Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
CHIMERIC AAV5 CAPSIDS
Document Type and Number:
WIPO Patent Application WO/2022/226294
Kind Code:
A1
Abstract:
The invention relates to chimeric AAV5 capsids, virus vectors comprising the same, and methods of using the vectors such as to target the liver. The invention further relates to chimeric AAV5 capsids with improved infectivity to hepatocytes, virus vectors comprising the same, and methods of using the vectors to target hepatocytes with improved infectivity.

Inventors:
XIAO BIN (US)
XIAO XIAO (US)
LI JUAN (US)
Application Number:
PCT/US2022/025925
Publication Date:
October 27, 2022
Filing Date:
April 22, 2022
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
UNIV NORTH CAROLINA CHAPEL HILL (US)
International Classes:
C07K14/005; A61K48/00; A61P7/02; C12N15/86
Foreign References:
US20200354743A12020-11-12
Other References:
QIAN RANDOLPH: "BIOENGINEERING OF ADENO-ASSOCIATED VIRUS SEROTYPE 5 FOR INCREASED LIVER TRANSDUCTION AND RETENTION OF LOW HUMORAL SEROREACTIVITY", PHD DISSERTATION, UNIVERSITY OF NORTH CAROLINA AT CHAPEL HILL, 1 January 2020 (2020-01-01), XP055979075
GOVINDASAMY LAKSHMANAN, DIMATTIA MICHAEL A., GURDA BRITTNEY L., HALDER SUJATA, MCKENNA ROBERT, CHIORINI JOHN A., MUZYCZKA NICHOLAS: "Structural Insights into Adeno-Associated Virus Serotype 5", JOURNAL OF VIROLOGY, THE AMERICAN SOCIETY FOR MICROBIOLOGY, US, vol. 87, no. 20, 15 October 2013 (2013-10-15), US , pages 11187 - 11199, XP055865805, ISSN: 0022-538X, DOI: 10.1128/JVI.00867-13
DATABASE PROTEIN 13 August 2018 (2018-08-13), ANONYMOUS : "capsid protein [adeno-associated virus 5] S", XP055979080, retrieved from NCBI Database accession no. YP_068409.1
QIAN RANDOLPH, XIAO BIN, LI JUAN, XIAO XIAO: "Directed Evolution of AAV Serotype 5 for Increased Hepatocyte Transduction and Retained Low Humoral Seroreactivity", MOLECULAR THERAPY- METHODS & CLINICAL DEVELOPMENT, NATURE PUBLISHING GROUP, GB, vol. 20, 1 March 2021 (2021-03-01), GB , pages 122 - 132, XP055864144, ISSN: 2329-0501, DOI: 10.1016/j.omtm.2020.10.010
HIDA, K. ; WON, S.Y. ; DI PASQUALE, G. ; HANES, J. ; CHIORINI, J.A. ; OSTERMEIER, M.: "Sites in the AAV5 capsid tolerant to deletions and tandem duplications", ARCHIVES OF BIOCHEMISTRY AND BIOPHYSICS, ACADEMIC PRESS, US, vol. 496, no. 1, 1 April 2010 (2010-04-01), US , pages 1 - 8, XP026965201, ISSN: 0003-9861
Attorney, Agent or Firm:
PASMAN, Lesley (US)
Download PDF:
Claims:
THAT WHICH IS CLAIMED IS: 1. A chimeric adeno-associated virus 5 (AAV5) capsid comprising: a) a VP1 capsid protein of a first AAV serotype that is not AAV5; and/or b) a VP2 capsid protein of a second AAV serotype that is not AAV5 and that is different from said first AAV serotype or the same as said first AAV serotype. 2. The chimeric AAV5 capsid of claim 1, wherein the first AAV serotype and the second AAV serotype are different. 3. The chimeric AAV5 capsid of claim 1, wherein the first AAV serotype and the second AAV serotype are the same. 4. The chimeric AAV5 capsid of any one of claims 1-3, wherein the first and/or second, AAV serotype is AAV1, AAV2, AAV3, AAV4, AAV6, AAV7, AAV8, and/or AAV9. 5. The chimeric AAV5 capsid of claim 4, comprising, consisting essentially of, or consisting of an amino acid sequence at least 90% identical to the amino acid sequence of SEQ ID NO:8. 6. The chimeric AAV5 capsid of claim 4, comprising, consisting essentially of, or consisting of an amino acid sequence at least 90% identical to the amino acid sequence of SEQ ID NO:9. 7. The chimeric AAV5 capsid of claim 4, comprising, consisting essentially of, or consisting of an amino acid sequence at least 90% identical to the amino acid sequence of SEQ ID NO:10. 8. The chimeric AAV5 capsid of claim 4, comprising, consisting essentially of, or consisting of an amino acid sequence at least 90% identical to the amino acid sequence of SEQ ID NO:11.

9. The chimeric AAV5 capsid of claim 4, comprising, consisting essentially of, or consisting of an amino acid sequence at least 90% identical to the amino acid sequence of SEQ ID NO:12. 10. The chimeric AAV5 capsid of claim 4, comprising, consisting essentially of, or consisting of an amino acid sequence at least 90% identical to the amino acid sequence of SEQ ID NO:13. 11. The chimeric AAV5 capsid of claim 4, comprising, consisting essentially of, or consisting of an amino acid sequence at least 90% identical to the amino acid sequence of SEQ ID NO:14. 12. The chimeric AAV5 capsid of claim 4, comprising, consisting essentially of, or consisting of an amino acid sequence at least 90% identical to the amino acid sequence of SEQ ID NO:15. 13. A chimeric adeno-associated virus 5 (AAV5) capsid comprising an AAV5 VP3 capsid protein comprising an insertion following amino acid residue Q574 in the VP3 variable region (VR)-VIII region of an AAV VP3 VR from an AAV serotype that is different from AAV5, wherein the numbering corresponds to the amino acid sequence of SEQ ID NO:20. 14. The chimeric AAV5 capsid of claim 13, wherein the chimeric AAV5 capsid comprising the AAV5 VP3 capsid protein comprising an insertion following amino acid residue Q574 in the VP3 variable region (VR)-VIII region of an AAV VP3 VR from an AAV serotype that is different from AAV5 has enhanced liver tropism as compared to the liver tropism of a corresponding wildtype AAV5 capsid. 15. The chimeric AAV5 capsid of claim 14, wherein the enhanced liver tropism is enhanced about 5.0 fold or higher as compared to the liver tropism of a corresponding wildtype AAV5 capsid.

16. The chimeric AAV5 capsid of any one of claims 13-15, wherein the AAV VP3 VR insertion from an AAV serotype that is different from AAV5 is a VP3 VR-I, VR-II, VR-III, VR-IV, VR-V, VR-VI, VR-VII, VR-VIII, and/or VR-IX. 17. The chimeric AAV5 capsid of claim 16, wherein the AAV VP3 VR insertion from an AAV serotype that is different from AAV5 is VP3 VR-I. 18. The chimeric AAV5 capsid of any one of claims 13-17, wherein the AAV VP3 VR insertion from an AAV serotype that is different from AAV5 is a VR of AAV1, AAV2, AAV3, AAV4, AAV5, AAV6, AAV7, AAV8, and/or AAV9. 19. The chimeric AAV5 capsid of claim 18, wherein the AAV VP3 VR insertion from an AAV serotype that is different from AAV5 is a VR of AAV6 or AAV8. 20. The chimeric AAV5 capsid of any one of claims 13-19, wherein the AAV VP3 VR insertion from an AAV serotype that is different from AAV5 comprises an amino acid sequence at least 90% identical to the amino acid sequence of any one of SEQ ID NOs:3-7. 21. The chimeric AAV5 capsid of claim 20, wherein the AAV VP3 VR insertion from an AAV serotype that is different from AAV5 comprises the amino acid sequence of any of SEQ ID NOs:3-7. 22. The chimeric AAV5 capsid of claim 20 or 21, comprising an amino acid sequence at least 90% identical to the amino acid sequence of SEQ ID NO:16. 23. The chimeric AAV5 capsid of claim 20 or 21, comprising an amino acid sequence at least 90% identical to the amino acid sequence of SEQ ID NO:18. 24. A chimeric adeno-associated virus 5 (AAV5) capsid comprising: a) a VP1 capsid protein of a first AAV serotype that is not AAV5 and a VP2 capsid protein of a second AAV serotype that is not AAV5 and that is different from said first AAV serotype or the same as said first AAV serotype; and b) an AAV5 VP3 capsid protein comprising an insertion following amino acid residue Q574 in the VP3 variable region (VR)-VIII region of an AAV VP3 VR from an AAV serotype that is different from AAV5, wherein the numbering corresponds to the amino acid sequence of SEQ ID NO:20. 25. The chimeric AAV5 capsid of claim 24, wherein the chimeric AAV5 capsid comprising the AAV5 VP3 capsid protein comprising said insertion following amino acid residue Q574 in the VP3 variable region (VR)-VIII region of an AAV VP3 VR from an AAV serotype that is different from AAV5 has enhanced liver tropism as compared to the liver tropism of a corresponding wildtype AAV5 capsid. 26. The chimeric AAV5 capsid of claim 25, wherein the enhanced liver tropism is enhanced about 5.0 fold or higher as compared to the liver tropism of a corresponding wildtype AAV5 capsid. 27. The chimeric AAV5 capsid of any one of claims 24-26, wherein the AAV VP3 VR insertion from an AAV serotype that is different from AAV5 is a VP3 VR-I, VR-II, VR-III, VR-IV, VR-V, VR-VI, VR-VII, VR-VIII, and/or VR-IX. 28. The chimeric AAV5 capsid of claim 27, wherein the AAV VP3 VR insertion from an AAV serotype that is different from AAV5 is a VP3 VR-I. 29. The chimeric AAV5 capsid of any one of claims 24-28, wherein the first AAV serotype and the second AAV serotype are different. 30. The chimeric AAV5 capsid of any one of claims 24-28, wherein the first AAV serotype and the second AAV serotype are the same. 31. The chimeric AAV5 capsid of any one of claims 24-30, wherein the serotype of the AAV VP3 VR insertion from an AAV serotype that is different from AAV5 is the same AAV serotype as the first and/or the second AAV serotype.

32. The chimeric AAV5 capsid of any one of claims 24-30, wherein the serotype of the AAV VP3 VR insertion from an AAV serotype that is different from AAV5 is different from the first and the second AAV serotype. 33. The chimeric AAV5 capsid of any one of claims 24-32, wherein the serotype of the first, second, and/or AAV VP3 VR insertion from an AAV serotype that is different from AAV5 is AAV1, AAV2, AAV3, AAV4, AAV6, AAV7, AAV8, and/or AAV9. 34. The chimeric AAV5 capsid of claim 33, wherein the first and second AAV serotypes are AAV9. 35. The chimeric AAV5 capsid of claim 33 or 34, wherein the serotype of the AAV VP3 VR insertion from an AAV serotype that is different from AAV5 is AAV6 or AAV8. 36. The chimeric AAV5 capsid of any one of claims 24-35, wherein the AAV VP3 VR insertion from an AAV serotype that is different from AAV5 comprises an amino acid sequence at least 90% identical to the amino acid sequence of any one of SEQ ID NOs:3-7. 37. The chimeric AAV5 capsid of claim 36, wherein the AAV VP3 VR insertion from an AAV serotype that is different from AAV5 comprises the amino acid sequence of any of SEQ ID NOs:3-7. 38. The chimeric AAV5 capsid of claim 36 or 37, comprising, consisting essentially of, or consisting of an amino acid sequence at least 90% identical to the amino acid sequence of SEQ ID NO:17. 39. The chimeric AAV5 capsid of claim 36 or 37, comprising, consisting essentially of, or consisting of an amino acid sequence at least 90% identical to the amino acid sequence of SEQ ID NO:19. 40. The chimeric AAV5 capsid of any one of claims 1-39, covalently linked, bound to, or encapsidating a compound selected from the group consisting of a DNA molecule, an RNA molecule, a polypeptide, a carbohydrate, a lipid, a small organic molecule, and any combination thereof.

41. An AAV particle comprising: the chimeric AAV5 capsid of any one of claims 1-40; and an AAV vector genome, wherein the AAV capsid encapsidates the AAV vector genome. 42. The AAV particle of claim 41, wherein the AAV vector genome comprises a heterologous nucleic acid molecule. 43. The AAV particle of claim 42, wherein the heterologous nucleic acid molecule encodes an antisense RNA, microRNA, or RNAi. 44. The AAV particle of claim 42, wherein the heterologous nucleic acid molecule encodes a polypeptide. 45. The AAV particle of claim 44, wherein the heterologous nucleic acid molecule encodes a therapeutic polypeptide. 46. A nucleic acid molecule encoding the chimeric AAV5 capsid of any one of claims 1- 39. 47. A vector comprising the nucleic acid molecule of claim 46. 48. The vector of claim 47, wherein the vector is a plasmid, phage, viral vector, bacterial artificial chromosome, or yeast artificial chromosome. 49. The vector of claim 48, wherein the viral vector is an AAV vector, an adenovirus vector, a herpesvirus vector, a lentivirus vector, an alphavirus vector or a baculovirus vector (e.g., an AAV particle, an adenovirus particle, a herpesvirus particle, a lentivirus particle, an alphavirus particle, a baculovirus particle, etc.). 50. The AAV vector of claim 49, wherein the nucleic acid molecule further comprises an AAV rep coding sequence.

51. A cell (e.g., an in vitro cell) comprising the chimeric AAV5 capsid of any one of claims 1-40, the particle of any one of claims 41-45, the nucleic acid molecule of claim 46 or the vector of any one of claims 47-50. 52. The cell of claim 51, wherein the nucleic acid molecule is stably incorporated into the genome of the cell. 53. A method of producing a recombinant AAV particle comprising an AAV capsid, the method comprising: providing/introducing into a cell in vitro with the nucleic acid molecule of claim 46, an AAV rep coding sequence, an AAV vector genome comprising a heterologous nucleic acid molecule, and helper functions for generating a productive AAV infection under conditions whereby assembly of the recombinant AAV particle comprising the AAV capsid and encapsidation of the AAV vector genome can occur. 54. An AAV5 particle produced by the method of claim 53. 55. A pharmaceutical formulation or composition comprising the AAV5 particle of any one of claims 41-45, the nucleic acid of claim 46, and/or the vector of any one of claims 47- 50 in a pharmaceutically acceptable carrier. 56. A method of delivering a nucleic acid molecule to a hepatocyte, the method comprising contacting the hepatocyte with the AAV5 particle of any one of claims 41-45 or 54, the nucleic acid molecule of claim 46, the vector of any one of claims 47-50, and/or the pharmaceutical formulation of claim 55. 57. A method of delivering a nucleic acid molecule to a hepatocyte in a mammalian subject, the method comprising: administering an effective amount of the AAV5 particle of any one of claims 41-45 or 54, the nucleic acid molecule of claim 46, the vector of any one of claims 47-50, and/or the pharmaceutical formulation of claim 55 to a mammalian subject, thereby delivering the nucleic acid molecule to a hepatocyte in the mammalian subject. 58. The method of claim 57, wherein the mammalian subject is a human subject.

59. The method of claim 57 or 58, wherein the AAV particle is delivered to the liver. 60. The method of claim 59, wherein the AAV particle is delivered to the liver by injection into the liver, injection into the portal vein, or any combination thereof. 61. A method of treating a disorder in a mammalian subject in need thereof, wherein the disorder is treatable by expressing a nucleic acid molecule encoding a therapeutic product in the liver of the subject, the method comprising administering a therapeutically effective amount of the AAV5 particle of any one of claims 41-45 or 54, the nucleic acid molecule of claim 46, the vector of any one of claims 47-50, and/or the pharmaceutical formulation of claim 55 to the mammalian subject under conditions whereby the nucleic acid molecule encoding the therapeutic product is expressed in the liver, thereby treating the disorder. 62. The method of claim 61, wherein the disorder is a blood clotting disorder (e.g., hemophilia A, hemophilia B, von Willebrand disease, Factor I deficiency, Factor II deficiency, Factor V deficiency, Factor VII deficiency, Factor X deficiency, Factor XI deficiency, Factor XII deficiency, and/or Factor XIII deficiency). 63. The method of claim 57 or 62, wherein the mammalian subject has previously received gene therapy treatment with an AAV particle of a serotype that is not AAV5 (e.g., a non-AAV5 particle). 64. The method of any one of claims 57-63, wherein the therapeutically effective amount of the AAV5 particle is between about 4×1012 vg (particles) /kg to about 6 x 1013 vg (particles) /kg. 65. The method of claim 64, wherein the therapeutically effective amount of the AAV5 particle is about 4 x 1012 vg (particles) /kg.

Description:
CHIMERIC AAV5 CAPSIDS STATEMENT OF PRIORITY This application claims the benefit, under 35 U.S.C. § 119(e), of U.S. Provisional Application No. 63/178,885, filed on April 23, 2021, the entire contents of which are incorporated by reference herein. STATEMENT REGARDING ELECTRONIC FILING OF A SEQUENCE LISTING A Sequence Listing in ASCII text format, submitted under 37 C.F.R. § 1.821, entitled 5470-894WO_ST25.txt, 145,681 bytes in size, generated on April 22, 2022 and filed via EFS- Web, is provided in lieu of a paper copy. This Sequence Listing is hereby incorporated herein by reference into the specification for its disclosures. FIELD OF THE INVENTION The invention relates to chimeric AAV5 capsids, virus vectors comprising the same, and methods of using the vectors such as to target the liver. The invention further relates to chimeric AAV5 capsids with improved infectivity to hepatocytes, virus vectors comprising the same, and methods of using the vectors to target hepatocytes with improved infectivity. BACKGROUND OF THE INVENTION Hemophilia A is a genetic disease with deficient activity of FVIII, which plays a critical role in the coagulation cascade by accelerating the conversion of factor X to factor Xa. Deficiency in FVIII activity increases bleeding time and is responsible for the bleeding disorder. In most cases hemophilia A is inherited as an X-linked recessive trait, and usually affects males, but there are cases which are caused by spontaneous mutations. The current treatment for hemophilia A is intravenous infusion of plasma-derived or recombinant FVIII protein. Despite this treatment being effective in controlling bleeding episodes, it is not a cure for hemophilia. Life-long frequent infusion is required because of the short half-life of FVIII (8-12 hours). Gene therapy has emerged as an attractive strategy for the eventual cure of this disease. AAV vectors have been applied in clinical trials as gene delivery tools to treat hemophilia diseases, but AAV exists in nature and infects human and induces human immune system to develop anti-AAV neutralizing antibodies (NAbs). The pre-existing NAbs are obstructions in the clinical application of AAV, because they could prevent AAV vector transduction and usually cause the failure of gene therapy. To pursue success of AAV vectors as a therapeutic tool, clinical trials must rule out many patients with pre-existing anti-AAV NAbs. The prevalence of NAbs against AAV serotypes has been reported as anti-AAV5 antibody at about 3.2%, and others were AAV1 (50.5%), AAV2 (59%), AAV6 (37%), AAV8 (19%) and AAV9 (33.5%). While AAV5 had the lowest preexisting NAbs in human body, AAV5 has poor infectivity, which caused the failure of one clinical trial. AAV5’s infectivity is over 10 times less than AAV6 and AAV8 in vivo, which limits its application in gene therapy of liver diseases. One strategy to obtain a significant clinical efficacy has been to use an increased dose of AAV5 up to 6x10 13 /kg, which is about 10 times over other AAV serotypes, which increased FVIII to a normal value while the consumption of injectable FVIII was reduced with low annualized bleeding rate. The present invention overcomes shortcomings in the art by providing chimeric AAV5 capsids with improved infectivity in vitro and in vivo, enhanced liver tropism, and improved treatment of hemophilia A at a reduced dosage. SUMMARY OF THE INVENTION One aspect of the present invention comprises a chimeric adeno-associated virus 5 (AAV5) capsid comprising: a) a VP1 capsid protein of a first AAV serotype that is not AAV5; and/or b) a VP2 capsid protein of a second AAV serotype that is not AAV5 and that is different from said first AAV serotype or the same as said first AAV serotype. Another aspect of the present invention comprises a chimeric adeno-associated virus 5 (AAV5) capsid comprising an AAV5 VP3 capsid protein comprising an insertion following amino acid residue Q574 in the VP3 variable region (VR)-VIII region of an AAV VP3 VR from an AAV serotype that is different from AAV5, wherein the numbering corresponds to the amino acid sequence of SEQ ID NO:20. Another aspect of the present invention comprises a chimeric adeno-associated virus 5 (AAV5) capsid comprising: a) a VP1 capsid protein of a first AAV serotype and a VP2 capsid protein of a second AAV serotype that is different from said first AAV serotype or the same as said first AAV serotype; and b) an AAV5 VP3 capsid protein comprising an insertion following amino acid residue Q574 in the VP3 variable region (VR)-VIII region of an AAV VP3 VR from an AAV serotype that is different from AAV5, wherein the numbering corresponds to the amino acid sequence of SEQ ID NO:20. In some embodiments, a chimeric AAV5 capsid of the present invention may have enhanced liver tropism as compared to wildtype AAV5 capsid. In some embodiments, a chimeric AAV5 capsid of the present invention may be covalently linked, bound to, or encapsidating a compound selected from the group consisting of a DNA molecule, an RNA molecule, a polypeptide, a carbohydrate, a lipid, a small organic molecule, and any combination thereof. Another aspect of the present invention provides an AAV particle comprising: a chimeric AAV5 capsid of the present invention; and an AAV vector genome; wherein the AAV capsid encapsidates the AAV vector genome. In another aspect, the present invention provides a nucleic acid molecule encoding a chimeric AAV5 capsid of the present invention. In another aspect, the present invention provides a vector comprising the nucleic acid molecule of this invention. In another aspect, the present invention provides a cell (e.g., an in vitro cell) comprising a chimeric particle, nucleic acid molecule, and/or vector of the present invention. Also provided are pharmaceutical formulations comprising an AAV particle, nucleic acid molecule, and/or vector of the present invention in a pharmaceutically acceptable carrier. Additionally provided is a method of producing a recombinant AAV particle comprising an AAV capsid, the method comprising: providing/introducing into a cell in vitro with a nucleic acid molecule of the present invention, an AAV rep coding sequence, an AAV vector genome comprising a heterologous nucleic acid molecule, and helper functions for generating a productive AAV infection under conditions whereby assembly of the recombinant AAV particle comprising the AAV capsid and encapsidation of the AAV vector genome can occur. Another aspect of the present invention provides a method of delivering a nucleic acid molecule of interest to a hepatocyte, the method comprising contacting the hepatocyte with an AAV particle, nucleic acid molecule, vector, and/or pharmaceutical formulation of the present invention. Another aspect of the present invention provides a method of delivering a nucleic acid molecule of interest to a hepatocyte in a mammalian subject, the method comprising: administering an effective amount of an AAV particle, nucleic acid molecule, vector, and/or pharmaceutical formulation to a mammalian subject, thereby delivering the nucleic acid molecule of interest to a hepatocyte in the mammalian subject. A further aspect of the present invention provides a method of treating a disorder in a mammalian subject in need thereof, wherein the disorder is treatable by expressing a nucleic acid molecule encoding a therapeutic product in the liver of the subject, the method comprising administering a therapeutically effective amount of an AAV particle, nucleic acid molecule, vector, and/or pharmaceutical formulation to the mammalian subject under conditions whereby the nucleic acid molecule encoding the therapeutic product is expressed in the liver, thereby treating the disorder. BRIEF DESCRIPTION OF THE DRAWINGS FIGS.1A-1I show schematics of the construction of the chimeric AAV5 vectors (AAV5n). VP1 and VP2 of AAV5 are replaced individually with VP1 and VP2 from AAV1, AAV2, AAV3, AAV4, AAV6, AAV7, AAV8 and AAV9. The VP3 of AAV5 is kept unchanged. FIG.2 shows western blot data for the VP1, VP2, VP3 proteins of AAV5n. The western blot shows AAV5n capsids (Cap) protein including VP1, VP2, VP3 proteins. VP2 of AAV57 and AAV58 includes two bands, to AAV7 and AAV8. The total dose of AAV vectors is 1×10 10 vg/channel. FIG.3A shows images of LacZ expression in HEK293 cells mediated by AAV5n vectors with CB promoter. LacZ expression mediated by AAV52, AAV57, AAV58 and AAV59 is enhanced significantly compared with AAV5. The left panel shows high MOI treatment (MOI=1×10 5 /cell, n=4), and the right panel shows low-MOI treatment (MOI=2×10 4 /cell, n=4). Scale bar: 400 μm (left panel), 200 μm (right panel). FIG.3B shows a bar graph of LacZ activity mediated by AAV5n viruses in 293 cell line. The activity of LacZ in HEK293 cells mediated by AAV5n vectors increased 4.5~7.7 times as compared to AAV5 treatment. ***P<0.001, vs. AAV5, n=4. FIG.4A shows images of LacZ expression mediated by AAV5n vectors with CB promoter in Huh-7 cells. LacZ expression mediated by AAV52, AAV57, AAV58 and AAV59 is enhanced significantly as compared to AAV5 treatment. The left panel shows high-MOI treatment (MOI=1×10 5 /cell, n=4), and the right panel shows low-MOI treatment (MOI=2×10 4 /cell, n=4). Scale bar: 400μm (left panel), 200 μm (right panel). FIG.4B shows a bar graph of LacZ activity mediated by AAV5n viruses in Huh-7 cell line. The activity of LacZ mediated by AAV5n vectors in Huh-7 cells increased 8.8~9.6 fold as compared to AAV5 treatment. *** P<0.001 vs. AAV5, n=4. FIG.5A shows images of LacZ expression mediated by AAV5 and the chimeric AAV5n vectors in the liver of C57/B6 mice (3×10 11 /mouse). No significant differences were found between the groups; Scale bar: 400 μm (upper panel), 200 μm (lower panel). FIG.5B shows a bar graph of LacZ activity mediated by AAV5 and the chimeric AAV5n vectors in the liver of C57/B6 mice (3×10 11 /mouse). The up-panel shows low power pictures, and the lower-panel shows high power pictures. There are no significant differences between groups. t-test, n=4. FIG.6A shows a schematic of the construction of AAV5 VP1, VP2 and VP3, and the chimeric vectors. Based on AAV5 construction, AAV59 is generated by replacing the VP1 and VP2 of AAV5 with the VP1 and VP2 of AAV9. AAV596 is inserted with the VR-I sequence of AAV6 at VR-VIII (Q574), based on AAV59. Similarly, AAV598 is inserted with the VR-I sequence of AAV8 at VR-VIII (Q574), based on AAV59. FIG.6B shows the partial VP3 sequences of AAV5 and related chimeric vectors. AAV5 and AAV59 have