facilitate scalable AAV vector production schemes. A hybrid herpes simplex virus type I (HSV-1) vector expressing the AAV-2 rep and cap genes has been described (Conway et al., (1999) Gene Therapy 6:986 and WO 00/17377.


As a further alternative, virus vectors can be produced in insect cells using baculovirus vectors to deliver the rep/cap genes and rAAV template as described, for example, by Urabe et al., (2002) Human Gene Therapy 13: 1935-43.


AAV vector stocks free of contaminating helper virus may be obtained by any method known in the art. For example, AAV and helper virus may be readily differentiated based on size. AAV may also be separated away from helper virus based on affinity for a heparin substrate (Zolotukhin et al. (1999) Gene Therapy 6:973). Deleted replication-defective helper viruses can be used so that any contaminating helper virus is not replication competent. As a further alternative, an adenovirus helper lacking late gene expression may be employed, as only adenovirus early gene expression is required to mediate packaging of AAV virus. Adenovirus mutants defective for late gene expression are known in the art (e.g., ts100K and ts149 adenovirus mutants).


Recombinant Virus Vectors


The virus vectors described herein are useful for the delivery of nucleic acids to cells in vitro, ex vivo, and in vivo. In particular, the virus vectors can be advantageously employed to deliver or transfer nucleic acids to animal, including mammalian, cells. Thus, in some embodiments, a nucleic acid may be encapsidated by a capsid protein described herein. In some embodiments, the nucleic acid is a cargo nucleic acid. In some embodiments, the cargo nucleic acid comprises a vector genome (e.g., 5′ ITR, transgene, and 3′ ITR).


The cargo nucleic acid sequence delivered by the virus vectors may be any heterologous nucleic acid sequence(s) of interest. Nucleic acids of interest include nucleic acids encoding polypeptides, including therapeutic (e.g., for medical or veterinary uses) or immunogenic (e.g., for vaccines) polypeptides or RNAs. In some embodiments, the cargo nucleic acid comprises a 5′ ITR and a 3′ ITR. In some embodiments, the cargo nucleic acid comprises a 5′ ITR, a transgene, and a 3′ITR. In some embodiments, the transgene encodes a therapeutic protein or RNA.


Therapeutic polypeptides include, but are not limited to, cystic fibrosis transmembrane regulator protein (CFTR), dystrophin (including mini- and micro-dystrophins, see, e.g., Vincent et al, (1993) Nature Genetics 5: 130; U.S. Patent Publication No. 2003/017131; International publication WO/2008/088895, Wang et al., Proc. Natl. Acad. Sci. USA 97: 1 3714-13719 (2000); and Gregorevic et al., Mol. Ther. 16:657-64 (2008)), myostatin propeptide, follistatin, activin type 11 soluble receptor, IGF-1, apolipoproteins such as apoA (apoA1, apoA2, apoA4, apoA-V), apoB (apoB100, ApoB48), apoC (apoCI, apoCII, apoCIII, apoCIV), apoD, apoE, apoH, apoL, apo(a), anti-inflammatory polypeptides such as the Ikappa B dominant mutant, amyloid beta, tau, sarcospan, utrophin (Tinsley et al, (1996) Nature 384:349), mini-utrophin, clotting factors (e.g., Factor VIII, Factor IX, Factor X, etc.), erythropoietin, angiostatin, endostatin, catalase, tyrosine hydroxylase, superoxide dismutase, leptin, the LDL receptor, lipoprotein lipase, progranulin, ornithine transcarbamylase, β-globin, α-globin, spectrin, alpha-1-antitrypsin, adenosine deaminase, hypoxanthine guanine phosphoribosyl transferase, β-glucocerebrosidase, battenin, sphingomyelinase, lysosomal hexosaminidase A, branched-chain keto acid dehydrogenase, frataxin, RP65 protein, cytokines (e.g., alpha-interferon, beta-interferon, gamma-interferon, interleukin-2, interleukin-4, alpha synuclein, parkin, granulocyte-macrophage colony stimulating factor, lymphotoxin, and the like), peptide growth factors, neurotrophic factors and hormones (e.g., somatotropin, insulin, insulin-like growth factors 1 and 2, platelet derived growth factor, epidermal growth factor, fibroblast growth factor, nerve growth factor, neurotrophic factor-3 and -4, brain-derived neurotrophic factor, bone morphogenic proteins [including RANKL and VEGF], glial derived growth factor, transforming growth factor-α and -β, and the like), huntingin, lysosomal acid alpha-glucosidase, iduronate-2-sulfatase, N-sulfoglucosamine sulfohydrolase, alpha-galactosidase A, receptors (e.g., the tumor necrosis growth factor soluble receptor), S100A1, ubiquitin protein ligase E3, parvalbumin, adenylyl cyclase type 6, a molecule that modulates calcium handling (e.g., SERCA2A, Inhibitor 1 of PP1 and fragments thereof [e.g., WO 2006/029319 and WO 2007/100465]), a molecule that effects G-protein coupled receptor kinase type 2 knockdown such as a truncated constitutively active bARKct, anti-inflammatory factors such as IRAP, anti-myostatin proteins, aspartoacylase, monoclonal antibodies (including single chain monoclonal antibodies; an exemplary Mab is the Herceptin® Mab), neuropeptides and fragments thereof (e.g., galanin, Neuropeptide Y (see, U.S. Pat. No. 7,071,172)), angiogenesis inhibitors such as Vasohibins and other VEGF inhibitors (e.g., Vasohibin 2 [see, WO JP2006/073052]). Other illustrative heterologous nucleic acid sequences encode suicide gene products (e.g., thymidine kinase, cytosine deaminase, diphtheria toxin, and tumor necrosis factor), proteins that enhance or inhibit transcription of host factors (e.g., nuclease-dead Cas9 linked to a transcription enhancer or inhibitor element, zinc-finger proteins linked to a transcription enhancer or inhibitor element, transcription activator-like (TAL) effectors linked to a transcription enhancer or inhibitor element), proteins conferring resistance to a drug used in cancer therapy, tumor suppressor gene products (e.g., p53, Rb, Wt-1), TRAIL, FAS-ligand, and any other polypeptide that has a therapeutic effect in a subject in need thereof. AAV vectors can also be used to deliver monoclonal antibodies and antibody fragments, for example, an antibody or antibody fragment directed against myostatin (see, e.g., Fang et al., Nature Biotechnology 23:584-590 (2005)). Heterologous nucleic acid sequences encoding polypeptides include those encoding reporter polypeptides (e.g., an enzyme). Reporter polypeptides are known in the art and include, but are not limited to, Green Fluorescent Protein, β-galactosidase, alkaline phosphatase, luciferase, and chloramphenicol acetyltransferase gene.


Optionally, the heterologous nucleic acid encodes a secreted polypeptide (e.g., a polypeptide that is a secreted polypeptide in its native state or that has been engineered to be secreted, for example, by operable association with a secretory signal sequence as is known in the art).


Alternatively, in some embodiments, the heterologous nucleic acid may encode an antisense nucleic acid, a ribozyme (e.g., as described in U.S. Pat. No. 5,877,022), RNAs that effect spliceosome-mediated/ram-splicing (see, Puttaraju et al, (1999) Nature Biotech. 17:246; U.S. Pat. Nos. 6,013,487; 6,083,702), interfering RNAs (RNAi) including siRNA, shRNA or miRNA that mediate gene silencing (see, Sharp et al, (2000) Science 287:2431), and other non-translated RNAs, such as “guide” RNAs (Gorman et al., (1998) Proc. Nat. Acad. Sci. USA 95:4929; U.S. Pat. No. 5,869,248 to Yuan et al.), and the like. Exemplary untranslated RNAs include RNAi against a multiple drug resistance (MDR) gene product (e.g., to treat and/or prevent tumors and/or for administration to the heart to prevent damage by chemotherapy), RNAi against myostatin (e.g., for Duchenne muscular dystrophy), RNAi against VEGF (e.g., to treat and/or prevent tumors), RNAi against phospholamban (e.g., to treat cardiovascular disease, see, e.g., Andino et al., J. Gene Med. 10: 132-142 (2008) and Li et al., Acta Pharmacol Sin. 26:51-55 (2005)); phospholamban inhibitory or dominant-negative molecules such as phospholamban S16E (e.g., to treat cardiovascular disease, see, e.g., Hoshijima et al. Nat. Med. 8:864-871 (2002)), RNAi to adenosine kinase (e.g., for epilepsy), and RNAi directed against pathogenic organisms and viruses (e.g., hepatitis B and/or C virus, human immunodeficiency virus, CMV, herpes simplex virus, human papilloma virus, etc.).


Further, a nucleic acid sequence that directs alternative splicing can be delivered. To illustrate, an antisense sequence (or other inhibitory sequence) complementary to the 5′ and/or 3′ splice site of dystrophin exon 51 can be delivered in conjunction with a U1 or U7 small nuclear (sn) RNA promoter to induce skipping of this exon. For example, a DNA sequence comprising a U1 or U7 snRNA promoter located 5′ to the antisense/inhibitory sequence(s) can be packaged and delivered in a modified capsid.


In some embodiments, a nucleic acid sequence that directs gene editing can be delivered. For example, the nucleic acid may encode a guide RNA. In some embodiments, the guide RNA is a single guide RNA (sgRNA) comprising a crRNA sequence and a tracrRNA sequence. In some embodiments, the nucleic acid may encode a nuclease. In some embodiments, the nuclease is a zinc-finger nuclease, a homing endonuclease, a TALEN (transcription activator-like effector nuclease), a NgAgo (agronaute endonuclease), a SGN (structure-guided endonuclease), a RGN (RNA-guided nuclease), or modified or truncated variants thereof. In some embodiments, the RNA-guided nuclease is a Cas9 nuclease, a Cas12(a) nuclease (Cpf1), a Cas12b nuclease, a Cas12c nuclease, a TrpB-like nuclease, a Cas13a nuclease (C2c2), a Cas13b nuclease, or modified or truncated variants thereof. In some embodiments, the Cas9 nuclease is isolated or derived from S. pyogenes or S. aureus.


In some embodiments, a nucleic acid sequence that directs gene knockdown can be delivered. For example, the nucleic acid sequence may encode a siRNA, an shRNA, a microRNA, or an antisense nucleic acid. The virus vector may also comprise a heterologous nucleic acid that shares homology with and recombines with a locus on a host chromosome. This approach can be utilized, for example, to correct a genetic defect in the host cell.


Also provided are virus vectors that express an immunogenic polypeptide, e.g., for vaccination. The nucleic acid may encode any immunogen of interest known in the art including, but not limited to, immunogens from human immunodeficiency virus (HIV), simian immunodeficiency virus (SIV), influenza virus, HIV or SIV gag proteins, tumor antigens, cancer antigens, bacterial antigens, viral antigens, and the like.


The use of parvoviruses as vaccine vectors is known in the art (see, e.g., Miyamura el al, (1994) Proc. Nat. Acad. Sci USA 91:8507; U.S. Pat. No. 5,916,563 to Young et al, U.S. Pat. No. 5,905,040 to Mazzara et al, U.S. Pat. No. 5,882,652, 5,863,541 to Samulski et al). The antigen may be presented in the parvovirus capsid.


Alternatively, the antigen may be expressed from a heterologous nucleic acid introduced into a recombinant vector genome. In some embodiments, any immunogen of interest as described herein and/or as is known in the art can be provided by the virus vectors described herein.


An immunogenic polypeptide can be any polypeptide suitable for eliciting an immune response and/or protecting the subject against an infection and/or disease, including, but not limited to, microbial, bacterial, protozoal, parasitic, fungal and/or viral infections and diseases. For example, the immunogenic polypeptide can be an orthomyxovirus immunogen (e.g., an influenza virus immunogen, such as the influenza virus hemagglutinin (HA) surface protein or the influenza virus nucleoprotein, or an equine influenza virus immunogen) or a lentivirus immunogen (e.g., an equine infectious anemia virus immunogen, a Simian Immunodeficiency Virus (SIV) immunogen, or a Human Immunodeficiency Virus (HIV) immunogen, such as the HIV or SIV envelope GP 160 protein, the HIV or SIV matrix/capsid proteins, and the HIV or SIV gag, pol and env genes products). The immunogenic polypeptide can also be an arenavirus immunogen (e.g., Lassa fever virus immunogen, such as the Lassa fever virus nucleocapsid protein and the Lassa fever envelope glycoprotein), a poxvirus immunogen (e.g., a vaccinia virus immunogen, such as the vaccinia LI or L8 gene products), a flavivirus immunogen (e.g., a yellow fever virus immunogen or a Japanese encephalitis virus immunogen), a filovirus immunogen (e.g., an Ebola virus immunogen, or a Marburg virus immunogen, such as NP and GP gene products), a bunyavirus immunogen (e.g., RVFV, CCHF, and/or SFS virus immunogens), or a coronavirus immunogen (e.g., an infectious human coronavirus immunogen, such as the human coronavirus envelope glycoprotein, or a porcine transmissible gastroenteritis virus immunogen, or an avian infectious bronchitis virus immunogen). The immunogenic polypeptide can further be a polio immunogen, a herpes immunogen (e.g., CMV, EBV, HSV immunogens), a mumps immunogen, a measles immunogen, a rubella immunogen, a diphtheria toxin or other diphtheria immunogen, a pertussis antigen, a hepatitis (e.g., hepatitis A, hepatitis B, hepatitis C, etc.) immunogen, and/or any other vaccine immunogen now known in the art or later identified as an immunogen.


Alternatively, the immunogenic polypeptide can be any tumor or cancer cell antigen. Optionally, the tumor or cancer antigen is expressed on the surface of the cancer cell.


Exemplary cancer and tumor cell antigens are described in S. A. Rosenberg (Immunity 10:281 (1991)). Other illustrative cancer and tumor antigens include, but are not limited to: BRCA1 gene product, BRCA2 gene product, gp100, tyrosinase, GAGE-1/2, BAGE, RAGE, LAGE, NY-ESO-1, CDK-4, β-catenin, MUM-1, Caspase-8, KIAA0205, HPVE, SART-1, FRAME, p15, melanoma tumor antigens (Kawakami et al., (1994) Proc. Natl. Acad. Sci. USA 91:3515; Kawakami et al., (1994) J. Exp. Med., 180:347; Kawakami et al., (1994) Cancer Res. 54:3124), MART-1, gp100, MAGE-1, MAGE-2, MAGE-3, CEA, TRP-1, TRP-2, P-15, tyrosinase (Brichard et al., (1993) J Exp. Med. 178:489); HER-2/neu gene product (U.S. Pat. No. 4,968,603), CA 125, LK26, FB5 (endosialin), TAG 72, AFP, CA 19-9, NSE, DU-PAN-2, CA50, SPan-1, CA72-4, HCG, STN (sialyl Tn antigen), c-erbB-2 proteins, PSA, L-CanAg, estrogen receptor, milk fat globulin, p53 tumor suppressor protein (Levine, (1993) Ann. Rev. Biochem. 62:623); mucin antigens (International Patent Publication No. WO 90/05142); telomerases; nuclear matrix proteins; prostatic acid phosphatase; papilloma virus antigens; and/or antigens now known or later discovered to be associated with the following cancers: melanoma, adenocarcinoma, thymoma, lymphoma (e.g., non-Hodgkin's lymphoma, Hodgkin's lymphoma), sarcoma, lung cancer, liver cancer, colon cancer, leukemia, uterine cancer, breast cancer, prostate cancer, ovarian cancer, cervical cancer, bladder cancer, kidney cancer, pancreatic cancer, brain cancer and any other cancer or malignant condition or metastasis thereof now known or later identified (see, e.g., Rosenberg, (1996) Ann. Rev. Med. 47:481-91).


As a further alternative, the heterologous nucleic acid can encode any polypeptide that is desirably produced in a cell in vitro, ex vivo, or in vivo. For example, the virus vectors may be introduced into cultured cells and the expressed gene product isolated therefrom.


It will be understood by those skilled in the art that the heterologous nucleic acid(s) of interest can be operably associated with appropriate control sequences. For example, the heterologous nucleic acid can be operably associated with expression control elements, such as transcription/translation control signals, origins of replication, polyadenylation signals, internal ribosome entry sites (IRES), promoters, and/or enhancers, and the like.


Further, regulated expression of the heterologous nucleic acid(s) of interest can be achieved at the post-transcriptional level, e.g., by regulating selective splicing of different introns by the presence or absence of an oligonucleotide, small molecule and/or other compound that selectively blocks splicing activity at specific sites (e.g., as described in WO 2006/119137).


Those skilled in the art will appreciate that a variety of promoter/enhancer elements can be used depending on the level and tissue-specific expression desired. The promoter/enhancer can be constitutive or inducible, depending on the pattern of expression desired. The promoter/enhancer can be native or foreign and can be a natural or a synthetic sequence. By foreign, it is intended that the transcriptional initiation region is not found in the wild-type host into which the transcriptional initiation region is introduced.


In some embodiments, the promoter/enhancer elements can be native to the target cell or subject to be treated. In some embodiments, the promoters/enhancer element can be native to the heterologous nucleic acid sequence. The promoter/enhancer element is generally chosen so that it functions in the target cell(s) of interest. Further, in some embodiments the promoter/enhancer element is a mammalian promoter/enhancer element. The promoter/enhancer element may be constitutive or inducible.


Inducible expression control elements are typically advantageous in those applications in which it is desirable to provide regulation over expression of the heterologous nucleic acid sequence(s). Inducible promoters/enhancer elements for gene delivery can be tissue-specific or -preferred promoter/enhancer elements, and include muscle specific or preferred (including cardiac, skeletal and/or smooth muscle specific or preferred), neural tissue specific or preferred (including brain-specific or preferred), eye specific or preferred (including retina-specific and cornea-specific), liver specific or preferred, bone marrow specific or preferred, pancreatic specific or preferred, spleen specific or preferred, and lung specific or preferred promoter/enhancer elements. Other inducible promoter/enhancer elements include hormone-inducible and metal-inducible elements. Exemplary inducible promoters/enhancer elements include, but are not limited to, a Tet on/off element, a RU486-inducible promoter, an ecdysone-inducible promoter, a rapamycin-inducible promoter, and a metallothionein promoter.


In some embodiments wherein the heterologous nucleic acid sequence(s) is transcribed and then translated in the target cells, specific initiation signals are generally included for efficient translation of inserted protein coding sequences. These exogenous translational control sequences, which may include the ATG initiation codon and adjacent sequences, can be of a variety of origins, both natural and synthetic.


The virus vectors described herein provide a means for delivering heterologous nucleic acids into a broad range of cells, including dividing and non-dividing cells. The virus vectors can be employed to deliver a nucleic acid of interest to a cell in vitro, e.g., to produce a polypeptide in vitro or for ex vivo gene therapy. The virus vectors are additionally useful in a method of delivering a nucleic acid to a subject in need thereof e.g., to express an immunogenic or therapeutic polypeptide or a functional RNA. In this manner, the polypeptide or functional RNA can be produced in vivo in the subject. The subject can be in need of the polypeptide because the subject has a deficiency of the polypeptide. Further, the method can be practiced because the production of the polypeptide or functional RNA in the subject may impart some beneficial effect.


The virus vectors can also be used to produce a polypeptide of interest or functional RNA in cultured cells or in a subject (e.g., using the subject as a bioreactor to produce the polypeptide or to observe the effects of the functional RNA on the subject, for example, in connection with screening methods).


In general, the virus vectors of the described herein can be employed to deliver a heterologous nucleic acid encoding a polypeptide or functional RNA to treat and/or prevent any disease state for which it is beneficial to deliver a therapeutic polypeptide or functional RNA. Illustrative disease states include, but are not limited to: cystic fibrosis (cystic fibrosis transmembrane regulator protein) and other diseases of the lung, hemophilia A (Factor VIII), hemophilia B (Factor IX), thalassemia (β-globin), anemia (erythropoietin) and other blood disorders. Alzheimer's disease (GDF; neprilysin), multiple sclerosis (β-interferon), Parkinson's disease (glial-cell line derived neurotrophic factor [GDNF]), Huntington's disease (RNAi to remove repeats), Canavan's disease, amyotrophic lateral sclerosis, epilepsy (galanin, neurotrophic factors), and other neurological disorders, cancer (endostatin, angiostatin, TRAIL, FAS-ligand, cytokines including interferons; RNAi including RNAi against VEGF or the multiple drug resistance gene product, mir-26a [e.g., for hepatocellular carcinoma]), diabetes mellitus (insulin), muscular dystrophies including Duchenne (dystrophin, mini-dystrophin, insulin-like growth factor I, a sarcoglycan [e.g., α, β, γ], RNAi against myostatic myostatin propeptide, follistatin, activin type II soluble receptor, anti-inflammatory polypeptides such as the Ikappa B dominant mutant, sarcospan, utrophin, mini-utrophin, antisense or RNAi against splice junctions in the dystrophin gene to induce exon skipping [see, e.g., WO/2003/095647], antisense against U7 snRNAs to induce exon skipping [see, e.g., WO/2006/021724], and antibodies or antibody fragments against myostatin or myostatin propeptide) and Becker, Myotonic dystrophy 1 or 2, facioscapulohumeral muscular dystrophy (FSHD), Gaucher disease (glucocerebrosidase), Hurler's disease (a-L-iduronidase), adenosine deaminase deficiency (adenosine deaminase), glycogen storage diseases (e.g., Fabry disease [a-galactosidase] and Pompe disease [lysosomal acid alpha-glucosidase]) and other metabolic disorders, congenital emphysema (alpha-1-antitrypsin), Lesch-Nyhan Syndrome (hypoxan thine guanine phosphoribosyl transferase), Niemann-Pick disease (sphingomyelinase), Tay-Sachs disease (lysosomal hexosaminidase A), frontotemporal dementia, Maple Syrup Urine Disease (branched-chain keto acid dehydrogenase), retinal degenerative diseases (and other diseases of the eye and retina; e.g., PDGF for macular degeneration and/or vasohibin or other inhibitors of VEGF or other angiogenesis inhibitors to treat/prevent retinal disorders, e.g., in Type I diabetes), diseases of solid organs such as brain (including Parkinson's Disease [GDNF], astrocytomas [endostatin, angiostatin and/or RNAi against VEGF], glioblastomas [endostatin, angiostatin and/or RNAi against VEGF]), liver, kidney, heart including congestive heart failure or peripheral artery disease (PAD) (e.g., by delivering protein phosphatase inhibitor 1 (1-1) and fragments thereof (e.g., IIC), serca2a, zinc finger proteins that regulate the phospholamban gene, Barkct, [32-adrenergic receptor, 2-adrenergic receptor kinase (BARK), phosphoinositide-3 kinase (PI3 kinase), S100A1, parvalbumin, adenylyl cyclase type 6, a molecule that effects G-protein coupled receptor kinase type 2 knockdown such as a truncated constitutively active bARKct; calsarcin, RNAi against phospholamban; phospholamban inhibitory or dominant-negative molecules such as phospholamban S16E, etc.), arthritis (insulin-like growth factors), joint disorders (insulin-like growth factor 1 and/or 2), intimal hyperplasia (e.g., by delivering enos, inos), improve survival of heart transplants (superoxide dismutase), AIDS (soluble CD4), muscle wasting (insulin-like growth factor I), kidney deficiency (erythropoietin), anemia (erythropoietin), arthritis (anti-inflammatory factors such as I RAP and TNFa soluble receptor), hepatitis (a-interferon), LDL receptor deficiency (LDL receptor), hyperammonemia (ornithine transcarbamylase), Krabbe's disease (galactocerebrosidase), Batten's disease, spinal cerebral ataxias including SCA1, SCA2 and SCA3, phenylketonuria (phenylalanine hydroxylase), autoimmune diseases, and the like. The compositions and methods disclosed herein can further be used following organ transplantation to increase the success of the transplant and/or to reduce the negative side effects of organ transplantation or adjunct therapies (e.g., by administering immunosuppressant agents or inhibitory nucleic acids to block cytokine production). As another example, bone morphogenic proteins (including BNP 2, 7, etc., RANKL and/or VEGF) can be administered with a bone allograft, for example, following a break or surgical removal in a cancer patient.


