Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
ENHANCING AAV-MEDIATED TRANSDUCTION OF OCULAR TISSUES WITH HYALURONIC ACID
Document Type and Number:
WIPO Patent Application WO/2020/264438
Kind Code:
A2
Abstract:
Disclosed herein are compositions of rAAV particles and methods for administrating rAAV particles having enhanced transduction properties.

Inventors:
BOYE SHANNON (US)
BOYE SANFORD (US)
Application Number:
PCT/US2020/040004
Publication Date:
December 30, 2020
Filing Date:
June 26, 2020
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
UNIV FLORIDA (US)
International Classes:
A61K31/728
Attorney, Agent or Firm:
GREENWALD, Eric, P. et al. (US)
Download PDF:
Claims:
CLAIMS

What is claimed is:

1. A method of delivering a cargo to an eye of a subject, the method comprising

administering to the eye of the subject a rAAV particle comprising: (a) a capsid that is admixed with hyaluronic acid (HA) and (b) a cargo, wherein the cargo is delivered to the eye.

2. The method of claim 1, wherein the capsid comprises one or more surface-exposed patches of positively-charged amino residues.

3. The method of claim 1 or 2, wherein the serotype of the capsid is AAV2 or a variant thereof.

4. The method of any one of claims 1-3, wherein the capsid comprises non-native amino acid substitutions at amino acid residues of a wild-type AAV2 capsid as set forth in SEQ ID NO: 2, wherein the non-native amino acid substitutions comprise one or more of Y272F, Y444F, T491V, Y500F, Y700F, Y704F or Y730F.

5. The method of any one of claims 1-3, wherein the capsid is an AAV2G9 capsid.

6. The method of claim 1 or 2, wherein the capsid comprises non-native amino acid substitutions of a wild-type AAV2 capsid as set forth in SEQ ID NO: 2, wherein the non-native amino acid substitutions comprise:

(a) Y444F;

(b) Y444F+Y500F+Y730F;

(c) Y272F+Y444F+Y500F+Y730F;

(d) Y 444F+ Y 500F+ Y730F+T491 V ; or

(e) Y272F+Y444F+Y500F+Y730F+T491V, or at equivalent amino acid positions corresponding thereto in any one of the wild-type AAV1, AAV3, AAV4, AAV5, AAV7, AAV9, or AAV10 capsid proteins, as set forth, respectively, in SEQ ID NO: 1, SEQ ID NO: 3, SEQ ID NO: 4, SEQ ID NO: 5, SEQ ID NO: 7, SEQ ID NO: 9, or SEQ ID NO: 10.

7. The method of claim 1 or 2, wherein the serotype of the capsid is AAV6 or a variant thereof.

8. The method of any one of claims 1, 2 and 7, wherein the capsid comprises non-native amino acid substitutions at amino acid residues of a wild-type AAV6 capsid as set forth in SEQ ID NO: 6, wherein the non-native amino acid substitutions comprise one or more of Y445F, Y705F, Y731F, T492V and S663V.

9. The method of any one of claims 1-3 and 7, wherein the capsid comprises non-native amino acid substitutions of a wild-type AAV6 capsid as set forth in SEQ ID NO: 6, wherein the non-native amino acid substitutions comprise:

(a) Y445F;

(b) Y705F+Y731F;

(c) T492V;

(d) Y705F+Y731F+T492V;

(e) S663V; or

(f) S663V+T492V.

10. The method of claim 1 or 2, wherein the capsid is selected from the group consisting of a AAV7m8 capsid, a AAV-DJ capsid, a AAV2/2-MAX capsid, a AAVSHhlO capsid, and a AAVSHhlOY capsid.

11. The method of claim 1 or 2, wherein the capsid is selected from the group consisting of a AAV3 capsid, a AAV3b capsid, and a AAVFK03 capsid.

12. The method of claim 1 or 2, wherein the capsid comprises a non-native amino acid substitution at amino acid residue 531 of a wild-type AAV1 capsid as set forth in SEQ ID NO: 1, wherein the non-native amino acid substitution is E531K.

13. The method of claim 1 or 2, wherein the capsid is AAV7BP2 capsid.

14. The method of any claim 1 or 2, wherein the capsid comprises non-native amino acid substitutions at amino acid residues 533 and/or 733 of a wild-type AAV8 capsid as set forth in SEQ ID NO: 8, wherein the non-native amino acid substitution is E533K and/or Y733F.

15. The method of any one of claims 1-14, wherein the capsid is pre-incubated with the

HA prior to the step of administering.

16. The method of any one of claims 1-15, wherein the capsid is pre-incubated with a buffer that comprises the HA.

17. The method of any one of claims 15-16, wherein the capsid is pre-incubated with the

HA for a duration of about 5 minutes, about 15 minutes, about 30 minutes, about 45 minutes, about 60 minutes or about 75 minutes.

18. The method of claim 17, wherein the capsid is pre-incubated for a duration of about

15 minutes.

19. The method of any one of claims 15-18, wherein the capsid is pre-incubated with the

HA in a concentration of 0.1%, 0.2%, 0.25%, 0.3%, 0.35%, 0.4%, 0.45%, 0.5%, 0.6%, 0.75%, or 1.0% weight by volume.

20. The method of any one of claims 1-19, wherein the rAAV particle is administered to the eye of the subject in a titer of about lxlO10 vg/ml, 5 xlO10 vg/ml, lxlO11 vg/ml, 5 xlO11 vg/ml, lxlO12 vg/ml, 2xl012 vg/ml, 3xl012 vg/ml, 4xl012 vg/ml, about 5xl012 vg/ml, about lxlO13 vg/ml, or about 5xl013 vg/ml.

21. The method of any one of claims 1-20, wherein the rAAV particle is administered to the eye of the subject in a titer of less than 5 xlO11 vg/ml.

22. The method of any one of claims 1-22, wherein the rAAV particle is administered by intravitreal injection.

23. The method of claim 22, wherein the intravitreal injection is provided in a volume of about 200 pL, about 175 pL, about 160 pL, about 145 pL, about 130 pL, about 115 pL, about 100 pL, about 90 pL, about 80 pL, about 70 pL, about 60 pL, about 55 pL, about 50 pL, about 45 pL, about 35 pL, about 20 pL, about 10 pL, or about 5 pL.

24. The method of claim 23, wherein the intravitreal injection is administered in a volume of about 50 pL.

25. The method of any one of claims 1-24, wherein the cargo comprises a polynucleotide comprising a heterologous nucleic acid sequence.

26. The method of claim 25, wherein the heterologous nucleic acid sequence is operably linked to a regulatory sequence that direct expression of the heterologous nucleic acid sequence in a photoreceptor cell, retinal pigment epithelium cell, retinal ganglion cell, bipolar cell, Miiller glial cell or astrocyte cell.

27. The method of claim 26, wherein the regulatory sequence is selected from the group consisting of: a woodchuck hepatitis vims post-transcription regulatory element (WPRE), a polyadenylation signal sequence, an intron/exon junctions/splicing signal, and any combination thereof.

28. The method of any one of claims 25-27, wherein the heterologous nucleic acid

sequence encodes a therapeutic agent.

29. The method of claim 28, wherein the therapeutic agent is a neurotrophic factor or an optogenetic actuator.

30. The method of claim 29, wherein the therapeutic agent is the neurotrophic factor.

31. The method of claim 30, wherein the neurotrophic factor is selected from the group consisting of: brain derived neurotrophic factor (BDNF), nerve growth factor (NGF), neurotrophin-3, ciliary neurotrophic factor (CNTF), an ephrin, glial cell line-derived neurotrophic factors (GDNF), and a combination thereof.

32. The method of claim 29, wherein the therapeutic agent is the optogenetic actuator.

33. The method of claim 32, wherein the optogenetic actuator is selected from the group consisting of: a bacteriorhodopsin, a halorhodopsin, a channelrhodopsin, a microbial sensory rhodopsin, a mammalian rhodopsin, a cone opsin, a melanopsin, and a combination thereof.

34. The method of any one of claims 28-33, wherein the subject is in need of a

therapeutically effective amount of the therapeutic agent.

35. The method of any one of claims 28-34, wherein the a rAAV particle is administered for a time effective to provide a therapeutically-effective amount of the therapeutic agent.

36. A method for treating or ameliorating one or more symptoms of a disease, disorder or condition, the method comprising intravitreally administering to the subject a rAAV particle comprising a capsid admixed with hyaluronic acid (HA) to an eye of a subject for a time sufficient to treat or ameliorate the one or more symptoms of the disease, disorder or condition in the subject, wherein the rAAV particle comprises (a) a polynucleotide encoding a therapeutic agent and (b) the serotype of the capsid is AAV2 or AAV6.