the same VP3 sequences. The partial amino acid sequences (amino acid positions 546-625) are shown (SEQ ID NO:2 (AAV5) and SEQ ID NO:15 (AAV59)). The site Q574 of AAV5 or Q578 of AAV59 can tolerate extra inserted sequences. AAV596 VP3 and AAV598 VP3 are generated via insertion at Q578 with the VR-I sequences of AAV6 and AAV8 (underlined), respectively. The sequence of amino acid positions 550-635 from AAV596 and amino acid positions 550-637 from AAV598 are shown in the figure (SEQ ID NO:17 (AAV596) and SEQ ID NO:19 (AAV598). FIG.7 shows schematics of the partial VP3 construction of AAV5 and related chimeric vectors including AAV59, AAV596 and AAV598. Three models are shown, including a line model (upper panel), a cartoon model (middle panel) and a surface model (lower panel). The VR-VIII of AAV5/AAV59 sticks out of AAV capsids. The insertion of the VR-I of AAV6/AAV8 into the VP3 changes the surface conformation of the VR-VIII region of AAV5. FIG.8 shows images of chimeric AAV vectors which could be successfully packaged. After the first ultra-speed spin with gradient CsCl, the empty (upper band) and full AAV (two lower bands), particles were separated into different bands. FIG.9A shows images of LacZ expression mediated by the chimeric AAV vectors in Huh-7 cells. Compared with original AAV5 vector, the chimeric AAV vectors increased LacZ gene expression in Huh-7 cells (MOI= 5x10 5 vg/cell). FIG.9B shows a bar graph of LacZ activity mediated by the chimeric AAV vectors in Huh-7 cells (MOI=5x10 5 vg/cell). Compared with original AAV5 vector, AAV59 with VP1 from AAV9 enhanced LacZ expression about 5 times; AAV596 and AAV598 inserted with VR-I from AAV6 or AAV8 increased LacZ activity about 7.9- or 20.6-fold vs. AAV5, respectively. ***P< 0.001 vs. AAV5, n=4. FIG.10 shows images of LacZ expression mediated by the chimeric AAV vectors in vivo. AAV viruses were injected into C57/B6 mice by tail vein (3x10 11 vg/mouse, n=4). The chimeric AAV vector AAV596 significantly increased liver tropism compared with original AAV5 vector and AAV59 vector, while there was no significant increase in heart and skeletal muscles GAS. Scale bar: 400 μm (upper panel), 200 μm (lower panel). FIG.11 shows a bar graph of LacZ activity mediated by the chimeric AAV vectors in different organs. AAV viruses were injected into C57/B6 mice by tail vein (3x10 11 vg/mouse). The chimeric AAV vector AAV596 significantly increased liver tropism 13.4 or 7.5-fold, compared with original AAV5 vector and AAV59 vector, respectively. There were no significant differences found between groups in heart, GAS, lung, intestine, kidney, spleen, and pancreas. Furthermore, the LacZ expression level was very low in these organs. **P< 0.01, vs. AAV5; ##P<0.01 vs AAV59; n=4, t-test. FIG.12 shows a bar graph of AAV596 and AAV598 vectors mediating high level gene expression of FVIII in plasma of a hemophilia-A mouse model. The chimeric AAV5 vectors packaged with FVIII were injected into the hemophilia-A mice by tail vein (4×10 12 vg/kg, n=5/group). After 2 week later, the FVIII activity of blood was measured by FVIII- specific ELISA. The results showed AAV596-mediated FVIII expression was significantly enhanced by 38.9- or 20.6-fold as compared to AAV5 or AAV59, respectively. AAV598- mediated FVIII expression was also enhanced by 10.8- or 5.7-fold compared with AAV5 or AAV59, respectively. *P< 0.01 vs. AAV5; ##P< 0.01 vs. AAV59. FIG.13 shows a bar graph of neutralizing antibody (Nab) titers of IVIG in response to chimeric AAV5 vectors and AAV9. Nab titers are IVIG dilutions that inhibited vector transduction by ^50% (red dotted line). AAV596 (1:80) and AAV598 (1:20) showed 8- and 32-fold greater resistance to neutralization than AAV9 (1:640). FIG.14 shows an alignment of capsid protein sequences of AAV1 (SEQ ID NO:21; GenBank® Accession No. AAD27757.1); AAV2 (SEQ ID NO:22; GenBank® Accession No. AAC03780.1), AAV3 (SEQ ID NO:23; GenBank® Accession No. NP_043941.1), AAV4 (SEQ ID NO:24; GenBank® Accession No. AAC58045.1), AAV5 (SEQ ID NO:20; GenBank® Accession No. YP_068409.1), AAV6 (SEQ ID NO:25; GenBank® Accession No. AAB95450.1), AAV7 (SEQ ID NO:26; GenBank® Accession No. YP_077178.1), AAV8 (SEQ ID NO:27; GenBank® Accession No. YP_077180.1), AAV9 (SEQ ID NO:28; GenBank® Accession No. AAS99264.1), and AAVrh10 (SEQ ID NO:29; GenBank® Accession No. AAO88201.1). DETAILED DESCRIPTION The present invention now will be described hereinafter with reference to the accompanying drawings and examples, in which embodiments of the invention are shown. This description is not intended to be a detailed catalog of all the different ways in which the invention may be implemented, or all the features that may be added to the instant invention. For example, features illustrated with respect to one embodiment may be incorporated into other embodiments, and features illustrated with respect to a particular embodiment may be deleted from that embodiment. Thus, the invention contemplates that in some embodiments of the invention, any feature or combination of features set forth herein can be excluded or omitted. In addition, numerous variations and additions to the various embodiments suggested herein will be apparent to those skilled in the art in light of the instant disclosure, which do not depart from the instant invention. Hence, the following descriptions are intended to illustrate some particular embodiments of the invention, and not to exhaustively specify all permutations, combinations, and variations thereof. 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 invention belongs. The terminology used in the description of the invention herein is for the purpose of describing particular embodiments only and is not intended to be limiting of the invention. All publications, patent applications, patents and other references cited herein are incorporated by reference in their entireties for the teachings relevant to the sentence and/or paragraph in which the reference is presented. Unless the context indicates otherwise, it is specifically intended that the various features of the invention described herein can be used in any combination. Moreover, the present invention also contemplates that in some embodiments of the invention, any feature or combination of features set forth herein can be excluded or omitted. To illustrate, if the specification states that a composition comprises components A, B and C, it is specifically intended that any of A, B or C, or a combination thereof, can be omitted and disclaimed singularly or in any combination. As used in the description of the invention 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. For example, "a" cell can mean a single cell or a multiplicity of cells. 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"). The term "about," as used herein when referring to a measurable value such as an amount or concentration and the like, is meant to encompass variations of ± 10%, ± 5%, ± 1%, ± 0.5%, or even ± 0.1% of the specified value as well as the specified value. For example, "about X" where X is the measurable value, is meant to include X as well as variations of ± 10%, ± 5%, ± 1%, ± 0.5%, or even ± 0.1% of X. A range provided herein for a measurable value may include any other range and/or individual value therein. As used herein, phrases such as "between X and Y" and "between about X and Y" should be interpreted to include X and Y. As used herein, phrases such as "between about X and Y" mean "between about X and about Y" and phrases such as "from about X to Y" mean "from about X to about Y." The term "comprise," "comprises" and "comprising" as used herein, specify the presence of the stated features, integers, steps, operations, elements, and/or components, but do not preclude the presence or addition of one or more other features, integers, steps, operations, elements, components, and/or groups thereof. As used herein, the transitional phrase "consisting essentially of" means that the scope of a claim is to be interpreted to encompass the specified materials or steps recited in the claim and those that do not materially affect the basic and novel characteristic(s) of the claimed invention. See, In re Herz, 537 F.2d 549, 551-52, 190 USPQ 461, 463 (CCPA 1976) (emphasis in the original); see also MPEP § 2111.03. Thus, the term "consisting essentially of" when used in a claim of this invention is not intended to be interpreted to be equivalent to "comprising."   Except as otherwise indicated, standard methods known to those skilled in the art may be used for cloning genes, amplifying and detecting nucleic acids, and the like. Such techniques are known to those skilled in the art. See, e.g., Sambrook et al., Molecular Cloning: A Laboratory Manual 2nd Ed. (Cold Spring Harbor, NY, 1989); Ausubel et al. Current Protocols in Molecular Biology (Green Publishing Associates, Inc. and John Wiley & Sons, Inc., New York). Nucleotide sequences are presented herein by single strand only, in the 5' to 3' direction, from left to right, unless specifically indicated otherwise. Nucleotides and amino acids are represented herein in the manner recommended by the IUPAC-IUB Biochemical Nomenclature Commission, or (for amino acids) by either the one-letter code, or the three letter code, both in accordance with 37 C.F.R. §1.822 and established usage. 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 sub combination is expressly set forth herein. Moreover, such language also indicates that one or more of the specified amino acids can be disclaimed (e.g., by negative proviso). For example, in particular 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. The designation of all amino acid positions in the AAV capsid proteins in the AAV vectors and recombinant AAV nucleic acid molecules of the invention is with respect to VP1, VP2, and/or VP3 capsid subunit numbering identified as SEQ ID NO:1 (AAV5 VP1+VP2) and SEQ ID NO:2 (AAV5 VP3), and/or wildtype AAV5 VP1, VP2, and VP3 capsid protein GenBank Accession No. YP_068409.1 (SEQ ID NO:20). It will be understood by those skilled in the art that modifications as 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). As used herein, the terms "reduce," "reduces," "reduction," "diminish," "inhibit" and similar terms mean a decrease of at least about 5%, 10%, 15%, 20%, 25%, 35%, 50%, 75%, 80%, 85%, 90%, 95%, 97% or more. As used herein, the terms "enhance," "enhances," "enhancement" and similar terms indicate an increase of at least about 25%, 50%, 75%, 100%, 150%, 200%, 300%, 400%, 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 Parvovirus, Erythrovirus, Densovirus, Iteravirus, and Contravirus. 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). 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, avian AAV, bovine AAV, canine AAV, equine AAV, ovine AAV, 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 additional AAV serotypes and clades have been identified (see, e.g., Gao et al., (2004) J. Virology 78:6381-6388; Mori et al., (2004) Virology 33-:375-383; and Table 1). 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. These sequences include known amino acid sequences of the serotype capsid proteins, including but not limited to, AAD27757.1 (AAV1), YP_068409.1 (AAV5), AAC03780.1 (AAV2), AAC58045.1 (AAV4), NP_043941.1 (AAV3), AAB95450.1 (AAV6), YP_077178.1 (AAV7), YP_077180.1 (AAV8), AAS99264.1 (AAV9), and AAO88201.1 (AAVrh10). See in addition, 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. Patent 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 1. 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); AAV4 (Padron et al. (2005) J. Virol.79: 5047- 58); AAV5 (Walters et al. (2004) J. Virol.78:3361-71); and CPV (Xie et al. (1996) J. Mol. Biol.6:497-520 and Tsao et al. (1991) Science 251:1456-64). 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 heterologous nucleic acid(s) of interest. As used herein, "transduction" of a cell by a virus vector (e.g., an AAV vector) means entry of the vector into the cell and transfer of genetic material into the cell by the incorporation of nucleic acid into the virus vector and subsequent transfer into the cell via the virus vector. Unless indicated otherwise, "efficient transduction" or "efficient tropism," or similar terms, can be determined by reference to a suitable positive or negative control (e.g., at least about 50%, 60%, 70%, 80%, 85%, 90%, 95% or more of the transduction or tropism, respectively, of a positive control or at least about 110%, 120%, 150%, 200%, 300%, 500%, 1000% or more of the transduction or tropism, respectively, of a negative control). 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 particular embodiments, the virus vector does not efficiently transduce (i.e., does not have efficient tropism for) tissues outside the liver, e.g., CNS, kidney, gonads and/or germ cells. In particular embodiments, undesirable transduction of tissue(s) is 20% or less, 10% or less, 5% or less, 1% or less, 0.1% or less of the level of transduction of the desired target tissue(s). As used herein, the term "polypeptide" encompasses both peptides and proteins, unless indicated otherwise. 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 nucleotides), but in representative embodiments are either single or double stranded DNA sequences. 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 representative embodiments an "isolated" nucleotide is enriched by at least about 10-fold, 100-fold, 1000-fold, 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 representative embodiments an "isolated" polypeptide is enriched by at least about 10-fold, 100-fold, 1000-fold, 10,000-fold or more as compared with the starting material. A "nucleic acid," "nucleic acid molecule," or "nucleotide sequence" 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), but is preferably either single or double stranded DNA sequences. As used herein, an "isolated" nucleic acid or nucleotide sequence (e.g., an "isolated DNA" or an "isolated RNA") means a nucleic acid or nucleotide sequence separated or substantially free 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 nucleic acid or nucleotide sequence. Likewise, an "isolated" polypeptide means a polypeptide that is separated or substantially free 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. An "isolated cell" refers to a cell that is separated from other components with which it is normally associated in its natural state. For example, an isolated cell can be a cell in culture medium and/or a cell in a pharmaceutically acceptable carrier of this invention. Thus, an isolated cell can be delivered to and/or introduced into a subject. In some embodiments, an isolated cell can be a cell that is removed from a subject and manipulated as described herein ex vivo and then returned to the subject. As used herein, by "isolate" or "purify" (or grammatical equivalents) a virus vector or virus particle or population of virus particles, it is meant that the virus vector or virus particle or population of virus particles is at least partially separated from at least some of the other components in the starting material. In representative embodiments an "isolated" or "purified" virus vector or virus particle or population of virus particles is enriched by at least about 10-fold, 100-fold, 1000-fold, 10,000-fold or more as compared with the starting material. The term "endogenous" refers to a component naturally found in an environment, i.e., a gene, nucleic acid, miRNA, protein, cell, or other natural component expressed in the subject, as distinguished from an introduced component, i.e., an "exogenous" component. As used herein, the term "heterologous" refers to a nucleotide/polypeptide that originates from a foreign species, or, if from the same species, is substantially modified from its native form in composition and/or genomic locus by deliberate human intervention. A "heterologous nucleotide sequence" or "heterologous nucleic acid" is a sequence that is not naturally occurring in the virus. Generally, the heterologous nucleic acid or nucleotide sequence comprises an open reading frame that encodes a polypeptide and/or a nontranslated RNA. A "therapeutic polypeptide" is a polypeptide 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 or induction of an immune response. 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. By "substantially retain" a property and/or to maintain a property "substantially the same" as a comparison (e.g., a control), it is meant that at least about 75%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% of the property (e.g., activity or other measurable characteristic) is retained. 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 methods of the invention. 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 are substantially less than what would occur in the absence of the present invention. A "treatment effective" or "effective" amount as used herein is an amount that is sufficient to provide some improvement or benefit to the subject. Alternatively stated, a "treatment effective" or "effective" amount is an amount that will provide some alleviation, mitigation, decrease or stabilization in at least one clinical symptom in the subject. Those skilled in the art will appreciate that the therapeutic effects need not be complete or curative, as long as some benefit is provided to the subject. A "prevention effective" amount as used herein is an amount that is sufficient to prevent and/or delay the onset of a disease, disorder and/or clinical symptoms in a subject and/or to reduce and/or delay the severity of the onset of a disease, disorder and/or clinical symptoms in a subject relative to what would occur in the absence of the methods of the invention. Those skilled in the art will appreciate that the level of prevention need not be complete, as long as some preventative benefit is provided to the subject. As used herein the term "bleeding disorder" reflects any defect, congenital, acquired or induced, of cellular, physiological, or molecular origin that is manifested in bleedings. Examples are clotting factor deficiencies (e.g., hemophilia A and B or deficiency of coagulation Factors XI or VII), clotting factor inhibitors, defective platelet function, thrombocytopenia, von Willebrand's disease, or bleeding induced by surgery or trauma. As used therein the term "excessive bleedings" refers to bleeding that occurs in subjects with a normally functioning blood clotting cascade (no clotting factor deficiencies or inhibitors against any of the coagulation factors) and may be caused by a defective platelet function, thrombocytopenia or von Willebrand's disease. In such cases, the bleedings may be likened to those bleedings caused by hemophilia because the hemostatic system, as in hemophilia, lacks or has abnormal essential clotting "compounds" (such as platelets or von Willebrand factor protein), causing major bleedings. In subjects who experience extensive tissue damage in association with surgery or trauma, the normal hemostatic mechanism may be overwhelmed by the demand of immediate hemostasis and they may develop bleeding in spite of a normal hemostatic mechanism. Achieving satisfactory hemostasis also is a problem when bleedings occur in organs such as the brain, inner ear region and eyes, with limited possibility for surgical hemostasis. The same problem may arise in the process of taking biopsies from various organs (liver, lung, tumor tissue, gastrointestinal tract) as well as in laparoscopic surgery. Common for all these situations is the difficulty to provide hemostasis by surgical techniques (sutures, clips, etc.), which also is the case when bleeding is diffuse (hemorrhagic gastritis and profuse uterine bleeding). Acute and profuse bleedings may also occur in subjects on anticoagulant therapy in whom a defective hemostasis has been induced by the therapy given. Such subjects may need surgical interventions in case the anticoagulant effect has to be counteracted rapidly. Radical retropubic prostatectomy is a commonly performed procedure for subjects with localized prostate cancer. The operation is frequently complicated by significant and sometimes massive blood loss. The considerable blood loss during prostatectomy is mainly related to the complicated anatomical situation, with various densely vascularized sites that are not easily accessible for surgical hemostasis, and which may result in diffuse bleeding from a large area. Also, intracerebral hemorrhage is the least treatable form of stroke and is associated with high mortality and hematoma growth in the first few hours following intracerebral hemorrhage. Another situation that may cause problems in the case of unsatisfactory hemostasis is when subjects with a normal hemostatic mechanism are given anticoagulant therapy to prevent thromboembolic disease. Such therapy may include heparin, other forms of proteoglycans, warfarin or other forms of vitamin K-antagonists as well as aspirin and other platelet aggregation inhibitors. The term "bleeding episode" is meant to include uncontrolled and excessive bleeding. Bleeding episodes may be a major problem both in connection with surgery and other forms of tissue damage. Uncontrolled and excessive bleeding may occur in subjects having a normal coagulation system and subjects having coagulation or bleeding disorders. The terms "nucleotide sequence of interest (NOI)," "heterologous nucleotide sequence" and "heterologous nucleic acid molecule" are used interchangeably herein and refer to a nucleic acid sequence that is not naturally occurring (e.g., engineered). Generally, the NOI, heterologous nucleic acid molecule or heterologous nucleotide sequence comprises an open reading frame that encodes a polypeptide and/or nontranslated RNA of interest (e.g., for delivery to a cell and/or subject). 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 a viral genome (e.g., viral DNA [vDNA]) and/or replicon nucleic acid molecule packaged within a virus particle. Alternatively, in some contexts, the term "vector" may be used to refer to the vector genome/vDNA alone. The term "vector," as used herein, means any nucleic acid entity capable of amplification in a host cell. Thus, the vector may be an autonomously replicating vector, i.e., a vector, which exists as an extrachromosomal entity, the replication of which is independent of chromosomal replication, e.g., a plasmid. Alternatively, the vector may be one which, when introduced into a host cell, is integrated into the host cell genome and replicated together with the chromosome(s) into which it has been integrated. The choice of vector will often depend on the host cell into which it is to be introduced. Vectors include, but are not limited to plasmid vectors, phage vectors, viruses or cosmid vectors. Vectors usually contain a replication origin and at least one selectable gene, i.e., a gene which encodes a product which is readily detectable or the presence of which is essential for cell growth A "rAAV vector genome" or "rAAV genome" is an AAV genome (i.e., vDNA) that comprises at least one terminal repeat (e.g., two terminal repeats) and one or more heterologous nucleotide sequences. rAAV vectors generally require only the 145 base terminal repeat(s) (TR(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 minimal TR sequence(s) 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). The rAAV vector genome optionally comprises two AAV TRs, which generally will be at the 5’ and 3’ ends of the heterologous nucleotide sequence(s), but need not be contiguous thereto. The TRs can be the same or different from each other. A "rAAV particle" comprises a rAAV vector genome packaged within an AAV capsid. The term "terminal repeat" or "TR" or "inverted terminal repeat (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 TR can be an AAV TR or a non-AAV TR. For example, a non-AAV TR 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 a TR, which can further be modified by truncation, substitution, deletion, insertion and/or addition. Further, the TR can be partially or completely synthetic, such as the "double-D sequence" as described in United States Patent No. 5,478,745 to Samulski et al., which is hereby incorporated by reference in its entirety. An "AAV terminal repeat" or "AAV TR" 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 1). An AAV terminal repeat need not have the native terminal repeat sequence (e.g., a native AAV TR 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. AAV proteins VP1, VP2 and VP3 are capsid proteins that interact together to form an AAV capsid of an icosahedral symmetry. VP1.5 is an AAV capsid protein described in US Publication No. 2014/0037585, which is hereby incorporated by reference in its entirety The virus vectors of the invention can further be "targeted" virus vectors (e.g., having a directed tropism) and/or a "hybrid" parvovirus (i.e., in which the viral TRs and viral capsid are from different parvoviruses) as described in international patent publication WO 00/28004 and Chao et al., (2000) Molecular Therapy 2:619, which is hereby incorporated by reference in its entirety. The virus vectors of the invention can further be duplexed parvovirus 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 of the invention. Further, the viral capsid or genomic elements can contain other modifications, including insertions, deletions and/or substitutions. A "chimeric" capsid protein and/or "chimeric" or "modified" capsid as used herein means an AAV capsid protein or capsid that has been modified by substitutions in one or more (e.g., 2, 3, 4, 5, 6, 7, 8, 9, 10, etc.) amino acid residues in the amino acid sequence of a capsid protein relative to wild type, as well as insertions and/or deletions of one or more (e.g., 2, 3, 4, 5, 6, 7, 8, 9, 10, etc.) amino acid residues in the amino acid sequence relative to wild type. In some embodiments, complete or partial domains, functional regions, epitopes, etc., from one AAV serotype can replace the corresponding wildtype domain, functional region, epitope, etc. of a different AAV serotype, in any combination, to produce a chimeric capsid protein or modified capsid of this invention. Production of a chimeric capsid protein or modified capsid can be carried out according to protocols well known in the art and a large number of chimeric capsid proteins are described in the literature as well as herein that can be included in the capsid of this invention. As used herein, the term "amino acid" or "amino acid residue" encompasses any naturally occurring amino acid, modified forms thereof, and synthetic amino acids. Naturally occurring, levorotatory (L-) amino acids are shown in Table 2. Conservative amino acid substitutions are known in the art. In particular embodiments, a conservative amino acid substitution includes substitutions within one or more of the following groups: glycine, alanine; valine, isoleucine, leucine; aspartic acid, glutamic acid; asparagine, glutamine; serine, threonine; lysine, arginine; and/or phenylalanine, tyrosine. Alternatively, the amino acid can be a modified amino acid residue (nonlimiting examples are shown in Table 3) and/or can be an amino acid that is modified by post- translation modification (e.g., acetylation, amidation, formylation, hydroxylation, methylation, phosphorylation or sulfatation). 