In some embodiments, the virus vectors described herein can be employed to deliver a heterologous nucleic acid encoding a polypeptide or functional RNA to treat and/or prevent a liver disease or disorder. The liver disease or disorder may be, for example, primary biliary cirrhosis, nonalcoholic fatty liver disease (NAFLD), non-alcoholic steatohepatitis (NASH), autoimmune hepatitis, hepatitis B, hepatitis C, alcoholic liver disease, fibrosis, jaundice, primary sclerosing cholangitis (PSC), Budd-Chiari syndrome, hemochromatosis, Wilson's disease, alcoholic fibrosis, non-alcoholic fibrosis, liver steatosis, Gilbert's syndrome, biliary atresia, alpha-1-antitrypsin deficiency, alagille syndrome, progressive familial intrahepatic cholestasis, Hemophilia B, Hereditary Angioedema (HAE), Homozygous Familial Hypercholesterolemia (HoFH), Heterozygous Familial Hypercholesterolemia (HeFH), Von Gierke's Disease (GSD 1), Hemophilia A, Methylmalonic Acidemia, Propionic Acidemia, Homocystinuria, Phenylketonuria (PKU), Tyrosinemia Type 1, Arginase 1 Deficiency, Argininosuccinate Lyase Deficiency, Carbamoyl-phosphate synthetase 1 deficiency, Citrullinemia Type 1, Citrin Deficiency, Crigler-Najjar Syndrome Type 1, Cystinosis, Fabry Disease, Glycogen Storage Disease 1b, LPL Deficiency, N-Acetylglutamate Synthetase Deficiency, Ornithine Transcarbamylase Deficiency, Ornithine Translocase Deficiency, Primary Hyperoxaluria Type 1, or ADA SCID.


The compositions and methods described herein can also be used to produce induced pluripotent stem cells (iPS). For example, a virus vector described herein can be used to deliver stem cell associated nucleic acid(s) into a non-pluripotent cell, such as adult fibroblasts, skin cells, liver cells, renal cells, adipose cells, cardiac cells, neural cells, epithelial cells, endothelial cells, and the like.


Nucleic acids encoding factors associated with stem cells are known in the art. Nonlimiting examples of such factors associated with stem cells and pluripotency include Oct-3/4, the SOX family (e.g., SOX 1, SOX2, SOX3 and/or SOX 15), the KIf family (e.g., KIf1, KHZ KIf4 and/or KIf5), the Myc family (e.g., C-myc, L-myc and/or N-myc), NANOG and/or LIN28.


The methods described herein can also be practiced to treat and/or prevent a metabolic disorder such as diabetes (e.g., insulin), hemophilia (e.g., Factor IX or Factor VIII), a lysosomal storage disorder such as a mucopolysaccharidosis disorder (e.g., Sly syndrome [β-glucuronidase], Hurler Syndrome [alpha-L-iduronidase], Scheie Syndrome [alpha-L-iduronidase], Hurler-Scheie Syndrome [alpha-L-iduronidase], Hunter's Syndrome [iduronate sulfatase], Sanfilippo Syndrome A [heparan sulfamidase], B [N-acetylglucosaminidase], C [acetyl-CoA:alpha-glucosaminide acetyltransferase], D [N-acetylglucosamine 6-sulfatase], Morquio Syndrome A [galactoses-sulfate sulfatase], B [3-galactosidase], Maroteaux-Lamy Syndrome [N-acetylgalactosamine-4-sulfatase], etc.), Fabry disease (alpha-galactosidase), Gaucher's disease (glucocerebrosidase), or a glycogen storage disorder (e.g., Pompe disease; lysosomal acid alpha-glucosidase).


Gene transfer has substantial use for understanding and providing therapy for disease states. There are a number of inherited diseases in which defective genes are known and have been cloned. In general, the above disease states fall into two classes: deficiency states, usually of enzymes, which are generally inherited in a recessive manner, and unbalanced states, which may involve regulatory or structural proteins, and which are typically inherited in a dominant manner. For deficiency state diseases, gene transfer can be used to bring a normal gene into affected tissues for replacement therapy, as well as to create animal models for the disease using antisense mutations. For unbalanced disease states, gene transfer can be used to create a disease state in a model system, which can then be used in efforts to counteract the disease state. Thus, virus vectors as described herein permit the treatment and/or prevention of genetic diseases.


The virus vectors described herein may also be employed to provide a functional RNA to a cell in vitro or in vivo. The functional RNA may be, for example, a non-coding RNA. In some embodiments, expression of the functional RNA in the cell can diminish expression of a particular target protein by the cell. Accordingly, functional RNA can be administered to decrease expression of a particular protein in a subject in need thereof. In some embodiments, expression of the functional RNA in the cell can increase expression of a particular target protein by the cell. Accordingly, functional RNA can be administered to increase expression of a particular protein in a subject in need thereof. In some embodiments, expression of the functional RNA can regulate splicing of a particular target RNA in a cell. Accordingly, functional RNA can be administered to regulate splicing a particular RNA in a subject in need thereof. In some embodiments, expression of the functional RNA in the cell can regulate the function of a particular target protein by the cell. Accordingly, functional RNA can be administered to regulate the function of a particular protein in a subject in need thereof. Functional RNA can also be administered to cells in vitro to regulate gene expression and/or cell physiology, e.g., to optimize cell or tissue culture systems or in screening methods.


In addition, virus vectors as described herein find use in diagnostic and screening methods, whereby a nucleic acid of interest is transiently or stably expressed in a cell culture system, or alternatively, a transgenic animal model.


The virus vectors can also be used for various non-therapeutic purposes, including but not limited to use in protocols to assess gene targeting, clearance, transcription, translation, etc., as would be apparent to one skilled in the art. The virus vectors can also be used for the purpose of evaluating safety (spread, toxicity, immunogenicity, etc.). Such data, for example, are considered by the United States Food and Drug Administration as part of the regulatory approval process prior to evaluation of clinical efficacy.


In some embodiments, the virus vectors may be used to produce an immune response in a subject. According to this embodiment, a virus vector comprising a heterologous nucleic acid sequence encoding an immunogenic polypeptide can be administered to a subject, and an active immune response is mounted by the subject against the immunogenic polypeptide. Immunogenic polypeptides are as described hereinabove. In some embodiments, a protective immune response is elicited.


Alternatively, the virus vector may be administered to a cell ex vivo and the altered cell is administered to the subject. The virus vector comprising the heterologous nucleic acid is introduced into the cell, and the cell is administered to the subject, where the heterologous nucleic acid encoding the immunogen can be expressed and induce an immune response in the subject against the immunogen. In some embodiments, the cell is an antigen-presenting cell (e.g., a dendritic cell).


An “active immune response” or “active immunity” is characterized by “participation of host tissues and cells after an encounter with the immunogen. It involves differentiation and proliferation of immunocompetent cells in lymphoreticular tissues, which lead to synthesis of antibody or the development of cell-mediated reactivity, or both.” Herbert B. Herscowitz, Immunophysiology: Cell Function and Cellular Interactions in Antibody Formation, in IMMUNOLOGY: BASIC PROCESSES 117 (Joseph A. Bellanti ed., 1985). Alternatively stated, an active immune response is mounted by the host after exposure to an immunogen by infection or by vaccination. Active immunity can be contrasted with passive immunity, which is acquired through the transfer of preformed substances (antibody, transfer factor, thymic graft, interleukin-2) from an actively immunized host to a non-immune host.


A “protective” immune response or “protective” immunity as used herein indicates that the immune response confers some benefit to the subject in that it prevents or reduces the incidence of disease. Alternatively, a protective immune response or protective immunity may be useful in the treatment and/or prevention of disease, in particular cancer or tumors (e.g., by preventing cancer or tumor formation, by causing regression of a cancer or tumor and/or by preventing metastasis and/or by preventing growth of metastatic nodules). The protective effects may be complete or partial, as long as the benefits of the treatment outweigh any disadvantages thereof.


In some embodiments, the virus vector or cell comprising the heterologous nucleic acid can be administered in an immunogenically effective amount, as described below.


In some embodiments, the virus vectors can be administered for cancer immunotherapy by administration of a virus vector expressing one or more cancer cell antigens (or an immunologically similar molecule) or any other immunogen that produces an immune response against a cancer cell. To illustrate, an immune response can be produced against a cancer cell antigen in a subject by administering a virus vector comprising a heterologous nucleic acid encoding the cancer cell antigen, for example to treat a patient with cancer and/or to prevent cancer from developing in the subject. The virus vector may be administered to a subject in vivo or by using ex vivo methods, as described herein.


Alternatively, the cancer antigen can be expressed as part of the virus capsid or be otherwise associated with the virus capsid (e.g., as described above).


As another alternative, any other therapeutic nucleic acid (e.g., RNAi) or polypeptide (e.g., cytokine) known in the art can be administered to treat and/or prevent cancer.


As used herein, the term “cancer” encompasses tumor-forming cancers. Likewise, the term “cancerous tissue” encompasses tumors. A “cancer cell antigen” encompasses tumor antigens.


The term “cancer” has its understood meaning in the art, for example, an uncontrolled growth of tissue that has the potential to spread to distant sites of the body (i.e., metastasize). Exemplary cancers include, but are not limited to melanoma, adenocarcinoma, thymoma, lymphoma (e.g., non-Hodgkin's lymphoma, Hodgkin's lymphoma), sarcoma, lung cancer, liver cancer, colon cancer, leukemia, uterine cancer, breast cancer, prostate cancer, ovarian cancer, cervical cancer, bladder cancer, kidney cancer, pancreatic cancer, brain cancer and any other cancer or malignant condition now known or later identified. In some embodiments, a method of treating and/or preventing tumor-forming cancers is provided.


The term “tumor” is also understood in the art, for example, as an abnormal mass of undifferentiated cells within a multicellular organism. Tumors can be malignant or benign. In some embodiments, the methods disclosed herein are used to prevent and treat malignant tumors.


By the terms “treating cancer,” “treatment of cancer” and equivalent terms it is intended that the severity of the cancer is reduced or at least partially eliminated and/or the progression of the disease is slowed and/or controlled and/or the disease is stabilized. In some embodiments, these terms indicate that metastasis of the cancer is prevented or reduced or at least partially eliminated and/or that growth of metastatic nodules is prevented or reduced or at least partially eliminated.


By the terms “prevention of cancer” or “preventing cancer” and equivalent terms it is intended that the methods at least partially eliminate or reduce and/or delay the incidence and/or severity of the onset of cancer. Alternatively stated, the onset of cancer in the subject may be reduced in likelihood or probability and/or delayed.


In some embodiments, cells may be removed from a subject with cancer and contacted with a virus vector expressing a cancer cell antigen as described herein. The modified cell is then administered to the subject, whereby an immune response against the cancer cell antigen is elicited. This method can be advantageously employed with immunocompromised subjects that cannot mount a sufficient immune response in vivo (i.e., cannot produce enhancing antibodies in sufficient quantities).


It is known in the art that immune responses may be enhanced by immunomodulatory cytokines (e.g., alpha-interferon, beta-interferon, gamma-interferon, omega-interferon, tau-interferon, interleukin-1-alpha, interleukin-1p, interleukin-2, interleukin-3, interleukin-4, interleukin 5, interleukin-6, interleukin-7, interleukin-8, interleukin-9, interleukin-10, interleukin-11, interleukin-12, interleukin-13, interleukin-14, interleukin-18, B cell Growth factor, CD40 Ligand, tumor necrosis factor-alpha, tumor necrosis factor-β, monocyte chemoattractant protein-1, granulocyte-macrophage colony stimulating factor, and lymphotoxin). Accordingly, immunomodulatory cytokines (preferably, CTL inductive cytokines) may be administered to a subject in conjunction with the virus vector. Cytokines may be administered by any method known in the art. Exogenous cytokines may be administered to the subject, or alternatively, a nucleic acid encoding a cytokine may be delivered to the subject using a suitable vector, and the cytokine produced in vivo.


Subjects, Pharmaceutical Formulations, and Modes of Administration


Virus vectors and capsids as described herein find use in both veterinary and medical applications. Suitable subjects include both avians and mammals. The term “avian” as used herein includes, but is not limited to, chickens, ducks, geese, quail, turkeys, pheasant, parrots, parakeets, and the like. The term “mammals” as used herein includes, but is not limited to, humans, non-human primates, bovines, ovines, caprines, equines, felines, canines, lagomorphs, etc. Human subjects include neonates, infants, juveniles, adults and geriatric subjects.


In some embodiments, the subject is “in need” of the methods described herein.


In some embodiments, a pharmaceutical composition is provided comprising a virus vector and/or capsid and/or capsid protein and/or virus particle in a pharmaceutically acceptable carrier and, optionally, other medicinal agents, pharmaceutical agents, stabilizing agents, buffers, carriers, adjuvants, diluents, etc. For injection, the carrier will typically be a liquid. For other methods of administration, the carrier may be either solid or liquid. For inhalation administration, the carrier will be respirable, and optionally can be in solid or liquid particulate form.


By “pharmaceutically acceptable” it is meant a material that is not toxic or otherwise undesirable, i.e., the material may be administered to a subject without causing any undesirable biological effects.


Also provided herein are method of transferring a nucleic acid to a cell in vitro. The virus vector may be introduced into the cells at the appropriate multiplicity of infection according to standard transduction methods suitable for the particular target cells. Titers of virus vector to administer can vary, depending upon the target cell type and number, and the particular virus vector, and can be determined by those of skill in the art without undue experimentation. In some embodiments, at least about 103 infectious units, optionally at least about 105 infectious units are introduced to the cell.


The cell(s) into which the virus vector is introduced can be of any type, including but not limited to neural cells (including cells of the peripheral and central nervous systems, in particular, brain cells such as neurons and oligodendrocytes), lung cells, cells of the eye (including retinal cells, retinal pigment epithelium, and corneal cells), epithelial cells (e.g., gut and respiratory epithelial cells), muscle cells (e.g., skeletal muscle cells, cardiac muscle cells, smooth muscle cells and/or diaphragm muscle cells), dendritic cells, pancreatic cells (including islet cells), hepatic cells, myocardial cells, bone cells (e.g., bone marrow stem cells), hematopoietic stem cells, spleen cells, keratinocytes, fibroblasts, endothelial cells, prostate cells, germ cells, and the like. In some embodiments, the cell can be any progenitor cell. As a further possibility, the cell can be a stem cell (e.g., neural stem cell, liver stem cell). As still a further alternative, the cell can be a cancer or tumor cell. Moreover, the cell can be from any species of origin, as indicated above.


The virus vector can be introduced into cells in vitro for the purpose of administering the modified cell to a subject. In some embodiments, the cells have been removed from a subject, the virus vector is introduced therein, and the cells are then administered back into the subject. Methods of removing cells from the subject for manipulation ex vivo, followed by introduction back into the subject are known in the art (see, e.g., U.S. Pat. No. 5,399,346). Alternatively, the recombinant virus vector can be introduced into cells from a donor subject, into cultured cells, or into cells from any other suitable source, and the cells are administered to a subject in need thereof (i.e., a “recipient” subject).


Suitable cells for ex vivo nucleic acid delivery are as described above. Dosages of the cells to administer to a subject will vary upon the age, condition and species of the subject, the type of cell, the nucleic acid being expressed by the cell, the mode of administration, and the like. Typically, at least about 102 to about 108 cells or at least about 103 to about 106 cells will be administered per dose in a pharmaceutically acceptable carrier. In some embodiments, the cells transduced with the virus vector are administered to the subject in a therapeutically effective amount in combination with a pharmaceutical carrier.


In some embodiments, the virus vector is introduced into a cell and the cell can be administered to a subject to elicit an immunogenic response against the delivered polypeptide (e.g., expressed as a transgene or in the capsid). Typically, a quantity of cells expressing an immunogenically effective amount of the polypeptide in combination with a pharmaceutically acceptable carrier is administered. An “immunogenically effective amount” is an amount of the expressed polypeptide that is sufficient to evoke an active immune response against the polypeptide in the subject to which the pharmaceutical formulation is administered. In some embodiments, the dosage is sufficient to produce a protective immune response (as defined above). The degree of protection conferred need not be complete or permanent, as long as the benefits of administering the immunogenic polypeptide outweigh any disadvantages thereof.


Thus, in some embodiments, a method of administering a nucleic acid to a cell comprises contacting the cell with the virus vector, virus particle and/or composition as described herein.


Also provided herein is a method of administering the virus vector, virus particle and/or virus capsid as described herein to a subject. In some embodiments, a method of delivering a nucleic acid to a subject comprises administering to the subject a virus particle, virus vector and/or composition as described herein. Administration of the virus vectors, virus particles and/or capsids to a human subject or an animal in need thereof can be by any means known in the art. Optionally, the virus vector, virus particle and/or capsid is delivered in a therapeutically effective dose in a pharmaceutically acceptable carrier. In some embodiments, a therapeutically effective amount of the virus vector, virus particle and/or capsid is delivered.


The virus vectors and/or capsids described herein can further be administered to elicit an immunogenic response (e.g., as a vaccine). Typically, immunogenic compositions comprise an immunogenically effective amount of virus vector and/or capsid in combination with a pharmaceutically acceptable carrier. Optionally, the dosage is sufficient to produce a protective immune response (as defined above). The degree of protection conferred need not be complete or permanent, as long as the benefits of administering the immunogenic polypeptide outweigh any disadvantages thereof. Subjects and immunogens are as described above.


Dosages of the virus vector and/or capsid to be administered to a subject depend upon the mode of administration, the disease or condition to be treated and/or prevented, the individual subject's condition, the particular virus vector or capsid, and the nucleic acid to be delivered, and the like, and can be determined in a routine manner. Exemplary doses for achieving therapeutic effects are titers of at least about 105, about 106, about 107, about 108, about 109, about 1010, about 1011, about 1012, about 1013, about 1014, or about 1015 transducing units, optionally about 108-1013 transducing units. In some embodiments, the dose of AAV may be from about 2.0×1013 vg/kg body weight of the subject to about 4.0×1013 vg/kg body weight of the subject, such as about 2.0×1013 vg/kg, about 2.1×1013 vg/kg, about 2.2×1013 vg/kg, about 2.3×1013 vg/kg, about 2.4×1013 vg/kg, about 2.5×1013 vg/kg, about 2.6×1013 vg/kg, about 2.7×1013 vg/kg, about 2.8×1013 vg/kg, about 2.9×1013 vg/kg, about 3.0×1013 vg/kg, about 3.1×1013 vg/kg, about 3.2×1013 vg/kg, about 3.3×1013 vg/kg, about 3.4×1013 vg/kg, about 3.5×1013 vg/kg, about 3.6×1013 vg/kg, about 3.7×1013 vg/kg, about 3.8×1013 vg/kg, about 3.9×1013 vg/kg, or about 4.0×1013 vg/kg. In some embodiments, the dose of AAV may be from about 2×1013 vg to about 4.0×1013 vg, such as about 2.0×1013 vg, about 2.1×1013 vg, about 2.2×1013 vg, about 2.3×1013 vg, about 2.4×1013 vg, about 2.5×1013 vg, about 2.6×1013 vg, about 2.7×1013 vg, about 2.8×1013 vg, about 2.9×1013 vg, about 3.0×1013 vg, about 3.1×1013 vg, about 3.2×1013 vg, about 3.3×1013 vg, about 3.4×1013 vg, about 3.5×1013 vg, about 3.6×1013 vg, about 3.7×1013 vg, about 3.8×1013 vg, about 3.9×1013 vg, or about 4.0×1013 vg.


In some embodiments, more than one administration (e.g., two, three, four or more administrations) may be employed to achieve the desired level of gene expression over a period of various intervals, e.g., daily, weekly, monthly, yearly, etc.


Exemplary modes of administration include oral, rectal, transmucosal, intranasal, inhalation (e.g., via an aerosol), buccal (e.g., sublingual), vaginal, intrathecal, intraocular, transdermal, in utero (or in ovo), parenteral (e.g., intravenous, subcutaneous, intradermal, intramuscular [including administration to skeletal, diaphragm and/or cardiac muscle], intradermal, intrapleural, intracerebral, and intraarticular), topical (e.g., to both skin and mucosal surfaces, including airway surfaces, and transdermal administration), intralymphatic, and the like, as well as direct tissue or organ injection (e.g., to liver, skeletal muscle, cardiac muscle, diaphragm muscle or brain). Administration can also be to a tumor (e.g., in or near a tumor or a lymph node). The most suitable route in any given case will depend on the nature and severity of the condition being treated and/or prevented and on the nature of the particular vector that is being used.


Administration to skeletal muscle includes but is not limited to administration to skeletal muscle in the limbs (e.g., upper arm, lower arm, upper leg, and/or lower leg), back, neck, head (e.g., tongue), thorax, abdomen, pelvis/perineum, and/or digits. Suitable skeletal muscles include but are not limited to abductor digiti minimi (in the hand), abductor digiti minimi (in the foot), abductor hallucis, abductor ossis metatarsi quinti, abductor pollicis brevis, abductor pollicis longus, adductor brevis, adductor hallucis, adductor longus, adductor magnus, adductor pollicis, anconeus, anterior scalene, articularis genus, biceps brachii, biceps femoris, brachialis, brachioradialis, buccinator, coracobrachialis, corrugator supercilii, deltoid, depressor anguli oris, depressor labii inferioris, digastric, dorsal interossei (in the hand), dorsal interossei (in the foot), extensor carpi radialis brevis, extensor carpi radialis longus, extensor carpi ulnaris, extensor digiti minimi, extensor digitorum, extensor digitorum brevis, extensor digitorum longus, extensor hallucis brevis, extensor hallucis longus, extensor indicis, extensor pollicis brevis, extensor pollicis longus, flexor carpi radialis, flexor carpi ulnaris, flexor digiti minimi brevis (in the hand), flexor digiti minimi brevis (in the foot), flexor digitorum brevis, flexor digitorum longus, flexor digitorum profundus, flexor digitorum superficial is, flexor hallucis brevis, flexor hallucis longus, flexor pollicis brevis. flexor pollicis longus, frontalis, gastrocnemius, geniohyoid, gluteus maximus, gluteus medius, gluteus minimus, gracilis, iliocostalis cervicis, iliocostalis lumborum, iliocostalis thoracis, illiacus, inferior gemellus, inferior oblique, inferior rectus, infraspinatus, interspinalis, intertransversi, lateral pterygoid, lateral rectus, latissimus dorsi, levator anguli oris, levator labii superioris, levator labii superioris alaeque nasi, levator palpebrae superioris, levator scapulae, long rotators, longissimus capitis, longissimus cervicis, longissimus thoracis, longus capitis, longus colli, lumbricals (in the hand), lumbricals (in the foot), masseter, medial pterygoid, medial rectus, middle scalene, multifidus, mylohyoid, obliquus capitis inferior, obliquus capitis superior, obturator externus, obturator internus, occipitalis, omohyoid, opponens digiti minimi, opponens pollicis, orbicularis oculi, orbicularis oris, palmar interossei, palmaris brevis, palmaris longus, pectineus, pectoralis major, pectoralis minor, peroneus brevis, peroneus longus, peroneus tertius, piriformis, plantar interossei, plantaris, platysma, popliteus, posterior scalene, pronator quadratus, pronator teres, psoas major, quadratus femoris, quadratus plantae, rectus capitis anterior, rectus capitis lateralis, rectus capitis posterior major, rectus capitis posterior minor, rectus femoris, rhomboid major, rhomboid minor, risorius, sartorius, scalenus minimus, semimembranosus, semispinalis capitis, semispinalis cervicis, semispinalis thoracis, semitendinosus, serratus anterior, short rotators, soleus, spinalis capitis, spinalis cervicis, spinalis thoracis, splenius capitis, splenius cervicis, sternocleidomastoid, sternohyoid, sternothyroid, stylohyoid, subclavius, subscapularis, superior gemellus, superior oblique, superior rectus, supinator, supraspinatus, temporalis, tensor fascia lata, teres major, teres minor, thoracis, thyrohyoid, tibialis anterior, tibialis posterior, trapezius, triceps brachii, vastus intermedius, vastus lateralis, vastus medialis, zygomaticus major, and zygomaticus minor, and any other suitable skeletal muscle as known in the art.