37. A method for providing a mammal in need thereof with a therapeutically effective amount of a therapeutic agent, the method comprising intravitreally administering to the subject a rAAV particle comprising a capsid that is admixed with hyaluronic acid (HA) to an eye of the subject for a time effective to provide the subject with a therapeutically-effective amount of the therapeutic agent, wherein the rAAV particle comprises an AAV7m8, an AAV-DJ, an AAV2/2-MAX, an AAVSHhlO, an

AAVSHhlOY, an AAV3, an AAV3b, or an AAVLK03 capsid.

38. A method for treating or ameliorating one or more symptoms of a disease, disorder or condition, the method comprising intravitreally administering to the subject a rAAV particle comprising a capsid that is admixed with hyaluronic acid (HA) to an eye of the subject in need thereof for a time sufficient to treat or ameliorate the one or more symptoms of the disease, disorder or condition in the subject, wherein the rAAV particle comprises (a) a polynucleotide encoding a therapeutic agent and (a) an AAV7m8, an AAV-DJ, an AAV2/2-MAX, an AAVSHhlO, an AAVSHhlOY, an AAV3, an AAV3b, or an AAVLK03 capsid.

39. The method of any one of claims 36-38, wherein the disease, disorder or condition is age-related macular degeneration (AMD), wet AMD, dry AMD, or geographic atrophy.

40. The method of any one of claims 36-39, wherein the subject is a human.

41. The method of any one of claims 36-40, wherein production of the therapeutic agent a) preserves one or more photoreceptor cells or one or more RPE cells, b) restores one or more rod- and/or cone-mediated functions, c) restores visual behavior in one or both eyes, or d) any combination thereof.

42. The method of claim 41, wherein production of the therapeutic agent persists in the one or more photoreceptor cells or the one or more RPE cells substantially for a period of at least three months following an initial administration of the rAAV particle into the one or both eyes of the mammal.

43. The method of any one of claims 36-40, wherein production of the therapeutic agent preserves one or more retinal ganglion cells, Miiller glial cells or astrocyte cells.

44. The method of claim 43, wherein the therapeutic agent comprises a neurotrophic

factor.

45. The method of claim 43 or 44, wherein the disease, disorder or condition is retinitis pigmentosa or glaucoma.

46. The method of any one of claims 36-40, wherein production of the therapeutic agent preserves one or more retinal ganglion cells or retinal bipolar cells.

47. The method of claim 46, wherein the therapeutic agent comprises an optogenetic

actuator.

48. The method of claim 47, wherein the optogenetic actuator is selected from the group consisting of: a bacteriorhodopsin, a halorhodopsin, a channelrhodopsin, a microbial sensory rhodopsin, a mammalian rhodopsin, a cone opsin, a melanopsin, and a combination thereof.

49. A buffer for storing a mixture of AAV and hyaluronic acid (HA), comprising:

(a) HA in a concentration of 0.1%, 0.2%, 0.25%, 0.3%, 0.35%, 0.4%, 0.45%, 0.5%,

0.6%, 0.75%, or 1.0% weight by volume;

(b) balanced salt solution (BSS);

(c) artificial cerebrospinal fluid; and

(d) phosphate buffered saline (PBS).

50. The buffer of claim 49 further comprising: a. Ringer’s lactate solution;

b. TMN200 solution; c. Polysorbate 20;

d. poloxamer 188; or

e. a combination thereof.

51. The buffer of claim 49 or 50, wherein the HA is in a concentration of 0.4% weight by volume.

Description:
ENHANCING AA V-MEDIATED TRANSDUCTION OF OCULAR TISSUES WITH

HYALURONIC ACID

STATEMENT REGARDING FEDERALLY SPONSORED RESEARCH

[0001] This invention was made with government support under Grant No. EY024280 awarded by the National Institutes of Health. The government has certain rights in the invention.

RELATED APPLICATIONS

[0002] This application claims the benefit of U.S. Provisional Application No. 62/867,596, filed on June 27, 2019, which is incorporated herein by reference in its entirety.

BACKGROUND OF THE INVENTION

[0003] Major advances in the field of gene therapy have been achieved by using viruses to deliver therapeutic genetic material. The adeno-associated virus (AAV) has attracted considerable attention as a highly effective viral vector for gene therapy due to its low immunogenicity and ability to effectively transduce non-dividing cells. AAV has been shown to infect a variety of cell and tissue types, and significant progress has been made over the last decade to adapt this viral system for use in human gene therapy. AAV treatments for ocular tissues have been the focus of much research this decade, and a handful of AAV therapies have recently been approved for use in patients by the FDA.

SUMMARY OF THE INVENTION

[0004] Described herein are methods of delivering a cargo to an eye of a subject. In some embodiments, a subject is in need thereof. In some embodiments, the method comprises administering to an eye of a subject a rAAV particle. In some embodiments, a rAAV particle comprises (a) a capsid that is admixed with hyaluronic acid (HA) and (b) a cargo. In some embodiments, a rAAV particle comprises one or both of (a) a capsid that is admixed with hyaluronic acid (HA) and (b) a cargo. In some embodiments, a cargo is delivered to an eye.

In some embodiments, the HA is in direct contact with a rAAV particle capsid. In some embodiments, a rAAV capsid is at least partially coated with HA. These methods may be used in gene therapy-based treatment of several diseases of the eye. In some embodiments, a rAAV particle is administered intravitreally. In some embodiments, capsid comprises one or more surface-exposed patches of positively-charged residues. In some embodiments, the serotype of a capsid is rAAV2, or a variant thereof. In some embodiments, the serotype of a capsid is rAAV6, or a variant thereof. In some embodiments, the method further comprises pre-incubating a capsid with HA prior to administering the rAAV particle to the eye. In some embodiments, the capsid is pre-incubated with a buffer that comprises HA. In some embodiments, a buffer comprises HA in a concentration of 0.1%, 0.2%, 0.25%, 0.3%, 0.35%, 0.4%, 0.45%, 0.5%, 0.6%, 0.75%, or 1.0% weight by volume. In some embodiments, a rAAV particle is administered to the eye of a subject in a titer of about lxlO 10 vg/ml, 5 xlO 10 vg/ml, lxlO 11 vg/ml, 5 xlO 11 vg/ml, lxlO 12 vg/ml, 2xl0 12 vg/ml, 3xl0 12 vg/ml, 4xl0 12 vg/ml, about 5xl0 12 vg/ml, about lxlO 13 vg/ml, or about 5xl0 13 vg/ml. In some embodiments, a rAAV particle is administered to the eye of a subject in a titer of less than 5 xlO 11 vg/ml. In some embodiments, a cargo comprises a polynucleotide comprising a heterologous nucleic acid sequence. In some embodiments, a heterologous nucleic acid sequence is operably linked to a regulatory sequence that direct expression of the heterologous nucleic acid sequence in a photoreceptor cell, retinal pigment epithelium cell, retinal ganglion cell, bipolar cell, Miiller glial cell or astrocyte cell. In some embodiments, a regulatory sequence is selected from the group consisting of: a woodchuck hepatitis virus post-transcription regulatory element (WPRE), a polyadenylation signal sequence, an intron/exon junctions/splicing signal and any combination thereof. In some embodiments, a heterologous nucleic acid sequence encodes a therapeutic agent. In some embodiments, a therapeutic agent is a neurotrophic factor. In some embodiments, a neurotrophic factor is selected from the group consisting of: brain derived neurotrophic factor (BDNF), nerve growth factor (NGF), neurotrophin-3, ciliary neurotrophic factor (CNTF), an ephrin, glial cell line-derived neurotrophic factors (GDNF) and a combination thereof. In some embodiments, a therapeutic agent is an optogenetic actuator. In some embodiments, a optogenetic actuator is selected from the group consisting of: a bacteriorhodopsin, a halorhodopsin, a channelrhodopsin, a microbial sensory rhodopsin, a mammalian rhodopsin, a cone opsin, a melanopsin and a combination thereof. In some embodiments, a cargo is administered to treat a disease selected from the group consisting of: retinitis pigmentosa, leber Congenital Amaurosis, age related macular degeneration (AMD), wet AMD, dry AMD, uveitis, best disease, stargardts disease, usher syndrome, geographic atrophy, diabetic retinopathy, retinoschisis, achromatopsia, choroideremia, bardet biedl syndrome, a glycogen storage disease and a combination thereof. In some embodiments, the rAAV particle comprises an AAV7m8, an AAV-DJ, an AAV2/2-MAX, an AAVSHhlO, an AAVSHhlOY, an AAV3, an AAV3b, or an AAVFK03 capsid. In some embodiments, a capsid sequence is at least about 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% sequence identity with SEQ ID NO:l, SEQ ID NO:2, SEQ ID NO:3, SEQ ID NO:4, SEQ ID NO:5, SEQ ID NO:6, SEQ ID NO:7, SEQ ID NO:8, SEQ ID NO:9, or

SEQ ID NO: 10.