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. Compositions of the invention The invention, in part, relates to compositions and methods of using a chimeric AAV5 capsid in the treatment of diseases such as bleeding disorders. Bleeding disorders are a group of conditions that result when the blood cannot clot properly. Such a condition may be genetic (i.e., inherited from a family member) or acquired (e.g., autoimmune disorders; drug treatment, etc.). In normal clotting (also known as coagulation), platelets, a type of blood cell, stick together and form a plug at the site of an injured blood vessel. Proteins in the blood called clotting factors then interact to form a fibrin clot, essentially a gel plug, which holds the platelets in place and allows healing to occur at the site of the injury while preventing blood from escaping the blood vessel. Typically, in bleeding disorders a deficiency of at least one clotting factor required for clotting is present. For example, deficiencies in clotting factor(s) II, V, VII, X, XI, or XII result in bleeding disorders and/or abnormal bleeding problems. Hemophilia is another example of a bleeding disorder and is classified as type A or type B, based on which type of clotting factor is deficient (factor VIII in type A and factor IX in type B). Possible treatment options for subjects suffering from bleeding disorders, such as Hemophilia A (HA) and Hemophilia B (HB) is protein replacement therapy. Clotting factors are replaced by injecting (infusing) a clotting factor concentrate into a vein to help blood to clot normally. For example, clotting factor VIIa has been used to control bleeding disorders by stimulating the coagulation cascade in a subject. In some embodiments, the subject has a normal functioning clotting cascade (i.e., no clotting factor deficiencies) and requires control of excessive bleeding caused by defective platelet function, thrombocytopenia, von Willebrand disease, surgery, and other forms of trauma. Another option may be delivery of FVa, which is a cofactor that binds to FXa during the formation of the prothrombinase complex, which activates prothrombin to thrombin. FVa is able to enhance the rate of thrombin generation by approximately 10,000 fold. Thrombin plays an important role in the coagulation cascade, e.g., it promotes platelet activation and aggregation and it converts FXI to FXIa, VIII to VIIIa, V to Va, fibrinogen to fibrin, and XIII to XIIIa. A clotting factor (e.g., FVa, FVIIa, FVIII, or any variant and/or derivative thereof) alone or in combination with other clotting factors may be administered to a subject in need thereof using any known method in the art, e.g., using a viral vector such as adeno-associated virus (AAV), retrovirus, lentivirus, poxvirus, alphavirus, baculovirus, vaccinia virus, herpes virus, and Epstein-Barr virus. AAV is a small (25-nm), nonenveloped virus that packages a linear single-stranded DNA genome. AAV can infect both dividing and quiescent cells and persist in an extrachromosomal state without integrating into the genome of the host cell, although in the native virus some integration of virally carried genes into the host genome does occur. The present invention relates to the design of a chimeric adeno-associated virus (AAV) capsid with improved infectivity in vitro and in vivo, enhanced tissue tropism, and improved treatment of disease at a reduced dosage as compared to established AAV gene therapy. In particular, the present invention provides a chimeric AAV5 capsid with improved infectivity in vitro and in vivo, enhanced liver tropism, and improved treatment of hemophilia A at a reduced dosage. Thus, one aspect of the present invention provides a chimeric adeno-associated virus 5 (AAV5) capsid comprising one or more of the following: a) a VP1 capsid protein of a first AAV serotype; b) a VP2 capsid protein of a second AAV serotype that is different from said first AAV serotype or the same as said first AAV serotype; and/or c) an AAV5 VP3 capsid protein comprising an insertion in the VP3 variable region (VR)-VIII region of an exogenous AAV VP3 VR. In some embodiments, the exogenous AAV VP3 VR insertion may be from an AAV serotype that is different from AAV5. In some embodiments, the present invention comprises a chimeric adeno-associated virus 5 (AAV5) capsid comprising: a) a VP1 capsid protein of a first AAV serotype that is not AAV5; and/or b) a VP2 capsid protein of a second AAV serotype that is not AAV5 and that is different from said first AAV serotype or the same as said first AAV serotype. In some embodiments, the present invention comprises a chimeric adeno-associated virus 5 (AAV5) capsid comprising an AAV5 VP3 capsid protein comprising an insertion following amino acid residue Q574 in the VP3 variable region (VR)-VIII region of an AAV VP3 VR from a serotype that is different from AAV5, wherein the numbering corresponds to the amino acid sequence of SEQ ID NO:20. In some embodiments, the present invention comprises a chimeric adeno-associated virus 5 (AAV5) capsid comprising: a) a VP1 capsid protein of a first AAV serotype and a VP2 capsid protein of a second AAV serotype that is different from said first AAV serotype or the same as said first AAV serotype; and b) an AAV5 VP3 capsid protein comprising an insertion following amino acid residue Q574 in the VP3 variable region (VR)-VIII region of an exogenous AAV VP3 VR (e.g., an AAV VP3 VR from a serotype that is different from AAV5), wherein the numbering corresponds to the amino acid sequence of SEQ ID NO:20. The insertion may be of any length which, when inserted into the VP3 VR-VIII region of an AAV5 VP3 capsid protein, retains functionality of said VP3. In some embodiments, the insertion (i.e., the inserted exogenous AAV VP3 VR) may of a length of about 1 to about 35 amino acid residues, e.g., a length of about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, or 35 amino acid residues or more, or any value or range therein. For example, in some embodiments, the insertion may be of about 1 to about 14 amino acid residues, about 10 to about 35 amino acid residues, about 5 to about 32 amino acid residues, about 14 to about 35 amino acid residues, or about 10 amino acid residues, about 14 amino acid residues, or about 32 amino acid residues. In some embodiments, the insertion in the VP3 VR-VIII region may be following amino acid residue Q574, wherein the numbering corresponds to the amino acid sequence of SEQ ID NO:20 (NCBI Accession No. Y18065.1; SEQ ID NO:1 + SEQ ID NO:2). In some embodiments, the insertion in the VP3 VR-VIII region may be immediately following amino acid residue Q574, i.e., between residue 574 and 575, wherein the numbering corresponds to the amino acid sequence of SEQ ID NO:20 (NCBI Accession No. Y18065.1; SEQ ID NO:1 + SEQ ID NO:2). In some embodiments, the insertion in the VP3 VR-VIII region is downstream of amino acid residue Q574, i.e., following position 574, 575, or 576, etc., wherein the numbering corresponds to the amino acid sequence of SEQ ID NO:20 (NCBI Accession No. Y18065.1; SEQ ID NO:1 + SEQ ID NO:2). SEQ ID NO:1 WT AAV5 VP1/VP2 (192AA) MSFVDHPPDWLEEVGEGLREFLGLEAGPPKPKPNQQHQDQARGLVLPGYNYLGPGN GLDRGEPVNRADEVAREHDISYNEQLEAGDNPYLKYNHADAEFQEKLADDTSFGGN LGKAVFQAKKRVLEPFGLVEEGAKTAPTGKRIDDHFPKRKKARTEEDSKPSTSSDAEA GPSGSQQLQIPAQPASSLGADT SEQ ID NO:2 WT AAV5 VP3 (532AAs; Q574 underlined) MSAGGGGPLGDNNQGADGVGNASGDWHCDSTWMGDRVVTKSTRTWVLPSYNNH QYREIKSGSVDGSNANAYFGYSTPWGYFDFNRFHSHWSPRDWQRLINNYWGFRPRS LRVKIFNIQVKEVTVQDSTTTIANNLTSTVQVFTDDDYQLPYVVGNGTEGCLPAFPPQ VFTLPQYGYATLNRDNTENPTERSSFFCLEYFPSKMLRTGNNFEFTYNFEEVPFHSSFA PSQNLFKLANPLVDQYLYRFVSTNNTGGVQFNKNLAGRYANTYKNWFPGPMGRTQG WNLGSGVNRASVSAFATTNRMELEGASYQVPPQPNGMTNNLQGSNTYALENTMIFN SQPANPGTTATYLEGNMLITSESETQPVNRVAYNVGGQMATNNQSSTTAPATGTYNLQ EIVPGSVWMERDVYLQGPIWAKIPETGAHFHPSPAMGGFGLKHPPPMMLIKNTPVPG NITSFSDVPVSSFITQYSTGQVTVEMEWELKKENSKRWNPEIQYTNNYNDPQFVDFAP DSTGEYRTTRPIGTRYLTRPL SEQ ID NO:20. [WT AAV5 capsid protein (VP1, VP2 (SEQ ID NO:1) and VP3 (SEQ ID NO:2)) Accession No. YP_068409.1; 574Q underlined] MSFVDHPPDWLEEVGEGLREFLGLEAGPPKPKPNQQHQDQARGLVLPGYNYLGPGN GLDRGEPVNRADEVAREHDISYNEQLEAGDNPYLKYNHADAEFQEKLADDTSFGGN LGKAVFQAKKRVLEPFGLVEEGAKTAPTGKRIDDHFPKRKKARTEEDSKPSTSSDAE AGPSGSQQLQIPAQPASSLGADTMSAGGGGPLGDNNQGADGVGNASGDWHCDSTW MGDRVVTKSTRTWVLPSYNNHQYREIKSGSVDGSNANAYFGYSTPWGYFDFNRFH SHWSPRDWQRLINNYWGFRPRSLRVKIFNIQVKEVTVQDSTTTIANNLTSTVQVFTD DDYQLPYVVGNGTEGCLPAFPPQVFTLPQYGYATLNRDNTENPTERSSFFCLEYFPS KMLRTGNNFEFTYNFEEVPFHSSFAPSQNLFKLANPLVDQYLYRFVSTNNTGGVQFN KNLAGRYANTYKNWFPGPMGRTQGWNLGSGVNRASVSAFATTNRMELEGASYQV PPQPNGMTNNLQGSNTYALENTMIFNSQPANPGTTATYLEGNMLITSESETQPVNRV AYNVGGQMATNNQSSTTAPATGTYNLQEIVPGSVWMERDVYLQGPIWAKIPETGA HFHPSPAMGGFGLKHPPPMMLIKNTPVPGNITSFSDVPVSSFITQYSTGQVTVEMEWE LKKENSKRWNPEIQYTNNYNDPQFVDFAPDSTGEYRTTRPIGTRYLTRPL The insertion may be from any VP3 variable region. In some embodiments, the inserted VP3 VR is a VP3 VR-I, VR-II, VR-III, VR-IV, VR-V, VR-VI, VR-VII, VR-VIII, and/or VR-IX. In some embodiments, inserted VP3 VR may be VP3 VR-I. In some embodiments, the first AAV serotype and the second AAV serotype are the same. In some embodiments, the first AAV serotype and the second AAV serotype are different. In some embodiments, the AAV serotype of the exogenous inserted AAV VP3 VR may be the same AAV serotype as the first and/or the second AAV serotype. In some embodiments, the AAV serotype of the exogenous inserted AAV VP3 VR may be different from the first and the second AAV serotype. In some embodiments, the serotype of the first serotype, second serotype and/or the AAV serotype of the exogenous inserted AAV VP3 VR may be AAV1, AAV2, AAV3, AAV4, AAV6, AAV7, AAV8, and/or AAV9. In some embodiments, the serotype of the first serotype, second serotype and/or the AAV serotype of the exogenous inserted AAV VP3 VR may be AAV2, AAV6, AAV7, AAV8, and/or AAV9. For example, in some embodiments, the first and second AAV serotypes may be AAV9. In some embodiments, the AAV serotype of the exogenous inserted AAV VP3 VR may be AAV6. In some embodiments, the AAV serotype of the exogenous inserted AAV VP3 VR may be AAV8. In some embodiments, a chimeric AAV5 capsid of the present invention comprising the AAV5 VP3 capsid protein comprising an insertion following amino acid residue Q574 in the VP3 variable region (VR)-VIII region of an exogenous AAV VP3 VR (e.g., from an AAV serotype that is different from AAV5) may have enhanced liver tropism as compared to the liver tropism of a corresponding wildtype AAV5 capsid. The liver tropism may be enhanced about 5.0 fold or higher (e.g., about 5, 5.5, 6, 6.5, 7, 7.5, 8, 8.5, 9, 9.5, 10, 10.5, 11, 11.5, 12, 12.5, 13, 13.5, 14, 14.5, 15, 15.5, 16, 16.5, 17, 17.5, 18, 18.5, 19, 19.5, or 20-fold or higher, or any value or range therein) as compared to the liver tropism of a corresponding wildtype AAV5 capsid. For example, in some embodiments, the liver tropism of the chimeric AAV5 capsid of the present invention may be about 5.0 fold to about 20 fold, about 10 fold to about 19.5 fold, about 7 fold to about 25 fold, about 5.5 fold to about 18 fold, or about 5.0 fold, about 7.0 fold, about 9.3 fold, about 18 fold, about 20 fold, or about 25 fold or higher. In some embodiments, a VP3 VR insertion of the present invention may comprise, consist essentially of, or consist of the amino acid sequence SEQ ID NO:3, or a sequence at least about 70% identical thereto (e.g., at least about 70, 75, 80, 85, 90, 91, 92, 93, 94, 95, 96, 97, 98, or 99% identical thereto). SEQ ID NO:3 AAV2 VR-I SQSGAS In some embodiments, a VP3 VR insertion of the present invention may comprise, consist essentially of, or consist of the amino acid sequence SEQ ID NO:4, or a sequence at least about 70% identical thereto (e.g., at least about 70, 75, 80, 85, 90, 91, 92, 93, 94, 95, 96, 97, 98, or 99% identical thereto). SEQ ID NO:4 AAV6 VR-I SASTGA In some embodiments, a VP3 VR insertion of the present invention may comprise, consist essentially of, or consist of the amino acid sequence SEQ ID NO:5, or a sequence at least about 70% identical thereto (e.g., at least about 70, 75, 80, 85, 90, 91, 92, 93, 94, 95, 96, 97, 98, or 99% identical thereto). SEQ ID NO:5 AAV7 VR-I SETAGST In some embodiments, a VP3 VR insertion of the present invention may comprise, consist essentially of, or consist of the amino acid sequence SEQ ID NO:6, or a sequence at least about 70% identical thereto (e.g., at least about 70, 75, 80, 85, 90, 91, 92, 93, 94, 95, 96, 97, 98, or 99% identical thereto). SEQ ID NO:6 AAV8 VR-I NGTSGGAT In some embodiments, a VP3 VR insertion of the present invention may comprise, consist essentially of, or consist of the amino acid sequence SEQ ID NO:7, or a sequence at least about 70% identical thereto (e.g., at least about 70, 75, 80, 85, 90, 91, 92, 93, 94, 95, 96, 97, 98, or 99% identical thereto). SEQ ID NO:7 AAV9 VR-I NSTSGGSS In some embodiments, a chimeric AAV5 capsid of the present invention may comprise, consist essentially of, or consist of an amino acid sequence at least 90% identical (e.g., at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical) to the amino acid sequence of SEQ ID NO:8 [AAV51 VP1/2 and VP3]. SEQ ID NO:8 [AAV51 VP1/2 and VP3] MAADGYLPDWLEDNLSEGIREWWDLKPGAPKPKANQQKQDDGRGLVLPGYKYLG PFNGLDKGEPVNAADAAALEHDKAYDQQLKAGDNPYLRYNHADAEFQERLQEDTS FGGNLGRAVFQAKKRVLEPLGLVEEGAKTAPGKKRPVEQSPQEPDSSSGIGKTGQQP AKKRLNFGQTGDSESVPDPQPLGEPPATPAAVGPTTMSAGGGGPLGDNNQGADGVG NASGDWHCDSTWMGDRVVTKSTRTWVLPSYNNHQYREIKSGSVDGSNANAYFGY STPWGYFDFNRFHSHWSPRDWQRLINNYWGFRPRSLRVKIFNIQVKEVTVQDSTTTI ANNLTSTVQVFTDDDYQLPYVVGNGTEGCLPAFPPQVFTLPQYGYATLNRDNTENP TERSSFFCLEYFPSKMLRTGNNFEFTYNFEEVPFHSSFAPSQNLFKLANPLVDQYLYR FVSTNNTGGVQFNKNLAGRYANTYKNWFPGPMGRTQGWNLGSGVNRASVSAFAT TNRMELEGASYQVPPQPNGMTNNLQGSNTYALENTMIFNSQPANPGTTATYLEGNM LITSESETQPVNRVAYNVGGQMATNNQSSTTAPATGTYNLQEIVPGSVWMERDVYL QGPIWAKIPETGAHFHPSPAMGGFGLKHPPPMMLIKNTPVPGNITSFSDVPVSSFITQY STGQVTVEMEWELKKENSKRWNPEIQYTNNYNDPQFVDFAPDSTGEYRTTRPIGTR YLTRPL In some embodiments, a chimeric AAV5 capsid of the present invention may comprise, consist essentially of, or consist of an amino acid sequence at least 90% identical (e.g., at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical) to the amino acid sequence of SEQ ID NO:9 [AAV52 VP1/2 and VP3]: SEQ ID NO:9 [AAV52 VP1/2 and VP3] MAADGYLPDWLEDTLSEGIRQWWKLKPGPPPPKPAERHKDDSRGLVLPGYKYLGPF NGLDKGEPVNEADAAALEHDKAYDRQLDSGDNPYLKYNHADAEFQERLKEDTSFG GNLGRAVFQAKKRVLEPLGLVEEPVKTAPGKKRPVEHSPVEPDSSSGTGKAGQQPA RKRLNFGQTGDADSVPDPQPLGQPPAAPSGLGTNTMSAGGGGPLGDNNQGADGVG NASGDWHCDSTWMGDRVVTKSTRTWVLPSYNNHQYREIKSGSVDGSNANAYFGY STPWGYFDFNRFHSHWSPRDWQRLINNYWGFRPRSLRVKIFNIQVKEVTVQDSTTTI ANNLTSTVQVFTDDDYQLPYVVGNGTEGCLPAFPPQVFTLPQYGYATLNRDNTENP TERSSFFCLEYFPSKMLRTGNNFEFTYNFEEVPFHSSFAPSQNLFKLANPLVDQYLYR FVSTNNTGGVQFNKNLAGRYANTYKNWFPGPMGRTQGWNLGSGVNRASVSAFAT TNRMELEGASYQVPPQPNGMTNNLQGSNTYALENTMIFNSQPANPGTTATYLEGNM LITSESETQPVNRVAYNVGGQMATNNQSSTTAPATGTYNLQEIVPGSVWMERDVYL QGPIWAKIPETGAHFHPSPAMGGFGLKHPPPMMLIKNTPVPGNITSFSDVPVSSFITQY STGQVTVEMEWELKKENSKRWNPEIQYTNNYNDPQFVDFAPDSTGEYRTTRPIGTR YLTRPL In some embodiments, a chimeric AAV5 capsid of the present invention may comprise, consist essentially of, or consist of an amino acid sequence at least 90% identical (e.g., at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical) to the amino acid sequence of SEQ ID NO:10 [AAV53 VP1/2 and VP3]. SEQ ID NO:10 [AAV53 VP1/2 and VP3] MAADGYLPDWLEDNLSEGIREWWALKPGVPQPKANQQHQDNRRGLVLPGYKYLG PGNGLDKGEPVNEADAAALEHDKAYDQQLKAGDNPYLKYNHADAEFQERLQEDTS FGGNLGRAVFQAKKRILEPLGLVEEAAKTAPGKKGAVDQSPQEPDSSSGVGKSGKQ PARKRLNFGQTGDSESVPDPQPLGEPPAAPTSLGSNTMSAGGGGPLGDNNQGADGV GNASGDWHCDSTWMGDRVVTKSTRTWVLPSYNNHQYREIKSGSVDGSNANAYFG YSTPWGYFDFNRFHSHWSPRDWQRLINNYWGFRPRSLRVKIFNIQVKEVTVQDSTTT IANNLTSTVQVFTDDDYQLPYVVGNGTEGCLPAFPPQVFTLPQYGYATLNRDNTENP TERSSFFCLEYFPSKMLRTGNNFEFTYNFEEVPFHSSFAPSQNLFKLANPLVDQYLYR FVSTNNTGGVQFNKNLAGRYANTYKNWFPGPMGRTQGWNLGSGVNRASVSAFAT TNRMELEGASYQVPPQPNGMTNNLQGSNTYALENTMIFNSQPANPGTTATYLEGNM LITSESETQPVNRVAYNVGGQMATNNQSSTTAPATGTYNLQEIVPGSVWMERDVYL QGPIWAKIPETGAHFHPSPAMGGFGLKHPPPMMLIKNTPVPGNITSFSDVPVSSFITQY STGQVTVEMEWELKKENSKRWNPEIQYTNNYNDPQFVDFAPDSTGEYRTTRPIGTR YLTRPL In some embodiments, a chimeric AAV5 capsid of the present invention may comprise, consist essentially of, or consist of an amino acid sequence at least 90% identical (e.g., at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical) to the amino acid sequence of SEQ ID NO:11 [AAV54 VP1/2 and VP3]. SEQ ID NO:11 [AAV54 VP1/2 and VP3] MTDGYLPDWLEDNLSEGVREWWALQPGAPKPKANQQHQDNARGLVLPGYKYLGP GNGLDKGEPVNAADAAALEHDKAYDQQLKAGDNPYLKYNHADAEFQQRLQGDTS FGGNLGRAVFQAKKRVLEPLGLVEQAGETAPGKKRPLIESPQQPDSSTGIGKKGKQP AKKKLVFEDETGAGDGPPEGSTSGAMSDDSEMSAGGGGPLGDNNQGADGVGNASG DWHCDSTWMGDRVVTKSTRTWVLPSYNNHQYREIKSGSVDGSNANAYFGYSTPW GYFDFNRFHSHWSPRDWQRLINNYWGFRPRSLRVKIFNIQVKEVTVQDSTTTIANNL TSTVQVFTDDDYQLPYVVGNGTEGCLPAFPPQVFTLPQYGYATLNRDNTENPTERSS FFCLEYFPSKMLRTGNNFEFTYNFEEVPFHSSFAPSQNLFKLANPLVDQYLYRFVSTN NTGGVQFNKNLAGRYANTYKNWFPGPMGRTQGWNLGSGVNRASVSAFATTNRME LEGASYQVPPQPNGMTNNLQGSNTYALENTMIFNSQPANPGTTATYLEGNMLITSES ETQPVNRVAYNVGGQMATNNQSSTTAPATGTYNLQEIVPGSVWMERDVYLQGPIW AKIPETGAHFHPSPAMGGFGLKHPPPMMLIKNTPVPGNITSFSDVPVSSFITQYSTGQV TVEMEWELKKENSKRWNPEIQYTNNYNDPQFVDFAPDSTGEYRTTRPIGTRYLTRPL In some embodiments, a chimeric AAV5 capsid of the present invention may comprise, consist essentially of, or consist of an amino acid sequence at least 90% identical (e.g., at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical) to the amino acid sequence of SEQ ID NO:12 [AAV56 VP1/2 and VP3]. SEQ ID NO:12 [AAV56 VP1/2 and VP3] MAADGYLPDWLEDNLSEGIREWWDLKPGAPKPKANQQKQDDGRGLVLPGYKYLG PFNGLDKGEPVNAADAAALEHDKAYDQQLKAGDNPYLRYNHADAEFQERLQEDTS FGGNLGRAVFQAKKRVLEPFGLVEEGAKTAPGKKRPVEQSPQEPDSSSGIGKTGQQP AKKRLNFGQTGDSESVPDPQPLGEPPATPAAVGPTTMSAGGGGPLGDNNQGADGVG NASGDWHCDSTWMGDRVVTKSTRTWVLPSYNNHQYREIKSGSVDGSNANAYFGY STPWGYFDFNRFHSHWSPRDWQRLINNYWGFRPRSLRVKIFNIQVKEVTVQDSTTTI ANNLTSTVQVFTDDDYQLPYVVGNGTEGCLPAFPPQVFTLPQYGYATLNRDNTENP TERSSFFCLEYFPSKMLRTGNNFEFTYNFEEVPFHSSFAPSQNLFKLANPLVDQYLYR FVSTNNTGGVQFNKNLAGRYANTYKNWFPGPMGRTQGWNLGSGVNRASVSAFAT TNRMELEGASYQVPPQPNGMTNNLQGSNTYALENTMIFNSQPANPGTTATYLEGNM LITSESETQPVNRVAYNVGGQMATNNQSSTTAPATGTYNLQEIVPGSVWMERDVYL QGPIWAKIPETGAHFHPSPAMGGFGLKHPPPMMLIKNTPVPGNITSFSDVPVSSFITQY STGQVTVEMEWELKKENSKRWNPEIQYTNNYNDPQFVDFAPDSTGEYRTTRPIGTR YLTRPL In some embodiments, a chimeric AAV5 capsid of the present invention may comprise, consist essentially of, or consist of an amino acid sequence at least 90% identical (e.g., at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical) to the amino acid sequence of SEQ ID NO:13 [AAV57 VP1/2 and VP3]. SEQ ID NO:13 [AAV57 VP1/2 and VP3] MAADGYLPDWLEDNLSEGIREWWDLKPGAPKPKANQQKQDNGRGLVLPGYKYLG PFNGLDKGEPVNAADAAALEHDKAYDQQLKAGDNPYLRYNHADAEFQERLQEDTS FGGNLGRAVFQAKKRVLEPLGLVEEGAKTAPAKKRPVEPSPQRSPDSSTGIGKKGQQ PARKRLNFGQTGDSESVPDPQPLGEPPAAPSSVGSGTVAAGGGAPMSAGGGGPLGD NNQGADGVGNASGDWHCDSTWMGDRVVTKSTRTWVLPSYNNHQYREIKSGSVDG SNANAYFGYSTPWGYFDFNRFHSHWSPRDWQRLINNYWGFRPRSLRVKIFNIQVKE VTVQDSTTTIANNLTSTVQVFTDDDYQLPYVVGNGTEGCLPAFPPQVFTLPQYGYAT LNRDNTENPTERSSFFCLEYFPSKMLRTGNNFEFTYNFEEVPFHSSFAPSQNLFKLANP LVDQYLYRFVSTNNTGGVQFNKNLAGRYANTYKNWFPGPMGRTQGWNLGSGVNR ASVSAFATTNRMELEGASYQVPPQPNGMTNNLQGSNTYALENTMIFNSQPANPGTT ATYLEGNMLITSESETQPVNRVAYNVGGQMATNNQSSTTAPATGTYNLQEIVPGSV WMERDVYLQGPIWAKIPETGAHFHPSPAMGGFGLKHPPPMMLIKNTPVPGNITSFSD VPVSSFITQYSTGQVTVEMEWELKKENSKRWNPEIQYTNNYNDPQFVDFAPDSTGE YRTTRPIGTRYLTRPL In some embodiments, a chimeric AAV5 capsid of the present invention may comprise, consist essentially of, or consist of an amino acid sequence at least 90% identical (e.g., at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical) to the amino acid sequence of SEQ ID NO:14 [AAV58 VP1/2 and VP3]. SEQ ID NO:14 [AAV58 VP1/2 and VP3] MAADGYLPDWLEDNLSEGIREWWALKPGAPKPKANQQKQDDGRGLVLPGYKYLG PFNGLDKGEPVNAADAAALEHDKAYDQQLQAGDNPYLRYNHADAEFQERLQEDTS FGGNLGRAVFQAKKRVLEPLGLVEEGAKTAPGKKRPVEPSPQRSPDSSTGIGKKGQQ PARKRLNFGQTGDSESVPDPQPLGEPPAAPSGVGPNTMSAGGGGPLGDNNQGADGV GNASGDWHCDSTWMGDRVVTKSTRTWVLPSYNNHQYREIKSGSVDGSNANAYFG YSTPWGYFDFNRFHSHWSPRDWQRLINNYWGFRPRSLRVKIFNIQVKEVTVQDSTTT IANNLTSTVQVFTDDDYQLPYVVGNGTEGCLPAFPPQVFTLPQYGYATLNRDNTENP TERSSFFCLEYFPSKMLRTGNNFEFTYNFEEVPFHSSFAPSQNLFKLANPLVDQYLYR FVSTNNTGGVQFNKNLAGRYANTYKNWFPGPMGRTQGWNLGSGVNRASVSAFAT TNRMELEGASYQVPPQPNGMTNNLQGSNTYALENTMIFNSQPANPGTTATYLEGNM LITSESETQPVNRVAYNVGGQMATNNQSSTTAPATGTYNLQEIVPGSVWMERDVYL QGPIWAKIPETGAHFHPSPAMGGFGLKHPPPMMLIKNTPVPGNITSFSDVPVSSFITQY STGQVTVEMEWELKKENSKRWNPEIQYTNNYNDPQFVDFAPDSTGEYRTTRPIGTR YLTRPL In some embodiments, a chimeric AAV5 capsid of the present invention may comprise, consist essentially of, or consist of an amino acid sequence at least 90% identical (e.g., at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical) to the amino acid sequence of SEQ ID NO:15 [AAV59 VP1/2 and VP3]. SEQ ID NO:15 [AAV59 VP1/2 and VP3] MAADGYLPDWLEDNLSEGIREWWALKPGAPQPKANQQHQDNARGLVLPGYKYLG PGNGLDKGEPVNAADAAALEHDKAYDQQLKAGDNPYLKYNHADAEFQERLKEDT SFGGNLGRAVFQAKKRLLEPLGLVEEAAKTAPGKKRPVEQSPQEPDSSAGIGKSGAQ PAKKRLNFGQTGDTESVPDPQPIGEPPAAPSGVGSLTMSAGGGGPLGDNNQGADGV GNASGDWHCDSTWMGDRVVTKSTRTWVLPSYNNHQYREIKSGSVDGSNANAYFG YSTPWGYFDFNRFHSHWSPRDWQRLINNYWGFRPRSLRVKIFNIQVKEVTVQDSTTT IANNLTSTVQVFTDDDYQLPYVVGNGTEGCLPAFPPQVFTLPQYGYATLNRDNTENP TERSSFFCLEYFPSKMLRTGNNFEFTYNFEEVPFHSSFAPSQNLFKLANPLVDQYLYR FVSTNNTGGVQFNKNLAGRYANTYKNWFPGPMGRTQGWNLGSGVNRASVSAFAT TNRMELEGASYQVPPQPNGMTNNLQGSNTYALENTMIFNSQPANPGTTATYLEGNM LITSESETQPVNRVAYNVGGQMATNNQSSTTAPATGTYNLQEIVPGSVWMERDVYL QGPIWAKIPETGAHFHPSPAMGGFGLKHPPPMMLIKNTPVPGNITSFSDVPVSSFITQY STGQVTVEMEWELKKENSKRWNPEIQYTNNYNDPQFVDFAPDSTGEYRTTRPIGTR YLTRPL In some embodiments, a chimeric AAV5 capsid of the present invention may comprise, consist essentially of, or consist of an amino acid sequence at least 90% identical (e.g., at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical) to the amino acid sequence of SEQ ID NO:16 [AAV5-6 VP3]. SEQ ID NO:16 [AAV5-6 VP3] MSAGGGGPLGDNNQGADGVGNASGDWHCDSTWMGDRVVTKSTRTWVLPSYNNH QYREIKSGSVDGSNANAYFGYSTPWGYFDFNRFHSHWSPRDWQRLINNYWGFRPRS LRVKIFNIQVKEVTVQDSTTTIANNLTSTVQVFTDDDYQLPYVVGNGTEGCLPAFPPQ VFTLPQYGYATLNRDNTENPTERSSFFCLEYFPSKMLRTGNNFEFTYNFEEVPFHSSF APSQNLFKLANPLVDQYLYRFVSTNNTGGVQFNKNLAGRYANTYKNWFPGPMGRT QGWNLGSGVNRASVSAFATTNRMELEGASYQVPPQPNGMTNNLQGSNTYALENTM IFNSQPANPGTTATYLEGNMLITSESETQPVNRVAYNVGGQMATNNQSASTGASSTT APATGTYNLQEIVPGSVWMERDVYLQGPIWAKIPETGAHFHPSPAMGGFGLKHPPP MMLIKNTPVPGNITSFSDVPVSSFITQYSTGQVTVEMEWELKKENSKRWNPEIQYTN NYNDPQFVDFAPDSTGEYRTTRPIGTRYLTRPL In some embodiments, a chimeric AAV5 capsid of the present invention may comprise, consist essentially of, or consist of an amino acid sequence at least 90% identical (e.g., at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical) to the amino acid sequence of SEQ ID NO:17 [AAV596 VP1/2 and VP3]. SEQ ID NO:17 [AAV596 VP1/2 and VP3] MAADGYLPDWLEDNLSEGIREWWALKPGAPQPKANQQHQDNARGLVLPGYKYLG PGNGLDKGEPVNAADAAALEHDKAYDQQLKAGDNPYLKYNHADAEFQERLKEDT SFGGNLGRAVFQAKKRLLEPLGLVEEAAKTAPGKKRPVEQSPQEPDSSAGIGKSGAQ PAKKRLNFGQTGDTESVPDPQPIGEPPAAPSGVGSLTMSAGGGGPLGDNNQGADGV GNASGDWHCDSTWMGDRVVTKSTRTWVLPSYNNHQYREIKSGSVDGSNANAYFG YSTPWGYFDFNRFHSHWSPRDWQRLINNYWGFRPRSLRVKIFNIQVKEVTVQDSTTT IANNLTSTVQVFTDDDYQLPYVVGNGTEGCLPAFPPQVFTLPQYGYATLNRDNTENP TERSSFFCLEYFPSKMLRTGNNFEFTYNFEEVPFHSSFAPSQNLFKLANPLVDQYLYR FVSTNNTGGVQFNKNLAGRYANTYKNWFPGPMGRTQGWNLGSGVNRASVSAFAT TNRMELEGASYQVPPQPNGMTNNLQGSNTYALENTMIFNSQPANPGTTATYLEGNM LITSESETQPVNRVAYNVGGQMATNNQSASTGASSTTAPATGTYNLQEIVPGSVWM ERDVYLQGPIWAKIPETGAHFHPSPAMGGFGLKHPPPMMLIKNTPVPGNITSFSDVPV SSFITQYSTGQVTVEMEWELKKENSKRWNPEIQYTNNYNDPQFVDFAPDSTGEYRTT RPIGTRYLTRPL In some embodiments, a chimeric AAV5 capsid of the present invention may comprise, consist essentially of, or consist of an amino acid sequence at least 90% identical (e.g., at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical) to the amino acid sequence of SEQ ID NO:18 [AAV5-8 VP3]. SEQ ID NO:18 [AAV5-8 VP3] MSAGGGGPLGDNNQGADGVGNASGDWHCDSTWMGDRVVTKSTRTWVLPSYNNH QYREIKSGSVDGSNANAYFGYSTPWGYFDFNRFHSHWSPRDWQRLINNYWGFRPRS LRVKIFNIQVKEVTVQDSTTTIANNLTSTVQVFTDDDYQLPYVVGNGTEGCLPAFPPQ VFTLPQYGYATLNRDNTENPTERSSFFCLEYFPSKMLRTGNNFEFTYNFEEVPFHSSF APSQNLFKLANPLVDQYLYRFVSTNNTGGVQFNKNLAGRYANTYKNWFPGPMGRT QGWNLGSGVNRASVSAFATTNRMELEGASYQVPPQPNGMTNNLQGSNTYALENTM IFNSQPANPGTTATYLEGNMLITSESETQPVNRVAYNVGGQMATNNQNGTSGGATSS TTAPATGTYNLQEIVPGSVWMERDVYLQGPIWAKIPETGAHFHPSPAMGGFGLKHPP PMMLIKNTPVPGNITSFSDVPVSSFITQYSTGQVTVEMEWELKKENSKRWNPEIQYT NNYNDPQFVDFAPDSTGEYRTTRPIGTRYLTRPL In some embodiments, a chimeric AAV5 capsid of the present invention may comprise, consist essentially of, or consist of an amino acid sequence at least 90% identical (e.g., at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical) to the amino acid sequence of SEQ ID NO:19 [AAV598 VP1/2 and VP3]. SEQ ID NO:19 [AAV598 VP1/2 and VP3] MAADGYLPDWLEDNLSEGIREWWALKPGAPQPKANQQHQDNARGLVLPGYKYLG PGNGLDKGEPVNAADAAALEHDKAYDQQLKAGDNPYLKYNHADAEFQERLKEDT SFGGNLGRAVFQAKKRLLEPLGLVEEAAKTAPGKKRPVEQSPQEPDSSAGIGKSGAQ PAKKRLNFGQTGDTESVPDPQPIGEPPAAPSGVGSLTMSAGGGGPLGDNNQGADGV GNASGDWHCDSTWMGDRVVTKSTRTWVLPSYNNHQYREIKSGSVDGSNANAYFG YSTPWGYFDFNRFHSHWSPRDWQRLINNYWGFRPRSLRVKIFNIQVKEVTVQDSTTT IANNLTSTVQVFTDDDYQLPYVVGNGTEGCLPAFPPQVFTLPQYGYATLNRDNTENP TERSSFFCLEYFPSKMLRTGNNFEFTYNFEEVPFHSSFAPSQNLFKLANPLVDQYLYR FVSTNNTGGVQFNKNLAGRYANTYKNWFPGPMGRTQGWNLGSGVNRASVSAFAT TNRMELEGASYQVPPQPNGMTNNLQGSNTYALENTMIFNSQPANPGTTATYLEGNM