The virus vector and/or capsid can be delivered to skeletal muscle by intravenous administration, intra-arterial administration, intraperitoneal administration, limb perfusion, (optionally, isolated limb perfusion of a leg and/or arm; see, e.g. Arruda et al., (2005) Blood 105: 3458-3464), and/or direct intramuscular injection. In some embodiments, the virus vector and/or capsid is administered to a limb (arm and/or leg) of a subject (e.g., a subject with muscular dystrophy such as Duchenne muscular dystrophy (DMD) or limb-girdle muscular dystrophy (LGMD)) by limb perfusion, optionally isolated limb perfusion (e.g., by intravenous or intra-articular administration). In some embodiments, the virus vectors and/or capsids can advantageously be administered without employing “hydrodynamic” techniques. Tissue delivery (e.g., to muscle) of prior art vectors is often enhanced by hydrodynamic techniques (e.g., intravenous/intravenous administration in a large volume), which increase pressure in the vasculature and facilitate the ability of the vector to cross the endothelial cell barrier. In some embodiments, the viral vectors and/or capsids can be administered in the absence of hydrodynamic techniques such as high volume infusions and/or elevated intravascular pressure (e.g., greater than normal systolic pressure, for example, less than or equal to a 5%, 10%, 15%, 20%, 25% increase in intravascular pressure over normal systolic pressure). Such methods may reduce or avoid the side effects associated with hydrodynamic techniques such as edema, nerve damage and/or compartment syndrome. Administration to cardiac muscle includes administration to the left atrium, right atrium, left ventricle, right ventricle and/or septum. The virus vector and/or capsid can be delivered to cardiac muscle by intravenous administration, intra-arterial administration such as intra-aortic administration, direct cardiac injection (e.g., into left atrium, right atrium, left ventricle, right ventricle), and/or coronary artery perfusion.


Administration to diaphragm muscle can be by any suitable method including intravenous administration, intra-arterial administration, and/or intra-peritoneal administration.


Delivery to a target tissue can also be achieved by delivering a depot comprising the virus vector and/or capsid. In some embodiments, a depot comprising the virus vector and/or capsid is implanted into skeletal, cardiac and/or diaphragm muscle tissue or the tissue can be contacted with a film or other matrix comprising the virus vector and/or capsid. Such implantable matrices or substrates are described in U.S. Pat. No. 7,201,898.


In some embodiments, a virus vector and/or virus capsid according is administered to skeletal muscle, diaphragm muscle and/or cardiac muscle (e.g., to treat and/or prevent muscular dystrophy, heart disease [for example, PAD or congestive heart failure]).


In some embodiments, the compositions and methods described herein are used to treat and/or prevent diseases or disorders of skeletal, cardiac and/or diaphragm muscle. The diseases or disorders of the muscle may be, for example, muscular dystrophy, myopathy, motor neuron disease, and cardiomyopathy. The diseases or disorders of the muscle may be, for example, dystrophinopathies, Duchenne muscular dystrophy, Becker muscular dystrophy, myotonic dystrophies (e.g., myotonic dystrophy 1 and 2), facioscapulohumeral muscular dystrophy (FDHD), Eimery-Dreifuss muscular dystrophy, limb-girdle disease, facioscapulohumeral muscular dystrophy, oculopharyngeal muscular dystrophy, distal muscular dystrophy, congenital muscular dystrophy, juvenile macular dystrophy, centronuclear myopathy, central core myopathy, and inclusion body myositis.


In some embodiments, a method of treating and/or preventing muscular dystrophy in a subject in need thereof is provided, the method comprising: administering a treatment or prevention effective amount of a virus vector to a mammalian subject, wherein the virus vector comprises a heterologous nucleic acid encoding dystrophin, a mini-dystrophin, a micro-dystrophin, myostatin propeptide, follistatin, activin type II soluble receptor, IGF-1, anti-inflammatory polypeptides such as the Ikappa B dominant mutant, sarcospan, utrophin, a micro-dystrophin, laminin-a2, alpha-sarcoglycan, beta-sarcoglycan, gamma-sarcoglycan, delta-sarcoglycan, IGF-1, an antibody or antibody fragment against myostatin or myostatin propeptide, and/or RNAi against myostatin. In some embodiments, the virus vector can be administered to skeletal, diaphragm and/or cardiac muscle as described elsewhere herein.


Alternatively, methods described herein can be practiced to deliver a nucleic acid to skeletal, cardiac or diaphragm muscle, which is used as a platform for production of a polypeptide (e.g., an enzyme) or functional RNA (e.g., RNAi, micro RNA, antisense RNA) that normally circulates in the blood or for systemic delivery to other tissues to treat and/or prevent a disorder (e.g., a metabolic disorder, such as diabetes [e.g., insulin], hemophilia [e.g., Factor IX or Factor VIII], a mucopolysaccharide disorder [e.g., Sly syndrome, Hurler Syndrome, Scheie Syndrome, Hurler-Scheie Syndrome, Hunter's Syndrome, Sanfilippo Syndrome A, B, C, D, Morquio Syndrome, Maroteaux-Lamy Syndrome, etc.] or a lysosomal storage disorder such as Gaucher's disease [glucocerebrosidase] or Fabry disease [a-galactosidase A] or a glycogen storage disorder such as Pompe disease [Iysosomal acid alpha glucosidase]). Other suitable proteins for treating and/or preventing metabolic disorders are described herein. The use of muscle as a platform to express a nucleic acid of interest is described in U.S. Patent publication US 2002/0192189.


In some embodiments, a method of treating and/or preventing a metabolic disorder in a subject in need thereof comprises administering a treatment or prevention effective amount of a virus vector to skeletal muscle of a subject, wherein the virus vector comprises a heterologous nucleic acid encoding a polypeptide, wherein the metabolic disorder is a result of a deficiency and/or defect in the polypeptide. Illustrative metabolic disorders and heterologous nucleic acids encoding polypeptides are described herein. Optionally, the polypeptide is secreted (e.g., a polypeptide that is a secreted polypeptide in its native state or that has been engineered to be secreted, for example, by operable association with a secretory signal sequence as is known in the art). Without being limited by any particular theory, according to this embodiment, administration to the skeletal muscle can result in secretion of the polypeptide into the systemic circulation and delivery to target tissue(s). Methods of delivering virus vectors to skeletal muscle is described in more detail herein.


The methods described herein can also be practiced to produce noncoding RNA, such as antisense RNA, RNAi or other functional RNA (e.g., a ribozyme) for systemic delivery.


In some embodiments, a method of treating and/or preventing congenital heart failure or PAD in a subject in need thereof comprises administering a treatment or prevention effective amount of a virus vector to a mammalian subject, wherein the virus vector comprises a heterologous nucleic acid encoding, for example, a sarcoplasmic endoreticulum Ca2+-ATPase (SERCA2a), an angiogenic factor, phosphatase inhibitor I (I-1) and fragments thereof (e.g., 11C), RNAi against phospholamban; a phospholamban inhibitory or dominant-negative molecule such as phospholamban S16E, a zinc finger protein that regulates the phospholamban gene, beta-2-adrenergic receptor, beta-2-adrenergic receptor kinase (BARK), PI3 kinase, calsarcan, a β-adrenergic receptor kinase inhibitor (PARKct), inhibitor 1 of protein phosphatase 1 and fragments thereof (e.g., I1 C), S100A1, parvalbumin, adenylyl cyclase type 6, a molecule that effects G-protein coupled receptor kinase type 2 knockdown such as a truncated constitutively active bARKct, Pim-1, PGC-I α, SOD-1, SOD-2, EC-SOD, kallikrein, HIF, thymosin-p4, mir-1, mir-133, mir-206, mir-208 and/or mir-26a.


Injectables can be prepared in conventional forms, either as liquid solutions or suspensions, solid forms suitable for solution or suspension in liquid prior to injection, or as emulsions. Alternatively, one may administer the virus vector and/or virus capsids in a local rather than systemic manner, for example, in a depot or sustained-release formulation. Further, the virus vector and/or virus capsid can be delivered adhered to a surgically implantable matrix (e.g., as described in U.S. Patent Publication No. US-2004-0013645-A1).


The virus vectors and/or virus capsids disclosed herein can be administered to the lungs of a subject by any suitable means, optionally by administering an aerosol suspension of respirable particles comprised of the virus vectors and/or virus capsids, which the subject inhales. The respirable particles can be liquid or solid. Aerosols of liquid particles comprising the virus vectors and/or virus capsids may be produced by any suitable means, such as with a pressure-driven aerosol nebulizer or an ultrasonic nebulizer, as is known to those of skill in the art. See, e.g., U.S. Pat. No. 4,501,729. Aerosols of solid particles comprising the virus vectors and/or capsids may likewise be produced with any solid particulate medicament aerosol generator, by techniques known in the pharmaceutical art.


The virus vectors and virus capsids can be administered to tissues of the CNS (e.g., brain, eye) and may advantageously result in broader distribution of the virus vector or capsid than would be observed in the absence of the compositions and methods described herein.


In some embodiments, the delivery vectors described herein may be administered to treat diseases of the CNS, including genetic disorders, neurodegenerative disorders, psychiatric disorders and tumors. Illustrative diseases of the CNS include, but are not limited to Adrenomyeloneuropathy (AMN), Alzheimer's disease, Angelman Syndrome, Frontotemporal Dementia, Parkinson's disease, Huntington's disease, Fragile X syndrome, Canavan disease, Leigh's disease, Refsum disease, Tourette syndrome, primary lateral sclerosis, amyotrophic lateral sclerosis, progressive muscular atrophy, Pick's disease, muscular dystrophy, multiple sclerosis, myasthenia gravis, Binswanger's disease, trauma due to spinal cord or head injury, Tay Sachs disease (GM2 Gangliosidosis), Lesch-Nyhan disease, MC4R Obesity, Metachromatic Leukodystrophy (MLD), MPS I (Hurler/Scheie), MPS IIIA (Sanfilippo A), Niemann Pick C1, Rett Syndrome, Spinal Muscular Atrophy (SMA), AADC Deficiency, Monogenic Amyotropic Lateral Sclerosis (ALS), Alpha mannosidosis, Aspartylglucosaminuria, Dravet Syndrome, Giant Axonal Neuropathy, Globoid Cell Leukodystrophy (Krabbe), Glut 1 Deficiency, GM1 Gangliosidosis, Infantile Neuronal Ceroid Lipfuscinosis (INCL, Batten), Juvenile Neuronal Ceroid Lipfuscinosis (JNCL, Batten), Late Infantile Neuronal Ceroid Lipfuscinosis (LINCL, Batten), MPS II (Hunter), MPS IIIB (Sanfilippo B), MPS IIIC (Sanfilippo C), MPS IVA (Morquio Syndrome), MPS VI (Maroteaux-Lamy), Peroxisome Biogenesis Disorders (Zellweger Syndrome Spectrum), Sandhoff Disease (GM2 Gangliosidosis), epilepsy, cerebral infarcts, psychiatric disorders including mood disorders (e.g., depression, bipolar affective disorder, persistent affective disorder, secondary mood disorder), schizophrenia, drug dependency (e.g., alcoholism and other substance dependencies), neuroses (e.g., anxiety, obsessional disorder, somatoform disorder, dissociative disorder, grief, post-partum depression), psychosis (e.g., hallucinations and delusions), dementia, paranoia, attention deficit disorder, psychosexual disorders, sleeping disorders, pain disorders, eating or weight disorders (e.g., obesity, cachexia, anorexia nervosa, and bulemia) and cancers and tumors (e.g., pituitary tumors) of the CNS.


Disorders of the CNS include ophthalmic disorders involving the retina, posterior tract, and optic nerve (e.g., retinitis pigmentosa, diabetic retinopathy and other retinal degenerative diseases, uveitis, age-related macular degeneration, glaucoma).


Most, if not all, ophthalmic diseases and disorders are associated with one or more of three types of indications: (1) angiogenesis, (2) inflammation, and (3) degeneration. The viral vectors described herein can be employed to deliver anti-angiogenic factors; anti-inflammatory factors; factors that retard cell degeneration, promote cell sparing, or promote cell growth and combinations of the foregoing.


Diabetic retinopathy, for example, is characterized by angiogenesis. Diabetic retinopathy can be treated by delivering one or more anti-angiogenic factors either intraocularly (e.g., in the vitreous) or periocularly (e.g., in the sub-Tenon's region). One or more neurotrophic factors may also be co-delivered, either intraocularly (e.g., intravitreally) or periocularly.


Uveitis involves inflammation. One or more anti-inflammatory factors can be administered by intraocular (e.g., vitreous or anterior chamber) administration of a delivery vector.


Retinitis pigmentosa, by comparison, is characterized by retinal degeneration. In some embodiments, retinitis pigmentosa can be treated by intraocular (e.g., vitreal administration) of a delivery vector encoding one or more neurotrophic factors.


Age-related macular degeneration involves both angiogenesis and retinal degeneration. This disorder can be treated by administering the inventive delivery vectors encoding one or more neurotrophic factors intraocularly (e.g., vitreous) and/or one or more anti-angiogenic factors intraocularly or periocularly (e.g., in the sub-Tenon's region).


Glaucoma is characterized by increased ocular pressure and loss of retinal ganglion cells. Treatments for glaucoma include administration of one or more neuroprotective agents that protect cells from excitotoxic damage using the inventive delivery vectors. Such agents include N-methyl-D-aspartate (NMDA) antagonists, cytokines, and neurotrophic factors, delivered intraocularly, optionally intravitreally.


In some embodiments, the compositions and methods described herein may be used to treat seizures, e.g., to reduce the onset, incidence or severity of seizures. The efficacy of a therapeutic treatment for seizures can be assessed by behavioral (e.g., shaking, ticks of the eye or mouth) and/or electrographic means (most seizures have signature electrographic abnormalities). Thus, epilepsy, which is marked by multiple seizures over time, may also be treated.


In some embodiments, a method of treating a subject in need thereof comprises administering to the subject an AAV vector comprising a capsid protein, wherein the capsid protein comprises the amino acid sequence of any one of SEQ ID NO: 165-187. In some embodiments, the AAV vector comprises a capsid protein comprising the amino acid sequence of SEQ ID NO: 175, or a sequence at least 95% identical thereto. In some embodiments, the AAV vector comprises a capsid protein comprising the amino acid sequence of SEQ ID NO: 175, or a sequence at least 95% identical thereto. In some embodiments, the subject has Dravet syndrome. In some embodiments, the subject has Rett syndrome. In some embodiments, the subject has Angelman syndrome. In some embodiments, the subject has Niemann-Pick disease. In some embodiments, the subject has Fragile X syndrome. In some embodiments, the subject has Alzheimer's disease. In some embodiments, the subject has Gaucher's disease. In some embodiments, the subject has Huntington's disease. In some embodiments, the subject has Parkinson's disease. In some embodiments, the subject has Friedrich's ataxia. In some embodiments, the AAV vector is administered to the subject by intracerebroventricular (ICV) injection. In some embodiments, the AAV vector is administered to the subject by intrathecal (IT) injection. In some embodiments, the AAV vector is administered to the subject by intravenous (IV) injection.


In some embodiments, a method of treating a subject in need thereof comprises administering to the subject an AAV vector comprising a capsid protein, wherein the capsid protein comprises the amino acid sequence of SEQ ID NO: 175 or 180, wherein the subject has Dravet syndrome, Rett syndrome, Angelman syndrome, Niemann-Pick disease, or Fragile X syndrome, and wherein the AAV vector is administered to the subject by ICV or IT injection.


In some embodiments, a method of treating a subject in need thereof comprises administering to the subject an AAV vector comprising a capsid protein, wherein the capsid protein comprises the amino acid sequence of SEQ ID NO: 175 or 180, wherein the subject has Gaucher's disease, Huntington's disease, Parkinson's disease, or Friedrich's ataxia, and wherein the AAV vector is administered to the subject by ICV or IT injection.


In some embodiments, somatostatin (or an active fragment thereof) is administered to the brain using a delivery vector to treat a pituitary tumor. According to this embodiment, the delivery vector encoding somatostatin (or an active fragment thereof) is administered by microinfusion into the pituitary. Likewise, such treatment can be used to treat acromegaly (abnormal growth hormone secretion from the pituitary). The nucleic acid (e.g., GenBank Accession No. J00306) and amino acid (e.g., GenBank Accession No. P01166; contains processed active peptides somatostatin-28 and somatostatin-14) sequences of somatostatins are known in the art.


In some embodiments, the vector can comprise a secretory signal as described in U.S. Pat. No. 7,071,172.


In some embodiments, the virus vector and/or virus capsid is administered to the CNS (e.g., to the brain or to the eye). The virus vector and/or capsid may be introduced into the spinal cord, brainstem (medulla oblongata, pons), midbrain (hypothalamus, thalamus, epithalamus, pituitary gland, substantia nigra, pineal gland), cerebellum, telencephalon (corpus striatum, cerebrum including the occipital, temporal, parietal and frontal lobes, cortex, basal ganglia, hippocampus and portaamygdala), limbic system, neocortex, corpus striatum, cerebrum, and inferior colliculus. The virus vector and/or capsid may also be administered to different regions of the eye such as the retina, cornea and/or optic nerve.


The virus vector and/or capsid may be delivered into the cerebrospinal fluid (e.g., by lumbar puncture) for more disperse administration of the delivery vector. The virus vector and/or capsid may further be administered intravascularly to the CNS in situations in which the blood-brain barrier has been perturbed (e.g., brain tumor or cerebral infarct).


The virus vector and/or capsid can be administered to the desired region(s) of the CNS by any route known in the art, including but not limited to, intrathecal, intra-ocular, intracerebral, intraventricular, intravenous (e.g., in the presence of a sugar such as mannitol), intranasal, intra-aural, intra-ocular (e.g., intra-vitreous, sub-retinal, anterior chamber) and peri-ocular (e.g., sub-Tenon's region) delivery as well as intramuscular delivery with retrograde delivery to motor neurons. In some embodiments, the virus vector and/or capsid is administered in a liquid formulation by direct injection (e.g., stereotactic injection) to the desired region or compartment in the CNS. In some embodiments, the virus vector and/or capsid may be provided by topical application to the desired region or by intra-nasal administration of an aerosol formulation. Administration to the eye, may be by topical application of liquid droplets. As a further alternative, the virus vector and/or capsid may be administered as a solid, slow-release formulation (see, e.g., U.S. Pat. No. 7,201,898).


In some embodiments, the virus vector can used for retrograde transport to treat and/or prevent diseases and disorders involving motor neurons (e.g., amyotrophic lateral sclerosis (ALS); spinal muscular atrophy (SMA), etc.). For example, the virus vector can be delivered to muscle tissue from which it can migrate into neurons.


Numbered Embodiments

Notwithstanding the appended claims, the disclosure sets forth the following numbered embodiments:


1. An adeno-associated virus (AAV) vector comprising (i) a recombinant capsid protein and (ii) a cargo nucleic acid encapsidated by the capsid protein, wherein the capsid protein comprises a peptide having the sequence of any one of SEQ ID NO: 12-20.


2. The AAV vector of embodiment 1, wherein the cargo nucleic acid comprises 5′ and 3′ AAV inverted terminal repeats.


3. The AAV vector of embodiment 1 or 2, wherein the cargo nucleic acid comprises a transgene.


4. The AAV vector of embodiment 3, wherein the transgene encodes a therapeutic protein or RNA.


5. The AAV vector of any one of embodiments 1-4, wherein the recombinant capsid protein has at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99% sequence identity to the native sequence of the AAV1, AAV2, AAV3, AAV4, AAV5, AAV6, AAV7, AAV8, AAV9, AAV10, AAV11, AAV12, AAVrh.8, AAVrh.10, AAVrh32.33, AAVrh74, bovine AAV or avian AAV capsid.


6. The AAV vector of embodiment 5, wherein the recombinant capsid protein has at least 90% sequence identity to the native sequence of the AAV9 capsid.


7. The AAV vector of any one of embodiments 1-6, wherein the peptide is located at the amino acid positions corresponding to amino acids 451-458 of the native AAV9 capsid, or the equivalent amino acid residues in AAV1, AAV2, AAV3, AAV4, AAV5, AAV6, AAV7, AAV8, AAV10, AAV11, AAV12, AAVrh.8, AAVrh.10, AAVrh32.33, AAVrh74, bovine AAV or avian AAV, and wherein the peptide is selected from any one of SEQ ID NO: 12-18.


8. The AAV vector of any one of embodiments 1-6, wherein the peptide is located at the amino acid positions corresponding to amino acids 587-594 of the native AAV9 capsid, or the equivalent amino acid residues in AAV1, AAV2, AAV3, AAV4, AAV5, AAV6, AAV7, AAV8, AAV10, AAV11, AAV12, AAVrh.8, AAVrh.10, AAVrh32.33, AAVrh74, bovine AAV or avian AAV, and wherein the peptide is selected from SEQ ID NO: 19 or 20.


9. The AAV vector of embodiment 1, wherein the recombinant capsid protein comprises: a) a first peptide having a sequence of any one of SEQ ID NO: 12-18; and b) a second peptide having a sequence of any one of SEQ ID NO: 19-20.


10. The AAV vector of embodiment 9, wherein the first peptide is at amino acid positions 451-458, and the second peptide is at amino acids 587-594, wherein the amino acid numbering is based on the native AAV9 capsid, or the equivalent amino acid residues in AAV1, AAV2, AAV3, AAV4, AAV5, AAV6, AAV7, AAV8, AAV10, AAV11, AAV12, AAVrh.8, AAVrh.10, AAVrh32.33, AAVrh74, bovine AAV or avian AAV.