[0005] Described herein, in certain embodiments, is a method comprising co-administering an rAAV particle with hyaluronic acid. In some embodiments, the rAAV particle with hyaluronic acid is administered by intravitreal injection to one or both eyes of a mammal. In some embodiments, the AAV particle comprises a capsid comprising one or more surface- exposed patches of positively-charged residues. In some embodiments, the serotype of the capsid is AAV2 or a variant thereof. In some embodiments, the serotype of the capsid is AAV6 or a variant thereof. In some embodiments, the rAAV particle comprises an AAV7m8, an AAV-DJ, an AAV2/2-MAX, an AAVSHhlO, an AAVSHhlOY, an AAV3, an AAV3b, or an AAVLK03 capsid. In some embodiments, the capsid of the rAAV particle is pre-incubated with hyaluronic acid (HA) prior to administration to one or both eyes of a mammal. In some embodiments, the capsid is pre-incubated with a buffer that comprises HA. In some embodiments, the capsid is pre-incubated with HA for a duration of at least about 5 minutes, about 15 minutes, about 30 minutes, about 45 minutes, about 60 minutes or about 75 minutes. In some embodiments, the capsid is pre-incubated for a duration of about 15 minutes. In some embodiments, the capsid is pre-incubated with HA in a concentration of at least about 0.1%, 0.2%, 0.25%, 0.3%, 0.35%, 0.4%, 0.45%, 0.5%, 0.6%, 0.75%, 1.0%, 1.5%, 2.5%,

3.0%, 3.5%, 4%, 5% or 10% weight by volume. In some embodiments, the rAAV particle is administered in a titer of about lxlO 10 vg/ml, 5 xlO 10 vg/ml, lxlO 11 vg/ml, 5 xlO 11 vg/ml, lxlO 12 vg/ml, 2xl0 12 vg/ml, 3xl0 12 vg/ml, 4xl0 12 vg/ml, about 5xl0 12 vg/ml, about lxlO 13 vg/ml, or about 5xl0 13 vg/ml. In some embodiments, the rAAV particle is administered in a titer of less than 5 xlO 11 vg/ml. In some embodiments, the intravitreal injection is provided in a volume of about 200 pL, about 175 pL, about 160 pL, about 145 pL, about 130 pL, about 115 pL, about 100 pL, about 90 pL, about 80 pL, about 70 pL, about 60 pL, about 55 pL, about 50 pL, about 45 pL, about 35 pL, about 20 pL, about 10 pL, or about 5 pL. In some embodiments, the intravitreal injection is administered in a volume of about 50 pL. In some embodiments, the rAAV particle further comprises a polynucleotide comprising a

heterologous nucleic acid sequence. In some embodiments, a heterologous nucleic acid sequence or protein is a cargo of the rAAV. In some embodiments, the heterologous nucleic acid sequence is operably linked to regulatory sequences which direct expression of the heterologous nucleic acid sequence in a photoreceptor cell, retinal pigment epithelium cell, retinal ganglion cell, bipolar cell, Miiller glial cell or astrocyte cell. In some embodiments, the heterologous nucleic acid sequence encodes a therapeutic agent. In some embodiment, the therapeutic agent is for the treatment of a disease, condition or condition. In some

embodiments, the therapeutic agent is selected from an neurotrophic factor or an optogenetic actuator. In some embodiments, the disease, disorder or condition is age-related macular degeneration (AMD), wet AMD, dry AMD, or geographic atrophy. In some embodiments, the mammal is human. In some embodiments, production of the therapeutic agent a) preserves one or more photoreceptor cells or one or more RPE cells, b) restores one or more rod- and/or cone-mediated functions, c) restores visual behavior in one or both eyes, or d) any combination thereof. In some embodiments, production of the therapeutic agent persists in the one or more photoreceptor cells or the one or more RPE cells substantially for a period of at least three months following an initial administration of the rAAV particle into the one or both eyes of the mammal. In some embodiments, production of the therapeutic agent preserves one or more retinal ganglion cells, Miiller glial cells or astrocyte cells. In some embodiments, the therapeutic agent comprises a neurotrophic factor. In some embodiments, a neurotrophic factor is selected from the group consisting of: brain derived neurotrophic factor (BDNF), nerve growth factor (NGF), neurotrophin-3, ciliary neurotrophic factor (CNTF), an ephrin, glial cell line-derived neurotrophic factors (GDNF) and a combination thereof. In some embodiments, the disease, disorder or condition is retinitis pigmentosa or glaucoma. In some embodiments, production of the therapeutic agent preserves one or more retinal ganglion cells or retinal bipolar cells. In some embodiments, the therapeutic agent comprises an optogenetic actuator. In some embodiments, the optogenetic actuator is selected from a bacteriorhodopsin, a halorhodopsin, a channelrhodopsin, a microbial sensory rhodopsin, a mammalian rhodopsin, a cone opsin, a melanopsin, or a combination thereof. In some embodiment, the rAAV particle is not comprised in a chimeric viral/non- viral nanoparticle. In some embodiments, a capsid sequence is at least about 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% sequence identity with SEQ ID NO:l, SEQ ID NO:2,

SEQ ID NOG, SEQ ID NO:4, SEQ ID NOG, SEQ ID NOG, SEQ ID NOG, SEQ ID NOG, SEQ ID NO:9, or SEQ ID NO: 10.

[0006] In some embodiments, described herein is a method for providing a mammal in need thereof with a therapeutically effective amount of a therapeutic agent. In some embodiments, the method comprises intravitreally co-administering an rAAV particle with hyaluronic acid (HA) to one or both eyes of a mammal. In some embodiments, HA is in direct association with the rAAV capsid. In some embodiments, co-administering of an rAAV particle with hyaluronic acid is for a time effective to provide the mammal with a therapeutically-effective amount of the therapeutic agent. In some embodiments, the rAAV particle comprises an AAV2 or an AAV6 capsid, or a variant thereof. In some embodiments, the rAAV particle comprises a capsid variant selected from AAV7m8, an AAV-DJ, an AAV2/2-MAX, an AAVSHhlO, an AAVSHhlOY, an AAV3, an AAV3b, and an AAVLK03 capsid. In some embodiments, the therapeutic agent is a heterologous nucleic acid sequence. In some embodiments, the rAAV particle comprises a polynucleotide comprising a heterologous nucleic acid sequence. In some embodiments, the heterologous nucleic acid sequence is operably linked to regulatory sequences which direct expression of the heterologous nucleic acid sequence in a photoreceptor cell, retinal pigment epithelium cell, retinal ganglion cell, bipolar cell, Miiller glial cell or astrocyte cell. In some embodiments, the heterologous nucleic acid sequence encodes the therapeutic agent. In some embodiments, the therapeutic agent is selected from an neurotrophic factor or an optogenetic actuator.