LITSESETQPVNRVAYNVGGQMATNNQNGTSGGATSSTTAPATGTYNLQEIVPGSV WMERDVYLQGPIWAKIPETGAHFHPSPAMGGFGLKHPPPMMLIKNTPVPGNITSFSD VPVSSFITQYSTGQVTVEMEWELKKENSKRWNPEIQYTNNYNDPQFVDFAPDSTGE YRTTRPIGTRYLTRPL It is to be understood that these examples are not intended to be limiting and any AAV serotypes can be combined with any other AAV serotypes, in any combination of said serotypes. Furthermore, the chimeric AAV5 capsid produced from the VP1, VP2, and VP3 of the respective AAV serotypes can be included in the methods and compositions of this invention in any combination and/or in any ratio relative to one another, as would be well understood to one of ordinary skill in the art. The amino acid residue positions of the substitutions that can be made to produce the desired chimeric AAV5 capsid can be readily determined by one of ordinary skill in the art according to the teachings herein and according to protocols well known in the art. The amino acid residue numbering provided in the amino acid sequences set forth here is based on the reference sequences of AAV5 wild type VP1, VP2, and VP3 capsid protein amino acid sequences, as provided herein (SEQ ID NO:1 (AAV5 VP1 and VP2), SEQ ID NO:2 (AAV5 VP3), and SEQ ID NO:20 (AAV5 VP1, VP2, and VP3, NCBI Accession No. YP_068409.1). However it would be readily understood by one of ordinary skill in the art that the equivalent amino acid positions in other AAV serotype sequences (e.g., including, but not limited to, the amino acid sequences of AAD27757.1 (AAV1), YP_068409.1 (AAV5), AAC03780.1 (AAV2), AAC58045.1 (AAV4), NP_043941.1 (AAV3), AAB95450.1 (AAV6), YP_077178.1 (AAV7), YP_077180.1 (AAV8), AAS99264.1 (AAV9), AAO88201.1 (AAVrh10), and any serotype of Table 1) can be readily identified and employed in the production of the chimeric AAV5 capsids of this invention. It would be understood that the modifications described above provide multiple examples of how the amino acid sequences described herein can be obtained and that, due to the degeneracy of the amino acid codons, numerous other modifications can be made to a nucleotide sequence encoding a capsid or fragment thereof (e.g., VP1, VP2, and/or VP3) to obtain the desired amino acid sequence. The present invention provides additional non limiting examples of nucleic acids and/or polypeptides of this invention that can be used in the compositions and methods described herein in the SEQUENCES section provided herein. It will be apparent to those skilled in the art that the amino acid sequences of the chimeric AAV capsid proteins of the present invention (including, but not limited to, SEQ ID NOs:8-19) can further be modified to incorporate other modifications as known in the art to impart desired properties. As nonlimiting possibilities, the capsid and/or capsid protein(s) can be modified to incorporate targeting sequences (e.g., clotting factors) or sequences that facilitate purification and/or detection. For example, the capsid and/or capsid protein(s) can be fused to all or a portion of glutathione-S-transferase, maltose-binding protein, a heparin/heparan sulfate binding domain, poly-His, a ligand, and/or a reporter protein (e.g., Green Fluorescent Protein, ^-glucuronidase, ^-galactosidase, luciferase, etc.), an immunoglobulin Fc fragment, a single-chain antibody, hemagglutinin, c-myc, FLAG epitope, and the like to form a fusion protein. Methods of inserting targeting peptides into an AAV capsid are known in the art (see, e.g., international patent publication WO 00/28004; Nicklin et al., (2001) Mol. Ther.474-181; White et al., (2004) Circulation 109:513 In some embodiments, a chimeric AAV5 capsid of the present invention may be covalently linked, bound to, or encapsidate a compound selected from the group consisting of a DNA molecule, an RNA molecule, a polypeptide, a carbohydrate, a lipid, a small organic molecule, and any combination thereof. The invention also provides chimeric AAV5 capsids of the invention and virus particles (i.e., virions) comprising the same, wherein the virus particle packages (i.e., encapsidates) a vector genome, optionally an AAV vector genome. In particular embodiments, the invention provides an AAV particle comprising an AAV capsid comprising an AAV capsid protein of the invention, wherein the AAV capsid packages an AAV vector genome. The invention also provides an AAV particle comprising an AAV capsid or AAV capsid protein encoded by a modified nucleic acid capsid coding sequence(s) of the invention. The chimeric capsid proteins and capsids can further comprise any other modification, now known or later identified. Those skilled in the art will appreciate that for some AAV capsids the corresponding modification(s) may 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. Likewise, when modifying AAV other than AAV5, the specific amino acid position(s) may be different than the position in AAV5. As discussed elsewhere herein, the corresponding amino acid position(s) will be readily apparent to those skilled in the art using well-known techniques. Another aspect of the present invention provides an AAV particle comprising: a chimeric AAV5 capsid of the present invention; and an AAV vector genome; wherein the AAV capsid encapsidates the AAV vector genome. In some embodiments, the AAV vector genome may comprise a heterologous nucleic acid molecule. In some embodiments, the heterologous nucleic acid molecule may encode an antisense RNA, microRNA, or RNAi. In particular embodiments, the virion is a recombinant vector comprising a heterologous nucleic acid (e.g., a nucleic acid of interest), e.g., for delivery to a cell. Thus, the present invention is useful for the delivery of nucleic acids to cells in vitro, ex vivo, and in vivo. In representative embodiments, the recombinant vector of the invention can be advantageously employed to deliver or transfer nucleic acids to animal (e.g., mammalian) cells. Heterologous molecules (e.g., nucleic acid, proteins, peptides, etc.) 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 a transgene for transfer of the molecules into host target cells. Such associated molecules can include DNA and/or RNA. Any heterologous nucleotide sequence(s) may be delivered by a virus vector of the present invention. Nucleic acids of interest include nucleic acids encoding polypeptides, optionally therapeutic (e.g., for medical or veterinary uses) and/or immunogenic (e.g., for vaccines) polypeptides. In some embodiments, the heterologous nucleic acid molecule may encode a polypeptide. In some embodiments, the heterologous nucleic acid molecule may encode a therapeutic polypeptide. Therapeutic polypeptides include, but are not limited to, cystic fibrosis transmembrane regulator protein (CFTR), dystrophin (including the protein product of dystrophin mini-genes or micro-genes, see, e.g., Vincent et al., (1993) Nature Genetics 5:130; U.S. Patent Publication No.2003017131; Wang et al., (2000) Proc. Natl. Acad. Sci. USA 97:13714-9 [mini-dystrophin]; Harper et al., (2002) Nature Med.8:253-61 [micro- dystrophin]); mini-agrin, a laminin-α2, a sarcoglycan (α, β, γ or δ), Fukutin-related protein, myostatin pro-peptide, follistatin, dominant negative myostatin, an angiogenic factor (e.g., VEGF, angiopoietin-1 or 2), an anti-apoptotic factor (e.g., heme-oxygenase-1, TGF-β, inhibitors of pro-apoptotic signals such as caspases, proteases, kinases, death receptors [e.g., CD-095], modulators of cytochrome C release, inhibitors of mitochondrial pore opening and swelling); activin type II soluble receptor, anti-inflammatory polypeptides such as the Ikappa B dominant mutant, sarcospan, utrophin, mini-utrophin, antibodies or antibody fragments against myostatin or myostatin propeptide, cell cycle modulators, Rho kinase modulators such as Cethrin, which is a modified bacterial C3 exoenzyme [available from BioAxone Therapeutics, Inc., Saint-Lauren, Quebec, Canada], BCL-xL, BCL2, XIAP, FLICEc-s, dominant-negative caspase-8, dominant negative caspase-9, SPI-6 (see, e.g., U.S. Patent Application No.20070026076), transcriptional factor PGC-α1, Pinch gene, ILK gene and thymosin β4 gene), clotting factors (e.g., Factor VIII, Factor IX, Factor X, etc.), erythropoietin, angiostatin, endostatin, catalase, tyrosine hydroxylase, an intracellular and/or extracellular superoxide dismutase, leptin, the LDL receptor, neprilysin, lipoprotein lipase, ornithine transcarbamylase, β-globin, α-globin, spectrin, α 1 -antitrypsin, methyl cytosine binding protein 2, adenosine deaminase, hypoxanthine guanine phosphoribosyl transferase, β- glucocerebrosidase, sphingomyelinase, lysosomal hexosaminidase A, branched-chain keto acid dehydrogenase, RP65 protein, a cytokine (e.g., α-interferon, β-interferon, interferon-γ, interleukins-1 through -14, granulocyte-macrophage colony stimulating factor, lymphotoxin, and the like), peptide growth factors, neurotrophic factors and hormones (e.g., somatotropin, insulin, insulin-like growth factors including IGF-1 and IGF-2, GLP-1, platelet derived growth factor, epidermal growth factor, fibroblast growth factor, nerve growth factor, neurotrophic factor –3 and –4, brain-derived neurotrophic factor, glial derived growth factor, transforming growth factor –α and –β, and the like), bone morphogenic proteins (including RANKL and VEGF), a lysosomal protein, a glutamate receptor, a lymphokine, soluble CD4, an Fc receptor, a T cell receptor, ApoE, ApoC, inhibitor 1 of protein phosphatase inhibitor 1 (I-1), phospholamban, serca2a, lysosomal acid α-glucosidase, α-galactosidase A, Barkct, β2- adrenergic receptor, β2-adrenergic receptor kinase (BARK), phosphoinositide-3 kinase (PI3 kinase), calsarcin, a receptor (e.g., the tumor necrosis growth factor-α soluble receptor), an anti-inflammatory factor such as IRAP, Pim-1, PGC-1α, SOD-1, SOD-2, ECF-SOD, kallikrein, thymosin-β4, hypoxia-inducible transcription factor [HIF], an angiogenic factor, 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; phospholamban inhibitory or dominant-negative molecules such as phospholamban S16E, a monoclonal antibody (including single chain monoclonal antibodies) or a suicide gene product (e.g., thymidine kinase, cytosine deaminase, diphtheria toxin, and tumor necrosis factors such as TNF-α), and any other polypeptide that has a therapeutic effect in a subject in need thereof. Heterologous nucleotide 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, a fluorescent protein (e.g., EGFP, GFP, RFP, BFP, YFP, or dsRED2), an enzyme that produces a detectable product, such as luciferase (e.g., from Gaussia, Renilla, or Photinus), ^-galactosidase, ^-glucuronidase, alkaline phosphatase, and chloramphenicol acetyltransferase gene, or proteins that can be directly detected. Virtually any protein can be directly detected by using, for example, specific antibodies to the protein. Additional markers (and associated antibiotics) that are suitable for either positive or negative selection of eukaryotic cells are disclosed in Sambrook and Russell (2001), Molecular Cloning, 3rd Ed., Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y., and Ausubel et al. (1992), Current Protocols in Molecular Biology, John Wiley & Sons, including periodic updates. Alternatively, the heterologous nucleic acid may encode a functional RNA, e.g., an antisense oligonucleotide, a ribozyme (e.g., as described in U.S. Patent No.5,877,022), RNAs that effect spliceosome-mediated trans-splicing (see, Puttaraju et al., (1999) Nature Biotech.17:246; U.S. Patent No.6,013,487; U.S. Patent No.6,083,702), interfering RNAs (RNAi) including small interfering RNAs (siRNA) that mediate gene silencing (see, Sharp et al., (2000) Science 287:2431), microRNA, or other non-translated "functional" RNAs, such as "guide" RNAs (Gorman et al., (1998) Proc. Nat. Acad. Sci. USA 95:4929; U.S. Patent No. 5,869,248 to Yuan et al.), and the like. Exemplary untranslated RNAs include RNAi or antisense RNA against the multiple drug resistance (MDR) gene product (e.g., to treat tumors and/or for administration to the heart to prevent damage by chemotherapy), RNAi or antisense RNA against myostatin (Duchenne or Becker muscular dystrophy), RNAi or antisense RNA against VEGF or a tumor immunogen including but not limited to those tumor immunogens specifically described herein (to treat tumors), RNAi or antisense oligonucleotides targeting mutated dystrophins (Duchenne or Becker muscular dystrophy), RNAi or antisense RNA against the hepatitis B surface antigen gene (to prevent and/or treat hepatitis B infection), RNAi or antisense RNA against the HIV tat and/or rev genes (to prevent and/or treat HIV) and/or RNAi or antisense RNA against any other immunogen from a pathogen (to protect a subject from the pathogen) or a defective gene product (to prevent or treat disease). RNAi or antisense RNA against the targets described above or any other target can also be employed as a research reagent. As is known in the art, anti-sense nucleic acids (e.g., DNA or RNA) and inhibitory RNA (e.g., microRNA and RNAi such as siRNA or shRNA) sequences can be used to induce "exon skipping" in patients with muscular dystrophy arising from defects in the dystrophin gene. Thus, the heterologous nucleic acid can encode an antisense nucleic acid or inhibitory RNA that induces appropriate exon skipping. Those skilled in the art will appreciate that the particular approach to exon skipping depends upon the nature of the underlying defect in the dystrophin gene, and numerous such strategies are known in the art. Exemplary antisense nucleic acids and inhibitory RNA sequences target the upstream branch point and/or downstream donor splice site and/or internal splicing enhancer sequence of one or more of the dystrophin exons (e.g., exons 19 or 23). For example, in particular embodiments, the heterologous nucleic acid encodes an antisense nucleic acid or inhibitory RNA directed against the upstream branch point and downstream splice donor site of exon 19 or 23 of the dystrophin gene. Such sequences can be incorporated into an AAV vector delivering a modified U7 snRNA and the antisense nucleic acid or inhibitory RNA (see, e.g., Goyenvalle et al., (2004) Science 306:1796-1799). As another strategy, a modified U1 snRNA can be incorporated into an AAV vector along with siRNA, microRNA or antisense RNA complementary to the upstream and downstream splice sites of a dystrophin exon (e.g., exon 19 or 23) (see, e.g., Denti et al., (2006) Proc. Nat. Acad. Sci. USA 103:3758-3763). Further, antisense nucleic acids and inhibitory RNA can target the splicing enhancer sequences within exons 19, 43, 45 or 53 (see, e.g., U.S. Patent No.6,653,467; U.S. Patent No.6,727,355; and U.S. Patent No.6,653,466). Ribozymes are RNA-protein complexes that cleave nucleic acids in a site-specific fashion. Ribozymes have specific catalytic domains that possess endonuclease activity (Kim et al., (1987) Proc. Natl. Acad. Sci. USA 84:8788; Gerlach et al., (1987) Nature 328:802; Forster and Symons, (1987) Cell 49:211). For example, a large number of ribozymes accelerate phosphoester transfer reactions with a high degree of specificity, often cleaving only one of several phosphoesters in an oligonucleotide substrate (Michel and Westhof, (1990) J. Mol. Biol.216:585; Reinhold-Hurek and Shub, (1992) Nature 357:173). This specificity has been attributed to the requirement that the substrate bind via specific base- pairing interactions to the internal guide sequence ("IGS") of the ribozyme prior to chemical reaction. Ribozyme catalysis has primarily been observed as part of sequence-specific cleavage/ligation reactions involving nucleic acids (Joyce, (1989) Nature 338:217). For example, U.S. Pat. No.5,354,855 reports that certain ribozymes can act as endonucleases with a sequence specificity greater than that of known ribonucleases and approaching that of the DNA restriction enzymes. Thus, sequence-specific ribozyme-mediated inhibition of nucleic acid expression may be particularly suited to therapeutic applications (Scanlon et al., (1991) Proc. Natl. Acad. Sci. USA 88:10591; Sarver et al., (1990) Science 247:1222; Sioud et al., (1992) J. Mol. Biol.223:831). MicroRNAs (mir) are natural cellular RNA molecules that can regulate the expression of multiple genes by controlling the stability of the mRNA. Over-expression or diminution of a particular microRNA can be used to treat a dysfunction and has been shown to be effective in a number of disease states and animal models of disease (see, e.g., Couzin, (2008) Science 319:1782-4). The chimeric AAV can be used to deliver microRNA into cells, tissues and subjects for the treatment of genetic and acquired diseases, or to enhance functionality and promote growth of certain tissues. For example, mir-1, mir-133, mir-206 and/or mir-208 can be used to treat cardiac and skeletal muscle disease (see, e.g., Chen et al., (2006) Genet. 38:228-33; van Rooij et al., (2008) Trends Genet.24:159-66). MicroRNA can also be used to modulate the immune system after gene delivery (Brown et al., (2007) Blood 110:4144- 52). The term "antisense oligonucleotide" (including "antisense RNA") as used herein, refers to a nucleic acid that is complementary to and specifically hybridizes to a specified DNA or RNA sequence. Antisense oligonucleotides and nucleic acids that encode the same can be made in accordance with conventional techniques. See, e.g., U.S. Patent No. 5,023,243 to Tullis; U.S. Patent No.5,149,797 to Pederson et al. Those skilled in the art will appreciate that it is not necessary that the antisense oligonucleotide be fully complementary to the target sequence as long as the degree of sequence similarity is sufficient for the antisense nucleotide sequence to specifically hybridize to its target (as defined above) and reduce production of the protein product (e.g., by at least about 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95% or more). To determine the specificity of hybridization, hybridization of such oligonucleotides to target sequences can be carried out under conditions of reduced stringency, medium stringency or even stringent conditions. Suitable conditions for achieving reduced, medium and stringent hybridization conditions are as described herein. Alternatively stated, in particular embodiments, antisense oligonucleotides of the invention have at least about 60%, 70%, 80%, 90%, 95%, 97%, 98% or higher sequence identity with the complement of the target sequence and reduce production of the protein product (as defined above). In some embodiments, the antisense sequence contains 1, 2, 3, 4, 5, 6, 7, 8, 9 or 10 mismatches as compared with the target sequence. Methods of determining percent identity of nucleic acid sequences are described in more detail elsewhere herein. The length of the antisense oligonucleotide is not critical as long as it specifically hybridizes to the intended target and reduces production of the protein product (as defined above) and can be determined in accordance with routine procedures. In general, the antisense oligonucleotide is at least about eight, ten or twelve or fifteen nucleotides in length and/or less than about 20, 30, 40, 50, 60, 70, 80, 100 or 150 nucleotides in length. RNA interference (RNAi) is another useful approach for reducing production of a protein product (e.g., shRNA or siRNA). RNAi is a mechanism of post-transcriptional gene silencing in which double-stranded RNA (dsRNA) corresponding to a target sequence of interest is introduced into a cell or an organism, resulting in degradation of the corresponding mRNA. The mechanism by which RNAi achieves gene silencing has been reviewed in Sharp et al., (2001) Genes Dev 15: 485-490; and Hammond et al., (2001) Nature Rev. Gen.2:110- 119). The RNAi effect persists for multiple cell divisions before gene expression is regained. RNAi is therefore a powerful method for making targeted knockouts or "knockdowns" at the RNA level. RNAi has proven successful in human cells, including human embryonic kidney and HeLa cells (see, e.g., Elbashir et al., Nature (2001) 411:494-8). Initial attempts to use RNAi in mammalian cells resulted in antiviral defense mechanisms involving PKR in response to the dsRNA molecules (see, e.g., Gil et al., (2000) Apoptosis 5:107). It has since been demonstrated that short synthetic dsRNA of about 21 nucleotides, known as "short interfering RNAs" (siRNA) can mediate silencing in mammalian cells without triggering the antiviral response (see, e.g., Elbashir et al., Nature (2001) 411:494-8; Caplen et al., (2001) Proc. Nat. Acad. Sci. USA 98:9742). The RNAi molecule (including an siRNA molecule) can be a short hairpin RNA (shRNA; see Paddison et al., (2002), Proc. Nat. Acad. Sci. USA 99:1443-1448), which is believed to be processed in the cell by the action of the RNase III like enzyme Dicer into 20- 25mer siRNA molecules. The shRNAs generally have a stem-loop structure in which two inverted repeat sequences are separated by a short spacer sequence that loops out. There have been reports of shRNAs with loops ranging from 3 to 23 nucleotides in length. The loop sequence is generally not critical. Exemplary loop sequences include the following motifs: AUG, CCC, UUCG, CCACC, CTCGAG, AAGCUU, CCACACC and UUCAAGAGA. The RNAi can further comprise a circular molecule comprising sense and antisense regions with two loop regions on either side to form a "dumbbell" shaped structure upon dsRNA formation between the sense and antisense regions. This molecule can be processed in vitro or in vivo to release the dsRNA portion, e.g., a siRNA. International patent publication WO 01/77350 describes a vector for bi-directional transcription to generate both sense and antisense transcripts of a heterologous sequence in a eukaryotic cell. This technique can be employed to produce RNAi for use according to the invention. Shinagawa et al., (2003) Genes Dev.17:1340 reported a method of expressing long dsRNAs from a CMV promoter (a pol II promoter), which method is also applicable to tissue specific pol II promoters. Likewise, the approach of Xia et al., (2002) Nature Biotech. 20:1006, avoids poly(A) tailing and can be used in connection with tissue-specific promoters. Methods of generating RNAi include chemical synthesis, in vitro transcription, digestion of long dsRNA by Dicer (in vitro or in vivo), expression in vivo from a delivery vector, and expression in vivo from a PCR-derived RNAi expression cassette (see, e.g., TechNotes 10(3) "Five Ways to Produce siRNAs," from Ambion, Inc., Austin TX). Guidelines for designing siRNA molecules are available (see e.g., literature from Ambion, Inc., Austin TX; available at www.ambion.com). In particular embodiments, the siRNA sequence has about 30-50% G/C content. Further, long stretches of greater than four T or A residues are generally avoided if RNA polymerase III is used to transcribe the RNA. Online siRNA target finders are available, e.g., from Ambion, Inc., through the Whitehead Institute of Biomedical Research or from Dharmacon Research, Inc. The antisense region of the RNAi molecule can be completely complementary to the target sequence, but need not be as long as it specifically hybridizes to the target sequence (as defined above) and reduces production of the protein product (e.g., by at least about 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95% or more). In some embodiments, hybridization of such oligonucleotides to target sequences can be carried out under conditions of reduced stringency, medium stringency or even stringent conditions, as defined above. In some embodiments, the antisense region contains 1, 2, 3, 4, 5, 6, 7, 8, 9 or 10 mismatches as compared with the target sequence. Mismatches are generally tolerated better at the ends of the dsRNA than in the center portion. In particular embodiments, the RNAi is formed by intermolecular complexing between two separate sense and antisense molecules. The RNAi comprises a ds region formed by the intermolecular basepairing between the two separate strands. In other embodiments, the RNAi comprises a ds region formed by intramolecular basepairing within a single nucleic acid molecule comprising both sense and antisense regions, typically as an inverted repeat (e.g., a shRNA or other stem loop structure, or a circular RNAi molecule). The RNAi can further comprise a spacer region between the sense and antisense regions. Generally, RNAi molecules are highly selective. If desired, those skilled in the art can readily eliminate candidate RNAi that are likely to interfere with expression of nucleic acids other than the target by searching relevant databases to identify RNAi sequences that do not have substantial sequence homology with other known sequences, for example, using BLAST (available at ncbi.nlm.nih.gov/BLAST). Kits for the production of RNAi are commercially available, e.g., from New England Biolabs, Inc. and Ambion, Inc. The recombinant virus vector may also comprise a heterologous nucleotide sequence that shares homology with and recombines with a locus on the host chromosome. This approach may be utilized to correct a genetic defect in the host cell. The present invention also provides recombinant virus vectors that express an immunogenic polypeptide, e.g., for vaccination. The heterologous nucleic acid may encode any immunogen of interest known in the art including, but are not limited to, immunogens from human immunodeficiency virus, influenza virus, gag proteins, tumor antigens, cancer antigens, bacterial antigens, viral antigens, and the like. Alternatively, the immunogen can be presented in the virus capsid (e.g., incorporated therein) or tethered to the virus capsid (e.g., by covalent modification). The use of parvoviruses as vaccines is known in the art (see, e.g., Miyamura et al., (1994) Proc. Nat. Acad. Sci. USA 91:8507; U.S. Patent No.5,916,563 to Young et al., 5,905,040 to Mazzara et al., U.S. Patent No.5,882,652, U.S. Patent No.5,863,541 to Samulski et al.; the disclosures of which are incorporated herein in their entireties by reference). The antigen may be presented in the virus capsid. Alternatively, the antigen may be expressed from a heterologous nucleic acid introduced into a recombinant vector genome. An immunogenic polypeptide, or immunogen, may be any polypeptide suitable for protecting the subject against a disease, including but