11. The AAV vector of any one of embodiments 1-10, wherein the peptide inhibits binding of at least one antibody to the capsid protein.


12. The AAV vector of embodiment 11, wherein the peptide inhibits neutralization of infectivity of the AAV vector by the antibody.


13. The AAV vector of any one of embodiments 1-12, wherein the peptide selectively binds to a receptor expressed on the surface of a cell in the central nervous system (CNS).


14. The AAV vector of embodiment 13, wherein the cell is in the premotor cortex, the thalamus, the cerebellar cortex, the dentate nucleus, the spinal cord, or the dorsal root ganglion.


15. The AAV vector of any one of embodiments 1-14, wherein the peptide selectively binds to a receptor expressed on the surface of a cell in the heart.


16. The AAV vector of any one of embodiments 1-15, wherein the capsid protein further comprises a peptide that modifies the HI loop of the capsid.


17. An adeno-associated virus (AAV) vector comprising (i) a mutant AAV9 capsid protein and (ii) a cargo nucleic acid encapsidated by the capsid protein, wherein the capsid protein comprises a peptide having the sequence X1-X2-X3-X4-X5-X6-X7-X8 (SEQ ID NO: 158) at amino acids 451-458 of the native AAV9 capsid protein sequence, wherein the peptide does not occur in the native AAV9 capsid protein sequence.


18. The AAV vector of embodiment 17, wherein X1 is not I, X2 is not N, X3 is not G, X4 is not S, X5 is not G, X6 is not Q, X7 is not N, and/or X8 is not Q.


19. The AAV vector of embodiment 18, wherein X1 is S, F, Q, G, K, or R.


20. The AAV vector of embodiment 18 or 19, wherein X2 is C, G, R, D, T, or Q.


21. The AAV vector of any one of embodiments 18-20, wherein X3 is Q, V, G, Y, R, F, or D.


22. The AAV vector of any one of embodiments 18-21, wherein X4 is P, Q, A, or R.


23. The AAV vector of any one of embodiments 18-22, wherein X5 is T, N, A, P, or I.


24. The AAV vector of anyone of embodiments 18-23, wherein X6 is V, Q, A, or I.


25. The AAV vector of any one of embodiments 18-24, wherein X7 is M, P, R, Q, or N.


26. The AAV vector of any one of embodiments 18-25, wherein X8 is N, L, F, E, H, or A.


27. The AAV vector of embodiment 17, wherein X1 is S, X2 is C, X3 is Q, X4 is P, X5 is T, X6 is V, X7 is M, and X8 is N.


28. The AAV vector of embodiment 17, wherein X1 is F, X2 is G, X3 is V, X4 is P, X5 is N, X6 is Q, X7 is P, and X8 is L.


29. The AAV vector of embodiment 17, wherein X1 is Q, X2 is R, X3 is G, X4 is Q, X5 is A, X6 is A, X7 is P, and X8 is F.


30. The AAV vector of embodiment 17, wherein X1 is G, X2 is D, X3 is Y, X4 is A, X5 is P, X6 is I, X7 is R, and X8 is E.


31. The AAV vector of embodiment 17, wherein X1 is K, X2 is T, X3 is R, X4 is R, X5 is I, X6 is V, X7 is Q, and X8 is H.


32. The AAV vector of embodiment 17, wherein X1 is F, X2 is G, X3 is F, X4 is P, X5 is N, X6 is Q, X7 is P, and X8 is L.


33. The AAV vector of embodiment 17, wherein X1 is R, X2 is Q, X3 is D, X4 is Q, X5 is P, X6 is I, X7 is N, and X8 is A.


34. An adeno-associated virus (AAV) vector comprising (i) a mutant AAV9 capsid protein and (ii) a cargo nucleic acid encapsidated by the capsid protein, wherein the capsid protein comprises a peptide having the sequence X1-X2-X3-X4-X5-X6-X7-X8 (SEQ ID NO: 158) at amino acids 587-594 of the native AAV9 capsid protein sequence, wherein the peptide does not occur in the native AAV9 capsid protein sequence.


35. The AAV vector of embodiment 34, wherein X1 is not A, X2 is not Q, X3 is not A, X4 is not Q, X5 is not A, X6 is not Q, X7 is not T, and/or X8 is not G.


36. The AAV vector of embodiment 35, wherein X1 is S.


37. The AAV vector of embodiment 35 or 36, wherein X2 is K or T.


38. The AAV vector of any one of embodiments 35-37, wherein X3 is V.


39. The AAV vector of any one of embodiments 35-38, wherein X4 is E or D.


40. The AAV vector of any one of embodiments 35-39, wherein X5 is S.


41. The AAV vector of any one of embodiments 35-40, wherein X6 is W or I.


42. The AAV vector of any one of embodiments 35-41, wherein X7 is T or A.


43. The AAV vector of any one of embodiments 35-42, wherein X8 is E or I.


44. The AAV vector of embodiment 34, wherein X1 is S, X2 is K, X3 is V, X4 is E, X5 is S, X6 is W, X7 is T, and X8 is E.


45. The AAV vector of embodiment 34, wherein X1 is S, X2 is T, X3 is V, X4 is D, X5 is S, X6 is I, X7 is A, and X8 is I.


46. An adeno-associated virus (AAV) vector comprising (i) a recombinant capsid protein and (ii) a cargo nucleic acid encapsidated by the capsid protein, wherein the capsid protein comprises an amino acid sequence that is at least 95%, at least 96%, at least 97%, at least 98%, or at least 99% identical to any one of SEQ ID NO: 165-187.


47. The AAV vector of embodiment 46, wherein the capsid protein comprises the amino acid sequence of any one of SEQ ID NO: 165-187.


48. The AAV vector of embodiment 47, wherein the capsid protein comprises the amino acid sequence of SEQ ID NO: 175.


49. The AAV vector of embodiment 47, wherein the capsid protein comprises the amino acid sequence of SEQ ID NO: 180.


50. The AAV vector of any one of embodiments 46-49, wherein the AAV vector selectively delivers the cargo nucleic acid to a cell or tissue of the central nervous system.


51. The AAV vector of embodiment 50, wherein the tissue of the central nervous system is the premotor cortex, the thalamus, the cerebellar cortex, the dentate nucleus, the spinal cord, or the dorsal root ganglion.


52. The AAV vector of any one of embodiments 46-49, wherein the AAV vector delivers the cargo nucleic acid to the brain, but does not deliver the AAV vector to the heart.


53. The AAV vector of any one of embodiments 46-49, wherein the AAV vector delivers the cargo nucleic acid to the brain and to the heart.


54. The AAV vector of embodiment 53, wherein delivery of the cargo nucleic acid is greater to the brain than to the heart.


55. The AAV vector of embodiment 53, wherein delivery of the cargo nucleic acid is approximately equal in the brain in the heart.


56. A nucleic acid sequence encoding the recombinant capsid protein of the AAV vector of any one of embodiments 1 to 55.


57. The nucleic acid sequence of embodiment 56, wherein the nucleic acid sequence is a DNA sequence.


58. The nucleic acid sequence of embodiment 56, wherein the nucleic acid sequence is an RNA sequence.


59. An expression vector comprising the nucleic acid sequence of any one of embodiments 56-58.


60. A cell comprising the nucleic acid sequence of any one of embodiments 56-58.


61. A cell comprising the expression vector of embodiment 59.


62. A pharmaceutical composition comprising the AAV vector of any one of embodiments 1-55.


63. The pharmaceutical composition of embodiment 62, wherein the composition further comprises a pharmaceutically acceptable carrier.


64. A pharmaceutical composition comprising the cell of embodiment 60 or 61.


65. The pharmaceutical composition of embodiment 64, wherein the composition further comprises a pharmaceutically acceptable carrier.


66. A method of treating a subject in need thereof comprising administering to the subject a therapeutically effective amount of the AAV vector of any one of embodiments 1-55.


67. The method of embodiment 66, wherein the subject has a disease or disorder of the central nervous system.


68. The method of embodiment 67, wherein the disease or disorder of the central nervous system is Dravet syndrome.


69. The method of embodiment 67, wherein the disease or disorder of the central nervous system is Rett syndrome.


70. The method of embodiment 67, wherein the disease or disorder of the central nervous system is Angelman syndrome.


71. The method of embodiment 67, wherein the disease or disorder of the central nervous system is Niemann-Pick disease.


72. The method of embodiment 67, wherein the disease or disorder of the central nervous system is Fragile X syndrome.


73. The method of embodiment 67, wherein the disease or disorder of the central nervous system is Gaucher's disease.


74. The method of embodiment 67, wherein the disease or disorder of the central nervous system is Huntington's disease.


75. The method of embodiment 67, wherein the disease or disorder of the central nervous system is Parkinson's disease.


76. The method of embodiment 67, wherein the disease or disorder of the central nervous system is Friedrich's ataxia.


77. The method of any one of embodiments 66-76, wherein AAV vector comprises a capsid protein, wherein the capsid protein comprises the amino acid sequence of any one of SEQ ID NO: 165-187.


78. The method of any one of embodiments 66-76, wherein AAV vector comprises a capsid protein, wherein the capsid protein comprises the amino acid sequence of SEQ ID NO: 175.


79. The method of any one of embodiments 66-76, wherein AAV vector comprises a capsid protein, wherein the capsid protein comprises the amino acid sequence of SEQ ID NO: 180.


80. The method of any one of embodiments 66-79, wherein the AAV vector is administered to the subject by intracerebroventricular (ICV) injection.


81. The method of any one of embodiments 66-79, wherein the AAV vector is administered to the subject by intrathecal (IT) injection.


82. The method of any one of embodiments 66-79, wherein the AAV vector is administered to the subject by intravenous (IV) injection.


83. The method of any one of embodiments 66-82, wherein the subject is a mammal.


84. The method of embodiment 83, wherein the subject is a human.


85. An in vitro method of introducing a nucleic acid molecule into a cell, comprising contacting the cell with the AAV vector of any one of embodiments 1-55.


86. An AAV vector of any one of embodiments 1-55 for use as a medicament.


87. An AAV vector of any one of embodiments 1-55 for use in a method of treatment of a subject in need thereof.


88. An AAV vector of any one of embodiments 1-55 for use in a method of treating or preventing a disease or disorder of the CNS in a subject in need thereof.


EXAMPLES

The following examples, which are included herein for illustration purposes only, are not intended to be limiting. As used herein, the terms STRD.101 and STRD.102 are used to describe capsid protein sequences, and AAV-STRD.101 and AAV-STRD.102 are used to describe AAV vectors comprising the capsid proteins. However, the terms STRD.101 and STRD.102 may be used in some contexts to describe AAV vectors comprising the named capsids, as will be apparent to the skilled artisan.


Example 1. Combinatorial Engineering and Selection of Antibody-Evading AAV Vectors

The method for generating antibody evading AAV mutants is as follows. The first step involves identification of conformational 3D antigenic epitopes on the AAV capsid surface, for example using cryo-electron microscopy. Selected residues within antigenic motifs are then subjected to mutagenesis using degenerate primers with each codon substituted by nucleotides NNK and gene fragments combined together by Gibson assembly and/or multistep PCR. Capsid-encoding genes containing a degenerate library of mutated antigenic motifs are cloned into a wild type AAV genome to replace the original Cap encoding DNA sequence, yielding a plasmid library. Plasmid libraries are then transfected into 293 producer cell lines with an adenoviral helper plasmid to generate AAV capsid libraries, which can then be subjected to selection. Successful generation of AAV libraries is confirmed via DNA sequencing.


In order to select for new AAV strains that can escape neutralizing antibodies (NAbs) and/or target the central nervous system (CNS), AAV libraries are subjected to multiple rounds of infection in non-human primates. At each stage, tissues of interest are isolated from animal subjects. Cell lysates harvested from the tissues of interest are sequenced to identify AAV isolates escaping antibody neutralization. After multiple rounds of infection in non-human primates, the isolated sequences from each mutagenized region are combined in all permutations and combinations.


As a specific example, a common antigenic motif on an AAV capsid protein (VP1) was subjected to mutagenesis as described above. The degenerate libraries (FIG. 1A) were then subjected to a first round of infection in a non-human primate (intravenous injection). Tissues were harvested at day 7 post-infection and sequenced to identify single AAV isolates.


Various recombinant AAV isolates were identified in tissue samples, including the spinal cord, dorsal root ganglion, frontal lobe, temporal lobe, occipital lobe, putamen, globus pallidus, thalamus, amygdala, hippocampus, substantia nigra, pons, cerebellum, medulla. Results from this first round of evolution are shown in FIG. 1B.


The recombinant AAVs isolated during the first round of evolution (FIG. 1B) were then reintroduced into a second non-human primate. Tissues were harvested at day 7 post-infection and sequenced to identify single AAV isolates. The results from this second round of evolution are shown in FIG. 1C.


Recombinant AAVs with the highest frequency were sequenced. Substitutions present in these AAVs are shown in Tables 6.1 and 6.2. These data demonstrate that recombinant AAV virions having capsid proteins comprising the substitutions listed in Tables 6.1 and 6.2 evade neutralizing antibodies in vivo in non-human primates and have a tropism for the desired target tissues.


Example 2: Manufacturability of Recombinant AAV Vectors

To determine whether various recombinant AAVs identified in Example 1 may be manufactured in large-scale systems, the AAVs were produced according to standard methods, and yield was compared to that of wildtype AAV vectors.


AAVs were produced in HEK293 cells according to a standard triple transfection protocol. Briefly, the cells were transfected with (i) a plasmid comprising either the wildtype AAV9 capsid sequence, the STRD.101 capsid variant sequence (SEQ ID NO: 180), or the STRD.102 capsid variant sequence (SEQ ID NO: 175), (ii) a plasmid comprising a 5′ITR, a transgene, and a 3′ ITR sequence, and (ii) a plasmid comprising helper genes necessary for AAV production. Two different transgenes were used with each capsid, in self-complementary constructs. The cells were subsequently lysed and the virions were purified using an affinity column, CsCl density ultracentrifugation, and dialysis. Subsequently, yield of each AAV was measured using a PCR-based quantification approach.


As shown in FIG. 2, recombinant AAV vectors comprising the STRD.101 and STRD.102 capsids had a yield that was similar to the yield of wildtype AAV9. This data confirms that recombinant AAVs comprising the recombinant capsid proteins are suitable for commercial manufacturing.


Example 3: In Vitro Transduction Using Recombinant AAV Viral Vectors

To confirm whether the recombinant AAV vectors of Example 1 are generally infective and able to transduce cells in culture, various AAV vectors were prepared according to a standard protocol.


The infectivity of the recombinant AAVs was tested using a standard TCID50 assay. Briefly, HeLaRC32 cells were infected with recombinant AAV particles in the presence of Adenovirus (Ad5) at doses spanning 5 orders of magnitude. After 72 hours, DNA was extracted and vector genome replication was quantified by qPCR.


The particle to infectivity ratio was calculated to determine infectivity. As shown in FIG. 3, the infectivity ratio of an AAV-STRD.101 vector was lower compared to that of wildtype AAV9. Because a lower infectivity ratio translates to a higher potency, AAV-STRD.101 is more infectious than wildtype AAV9.


Separately, infectivity was also determined in various cell lines. Recombinant AAVs packaging a luciferase transgene were generated and contacted with the cells in culture at a dose of 10,000 vector genomes (vg) per cell. 48-hours post infection, cells were lysed. The lysate was contacted with a bioluminescent substrate, and relative fluorescence units (RFUs) were measured. As shown in FIG. 4A-4D, AAV-STRD.101 vectors infected U87 cells (human glioblastoma cell line, FIG. 4A), N2A cells (mouse neural crest-derived cell line, FIG. 4B), SY5Y cells (human neuroblastoma cell line, FIG. 4C), and U2OS cells (human osteosarcoma cell line, FIG. 4D) at levels comparable to wildtype AAV9.


Accordingly, this data demonstrates that the recombinant AAV vectors of Example 1 can effectively transduce cells in culture.


Example 4: In Vivo Characterization of Recombinant AAVs Targeting the Central Nervous System

Two recombinant capsid proteins, STRD.101 and STRD.102 were selected for in vivo characterization. Recombinant AAVs comprising these capsid proteins and packaging a native tdTomato fluorescent transgene were generated. The recombinant AAVs were administered to neonatal mice by intracerebroventricular injection at day 0. At three weeks post-injection, brain tissues were harvested and fixed to evaluate the expression by visual assessment of the tdTomato fluorescence. FIG. 5 provides representative images showing tdTomato expression in coronal vibratome sections after 24 hours post-fixation with 4% PFA. These same sections were also visualized using immunohistochemistry (FIG. 6). As shown in the images of FIG. 5 and FIG. 6, AAV9, AAV-STRD.102 and AAV-STRD.101 vectors each had different distribution in the brain tissues, with the highest transgene expression localized near the site of injection. Taken together, this data shows that the recombinant AAVs tested successfully deliver a transgene to target cells in vivo after intracerbroventricular injection.


The AAV-STRD.101 and AAV-STRD.102 vectors packaging tdTomato were also administered to four adult mice by intravenous injection at a dose of 5.5×1013 vg/kg. Three weeks post-injection, liver and heart were harvested and fixed to evaluate the expression profile by visual assessment of tdTomato fluorescence.


Representative images from one mouse showing TdTomato expression in vibratrome liver sections after 24 hours post-fixation with 4% PFA are provided in FIG. 7. Notably, the AAV-STRD.102 and AAV-STRD.101 vectors were detargeted to the liver compared to wildtype AAV9. This desirable property was unexpected, as no counter screen in the liver was performed during evolution.


Representative images from one mouse showing TdTomato expression in vibratrome heart sections after 24 hours post-fixation with 4% PFA are provided in FIG. 8. Notably, the vectors tested had different tropism for the heart. Specifically, the AAV-STRD.102 vector was less infective in heart compared to AAV-STRD.101. Because no heart screen was performed during evolution, this differential transduction was wholly unexpected.


Taken together, this data indicates that the AAV-STRD.102 and AAV-STRD.101 vectors can be successfully used to target CNS tissues in vivo, avoid clearance by the liver, and are powerful tools for gene therapy. Given their different tropisms (i.e., AAV-STRD.101 was more infective in the heart than AAV-STRD.102), these vectors will be powerful tools for targeting gene therapy treatments to specifically desired tissues.


Example 5: Biodistribution of Recombinant AAVs in Non-Human Primates

Recombinant AAVs were administered to non-human primates, in order to determine biodistribution. Recombinant AAVs were administered by intravenous (IV) and intracerebrovascular (ICV) injection (FIG. 9). AAV-STRD.101 was administered at a dose of 2.9×1013 vg/kg by IV injection, and 2.1×1013 vg by ICV injection (black dots). AAV-STRD.102 was administered at a dose of 2.8×1013 vg/kg by IV injection, and 3.0×1013 vg by ICV injection (white dots). After 30 days, the animals were sacrificed, and viral load in various CNS tissues was measured by qPCR.


As shown in FIG. 9, both AAV-STRD.102 and AAV-STRD.101 infected various CNS tissues. Additionally, because the AAVs showed high levels of transduction, this data suggest that these AAVs are likely to avoid neutralizing AAVs in vivo.


Example 6: Cell Therapy Method for Treating a Subject in Need Thereof

Cells are transduced using an AAV vector ex vivo. For some purposes, the cells may be autologous (i.e., derived from the subject to be treated) or allogenic (i.e., derived from a different subject/donor). After transduction of the cells using an AAV, and after expression of a transgene has been verified, the cells are administered to the subject using standard clinical methods.


Cells may be administered to the subject once, or administration may be repeated multiple times at therapeutically effective intervals. The number of cells administered varies depending on, for example, the disease or condition to be treated, the severity of the subject's disease/condition, and the subject's height and weight.


Example 7: Gene Therapy Method for Treating a Subject in Need Thereof

An AAV vector described herein (e.g., an AAV vector comprising a capsid having the sequence of SEQ ID NO: 175 or 180) is administered to a subject in need thereof, wherein the subject has a disease or disorder of the CNS. The AAV vector is administered to the subject once, or administration may be repeated multiple times at therapeutically effective intervals. The administration is by one or more therapeutically effective routes, such as intravenous (IV), intracerebroventricular (ICV), or intrathecal (IT) injection. The dose of AAV vector varies depending on, for example, the disease or condition to be treated, the severity of the subject's disease/condition, and the subject's height and weight. For example, the dose of AAV administered to the subject may be 2.8×1013 vg/kg or 2.9×1013 vg/kg when the AAV vector is administered by IV injection. When the AAV vector is administered by ICV injection, the dose may be 2.1×1013 vg or 3.0×1013 vg. In some protocols, the AAV vector may be administered to the subject by both IV and ICV injection.


The foregoing is illustrative of the present invention, and is not to be construed as limiting thereof. The invention is defined by the following claims, with equivalents of the claims to be included therein.

Claims
  • 1. An adeno-associated virus (AAV) vector comprising an AAV capsid protein, wherein the AAV capsid protein comprises the amino acid sequence of SEQ ID NO: 180.
  • 2. The AAV vector of claim 1, wherein the AAV vector comprises a cargo nucleic acid, wherein the cargo nucleic acid is encapsidated by the AAV capsid protein.
  • 3. The AAV vector of claim 2, wherein the cargo nucleic acid comprises at least one terminal repeat sequence.
  • 4. The AAV vector of claim 3, wherein the cargo nucleic acid comprises a heterologous nucleic acid sequence.
  • 5. The AAV vector of claim 4, wherein the cargo nucleic acid comprises inverted terminal repeat (ITR) sequences located 3′ and 5′ of the heterologous sequence.
  • 6. The AAV vector of claim 4, wherein the heterologous nucleic acid sequence encodes a polypeptide.
  • 7. The AAV vector of claim 6, wherein the polypeptide is a therapeutic polypeptide.
  • 8. The AAV vector of claim 6, wherein the polypeptide is an immunogenic polypeptide.
  • 9. The AAV vector of claim 6, wherein the polypeptide is nuclease.
  • 10. The AAV vector of claim 9, wherein the nuclease is a Cas9 nuclease, a Cas12(a) nuclease (Cpf1), a Cas12b nuclease, a Cas12c nuclease, a TrpB-like nuclease, a Cas13a nuclease (C2c2), a Cas13b nuclease, or a modified or truncated variant of any thereof.
  • 11. The AAV vector of claim 4, wherein the heterologous nucleic acid sequence encodes an untranslated RNA.
  • 12. The AAV vector of claim 11, wherein the untranslated RNA is a guide RNA.
  • 13. The AAV vector of claim 11, wherein the untranslated RNA is an antisense RNA, a ribozyme, or an interfering RNA.
  • 14. A pharmaceutical composition comprising the AAV vector of claim 2 and a pharmaceutically acceptable carrier.
  • 15. A method of introducing a cargo nucleic acid molecule into a cell, comprising contacting the cell with the AAV vector of claim 2.
  • 16. An adeno-associated virus (AAV) capsid protein comprising the amino acid sequence of SEQ ID NO: 180.
  • 17. A nucleic acid comprising a nucleic acid sequence encoding the AAV capsid protein of claim 16.
  • 18. A cell comprising the nucleic acid of claim 17.
  • 19. An expression vector comprising a nucleic acid sequence encoding the AAV capsid protein of claim 16.
  • 20. A method of producing an adeno-associated virus (AAV) vector, the method comprising: a. culturing a cell that comprises (i) the nucleic acid of claim 17, (ii) a cargo nucleic acid comprising a 5′ inverted terminal repeat (ITR), a heterologous acid sequence, and a 3′ ITR, and (iii) AAV sequences sufficient for replication and encapsidation of nucleic acid, wherein the cell is cultured under conditions such that it produces the AAV vector; andb. collecting the AAV vector from the cell.
CROSS-REFERENCE TO RELATED APPLICATION

This application is a continuation of U.S. patent application Ser. No. 17/479,577, filed Sep. 20, 2021, which is a continuation of International Patent Application No. PCT/US2020/023877, filed Mar. 20, 2020, which claims the benefit of U.S. Provisional Application No. 62/821,710, filed Mar. 21, 2019, which is incorporated herein by reference in its entirety.