[0007] In some embodiment, the therapeutic agent is for treatment of a disease, condition or disorder. In some embodiments, the disease, disorder or condition is age-related macular degeneration (AMD), wet AMD, dry AMD, or geographic atrophy. In some embodiments, the mammal is human. In some embodiments, production of the therapeutic agent a) preserves one or more photoreceptor cells or one or more RPE cells, b) restores one or more rod- and/or cone-mediated functions, c) restores visual behavior in one or both eyes, or d) any combination thereof. In some embodiments, production of the therapeutic agent persists in the one or more photoreceptor cells or the one or more RPE cells substantially for a period of at least three months following an initial administration of the rAAV particle into the one or both eyes of the mammal. In some embodiments, production of the therapeutic agent preserves one or more retinal ganglion cells, Miiller glial cells or astrocyte cells. In some embodiments, the therapeutic agent comprises a neurotrophic factor. In some embodiments, a neurotrophic factor is selected from the group consisting of: brain derived neurotrophic factor (BDNF), nerve growth factor (NGF), neurotrophin-3, ciliary neurotrophic factor (CNTF), an ephrin, glial cell line-derived neurotrophic factors (GDNF) and a combination thereof. In some embodiments, the disease, disorder or condition is retinitis pigmentosa or glaucoma. In some embodiments, production of the therapeutic agent preserves one or more retinal ganglion cells or retinal bipolar cells. In some embodiments, the therapeutic agent comprises an optogenetic actuator. In some embodiments, the optogenetic actuator is selected from a bacteriorhodopsin, a halorhodopsin, a channelrhodopsin, a microbial sensory rhodopsin, a mammalian rhodopsin, a cone opsin, a melanopsin, or a combination thereof. In some embodiment, the rAAV particle is not comprised in a chimeric viral/non- viral nanoparticle. In some embodiments, a capsid sequence is at least about 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% sequence identity with SEQ ID NO:l, SEQ ID NO:2,

SEQ ID NO:3, SEQ ID NO:4, SEQ ID NO:5, SEQ ID NO:6, SEQ ID NO:7, SEQ ID NO:8, SEQ ID NO:9, or SEQ ID NO: 10.

[0008] In some embodiments, described herein is a method for treating or ameliorating one or more symptoms of a disease, disorder or condition. In some embodiments, the method comprises intravitreally co-administering an rAAV particle with hyaluronic acid to one or both eyes of a mammal in need thereof. In some embodiments, the co-administration is for a time sufficient to treat or ameliorate one or more symptoms of a disease, disorder or condition in the mammal. In some embodiments, the rAAV particle comprises i) a polynucleotide encoding a therapeutic agent and ii) an AAV2 or an AAV6 capsid, or a variant thereof. In some embodiments, the rAAV particle comprises an AAV7m8, an AAV- DJ, an AAV2/2-MAX, an AAVSHhlO, an AAVSHhlOY, an AAV3, an AAV3b, or an AAVLK03 capsid. In some embodiments, the disease, disorder or condition is age-related macular degeneration (AMD), wet AMD, dry AMD, or geographic atrophy. In some embodiments, the mammal is human. In some embodiments, production of the therapeutic agent a) preserves one or more photoreceptor cells or one or more RPE cells, b) restores one or more rod- and/or cone-mediated functions, c) restores visual behavior in one or both eyes, or d) any combination thereof. In some embodiments, production of the therapeutic agent persists in the one or more photoreceptor cells or the one or more RPE cells substantially for a period of at least three months following an initial administration of the rAAV particle into the one or both eyes of the mammal. In some embodiments, production of the therapeutic agent preserves one or more retinal ganglion cells, Miiller glial cells or astrocyte cells. In some embodiments, the therapeutic agent comprises a neurotrophic factor. In some embodiments, a neurotrophic factor is selected from the group consisting of: brain derived neurotrophic factor (BDNF), nerve growth factor (NGF), neurotrophin-3, ciliary neurotrophic factor (CNTF), an ephrin, glial cell line-derived neurotrophic factors (GDNF) and a combination thereof. In some embodiments, the disease, disorder or condition is retinitis pigmentosa or glaucoma. In some embodiments, production of the therapeutic agent preserves one or more retinal ganglion cells or retinal bipolar cells. In some embodiments, the therapeutic agent comprises an optogenetic actuator. In some embodiments, the optogenetic actuator is selected from a bacteriorhodopsin, a halorhodopsin, a channelrhodopsin, a microbial sensory rhodopsin, a mammalian rhodopsin, a cone opsin, a melanopsin, or a combination thereof. In some embodiment, the rAAV particle is not comprised in a chimeric viral/non-viral nanoparticle.

[0009] In some embodiments, described herein is a method for providing a mammal in need thereof with a therapeutically effective amount of a therapeutic agent. In some embodiments the method comprises intravitreally co-administering an rAAV particle with hyaluronic acid to one or both eyes of a mammal. In some embodiments, the co-administering of an rAAV particle with hyaluronic acid to one or both eyes of a mammal is for a time effective to provide the mammal with a therapeutically-effective amount of the therapeutic agent. In some embodiments, the rAAV particle comprises an AAV7m8, an AAV-DJ, an AAV2/2- MAX, an AAVSHhlO, an AAVSHhlOY, an AAV3, an AAV3b, or an AAVLK03 capsid. In some embodiments, a capsid sequence is at least about 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% sequence identity with SEQ ID NO:l, SEQ ID NO:2,

SEQ ID NO:3, SEQ ID NO:4, SEQ ID NO:5, SEQ ID NO:6, SEQ ID NO:7, SEQ ID NO:8, SEQ ID NO:9, or SEQ ID NO: 10.

[0010] In certain aspects, described herein is a method for treating or ameliorating one or more symptoms of a disease, disorder or condition. In some embodiments, a method comprises intravitreally co-administering an rAAV particle with hyaluronic acid to one or both eyes of a mammal in need thereof for a time sufficient to treat or ameliorate the one or more symptoms of the disease, disorder or condition in the mammal. In some embodiments, the rAAV particle comprises i) a polynucleotide encoding a therapeutic agent and ii) an AAV7m8, an AAV-DJ, an AAV2/2-MAX, an AAVSHhlO, an AAVSHhlOY, an AAV3, an AAV3b, or an AAVLK03 capsid. In some embodiments, the capsid is selected from

AAV7m8, AAV-DJ, AAV2/2-MAX, AAVSHhlO, and AAVSHhlOY. In some embodiments, the capsid is selected from AAV3, AAV3b, and AAVLK03. In some embodiments, the capsid comprises non-native amino acid substitutions at amino acid residues of a wild-type AAV2 capsid as set forth in SEQ ID NO: 2. In some embodiments, the non-native amino acid substitutions comprise one or more of Y272F, Y444F, T491V, Y500F, Y700F, Y704F and Y730F. In some embodiments, the capsid comprises non-native amino acid substitutions at amino acid residues of a wild-type AAV6 capsid as set forth in SEQ ID NO: 6. In some embodiments, the non-native amino acid substitutions comprise one or more of Y445F, Y705F, Y731F, T492V and S663V. In some embodiments, the capsid comprises a non-native amino acid substitution at amino acid residue 531 of a wild-type AAV1 capsid as set forth in SEQ ID NO: 1. In some embodiments, the non-native amino acid substitution is E531K. In some embodiments, the capsid comprises AAV7BP2. In some embodiments, the capsid comprises AAV2G9. In some embodiments, the capsid comprises non-native amino acid substitutions at amino acid residues 533 and/or 733 of a wild-type AAV8 capsid as set forth in SEQ ID NO: 8. In some embodiments, the non-native amino acid substitution is E533K and/or Y733F. In some embodiments, the capsid comprises non-native amino acid

substitutions of a wild-type AAV2 capsid as set forth in SEQ ID NO: 2. In some

embodiments, the non-native amino acid substitutions comprise: (a) Y444F; (b)

Y444F+Y500F+Y730F; (c) Y272F+Y444F+Y500F+Y730F; (d)

Y 444F+ Y 500F+ Y730F+T491 V ; or (e) Y272F+Y444F+Y500F+Y730F+T491V, or at equivalent amino acid positions corresponding thereto in any one of the wild-type AAV1, AAV3, AAV4, AAV5, AAV7, AAV9, or AAV10 capsid proteins, as set forth, respectively, in SEQ ID NO: 1, SEQ ID NO: 3, SEQ ID NO: 4, SEQ ID NO: 5, SEQ ID NO: 7, SEQ ID NO: 9, or SEQ ID NO: 10. In some embodiments, the capsid comprises non-native amino acid substitutions of a wild-type AAV6 capsid as set forth in SEQ ID NO: 6, wherein the non-native amino acid substitutions comprise: (a) Y445F; (b) Y705F+Y731F; (c) T492V; (d) Y705F+Y731F+T492V; (e) S663V; or (f) S663V+T492V. In some embodiments, the disease, disorder or condition is age-related macular degeneration (AMD), wet AMD, dry AMD, or geographic atrophy. In some embodiments, the mammal is human. In some embodiments, production of the therapeutic agent a) preserves one or more photoreceptor cells or one or more RPE cells, b) restores one or more rod- and/or cone-mediated functions, c) restores visual behavior in one or both eyes, or d) any combination thereof. In some embodiments, production of the therapeutic agent persists in the one or more photoreceptor cells or the one or more RPE cells substantially for a period of at least three months following an initial administration of the rAAV particle into the one or both eyes of the mammal. In some embodiments, production of the therapeutic agent preserves one or more retinal ganglion cells, Miiller glial cells or astrocyte cells. In some embodiments, the therapeutic agent comprises a neurotrophic factor. In some embodiments, a neurotrophic factor is selected from the group consisting of: brain derived neurotrophic factor (BDNF), nerve growth factor (NGF), neurotrophin-3, ciliary neurotrophic factor (CNTF), an ephrin, glial cell line-derived neurotrophic factors (GDNF) and a combination thereof. In some embodiments, the disease, disorder or condition is retinitis pigmentosa or glaucoma. In some embodiments, production of the therapeutic agent preserves one or more retinal ganglion cells or retinal bipolar cells. In some embodiments, the therapeutic agent comprises an optogenetic actuator. In some embodiments, the optogenetic actuator is selected from a bacteriorhodopsin, a halorhodopsin, a channelrhodopsin, a microbial sensory rhodopsin, a mammalian rhodopsin, a cone opsin, a melanopsin, or a combination thereof. In some embodiment, the rAAV particle is not comprised in a chimeric viral/non-viral nanoparticle. In some embodiments, a capsid sequence is at least about 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% sequence identity with SEQ ID NO:l, SEQ ID NO:2, SEQ ID NO:3, SEQ ID NO:4, SEQ ID NO:5, SEQ ID NO:6, SEQ ID NO:7, SEQ ID NO:8, SEQ ID NO:9, or