not limited to microbial, bacterial, protozoal, parasitic, fungal and viral diseases. For example, the immunogen may be an orthomyxovirus immunogen (e.g., an influenza virus immunogen, such as the influenza virus hemagglutinin (HA) surface protein or the influenza virus nucleoprotein gene, 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 GP160 protein, the HIV or SIV matrix/capsid proteins, and the HIV or SIV gag, pol and env genes products). The immunogen may also be an arenavirus immunogen (e.g., Lassa fever virus immunogen, such as the Lassa fever virus nucleocapsid protein gene and the Lassa fever envelope glycoprotein gene), a poxvirus immunogen (e.g., vaccinia, such as the vaccinia L1 or L8 genes), 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 genes), a bunyavirus immunogen (e.g., RVFV, CCHF, and SFS viruses), or a coronavirus immunogen (e.g., an infectious human coronavirus immunogen, such as the human coronavirus envelope glycoprotein gene, or a porcine transmissible gastroenteritis virus immunogen, or an avian infectious bronchitis virus immunogen, or a severe acute respiratory syndrome (SARS) immunogen such as a S [S1 or S2], M, E, or N protein or an immunogenic fragment thereof). The immunogen may further be a polio immunogen, herpes immunogen (e.g., CMV, EBV, HSV immunogens) mumps immunogen, measles immunogen, rubella immunogen, diphtheria toxin or other diphtheria immunogen, pertussis antigen, hepatitis (e.g., hepatitis A, hepatitis B or hepatitis C) immunogen, or any other vaccine immunogen known in the art. Alternatively, the immunogen may 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, (1999) Immunity 10:281). Illustrative cancer and tumor antigens include, but are not limited to: BRCA1 gene product, BRCA2 gene product, gp100, tyrosinase, GAGE-1/2, BAGE, RAGE, NY-ESO-1, CDK-4, ^- catenin, MUM-1, Caspase-8, KIAA0205, HPVE, SART-1, PRAME, 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) including MART-1 (Coulie et al., (1991) J. Exp. Med.180:35), gp100 (Wick et al., (1988) J. Cutan. Pathol.4:201) and MAGE antigen (MAGE-1, MAGE-2 and MAGE-3) (Van der Bruggen et al., (1991) Science, 254:1643), CEA, TRP-1; TRP-2; P-15 and tyrosinase (Brichard et al., (1993) J. Exp. Med.178:489); HER-2/neu gene product (U.S. Pat. No.4,968,603); CA 125; HE4; LK26; FB5 (endosialin); TAG 72; AFP; CA19-9; 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 WO 90/05142); telomerases; nuclear matrix proteins; prostatic acid phosphatase; papilloma virus antigens; and antigens associated with the following cancers: melanomas, adenocarcinoma, thymoma, sarcoma, lung cancer, liver cancer, colorectal cancer, non-Hodgkin's lymphoma, Hodgkin's lymphoma, leukemias, uterine cancer, breast cancer, prostate cancer, ovarian cancer, cervical cancer, bladder cancer, kidney cancer, pancreatic cancer, brain cancer, kidney cancer, stomach cancer, esophageal cancer, head and neck cancer and others (see, e.g., Rosenberg, (1996) Annu. Rev. Med.47:481-91). The present invention further provides a composition, which can be a pharmaceutical formulation comprising the virus vector or AAV particle of this invention and a pharmaceutically acceptable carrier. Alternatively, the heterologous nucleotide sequence may 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 protein product isolated therefrom. The present invention further provides a nucleic acid molecule encoding a chimeric AAV5 capsid of the present invention. It will be understood by those skilled in the art that the heterologous nucleic acid(s) of interest may be operably associated with appropriate control sequences. For example, the heterologous nucleic acid may be operably associated with expression control elements, such as transcription/translation control signals, origins of replication, polyadenylation signals, internal ribosome entry sites (IRES), promoters, enhancers, and the like. Those skilled in the art will further appreciate that a variety of promoter/enhancer elements may be used depending on the level and tissue-specific expression desired. The promoter/enhancer may be constitutive or inducible, depending on the pattern of expression desired. The promoter/enhancer may 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. Promoter/enhancer elements can be native to the target cell or subject to be treated and/or native to the heterologous nucleic acid sequence. The promoter/enhancer element is generally chosen so that it will function in the target cell(s) of interest. In representative embodiments, the promoter/enhancer element is a mammalian promoter/enhancer element. The promoter/enhance element may be constitutive or inducible. Inducible expression control elements are generally used 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 tissue-preferred promoter/enhancer elements, and include muscle specific or preferred (including cardiac, skeletal and/or smooth muscle), neural tissue specific or preferred (including brain-specific), eye (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. In one embodiment, a CNS cell-specific or CNS cell-preferred promoter is used. Examples of neuron-specific or preferred promoters include, without limitation, neuronal-specific enolase, synapsin, and MeCP2. Examples of astrocyte-specific or preferred promoters include, without limitation, glial fibrillary acidic protein and S100β. Examples of ependymal cell-specific or preferred promoters include, without limitation, wdr16, Foxj1, and LRP2. Examples of microglia- specific or preferred promoters include, without limitation, F4/80, CX3CR1, and CD11b. Examples of oligodendrocyte-specific or preferred promoters include, without limitation, myelin basic protein, cyclic nucleotide phosphodiesterase, proteolipid protein, Gtx, and Sox10. Use of a CNS cell-specific or preferred promoter can increase the specificity achieved by the chimeric AAV vector by further limiting expression of the heterologous nucleic acid to the CNS. 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 embodiments wherein the heterologous nucleic acid sequence(s) is transcribed and then translated in the target cells, specific initiation signals are generally employed 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. In other embodiments, nucleic acid sequences encoding a variant capsid or capsid protein of the invention have at least about 70%, 71%, 72%, 73%, 74%, 75%, 76%, 77%, 78%, 79%, 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99%, or higher sequence identity with a nucleic acid sequence encoding the amino acid sequence of SEQ ID NOs:8-19 and optionally encode a variant capsid or capsid protein that substantially retains at least one property of the capsid or capsid protein of the amino acid sequence of SEQ ID NOs:8-19. As is known in the art, a number of different programs can be used to identify whether a nucleic acid or polypeptide has sequence identity to a known sequence. Percent identity as used herein means that a nucleic acid or fragment thereof shares a specified percent identity to another nucleic acid, when optimally aligned (with appropriate nucleotide insertions or deletions) with the other nucleic acid (or its complementary strand), using BLASTN. To determine percent identity between two different nucleic acids, the percent identity is to be determined using the BLASTN program "BLAST 2 sequences". This program is available for public use from the National Center for Biotechnology Information (NCBI) over the Internet (Altschul et al., (1997) Nucleic Acids Res.25(17):3389-3402). The parameters to be used are whatever combination of the following yields the highest calculated percent identity (as calculated below) with the default parameters shown in parentheses: Program--blastn Matrix--0 BLOSUM62 Reward for a match--0 or 1 (1) Penalty for a mismatch--0, -1, -2 or -3 (-2) Open gap penalty--0, 1, 2, 3, 4 or 5 (5) Extension gap penalty-- 0 or 1 (1) Gap x_dropoff--0 or 50 (50) Expect--10. Percent identity or similarity when referring to polypeptides, indicates that the polypeptide in question exhibits a specified percent identity or similarity when compared with another protein or a portion thereof over the common lengths as determined using BLASTP. This program is also available for public use from the National Center for Biotechnology Information (NCBI) over the Internet (Altschul et al., (1997) Nucleic Acids Res.25(17):3389-3402). Percent identity or similarity for polypeptides is typically measured using sequence analysis software. See, e.g., the Sequence Analysis Software Package of the Genetics Computer Group, University of Wisconsin Biotechnology Center, 910 University Avenue, Madison, Wis.53705. Protein analysis software matches similar sequences using measures of homology assigned to various substitutions, deletions and other modifications. Conservative substitutions typically include substitutions within the following groups: glycine, alanine; valine, isoleucine, leucine; aspartic acid, glutamic acid; asparagine, glutamine; serine, threonine; lysine, arginine; and phenylalanine, tyrosine. The invention also provides chimeric AAV particles comprising an AAV capsid and an AAV genome, wherein the AAV genome "corresponds to" (i.e., encodes) the AAV capsid. Also provided are collections or libraries of such chimeric AAV particles, wherein the collection or library comprises 2 or more, 10 or more, 50 or more, 100 or more, 1000 or more, 104 or more, 105 or more, or 106 or more distinct sequences. The present invention further encompasses "empty" capsid particles (i.e., in the absence of a vector genome) comprising, consisting of, or consisting essentially of the chimeric AAV capsid proteins of the invention. The chimeric AAV capsids of the invention can be used as "capsid vehicles," as has been described in U.S. Patent No.5,863,541. Molecules that can be covalently linked, bound to or packaged by the virus capsids and transferred into a cell include DNA, RNA, a lipid, a carbohydrate, a polypeptide, a small organic molecule, or combinations of the same. Further, molecules can be associated with (e.g., "tethered to") the outside of the virus capsid for transfer of the molecules into host target cells. In one embodiment of the invention the 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 virus capsids of the invention 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 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. The invention also provides nucleic acids (e.g., isolated nucleic acids) encoding the chimeric virus capsids and chimeric capsid proteins of the invention. Further provided are vectors comprising the nucleic acids, and cells (in vivo or in culture) comprising the nucleic acids and/or vectors of the invention. Such nucleic acids, vectors and cells can be used, for example, as reagents (e.g., helper constructs or packaging cells) for the production of virus vectors as described herein. In some embodiments, a vector of the present invention may be a plasmid, phage, viral vector, bacterial artificial chromosome, or yeast artificial chromosome. In some embodiments, a viral vector of the present invention may be an AAV vector, an adenovirus vector, a herpesvirus vector, a lentivirus vector, an alphavirus vector or a baculovirus vector (e.g., an AAV particle, an adenovirus particle, a herpesvirus particle, a lentivirus particle, an alphavirus particle, a baculovirus particle, etc.). In some embodiments, the nucleic acid encoding the chimeric AAV capsid protein further comprises an AAV rep coding sequence. For example, the nucleic acid can be a helper construct for producing viral stocks. In another aspect of the invention, the chimeric/modified AAV capsid and vectors of the invention are fully- or nearly fully-detargeted vectors that can be further modified to a desirable tropic profile for targeting of one or more peripheral organs or tissues. The invention also provides packaging cells stably comprising a nucleic acid of the invention. For example, the nucleic acid can be stably incorporated into the genome of the cell or can be stably maintained in an episomal form (e.g., an "EBV based nuclear episome"). The nucleic acid can be incorporated into a delivery vector, such as a viral delivery vector. To illustrate, the nucleic acid of the invention can be packaged in an AAV particle, an adenovirus particle, a herpesvirus particle, a baculovirus particle, or any other suitable virus particle. Moreover, the nucleic acid can be operably associated with a promoter element. Promoter elements are described in more detail herein. Further provided is a pharmaceutical formulation comprising a AAV particle, nucleic acid molecule, and/or the vector of the present invention in a pharmaceutically acceptable carrier. In some embodiments, the present invention provides a pharmaceutical composition comprising a virus vector of the invention 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 will preferably 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. Methods of making the invention The present invention further provides methods of producing the virus capsid and/or virus particles of the present invention. In some embodiments, the present invention provides a method of producing a recombinant AAV particle comprising an AAV capsid, the method comprising: providing/introducing into a cell in vitro with a nucleic acid molecule of the present invention, an AAV rep coding sequence, an AAV vector genome comprising a heterologous nucleic acid molecule, and helper functions for generating a productive AAV infection under conditions whereby assembly of the recombinant AAV particle comprising the AAV capsid and encapsidation of the AAV vector genome can occur. The template and AAV replication and capsid sequences are provided under conditions such that recombinant virus particles comprising the template packaged within the capsid are produced in the cell. The method can further comprise the step of collecting the virus particles from the cell. Virus particles may be collected from the medium and/or by lysing the cells. Further provided herein is the AAV particle(s) produced by such a method. The cell is typically a cell that is permissive for AAV viral replication. Any suitable cell known in the art may be employed, such as mammalian cells. Also suitable are trans- complementing packaging cell lines that provide 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. As a further alternative, the rep/cap sequences may be stably carried (episomal or integrated) within a cell. Typically, the AAV rep/cap sequences will not be flanked by the AAV packaging sequences (e.g., AAV ITRs), to prevent rescue and/or packaging of these sequences. The template (e.g., an rAAV vector genome) can be provided to the cell using any method known in the art. For example, the template may be supplied by a non-viral (e.g., plasmid) or viral vector. In particular embodiments, the 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. Virol.72:5025, describe a baculovirus vector carrying a reporter gene flanked by the AAV ITRs. EBV vectors may also be employed to deliver the template, as described above with respect to the rep/cap genes. In another representative embodiment, the template is provided by a replicating rAAV virus. In still other embodiments, an AAV provirus is stably integrated into the chromosome of the cell. To obtain maximal virus titers, helper virus functions (e.g., adenovirus or herpesvirus) essential for a productive AAV infection are generally provided to the cell. Helper virus sequences necessary for AAV replication are known in the art. Typically, these sequences are 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 non- infectious adenovirus miniplasmid that carries all of the helper genes required for efficient AAV production as described by Ferrari et al., (1997) Nature Med.3:1295, and U.S. Patent Nos.6,040,183 and 6,093,570. Further, the helper virus functions may be provided by a packaging cell with the helper genes integrated in the chromosome or maintained as a stable extrachromosomal element. In representative embodiments, the helper virus sequences cannot be packaged into AAV virions, e.g., are not flanked by AAV ITRs. 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, but is optionally a hybrid adenovirus or hybrid herpesvirus comprising the AAV rep/cap genes. In one particular embodiment, the AAV rep/cap sequences and the adenovirus helper sequences are supplied by a single adenovirus helper vector. This vector further contains the rAAV template. The AAV rep/cap sequences and/or the rAAV template may be inserted into a deleted region (e.g., the E1a or E3 regions) of the adenovirus. In a further embodiment, the AAV rep/cap sequences and the adenovirus helper sequences are supplied by a single adenovirus helper vector. The rAAV template is provided as a plasmid template. In another illustrative embodiment, 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 a "EBV based nuclear episome," see Margolski, (1992) Curr. Top. Microbiol. Immun.158:67). In a further exemplary embodiment, the AAV rep/cap sequences and adenovirus helper sequences are provided by a single adenovirus helper. The rAAV template is provided as a separate replicating viral vector. For example, the rAAV template may 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, in representative embodiments, the adenovirus helper sequences and the AAV rep/cap sequences are not flanked by the AAV packaging sequences (e.g., the AAV ITRs), so that these sequences are not packaged into the AAV virions. Herpesvirus may also be used as a helper virus in AAV packaging methods. Hybrid herpesviruses encoding the AAV rep protein(s) may advantageously facilitate for more 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, the disclosures of which are incorporated herein in their entireties). As a further alternative, the virus vectors of the invention can be produced in insect cells using baculovirus vectors to deliver the rep/cap genes and rAAV template as described by Urabe et al., (2002) Human Gene Therapy 13:1935-43. Other methods of producing AAV use stably transformed packaging cells (see, e.g., U.S. Patent No.5,658,785). 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). In representative embodiments, deleted replication-defective helper viruses are 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). The inventive packaging methods may be employed to produce high titer stocks of virus particles. In particular embodiments, the virus stock has a titer of at least about 105 transducing units (tu)/ml, at least about 106 tu/ml, at least about 107 tu/ml, at least about 108 tu/ml, at least about 109 tu/ml, or at least about 1010 tu/ml. The novel capsid protein and capsid structures find use in raising antibodies, for example, for diagnostic or therapeutic uses or as a research reagent. Thus, the invention also provides antibodies against the novel capsid proteins and capsids of the invention. The term "antibody" or "antibodies" as used herein refers to all types of immunoglobulins, including IgG, IgM, IgA, IgD, and IgE. The antibody can be monoclonal or polyclonal and can be of any species of origin, including (for example) mouse, rat, rabbit, horse, goat, sheep or human, or can be a chimeric antibody. See, e.g., Walker et al., Mol. Immunol.26, 403-11 (1989). The antibodies can be recombinant monoclonal antibodies, for example, produced according to the methods disclosed in U.S. Patent No.4,474,893 or U.S. Patent No.4,816,567. The antibodies can also be chemically constructed, for example, according to the method disclosed in U.S. Patent No.4,676,980. Antibody fragments included within the scope of the present invention include, for example, Fab, F(ab')2, and Fc fragments, and the corresponding fragments obtained from antibodies other than IgG. Such fragments can be produced by known techniques. For example, F(ab´)2 fragments can be produced by pepsin digestion of the antibody molecule, and Fab fragments can be generated by reducing the disulfide bridges of the F(ab´)2 fragments. Alternatively, Fab expression libraries can be constructed to allow rapid and easy identification of monoclonal Fab fragments with the desired specificity (Huse et al., (1989) Science 254, 1275-1281). Polyclonal antibodies can be produced by immunizing a suitable animal (e.g., rabbit, goat, etc.) with an antigen to which a monoclonal antibody to the target binds, collecting immune serum from the animal, and separating the polyclonal antibodies from the immune serum, in accordance with known procedures. Monoclonal antibodies can be produced in a hybridoma cell line according to the technique of Kohler and Milstein, (1975) Nature 265, 495-97. For example, a solution containing the appropriate antigen can be injected into a mouse and, after a sufficient time, the mouse sacrificed and spleen cells obtained. The spleen cells are then immortalized by fusing them with myeloma cells or with lymphoma cells, typically in the presence of polyethylene glycol, to produce hybridoma cells. The hybridoma cells are then grown in a suitable medium and the supernatant screened for monoclonal antibodies having the desired specificity. Monoclonal Fab fragments can be produced in E. coli by recombinant techniques known to those skilled in the art. See, e.g., W. Huse, (1989) Science 246, 1275-81. Antibodies specific to a target polypeptide can also be obtained by phage display techniques known in the art. Various immunoassays can be used for screening to identify antibodies having the desired specificity. Numerous protocols for competitive binding or immunoradiometric assays using either polyclonal or monoclonal antibodies with established specificity are well known in the art. Such immunoassays typically involve the measurement of complex formation between an antigen and its specific antibody (e.g., antigen/antibody complex formation). A two-site, monoclonal-based immunoassay utilizing monoclonal antibodies reactive to two non-interfering epitopes can be used as well as a competitive binding assay. Antibodies can be conjugated to a solid support (e.g., beads, plates, slides or wells formed from materials such as latex or polystyrene) in accordance with known techniques. Antibodies can likewise be directly or indirectly conjugated to detectable groups such as radiolabels (e.g., 35S, 125I, 131I), enzyme labels (e.g., horseradish peroxidase, alkaline phosphatase), and fluorescence labels (e.g., fluorescein) in accordance with known techniques. Determination of the formation of an antibody/antigen complex in the methods of this invention can be by detection of, for example, precipitation, agglutination, flocculation, radioactivity, color development or change, fluorescence, luminescence, etc., as is well known in the art. Methods of using the invention The present invention also relates to methods for delivering heterologous nucleotide sequences into preferred tissues (e.g., the liver). The virus vectors of the invention may be employed to deliver a nucleotide sequence to a cell in vitro, e.g., to produce a polypeptide or nucleic acid in vitro or for ex vivo gene therapy. The vectors are additionally useful in a method of delivering a nucleotide sequence to a subject in need thereof, e.g., to express a therapeutic or immunogenic polypeptide or nucleic acid. In this manner, the polypeptide or nucleic acid may thus be produced in vivo in the subject. The subject may be in need of the polypeptide or nucleic acid because the subject has a deficiency of the polypeptide, or because the production of the polypeptide or nucleic acid in the subject may impart some therapeutic effect, as a method of treatment or otherwise, and as explained further below. In particular embodiments, the vectors are useful to express a polypeptide or nucleic acid that provides a beneficial effect to the liver, e.g., to deliver clotting factors and/or otherwise treat bleeding disorders such as, but not limited to, hemophilia. Accordingly, the ability to target vectors to the liver may be particularly useful to treat diseases or disorders involving liver dysfunction. Accordingly, one aspect of the present invention provides a method of delivering a nucleic acid molecule (e.g., a nucleic acid molecule of interest) to a hepatocyte, the method comprising contacting the hepatocyte with an AAV particle, nucleic acid molecule, vector, and/or pharmaceutical formulation of the present invention. Further provided is a method of delivering a nucleic acid molecule of interest (e.g., a nucleic acid molecule of interest) to a hepatocyte in a mammalian subject, the method comprising: administering an effective amount of an AAV particle, nucleic acid molecule, vector, and/or pharmaceutical formulation of the present invention to a mammalian subject, thereby delivering the nucleic acid molecule to a hepatocyte in the mammalian subject. A further aspect of the present invention provides a method of treating a disorder in a mammalian subject in need thereof, wherein the disorder is treatable by expressing a nucleic acid molecule encoding a therapeutic product in the liver of the subject, the method comprising administering a therapeutically effective amount of an AAV particle, nucleic acid molecule, vector, and/or pharmaceutical formulation of the present invention to the mammalian subject under conditions whereby the nucleic acid molecule encoding the therapeutic product is expressed in the liver, thereby treating the disorder. In general, the virus vectors of the invention may be employed to deliver any foreign nucleic acid with a biological effect to treat or ameliorate the symptoms associated with any disorder related to gene expression. Further, the invention can be used to treat any disease state for which it is beneficial to deliver a therapeutic polypeptide. 