US Referenced Citations (257)
Number Name Date Kind
4039388 Gal et al. Aug 1977 A
4501729 Boucher et al. Feb 1985 A
4968603 Slamon et al. Nov 1990 A
5139941 Muzyczka et al. Aug 1992 A
5328470 Nabel et al. Jul 1994 A
5399346 Anderson et al. Mar 1995 A
5478745 Samulski et al. Dec 1995 A
5658776 Flotte et al. Aug 1997 A
5686240 Schuchman et al. Nov 1997 A
5863541 Samulski et al. Jan 1999 A
5869248 Yuan et al. Feb 1999 A
5877022 Stinchcomb et al. Mar 1999 A
5905040 Mazzara et al. May 1999 A
5916563 Young et al. Jun 1999 A
5962313 Podsakoff et al. Oct 1999 A
6013487 Mitchell Jan 2000 A
6040183 Ferrari et al. Mar 2000 A
6083702 Mitchell et al. Jul 2000 A
6093570 Ferrari et al. Jul 2000 A
6156303 Russell et al. Dec 2000 A
6180613 Kaplitt et al. Jan 2001 B1
6426198 Carstea et al. Jul 2002 B1
6468524 Chiorini et al. Oct 2002 B1
6468798 Debs et al. Oct 2002 B1
6503888 Kaplitt et al. Jan 2003 B1
6544785 Palese et al. Apr 2003 B1
6562958 Breton et al. May 2003 B1
6733757 Patel et al. May 2004 B2
6822071 Stephens et al. Nov 2004 B1
6962815 Bartlett Nov 2005 B2
6984517 Chiorini et al. Jan 2006 B1
7045675 Carstea et al. May 2006 B2
7071172 McCown et al. Jul 2006 B2
7105345 Wilson et al. Sep 2006 B2
7198951 Gao et al. Apr 2007 B2
7201898 Monahan et al. Apr 2007 B2
7214786 Kovalic et al. May 2007 B2
7252997 Hallek et al. Aug 2007 B1
7259151 Arbetman et al. Aug 2007 B2
7282199 Gao et al. Oct 2007 B2
7314912 Hallek et al. Jan 2008 B1
7473531 Domon et al. Jan 2009 B1
7588772 Kay et al. Sep 2009 B2
7712893 Dobashi May 2010 B2
7718424 Chiorini et al. May 2010 B2
7749492 Bartlett et al. Jul 2010 B2
7777097 Glazebrook et al. Aug 2010 B2
7790449 Gao et al. Sep 2010 B2
7867484 Samulski et al. Jan 2011 B2
7892809 Bowles et al. Feb 2011 B2
7906111 Wilson et al. Mar 2011 B2
8067014 Kay et al. Nov 2011 B2
8299321 Cao Oct 2012 B2
8318480 Gao et al. Nov 2012 B2
8343764 Abad et al. Jan 2013 B2
8445267 Zhong et al. May 2013 B2
8628966 Chatterjee et al. Jan 2014 B2
8664475 Puzio et al. Mar 2014 B2
8679837 Zolotukhin et al. Mar 2014 B2
8734809 Gao et al. May 2014 B2
8802440 Zhong et al. Aug 2014 B2
8889641 Asokan et al. Nov 2014 B2
8906387 Kay et al. Dec 2014 B2
8906675 Gao et al. Dec 2014 B2
8927514 Chatterjee et al. Jan 2015 B2
8952217 Puzio et al. Feb 2015 B2
8962332 Gao et al. Feb 2015 B2
9012224 Bowles et al. Apr 2015 B2
9066966 Puccio et al. Jun 2015 B2
9157098 Zhong et al. Oct 2015 B2
9409953 Asokan et al. Aug 2016 B2
9441244 Schaffer et al. Sep 2016 B2
9475845 Asokan et al. Oct 2016 B2
9567376 Cronin et al. Feb 2017 B2
9585971 Deverman et al. Mar 2017 B2
9587250 Gao et al. Mar 2017 B2
9598468 Weigel-Van Aken et al. Mar 2017 B2
9611302 Srivastava et al. Apr 2017 B2
9623120 Chatterjee et al. Apr 2017 B2
9677088 Nakai et al. Jun 2017 B2
9677089 Gao et al. Jun 2017 B2
9683268 Barouch et al. Jun 2017 B2
9695220 Vandenberghe et al. Jul 2017 B2
9719070 Vandenberghe et al. Aug 2017 B2
9725485 Srivastava et al. Aug 2017 B2
9737618 Wilson et al. Aug 2017 B2
9737619 Ansell et al. Aug 2017 B2
9775918 Zhong et al. Oct 2017 B2
9777291 Chatterjee et al. Oct 2017 B2
9783825 Chatterjee et al. Oct 2017 B2
9790472 Gao et al. Oct 2017 B2
9803218 Chatterjee et al. Oct 2017 B2
9834789 Chatterjee et al. Dec 2017 B2
9839696 Chatterjee et al. Dec 2017 B2
9879275 Nadzan et al. Jan 2018 B2
9890396 Chatterjee et al. Feb 2018 B2
9909142 Yazicioglu et al. Mar 2018 B2
9920097 Zhong et al. Mar 2018 B2
9944908 Vatèn et al. Apr 2018 B2
9976157 Poraty-Gavra et al. May 2018 B2
10011640 Srivastava et al. Jul 2018 B2
10072251 Gao et al. Sep 2018 B2
10077291 Asokan et al. Sep 2018 B2
10081659 Chiorini et al. Sep 2018 B2
10119125 Vandenberghe et al. Nov 2018 B2
10214566 Schaffer et al. Feb 2019 B2
10337027 Puccio et al. Jul 2019 B2
10369193 Passini et al. Aug 2019 B2
10385320 Kay et al. Aug 2019 B2
10392632 Wright et al. Aug 2019 B2
10406244 Kay et al. Sep 2019 B2
10414803 Nathwani et al. Sep 2019 B2
10426844 Agbandje-McKenna et al. Oct 2019 B2
10526627 Skuratowicz et al. Jan 2020 B2
10668094 Karlish et al. Jun 2020 B2
10745447 Asokan et al. Aug 2020 B2
10907176 Asokan et al. Feb 2021 B2
11077128 Karlish et al. Aug 2021 B2
20020192189 Xiao et al. Dec 2002 A1
20030017131 Park et al. Jan 2003 A1
20030053990 Rabinowitz et al. Mar 2003 A1
20030225017 Murdin et al. Dec 2003 A1
20040013645 Monahan et al. Jan 2004 A1
20040031072 La Rosa et al. Feb 2004 A1
20040071659 Chang et al. Apr 2004 A1
20040166519 Cargill et al. Aug 2004 A1
20040214272 La Rosa et al. Oct 2004 A1
20050287122 Bartlett et al. Dec 2005 A1
20060107345 Alexandrov et al. May 2006 A1
20060123505 Kikuchi et al. Jun 2006 A1
20060171926 Passini et al. Aug 2006 A1
20060236419 La Rosa et al. Oct 2006 A1
20070015238 Snyder et al. Jan 2007 A1
20070124833 Abad et al. May 2007 A1
20070196338 Samulski et al. Aug 2007 A1
20070243526 Kay et al. Oct 2007 A1
20080229439 La Rosa et al. Sep 2008 A1
20090215879 Diprimio et al. Aug 2009 A1
20090221620 Luke et al. Sep 2009 A1
20090275107 Lock et al. Nov 2009 A1
20090317417 Vandenberghe et al. Dec 2009 A1
20100037352 Alexandrov et al. Feb 2010 A1
20100047174 Kay et al. Feb 2010 A1
20100095387 Smith et al. Apr 2010 A1
20110061124 Nadzan et al. Mar 2011 A1
20110067143 Rosa et al. Mar 2011 A2
20110124048 Yun May 2011 A1
20110131679 La Rosa et al. Jun 2011 A2
20110209246 Kovalic et al. Aug 2011 A1
20110214206 La Rosa et al. Sep 2011 A1
20110236353 Wilson et al. Sep 2011 A1
20110294218 Chatterjee et al. Dec 2011 A1
20120009268 Asokan et al. Jan 2012 A1
20120137379 Gao et al. May 2012 A1
20120216318 La Rosa et al. Aug 2012 A1
20120255046 Kay et al. Oct 2012 A1
20120322861 Byrne et al. Dec 2012 A1
20130096182 Chatterjee et al. Apr 2013 A1
20130152224 Abad et al. Jun 2013 A1
20130185831 Kovalic et al. Jul 2013 A1
20130195801 Gao et al. Aug 2013 A1
20130203841 Zhong et al. Aug 2013 A1
20130216501 Zhong et al. Aug 2013 A1
20130224836 Muramatsu Aug 2013 A1
20130225666 Kaspar et al. Aug 2013 A1
20130326723 La Rosa et al. Dec 2013 A1
20140017212 Rebar Jan 2014 A1
20140037585 Wright et al. Feb 2014 A1
20140050701 Zhong et al. Feb 2014 A1
20140056854 Asokan et al. Feb 2014 A1
20140057969 Frost et al. Feb 2014 A1
20140130203 La Rosa et al. May 2014 A1
20140162319 Hareendran et al. Jun 2014 A2
20140199313 Plesch et al. Jul 2014 A1
20140223605 Puzio et al. Aug 2014 A1
20140259218 Kovalic et al. Sep 2014 A1
20140296486 Gao et al. Oct 2014 A1
20140335054 Gao et al. Nov 2014 A1
20140341852 Srivastava et al. Nov 2014 A1
20150050302 Thess Feb 2015 A1
20150079038 Deverman et al. Mar 2015 A1
20150082481 La Rosa et al. Mar 2015 A1
20150126588 Nakai et al. May 2015 A1
20150133530 Srivastava et al. May 2015 A1
20150184189 Abad et al. Jul 2015 A1
20150191739 La Rosa et al. Jul 2015 A1
20150197763 La Rosa et al. Jul 2015 A1
20150238550 McCown et al. Aug 2015 A1
20150344911 Chatterjee et al. Dec 2015 A1
20160017295 Schaffer et al. Jan 2016 A1
20160025657 Shahbazmohamadi et al. Jan 2016 A1
20160106865 Zhong et al. Apr 2016 A1
20160215024 Vandenberghe et al. Jul 2016 A1
20160222067 Gao et al. Aug 2016 A1
20160256571 Corral-Debrinski et al. Sep 2016 A1
20160264984 La Rosa et al. Sep 2016 A1
20160289275 Chiorini et al. Oct 2016 A1
20160319294 Kovalic et al. Nov 2016 A1
20160333372 Srivastava et al. Nov 2016 A1
20160361439 Agbandje-McKenna et al. Dec 2016 A1
20160369299 Boye et al. Dec 2016 A1
20170007720 Boye et al. Jan 2017 A1
20170028082 Wilson et al. Feb 2017 A1
20170049910 Cronin et al. Feb 2017 A1
20170067908 Nakai et al. Mar 2017 A1
20170088852 Dangoor et al. Mar 2017 A1
20170088858 Gao et al. Mar 2017 A1
20170096683 Scaria et al. Apr 2017 A1
20170130245 Kotin et al. May 2017 A1
20170159027 Wilson et al. Jun 2017 A1
20170166926 Deverman et al. Jun 2017 A1
20170204144 Deverman et al. Jul 2017 A1
20170211092 Chatterjee et al. Jul 2017 A1
20170211093 Chatterjee et al. Jul 2017 A1
20170211094 Chatterjee et al. Jul 2017 A1
20170211095 Chatterjee et al. Jul 2017 A1
20170240885 Deverman et al. Aug 2017 A1
20170275337 Srivastava et al. Sep 2017 A1
20170298323 Vandenberghe et al. Oct 2017 A1
20170349911 Gao et al. Dec 2017 A1
20180002722 Asokan et al. Jan 2018 A1
20180030096 Aslanidi et al. Feb 2018 A1
20180030479 Gao et al. Feb 2018 A1
20180036428 Zhong et al. Feb 2018 A1
20180066022 Chalberg et al. Mar 2018 A1
20180066285 Ojala et al. Mar 2018 A1
20180104289 Venditti et al. Apr 2018 A1
20180105559 Srivastava et al. Apr 2018 A1
20180112229 Nadzan et al. Apr 2018 A1
20180119167 Abad et al. May 2018 A1
20180135074 Srivastava et al. May 2018 A1
20180135076 Linden May 2018 A1
20180163227 Chatterjee et al. Jun 2018 A1
20180214576 Fitzgerald et al. Aug 2018 A1
20180244727 Zhong et al. Aug 2018 A1
20180265863 Esteves et al. Sep 2018 A1
20180355376 Chiorini et al. Dec 2018 A1
20180362592 Gao et al. Dec 2018 A1
20180371024 Asokan et al. Dec 2018 A1
20190048041 Asokan et al. Feb 2019 A1
20190055524 Vandenberghe et al. Feb 2019 A1
20190085301 Gao et al. Mar 2019 A1
20190100560 Vandenberghe et al. Apr 2019 A1
20190249195 Marsic et al. Aug 2019 A1
20190255192 Kirn et al. Aug 2019 A1
20190262373 Woodard et al. Aug 2019 A1
20190284576 Qu et al. Sep 2019 A1
20190292561 Qu et al. Sep 2019 A1
20190367562 Asokan et al. Dec 2019 A1
20200109418 Li et al. Apr 2020 A1
20200399321 Asokan et al. Dec 2020 A1
20210115474 McCoy et al. Apr 2021 A1
20210128652 Dismuke May 2021 A1
20210324418 Thomas et al. Oct 2021 A1
20220056478 O'Banion Feb 2022 A1
20220088152 Mikati et al. Mar 2022 A1
20220089651 Asokan et al. Mar 2022 A1
Foreign Referenced Citations (150)
Number Date Country
1033405 Sep 2000 EP
1777296 Apr 2007 EP
1887081 Feb 2008 EP
2194140 Jun 2010 EP
2359869 Aug 2011 EP
2492347 Aug 2012 EP
2660325 Nov 2013 EP
2315833 Apr 2015 EP
1453547 Sep 2016 EP
2007795 Nov 2016 EP
2675484 May 2018 EP
2263692 Sep 2018 EP
2206728 Oct 2018 EP
3244931 Oct 2018 EP
1633767 Nov 2018 EP
3060575 Dec 2018 EP
3250239 Dec 2018 EP
3459965 Mar 2019 EP
3511021 Jul 2019 EP
3108000 Aug 2019 EP
2014-534245 Dec 2014 JP
2457252 Jul 2012 RU
WO-9005142 May 1990 WO
WO-9811244 Mar 1998 WO
WO-9901555 Jan 1999 WO
WO-9961601 Dec 1999 WO
WO-0017377 Mar 2000 WO
WO-9961601 Mar 2000 WO
WO-0023477 Apr 2000 WO
WO-0028004 May 2000 WO
WO-0028061 May 2000 WO
WO-0028061 Nov 2000 WO
WO-0111034 Feb 2001 WO
WO-0181581 Nov 2001 WO
WO-200192551 Dec 2001 WO
WO-0210210 Feb 2002 WO
WO-03000906 Jan 2003 WO
WO-03008540 Jan 2003 WO
WO-03033515 Apr 2003 WO
WO-03042361 May 2003 WO
WO-2003052051 Jun 2003 WO
WO-03095647 Nov 2003 WO
WO-2004027019 Apr 2004 WO
WO-2006021724 Mar 2006 WO
WO-2006029319 Mar 2006 WO
WO-2006066066 Jun 2006 WO
WO-2006073052 Jul 2006 WO
WO-2006119137 Nov 2006 WO
WO-2006119432 Nov 2006 WO
WO-2007084773 Jul 2007 WO
WO-2007089632 Aug 2007 WO
WO-2007092563 Aug 2007 WO
WO-2007100465 Sep 2007 WO
WO-2007120542 Oct 2007 WO
WO-2007127264 Nov 2007 WO
WO-2008088895 Jul 2008 WO
WO-2009037279 Mar 2009 WO
WO-2009043936 Apr 2009 WO
WO-2009105612 Aug 2009 WO
WO-2009108274 Sep 2009 WO
WO-2010093784 Aug 2010 WO
WO-2010129021 Nov 2010 WO
WO-2010138263 Dec 2010 WO
WO-2011020118 Feb 2011 WO
WO-2011020710 Feb 2011 WO
WO-2011122950 Oct 2011 WO
WO-2011133890 Oct 2011 WO
WO-2012061744 May 2012 WO
WO-2012064960 May 2012 WO
WO-2012112578 Aug 2012 WO
WO-2013016315 Jan 2013 WO
WO-2013027223 Feb 2013 WO
WO-2013158879 Oct 2013 WO
WO-2013170078 Nov 2013 WO
WO-2013173512 Nov 2013 WO
WO-2013190059 Dec 2013 WO
WO-2014007858 Jan 2014 WO
WO-2014045674 Mar 2014 WO
WO-2014124282 Aug 2014 WO
WO-2014144229 Sep 2014 WO
WO-2014153083 Sep 2014 WO
WO-2014193716 Dec 2014 WO
WO-2014194132 Dec 2014 WO
WO-2015013313 Jan 2015 WO
WO-2015018860 Feb 2015 WO
WO-2015038958 Mar 2015 WO
WO-2015054653 Apr 2015 WO
WO-2015062516 May 2015 WO
WO-2015121501 Aug 2015 WO
WO-2015164757 Oct 2015 WO
WO-2015168666 Nov 2015 WO
WO-2015181823 Dec 2015 WO
WO-2015191508 Dec 2015 WO
WO-2016054557 Apr 2016 WO
WO-2016065001 Apr 2016 WO
WO-2016115382 Jul 2016 WO
WO-2016115503 Jul 2016 WO
WO-2016128558 Aug 2016 WO
WO-2016128559 Aug 2016 WO
WO-2016134338 Aug 2016 WO
WO-2016150964 Sep 2016 WO
WO-2016164642 Oct 2016 WO
WO-2016172008 Oct 2016 WO
WO-2016172155 Oct 2016 WO
WO-2016179644 Nov 2016 WO
WO-2017015102 Jan 2017 WO
WO-2017058892 Apr 2017 WO
WO-2017066764 Apr 2017 WO
WO-2017070516 Apr 2017 WO
WO-2017077451 May 2017 WO
WO-2017096164 Jun 2017 WO
WO-2017106236 Jun 2017 WO
WO-2017139643 Aug 2017 WO
WO-2017143100 Aug 2017 WO
WO-2017147123 Aug 2017 WO
WO-2017180854 Oct 2017 WO
WO-2017192750 Nov 2017 WO
WO-2017201248 Nov 2017 WO
WO-2018022608 Feb 2018 WO
WO-2018035213 Feb 2018 WO
WO-2018049226 Mar 2018 WO
WO-2018064624 Apr 2018 WO
WO-2018075798 Apr 2018 WO
WO-2018119330 Jun 2018 WO
WO-2018152333 Aug 2018 WO
WO-2018160582 Sep 2018 WO
WO-2018170310 Sep 2018 WO
WO-2018204764 Nov 2018 WO
WO-2018209154 Nov 2018 WO
WO-2018226785 Dec 2018 WO
WO-2018237066 Dec 2018 WO
WO-2019006418 Jan 2019 WO
WO-2019025984 Feb 2019 WO
WO-2019141765 Jul 2019 WO
WO-2019168961 Sep 2019 WO
WO-2019169004 Sep 2019 WO
WO-2019169132 Sep 2019 WO
WO-2019173434 Sep 2019 WO
WO-2019173538 Sep 2019 WO
WO-2019178412 Sep 2019 WO
WO-2019195423 Oct 2019 WO
WO-2019195444 Oct 2019 WO
WO-2019195449 Oct 2019 WO
WO-2020016318 Jan 2020 WO
WO-2020106916 May 2020 WO
WO-2020142653 Jul 2020 WO
WO-2020191300 Sep 2020 WO
WO-2020232297 Nov 2020 WO
WO-2021076911 Apr 2021 WO
WO-2021076925 Apr 2021 WO
Non-Patent Literature Citations (408)
Entry
ACS on STN, BD Registry, 1182714-10-8 [online] [retrieved on Apr. 30, 2019], 2009215879, Aug. 27, 2009, SEQ ID No. 7, 1 page.
ACS on STN, BD Registry, 1182714-97-1 [online] [retrieved on Apr. 30, 2019], 2009215879, Aug. 27, 2009, SEQ ID No. 210, 1 page.
Adachi et al., “Drawing a high-resolution functional map of adeno-associated virus capsid by massively parallel sequencing,” Nature Communications 5(1): 14 pages (2013).
Agbandje et al. “The Structure of Human Parvovirus B19 at 8 A; Resolution” Virology 203(1):106-115 (1994).
Agbandje-McKenna et al. “AAV Capsid Structure and Cell Interactions” Methods in Molecular Biology, 807:47-92 (2011).
Albright et al., “Mapping the Structural Determinants Required for AAVrh 0.10 Transport across the Blood-Brain Barrier,” Molecular Therapy 26(2), p. 1-14 (2017).
Albright, “Modulation of Sialic Acid Dependence Influences the Central Nervous System Transduction Profile of Adeno-associated Viruses,” Journal of Virology 93(11), pp. 1-15 (2019).
Altschul et al. “Basic Local Alignment Search Tool” Journal of Molecular Biology 215:403-410 (1990).
Altschul et al. “Local Alignment Statistics” Methods in Enzymology 266:460-480 (1996).
Altschul et al., “Gapped BLAST and PSI-BLAST: a new generation of protein database search programs,” Nucleic Acids Res. (1997) 25(17):3389-3402.
Altschul, SF et al., ‘Issues in searching molecular sequence databases,’ Nat. Genet., vol. 6, pp. 119-129, (Feb. 1994).
Andino et al. “AAV-mediated knockdown of phospholamban leads to improved contractility and calcium handling in cardiomyocytes” The Journal of Gene Medicine 10:132-142 (2008).
Arnold et al., “A calcium responsive element that regulates expression of two calcium binding proteins in Purkinje cells,” Proc Natl Acad Sci USA 94(16):8842-8847 (1997).
Arruda et al., “Regional intravascular delivery of AAV-2-F.IX to skeletal muscle achieves longterm correction of hemophilia B in a large animal model,” Blood 105:3458-3464 (2005).
Asokan et al. “Adeno-Associated Virus Type 2 Contains an Integrin a5 1 Binding Domain Essential for Viral Cell Entry” Journal of Virology, 80(18):8961-8969 (2006).
Asokan et al., “The AW Vector Toolkit: Poised at the Clinical Crossroads,” Molecular Therapy 20(4):699-708 (2012).
Asokan, et al., “Reengineering a receptor footprint of adeno-associated virus enables selective and systemic gene transfer to muscle”, Nat Biotechnol, (Jan. 2010); 28(1): 79-82.
Asuri et al., Directed Evolution of adeno-associated Virus for Enhanced Gene Delivery and Gene Targeting in Human Pluripotent Stem Cells, Molecular Therapy, Nature Publishing Group GB 20(2):329-338 (2013).
Ballabh et al. “The blood-brain barrier: an overview: structure, regulation, and clinical implications” Neurobiology of Disease, 16:1-13 (2004).
Bantel-Schaal et al. “Human adeno-Associated Virus Type 5 Is Only Distantly Related to Other Known Primate Helper-Dependent Parvovirus” Journal of Virology 73(2):939-947 (1999).
Bantel-Schaal et al., “Adeno-associated virus type 5 exploits two different entry pathways in human embryo fibroblast,” J Virology 73:939 (1999).
Bartlett, JS et al., ‘Selective and Rapid Uptake of Adeno-Associated Virus Type 2 in Brain,’ Hum. Gene Ther., 9(8):1181-1186, (May 1998).
Bell et al. “Identification of the Galactose Binding Domain of the Adeno-Associated Virus Serotype 9 Capsid” Journal of Virology, 86(13):7326-7333 (2012).
Bennett et al. “AAV6 K531 serves a dual function in selective receptor and antibody ADK6 recognition” Virology, 18:369-376 (2018).
Bleker et al. “Mutational Analysis of Narrow Pores at the Fivefold Symmetry Axes of Adeno-Associated Virus Type 2 Capsids Reveals a Dual Role in Genome Packaging and Activation of Phospholipase A2 Activity” Journal of Virology, 79(4):2528-2540 (2005).
Bordoli et al. “Protein structure homology modeling using SWISS-MODEL workspace” Nature Protocols, 4(1):1-13 (2008).
Bowie, et al., “Deciphering the Message in Protein Sequences: Tolerance to Amino Acid Substitutions.” Science (1990); 247: 1306-1310.
Bowles et al. “Phase 1 Gene Therapy for Duchenne Muscular Dystrophy Using a Translational Optimized AAV Vector” Molecular Therapy, 20(2):443-455 (2012).
Brichard et al. ““The Tyrosinase Gene Codes for an Antigen Recognized by Autologous Cytolytic T Lymphocytes on HLA-AZ Melanomas”” Journal of Experimental Medicine 178:489-495 (1993).
Brown et al. “Chimeric Parvovirus 19 Capsids for the Presentation of Foreign Epitopes” Virology 198(2):477-488 (1994).
Brown et al. “Erythrocyte P Antigen: Cellular Receptor for B19 Parvovirus” Science 262(5130):114-117 (1993).
Carrillo-Tripp et al. “VIPERdb2: an enhanced and web API enabled relational database for structural virology” Nucleic Acids Research, 37:D436-D442 (2009).
Carstea, ED et al. ‘Niemann-Pick C1 Disease Gene: Homology to Mediators of Cholesterol Homeostasis,’ Science, 277(5323): 228-231 (Jul. 1997).
Cearley et al. “Transduction Characteristics of Adeno-associated Virus Vectors Expressing Cap Serotypes 7, 8, 9, and Rh10 in the Mouse Brain” Molecular Therapy, 13(3):528-537 (2006).
Cearley, C.N. et al. (2008). “Expanded repertoire of AAV vector serotypes mediate unique patterns of transduction in mouse brain,” Mol. Ther. 16:1710-1718.
Chai et al., “Nab Escaping AAV Mutants Isolated from Mouse Muscles.” Bio Protoc., vol. 98, No. 9, pages Article: e2841 Section on “Procedure” (2018).