SEQ ID NO: 10.

[0011] In certain embodiments, described herein is a buffer for storing a mixture of AAV and hyaluronic acid (HA). In some embodiments, the buff comprises: (a) HA in a concentration of about 0.1%, 0.2%, 0.25%, 0.3%, 0.35%, 0.4%, 0.45%, 0.5%, 0.6%, 0.75%, 1.0%, 2.0%, 3.0%, 5.0% or 10%weight by volume; (b) balanced salt solution (BSS); (c) artificial cerebrospinal fluid; and/or (d) phosphate buffered saline (PBS). In some embodiments, the buffer further comprises (e) Ringer’s lactate solution; (f) TMN200 solution; (g) Polysorbate 20; and/or(h) poloxamer 188. In some embodiments, the HA is in a concentration of 0.4% weight by volume.

BRIEF DESCRIPTIONS OF THE DRAWINGS

[0012] The following drawings form part of the present specification and are included to demonstrate certain aspects of the present disclosure. The disclosure may be better understood by reference to the following description taken in conjunction with the accompanying drawings, in which like reference numerals identify like elements, and in which:

[0013] Fig. 1 is a schematic of the chemical structure of Hyaluronic Acid (HA), which is a linear concatemer of a dimer of D-glucaronic acid and N-axcetyl-D-glucosamine.

[0014] Figs. 2A-2B illustrate the effects of the administration to 661W murine photoreceptor cells in vitro of rAAV particles whose capsids had been pre-incubated with HA (brand name Healon ® ) at 5 minutes, 15 minutes, and 1 hour prior to infection. Pre-treatment with HA increases transduction of 661W cells in AAV2 (Fig. 2A). 661W cells in AAV5 are illustrated in Fig. 2B. Capsids were preincubated with Healon ® in a ratio of 3:1 (AAV:Healon ® ) and there after injected into cells at a multiplicity of infection (MOI) of 2000. Controls included uninfected cells and cells infected with vector alone.

[0015] Fig. 3 illustrates the effects of administration to HEK293T cells in vitro injected at MOIs of 5000 and 10,000 of self-complementary (sc) rAAV6-based particles expressing an mCherry reporter transgene operably controlled by an smCBA promoter. Where indicated, AAV6 capsid and AAV6 capsid variants were pre-incubated with HA prior to infection. Variants tested include AAV6(D532N) and AAV6-3pmut. mCherry expression was calculated by fluorescence-activated cell sorting (FACS) by multiplying the percentage of positive cells by the mean fluorescence intensity in each sample. Error bars represent -1 standard deviation. HA pre-treatment substantially increased transduction of HEK293T cells by these particles.

[0016] Fig. 4 illustrates the results of a validation experiment in which

immunohistochemistry (IHC) was evaluated against CD44 cell surface receptor in three cell lines. ARPE-19 cells (Fig. 4A), a human RPE line, exhibited CD44 expression (as indicated by arrows); whereas HEK293T (Fig. 4C) and 661W cells (Fig. 4B) did not express CD44.

[0017] Fig. 5 illustrates the results of a heparan binding elution profile of AAV2 capsid alone and AAV2 capsid pre-incubated with Healon ® at different salt concentrations between 50mM and 1M NaCl. HA pre-treatment does not alter interaction of AAV2 capsids with heparan sulfate proteoglycan (HSPG) on cell membranes.

[0018] Figs. 6A-6B illustrate the effects of HA pre-incubation and co-administration on transduction of AAV2-mediated mCherry expression after intravitreal injection of rAAV2 into whole mouse retina in vivo. Controls included uninfected cells and cells infected with vector alone. Fig. 6B represents a quantification of mCherry expression observed in Fig. 6A, as measured by flow cytometry. mCherry expression is indicated by arrows in Fig. 6A.

[0019] Figs. 7A-7C illustrate the effects of intravitreal injection of rAAV particles comprising AAV2 capsid variant DGE-DF, with or without pre-treatment of the capsid with HA, into three 4-week old Nrl-GFP mice. Fundus images taken at 2 weeks (Fig. 7A) and 4 weeks (Fig. 7B) post- injection are shown. Fig. 7C represents a quantification of the mCherry expression observed in Figs. 7A and 7B, as measured by flow cytometry data aggregated among the three mice. mCherry expression is indicated by arrows in Figs. 7A-7B.

[0020] Figs. 8A-8C illustrate the effects of intravitreal injection of rAAV particles comprising AAV2 capsid variant P2-V2, with or without pre-treatment of the capsid with HA, into three Nrl-GFP mice. Fundus images taken at 2 weeks (Fig. 8A) and 4 weeks (Fig. 8B) post-injection are shown. Fig. 8C represents a quantification of the mCherry expression observed in Figs. 8 A and 8B, as measured by flow cytometry data aggregated among the three mice. mCherry expression is indicated by arrows in Figs. 8A-8B.

[0021] Figs. 9A-9C illustrate the effects of intravitreal injection of rAAV particles comprising AAV2 capsid variant P2-V3, with or without pre-treatment of the capsid with HA, into Nrl-GFP mice. Fundus images taken at 2 weeks (Fig. 9A) and 4 weeks (Fig. 9B) post-injection are shown. Fig. 9C represents a quantification of the mCherry expression observed in Figs. 9A and 9B, as measured by flow cytometry data aggregated among the two mice. mCherry expression is indicated by arrows in Figs. 9A-9B.

[0022] Figs. 10A-10C illustrate the effects of intravitreal injection of rAAV particles comprising AAV2 capsid variant ME-B(Y-F+T-V), with or without pre-treatment of the capsid with HA, into three Nrl-GFP mice. Fundus images taken at 2 weeks (Fig. 10A) and 4 weeks (Fig. 10B) post-injection are shown. Fig. IOC represents a quantification of the mCherry expression observed in Figs. 10A and 10B, as measured by flow cytometry data aggregated among the three mice. mCherry expression is indicated by arrows in Figs. 10A- 10B.

[0023] Fig. 11 is a schematic depicting the aggregated flow cytometry data for four capsids: DGE-DF, P2-V2, P2-V3, and ME-B(Y-F+T-V).

[0024] Figs. 12A-12E are schematics showing the electroretinogram (ERG) results following intravitreal administration of HA-treated DGE-DF (Fig. 12A), P2-V2 (Fig. 12B), P2-V3 (Fig. 12C), ME-B(Y-F+T-V) (Fig. 12D) and aggregated data (Fig. 12E). Treatment and co administration with HA had no impact on retinal cell function. In the top graph of Fig. 12E, N=l l. P-value=0.478132. In the bottom graph of Fig. 12E, N=l l. P-value=0.524563.

DETAILED DESCRIPTION

[0025] The present disclosure provides novel methods, compositions and buffers for administration of rAAV particles having enhanced transduction properties, comprising the pre-incubation and/or co-administration of AAV capsids and hyaluronic acid. In some embodiments, methods disclosed herein rely on treatment with a natural material that is already comprised in the mammalian eye. In some embodiments, methods disclosed herein do not rely on the modification of the capsid and function through a mechanism that does not interfere with AAV adhesion, thus fulfilling a long-felt need in the art. Advantageously, the novel methods of rAAV particle administration disclosed herein have improved efficiency in transducing the retina of the mammalian eye, and in particular, in transducing the

photoreceptor (PR) and retinal pigment epithelial (RPE) cells in vivo.