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 (ß-globin), anemia (erythropoietin) and other blood disorders, Alzheimer’s disease (GDF; neprilysin), multiple sclerosis (ß-interferon), Parkinson’s disease (glial-cell line derived neurotrophic factor [GDNF]), Huntington’s disease (inhibitory RNA including without limitation RNAi such as siRNA or shRNA, antisense RNA or microRNA to remove repeats), amyotrophic lateral sclerosis, epilepsy (galanin, neurotrophic factors), and other neurological disorders, cancer (endostatin, angiostatin, TRAIL, FAS-ligand, cytokines including interferons; inhibitory RNA including without limitation RNAi (such as siRNA or shRNA), antisense RNA and microRNA including inhibitory RNA against VEGF, the multiple drug resistance gene product or a cancer immunogen), diabetes mellitus (insulin, PGC- α1, GLP-1, myostatin pro-peptide, glucose transporter 4), muscular dystrophies including Duchenne and Becker (e.g., dystrophin, mini-dystrophin, micro-dystrophin, insulin-like growth factor I, a sarcoglycan [e.g., α, β, γ], Inhibitory RNA [e.g., RNAi, antisense RNA or microRNA] against myostatin or myostatin propeptide, laminin-alpha2, Fukutin-related protein, dominant negative myostatin, follistatin, activin type II soluble receptor, anti-inflammatory polypeptides such as the Ikappa B dominant mutant, sarcospan, utrophin, mini-utrophin, inhibitory RNA [e.g., RNAi, antisense RNA or microRNA] against splice junctions in the dystrophin gene to induce exon skipping [see, e.g., WO/2003/095647], inhibitory RNA (e.g., RNAi, antisense RNA or micro RNA] against U7 snRNAs to induce exon skipping [see, e.g., WO/2006/021724], and antibodies or antibody fragments against myostatin or myostatin propeptide), Gaucher disease (glucocerebrosidase), Hurler’s disease (α-L-iduronidase), adenosine deaminase deficiency (adenosine deaminase), glycogen storage diseases (e.g., Fabry disease [α-galactosidase] and Pompe disease [lysosomal acid α-glucosidase]) and other metabolic defects including other lysosomal storage disorders and glycogen storage disorders, congenital emphysema (α1-antitrypsin), Lesch-Nyhan Syndrome (hypoxanthine guanine phosphoribosyl transferase), Niemann-Pick disease (sphingomyelinase), Maple Syrup Urine Disease (branched-chain keto acid dehydrogenase), retinal degenerative diseases (and other diseases of the eye and retina; e.g., PDGF, endostatin and/or angiostatin for macular degeneration), 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 (RNAi such as siRNA or shRNA, microRNA or antisense RNA for hepatitis B and/or hepatitis C genes), kidney, heart including congestive heart failure or peripheral artery disease (PAD) (e.g., by delivering protein phosphatase inhibitor I [I-1], phospholamban, sarcoplasmic endoreticulum Ca 2+ - ATPase [serca2a], zinc finger proteins that regulate the phospholamban gene, Pim-1, PGC- 1α, SOD-1, SOD-2, ECF-SOD, kallikrein, thymosin-β4, hypoxia-inducible transcription factor [HIF], βarkct, β2-adrenergic receptor, β2-adrenergic receptor kinase [βARK], phosphoinositide-3 kinase [PI3 kinase], calsarcin, an angiogenic factor, 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, an inhibitory RNA [e.g., RNAi, antisense RNA or microRNA] against phospholamban; phospholamban inhibitory or dominant-negative molecules such as phospholamban S16E, etc.), arthritis (insulin-like growth factors), joint disorders (insulin-like growth factors), 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, myostatin pro-peptide, an anti-apoptotic factor, follistatin), limb ischemia (VEGF, FGF, PGC-1α, EC-SOD, HIF), kidney deficiency (erythropoietin), anemia (erythropoietin), arthritis (anti-inflammatory factors such as IRAP and TNFα soluble receptor), hepatitis (α-interferon), LDL receptor deficiency (LDL receptor), hyperammonemia (ornithine transcarbamylase), spinal cerebral ataxias including SCA1, SCA2 and SCA3, phenylketonuria (phenylalanine hydroxylase), autoimmune diseases, and the like. The invention 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 RANKL and/or VEGF) can be administered with a bone allograft, for example, following a break or surgical removal in a cancer patient. Exemplary lysosomal storage diseases that can be treated according to the present invention include without limitation: Hurler’s Syndrome (MPS IH), Scheie’s Syndrome (MPS IS), and Hurler-Scheie Syndrome (MPS IH/S) (α-L-iduronidase); Hunter’s Syndrome (MPS II) (iduronate sulfate sulfatase); Sanfilippo A Syndrome (MPS IIIA) (Heparan-S- sulfate sulfaminidase), Sanfilippo B Syndrome (MPS IIIB) (N-acetyl-D-glucosaminidase), Sanfilippo C Syndrome (MPS IIIC) (Acetyl-CoA-glucosaminide N-acetyltransferase), Sanfilippo D Syndrome (MPS IIID) (N-acetyl-glucosaminine-6-sulfate sulfatase); Morquio A disease (MPS IVA) (Galactosamine-6-sulfate sulfatase), Morquio B disease (MPS IV B) (β- Galactosidase); Maroteaux-lmay disease (MPS VI) (arylsulfatase B); Sly Syndrome (MPS VII) (β-glucuronidase); hyaluronidase deficiency (MPS IX) (hyaluronidase); sialidosis (mucolipidosis I), mucolipidosis II (I-Cell disease) (N-actylglucos-aminyl-1- phosphotransferase catalytic subunit), mucolipidosis III (pseudo-Hurler polydystrophy) (N- acetylglucos-aminyl-1-phosphotransferase; type IIIA [catalytic subunit] and type IIIC [substrate recognition subunit]); GM1 gangliosidosis (ganglioside β-galactosidase), GM2 gangliosidosis Type I (Tay-Sachs disease) (β-hexaminidase A), GM2 gangliosidosis type II (Sandhoff’s disease) (β-hexosaminidase B); Niemann-Pick disease (Types A and B) (sphingomyelinase); Gaucher’s disease (glucocerebrosidase); Farber’s disease (ceraminidase); Fabry’s disease (α-galactosidase A); Krabbe’s disease (galactosylceramide β- galactosidase); metachromatic leukodystrophy (arylsulfatase A); lysosomal acid lipase deficiency including Wolman’s disease (lysosomal acid lipase); Batten disease (juvenile neuronal ceroid lipofuscinosis) (lysosomal trans-membrane CLN3 protein) sialidosis (neuraminidase 1); galactosialidosis (Goldberg’s syndrome) (protective protein/ cathepsin A); α-mannosidosis (α-D-mannosidase); β-mannosidosis (β-D-mannosidosis); fucosidosis (α-D- fucosidase); aspartylglucosaminuria (N-Aspartylglucosaminidase); and sialuria (Na phosphate cotransporter). Exemplary glycogen storage diseases that can be treated according to the present invention include, but are not limited to, Type Ia GSD (von Gierke disease) (glucose-6- phosphatase), Type Ib GSD (glucose-6-phosphate translocase), Type Ic GSD (microsomal phosphate or pyrophosphate transporter), Type Id GSD (microsomal glucose transporter), Type II GSD including Pompe disease or infantile Type IIa GSD (lysosomal acid α- glucosidase) and Type IIb (Danon) (lysosomal membrane protein-2), Type IIIa and IIIb GSD (Debrancher enzyme; amyloglucosidase and oligoglucanotransferase), Type IV GSD (Andersen's disease) (branching enzyme), Type V GSD (McArdle disease) (muscle phosphorylase), Type VI GSD (Hers' disease) (liver phosphorylase), Type VII GSD (Tarui's disease) (phosphofructokinase), GSD Type VIII/IXa (X-linked phosphorylase kinase), GSD Type IXb (Liver and muscle phosphorylase kinase), GSD Type IXc (liver phosphorylase kinase), GSD Type IXd (muscle phosphorylase kinase), GSD O (glycogen synthase), Fanconi-Bickel syndrome (glucose transporter-2), phosphoglucoisomerase deficiency, muscle phosphoglycerate kinase deficiency, phosphoglycerate mutase deficiency, fructose 1,6- diphosphatase deficiency, phosphoenolpyruvate carboxykinase deficiency, and lactate dehydrogenase deficiency. Nucleic acids and polypeptides that can be delivered to cardiac muscle include those that are beneficial in the treatment of damaged, degenerated or atrophied cardiac muscle and/or congenital cardiac defects. For example, angiogenic factors useful for facilitating vascularization in the treatment of heart disease include but are not limited to vascular endothelial growth factor (VEGF), VEGF II, VEGF-B, VEGF-C, VEGF-D, VEGF-E, VEGF121, VEGF138, VEGF145, VEGF165, VEGF189, VEGF206, hypoxia inducible factor 1α (HIF 1α), endothelial NO synthase (eNOS), iNOS, VEFGR-1 (Flt1), VEGFR-2 (KDR/Flk1), VEGFR-3 (Flt4), angiogenin, epidermal growth factor (EGF), angiopoietin, platelet-derived growth factor, angiogenic factor, transforming growth factor-α (TGF- α), transforming growth factor- β (TGF-β), vascular permeability factor (VPF), tumor necrosis factor alpha (TNF-α), interleukin-3 (IL-3), interleukin-8 (IL-8), platelet-derived endothelial growth factor (PD-EGF), granulocyte colony stimulating factor (G-CSF), hepatocyte growth factor (HGF), scatter factor (SF), pleitrophin, proliferin, follistatin, placental growth factor (PIGF), midkine, platelet-derived growth factor-BB (PDGF), fractalkine, ICAM-1, angiopoietin-1 and -2 (Ang1 and Ang2), Tie-2, neuropilin-1, ICAM-1, chemokines and cytokines that stimulate smooth muscle cell, monocyte, or leukocyte migration, anti-apoptotic peptides and proteins, fibroblast growth factors (FGF), FGF-1, FGF-1b, FGF-1c, FGF-2, FGF-2b, FGF-2c, FGF-3, FGF-3b, FGF-3c, FGF-4, FGF-5, FGF-7, FGF-9, acidic FGF, basic FGF, monocyte chemotactic protein-1, granulocyte macrophage-colony stimulating factor, insulin-like growth factor-1 (IGF-1), IGF-2, early growth response factor-1 (EGR-1), ETS-1, human tissue kallikrein (HK), matrix metalloproteinase, chymase, urokinase-type plasminogen activator and heparinase. (see, e.g., U.S. Patent Application No.20060287259 and U.S. Patent Application No.20070059288). The most common congenital heart disease found in adults is bicuspid aortic valve, whereas atrial septal defect is responsible for 30-40% of congenital heart disease seen in adults. The most common congenital cardiac defect observed in the pediatric population is ventricular septal defect. Other congenital heart diseases include Eisenmenger's syndrome, patent ductus arteriosus, pulmonary stenosis, coarctation of the aorta, transposition of the great arteries, tricuspid atresia, univentricular heart, Ebstein's anomaly, and double-outlet right ventricle. A number of studies have identified putative genetic loci associated with one or more of these congenital heart diseases. For example, the putative gene(s) for congenital heart disease associated with Down syndrome is 21q22.2-q22.3, between ETS2 and MX1. Similarly, most cases of DiGeorge syndrome result from a deletion of chromosome 22q11.2 (the DiGeorge syndrome chromosome region, or DGCR). Several genes are lost in this deletion including the putative transcription factor TUPLE1. This deletion is associated with a variety of phenotypes, e.g., Shprintzen syndrome; conotruncal anomaly face (or Takao syndrome); and isolated outflow tract defects of the heart including Tetralogy of Fallot, truncus arteriosus, and interrupted aortic arch. All of the foregoing disorders can be treated according to the present invention. Other significant diseases of the heart and vascular system are also believed to have a genetic, typically polygenic, etiological component. These diseases include, for example, hypoplastic left heart syndrome, cardiac valvular dysplasia, Pfeiffer cardiocranial syndrome, oculofaciocardiodental syndrome, Kapur-Toriello syndrome, Sonoda syndrome, Ohdo Blepharophimosis syndrome, heart-hand syndrome, Pierre-Robin syndrome, Hirschsprung disease, Kousseff syndrome, Grange occlusive arterial syndrome, Kearns-Sayre syndrome, Kartagener syndrome, Alagille syndrome, Ritscher-Schinzel syndrome, Ivemark syndrome, Young-Simpson syndrome, hemochromatosis, Holzgreve syndrome, Barth syndrome, Smith- Lemli-Opitz syndrome, glycogen storage disease, Gaucher-like disease, Fabry disease, Lowry-Maclean syndrome, Rett syndrome, Opitz syndrome, Marfan syndrome, Miller-Dieker lissencephaly syndrome, mucopolysaccharidosis, Bruada syndrome, humerospinal dysostosis, Phaver syndrome, McDonough syndrome, Marfanoid hypermobility syndrome, atransferrinemia, Cornelia de Lange syndrome, Leopard syndrome, Diamond-Blackfan anemia, Steinfeld syndrome, progeria, and Williams-Beuren syndrome. All of these disorders can be treated according to the present invention. Anti-apoptotic factors can be delivered to skeletal muscle, diaphragm muscle and/or cardiac muscle to treat muscle wasting diseases, limb ischemia, cardiac infarction, heart failure, coronary artery disease and/or type I or type II diabetes. Nucleic acids that can be delivered to skeletal muscle include those that are beneficial in the treatment of damaged, degenerated and/or atrophied skeletal muscle. The genetic defects that cause muscular dystrophy are known for many forms of the disease. These defective genes either fail to produce a protein product, produce a protein product that fails to function properly, or produce a dysfunctional protein product that interferes with the proper function of the cell. The heterologous nucleic acid may encode a therapeutically functional protein or a polynucleotide that inhibits production or activity of a dysfunctional protein. Polypeptides that may be expressed from delivered nucleic acids, or inhibited by delivered nucleic acids (e.g., by delivering RNAi, microRNA or antisense RNA), include without limitation dystrophin, a mini-dystrophin or a micro-dystrophin (Duchene's and Becker MD); dystrophin-associated glycoproteins β-sarcoglycan (limb-girdle MD 2E), δ-sarcoglycan (limb-girdle MD 22F), α-sarcoglycan (limb girdle MD 2D) and γ-sarcoglycan (limb-girdle MD 2C), utrophin, calpain (autosomal recessive limb-girdle MD type 2A), caveolin-3 (autosomal-dominant limb-girdle MD), laminin-alpha2 (merosin-deficient congenital MD), miniagrin (laminin-alpha2 deficient congenital MD), fukutin (Fukuyama type congenital MD), emerin (Emery-Dreifuss MD), myotilin, lamin A/C, calpain-3, dysferlin, and/or telethonin. Further, the heterologous nucleic acid can encode mir-1, mir-133, mir-206, mir- 208 or an antisense RNA, RNAi (e.g., siRNA or shRNA) or microRNA to induce exon skipping in a defective dystrophin gene. In particular embodiments, the nucleic acid is delivered to tongue muscle (e.g., to treat dystrophic tongue). Methods of delivering to the tongue can be by any method known in the art including direct injection, oral administration, topical administration to the tongue, intravenous administration, intra-articular administration and the like. The foregoing proteins can also be administered to diaphragm muscle to treat muscular dystrophy. Alternatively, a gene transfer vector may be administered that encodes any other therapeutic polypeptide. In particular embodiments, a virus vector according to the present invention is used to deliver a nucleic acid of interest as described herein to skeletal muscle, diaphragm muscle and/or cardiac muscle, for example, to treat a disorder associated with one or more of these tissues such as muscular dystrophy, heart disease (including PAD and congestive heart failure), and the like. Gene transfer has substantial potential use in 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 inhibitory RNA such as RNAi (e.g., siRNA or shRNA), microRNA or antisense RNA. 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, the virus vectors according to the present invention permit the treatment of genetic diseases. As used herein, a disease state is treated by partially or wholly remedying the deficiency or imbalance that causes the disease or makes it more severe. The use of site-specific recombination of nucleic sequences to cause mutations or to correct defects is also possible. In some embodiments, a disorder treatable by the methods of the present invention may be a blood clotting disorder, including, but not limited to, hemophilia A, hemophilia B, von Willebrand disease, Factor I deficiency, Factor II deficiency, Factor V deficiency, Factor VII deficiency, Factor X deficiency, Factor XI deficiency, Factor XII deficiency, and/or Factor XIII deficiency. In particular embodiments, the nucleic acid is delivered to the liver. Methods of delivering to the liver can be by any method known in the art including injection into the liver, injection into the portal vein, or any combination thereof. As a further aspect, the virus vectors of the present invention may be used to produce an immune response in a subject. According to this embodiment, a virus vector comprising a nucleic acid encoding an immunogen may be administered to a subject, and an active immune response (optionally, a protective immune response) is mounted by the subject against the immunogen. Immunogens are as described hereinabove. Alternatively, the virus vector may be administered to a cell ex vivo and the altered cell is administered to the subject. 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 is optionally expressed and induces an immune response in the subject against the immunogen. In particular 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 immunogens 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." Id. 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 immunogens 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." Id. The virus vectors of the present invention may also be administered for cancer immunotherapy by administration of a viral vector expressing a cancer cell antigen (or an immunologically similar molecule) or any other immunogen that produces an immune response against a cancer cell. To illustrate, an immune response may be produced against a cancer cell antigen in a subject by administering a viral vector comprising a heterologous nucleotide sequence encoding the cancer cell antigen, for example to treat a patient with cancer. The virus vector may be administered to a subject in vivo or by using ex vivo methods, as described herein. 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, leukemia, lymphoma (e.g., Hodgkin and non-Hodgkin lymphomas), colorectal cancer, renal cancer, liver cancer, breast cancer, lung cancer, prostate cancer, testicular cancer, ovarian cancer, uterine cancer, cervical cancer, brain cancer (e.g., gliomas and glioblastoma), bone cancer, sarcoma, melanoma, head and neck cancer, esophageal cancer, thyroid cancer, and the like. In embodiments of the invention, the invention is practiced to treat and/or prevent tumor-forming cancers. 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 representative embodiments, the methods disclosed herein are used to prevent and treat malignant tumors. Cancer cell antigens have been described hereinabove. By the terms "treating cancer" or "treatment of cancer," it is intended that the severity of the cancer is reduced or the cancer is prevented or at least partially eliminated. For example, in particular contexts, these terms indicate that metastasis of the cancer is prevented or reduced or at least partially eliminated. In further representative embodiments these terms indicate that growth of metastatic nodules (e.g., after surgical removal of a primary tumor) is prevented or reduced or at least partially eliminated. By the terms "prevention of cancer" or "preventing cancer" it is intended that the methods at least partially eliminate or reduce the incidence or onset of cancer. Alternatively stated, the onset or progression of cancer in the subject may be slowed, controlled, decreased in likelihood or probability, or delayed. In particular embodiments, cells may be removed from a subject with cancer and contacted with a virus vector according to the present invention. The modified cell is then administered to the subject, whereby an immune response against the cancer cell antigen is elicited. This method is particularly 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., α-interferon, β-interferon, γ-interferon, ^-interferon, ^-interferon, interleukin- 1α, interleukin-1β, 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-α, tumor necrosis factor-β, monocyte chemoattractant protein-1, granulocyte- macrophage colony stimulating factor, and lymphotoxin). Accordingly, immunomodulatory cytokines (e.g., CTL inductive cytokines) may be administered to a subject in conjunction with the virus vectors. Cytokines may be administered by any method known in the art. Exogenous cytokines may be administered to the subject, or alternatively, a nucleotide sequence encoding a cytokine may be delivered to the subject using a suitable vector, and the cytokine produced in vivo. The viral vectors are further useful for targeting liver cells for research purposes, e.g., for study of liver function in vitro or in animals or for use in creating and/or studying animal models of disease. In other embodiments, the viral vector can be used to specifically deliver to oligodendrocytes a toxic agent or an enzyme that produces a toxic agent (e.g., thymidine kinase) in order to kill some or all of the cells. Further, the virus vectors according to the present invention find further use in diagnostic and screening methods, whereby a gene of interest is transiently or stably expressed in a cell culture system, or alternatively, a transgenic animal model. The invention can also be practiced to deliver a nucleic acid for the purposes of protein production, e.g., for laboratory, industrial or commercial purposes. Recombinant virus vectors according to the present invention 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. The term "mammal" as used herein includes, but is not limited to, humans, primates, non-human primates (e.g., monkeys and baboons), cattle, sheep, goats, pigs, horses, cats, dogs, rabbits, rodents (e.g., rats, mice, hamsters, and the like), etc. Human subjects include neonates, infants, juveniles, and adults. Optionally, the subject is "in need of" the methods of the present invention, e.g., because the subject has or is believed at risk for a disorder including those described herein or that would benefit from the delivery of a nucleic acid including those described herein. For example, in particular embodiments, the subject has (or has had) or is at risk for a demyelinating disorder or a spinal cord or brain injury. As a further option, the subject can be a laboratory animal and/or an animal model of disease. In some embodiments, the mammalian subject (e.g., a human patient) may have previously received gene therapy treatment with an AAV particle of a serotype that is not the serotype of the backbone of the capsid, e.g., that is not AAV5 (e.g., a non-AAV5 particle). One aspect of the present invention is a method of transferring a nucleotide sequence to a cell in vitro. The virus vector may be introduced to the cells at the appropriate multiplicity of infection according to standard transduction methods appropriate for the particular target cells. Titers of the virus vector or capsid to administer can vary, depending upon the target cell type and number, and the particular virus vector or capsid, and can be determined by those of skill in the art without undue experimentation. In particular embodiments, at least about 10 3 infectious units, more preferably at least about 10 5 infectious units are introduced to the cell. The cell(s) into which the virus vector can be introduced may 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, oligodendrocytes, glial cells, astrocytes), lung cells, cells of the eye (including retinal cells, retinal pigment epithelium, and corneal cells), epithelial cells (e.g., gut and respiratory epithelial cells), skeletal muscle cells (including myoblasts, myotubes and myofibers), diaphragm muscle cells, dendritic cells, pancreatic cells (including islet cells), hepatic cells, a cell of the gastrointestinal tract (including smooth muscle cells, epithelial cells), heart cells (including cardiomyocytes), bone cells (e.g., bone marrow stem cells), hematopoietic stem cells, spleen cells, keratinocytes, fibroblasts, endothelial cells, prostate cells, joint cells (including, e.g., cartilage, meniscus, synovium and bone marrow), germ cells, and the like. Alternatively, the cell may be any progenitor cell. As a further alternative, the cell can be a stem cell (e.g., neural stem cell, liver stem cell). As still a further alternative, the cell may be a cancer or tumor cell (cancers and tumors are described above). Moreover, the cells can be from any species of origin, as indicated above. The virus vectors may be introduced to cells in vitro for the purpose of administering the modified cell to a subject. In particular embodiments, the cells have been removed from a subject, the virus vector is introduced therein, and the cells are then replaced back into the subject. Methods of removing cells from subject for treatment ex vivo, followed by introduction back into the subject are known in the art (see, e.g., U.S. patent No.5,399,346). Alternatively, the recombinant virus vector is introduced into cells from another subject, into cultured cells, or into cells from any other suitable source, and the cells are administered to a subject in need thereof. Suitable cells for ex vivo gene therapy 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 10 2 to about 10 8 or about 10 3 to about 10 6 cells will be administered per dose in a pharmaceutically acceptable carrier. In particular embodiments, the cells transduced with the virus vector are administered to the subject in an effective amount in combination with a pharmaceutical carrier. In some embodiments, cells that have been transduced with the virus vector may be administered 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 effective amount of the polypeptide in combination with a pharmaceutically acceptable carrier is administered. 