Chandler et al., “Systemic AAV9 gene therapy improves the lifespan of mice with Niemann-Pick disease, type C1 ,” Human Molecular Genetics 26(1):52-64 (2017).
Chao et al. “Several Log Increase in Therapeutic Transgene Delivery by Distinct Adeno-Associated Viral Serotype Vectors” Molecular Therapy 2(6):619-623 (2000).
Chapman et al. “Structure, Sequence, and Function Correlations among Parvoviruses” Virology 194(2):491-508 (1993).
Chen et al. “Efficient Transduction of Vascular Endothelial Cells with Recombinant Adeno-Associated Virus Serotype 1 and 5 Vectors” Human Gene Therapy, 16(2):235-247 (2005).
Chen, SH et al., ‘Gene therapy for brain tumors: Regression of experimental gliomas by adenovirus-mediated gene transfer in vivo,’ Proc. Natl Acad. Sci. USA, vol. 91, pp. 3054-3057, (Apr. 1994).
Chiorini et al. “Cloning and Characterization of adeno-Associated Virus Type 5” Journal of Virology 73(2):1309-1319 (1999).
Chiorini et al. “Cloning of adeno-Associated Virus Type 4 (AAV4). and Generation of Recombinant AAV4 Particles” Journal of Virology 71 (9):6823-6833 (1997).
Chipman et al. ““Cryo-electron microscopy studies of empty capsids of human parvovirus 819 complexed with its cellular receptor”” Proceedings of the National Academy of Sciences 93:7502-7506 (1996).
Chirmule et al., “Humoral immunity to adeno-associated virus type 2 vectors following administration to murine and nonhuman primate muscle,” Journal of Virology, The American Society for Microbiology, 74(5):2420-2425 (2000).
Choi et al., “Optimization of AAV expression cassettes to improve packaging capacity and transgene expression in neurons,” Molecular Brain, Biomed Central Ltd, London UK, 7(1):17 pp. 1-10 (2014).
Choudhury et al., “In Vivo Selection Yields AAV-B1 Capsid for Central Nervous System and Muscle Gene Therapy.” Mol Ther, vol. 24, No. 7, pp. 1247-1257 (2016).
Clapcote SJ, et al., “Mutation 1810N in the alpha3 isoform of Na+, K+-ATPase causes impairments in the sodium pump and hyperexcitability in the CNS,” Proc Natl Acad Sci USA. 106(33):14085-14090 (2009).
Clark, KR et al., ‘Highly Purified Recombinant Adeno-Associated Virus Vectors Are Biologically Active and Free of Detectable Helper and Wild-Type Viruses,’ Hum. Gene Ther., 10(6):1031-1039, (Apr. 1999).
Conway et al. “High-titer recombinant adeno-associated virus production utilizing a recombinant herpes simplex virus type 1 vector expressing AAV-2 Rep and Cap” Gene Therapy 6:986-993 (1999).
Corpet et al., ‘Multiple sequence alignment with hierarchical clustering,’ Nucleic Acids Research, vol. 16 No. 22, pp. 10881-10890, (Oct. 1988).
Cotmore et al.,“The Family Parvoviridae,” Archives of Virology 159:1239-1247 (2014).
DataBase GenBank: ABS91093.1, Aug. 10, 2007, [online] [retrieved on Feb. 14, 2020] Retrieved from Internet: https://www.ncbi.nlm.nih.gov/protein/ABS91093.1.
DataBase GenBank: ACW56705.1, Sep. 24, 2009, [online] [retrieved on May 7, 2019] Retrieved from Internet:https://www.ncbi.nlm.nih.gov/protein/ACW56705.1?report=genbank&log$=prottop&blast_rank= 1&RID=D2CZ8TP9014, 1 page.
De Jesus et al., “Telomerase gene therapy in adult and old mice delays aging and increases longevity without increasing cancer,” EMBO Mol. Med. 4(8): 691-704 (2012).
Devereux et al. “A comprehensive set of sequence analysis programs for the VAX” Nucleic Acids Research 12(1):387-395 (1984).
Deverman, BE, Cre-dependent selection yields AAV variants for widespread gene transfer to the adult brain, Nat. Biotechnol., 34(2) :204-209. doi: 10.1038/nbt.3440. PubMed PMID: 26829320 (Epub Feb. 1, 2016).
Dimattia et al. “Structural Insight into the Unique Properties of Adeno-Associated Virus Serotype 9,” Journal of Virology, 86(12):6947-6958 (2012).
Dipasquale et al. “Identification of PDGFR as a receptor for MV-5 transduction” Nature Medicine, 9:1306-1312 (2003). (Abstract only).
Diprimio, et al., “Surface loop dynamics in adeno-associated virus capsid assembly”, Journal of Virology (2008); vol. 82, No. 11, pp. 5178-5189.
Emsley et al. “Features and development of Coot” Acta Crystallographica Section D: Biological Crystallography, D66:486-501 (2010).
Fang et al., “Stable antibody expression at therapeutic levels using the 2A peptide,” Nature Biotechnology 23:584-590 (2005).
Felsenstein, Joseph “Confidence Limits on Phylogenies: An Approach Using the Bootstrap” Evolution, 39 (4):783-791 (1985).
Fisher, KJ et al., ‘Transduction with Recombinant Adeno-Associated Virus for Gene Therapy Is Limited by Leading-Strand Synthesis,’ J. Virol., 70(1):520-532 (LFU assay) (Jan. 1996).
Foster et al., “Emerging Immunotherapies for Autoimmune Kidney Disease,” Hyman Vaccines & Immunotherapeutics 15(4):876-890 (2019).
Foust et al. “Intravascular AAV9 preferentially targets neonatal neurons and adult astrocytes” Nature Biotechnology, 27(1):59-65 (2009).
Gao et al. “Adeno-associated viruses undergo substantial evolution in primates during natural infections” Proceedings of the National Academy of Sciences, 100(10):6081-6086 (2003).
Gao et al. “Clades of adeno-Associated Viruses are Widely Disseminated in Human Tissues” Journal of Virology 78(12):6381-6388 (2004).
Gao et al. “Novel adeno-associated viruses from Rhesus Monkeys as Vectors for human gene therapy,” Proceedings of the National Academy of Sciences 99(18):11854-11859 (2002).
Genbank Accession No. AAR26465, Bovine Adeno-Associated Virus, dated May 25, 2004, 2 pages.
Genbank Accession No. AAT46339, capsid protein [Adeno-associated virus 11], dated Nov. 30, 2004, 2 pages.
Genbank Accession No. ABI16639, VP1 [Adeno-associated virus 12, dated Feb. 20, 2008, 2 pages.
GenBank Accession No. AF028704 “Adeno-associated virus 6, complete genome,” Jan. 12, 1998 [online]. (Retrieved online Feb. 21, 2019].
GenBank Accession No. AF028705 “adeno-associated Virus 3B, complete genome” NCBI (2 pages). (Jan. 12, 1998).
GenBank Accession No. AF043303 “Adeno-associated virus 2, complete genome,” May 20, 2010 [online]. (Retrieved online Feb. 21, 2019].
Genbank Accession No. AF085716, Adeno-associated virus 5 DNA binding trs helicase (Rep22) and capsid protein (VP1) genes, complete cds., dated Feb. 9, 1999, 3 pages.
GenBank Accession No. AF258783.1 ‘Felis catus Niemann-Pick type C1 disease protein (NPC1) mRNA, complete eds’ (2000).
GenBank Accession No. AF288061 “Hamster parvovirus 5′ terminal hairpin gene sequence” NCBI (1 page). (Apr. 13, 2001), replaced by AH009962.
GenBank Accession No. AF513851 “Adeno-associated virus 7 nonstructural protein and capsid protein genes, complete cds,” Sep. 5, 2002 [online]. (Retrieved online Feb. 21, 2019].
GenBank Accession No. AF513852 “Adeno-associated virus 8 nonstructural protein and capsid protein genes, complete cds,” Sep. 5, 2002 [online]. (Retrieved online Feb. 21, 2019].
GenBank Accession No. AY028223 “B19 Virus isolate patient_A.1.1 genomic sequence” NCB/ (1 page). (Apr. 16, 2001).
GenBank Accession No. AY028226 “819 Virus isolate patient_A.2.1 genomic sequence” NCB/ (1 page). (Apr. 16, 2001).
Genbank Accession No. AY186198, Avian adeno-associated virus Atcc VR-865, complete genome, dated Jun. 5, 2003, 3 pages.
Genbank Accession No. AY242997, Non-Human primate Adeno-associated virus isolate AAVrh.8 capsid protein (VP1) gene, complete cds, dated May 14, 2003, 2 pages.
Genbank Accession No. AY242998, Non-Human primate Adeno-associated virus isolate AAVrh.37 capsid protein (VP1) gene, complete cds, dated May 14, 2003, 2 pages.
Genbank Accession No. AY242999, Non-Human primate Adeno-associated virus isolate AAVrh.36 capsid protein (VP1) gene, complete cds, dated May 14, 2003, 2 pages.
Genbank Accession No. AY243000, Non-Human primate Adeno-associated virus isolate AAVrh.35 capsid protein (VP1) gene, complete cds, dated May 14, 2003, 2 pages.
Genbank Accession No. AY243001, Non-Human Primate Adeno-associated Virus Isolate AAVrh.34 capsid protein (VP1) gene, complete cds., dated May 14, 2003, 2 pages.
Genbank Accession No. AY243002, Non-Human Primate Adeno-associated Virus Isolate AAVrh.33 capsid protein (VP1) gene, complete cds. dated May 14, 2003, 2 pages.
Genbank Accession No. AY243003, Non-Human Primate Adeno-associated Virus Isolate AAVrh.32 capsid protein (VP1) gene, complete cds, dated May 14, 2003, 2 pages.
Genbank Accession No. AY243007, Non-Human Primate Adeno-associated Virus Isolate AAVrh.2 capsid protein (VP1) gene, complete cds, dated May 14, 2003, 2 pages.
Genbank Accession No. AY243013, Non-Human primate Adeno-associated virus isolate AAVrh.13 capsid protein (VP1) gene, complete cds, dated May 14, 2003, 2 pages.
Genbank Accession No. AY243015, Non-Human primate Adeno-associated virus isolate AAVrh.10 capsid protein (VP1) gene, complete cds dated May 14, 2003, 2 pages.
Genbank Accession No. AY243016, Non-Human primate Adeno-associated virus isolate AAVcy.6 capsid protein (VP1) gene, complete cds dated May 14, 2003, 2 pages.
Genbank Accession No. AY243017, Non-Human primate Adeno-associated virus isolate AAVcy.5 capsid protein (VP1) gene, complete cds, dated May 14, 2003, 2 pages.
Genbank Accession No. AY243018, Non-Human primate Adeno-associated virus isolate AAVcy.4 capsid protein (VP1) gene, complete cds, dated May 14, 2003, 2 pages.
Genbank Accession No. AY243019, Non-Human primate Adeno-associated virus isolate AAVcy.3 capsid protein (VP1) gene, complete cds, dated May 14, 2003, 2 pages.
Genbank Accession No. AY243020, Non-Human primate Adeno-associated virus isolate AAVcy.2 capsid protein (VP1) gene, complete cds, dated May 14, 2003, 2 pages.
Genbank Accession No. AY243021, Non-Human primate Adeno-associated virus isolate AAVch.5 capsid protein (VP1) gene, complete cds, dated May 14, 2003, 2 pages.
Genbank Accession No. AY243022, Non-Human primate Adeno-associated virus isolate AAVbb.2 capsid protein (VP1) gene, complete cds, dated May 14, 2003, 2 pages.
Genbank Accession No. AY243023, Non-Human primate Adeno-associated virus isolate AAVbb.1 capsid protein (VP1) gene, complete cds, dated May 14, 2003, 2 pages.
Genbank Accession No. AY388617, Bovine adeno-associated virus, complete genome, dated May 25, 2004, 3 pages.
Genbank Accession No. AY530553, Adeno-associated virus isolate pi.1 capsid protein VP1 (cap) gene, complete cds, dated Jun. 24, 2004, 2 pages.
Genbank Accession No. AY530554, Adeno-associated virus isolate pi.2 capsid protein VP1 (cap) gene, complete cds, dated Jun. 24, 2004, 2 pages.
Genbank Accession No. AY530555, Adeno-associated virus isolate pi.3 capsid protein VP1 (cap) gene, complete cds, dated Jun. 24, 2004, 2 pages.
Genbank Accession No. AY530556, Adeno-associated virus isolate rh.1 capsid protein VP1 (cap) gene, complete cds, dated Jun. 24, 2004, 2 pages.
Genbank Accession No. AY530557, Adeno-associated virus isolate rh.25 capsid protein VP1 (cap) gene, complete cds, dated Jun. 24, 2004, 2 pages.
Genbank Accession No. AY530558, Adeno-associated virus isolate rh.38 capsid protein VP1 (cap) gene, complete cds, dated Jun. 24, 2004, 2 pages.
Genbank Accession No. AY530559, Adeno-associated virus isolate rh.40 capsid protein VP1 (cap) gene, complete cds, dated Jun. 24, 2004, 2 pages.
Genbank Accession No. AY530560, Adeno-associated virus isolate rh.43 capsid protein VP1 (cap) gene, complete cds, dated Jun. 24, 2004, 2 pages.
Genbank Accession No. AY530561, Adeno-associated virus isolate rh.48 capsid protein VP1 (cap) gene, complete cds, dated Jun. 24, 2004, 2 pages.
Genbank Accession No. AY530562, Adeno-associated virus isolate rh.49 capsid protein VP1 (cap) gene, complete cds, dated Jun. 24, 2004, 2 pages.
Genbank Accession No. AY530563, Adeno-associated virus isolate rh.50 capsid protein VP1 (cap) gene, complete cds, dated Jun. 24, 2004, 2 pages.
Genbank Accession No. AY530564, Adeno-associated virus isolate rh.51 capsid protein VP1 (cap) gene, complete cds, dated Jun. 24, 2004, 2 pages.
Genbank Accession No. AY530565, Adeno-associated virus isolate rh.52 capsid protein VP1 (cap) gene, complete cds, dated Jun. 24, 2004, 2 pages.
Genbank Accession No. AY530566, Adeno-associated virus isolate rh.53 capsid protein VP1 (cap) gene, complete cds, dated Jun. 24, 2004, 2 pages.
Genbank Accession No. AY530567, Adeno-associated virus isolate rh.54 capsid protein VP1 (cap) gene, complete cds, dated Jun. 24, 2004, 2 pages.
Genbank Accession No. AY530568, Adeno-associated virus isolate rh.55 capsid protein VP1 (cap) gene, complete cds, dated Jun. 24, 2004, 2 pages.
Genbank Accession No. AY530569, Adeno-associated virus isolate rh.57 capsid protein VP1 (cap) gene, complete cds, dated Jun. 24, 2004, 2 pages.
Genbank Accession No. AY530570, Adeno-associated virus isolate rh.58 capsid protein VP1 (cap) gene, complete cds, dated Jun. 24, 2004, 2 pages.
Genbank Accession No. AY530572, Adeno-associated virus isolate rh.61 capsid protein VP1 (cap) gene, complete cds, dated Jun. 24, 2004, 2 pages.
Genbank Accession No. AY530573, Adeno-associated virus isolate rh.62 capsid protein VP1 (cap) gene, complete cds, dated Jun. 24, 2004, 2 pages.
Genbank Accession No. AY530574, Adeno-associated virus isolate rh.64 capsid protein VP1 (cap) gene, complete cds, dated Jun. 24, 2004, 2 pages.
Genbank Accession No. AY530575, Adeno-associated virus isolate hu.1 capsid protein VP1 (cap) gene, complete cds, dated Jun. 24, 2004, 2 pages.
Genbank Accession No. AY530576, Adeno-associated virus isolate hu.10 capsid protein VP1 (cap) gene, complete cds, dated Jun. 24, 2004, 2 pages.
Genbank Accession No. AY530577, Adeno-associated virus isolate hu.11 capsid protein VP1 (cap) gene, complete cds, dated Jun. 24, 2004, 2 pages.
Genbank Accession No. AY530578, Adeno-associated virus isolate hu.13 capsid protein VP1 (cap) gene, complete cds, dated Jun. 24, 2004, 2 pages.
Genbank Accession No. AY530580, Adeno-associated virus isolate hu.15 capsid protein VP1 (cap) gene, complete cds, dated Jun. 24, 2004, 2 pages.
Genbank Accession No. AY530581, Adeno-associated virus isolate hu.16 capsid protein VP1 (cap) gene, complete cds, dated Jun. 24, 2004, 2 pages.
Genbank Accession No. AY530582, Adeno-associated virus isolate hu.17 capsid protein VP1 (cap) gene, complete cds, dated Jun. 24, 2004, 2 pages.
Genbank Accession No. AY530583, Adeno-associated virus isolate hu.18 capsid protein VP1 (cap) gene, complete cds, dated Jun. 24, 2004, 2 pages.
Genbank Accession No. AY530584, Adeno-associated virus isolate hu.19 capsid protein VP1 (cap) gene, complete cds, dated Jun. 24, 2004, 2 pages.
Genbank Accession No. AY530585, Adeno-associated virus isolate hu.2 capsid protein VP1 (cap) gene, complete cds, dated Jun. 24, 2004, 2 pages.
Genbank Accession No. AY530586, Adeno-associated virus isolate hu.20 capsid protein VP1 (cap) gene, complete cds, dated Jun. 24, 2004, 2 pages.
Genbank Accession No. AY530587, Adeno-associated virus isolate hu.21 capsid protein VP1 (cap) gene, complete cds, dated Jun. 24, 2004, 2 pages.
Genbank Accession No. AY530588, Adeno-associated virus isolate hu.22 capsid protein VP1 (cap) gene, complete cds, dated Jun. 24, 2004, 2 pages.
Genbank Accession No. AY530589, Adeno-associated virus isolate hu.23 capsid protein VP1 (cap) gene, complete cds, dated Jun. 24, 2004, 2 pages.
Genbank Accession No. AY530590, Adeno-associated virus isolate hu.24 capsid protein VP1 (cap) gene, complete cds, dated Jun. 24, 2004, 2 pages.
Genbank Accession No. AY530591, Adeno-associated virus isolate hu.25 capsid protein VP1 (cap) gene, complete cds, dated Jun. 24, 2004, 2 pages.
Genbank Accession No. AY530592, Adeno-associated virus isolate hu.27 capsid protein VP1 (cap) gene, complete cds, dated Jun. 24, 2004, 2 pages.
Genbank Accession No. AY530593, Adeno-associated virus isolate hu.28 capsid protein VP1 (cap) gene, complete cds, dated Jun. 24, 2004, 2 pages.
Genbank Accession No. AY530594, Adeno-associated virus isolate hu.29 capsid protein VP1 (cap) gene, complete cds, dated Jun. 24, 2004, 2 pages.
Genbank Accession No. AY530595, Adeno-associated virus isolate hu.3 capsid protein VP1 (cap) gene, complete cds, dated Jun. 24, 2004, 2 pages.
Genbank Accession No. AY530596, Adeno-associated virus isolate hu.31 capsid protein VP1 (cap) gene, complete cds, dated Jun. 24, 2004, 2 pages.
Genbank Accession No. AY530597, Adeno-associated virus isolate hu.32 capsid protein VP1 (cap) gene, complete cds, dated Jun. 24, 2004, 2 pages.
Genbank Accession No. AY530598, Adeno-associated virus isolate hu.34 capsid protein VP1 (cap) gene, complete cds, dated Jun. 24, 2004, 2 pages.
Genbank Accession No. AY530599, Adeno-associated virus isolate hu.35 capsid protein VP1 (cap) gene, complete cds, dated Jun. 24, 2004, 2 pages.
Genbank Accession No. AY530600, Adeno-associated virus isolate hu.37 capsid protein VP1 (cap) gene, complete cds, dated Jun. 24, 2004, 2 pages.
Genbank Accession No. AY530601, Adeno-associated virus isolate hu.39 capsid protein VP1 (cap) gene, complete cds, dated Jun. 24, 2004, 2 pages.
Genbank Accession No. AY530602,Adeno-associated virus isolate hu.4 capsid protein VP1 (cap) gene, complete cds, dated Jun. 24, 2004, 2 pages.
Genbank Accession No. AY530603, Adeno-associated virus isolate hu.40 capsid protein VP1 (cap) gene, complete cds, dated Jun. 24, 2004, 2 pages.
Genbank Accession No. AY530604, Adeno-associated virus isolate hu.41 capsid protein VP1 (cap) gene, complete cds, dated Jun. 24, 2004, 2 pages.
Genbank Accession No. AY530605, Adeno-associated virus isolate hu.42 capsid protein VP1 (cap) gene, complete cds, dated Jun. 24, 2004, 2 pages.
Genbank Accession No. AY530606, Adeno-associated virus isolate hu.43 capsid protein VP1 (cap) gene, complete cds, dated Jun. 24, 2004, 2 pages.
Genbank Accession No. AY530607, Adeno-associated virus isolate hu.44 capsid protein VP1 (cap) gene, complete cds, dated Jun. 24, 2004, 2 pages.
Genbank Accession No. AY530608, Adeno-associated virus isolate hu.45 capsid protein VP1 (cap) gene, complete cds, dated Jun. 24, 2004, 2 pages.
Genbank Accession No. AY530609, Adeno-associated virus isolate hu.46 capsid protein VP1 (cap) gene, complete cds, dated Jun. 24, 2004, 2 pages.
Genbank Accession No. AY530610, Adeno-associated virus isolate hu.47 capsid protein VP1 (cap) gene, complete cds, dated Jun. 24, 2004, 2 pages.
Genbank Accession No. AY530611, Adeno-associated virus isolate hu.48 capsid protein VP1 (cap) gene, complete cds, dated Jun. 24, 2004, 4 pages.
Genbank Accession No. AY530612, Adeno-associated virus isolate hu.49 capsid protein VP1 (cap) gene, complete cds, dated Jun. 24, 2004, 2 pages.
Genbank Accession No. AY530613, Adeno-associated virus isolate hu.51 capsid protein VP1 (cap) gene, complete cds, dated Jun. 24, 2004, 2 pages.