[0026] Intraocular AAV therapies generally suffer from poor transduction profiles.

Achieving adequate transduction of rAAV particles of the AAV2 serotype has been particularly elusive in view of the propensity of AAV2 capsids to aggregate. This aggregation is in large part due to the presence of positively-charged amino acid residues on the capsid surface. AAV6 capsids, along with several psuedotypes and variants of AAV6, AAV2 and other capsids, have similar positively-charged“patches” of residues on their surfaces that are exposed to a milieu.

[0027] Two common approaches of gene delivery into the retina are intravitreal and subretinal injections. Subretinal injections require administration into the subretinal space between the retinal pigment epithelium (RPE) and photoreceptors to transduce these neurons. To achieve delivery of the vector, a needle must penetrate the retina and, in doing so, detaches the photoreceptor cell layer from the RPE.

[0028] Intravitreal injection is less invasive, and is thus a preferred route of administration that is routinely performed for clinical human subjects. Nonetheless, few AAV serotypes have exhibited efficient transduction by intravitreal delivery. This occurs in part due to presence of the inner limiting membrane (ILM), a basement membrane between the vitreous and neural retina that is replete with heparan sulfate proteoglycans, which acts as a barrier for effective spread of the vector through the retina by sequestering capsids with high heparan sulfate affinity. In particular, AAV2 vectors have generally failed to transduce photoreceptor cells upon administration by intravitreal injection.

[0029] Inherited retinal degenerative diseases are a clinically promising focus of AAV- mediated gene therapy. These diseases arise from pathogenic mutations in mRNA transcripts expressed in the eye’s photoreceptor cells or retinal pigment epithelium (RPE), leading to cell death and structural deterioration. Prior methods designed to reduce aggregation of AAV2 and AAV6 capsids have involved pre-treatment of capsids with high-salt buffers. But these buffers are toxic to the eye, making administration after pre-treatment infeasible.

[0030] Hyaluronic acid (HA) is an anionic, non-sulfated glycosaminoglycan that is the major component of the human vitreous, where it is covalently linked to extracellular matrix. In addition, HA is an FDA-approved substance with optical and viscoelastic properties that make it a good substitute for human vitreous. HA typically enters cells through endocytosis by cell surface glycoproteins, and in particular the receptor CD44. HA can also enter the cell also by micropinocytosis.

[0031] Adenovirus transduction occurred via a mechanism dependent on CD44 which did not purport to impact the efficiency of adenovirus infectivity. Gains in transduction were primarily a function of increased transgene expression mediated by liberation of the intracellular domain of CD44 which occurred as a consequence of HA binding to CD44. However, results in adenoviral therapies cannot readily be recapitulated in recombinant AAV therapies. Further, because excipient binding to CD44 receptors may interfere with the ability of the AAV capsid to bind the heparan sulfate proteoglycan (HSPG) component of the ILM, which is critical for cell recognition and internalization, a need exists for administration with an excipient that functions through a CD44-independent mechanism.

[0032] An alternative,“subILM” injection method has been developed in which AAV vector is placed in a surgically induced, hydrodissected space, or bleb, between the ILM and neural retina. SubILM injections generally avoid the ILM barrier to AAV spread, as described above.

[0033] The described methods provide for effective vector transduction through noninvasive intravitreal delivery at potentially lower vector doses than those of subILM delivery. The disclosure is based, at least in part, on the discovery that coating AAV capsids having positively-charged patches on the surface prevents aggregation of these particles in the vitreous. As used herein,“surface-exposed patches” of the capsid refer to three-dimensional areas of contiguous or abutting (or substantially contiguous or abutting) cationic residues on the capsid that are exposed to milieu. Prevention of capsid aggregation enables the lateral spread of single particle suspensions of AAV to spread throughout the retina to reach target cells such as retinal ganglion cells (RGCs), Muller glia cells, and other PRs. This enhanced spread of AAV suggests that current intravitreal doses of AAV may be reduced to achieve the same transduction efficiency in target cells, which provides for a safer gene therapy.

[0034] As described herein, the pre-incubation of capsids having cationic patches with hyaluronic acid increases retinal transduction by intravitreally-delivered rAAV particles, and is independent of the capsid sequence. In some embodiments, enhancement of transduction by HA function independently of CD44. In some embodiments, HA as a vehicle does not interfere with AAV binding of HSPG in the ILM and vitreous.

[0035] In some embodiments, the described methods are suitable for use with a variety of AAV capsid serotypes and presudotypes. The described methods improve vector potency, thus lowering the concentration of rAAV particle required to achieve the desired effect. Accordingly, the described methods are capable of increasing safety and reducing expense of manufacturing or rAAV. These methods minimize the surgical risks associated with intravitreal injection, lower the cost of care, and increase the accessibility of gene therapies. The described methods increase retinal transduction of AAV in any clinical setting where AAV2 or AAV6-based capsids are delivered by intravitreal injection.

Certain Terminology

[0036] Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this disclosure belongs. Although any methods and compositions similar or equivalent to those described herein can be used in the practice or testing of the present disclosure, the preferred methods and compositions are described herein. For purposes of the present disclosure, the following terms are defined below:

[0037] As used herein, the terms“hyaluronic acid” and“HA” encompass hyaluronic acid, hyaluronan, Healon ® , and sodium hyaluronate, without regard to the molecular weight or mass thereof. That is, these terms are meant to encompass hyaluronic acids of any molecular weight known or used in the art. The term also encompasses variants of hyaluronic acid, including but not limited to truncated and chemically modified versions of HA. A“variant” of HA is at least about 80% identical, at least about 90% identical, at least about 95% identical, at least about 96% identical, at least about 97% identical, at least about 98% identical, at least about 99% identical, at least about 99.5% identical, or at least about 99.9% identical to a wild type HA sequence.

[0038] As used herein, the terms“intravitreal injection,”“intravitreal delivery,” and “intravitreal administration,” refer to injection of material in and to the vitreous of the eye. This term does not encompass the leakage of material, e.g. AAV vector or particles, from a bleb created by a subILM injection to the vitreous or retina.

[0039] As used herein, the term“optogenetic actuator” refers to light-sensitive ion channel proteins that are capable of modulating the activities of neurons in living tissue, e.g. neurons in retinal tissue, that have been genetically modified to express these proteins. Exemplary optogenic actuators include, but are not limited to, halorhodopsin, melanopsin, cone opsins, channel rhodopsins, bacteriorhodopsin, mammalian rhodopsin and archea-associated opsins.

[0040] The term“subject,” as used herein, describes an organism, including mammals such as primates, to which treatment with the compositions according to the present disclosure can be provided. Mammalian species that can benefit from the described methods of treatment include, but are not limited to, apes; chimpanzees; orangutans; humans; monkeys;

domesticated animals such as dogs and cats; livestock such as horses, cattle, pigs, sheep, goats, and chickens; and other animals such as mice, rats, guinea pigs, and hamsters.

[0041] The term“treatment” or any grammatical variation thereof ( e.g ., treat, treating, and treatment etc.), as used herein, includes but is not limited to, alleviating a symptom of a disease or condition; and/or reducing, suppressing, inhibiting, lessening, ameliorating or affecting the progression, severity, and/or scope of a disease or condition. Accordingly, the term "treating" includes the administration of the compounds, compositions, constructs or agents of the present invention to prevent, delay, alleviate, arrest or inhibit development of the symptoms or conditions associated with a disease. The term "therapeutic effect" refers to the reduction, elimination, or prevention of the disease, symptoms of the disease, or side effects of the disease in the subject.

[0042] As used herein, the term“pre-treatment” in some embodiments refers to the application of exogenous material to an AAV vector, prior to administration of the vector to a subject or subject cell.

[0043] The term“effective amount,” as used herein, refers to an amount that is capable of treating or ameliorating a disease or condition or otherwise capable of producing an intended therapeutic effect.

[0044] As used herein, the terms“engineered” and“recombinant” cells are intended to refer to a cell into which an exogenous polynucleotide segment (such as DNA segment that leads to the transcription of a biologically active molecule) has been introduced. Therefore, engineered cells are distinguishable from naturally occurring cells, which do not contain a recombinantly introduced exogenous DNA segment. Engineered cells are, therefore, cells that comprise at least one or more heterologous nucleic acid segments introduced through the hand of man.