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. A further aspect of the invention is a method of administering the virus vectors or capsids of the invention to subjects. In particular embodiments, the method comprises a method of delivering a nucleic acid of interest to an animal subject, the method comprising: administering an effective amount of a virus vector according to the invention to an animal subject. Administration of the virus vectors of the present invention to a human subject or an animal in need thereof can be by any means known in the art. Optionally, the virus vector is delivered in an effective dose in a pharmaceutically acceptable carrier. The virus vectors of the invention can further be administered to a subject to elicit an immunogenic response (e.g., as a vaccine). Typically, vaccines of the present invention comprise an effective amount of virus 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 vectors to be administered to a subject will depend upon the mode of administration, the disease or condition to be treated, the individual subject's condition, the particular virus vector, and the nucleic acid to be delivered, and can be determined in a routine manner. Exemplary doses for achieving therapeutic effects are virus titers of at least about 10 5 , 10 6 , 10 7 , 10 8 , 10 9 , 10 10 , 10 11 , 10 12 , 10 13 , 10 14 , 10 15 transducing units or more, preferably about 10 7 or 10 8 , 10 9 , 10 10 , 10 11 , 10 12 , 10 13 or 10 14 transducing units, yet more preferably about 1012 transducing units. In some embodiments, a therapeutically effective amount of the AAV particle is between about 4×10 12 vg (particles) /kg to about 6 x 10 13 particles/kg, e.g., about 4×10 12 , 5×10 12 , 6×10 12 , 7×10 12 , 8×10 12 , 9×10 12 , 1×10 13 , 2×10 13 , 3×10 13 , 4×10 13 , 5×10 13 , or 6×10 13 or any value or range therein. Exemplary modes of administration include oral, rectal, transmucosal, topical, 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), intro- lymphatic, 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 a 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 on the nature of the particular vector that is being used. In some embodiments, the viral vector is administered directly to the CNS, e.g., the brain or the spinal cord. Direct administration can result in high specificity of transduction of CNS cells, e.g., wherein at least 80%, 85%, 90%, 95% or more of the transduced cells are CNS cells. Any method known in the art to administer vectors directly to the CNS can be used. The vector 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 amygdala), limbic system, neocortex, corpus striatum, cerebrum, and inferior colliculus. The vector may also be administered to different regions of the eye such as the retina, cornea or optic nerve. The vector may be delivered into the cerebrospinal fluid (e.g., by lumbar puncture) for more disperse administration of the vector. The delivery vector may be administered to the desired region(s) of the CNS by any route known in the art, including but not limited to, intrathecal, intracerebral, intraventricular, intranasal, intra-aural, intra-ocular (e.g., intra-vitreous, sub-retinal, anterior chamber) and peri-ocular (e.g., sub-Tenon's region) delivery or any combination thereof. Administration to skeletal muscle according to the present invention includes but is not limited to administration to skeletal muscles 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 muscle tissues 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 superficialis, 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 can be delivered to skeletal muscle by any suitable method including without limitation intravenous administration, intra-arterial administration, intraperitoneal administration, isolated limb perfusion (of leg and/or arm; see, e.g., Arruda et al. (2005) Blood 105:3458-3464), and/or direct intramuscular injection. Administration to cardiac muscle includes without limitation administration to the left atrium, right atrium, left ventricle, right ventricle and/or septum. The virus vector can be delivered to cardiac muscle by any method known in the art including, e.g., 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 any of these tissues can also be achieved by delivering a depot comprising the virus vector, which can be implanted into the skeletal, cardiac and/or diaphragm muscle tissue or the tissue can be contacted with a film or other matrix comprising the virus vector. Examples of such implantable matrices or substrates are described in U.S. Patent No. 7,201,898). In particular embodiments, a virus vector according to the present invention is administered to skeletal muscle, diaphragm muscle and/or cardiac muscle (e.g., to treat muscular dystrophy or heart disease [for example, PAD or congestive heart failure]). The invention can be used to treat disorders of skeletal, cardiac and/or diaphragm muscle. Alternatively, the invention can be practiced to deliver a nucleic acid to skeletal, cardiac and/or diaphragm muscle, which is used as a platform for production of a protein product (e.g., an enzyme) or non-translated RNA (e.g., RNAi, microRNA, antisense RNA) that normally circulates in the blood or for systemic delivery to other tissues to treat a disorder (e.g., a metabolic disorder, such as diabetes (e.g., insulin), hemophilia (e.g., Factor IX or Factor VIII), or a lysosomal storage disorder (such as Gaucher's disease [glucocerebrosidase], Pompe disease [lysosomal acid α-glucosidase] or Fabry disease [α- galactosidase A]) or a glycogen storage disorder (such as Pompe disease [lysosomal acid α glucosidase]). Other suitable proteins for treating metabolic disorders are described above. 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. In some embodiments, a single administration (e.g., a single intravenous dose) may be employed to achieve the desired level of gene expression, e.g., to deliver a blood clotting factor such as FVa and/or FVIII for the treatment of hemophilia. In some embodiments, a single administration at a dose of between about 4×10 12 vg (particles) /kg to about 6 x 10 13 particles/kg (e.g., about 4×10 12 , 5×10 12 , 6×10 12 , 7×10 12 , 8×10 12 , 9×10 12 , 1×10 13 , 2×10 13 , 3×10 13 , 4×10 13 , 5×10 13 , or 6×10 13 or any value or range therein) may be employed to achieve the desired level of gene expression, e.g., to deliver a blood clotting factor such as FVa and/or FVIII for the treatment of hemophilia. Pharmaceutical compositions suitable for oral administration can be presented in discrete units, such as capsules, cachets, lozenges, or tablets, each containing a predetermined amount of the composition of this invention; as a powder or granules; as a solution or a suspension in an aqueous or non-aqueous liquid; or as an oil-in-water or water-in-oil emulsion. Oral delivery can be performed by complexing a virus vector of the present invention to a carrier capable of withstanding degradation by digestive enzymes in the gut of an animal. Examples of such carriers include plastic capsules or tablets, as known in the art. Such formulations are prepared by any suitable method of pharmacy, which includes the step of bringing into association the composition and a suitable carrier (which may contain one or more accessory ingredients as noted above). In general, the pharmaceutical composition according to embodiments of the present invention are prepared by uniformly and intimately admixing the composition with a liquid or finely divided solid carrier, or both, and then, if necessary, shaping the resulting mixture. For example, a tablet can be prepared by compressing or molding a powder or granules containing the composition, optionally with one or more accessory ingredients. Compressed tablets are prepared by compressing, in a suitable machine, the composition in a free-flowing form, such as a powder or granules optionally mixed with a binder, lubricant, inert diluent, and/or surface active/dispersing agent(s). Molded tablets are made by molding, in a suitable machine, the powdered compound moistened with an inert liquid binder. Pharmaceutical compositions suitable for buccal (sub-lingual) administration include lozenges comprising the composition of this invention in a flavored base, usually sucrose and acacia or tragacanth; and pastilles comprising the composition in an inert base such as gelatin and glycerin or sucrose and acacia. Pharmaceutical compositions suitable for parenteral administration can comprise sterile aqueous and non-aqueous injection solutions of the composition of this invention, which preparations are optionally isotonic with the blood of the intended recipient. These preparations can contain anti-oxidants, buffers, bacteriostats and solutes, which render the composition isotonic with the blood of the intended recipient. Aqueous and non-aqueous sterile suspensions, solutions and emulsions can include suspending agents and thickening agents. Examples of non-aqueous solvents are propylene glycol, polyethylene glycol, vegetable oils such as olive oil, and injectable organic esters such as ethyl oleate. Aqueous carriers include water, alcoholic/aqueous solutions, emulsions or suspensions, including saline and buffered media. Parenteral vehicles include sodium chloride solution, Ringer's dextrose, dextrose and sodium chloride, lactated Ringer's, or fixed oils. Intravenous vehicles include fluid and nutrient replenishers, electrolyte replenishers (such as those based on Ringer's dextrose), and the like. Preservatives and other additives may also be present such as, for example, antimicrobials, anti-oxidants, chelating agents, and inert gases and the like. The compositions can be presented in unit/dose or multi-dose containers, for example, in sealed ampoules and vials, and can be stored in a freeze-dried (lyophilized) condition requiring only the addition of the sterile liquid carrier, for example, saline or water-for- injection immediately prior to use. Extemporaneous injection solutions and suspensions can be prepared from sterile powders, granules and tablets of the kind previously described. For example, an injectable, stable, sterile composition of this invention in a unit dosage form in a sealed container can be provided. The composition can be provided in the form of a lyophilizate, which can be reconstituted with a suitable pharmaceutically acceptable carrier to form a liquid composition suitable for injection into a subject. The unit dosage form can be from about 1 µg to about 10 grams of the composition of this invention. When the composition is substantially water- insoluble, a sufficient amount of emulsifying agent, which is physiologically acceptable, can be included in sufficient quantity to emulsify the composition in an aqueous carrier. One such useful emulsifying agent is phosphatidyl choline. Pharmaceutical compositions suitable for rectal administration can be presented as unit dose suppositories. These can be prepared by admixing the composition with one or more conventional solid carriers, such as for example, cocoa butter and then shaping the resulting mixture. Pharmaceutical compositions of this invention suitable for topical application to the skin can take the form of an ointment, cream, lotion, paste, gel, spray, aerosol, or oil. Carriers that can be used include, but are not limited to, petroleum jelly, lanoline, polyethylene glycols, alcohols, transdermal enhancers, and combinations of two or more thereof. In some embodiments, for example, topical delivery can be performed by mixing a pharmaceutical composition of the present invention with a lipophilic reagent (e.g., DMSO) that is capable of passing into the skin. Pharmaceutical compositions suitable for transdermal administration can be in the form of discrete patches adapted to remain in intimate contact with the epidermis of the subject for a prolonged period of time. Compositions suitable for transdermal administration can also be delivered by iontophoresis (see, for example, Pharm. Res.3:318 (1986)) and typically take the form of an optionally buffered aqueous solution of the composition of this invention. Suitable formulations can comprise citrate or bis\tris buffer (pH 6) or ethanol/water and can contain from 0.1 to 0.2M active ingredient. The virus vectors disclosed herein may be administered to the lungs of a subject by any suitable means, for example, by administering an aerosol suspension of respirable particles comprised of the virus vectors, which the subject inhales. The respirable particles may be liquid or solid. Aerosols of liquid particles comprising the virus vectors 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. Patent No. 4,501,729. Aerosols of solid particles comprising the virus vectors may likewise be produced with any solid particulate medicament aerosol generator, by techniques known in the pharmaceutical art. The invention will now be described with reference to the following examples. It should be appreciated that these examples are not intended to limit the scope of the claims to the invention but are rather intended to be exemplary of certain embodiments. Any variations in the exemplified methods that occur to the skilled artisan are intended to fall within the scope of the invention. EXAMPLES 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. EXAMPLE 1: Anti-AAV5 antibody pre-exist less in the human body as compared to other AAV serotypes, suggesting that AAV5 would be a good choice in theory for AAV gene therapy. However, AAV5 serotype has poor infectivity in vitro and in vivo, which limits its application in clinical settings. To overcome the shortcomings of wildtype AAV5 and increase target gene expression in vitro and in vivo, it is necessary to modify AAV5’s capsid for more powerful infectivity. The invention focused on modifying the capsid gene of AAV5 to increase infectivity of an AAV5 vector. A two-step method was created to enhance AAV5 hepatocytic tropism. The first step was replacement of VP1 and VP2 of AAV5 with that of AAV1, AAV2, AAV3, AAV4, AAV6, AAV7, AAV8 or AAV9’s, while keeping the AAV5 VP3 unchanged. The new AAV vectors were named AAV5n (wherein n = the AAV serotype of the replacement VP1 and VP2, e.g., AAV51 being AAV5 capsid protein with replaced VP1 and VP2 from AAV1). The modified AAV5n vectors increased targeting gene expression in vitro but not in vivo. To improve the tissue tropism of AAV5n capsid in vivo, a second step was performed to generate a modified VP3. Because the capsid tissue tropism is mainly determined by variable regions (VR), an extra copy of VR-I from either AAV6 or AAV8 was inserted into VR-VIII of AAV59, so called AAV596 (with AAV6 VR-I inserted into the VP3 VR-VIII of AAV59) and AAV598 (with AAV8 VR-I inserted into the VP3 VR-VIII of AAV59), respectively. The insertions resulted in an increase of AAV5 vector infectivity and liver tropism. AAV596 and AAV598 greatly increased AAV5 liver tropism in vivo. For example, AAV596 increased LacZ expression about 13.4 and about 7.5-fold, respectively, compared with original AAV5 vector and AAV59 vector. Furthermore, the modified AAV596 and AAV598 were successfully applied to mediate the expression of FVIII in a hemophilia-A mouse model, and FVIII activity in plasma was increased 38.9- and 10.8-fold over AAV5, respectively, and increased 20.6-and 5.7-fold over AAV59, respectively, while their antigenicity was kept unchanged. Thus, the invention indicates the novel AAV5 capsids modified with two-step method could be regarded as ideal vectors for gene therapy of hemophilia A disease. EXAMPLE 2: Construction of the modified AAV5 vectors. Several pairs of primers were synthesized to amplify the sequences of VP1 and VP2 from AAV1, AAV2, AAV3, AAV4, AAV6, AAV7, AAV8 and AAV9 as inserts by PCR, which were used to replace the VP1 and VP2 of AAV5 by blunt ligation. FIGS.1A-1I show the construction of the modified AAV5n vectors. VP1 and VP2 of AAV5 were replaced individually by the VP1 and VP2 of AAV1, AAV2, AAV3, AAV4, AAV6, AAV7, AAV8 and AAV9. VP3 of AAV5 was kept unchanged at this step. All AAV vectors were produced by triple plasmids transfection in Human Embryonic Kidney (HEK) 293 cells. The AAV vector plasmid (containing the gene of LacZ or FVIII or FIX), AAV helper plasmid, and Ad helper plasmid AAV5n were transiently transfected into HEK293 cells. Forty-eight hours after transfection, the HEK293 cells were harvested, and freeze-thawed three times. The viruses were purified through polyethylene glycol (PEG8000) precipitation, followed by two courses of CsCl gradient ultracentrifugation in an OptimaTM L-100XP ultracentrifuge (Beckman Coulter, Indianapolis, IN). The AAV viruses were titered by standard dot-blot assay. The viral vectors were diluted to 5.0 ×10 12 viral genomes per milliliter (vg/ml). Western blots were used to identify whether the VP proteins VP1, VP2 and/or VP3 of AAV5n vectors were normally expressed and assembled into viruses. Briefly, AAV5n viruses (a total 1×10 10 vg/channel) were lysed in ice-cold Radio Immuno Precipitation Assay (RIPA) buffer (150 mmol/l NaCl, 1% Triton X-100, 50 mmol/l Tris, PH 8.0, 0.1% SDS, 0.5% sodium deoxycholate) plus protease inhibitor cocktail. All samples were separated in an 8% SDS-polyacrylamide gel electrophoresis (PAGE). Then, the gels were electrophoretically transferred to polyvinylidene fluoride (PVDF) membranes, which were next pre-incubated in 5% skim milk dissolved in Tris-Buffered Saline and Tween- 20 (TBST: 10 mM Tris-Cl, 150 M NaCl and 0.05% Tween-20, pH 7.6). Then, the membranes were incubated with mouse primary anti-VP antibody (1:500) overnight at 4℃. Afterwards, the membranes were incubated with goat anti-mouse secondary antibody (1:3000, Abcam, Cambridge, MA) conjugated with Horse Radish Peroxidase (HRP) at RT for one hour and washed with TBST. The membranes were developed with horseradish peroxidase/enhanced chemiluminescence (PerkinElmer, Waltham, MA) and were photographed and analyzed with the FluorChem M MultiFluor system. From these AAV5n vectors, AAV52, AAV57, AAV58 and AAV59 were used as examples, where their VP1, VP2 and VP3 proteins could be expressed successfully (FIG.2). VP2 of AAV57 and AAV58 included two bands, similar to wildtype AAV7 and AAV8. EXAMPLE 3: AAV5n vectors enhanced target gene expression in vitro. To identify whether the modified AAV5n vectors had acquired enhanced infectivity, HEK293 cell and Huh-7 cell line were infected with purified AAV5n viruses in vitro. The dose of vectors was administrated with high dose and low dose according to MOI (high dose: MOI=1×10 5 vg/cell; low dose: MOI=2×10 4 vg/cell). After 72h, the cells were washed with PBS for three times and then first fixed for five minutes in cold fixative solution (2% formaldehyde and 0.2% glutaraldehyde diluted in cold PBS). The cells were washed gently with PBS three times, followed by staining at 37℃ overnight in X-gal solution (1 mg/mL X- gal, 5 mM MgCl2, 5 mM potassium ferricyanide and 5 mM potassium ferrocyanide in PBS). FIG.3A shows LacZ expression in HEK293 cells mediated by AAV5n vectors with CB promoter. LacZ expression mediated by AAV52, AAV57, AAV58 and AAV59, for example, was enhanced significantly compared with AAV5 treatment. The left panel in FIG.3A shows high-MOI treatment (MOI=1×10 5 vg/cell), and the right panel shows low-MOI treatment (MOI=2×10 4 vg/cell). The quantitative LacZ enzyme activity assays were carried out using the Galacto- Light PlusTM System (Applied Biosystems, Bedford, MA) according to the manufacturer's instructions. Cells were lysed with lysis solution, and then centrifuged for two min to pellet debris. Then, 10 µl of the supernatant was transferred to microplate wells and incubated with 70 µl of reaction buffer for one hour. After injecting 100 µl of Accelerator-II, the signal was read with microplate luminometers. The LacZ activity was expressed as relative light units (RLU) per milligram of total protein (RLU/mg protein). FIG.3B shows the activity of LacZ mediated by AAV5n vectors increased 4.5-7.7 times compared with AAV5 in HEK293 cells. FIG.4A shows LacZ expression mediated by AAV52, AAV57, AAV58 and AAV59 was enhanced significantly compared with AAV5-treatment in different MOI treatment in vitro in the human liver cell line Huh-7, which is a well differentiated hepatocyte-derived carcinoma cell. The left panel shows high-MOI treatment (MOI=1×10 5 vg/cell), and the right panel shows low-MOI treatment (MOI=2×10 4 vg/cell). In the FIG.4B, the activity of LacZ in Huh-7 cells mediated by low MOI of AAV5n vectors was increased significantly by 8.8-9.6 times compared with AAV5 treatment. EXAMPLE 4: AAV5n vectors did not present powerful infectivity in vivo. AAV5n viruses (AAV52 and AAV59 vectors as examples) were injected in vivo into C57B6 mice by tail vein (3×10 11 vg/mouse, n=4/group). After 2 weeks, the livers were harvested and frozen into liquid nitrogen. The tissue sections were performed with 20 μm thickness. The cryosections were first fixed for five minutes in cold fixative solution (2% formaldehyde and 0.2% glutaraldehyde diluted in cold PBS). The sections were washed gently with PBS three times, followed by staining at 37℃ overnight in X-gal solution (1 mg/mL X-gal, 5 mM MgCl2, 5 mM potassium ferricyanide and 5 mM potassium ferrocyanide in PBS). FIG.5A shows LacZ expression mediated by AAV5n vectors with CB promoter in the liver of C57/B6 mice. LacZ expression mediated by AAV5n vectors was not significantly enhanced compared with original AAV5 vector treatment. The upper-panel of FIG.5B shows low power pictures, and the lower-panel shows high power pictures. In summary, AAV5n vectors significantly enhanced the target gene expression in vitro, but they did not greatly enhance their infectivity in vivo. Therefore, it was necessary to modify the capsid of AAV5n for higher infectivity in vivo. EXAMPLE 5: Remodeling the construction of the modified AAV5n vectors. A pair of primers were synthesized to amplify the sequences of VP1 and VP2 from AAV9 as an insert by PCR, which was used to replace the VP1 and VP2 of AAV5 by blunt ligation. FIG.1 shows the construction of the modified AAV59 vector, whose VP1 and VP2 were replaced by the VP1 and VP2 of AAV9, and whose VP3 was kept unchanged (i.e., the VP3 of AAV5). The VP3 capsid protein was then modified by inserting the sequences of variable region I (VR-I) of either AAV6 or AAV8 into VR-VIII at site Q574 (AAV596 and AAV598; FIG.6A) of AAV5 capsid plasmid. AAV596 was inserted with the VR-I sequence of AAV6 at VR-VIII (Q574), based on AAV59. Similarly, AAV598 was inserted with the VR-I sequence of AAV8 at VR-VIII (Q574), based on AAV59. The partial sequences of the VP3 of AAV5 and the modified vectors (amino acid positions 546-625) are shown in FIG. 6B. FIG.7 shows the construction of the VP3 of AAV5 and the modified vectors AAV596 and AAV598 in three models including line model, cartoon model and surface model. As can be seen in the models, the VR-VIII of AAV5/AAV59 physically sticks out of AAV capsids. It can be seen that insertion of VR-I from AAV6 or AAV8 changes the surface conformation of the VR-VIII of AAV5. While not wishing to be bound to theory, this physical conformation may change the tissue tropism of a vector. Next, all AAV vectors were produced by calcium phosphate-mediated triple plasmids co-transfection in HEK293 cells. The AAV vector plasmid (containing the gene of LacZ or FVIII), AAV helper plasmid, Ad helper plasmid, and AAV596 and AAV598 were transiently transfected into HEK293 cells. Forty-eight hours after transfection, the HEK293 cells were harvested, and frozen-thawed three times. The viruses were purified through polyethylene glycol (PEG8000) precipitation, followed by two courses of CsCl gradient ultracentrifugation in an OptimaTM L-100XP ultracentrifuge (Beckman Coulter, Indianapolis, IN; FIG.8). The AAV viruses were titered by standard dot-blot assay. The viral vectors were diluted to 5.0 ×10 12 viral genomes per milliliter (vg/ml). EXAMPLE 6: Modified AAV5n vectors enhanced target gene expression in vitro. To identify whether the VP3-modified AAV596 and AAV598 enhanced infectivity of the vectors, Huh-7 cell line were infected with purified AAV viruses packaging LacZ gene with CB promoter. The dose of vectors was administrated according to MOI (MOI = 5×10 5 vg/cell). In FIG.9A, the modified AAV5 vectors AAV596 and AAV598 greatly enhanced LacZ expression in Huh-7 cells compared with original AAV5 vector. The result of LacZ activity also showed that AAV596 and AAV598 inserted with the VR-I sequence of AAV6 or AAV8 had increased target gene expression of about 7.9 or 20.6 fold as compared to AAV5 in vitro, respectively (FIG.9B). EXAMPLE 7: AAV596 and AAV598 vectors significantly increased target gene expression in vivo. The modified AAV5 viruses were injected in vivo into C57B6 mice by tail vein (3×10 11 /mouse, n=4/group). After two weeks, the livers were harvested and frozen into liquid nitrogen. FIG.10 shows LacZ expression in the liver of C57B6 mice mediated by AAV5 vectors with CB promoter. The modified AAV5n vectors, especially AAV596, significantly increased liver tropism compared with original AAV5 vector and AAV59 vector, while there were no significant increases in heart and skeletal muscles GAS. Next, liver tissues were homogenized with lysis buffer and centrifuged for supernatant, which were used for measuring the LacZ activity. FIG.11 shows the activity of LacZ mediated by the modified AAV5 vectors in liver and other tissues. The results show the modified vectors AAV596 and AAV598 increased LacZ expression in liver. AAV596 significantly increased liver tropism up to 13.4 or 7.5 fold, compared with original AAV5 vector and AAV59 vector, respectively. Furthermore, in these organs the LacZ expression level was very low, and there are no significant differences between groups in heart, GAS, lung, intestine, kidney, spleen and pancreas, indicating that the modified AAV5n vectors significantly increased their liver targeting. EXAMPLE 8: AAV596 and AAV598 vectors mediate high level gene expression of FVIII in a hemophilia-A mouse model. The modified AAV5n vectors packaged with FVIII were injected into the hemophilia-A mice by tail vein (4×10 12 vg/kg, n=5/group). After two weeks, the FVIII activity of blood was measured by FVIII-specific ELISA. FIG.12 shows AAV596 mediated FVIII activity was significantly enhanced 38.9 and 10.8 fold compared with AAV5 or AAV59, respectively. AAV598-mediated FVIII expression was also enhanced by 10.8- and 5.7-fold compared with AAV5 or AAV59, respectively. Thus the two modified AAV5 vectors, especially AAV596, could mediate FVIII expression well in vivo, and could be used as ideal gene therapy tools to treat hemophilia A disease. EXAMPLE 9: AAV596 and AAV598 vectors presented the ability of immunologic escape. Intravenous immunoglobulin (IVIG) contains the pooled immunoglobulin G (IgG) from the plasma of thousands of blood donors. The average level of AAV neutralization in IVIG represents the repertoire of anti-AAV antibodies with heterogeneous specificities and affinities in the population. In the present invention, IVIG did not inhibit AAV596 and AAV598 mediated LacZ expression (FIG.13), thus demonstrating that the modified AAV5n vectors are significantly more resistant to neutralization by IVIG than AAV9. AAV596 (1:80) and AAV598 (1:20) showed an 8- and 32-fold greater resistance to neutralization