Genbank Accession No. AY530614, Adeno-associated virus isolate hu.52 capsid protein VP1 (cap) gene, complete cds, dated Jun. 24, 2004, 2 pages.
Genbank Accession No. AY530615, Adeno-associated virus isolate hu.53 capsid protein VP1 (cap) gene, complete cds, dated Jun. 24, 2004, 2 pages.
Genbank Accession No. AY530616, Adeno-associated virus isolate hu.54 capsid protein VP1 (cap) gene, complete cds, dated Jun. 24, 2004, 2 pages.
Genbank Accession No. AY530617, Adeno-associated virus isolate hu.55 capsid protein VP1 (cap) gene, complete cds, dated Jun. 24, 2004, 2 pages.
Genbank Accession No. AY530618, Adeno-associated virus isolate hu.56 capsid protein VP1 (cap) gene, complete cds, dated Jun. 24, 2004, 2 pages.
Genbank Accession No. AY530619, Adeno-associated virus isolate hu.57 capsid protein VP1 (cap) gene, complete cds, dated Jun. 24, 2004, 2 pages.
Genbank Accession No. AY530620, Adeno-associated virus isolate hu.58 capsid protein VP1 (cap) gene, complete cds, dated Jun. 24, 2004, 2 pages.
Genbank Accession No. AY530621, Adeno-associated virus isolate hu.6 capsid protein VP1 (cap) gene, complete cds, dated Jun. 24, 2004, 2 pages.
Genbank Accession No. AY530622, Adeno-associated virus isolate hu.60 capsid protein VP1 (cap) gene, complete cds, dated Jun. 24, 2004, 2 pages.
Genbank Accession No. AY530623, Adeno-associated virus isolate hu.61 capsid protein VP1 (cap) gene, complete cds, dated Jun. 24, 2004, 2 pages.
Genbank Accession No. AY530624, Adeno-associated virus isolate hu.63 capsid protein VP1 (cap) gene, complete cds, dated Jun. 24, 2004, 2 pages.
Genbank Accession No. AY530625, Adeno-associated virus isolate hu.64 capsid protein VP1 (cap) gene, complete cds, dated Jun. 24, 2004, 2 pages.
Genbank Accession No. AY530626, Adeno-associated virus isolate hu.66 capsid protein VP1 (cap) gene, complete cds, dated Jun. 24, 2004, 2 pages.
Genbank Accession No. AY530627, Adeno-associated virus isolate hu.67 capsid protein VP1 (cap) gene, complete cds, dated Jun. 24, 2004, 2 pages.
Genbank Accession No. AY530628, Adeno-associated virus isolate hu.7 capsid protein VP1 (cap) gene, complete cds, dated Jun. 24, 2004, 2 pages.
Genbank Accession No. AY530629, Adeno-associated virus isolate hu.9 capsid protein VP1 (cap) gene, complete cds, dated Jun. 24, 2004, 2 pages.
Genbank Accession No. AY629583, Avian adeno-associated virus strain DA-1, complete genome, dated Sep. 10, 2004, 3 pages.
Genbank Accession No. AY631966, Adeno-associated virus 11 nonstructural protein and capsid protein genes, complete cds, dated Nov. 30, 2004, 3 pages.
Genbank Accession No. AY695370, Adeno-associated virus isolate hu.T17 capsid protein VP1 (cap) gene, complete cds, dated Nov. 15, 2005, 3 pages.
Genbank Accession No. AY695371, Adeno-associated virus isolate hu.T32 Rep 78 protein and capsid protein VP1 (cap) genes, complete cds, dated Nov. 15, 2005, 3 pages.
Genbank Accession No. AY695372, Adeno-associated virus isolate hu.T40 Rep 78 protein and capsid protein VP1 (cap) genes, complete cds, dated Nov. 15, 2005, 3 pages.
Genbank Accession No. AY695373, Adeno-associated virus isolate hu.T70 Rep 78 protein and capsid protein VP1 (cap) genes, complete cds, dated Nov. 15, 2005, 3 pages.
Genbank Accession No. AY695374, Adeno-associated virus isolate hu.T32 Rep 71 protein and capsid protein VP1 (cap) genes, complete cds, dated Nov. 15, 2005, 3 pages.
Genbank Accession No. AY695375, Adeno-associated virus isolate hu.T88 Rep 78 protein and capsid protein VP1 (cap) genes, complete cds, dated Nov. 15, 2005, 3 pages.
Genbank Accession No. AY695376, Adeno-associated virus isolate hu.S17 Rep 78 protein and capsid protein VP1 (cap) genes, complete cds, dated Nov. 15, 2005, 3 pages.
Genbank Accession No. AY695377, Adeno-associated virus isolate hu.LG15 capsid protein VP1 (cap) gene, complete cds, dated Nov. 15, 2005, 3 pages.
Genbank Accession No. AY695378, Adeno-associated virus isolate hu.T41 capsid protein VP1 (cap) gene, complete cds, dated Nov. 15, 2005, 2 pages.
GenBank Accession No. BC002532 ‘Homo sapiens Niemann-Pick disease, type C2, mRNA (cDNA clone MGC:1333 Image:3140870), complete eds’ (2006).
GenBank Accession No. BC045895 ‘Dania rerio Niemann-Pick disease, type C2, mRNA (cDNA clone MGC:56070 Image:5409780), complete eds’ (2003).
GenBank Accession No. BC054539 ‘Mus musculus Niemann Pick type C1, mRNA (cDNA clone MGC:62352 Image:6405214), complete eds’ (2006).
GenBank Accession No. BC090541 ‘Dania rerio Niemann-Pick disease, type C1, mRNA (cDNA clone Image:7149020), partial eds’ (2016).
GenBank Accession No. BC102504 ‘Bos taurus Niemann-Pick disease, type C2, mRNA (cDNA clone MGC:127986 Image:7954223), complete eds’ (2007).
GenBank Accession No. BC117178 ‘Homo sapiens NPC1 (Niemann-Pickdisease, type C1, gene)-like 1, mRNA (cDNA clone MGC:150787 Image:40125729), complete eds’ (2006).
GenBank Accession No. BC143756 ‘Homo sapiens NPC1 (Niemann-Pickdisease, type C1, gene)-like 1, mRNA (cDNA clone MGC:177287 Image:9052270), complete eds’ (2009).
GenBank Accession No. BC151276 ‘Bos taurus Niemann-Pick disease, type C1, mRNA (cDNA clone MGC:152602 Image:8433293), complete eds’ (2007).
Genbank Accession No. DQ813647, Adeno-Associated Virus 12 Rep 78 and VP1 genes, complete cds., dated Feb. 20, 2008, 3 pages.
GenBank Accession No. J00306 “Human somatostatin I gene and flanks” NCBJ (2 pages). (Jan. 13, 1995).
GenBank Accession No. J01901 “adeno-associated Virus 2, complete genome” NCBJ (3 pages). (Apr. 27, 1993).
GenBank Accession No. J02275 “Minute Virus of mice, complete genome” NCBJ (4 pages). (May 22, 1995).
GenBank Accession No. KJ893081 ‘Synthetic construct Homo sapiens clone cosb BroadEn 02475 NPC2 gene, encodes complete protein’ (2015).
Genbank Accession No. MI332400.1, Sequence 20 from U.S. Pat. No. 9,839,696, dated Feb. 12, 2018, 2 pages.
Genbank Accession No. MI332401.1, Sequence 21 from U.S. Pat. No. 9,839,696, dated Feb. 12, 2018, 2 pages.
Genbank Accession No. MI332402.1, Sequence 22 from U.S. Pat. No. 9,839,696, dated Feb. 12, 2018, 2 pages.
Genbank Accession No. MI332403.1, Sequence 23 from U.S. Pat. No. 9,839,696, dated Feb. 12, 2018, 2 pages.
Genbank Accession No. MI332404.1, Sequence 24 from U.S. Pat. No. 9,839,696, dated Feb. 12, 2018, 2 pages.
Genbank Accession No. MI332405.1, Sequence 25 from U.S. Pat. No. 9,839,696, dated Feb. 12, 2018, 2 pages.
Genbank Accession No. MI332406.1, Sequence 26 from U.S. Pat. No. 9,839,696, dated Feb. 12, 2018, 2 pages.
Genbank Accession No. MI332407.1, Sequence 27 from U.S. Pat. No. 9,839,696, dated Feb. 12, 2018, 2 pages.
Genbank Accession No. MI332408.1, Sequence 28 from U.S. Pat. No. 9,839,696, dated Feb. 12, 2018, 2 pages.
Genbank Accession No. MI332409.1, Sequence 29 from U.S. Pat. No. 9,839,696, dated Feb. 12, 2018, 2 pages.
Genbank Accession No. MI332410.1, Sequence 30 from U.S. Pat. No. 9,839,696, dated Feb. 12, 2018, 2 pages.
Genbank Accession No. MI332411.1, Sequence 31 from U.S. Pat. No. 9,839,696, dated Feb. 12, 2018, 2 pages.
Genbank Accession No. MI332412.1, Sequence 32 from U.S. Pat. No. 9,839,696, dated Feb. 12, 2018, 2 pages.
Genbank Accession No. MI332413.1, Sequence 33 from U.S. Pat. No. 9,839,696, dated Feb. 12, 2018, 2 pages.
Genbank Accession No. MI332414.1, Sequence 34 from U.S. Pat. No. 9,839,696, dated Feb. 12, 2018, 2 pages.
Genbank Accession No. MI332415.1, Sequence 35 from U.S. Pat. No. 9,839,696, dated Feb. 12, 2018, 2 pages.
GenBank Accession No. NC_000883 “Human parvo Virus 819, complete genome” NCBI (4 pages). (Feb. 10, 2015).
GenBank Accession No. NC_001358 “ParvoVirus H1, complete genome” NCBI (3 pages). (Feb. 10, 2015).
GenBank Accession No. NC_001510 “Minute Virus of mice, complete genome” NCBI (5 pages). (Mar. 28, 2016).
Genbank Accession No. NC_001729, Adeno-associated virus-3, complete genome, dated Aug. 13, 2018, 3 pages.
Genbank Accession No. NC_001829, Adeno-associated virus-4, complete genome, dated Aug. 13, 2018, 3 pages.
Genbank Accession No. NC_001863, Adeno-associated virus 3B, complete genome, dated Jan. 12, 2004, 4 pages.
Genbank Accession No. NC_002077, Adeno-associated virus-1, dated Aug. 13, 2018, 3 pages.
Genbank Accession No. NC_004828, Avian adeno-associated virus ATCC VR-865, complete genome, dated Aug. 13, 2018, 3 pages.
Genbank Accession No. NC_005889, Bovine adeno-associated virus, complete genome, dated Aug. 13, 2018, 3 pages.
Genbank Accession No. NC_006148.1, Snake parvovirus 1, complete genome, dated Aug. 13, 2018, 3 pages.
GenBank Accession No. NC_006152 “adeno-associated Virus 5, complete genome” NCBI (3 pages). (Dec. 8, 2008).
GenBank Accession No. NC_006261 “adeno-associated Virus 8, complete genome” NCBI (3 pages). (Mar. 11, 2010).
Genbank Accession No. NC_006263, Avian adeno-associated virus strain DA-1, complete genome, dated Aug. 13, 2018, 3 pages.
GenBank Accession No. NM 000271.4 Homo sapiens cholesterol transporter 1 (NPC1), mRNA (2017).
GenBank Accession No. NM 008720.2 Mus musculus cholesterol transporter 1 (Npc1), mRNA (2017).
GenBank Accession No. NM 023409.4 Mus musculus NPC intracellular cholesterol transporter 2 (Npc2 mRNA' (2017).
GenBank Accession No. NM 173918 Bos taurus NPC intracellular cholesterol transporter 2 NPC2), mRNA-;-(2017).
GenBank Accession No. NM_006432.3 ‘Homo sapiens NPC intracellular cholesterol transporter 2 (NPC2), mRNA’ (2017).
GenBank Accession No. NM_214206 “Sus scrofa NPC intracellular cholesterol transporter 2 (NPC2), mRNA,” dated Jun. 20, 2021, 2 pages.
GenBank Accession No. NP_044927 “capsid [Adeno-associated Virus-4]” NCBI (2 pages). (Jan. 28, 2010).
GenBank Accession No. P01166 “Somatostatin precursor [Contains: Somatostatin 28; Somatostatin-14]” NCBI (2 pages). (Sep. 15, 2003).
GenBank Accession No. P61278 “Somatostatin precursor [Contains: Somatostatin 28; Somatostatin-14]” NCBI (2 pages). (Nov. 13, 2019).
GenBank Accession No. U89790 “Adeno-associated virus 4, complete genome,” Aug. 21, 1997 [online]. (Retrieved online Feb. 21, 2019].
GenBank Accession No. X01457 “Parvovirus h-1, complete genome” NCBI (3 pages). (Apr. 18, 2005).
Genbank Accession No. Y18065, adeno-associated virus type 5 partial genome (cap and rep genes complete), dated Jan. 15, 1999, 3 pages.
GenBank Accession No. AH009962 “Hamster parvovirus” NCBI (1 page). (Aug. 25, 2016), replaced AF288061.
GenBank Accession No. AY530579 “adeno-associated Virus 9 isolate hu.14 capsid protein VP1 (cap). gene, complete eds” NCBI (2 pages). (Jun. 24, 2004).
GenBank Accession No. NC_001401 “adeno-associated Virus-2, complete genome” NCBI (5 pages). (Dec. 2, 2014).
GenBank Accession No. NC_001540 “Bovine parvovirus complete genome” NCBI (4 pages). (Nov. 30, 2009).
GenBank Accession No. NC_001701 “Goose parvovirus, complete genome” NCBI (4 pages). (Jan. 28, 2010).
GenBank Accession No. NC_001729 “adeno-associated virus-3, complete genome” NCBI (3 pages). (Jun. 28, 2010).
GenBank Accession No. NC_001829 “adeno-associated Virus 4, complete genome” NCBI (3 pages). (Jan. 28, 2010).
GenBank Accession No. NC_002077 “adeno-associated Virus-1, complete genome” NCBI (3 pages). (Mar. 11, 2010).
Ghusayni R, et al., ““Magnetic resonance imaging volumetric analysis in patients with Alternating hemiplegia of childhood: A pilot study,”” Eur J Paediatr Neural. 26:15-19 (2020).
Gorman et al. “Stable alteration of pre-mRNA splicing patterns by modified U7 small nuclear RNAs” Proceedings of the National Academy of Sciences 95:4929:4934 (1998).
Govindasamy et al, “Structurally mapping the diverse phenotype of adeno-associated virus serotype 4,” J. Virol 80:11556-11570 (2006).
Govindasamy et al., “Structural Insights into Adeno-Associated Virus Serotype 5,” J. Virology 87: 11187-11199 (2013).
Gray et al. “Preclinical Differences of Intravascular MV9 Delivery to Neurons and Glia: A Comparative Study of Adult Mice and Nonhuman Primates” Molecular Therapy, 19(6):1058-1069 (2011).
Gregorevic et al. “Systemic Microdystrophin Gene Delivery Improves Skeletal Muscle Structure and Function in Old Dystrophic mdx Mice” Molecular Therapy 16(4):657-664 (2008).
Grieger, et al., “Separate Basic Region Motifs within the Adeno-Associated Virus Capsid Proteins Are Essential for infectivity and Assembly.” J. Virol. (2006), 80(11): 5199-5210.
Grifman, et al., “Incorporation of tumor-targeting peptides into recombinant adeno-associated virus capsids”. Molecular Therapy (2001); vol. 3, No. 6, pp. 964-975.
Grimm D., et al., “In Vitro and in Vivo Gene Therapy Vector Evolution Via Multispecies Interbreeding and Retargeting of Adeno-Associated Viruses,” Journal of Virology, Jun. 2008, vol. 82(12), pp. 5887-5911, XP002610286.
Gurda et al. “Capsid Antibodies to Different adeno-Associated Virus Serotypes Bind Common Regions” Journal of Virology, 87(16):9111-9124 (2013).
Gurda et al., “Mapping a Neutralizing Epitope onto the Capsid of Adeno-Associated Virus Serotype 8,” Journal of Virology 86(15): 7739-7751 (2012).
Hadaczek et al. “Transduction of Nonhuman Primate Brain with Adeno-Associated Virus Serotype 1: Vector Trafficking and Immune Response” Human Gene Therapy, 20(3):225-237 (2009).
Hajitou et al., “Vascular targeting: recent advances and therapeutic perspectives,” TCM 16:80-88 (2006).
Hauck et al. “Characterization of Tissue Tropism Determinants of Adeno-Associated Virus Type 1” Journal of Virology 77(4):2768-2774 (2003).
Heinzen EL, et al., “De nova mutations in ATP1A3 cause alternating hemiplegia of childhood,” Nat Genet. 44 (9):1030-1034 (2012).
Helseth AR, et al., “Novel E815K knock-in mouse model of alternating hemiplegia of childhood,” Neurobiol Dis. 119:100-112 (2018).
Higgins, Desmond G., and Sharp, Paul M. “CLUSTAL: a package for performing multiple sequence alignment on a microcomputer.” Gene (1988); 73.1: 237-244.
Holm R, et al., “B. Neurological disease mutations of a3 Na+, K+-ATPase: Structural and functional perspectives and rescue of compromised function,” Biochim Biophys Acta. 1857(11):1807-1828 (2016).
Hoshijima et al. ““Chronic suppression of heart-failure progression by a pseudophosphorylated mutant of phospholamban via in vivo cardiac rAAV gene delivery”” Nature Medicine 8:864-871 (2002).
Huang et al. “Characterization of the adeno-Associated Virus 1 and 6 Sialic Acid Binding Site” Journal of Virology, 9 (11 ):5219-5230 (2016).
Huang et al. “ParvoVirus glycan interactions” Current Opinion in Virology 7:108-118 (2014).
Huang, X et al., ‘Dynamic programming algorithms for restriction map comparison,’ Cabios, Vo1.8, No. 5., pp. 511-520, (1992).
Hughes et al., “AAV9 intracerebroventricular gene therapy improves lifespan, locomotor function and pathology in a mouse model of Niemann-Pick type C1 disease,” Human Molecular Genetics 27(17)3079-3098 (2018).
Hunanyan AS, et al., Knock-in mouse model of alternating hemiplegia of childhood: behavioral and electrophysiologic characterization. Epilepsia. 56(1):82-93 (2015).
Hunanyan AS, et al., “Mechanisms of increased hippocampal excitability in the Mashl+/− mouse model of Na+ /K+-ATPase dysfunction,” Epilepsia 59(7):1455-1468 (2018).
Ikeda K, et al., ““Knockout of sodium pump a3 subunit gene (Atp1a3-/-) results in perinatal seizure and defective respiratory rhythm generation,”” Brain Res. 1666:27-37 (2017).
Isaksen T J, et al., “Hypothermia-induced dystonia and abnormal cerebellar activity in a mouse model with a single disease-mutation in the sodium-potassium pump,” PLoS Genet. 13(5):e1006763, pp. 1-23 (2017).
Janson, C. et al., ‘Clinical protocol. Gene therapy of Canavan disease: AAV-2 vector for neurosurgical delivery of aspartoacylase gene (ASPA) to the human brain,’ Hum. Gene Ther., 13(11 ):1391-1412 (Jul. 2002).
Kailasan et al., “Structure of an enteric pathogen, bovine parvovirus,” Virology 89:2603-2614 (2015).
Kaplitt, M.G. et al. (1994). “Long-term gene expression and phenotypic correction using adenoassociated virus vectors in the mammalian brain,” Nature Genetics 6:148-154.
Karlin et al. “Applications and statistics for multiple high-scoring segments in molecular sequences” Proceedings of National Academy of Sciences 90:5873-5877 (1993).
Kashiwakura et al. “Hepatocyte Growth Factor Receptor Is a Coreceptor for Adeno-Associated Virus Type 2 Infection” Journal of Virology, 79(1).609-614 (2005).
Kauffman et al., “Mechanism Matters: A Taxonomy of Cell Penetrating Peptides,” Trends in Biochemcial Sciences, Elsevier, Amsterdam, NL 40(12):749-764 (2015).
Kawakami et al. “Cloning of the gene coding for a shared human melanoma antigen recognized by autologous T cells infiltrating into tumor” Proceedings of the National Academy of Sciences 91:3515-3519 (1994).
Kawakami et al. “Identification of the Immunodominant Peptides of the MART-1 Human Melanoma Antigen Recognized by the Majority of HLA-A2-restricted Tumor Infiltrating Lymphocytes” The Journal of Experimental Medicine 180:347-352 (1994).
Kells, A.P., et al., “AAV-Mediated Gene Delivery of BDNF or GDNF is Neuroprotective in a Model of Huntington Disease,” Molecular Therapy, May 2004, vol. 9(5), pp. 682-688.
Kirshenbaum GS, et al., “Alternating hemiplegia of childhood-related neural and behavioural phenotypes in Na+, K+-ATPase a3 missense mutant mice,” PLoS One. 8(3):e60141, pp. 1-15 (2013).
Koivunen et al., “Identification of Receptor Ligands with Phase Display Peptide Libraries,” J. Nucl. Med. 40:883-888 (1999).
Krissinel et al. “Secondary-structure matching (SSM)., a new tool for fast protein structure alignment in three dimensions” Acta Crystallographica Section D: Biological Crystallography, D60:2256-2268 (2004).
Kuck et al. “Development of AAV serotype-specific ELISAs using novel monoclonal antibodies” Journal of Virological Methods, 140(1-2):17-24 (2007) (Abstract only).
Kumar et al. “MEGA7: Molecular Evolutionary Genetics Analysis Version 7.0 for Bigger Datasets” Molecular Biology and Evolution, 33(7):1870-1874 (2016).
Lein et al. “Genome-wide atlas of gene expression in the adult mouse brain” Nature, 445(7124):168-176 (2007). (Abstract only).
Lerch et al., “The structure of adeno-associated virus serotype 3B (AAV-3B): insights into receptor binding and immune evasion,” Virology 403(1):26-36 (2010).
Levine et al. “The Tumor Suppressor Genes” Annual Review of Biochemistry 62:623-651 (1993).
Li et al. “Development of Patient-specific AAV Vectors After Neutralizing Antibody Selection for Enhanced Muscle Gene Transfer” Molecular Therapy, 24(1):53-65 (2016).
Li et al. “Engineering and Selection of Shuffled AAV Genomes: A New Strategy for Producing Targeted Biological Nanoparticles” Molecular Therapy, 16(7):1252-1260 (2008).