[0045] The term“promoter,” as used herein refers to a region or regions of a nucleic acid sequence that regulates transcription.

[0046] The term“regulatory element,” as used herein, refers to a region or regions of a nucleic acid sequence that regulates transcription. Exemplary regulatory elements include, but are not limited to, enhancers, post-transcriptional elements, transcriptional control sequences, and such like. [0047] The tern“vector,” as used herein, refers to a nucleic acid molecule (typically comprised of DNA) capable of replication in a host cell and/or to which another nucleic acid segment can be operatively linked so as to bring about replication of the attached segment. A plasmid, cosmid, or a virus is an exemplary vector.

[0048] The term“substantially corresponds to,”“substantially homologous,” or“substantial identity,” as used herein, denote a characteristic of a nucleic acid or an amino acid sequence, wherein a selected nucleic acid or amino acid sequence has at least about 70 or about 75 percent sequence identity as compared to a selected reference nucleic acid or amino acid sequence. More typically, the selected sequence and the reference sequence will have at least about 76, 77, 78, 79, 80, 81, 82, 83, 84 or even 85 percent sequence identity, and more preferably, at least about 86, 87, 88, 89, 90, 91, 92, 93, 94, or 95 percent sequence identity. More preferably still, highly homologous sequences often share greater than at least about 96, 97, 98, or 99 percent sequence identity between the selected sequence and the reference sequence to which it was compared. The percentage of sequence identity may be calculated over the entire length of the sequences to be compared, or may be calculated by excluding small deletions or additions which total less than about 25 percent or so of the chosen reference sequence. The reference sequence may be a subset of a larger sequence, such as a portion of a gene or flanking sequence, or a repetitive portion of a chromosome. However, in the case of sequence homology of two or more polynucleotide sequences, the reference sequence will typically comprise at least about 18-25 nucleotides, more typically at least about 26 to 35 nucleotides, and even more typically at least about 40, 50, 60, 70, 80, 90, or even 100 or so nucleotides.

[0049] When highly-homologous fragments are desired, the extent of percent identity between the two sequences will be at least about 80%, preferably at least about 85%, and more preferably about 90% or 95% or higher, as readily determined by one or more of the sequence comparison algorithms well-known to those of skill in the art, such as e.g., the FASTA program analysis described by Pearson and Lipman (1988) and blastn computer program based on the algorithm of Brutlag et al. (Comp. App. Biosci. 6:237-245 (1990). A preferred method for determining the best overall match between a query sequence (e.g., a sequence of the present disclosure) and a subject sequence, also referred to as a global sequence alignment, can be determined using the FASTA or blastn. In a sequence alignment the query and subject sequences are either both nucleotide sequences or both amino acid sequences. The result of said global sequence alignment is expressed as percent identity. Preferred parameters used in a FASTA amino acid alignment are: Matrix=PAM 0, k-tuple=2, Mismatch Penalty=l, Joining Penalty=20, Randomization Group Length=0, Cutoff Score=l, Window Size=sequence length, Gap Penalty=5, Gap Size Penalty=0.05, Window Size=500 or the length of the subject amino acid sequence, whichever is shorter. Whether a nucleotide is matched/aligned is determined by results of the FASTA sequence alignment. This percentage is then subtracted from the percent identity, calculated by the above FASTA program using the specified parameters, to arrive at a final percent identity score. This final percent identity score is what is used for the purposes of the present disclosure.

[0050]“Percent (%) identity” refers to the extent to which two sequences (nucleotide or amino acid) have the same residue at the same positions in an alignment. For example,“an amino acid sequence is X% identical to SEQ ID NO: Y” refers to % identity of the amino acid sequence to SEQ ID NO: Y and is elaborated as X% of residues in the amino acid sequence are identical to the residues of sequence disclosed in SEQ ID NO: Y. Generally, computer programs are employed for such calculations. Exemplary programs that compare and align pairs of sequences, include ALIGN (Myers and Miller, 1988), FASTA (Pearson and Lipman, 1988; Pearson, 1990) and gapped BLAST (Altschul et al., 1997), BLASTP, BLASTN, or GCG (Devereux et al., 1984).

[0051] The term“operably linked,” as used herein, refers to that the nucleic acid sequences being linked are typically contiguous, or substantially contiguous, and, where necessary to join two protein coding regions, contiguous and in reading frame. However, since enhancers generally function when separated from the promoter by several kilobases and intronic sequences may be of variable lengths, some polynucleotide elements may be operably linked but not contiguous.

[0052] As used herein, the term“variant” refers to a molecule (e.g. a capsid) having characteristics that deviate from what occurs in nature, e.g., a“variant” is at least about 80% identical, at least about 90% identical, at least about 95% identical, at least about 96% identical, at least about 97% identical, at least about 98% identical, at least about 99% identical, at least about 99.5% identical, or at least about 99.9% identical to the wild type capsid. Variants of a protein molecule, e.g. a capsid, may contain modifications to the amino acid sequence (e.g., having 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 10-15, or 15-20 amino acid

substitutions) relative to the wild type protein sequence, which arise from point mutations installed into the nucleic acid sequence encoding the capsid protein. These modifications include chemical modifications as well as truncations, such as truncations at the N- or C- terminus of a capsid protein sequence.

[0053] As used herein, singular forms“a”,“and,” and“the” include plural referents unless the context clearly indicates otherwise.

[0054] As used herein, all numerical values or numerical ranges include whole integers within or encompassing such ranges and fractions of the values or the integers within or encompassing ranges unless the context clearly indicates otherwise. Thus, for example, reference to a range of 90-100%, includes 91%, 92%, 93%, 94%, 95%, 95%, 96%, 97%, etc., as well as 91.1%, 91.2%, 91.3%, 91.4%, 91.5%, etc., 92.1%, 92.2%, 92.3%, 92.4%, 92.5%, etc., and so forth.

[0055]“About” a number, as used herein, refers to range including the number and ranging from 10% below that number to 10% above that number. “About” a range refers to 10% below the lower limit of the range, spanning to 10% above the upper limit of the range

Methods

[0056] Illustrative embodiments of the disclosure are described below. In some

embodiments, the present disclosure provides methods of use of HA as a vehicle for administration of AAV particles to the eyes of a mammal via intravitreal injection. In some embodiments, the presently described methods provide for pre-incubation with HA and/or co administration with HA. In some embodiments, the disclosure further provides buffers for storage of HA and AAV capsids.

[0057] In some embodiments, the intravitreal administration methods described herein are capable of enhancing the transduction capacity of any AAV capsid or capsid variant having one or more surface-exposed patches of positively-charged residues. In some embodiments, a patch of positively charged residues comprises a plurality of positively-charged residues. In some embodiments, a patch of positively-charged residues comprises at least about 2, 3, 4, 5, 6, 7, 8, 9, 10 ,11, 12, 13, 15, 20, 30 ,50 ,60 positively-charged residues. In some

embodiments, the negative charge of the hyaluronic acid interacts electrostatically with the positively-charged resides of the capsid. In some embodiments, the HA is in direct contact or association with the AAV capsid. In some embodiments, HA is not in direct contact with the AAV capsid. In some embodiments, the AAV capsid is at least partially coated by the HA. In some embodiments, the HA effectively coats the cationic patches of the capsid, shielding it from aggregating with the cationic patches of other capsids after administration to the eye. In some embodiments, this shielding effect liberates the capsid to spread laterally throughout the vitreous and transduce target RPE and PR cells, such as RGCs, Muller cells, astrocytes, and bipolar cells.

[0058] In some embodiments, the endogenous hyaluronic acid molecules covalently linked to the extracellular matrix of the vitreous and ILM of the host cell do not exert any deterrent effect on the diffusion or transduction of AAV capsids administered with HA.

[0059] In some embodiments, the mechanism of enhancement of AAV transduction and diffusion provided by pre-treatment and administration with HA is independent of interaction with the CD44 cell surface receptor. In some embodiments, the methods provided herein do not interfere with basal AAV capsid interaction with HSPG on target cell surfaces or cell surface recognition, while still facilitating AAV particles to diffuse freely throughout the vitreous.

[0060] The vitreous humor is composed of 99% water, collagen fibrils and

glycosaminoglycans (GAGs). The vast majority of GAGs is hyaluronic acid (HA), with the concentration of HA in the human eye being 200 ug/mL. HA is a linear concatemer of a dimer of D-glucaronic acid and N-acetyl-D-glucosamine (see Fig. 1). HA forms folds, loops and turns creating‘network’ structure, which confers a viscoelastic property to the molecule. HA may concatermize into a variety of lengths which results in a variety of different molecular weights, ranging from at least 20 kilodaltons (kDa) to 5,000 kDa.