than AAV9 (1:640). Thus, AAV596 and AAV598 vectors could be inhibited by IVIG, indicating that they have the ability of immunologic escape. EXAMPLE 10: Additional methods used in Examples 1-9. AAV capsid modification. The replacement of AAV5 VP1/2 with other serotypes was performed by blunt ligations with pairs of PCR products. In detail, the backbone including AAV5 VP3 was amplified by one pair of primers (forward primer, 5’- pATGTCTGCGGGAGGTGGCGGC-3’ (SEQ ID NO:30) and reverse primer: 5’- pACTCACCAGTCACAGAAAAGCATC 3’ (SEQ ID NO:31)). The insertions of VP1 and VP2 from AAV2, AAV7, AAV8 and AAV9 were generated by PCR with primers (common forward primer: 5’- p’ACTCAACCAAGTCATTCTGAGAATAG-3’ (SEQ ID NO:32); reverse primer of AAV2 VP1 and VP2: 5’- pCGTATTAGTTCCCAGACCAGAGG-3’ (SEQ ID NO:33); reverse primer of AAV7 VP1 and VP2: 5’ pTGTACCAGATCCCACACTAGAGGG-3’ (SEQ ID NO:34); reverse primer of AAV8 VP1 and VP2: 5’-pTGTATTAGGTCCCACACCAGAGG-3’ (SEQ ID NO:35); reverse primer of AAV9 VP1 and VP2: 5’- pTGTAAGAGATCCCACACCTGAGG-3’ (SEQ ID NO:36)). By ligating the backbones and the inserts with T4 DNA ligase (Promega, Madison, WI), AAV5n (AAV52, AAV57, AAV58 and AAV59) were produced successfully. The plasmid of AAV596 was constructed by inverse PCR, based on the plasmid of AAV59 with one pair of primers (forward primer: 5’-p GGGGCCAGCTCCACCACTGCCCCCGC-3’ (SEQ ID NO:37) and reverse primer: 5’- pCGTTGAAGCACTCTGGTTGTTGGTGGCCATCTG-3’ (SEQ ID NO:38)). Human- derived FVIII gene, driven by one liver promoter lpx2.1, was synthetized with B-domain deletion, as described in US 2016/0229904, incorporated herein by reference. AAV packaging and purification. All AAV vectors were produced by triple plasmids transduction in Human Embryonic Kidney (HEK) 293 cells. The AAV vector plasmid (containing the gene of LacZ or FVIII), AAV helper plasmid, and Ad helper plasmids pXX2, pXX5, pXX7, pXX8 or pXX9 were transiently transfected into HEK293 cells. The pAAV2.1-CB-LacZ-cyt vector plasmid was used to express LacZ in the cytoplasm. The pXX-lpx2.1-FVIII-ER vector plasmid was also constructed by insertion with human FVIII gene. Forty-eight hours after transfection, the HEK293 cells were harvested, and frozen/thawed three times. All viruses were purified through polyethylene glycol (PEG8000) precipitation, followed by two courses of CsC11radient ultracentrifugation in an OptimaTM L-100XP ultracentrifuge (Beckman Coulter, Indianapolis, IN). The AAV viruses were titered by standard dot-blot assay. The viral vectors were diluted to 2.0 to 6.0 × 10 12 viral genomes per milliliter (vg/ml). Cell culture and infection. HEK293 cell line and Huh-7 cell line were cultured in DMEM medium plus 10% FBS in an incubator with 5% CO2 at 37°C. To investigate the infectivity of different novel AAV vectors in vitro, the purified viruses packaged with CB- LacZ were added into the medium according to MOI (high MOI: 1×10 5 vg/cell; low MOI: 2×10 4 vg/cell). To measure the FVIII activity mediated by AAV5, AAV59 and AAV596 in vitro, Huh-7 cells were infected with the vectors (MOI=5×10 5 vg/cell). Animals and vector administration. Mice were divided into groups according to randomization principle. AAV vectors packaged with LacZ were injected by tail vein into 8- week old C57BL/6 mice (1.2×10 13 vg/kg). After two weeks, the tissues were harvested and frozen at -80 ℃. AAV-lpx2.1-FVIII vectors were delivered into FVIIIKO mice by i.v. injection (4×10 12 vg/kg). Blood samples were collected by retro-orbital bleeding at three time points (2, 4 and 10 weeks after virus injection), and mixed with sodium citrate (3.2%) and stored at -80 ℃ for analysis. Western blot. AAV vectors (1×10 10 vg/channel) were lysed in ice-cold Radio Immuno Precipitation Assay (RIPA) buffer (150 mmol/l NaCl, 1% Triton X-100, 50 mmol/l Tris, pH 8.0, 0.1% SDS, 0.5% sodium deoxycholate) plus protease inhibitor cocktail (Sigma-Aldrich, St. Louis, MO). After boiling for 10 minutes, the vectors were put on ice. Samples were separated in an 8% SDS-polyacrylamide gel electrophoresis (PAGE) for VP proteins. Gels were electrophoretically transferred to polyvinylidene fluoride (PVDF, Bio-Rad, Hercules, CA) membranes, which were next pre-incubated in 5% skim milk (Bio-Rad, Hercules, CA) dissolved in Tris-Buffered Saline and Tween-20 (TBST: 10 mM Tris-Cl, 150 M NaCl and 0.05% Tween-20, pH 7.6). Then, the membranes were incubated with primary antibodies including polyclonal mouse anti-VP antibody (1:500 Abcam, Cambridge, MA) overnight at 4℃. Afterwards the membranes were incubated with goat anti-mouse secondary antibody (1:3000, Abcam, Cambridge, MA) conjugated with Horse Radish Peroxidase (HRP) at RT for one hour and washed with TBST. The membranes were developed with horseradish peroxidase/enhanced chemiluminescence (PerkinElmer, Waltham, MA) and were photographed and analyzed with the FluorChem M MultiFluor system (Cell & Biosciences, Santa Clara, CA). X-gal staining and LacZ activity. In vitro, 72h later after infection with AAV-CB- LacZ vectors, the cultured cells were first washed with PBS for three times and then fixed for five minutes in cold fixative solution (2% formaldehyde and 0.2% glutaraldehyde diluted in cold PBS). Subsequently, the cells were washed gently with PBS three times, followed by staining at 37℃ overnight in X-gal solution (1mg/mL X-gal, 5 mM MgC12, 5 mM potassium ferricyanide and 5 mM potassium ferrocyanide in PBS). In vivo, two weeks after AAV injection, the livers were harvested and frozen into liquid nitrogen. The tissue sections were performed with 20 μm thickness. The cryosections were first fixed for five minutes in cold fixative solution (2% formaldehyde and 0.2% glutaraldehyde diluted in cold PBS). Subsequently, the cells were washed gently with PBS three times, followed by staining at 37℃ overnight in X-gal solution (1 mg/mL X-gal, 5 mM MgCl2, 5 mM potassium ferricyanide and 5 mM potassium ferrocyanide in PBS). The quantitative LacZ enzyme activity assays were carried out using the Galacto- Light PlusTM System (Applied Biosystems, Bedford, MA) according to the manufacturer's instructions. The cells or tissues were lysed with lysis solution, and then centrifuged for 10 minutes to pellet debris. Then, 10 µl of the supernatant was transferred to microplate wells, and incubated with 70 µl of reaction buffer for one hour. After injection of 100 µl Accelerator-II, the signal was read with microplate luminometers. The LacZ activity was expressed as relative light units (RLU) per milligram of total protein (RLU/mg protein). FVIII activity. After blood plasma samples were collected, the FVIII activity was quantified with one-stage FVIII activity assay (FVIII-specific aPTT), recorded on the STart 4 coagulation analyzer (Diagnostica Stago). 50 mL plasma samples diluted in Owren-Koller buffer (Diagnostica Stago) were incubated with 50 mL hFVIII-deficient plasma (George King Bio-Medical, Overland Park, KS) and 50 mL aPTT reagents for three minutes, followed by adding 50 mL 0.25 M calcium chloride. Recombinant human factor VIII (Advate®; Baxter, Westlake Village, CA) was used to make a standard curve. IVIG inhibition: AAV-CB-LacZ (4 × 10 9 vg/well) was first diluted with DMEM medium (serum-free), then preincubated with 2-fold serial dilutions of IVIG (initial dilution of IVIG, 1:20; Carimune NF; ZLB Behring) on DMEM at 37°C. After one hour, the serum- vector mixture was added to 96-well plates seeded with Huh7 (4 × 10 4 cells/well) and incubated at 37°C and 5% CO2 for another one hour. Subsequently, each well was gently supplemented with an equal volume of DMEM with 20% FBS and put back to the cell culture incubator with 5% CO2 at 37°C. After 24 hours, cells were washed two times with PBS and lysed with lysis solution. The LacZ activity was measured with the Galacto-Light PlusTM System (Applied Biosystems, Bedford, MA). The titer of NAbs was recorded as the highest serum dilution which could inhibit the infection of AAV-CB-LacZ by ≥50%, compared with the untreated control. Statistical analysis. All values are expressed as means ±SE. Student’s t-tests were adopted when comparing two groups. When comparing three groups or more, one-way ANOVA analysis of variance plus Dunnett post-test was performed by using SPSS 13.0 (IBM, Armonk, New York). P< 0.05 was accepted as statistically significant.

Table 1. Table 2.

Table 3. SEQUENCES SEQ ID NO:1 WT AAV5 VP1/VP2 (192AA; YP_068409.1) MSFVDHPPDWLEEVGEGLREFLGLEAGPPKPKPNQQHQDQARGLVLPGYNYLGPGNGLDR G EPVNRADEVAREHDISYNEQLEAGDNPYLKYNHADAEFQEKLADDTSFGGNLGKAVFQAK KRVLEPFGLVEEGAKTAPTGKRIDDHFPKRKKARTEEDSKPSTSSDAEAGPSGSQQLQIP AQP ASSLGADT SEQ ID NO:2 WT AAV5 VP3 (532AAs; YP_068409.1; 574Q underlined) MSAGGGGPLGDNNQGADGVGNASGDWHCDSTWMGDRVVTKSTRTWVLPSYNNHQYREI KSGSVDGSNANAYFGYSTPWGYFDFNRFHSHWSPRDWQRLINNYWGFRPRSLRVKIFNIQ V KEVTVQDSTTTIANNLTSTVQVFTDDDYQLPYVVGNGTEGCLPAFPPQVFTLPQYGYATL NR DNTENPTERSSFFCLEYFPSKMLRTGNNFEFTYNFEEVPFHSSFAPSQNLFKLANPLVDQ YLY RFVSTNNTGGVQFNKNLAGRYANTYKNWFPGPMGRTQGWNLGSGVNRASVSAFATTNRM ELEGASYQVPPQPNGMTNNLQGSNTYALENTMIFNSQPANPGTTATYLEGNMLITSESET QP VNRVAYNVGGQMATNNQSSTTAPATGTYNLQEIVPGSVWMERDVYLQGPIWAKIPETGAH FHPSPAMGGFGLKHPPPMMLIKNTPVPGNITSFSDVPVSSFITQYSTGQVTVEMEWELKK ENS KRWNPEIQYTNNYNDPQFVDFAPDSTGEYRTTRPIGTRYLTRPL SEQ ID NO:3 AAV2 VR-I SQSGAS SEQ ID NO:4 AAV6 VR-I SASTGA SEQ ID NO:5 AAV7 VR-I SETAGST SEQ ID NO:6 AAV8 VR-I NGTSGGAT SEQ ID NO:7 AAV9 VR-I NSTSGGSS SEQ ID NO:8 [AAV51 VP1/2 and VP3] MAADGYLPDWLEDNLSEGIREWWDLKPGAPKPKANQQKQDDGRGLVLPGYKYLG PFNGLDKGEPVNAADAAALEHDKAYDQQLKAGDNPYLRYNHADAEFQERLQEDTS FGGNLGRAVFQAKKRVLEPLGLVEEGAKTAPGKKRPVEQSPQEPDSSSGIGKTGQQP AKKRLNFGQTGDSESVPDPQPLGEPPATPAAVGPTTMSAGGGGPLGDNNQGADGVG NASGDWHCDSTWMGDRVVTKSTRTWVLPSYNNHQYREIKSGSVDGSNANAYFGY STPWGYFDFNRFHSHWSPRDWQRLINNYWGFRPRSLRVKIFNIQVKEVTVQDSTTTI ANNLTSTVQVFTDDDYQLPYVVGNGTEGCLPAFPPQVFTLPQYGYATLNRDNTENP TERSSFFCLEYFPSKMLRTGNNFEFTYNFEEVPFHSSFAPSQNLFKLANPLVDQYLYR FVSTNNTGGVQFNKNLAGRYANTYKNWFPGPMGRTQGWNLGSGVNRASVSAFAT TNRMELEGASYQVPPQPNGMTNNLQGSNTYALENTMIFNSQPANPGTTATYLEGNM LITSESETQPVNRVAYNVGGQMATNNQSSTTAPATGTYNLQEIVPGSVWMERDVYL QGPIWAKIPETGAHFHPSPAMGGFGLKHPPPMMLIKNTPVPGNITSFSDVPVSSFITQY STGQVTVEMEWELKKENSKRWNPEIQYTNNYNDPQFVDFAPDSTGEYRTTRPIGTR YLTRPL SEQ ID NO:9 [AAV52 VP1/2 and VP3] MAADGYLPDWLEDTLSEGIRQWWKLKPGPPPPKPAERHKDDSRGLVLPGYKYLGPF NGLDKGEPVNEADAAALEHDKAYDRQLDSGDNPYLKYNHADAEFQERLKEDTSFG GNLGRAVFQAKKRVLEPLGLVEEPVKTAPGKKRPVEHSPVEPDSSSGTGKAGQQPA RKRLNFGQTGDADSVPDPQPLGQPPAAPSGLGTNTMSAGGGGPLGDNNQGADGVG NASGDWHCDSTWMGDRVVTKSTRTWVLPSYNNHQYREIKSGSVDGSNANAYFGY STPWGYFDFNRFHSHWSPRDWQRLINNYWGFRPRSLRVKIFNIQVKEVTVQDSTTTI ANNLTSTVQVFTDDDYQLPYVVGNGTEGCLPAFPPQVFTLPQYGYATLNRDNTENP TERSSFFCLEYFPSKMLRTGNNFEFTYNFEEVPFHSSFAPSQNLFKLANPLVDQYLYR FVSTNNTGGVQFNKNLAGRYANTYKNWFPGPMGRTQGWNLGSGVNRASVSAFAT TNRMELEGASYQVPPQPNGMTNNLQGSNTYALENTMIFNSQPANPGTTATYLEGNM LITSESETQPVNRVAYNVGGQMATNNQSSTTAPATGTYNLQEIVPGSVWMERDVYL QGPIWAKIPETGAHFHPSPAMGGFGLKHPPPMMLIKNTPVPGNITSFSDVPVSSFITQY STGQVTVEMEWELKKENSKRWNPEIQYTNNYNDPQFVDFAPDSTGEYRTTRPIGTR YLTRPL SEQ ID NO:10 [AAV53 VP1/2 and VP3] MAADGYLPDWLEDNLSEGIREWWALKPGVPQPKANQQHQDNRRGLVLPGYKYLG PGNGLDKGEPVNEADAAALEHDKAYDQQLKAGDNPYLKYNHADAEFQERLQEDTS FGGNLGRAVFQAKKRILEPLGLVEEAAKTAPGKKGAVDQSPQEPDSSSGVGKSGKQ PARKRLNFGQTGDSESVPDPQPLGEPPAAPTSLGSNTMSAGGGGPLGDNNQGADGV GNASGDWHCDSTWMGDRVVTKSTRTWVLPSYNNHQYREIKSGSVDGSNANAYFG YSTPWGYFDFNRFHSHWSPRDWQRLINNYWGFRPRSLRVKIFNIQVKEVTVQDSTTT IANNLTSTVQVFTDDDYQLPYVVGNGTEGCLPAFPPQVFTLPQYGYATLNRDNTENP TERSSFFCLEYFPSKMLRTGNNFEFTYNFEEVPFHSSFAPSQNLFKLANPLVDQYLYR FVSTNNTGGVQFNKNLAGRYANTYKNWFPGPMGRTQGWNLGSGVNRASVSAFAT TNRMELEGASYQVPPQPNGMTNNLQGSNTYALENTMIFNSQPANPGTTATYLEGNM LITSESETQPVNRVAYNVGGQMATNNQSSTTAPATGTYNLQEIVPGSVWMERDVYL QGPIWAKIPETGAHFHPSPAMGGFGLKHPPPMMLIKNTPVPGNITSFSDVPVSSFITQY STGQVTVEMEWELKKENSKRWNPEIQYTNNYNDPQFVDFAPDSTGEYRTTRPIGTR YLTRPL SEQ ID NO:11 [AAV54 VP1/2 and VP3] MTDGYLPDWLEDNLSEGVREWWALQPGAPKPKANQQHQDNARGLVLPGYKYLGP GNGLDKGEPVNAADAAALEHDKAYDQQLKAGDNPYLKYNHADAEFQQRLQGDTS FGGNLGRAVFQAKKRVLEPLGLVEQAGETAPGKKRPLIESPQQPDSSTGIGKKGKQP AKKKLVFEDETGAGDGPPEGSTSGAMSDDSEMSAGGGGPLGDNNQGADGVGNASG DWHCDSTWMGDRVVTKSTRTWVLPSYNNHQYREIKSGSVDGSNANAYFGYSTPW GYFDFNRFHSHWSPRDWQRLINNYWGFRPRSLRVKIFNIQVKEVTVQDSTTTIANNL TSTVQVFTDDDYQLPYVVGNGTEGCLPAFPPQVFTLPQYGYATLNRDNTENPTERSS FFCLEYFPSKMLRTGNNFEFTYNFEEVPFHSSFAPSQNLFKLANPLVDQYLYRFVSTN NTGGVQFNKNLAGRYANTYKNWFPGPMGRTQGWNLGSGVNRASVSAFATTNRME LEGASYQVPPQPNGMTNNLQGSNTYALENTMIFNSQPANPGTTATYLEGNMLITSES ETQPVNRVAYNVGGQMATNNQSSTTAPATGTYNLQEIVPGSVWMERDVYLQGPIW AKIPETGAHFHPSPAMGGFGLKHPPPMMLIKNTPVPGNITSFSDVPVSSFITQYSTGQV TVEMEWELKKENSKRWNPEIQYTNNYNDPQFVDFAPDSTGEYRTTRPIGTRYLTRPL SEQ ID NO:12 [AAV56 VP1/2 and VP3] MAADGYLPDWLEDNLSEGIREWWDLKPGAPKPKANQQKQDDGRGLVLPGYKYLG PFNGLDKGEPVNAADAAALEHDKAYDQQLKAGDNPYLRYNHADAEFQERLQEDTS FGGNLGRAVFQAKKRVLEPFGLVEEGAKTAPGKKRPVEQSPQEPDSSSGIGKTGQQP AKKRLNFGQTGDSESVPDPQPLGEPPATPAAVGPTTMSAGGGGPLGDNNQGADGVG NASGDWHCDSTWMGDRVVTKSTRTWVLPSYNNHQYREIKSGSVDGSNANAYFGY STPWGYFDFNRFHSHWSPRDWQRLINNYWGFRPRSLRVKIFNIQVKEVTVQDSTTTI ANNLTSTVQVFTDDDYQLPYVVGNGTEGCLPAFPPQVFTLPQYGYATLNRDNTENP TERSSFFCLEYFPSKMLRTGNNFEFTYNFEEVPFHSSFAPSQNLFKLANPLVDQYLYR FVSTNNTGGVQFNKNLAGRYANTYKNWFPGPMGRTQGWNLGSGVNRASVSAFAT TNRMELEGASYQVPPQPNGMTNNLQGSNTYALENTMIFNSQPANPGTTATYLEGNM LITSESETQPVNRVAYNVGGQMATNNQSSTTAPATGTYNLQEIVPGSVWMERDVYL QGPIWAKIPETGAHFHPSPAMGGFGLKHPPPMMLIKNTPVPGNITSFSDVPVSSFITQY STGQVTVEMEWELKKENSKRWNPEIQYTNNYNDPQFVDFAPDSTGEYRTTRPIGTR YLTRPL SEQ ID NO:13 [AAV57 VP1/2 and VP3] MAADGYLPDWLEDNLSEGIREWWDLKPGAPKPKANQQKQDNGRGLVLPGYKYLG PFNGLDKGEPVNAADAAALEHDKAYDQQLKAGDNPYLRYNHADAEFQERLQEDTS FGGNLGRAVFQAKKRVLEPLGLVEEGAKTAPAKKRPVEPSPQRSPDSSTGIGKKGQQ PARKRLNFGQTGDSESVPDPQPLGEPPAAPSSVGSGTVAAGGGAPMSAGGGGPLGD NNQGADGVGNASGDWHCDSTWMGDRVVTKSTRTWVLPSYNNHQYREIKSGSVDG SNANAYFGYSTPWGYFDFNRFHSHWSPRDWQRLINNYWGFRPRSLRVKIFNIQVKE VTVQDSTTTIANNLTSTVQVFTDDDYQLPYVVGNGTEGCLPAFPPQVFTLPQYGYAT LNRDNTENPTERSSFFCLEYFPSKMLRTGNNFEFTYNFEEVPFHSSFAPSQNLFKLANP LVDQYLYRFVSTNNTGGVQFNKNLAGRYANTYKNWFPGPMGRTQGWNLGSGVNR ASVSAFATTNRMELEGASYQVPPQPNGMTNNLQGSNTYALENTMIFNSQPANPGTT ATYLEGNMLITSESETQPVNRVAYNVGGQMATNNQSSTTAPATGTYNLQEIVPGSV WMERDVYLQGPIWAKIPETGAHFHPSPAMGGFGLKHPPPMMLIKNTPVPGNITSFSD VPVSSFITQYSTGQVTVEMEWELKKENSKRWNPEIQYTNNYNDPQFVDFAPDSTGE YRTTRPIGTRYLTRPL SEQ ID NO:14 [AAV58 VP1/2 and VP3] MAADGYLPDWLEDNLSEGIREWWALKPGAPKPKANQQKQDDGRGLVLPGYKYLG PFNGLDKGEPVNAADAAALEHDKAYDQQLQAGDNPYLRYNHADAEFQERLQEDTS FGGNLGRAVFQAKKRVLEPLGLVEEGAKTAPGKKRPVEPSPQRSPDSSTGIGKKGQQ PARKRLNFGQTGDSESVPDPQPLGEPPAAPSGVGPNTMSAGGGGPLGDNNQGADGV GNASGDWHCDSTWMGDRVVTKSTRTWVLPSYNNHQYREIKSGSVDGSNANAYFG YSTPWGYFDFNRFHSHWSPRDWQRLINNYWGFRPRSLRVKIFNIQVKEVTVQDSTTT IANNLTSTVQVFTDDDYQLPYVVGNGTEGCLPAFPPQVFTLPQYGYATLNRDNTENP TERSSFFCLEYFPSKMLRTGNNFEFTYNFEEVPFHSSFAPSQNLFKLANPLVDQYLYR FVSTNNTGGVQFNKNLAGRYANTYKNWFPGPMGRTQGWNLGSGVNRASVSAFAT TNRMELEGASYQVPPQPNGMTNNLQGSNTYALENTMIFNSQPANPGTTATYLEGNM LITSESETQPVNRVAYNVGGQMATNNQSSTTAPATGTYNLQEIVPGSVWMERDVYL QGPIWAKIPETGAHFHPSPAMGGFGLKHPPPMMLIKNTPVPGNITSFSDVPVSSFITQY STGQVTVEMEWELKKENSKRWNPEIQYTNNYNDPQFVDFAPDSTGEYRTTRPIGTR YLTRPL SEQ ID NO:15 [AAV59 VP1/2 and VP3] MAADGYLPDWLEDNLSEGIREWWALKPGAPQPKANQQHQDNARGLVLPGYKYLG PGNGLDKGEPVNAADAAALEHDKAYDQQLKAGDNPYLKYNHADAEFQERLKEDT SFGGNLGRAVFQAKKRLLEPLGLVEEAAKTAPGKKRPVEQSPQEPDSSAGIGKSGAQ PAKKRLNFGQTGDTESVPDPQPIGEPPAAPSGVGSLTMSAGGGGPLGDNNQGADGV GNASGDWHCDSTWMGDRVVTKSTRTWVLPSYNNHQYREIKSGSVDGSNANAYFG YSTPWGYFDFNRFHSHWSPRDWQRLINNYWGFRPRSLRVKIFNIQVKEVTVQDSTTT IANNLTSTVQVFTDDDYQLPYVVGNGTEGCLPAFPPQVFTLPQYGYATLNRDNTENP TERSSFFCLEYFPSKMLRTGNNFEFTYNFEEVPFHSSFAPSQNLFKLANPLVDQYLYR FVSTNNTGGVQFNKNLAGRYANTYKNWFPGPMGRTQGWNLGSGVNRASVSAFAT TNRMELEGASYQVPPQPNGMTNNLQGSNTYALENTMIFNSQPANPGTTATYLEGNM LITSESETQPVNRVAYNVGGQMATNNQSSTTAPATGTYNLQEIVPGSVWMERDVYL QGPIWAKIPETGAHFHPSPAMGGFGLKHPPPMMLIKNTPVPGNITSFSDVPVSSFITQY STGQVTVEMEWELKKENSKRWNPEIQYTNNYNDPQFVDFAPDSTGEYRTTRPIGTR YLTRPL SEQ ID NO:16 [AAV5-6 VP3] MSAGGGGPLGDNNQGADGVGNASGDWHCDSTWMGDRVVTKSTRTWVLPSYNNH QYREIKSGSVDGSNANAYFGYSTPWGYFDFNRFHSHWSPRDWQRLINNYWGFRPRS LRVKIFNIQVKEVTVQDSTTTIANNLTSTVQVFTDDDYQLPYVVGNGTEGCLPAFPPQ VFTLPQYGYATLNRDNTENPTERSSFFCLEYFPSKMLRTGNNFEFTYNFEEVPFHSSF APSQNLFKLANPLVDQYLYRFVSTNNTGGVQFNKNLAGRYANTYKNWFPGPMGRT QGWNLGSGVNRASVSAFATTNRMELEGASYQVPPQPNGMTNNLQGSNTYALENTM IFNSQPANPGTTATYLEGNMLITSESETQPVNRVAYNVGGQMATNNQSASTGASSTT APATGTYNLQEIVPGSVWMERDVYLQGPIWAKIPETGAHFHPSPAMGGFGLKHPPP MMLIKNTPVPGNITSFSDVPVSSFITQYSTGQVTVEMEWELKKENSKRWNPEIQYTN NYNDPQFVDFAPDSTGEYRTTRPIGTRYLTRPL SEQ ID NO:17 [AAV596 VP1/2 and VP3] MAADGYLPDWLEDNLSEGIREWWALKPGAPQPKANQQHQDNARGLVLPGYKYLG PGNGLDKGEPVNAADAAALEHDKAYDQQLKAGDNPYLKYNHADAEFQERLKEDT SFGGNLGRAVFQAKKRLLEPLGLVEEAAKTAPGKKRPVEQSPQEPDSSAGIGKSGAQ PAKKRLNFGQTGDTESVPDPQPIGEPPAAPSGVGSLTMSAGGGGPLGDNNQGADGV GNASGDWHCDSTWMGDRVVTKSTRTWVLPSYNNHQYREIKSGSVDGSNANAYFG YSTPWGYFDFNRFHSHWSPRDWQRLINNYWGFRPRSLRVKIFNIQVKEVTVQDSTTT IANNLTSTVQVFTDDDYQLPYVVGNGTEGCLPAFPPQVFTLPQYGYATLNRDNTENP TERSSFFCLEYFPSKMLRTGNNFEFTYNFEEVPFHSSFAPSQNLFKLANPLVDQYLYR FVSTNNTGGVQFNKNLAGRYANTYKNWFPGPMGRTQGWNLGSGVNRASVSAFAT TNRMELEGASYQVPPQPNGMTNNLQGSNTYALENTMIFNSQPANPGTTATYLEGNM LITSESETQPVNRVAYNVGGQMATNNQSASTGASSTTAPATGTYNLQEIVPGSVWM ERDVYLQGPIWAKIPETGAHFHPSPAMGGFGLKHPPPMMLIKNTPVPGNITSFSDVPV SSFITQYSTGQVTVEMEWELKKENSKRWNPEIQYTNNYNDPQFVDFAPDSTGEYRTT RPIGTRYLTRPL SEQ ID NO:18 [AAV5-8 VP3] MSAGGGGPLGDNNQGADGVGNASGDWHCDSTWMGDRVVTKSTRTWVLPSYNNH QYREIKSGSVDGSNANAYFGYSTPWGYFDFNRFHSHWSPRDWQRLINNYWGFRPRS LRVKIFNIQVKEVTVQDSTTTIANNLTSTVQVFTDDDYQLPYVVGNGTEGCLPAFPPQ VFTLPQYGYATLNRDNTENPTERSSFFCLEYFPSKMLRTGNNFEFTYNFEEVPFHSSF APSQNLFKLANPLVDQYLYRFVSTNNTGGVQFNKNLAGRYANTYKNWFPGPMGRT QGWNLGSGVNRASVSAFATTNRMELEGASYQVPPQPNGMTNNLQGSNTYALENTM IFNSQPANPGTTATYLEGNMLITSESETQPVNRVAYNVGGQMATNNQNGTSGGATSS TTAPATGTYNLQEIVPGSVWMERDVYLQGPIWAKIPETGAHFHPSPAMGGFGLKHPP PMMLIKNTPVPGNITSFSDVPVSSFITQYSTGQVTVEMEWELKKENSKRWNPEIQYT NNYNDPQFVDFAPDSTGEYRTTRPIGTRYLTRPL SEQ ID NO:19 [AAV598 VP1/2 and VP3] MAADGYLPDWLEDNLSEGIREWWALKPGAPQPKANQQHQDNARGLVLPGYKYLG PGNGLDKGEPVNAADAAALEHDKAYDQQLKAGDNPYLKYNHADAEFQERLKEDT SFGGNLGRAVFQAKKRLLEPLGLVEEAAKTAPGKKRPVEQSPQEPDSSAGIGKSGAQ PAKKRLNFGQTGDTESVPDPQPIGEPPAAPSGVGSLTMSAGGGGPLGDNNQGADGV GNASGDWHCDSTWMGDRVVTKSTRTWVLPSYNNHQYREIKSGSVDGSNANAYFG YSTPWGYFDFNRFHSHWSPRDWQRLINNYWGFRPRSLRVKIFNIQVKEVTVQDSTTT IANNLTSTVQVFTDDDYQLPYVVGNGTEGCLPAFPPQVFTLPQYGYATLNRDNTENP TERSSFFCLEYFPSKMLRTGNNFEFTYNFEEVPFHSSFAPSQNLFKLANPLVDQYLYR FVSTNNTGGVQFNKNLAGRYANTYKNWFPGPMGRTQGWNLGSGVNRASVSAFAT TNRMELEGASYQVPPQPNGMTNNLQGSNTYALENTMIFNSQPANPGTTATYLEGNM LITSESETQPVNRVAYNVGGQMATNNQNGTSGGATSSTTAPATGTYNLQEIVPGSV WMERDVYLQGPIWAKIPETGAHFHPSPAMGGFGLKHPPPMMLIKNTPVPGNITSFSD VPVSSFITQYSTGQVTVEMEWELKKENSKRWNPEIQYTNNYNDPQFVDFAPDSTGE YRTTRPIGTRYLTRPL SEQ ID NO:20. [WT AAV5 capsid protein (VP1, VP2 (SEQ ID NO:1) and VP3 (SEQ ID NO:2)) Accession No. YP_068409.1; 574Q underlined] MSFVDHPPDWLEEVGEGLREFLGLEAGPPKPKPNQQHQDQARGLVLPGYNYLGPGN GLDRGEPVNRADEVAREHDISYNEQLEAGDNPYLKYNHADAEFQEKLADDTSFGGN LGKAVFQAKKRVLEPFGLVEEGAKTAPTGKRIDDHFPKRKKARTEEDSKPSTSSDAE AGPSGSQQLQIPAQPASSLGADTMSAGGGGPLGDNNQGADGVGNASGDWHCDSTW MGDRVVTKSTRTWVLPSYNNHQYREIKSGSVDGSNANAYFGYSTPWGYFDFNRFH SHWSPRDWQRLINNYWGFRPRSLRVKIFNIQVKEVTVQDSTTTIANNLTSTVQVFTD DDYQLPYVVGNGTEGCLPAFPPQVFTLPQYGYATLNRDNTENPTERSSFFCLEYFPS KMLRTGNNFEFTYNFEEVPFHSSFAPSQNLFKLANPLVDQYLYRFVSTNNTGGVQFN KNLAGRYANTYKNWFPGPMGRTQGWNLGSGVNRASVSAFATTNRMELEGASYQV PPQPNGMTNNLQGSNTYALENTMIFNSQPANPGTTATYLEGNMLITSESETQPVNRV AYNVGGQMATNNQSSTTAPATGTYNLQEIVPGSVWMERDVYLQGPIWAKIPETGA HFHPSPAMGGFGLKHPPPMMLIKNTPVPGNITSFSDVPVSSFITQYSTGQVTVEMEWE LKKENSKRWNPEIQYTNNYNDPQFVDFAPDSTGEYRTTRPIGTRYLTRPL SEQ ID NO:21. AAV1 VP1, VP2 and VP3 MAADGYLPDWLEDNLSEGIREWWDLKPGAPKPKANQQKQDDGRGLVLPGYKYLG PFNGLDKGEPVNAADAAALEHDKAYDQQLKAGDNPYLRYNHADAEFQERLQEDTS FGGNLGRAVFQAKKRVLEPLGLVEEGAKTAPGKKRPVEQSPQEPDSSSGIGKTGQQP AKKRLNFGQTGDSESVPDPQPLGEPPATPAAVGPTTMASGGGAPMADNNEGADGV GNASGNWHCDSTWLGDRVITTSTRTWALPTYNNHLYKQISSASTGASNDNHYFGYS TPWGYFDFNRFHCHFSPRDWQRLINNNWGFRPKRLNFKLFNIQVKEVTTNDGVTTIA NNLTSTVQVFSDSEYQLPYVLGSAHQGCLPPFPADVFMIPQYGYLTLNNGSQAVGRS SFYCLEYFPSQMLRTGNNFTFSYTFEEVPFHSSYAHSQSLDRLMNPLIDQYLYYLNRT QNQSGSAQNKDLLFSRGSPAGMSVQPKNWLPGPCYRQQRVSKTKTDNNNSNFTWT GASKYNLNGRESIINPGTAMASHKDDEDKFFPMSGVMIFGKESAGASNTALDNVMIT DEEEIKATNPVATERFGTVAVNFQSSSTDPATGDVHAMGALPGMVWQDRDVYLQG PIWAKIPHTDGHFHPSPLMGGFGLKNPPPQILIKNTPVPANPPAEFSATKFASFITQYST GQVSVEIEWELQKENSKRWNPEVQYTSNYAKSANVDFTVDNNGLYTEPRPIGTRYL TRPL SEQ ID NO:22. AAV2 VP1, VP2, and VP3 AAC03780.1 MAADGYLPDWLEDTLSEGIRQWWKLKPGPPPPKPAERHKDDSRGLVLPGYKYLGPF NGLDKGEPVNEADAAALEHDKAYDRQLDSGDNPYLKYNHADAEFQERLKEDTSFG GNLGRAVFQAKKRVLEPLGLVEEPVKTAPGKKRPVEHSPVEPDSSSGTGKAGQQPA RKRLNFGQTGDADSVPDPQPLGQPPAAPSGLGTNTMATGSGAPMADNNEGADGVG NSSGNWHCDSTWMGDRVITTSTRTWALPTYNNHLYKQISSQSGASNDNHYFGYSTP WGYFDFNRFHCHFSPRDWQRLINNNWGFRPKRLNFKLFNIQVKEVTQNDGTTTIAN NLTSTVQVFTDSEYQLPYVLGSAHQGCLPPFPADVFMVPQYGYLTLNNGSQAVGRS SFYCLEYFPSQMLRTGNNFTFSYTFEDVPFHSSYAHSQSLDRLMNPLIDQYLYYLSRT NTPSGTTTQSRLQFSQAGASDIRDQSRNWLPGPCYRQQRVSKTSADNNNSEYSWTG ATKYHLNGRDSLVNPAMASHKDDEEKFFPQSGVLIFGKQGSEKTNVNIEKVMITDEE EIGTTNPVATEQYGSVSTNLQRGNRQAATADVNTQGVLPGMVWQDRDVYLQGPIW AKIPHTDGHFHPSPLMGGFGLKHPPPQILIKNTPVPANPSTTFSAAKFASFITQYSTGQ VSVEIEWELQKENSKRWNPEIQYTSNYNKSVNRGLTVDTNGVYSEPRPIGTRYLTRN L SEQ ID NO:23. AAV3 VP1, VP3, and VP3 NP_043941.1 MAADGYLPDWLEDNLSEGIREWWALKPGVPQPKANQQHQDNRRGLVLPGYKYLG PGNGLDKGEPVNEADAAALEHDKAYDQQLKAGDNPYLKYNHADAEFQERLQEDTS FGGNLGRAVFQAKKRILEPLGLVEEAAKTAPGKKGAVDQSPQEPDSSSGVGKSGKQ PARKRLNFGQTGDSESVPDPQPLGEPPAAPTSLGSNTMASGGGAPMADNNEGADGV GNSSGNWHCDSQWLGDRVITTSTRTWALPTYNNHLYKQISSQSGASNDNHYFGYST PWGYFDFNRFHCHFSPRDWQRLINNNWGFRPKKLSFKLFNIQVRGVTQNDGTTTIAN NLTSTVQVFTDSEYQLPYVLGSAHQGCLPPFPADVFMVPQYGYLTLNNGSQAVGRS SFYCLEYFPSQMLRTGNNFQFSYTFEDVPFHSSYAHSQSLDRLMNPLIDQYLYYLNR TQGTTSGTTNQSRLLFSQAGPQSMSLQARNWLPGPCYRQQRLSKTANDNNNSNFPW TAASKYHLNGRDSLVNPGPAMASHKDDEEKFFPMHGNLIFGKEGTTASNAELDNV MITDEEEIRTTNPVATEQYGTVANNLQSSNTAPTTGTVNHQGALPGMVWQDRDVYL QGPIWAKIPHTDGHFHPSPLMGGFGLKHPPPQIMIKNTPVPANPPTTFSPAKFASFITQ YSTGQVSVEIEWELQKENSKRWNPEIQYTSNYNKSVNVDFTVDTNGVYSEPRPIGTR YLTRNL SEQ ID NO:24. AAV4 VP1, VP2, and VP3 AAC58045.1 MTDGYLPDWLEDNLSEGVREWWALQPGAPKPKANQQHQDNARGLVLPGYKYLGP GNGLDKGEPVNAADAAALEHDKAYDQQLKAGDNPYLKYNHADAEFQQRLQGDTS FGGNLGRAVFQAKKRVLEPLGLVEQAGETAPGKKRPLIESPQQPDSSTGIGKKGKQP AKKKLVFEDETGAGDGPPEGSTSGAMSDDSEMRAAAGGAAVEGGQGADGVGNAS GDWHCDSTWSEGHVTTTSTRTWVLPTYNNHLYKRLGESLQSNTYNGFSTPWGYFD FNRFHCHFSPRDWQRLINNNWGMRPKAMRVKIFNIQVKEVTTSNGETTVANNLTST VQIFADSSYELPYVMDAGQEGSLPPFPNDVFMVPQYGYCGLVTGNTSQQQTDRNAF YCLEYFPSQMLRTGNNFEITYSFEKVPFHSMYAHSQSLDRLMNPLIDQYLWGLQSTT TGTTLNAGTATTNFTKLRPTNFSNFKKNWLPGPSIKQQGFSKTANQNYKIPATGSDSL IKYETHSTLDGRWSALTPGPPMATAGPADSKFSNSQLIFAGPKQNGNTATVPGTLIFT SEEELAATNATDTDMWGNLPGGDQSNSNLPTVDRLTALGAVPGMVWQNRDIYYQG PIWAKIPHTDGHFHPSPLIGGFGLKHPPPQIFIKNTPVPANPATTFSSTPVNSFITQYST G QVSVQIDWEIQKERSKRWNPEVQFTSNYGQQNSLLWAPDAAGKYTEPRAIGTRYLT HHLZ SEQ ID NO:25. AAV6 VP1, VP2, and VP3 AAB95450.1 MAADGYLPDWLEDNLSEGIREWWDLKPGAPKPKANQQKQDDGRGLVLPGYKYLG PFNGLDKGEPVNAADAAALEHDKAYDQQLKAGDNPYLRYNHADAEFQERLQEDTS FGGNLGRAVFQAKKRVLEPFGLVEEGAKTAPGKKRPVEQSPQEPDSSSGIGKTGQQP AKKRLNFGQTGDSESVPDPQPLGEPPATPAAVGPTTMASGGGAPMADNNEGADGV GNASGNWHCDSTWLGDRVITTSTRTWALPTYNNHLYKQISSASTGASNDNHYFGYS TPWGYFDFNRFHCHFSPRDWQRLINNNWGFRPKRLNFKLFNIQVKEVTTNDGVTTIA NNLTSTVQVFSDSEYQLPYVLGSAHQGCLPPFPADVFMIPQYGYLTLNNGSQAVGRS SFYCLEYFPSQMLRTGNNFTFSYTFEDVPFHSSYAHSQSLDRLMNPLIDQYLYYLNRT QNQSGSAQNKDLLFSRGSPAGMSVQPKNWLPGPCYRQQRVSKTKTDNNNSNFTWT GASKYNLNGRESIINPGTAMASHKDDKDKFFPMSGVMIFGKESAGASNTALDNVMI TDEEEIKATNPVATERFGTVAVNLQSSSTDPATGDVHVMGALPGMVWQDRDVYLQ GPIWAKIPHTDGHFHPSPLMGGFGLKHPPPQILIKNTPVPANPPAEFSATKFASFITQYS TGQVSVEIEWELQKENSKRWNPEVQYTSNYAKSANVDFTVDNNGLYTEPRPIGTRY LTRPL SEQ ID NO:26. AAV7 VP1, VP2, VP3 YP_077178.1 MAADGYLPDWLEDNLSEGIREWWDLKPGAPKPKANQQKQDNGRGLVLPGYKYLG PFNGLDKGEPVNAADAAALEHDKAYDQQLKAGDNPYLRYNHADAEFQERLQEDTS FGGNLGRAVFQAKKRVLEPLGLVEEGAKTAPAKKRPVEPSPQRSPDSSTGIGKKGQQ PARKRLNFGQTGDSESVPDPQPLGEPPAAPSSVGSGTVAAGGGAPMADNNEGADGV GNASGNWHCDSTWLGDRVITTSTRTWALPTYNNHLYKQISSETAGSTNDNTYFGYS TPWGYFDFNRFHCHFSPRDWQRLINNNWGFRPKKLRFKLFNIQVKEVTTNDGVTTIA NNLTSTIQVFSDSEYQLPYVLGSAHQGCLPPFPADVFMIPQYGYLTLNNGSQSVGRSS FYCLEYFPSQMLRTGNNFEFSYSFEDVPFHSSYAHSQSLDRLMNPLIDQYLYYLART QSNPGGTAGNRELQFYQGGPSTMAEQAKNWLPGPCFRQQRVSKTLDQNNNSNFAW TGATKYHLNGRNSLVNPGVAMATHKDDEDRFFPSSGVLIFGKTGATNKTTLENVLM TNEEEIRPTNPVATEEYGIVSSNLQAANTAAQTQVVNNQGALPGMVWQNRDVYLQ GPIWAKIPHTDGNFHPSPLMGGFGLKHPPPQILIKNTPVPANPPEVFTPAKFASFITQYS TGQVSVEIEWELQKENSKRWNPEIQYTSNFEKQTGVDFAVDSQGVYSEPRPIGTRYL TRNL SEQ ID NO:27. AAV8 VP1, VP2, VP3, YP_077180.1 MAADGYLPDWLEDNLSEGIREWWALKPGAPKPKANQQKQDDGRGLVLPGYKYLG PFNGLDKGEPVNAADAAALEHDKAYDQQLQAGDNPYLRYNHADAEFQERLQEDTS FGGNLGRAVFQAKKRVLEPLGLVEEGAKTAPGKKRPVEPSPQRSPDSSTGIGKKGQQ PARKRLNFGQTGDSESVPDPQPLGEPPAAPSGVGPNTMAAGGGAPMADNNEGADG VGSSSGNWHCDSTWLGDRVITTSTRTWALPTYNNHLYKQISNGTSGGATNDNTYFG YSTPWGYFDFNRFHCHFSPRDWQRLINNNWGFRPKRLSFKLFNIQVKEVTQNEGTKT IANNLTSTIQVFTDSEYQLPYVLGSAHQGCLPPFPADVFMIPQYGYLTLNNGSQAVGR SSFYCLEYFPSQMLRTGNNFQFTYTFEDVPFHSSYAHSQSLDRLMNPLIDQYLYYLSR TQTTGGTANTQTLGFSQGGPNTMANQAKNWLPGPCYRQQRVSTTTGQNNNSNFAW TAGTKYHLNGRNSLANPGIAMATHKDDEERFFPSNGILIFGKQNAARDNADYSDVM LTSEEEIKTTNPVATEEYGIVADNLQQQNTAPQIGTVNSQGALPGMVWQNRDVYLQ GPIWAKIPHTDGNFHPSPLMGGFGLKHPPPQILIKNTPVPADPPTTFNQSKLNSFITQY STGQVSVEIEWELQKENSKRWNPEIQYTSNYYKSTSVDFAVNTEGVYSEPRPIGTRY LTRNL SEQ ID NO:28. AAV9 VP1, VP2, VP3 AAS99264.1 MAADGYLPDWLEDNLSEGIREWWALKPGAPQPKANQQHQDNARGLVLPGYKYLG PGNGLDKGEPVNAADAAALEHDKAYDQQLKAGDNPYLKYNHADAEFQERLKEDT SFGGNLGRAVFQAKKRLLEPLGLVEEAAKTAPGKKRPVEQSPQEPDSSAGIGKSGAQ PAKKRLNFGQTGDTESVPDPQPIGEPPAAPSGVGSLTMASGGGAPVADNNEGADGV GSSSGNWHCDSQWLGDRVITTSTRTWALPTYNNHLYKQISNSTSGGSSNDNAYFGY STPWGYFDFNRFHCHFSPRDWQRLINNNWGFRPKRLNFKLFNIQVKEVTDNNGVKTI ANNLTSTVQVFTDSDYQLPYVLGSAHEGCLPPFPADVFMIPQYGYLTLNDGSQAVG RSSFYCLEYFPSQMLRTGNNFQFSYEFENVPFHSSYAHSQSLDRLMNPLIDQYLYYLS KTINGSGQNQQTLKFSVAGPSNMAVQGRNYIPGPSYRQQRVSTTVTQNNNSEFAWP GASSWALNGRNSLMNPGPAMASHKEGEDRFFPLSGSLIFGKQGTGRDNVDADKVMI TNEEEIKTTNPVATESYGQVATNHQSAQAQAQTGWVQNQGILPGMVWQDRDVYLQ GPIWAKIPHTDGNFHPSPLMGGFGMKHPPPQILIKNTPVPADPPTAFNKDKLNSFITQ YSTGQVSVEIEWELQKENSKRWNPEIQYTSNYYKSNNVEFAVNTEGVYSEPRPIGTR YLTRNL SEQ ID NO:29. AAVrh10 VP1, VP2, VP3 AAO88201.1 MAADGYLPDWLEDNLSEGIREWWDLKPGAPKPKANQQKQDDGRGLVLPGYKYLG PFNGLDKGEPVNAADAAALEHDKAYDQQLKAGDNPYLRYNHADAEFQERLQEDTS FGGNLGRAVFQAKKRVLEPLGLVEEGAKTAPGKKRPVEPSPQRSPDSSTGIGKKGQQ PAKKRLNFGQTGDSESVPDPQPIGEPPAGPSGLGSGTMAAGGGAPMADNNEGADGV GSSSGNWHCDSTWLGDRVITTSTRTWALPTYNNHLYKQISNGTSGGSTNDNTYFGY STPWGYFDFNRFHCHFSPRDWQRLINNNWGFRPKRLNFKLFNIQVKEVTQNEGTKTI ANNLTSTIQVFTDSEYQLPYVLGSAHQGCLPPFPADVFMIPQYGYLTLNNGSQAVGR SSFYCLEYFPSQMLRTGNNFEFSYQFEDVPFHSSYAHSQSLDRLMNPLIDQYLYYLSR TQSTGGTAGTQQLLFSQAGPNNMSAQAKNWLPGPCYRQQRVSTTLSQNNNSNFAW TGATKYHLNGRDSLVNPGVAMATHKDDEERFFPSSGVLMFGKQGAGKDNVDYSSV MLTSEEEIKTTNPVATEQYGVVADNLQQQNAAPIVGAVNSQGALPGMVWQNRDVY LQGPIWAKIPHTDGNFHPSPLMGGFGLKHPPPQILIKNTPVPADPPTTFSQAKLASFIT QYSTGQVSVEIEWELQKENSKRWNPEIQYTSNYYKSTNVDFAVNTDGTYSEPRPIGT RYLTRNL