Li et al. “Single Amino Acid Modification of adeno-Associated Virus Capsid Changes Transduction and Humeral Immune Profiles” Journal of Virology, 86(15):7752-7759 (2012).
Lisowski L., et al., “Selection and Evaluation of Clinically Relevant AAV Variants in a Xenograft Liver Model,” Nature, Feb. 2014, vol. 506 (7488), pp. 382-386, XP055573596.
Loftus, SK et al., ‘Murine Model of Niemann-Pick C Disease: Mutation in a Cholesterol Homeostasis Gene,’ Science, 277(5323):232-235 (Jul. 1997).
Lux et al. “Green Fluorescent Protein-Tagged Adena-Associated Virus Particles Allow the Study of Cytosolic and Nuclear Trafficking” Journal of Virology, 79(18):11776-11787 (2005).
Madigan et al. “Engineering AAV receptor footprints for gene therapy” Current Opinion in Virology, 18:89-96 (2016).
Margolskee, R. F. “Epstein-Barr Virus Based Expression Vectors” Current Topics in Microbiology and Immunology 158:67-95 (1992).
Masoud M, et al., “Diagnosis and Treatment of Alternating Hemiplegia of Childhood,” Curr Treat Options Neural. 19(2):8 (2017).
Mauro et al., “A critical analysis of codon optimization in human therapeutics,” Trends in Molecular Medicine, Nov. 2014, vol. 20, No. 11, pp. 604-613.
McCarty, D.M., et al., “Self-complementary recombinant adeno-associated virus (scAAV) vectors promote efficient transduction independently of DNA synthesis,” Gene Therapy 8, 1248-1254 (2001).
McCraw et al. “structurE of adeno-associated virus-2 In Complex with Neutralizing Monoclonal antibodY A20” Virology, 431 (1-2):40-49 (2012).
McLaughlin et al., “Adeno-associated virus general transduction vectors: analysis of proviral structures,” J. Virol., (1988) 62:1963-1973.
Mikati et al., “Alternating hemiplegia of childhood: clinical manifestations and long-term outcome,” Pediatr Neurol. 23(2):134-141 (2000).
Miller et al. “Production, purification and preliminary X-ray crystallographic studies of adenoassociated virus serotype 1” Acta Crystallographica Section F: Structural Biology and Crystallization Communications, 62(Pt 12):1271-1274 (2006).
Mingozzi et al., “Overcoming the Host Immune Response to Adeno-Associated Virus Gene Delivery Vectors: The Race Between Clearance, Tolerance, Neutralization, and Escape,” Annual Review of Virology 1(1):511-534 (2017).
Miyamura et al. “ParvoVirus particles at platforms for protein presentation” Proceedings of National Academy of Sciences 91 :8507-8511, 1994.
Mori et al. “Two novel adeno-associated viruses from cynomolgus monkey: pseudotyping characterization of capsid protein” Virology 330:375-383 (2004).
Muller et al. “Random peptide libraries displayed on adeno-associated virus to select for targeted gene therapy vectors”, Nat Biotechnol, Sep. 2003; 21(9):1040-6. Epub Aug. 3, 2003.
Muramatsu et al. “Nucleotide Sequencing and Generation of an Infectious Clone of adeno-Associated Virus 3”, Virology, 221(0367):208-217 (1996).
Murlidharan et al. “Biology of adeno-associated viral vectors in the central nervous system” Frontiers in Molecular Neuroscience, 7(76):1-9 (2014), pp. 1-12.
Murlidharan et al. “Glymphatic fluid transport controls paravascular clearance of MV vectors from the brain” JCI Insight, 1 (14):e88034 (2016).
Murlidharan et al. “Unique Glycan Signatures Regulate adeno-Associated Virus Tropism in the Developing Brain” Journal of Virology 89(7):3976-3987 (2015).
Muzyczka, N. “Use of adeno-Associated Virus as a General Transduction Vector for Mammalian Cells,” Current Topics in Microbiology and Immunology 158:97-129 (1992).
Nam et al. “Structure of Adeno-Associated Virus Serotype 8, a Gene Therapy Vector” Journal of Virology, 81 (22):12260-12271 (2007).
Nathwani et al. “Long-Term Safety and Efficacy of Factor IX Gene Therapy in Hemophilia B” The New England Journal of Medicine, 371 (21):1994-2004 (2014).
Needleman and Wunsch, “A general method applicable to the search for similarities in the amino acid sequence of two proteins.” J Mol Biol. (1970); 48(3): 443-453.
Newton et al., Phage Peptide Display in Handbook of Experimental Pharmacology, pp. 145-163, Springer-Verlag, Berlin (2008).
Ng et al. “Structural Characterization of the Dual Glycan Binding adeno-Associated Virus Serotype 6” Journal of Virology, 84(24):12945-12957 (2010).
Nguyen Vu et al., “Cerebellar Purkinje cell activity drives motor learning”, Nature Neuroscience 16(12):1734-1736 (2013).
Padron et al. “Structure of adeno-Associated Virus Type 4” Journal of Virology 79(8):5047-5058 (2005).
Palombo et al. “Site-Specific Integration in Mammalian Cells Mediated by a New Hybrid Baculovirus-Adeno-Associated Virus Vector” Journal of Virology72(6):5025-5034 (1998).
Papadakis, ED et al., ‘Promoters and Control Elements: Designing Expression Cassettes for Gene Therapy,’ Curr. Gene Therapy, vol. 4, No. 1, pp. 89-113, (Mar. 2004).
Partial Supplementary European Search Report issued by the European Patent Office for Application No. 16852471.8, dated Apr. 24, 2019, 17 pages.
Passini, MA et al., ‘Distribution of a Lysosomal Enzyme in the Adult Brain by Axonal Transport and by Cells of the Rostral Migratory Stream,’ J. Neuroscience, 22(15):6437-6446 (Aug. 2002).
Paul, CA et al., ‘Adenovirus Expressing an NPCI-GFP Fusion Gene Corrects Neuronal and Nonneuronal Defects Associated With Niemann Pick Type C Disease,’ J. Neurosci. Res., vol. 81, No. 5, pp. 706-719 (Sep. 2005).
Pearson et al., “Improved tools for biological sequence comparison,” Proc. Natl. Acad. Sci. USA 85, 2444-2448 (1988).
Piguet et al, “Rapid and Complete Reversal of Sensory Ataxia by Gene Therapy in a Novel Model of Friedreich Ataxia”, Molecular Therapy, Nature Publishing Group, GB 26(8), pp. 1-13 (2018).
Pillay et al. “An essential receptor for adeno-associated virus infection” Nature, 530(7588):108-112 (2016).
Powell et al. Characterization of a Novel Adena-Associated Viral Vector with Preferential Oligodendrocyte Tropism. Gene Therapy, 2016. 23:807-814.
Pulicherla et al. “Engineering Liver-detargeted AAV9 Vectors for Cardiac and Musculoskeletal Gene Transfer” Molecular Therapy, 19(6):1070-1078 (2011).
Puttaraju et al. “Spliceosome-mediated RNA trans-splicing as a tool for gene therapy” Nature Biotechnology 17:246-252 (1999).
Robbins et al. “Recognition of Tyrosinase by Tumor-infiltrating Lymphocytes from a Patient Responding to Immunotherapy” Cancer Research 54:3124-3126 (1994).
Rosenberg “The Immunotherapy of Solid Cancers Based on Cloning the Genes Encoding Tumor-Rejection Antigens” Annual Review of Medicine 47:481-491 (1996).
Rosenberg et al. “A New Era for Cancer Immunotherapy Based on the Genes that Encode Cancer Antigens” Immunity 10:281-287 (1999).
Rosenberg et al. “Comparative Efficacy and Safety of Multiple Routes of Direct CNS Administration of Adeno-Associated Virus Gene Transfer Vector Serotype rh.10 Expressing the Human Arylsulfatase A cDNA to Nonhuman Primates” Human Gene Therapy Clinical Development, 25(3):164-177 (2014).
Saitou, N. et al. (1987). “The neighbor-joining method: A new method for reconstructing phylogenetic trees,” Mol. Biol. Evol. 4:406-425.
Salinas et al. “A hitchhiker's guide to the nervous system: the complex journey of viruses and toxins” Nature Reviews Microbiology, 8(9):645-655 (2010). (Abstract only).
Selot et al., “Developing Immunologically Inert Adeno-Associated Virus (AAV). Vectors for Gene Therapy: Possibilities and Limitations,” Current Pharmaceutical Biotechnology, Bentham Science Publishers, NL 14(12).1072-1082 (2013).
Severino M, et al., “White matter and cerebellar involvement in alternating hemiplegia of childhood,” J Neurol. 267 (5):1300-1311 (2020).
Shade et al. “Nucleotide Sequence and Genome Organization of Human Parvovirus B19 Isolated from the Serum of a Child during Aplastic Crisis” Journal of Virology 28(3):921-936 (1986).
Sharp et al. “RNA Interference” Science 287(5462):2431-2433 (2000).
Shen et al. “Engraftment of a Galactose Receptor Footprint onto adeno-associated Viral Capsids Improves Transduction Efficiency” The Journal of Biological Chemistry, 288(40):28814-28823 (2013).
Shen et al., Multiple Roles for Sialylated Glycans in Determining the Cardiopulmonary Tropism of Adeno-Associated Virus 4, Journal of Virology 87(24):13206-13213 (2013).
Shi et al. “Insertional Mutagenesis at Positions 520 and 584 of adeno-Associated Virus Type 2 (AAV2). Capsid Gene and Generation of AAV2 Vectors with Eliminated Heparin-Binding Ability and Introduced Novel Tropism” Human Gene Therapy 17:353-361 (2006).
Sirin S, Apgar JR, Bennett EM, Keating AE. AB-Bind: Antibody binding mutational database for computational affinity predictions. Protein Sci. Feb. 2016;25(2):393-409. Epub Nov. 6, 2015.
Smith et al, “Comparison of Biosequences”, Advanced in Applied Mathematics, vol. 2, Issue 4, Dec. 1981, pp. 482-489.
Sonntag et al. “Adeno-Associated Virus Type 2 Capsids with Externalized VP1NP2 Trafficking Domains Are Generated prior to Passage through the Cytoplasm and Are Maintained until Uncoating Occurs in the Nucleus” Journal of Virology, 80(22):11040-11054 (2006).
Srivastava et al. “Nucleotide Sequence and Organization of the Adeno Associated Virus 2 Genome.” Journal of Virology (1983); 45:2, p. 555-564.
Summerford et al. “Membrane-Associated Heparan Sulfate Proteoglycan Is a Receptor for adeno-Associated Virus Type 2 Virions” Journal of Virology, 72(2):1438-1445 (1998).
Tellez et al. “Characterization of Naturally-Occurring Humoral Immunity to AAV in Sheep” PLoS ONE, 8(9):e75142 (2013).
Tinsley et al. “Amelioration of the dystrophic phenotype of mdx mice using a truncated utrophin transgene” Nature 384(6607):349-353 (1996).
Titeux et al., “SIN Retroviral Vectors Expressing COL7A1 Under Human Promoters for Ex Vivo Gene Therapy of Recessive Dystrophic Epidermolysis Bullosa,” Mol. Ther., 2010 18:1509-1518.
Tsao et al. The Three-Dimensional Structure of Canine ParvoVirus and Its Functional Implications Science 251 (5000):1456-1464 (1991).
Tse et al., “Strategies to Circumvent Humoral Immunity to Adeno-Associated Viral Vectors,” Expert Opinion on Biological Therapy 15(6):845-855 (2015).
Tse L.V., et al., “Structure-Guided Evolution of Antigenically Distinct Adeno-Associated Virus Variants for Immune Evasion,” Proceedings of the National Academy of Sciences, Jun. 2017, vol. 114(24), pp. E4812-E4821, XP055590029.
Tseng et al. “Adeno-Associated Virus Serotype 1 (AAV1).- and AAV5-Antibody Complex Structures Reveal Evolutionary Commonalities in ParvoVirus Antigenic Reactivity” Journal of Virology, 89(3):1794-1808 (2015).
Tseng et al. “Generation and characterization of anti-adeno-associated Virus serotype 8 (AAV8). and anti-AAV9 monoclonal antibodies” Journal of Virological Methods, 236:105-110 (2016).
Tseng et al. “Mapping the AAV capsid host antibody response toward the development of second generation gene delivery vectors” Frontiers in Immunology, 5(9):1-11 (2014). UniProt Accession No. O15118, dated May 30, 2000, 21 pages.
Urabe et al. “Insect Cells as a Factory to Produce adeno-Associated Virus Type 2 Vectors” Human Gene Therapy 13:1935-1943 (2002).
Various: Abstracts , 20th Annual Meeting of the American-Society-of-Gene-and-Cell-Therapy (ASGCT); Washington, DC, USA; May 10-13. 2017 , Molecular Therapy: the Journal of the American Society of Gene Therapy 25:1-363 (2017).
Veldwijk, MR et al., ‘Development and optimization of a real-time quantitative PCR-based method for the titration of AAV-2 vector stocks,’ Mal. Ther.,6(2):272-8 (Aug. 2002).
Veron et al. ““Humeral and Cellular Capsid-Specific Immune Responses to Adena-Associated Virus Type 1 in andomized Healthy Donors”” The Journal of Immunology, 188:6418-6424 (2012).
Vincent et al. “Long-term correction of mouse dystrophic degeneration by adenovirusmediated transfer of a minidystrophin gene” Nature Genetics 5:130-134 (1993).
Walters et al. “Structure of adeno-Associated Virus Serotype 5” Journal of Virology 78(7):3361-3371 (2004).
Wang et al. “Adeno-associated Virus vector carrying human minidystrophin genes effectively ameliorates muscular dystrophy in mdx mouse model” Proceedings of the National Academy of Sciences 97(25):13714-13719 (2000).
Wang et al. “Expanding the genetic code” Annual Review of Biophysics and Biomolecular Structure 35:225-249 (2006).
Wang et al., “Identification of an adeno-associated Virus binding epitope for AVB sepharose affinity resin,” Molecular Therapy—Methods & Clinical Development vol. 2, pp. 1-6 (2015).
Wassif, CA et al., ‘High Incidence of Unrecognized Visceral/Neurological Lateonset Niemann-Pick Disease, type C1 Predicted by Analysis of Massively Parallel Sequencing Data Sets,’ Genet Med., 18(1):41-48 (Jan. 2016).
Weller et al. “Epidermal growth factor receptor is a co-receptor for adeno-associated virus serotype 6” Nature Medicine, 16(6):662-664 (2010).
Williams et al. ““Monocyte maturation, HIV susceptibility, and transmigration across the blood brain barrier are critical in HIV neuropathogenesis”” Journal of Leukocyte Biology, 91 (3):401-415 (2012).
Winkler et al., “Changing the Antigen Binding Specificity by Single Point Mutations of an Antip24 (HIV-1) Antibody”, The Journal of Immunology, 165: 4505-4514 (2000).
Wobus et al. “Monoclonal Antibodies against the Adena-Associated Virus Type 2 (AAV-2) Capsid: Epitope Mapping and Identification of Capsid Domains Involved in AAV-2-Cell Interaction and Neutralization of AAV-2 Infection,” J. of Virology, 74(19):9281-9293 (2000).
Work, et al., “Vascular bed-targeted in vivo gene delivery using tropism-modified adenoassociated viruses.” Mol. Ther.; vol. 13, No. 4, pp. 683-693 (Apr. 2006).
Xiao et al. “Gene Therapy Vectors Based on adeno-Associated Virus Type 1” Journal of Virology 73(5):3994-4003 (1999).
Xiao et al. “Interpretation of Electron Density with Stereographic Roadmap Projections” Journal of Structural Biology, 158(2):182-187 (2007).
Xiao et al., “Gene transfer by adeno-associated virus vectors into the central nervous system,” Exp. Neurobiol., (1997) 144:113-124.
Xie et al. “Canine ParvoVirus Capsid Structure, Analyzed at 2.9 A Resolution” Journal of Molecular Biology 264(3):497-420 (1996).
Xie et al. “The atomic structure of adeno-associated Virus (AAV-2)., a vector for human gene therapy” Proceeding of the National Academy of Sciences 99(16):10405-10410 (2002).
Xie, J. et al., “Short DNA Hairpins Compromise Recombinant Adeno-Associated Virus Genome Homogeneity,” Mol. Ther., 25(6): 1363-1374 (2017).
Yang et al. “Global CNS Transduction of Adult Mice by Intravenously Delivered rAAVrh.8 and rAAVrh.10 and Nonhuman Primates by rAAVrh.10” Molecular Therapy, 22(7):1299-1309 (2014).
Ye Q, et al., “The AAA+ ATPase TRIP13 remodels HORMA domains through N-terminal engagement and unfolding,” EMBO J. 36(16):2419-2434 (2017).
Zhang et al. “Recombinant adenoVirus expressing adeno-associated Virus cap and rep proteins supports production of high-titer recombinant adeno-associated virus” Gene Therapy 8:704-712 (2001).
Zhang et al. “Several rAAV Vectors Efficiently Cross the Blood-brain Barrier and Transduce Neurons and Astrocytes in the Neonatal Mouse Central Nervous System” Molecular Therapy, 19(8): 1440-1448 (2011 ).
Zhang, “Endocytic mechanisms and drug discovery in neurodegenerative diseases,” Frontiers in Bioscience 13:6086-6105 (2008).
Zhong et al. ““Tyrosine-phosphorylation of AA V2 vectors and its consequences on viral intracellular trafficking and transgene expression”” Virology, 381 (2):194-202 (2008).
Zhong et al. “Next generation of adeno-associated virus 2 vectors: Point mutations in tyrosines lead to high-efficienc ransduction at lower doses” Proceedings of the National Academy of Sciences USA, 105(22):7827-7832 (2008).
Zinn, E. et al., “In Silico Reconstruction of the Viral Evolutionary Lineage Yields a Potent Gene Therapy Vector,” Cell Reports, Aug. 2015; 12:1056-1068.
Zolotukhin et al. “Production and purification of serotype 1, 2, and 5 recombinant adeno-associated viral vectors” Methods, 28(2):158-167 (2002) (Abstract only).
Zolotukhin, et al., “Recombinant adeno-associated virus purification using novel methods improves infectious titer and yield.” Gene Therapy (1999); vol. 6, pp. 973-985.
Cleves, Ann E. “Protein transport: The nonclassical ins and outs” Current Biology7:R318-R320 (1997).
Ferrari et al. “New developments in the generation of Ad-free high-titer rAAV gene therapy vectors” Nature Medicine 3(11):1295-1297 (1997).
Gonzales, “Cross-Species Evolution of Synthetic A hetis AAV Strains for clinical Translation,” ASGCT, abstract #23 2 pages (2020).
Havlik et al, “Co-Evolution of AAV Capsid Antigenecity and Tropism Through a Structure-Guided Approach,” ASGCT, pp. 1-17 (2020).
Havlik, Engineering A Humanized AAV8 Capsid Through Iterative Structure-Guided Evolution ASGCT, 24 pages. (2019), abstract #100, 1 page.
Higgins, DG et al., ‘Fast and sensitive multiple sequence alignments on a microcomputer,’ Comput Appl Biosci., 5(2):151-3, (Apr. 1989).
Li et al. “Construction of phospholamban antisense RNA recombinant adeno-associated Virus vector and its effects in rat cardiomyocytes” Acta Pharmalogica Sinica 26(1).51-55 (2005).
Mingozzi et al. “Immune responses to AAV vectors: overcoming barriers to successful gene therapy” Blood, 122 (1):23-36 (2013).
Murlidharan et al. “265. Polysialic Acid as a Novel Regulator of AAV Tropism in the Developing Brain” Molecular Therapy 23(Supplement 1 ):S106 (2015), 1 page.
Murlidharan et al. “CNS-restricted Transduction and CRISPR/Cas9-mediated Gene pages Deletion with an Engineered AAV Vector” Molecular Therapy: Nucleic Acids, 5:e338 (2016), pp. 1-12.
Smith et al., “Structural Mapping of AAV9 Antigenic Sites and the Engineering of Immune Escape Variants,” Molecular Therapy; 20th Annual Meeting of the American Society of Gene and Cell Therapy (ASGCT).; Washington, DC, A; May 10-13, 2017, Nature Publishing Group, GB vol. 25, No. 5, Suppl 1 (2017), abstract #733, 1 page.
Smith, TF et al., ‘Identification of Common Molecular Subsequences,’ Journal of Molecular Biology, 147:195-197, PMID 7265238. doi:10.1016/0022-2836(81)90087-5, (1981).
Wang et al., “Selection of neutralizing antibody-resistant AAV8 variants with structure-guided site-specific saturated mutagenesis,” Molecular Therapy, 2011, vol. 19 Suppl. 1, S129, abstract #333 1 page.
Wu et al. “alpha2,3 and alpha2,6 N-Linked Sialic Acids Facilitate Efficient Binding and Transduction by Adeno-Associated Virus Types 1 and 6” Journal of Virology, 80(18):9093-9103 (2006).
Related Publications (1)
Number Date Country
20240067986 A1 Feb 2024 US
Provisional Applications (1)
Number Date Country
62821710 Mar 2019 US
Continuations (2)
Number Date Country
Parent 17479577 Sep 2021 US
Child 18372334 US
Parent PCT/US2020/023877 Mar 2020 US
Child 17479577 US