[0061] In some embodiments, provided herein are methods comprising administering rAAV particles and HA by intravitreal injection to the eyes of a subject, and/or pre-treating rAAV particles with HA prior to administration to a subject, wherein the rAAV capsid has one or more surface-exposed cationic patches, i.e., three-dimensional areas of contiguous or abutting (or substantially contiguous or abutting) cationic residues on surfaces of the capsid that are exposed to milieu. The relevant mileu may be the vitreous, the inner limiting membrane (ILM), the subretinal space, an extracellular matrix, a cell membrane, and/or a cell cytosol.

In some embodiments, administration is sequential administration or co-administration.

Pre-incubations

[0062] In some embodiments, in the methods described herein, the capsid is pre-incubated with the HA for a duration of at least about 1 minute, about 3 minutes, about 5 minutes, about 15 minutes, about 30 minutes, about 45 minutes, about 60 minutes, about 90 minutes, about 120 minutes or about 175 minutes or more prior to administration to a subject. In some embodiments, the HA is in direct contact with the AAV capsid. In some embodiments, the capsid is pre-incubated with HA for about 15 minutes. In some embodiments, the capsid is pre-incubated with HA for more than about 15 minutes.

[0063] In some embodiments, the capsid is pre-incubated with a buffer comprising HA for at least about 1 minute, about 3 minutes, about 5 minutes, about 15 minutes, about 30 minutes, about 45 minutes, about 60 minutes, about 90 minutes, about 120 minutes or about 175 minutes or more minutes. In some embodiments, the capsid is pre-incubated with a buffered comprising HA for about 15 minutes, or more than about 15 minutes. In some embodiments, the capsid is pre-incubated with a buffer comprising HA in a concentration of at least about 0.1%, 0.2%, 0.25%, 0.3%, 0.35%, 0.4%, 0.45%, 0.5%, 0.6%, 0.75%, 1.0%, 2%, 3%, 5%,

10%, 15% or 20.0% weight by volume (w/v) and BSS. In some embodiments, the capsid is pre-incubated with buffer comprising HA in a concentration of about 0.4% w/v, and one or more of BSS, artificial cerebrospinal fluid (CSF), and PBS. In some embodiments, the capsid is pre-incubated with a buffer comprising HA in a concentration of about 0.4% w/v, and one or more of BSS, artificial CSF, PBS, Ringer’s lactate solution, TMN200 solution, polysorbate 20, and poloxamer 188.

[0064] In some embodiments of the described methods, the capsid is pre-incubated with HA in a concentration of at least about 0.1%, 0.2%, 0.25%, 0.3%, 0.35%, 0.4%, 0.45%, 0.5%, 0.6%, 0.75%, 1.0%, 2%, 3%, 5%, 10%, 15% or 20.0% w/v. In some embodiments, the capsid is pre-incubated with HA in a concentration of about 0.4% w/v. In some embodiments, the capsid is pre-incubated with HA in a concentration of about 0.5% w/v.

Dosage

[0065] In some embodiments, the rAAV particle is administered via intravitreal injection. In some embodiments, the rAAV particle is administered via intravitreal administration in a titer of at least about lxlO 3 vg/ml, lxlO 4 vg/ml, lxlO 5 vg/ml, lxlO 6 vg/ml, lxlO 7 vg/ml, lxlO 8 vg/ml, lxlO 9 vg/ml, lxlO 10 vg/ml, 5 xlO 10 vg/ml, lxlO 11 vg/ml, 5 xlO 11 vg/ml, lxlO 12 vg/ml, 2xl0 12 vg/ml, 3xl0 12 vg/ml, 4xl0 12 vg/ml, about 5xl0 12 vg/ml, about lxlO 13 vg/ml, 5xl0 13 vg/ml, lxlO 14 vg/ml, 5xl0 15 vg/ml or lxlO 14 vg/ml In some embodiments, the rAAV particle is administered in a titer of less than 5 xlO 11 vg/ml. In some embodiments, the lower end of these titers represents substantially lower doses than those doses routinely used in subretinal AAV delivery.

[0066] In some embodiments, mixture of rAAV and HA is administered in an intravitreal injection. In some embodiments, the intravitreal injection is provided in a volume of about 1000 pL, about 900 pL, about 800 pL, about 700 pL, about 600 pL, about 500 pL, about 400 pL, about 300 pL, about 200 pL, about 175 pL, about 160 pL, about 145 pL, about 130 pL, about 115 pL, about 100 pL, about 90 pL, about 80 pL, about 70 pL, about 60 pL, about 55 pL, about 50 pL, about 45 pL, about 35 pL, about 20 pL, about 10 pL, about 5 pL, about 4 pL, about 3 pL, about 2 mL, about 1 pL or about 0.5 pL. In some embodiments, the intravitreal injection is provided in a volume of about 50 m L.

[0067] In certain aspects of the described methods, higher concentrations of AAV vector is delivered in a given volume of administration by intravitreal injection.

Hyaluronic Acid

[0068] In some embodiments, the HA suitable for use in the described methods is of any molecular weight known or used in the art. In some embodiments, the HA for administration and/or pre-treatment or use according the described methods has a molecular weight of at least about 4kDa, 5kDa, 20 kDa, 25 kDa, 30 kDa, 40 kDa, 50 kDa, 75 kDa, 100 kDa, 150 kDa, 200 kDa, 350 kDa, 500 kDa, 750 kDa, 1000 kDa, 1500 kDa, 2000 kDa, 2500 kDa, 3000 kDa, 3200 kDa, 3500 kDa, 4000 kDa or 5000 kDa. In some embodiments, the HA for administration and/or pre-treatment or use according the described methods has a molecular weight of less than about 4kDa, 5kDa, 20 kDa, 25 kDa, 30 kDa, 40 kDa, 50 kDa, 75 kDa, 100 kDa, 150 kDa, 200 kDa, 350 kDa, 500 kDa, 750 kDa, 1000 kDa, 1500 kDa, 2000 kDa, 2500 kDa, 3000 kDa, 3200 kDa, 3500 kDa, 4000 kDa or 5000 kDa. In some embodiments, the HA has a molecular weight in the range of about 20 kDa to about 200 kDa. In some

embodiments, the HA has a molecular weight of about 20 kDa or 25 kDa. In some embodiments, the HA is uniform in molecular weight. In some embodiments, the HA has varying molecular weight. In some embodiments, administration is sequential administration or co-administration.

Administration

[0069] In some embodiments, a method for providing a mammal in need thereof with a therapeutically-effective amount of a selected therapeutic agent is described herein. In some embodiments, the therapeutic agent is encoded in a heterologous nucleic acid, or transgene, that is inserted into a recombinant AAV nucleic acid vector. In some embodiments, the nucleic acid vector comprises one or more heterologous nucleic acids comprising a sequence encoding a protein or polypeptide of interest operably linked to a promoter, wherein the one or more transgenes are flanked on each side with an ITR sequence. In some embodiments, the nucleic acid vector further comprises a region encoding a Rep protein as described herein, either contained within the region flanked by ITRs or outside the region or nucleic acid) operably linked to a promoter (e.g. an hGRKl promoter), wherein the one or more nucleic acid regions. The ITR sequences can be derived from any AAV serotype (e.g., 1, 2,

3, 4, 5, 6, 7, 8, 9, or 10) or can be derived from more than one serotype. In some

embodiments, the ITR sequences are derived from AAV2 or AAV6. In some embodiments, the ITR sequences of the first serotype are derived from AAV3, AAV2 or AAV6. In other embodiments, the ITR sequences of the first serotype are derived from AAV 1, AAV5,

AAV8, AAV9 or AAV10. In some embodiments, the ITR sequences are the same serotype as the capsid (e.g., AAV3 ITR sequences and AAV3 capsid, etc.).

[0070] ITR sequences and plasmids containing ITR sequences are known in the art and commercially available (see, e.g., products and services available from Vector Biolabs, Philadelphia, PA; Cellbiolabs, San Diego, CA; Agilent Technologies, Santa Clara, Ca; and Addgene, Cambridge, MA; and Gene delivery to skeletal muscle results in sustained expression and systemic delivery of a therapeutic protein. In some embodiments, the nucleic acid vector comprises a pTR-UF-11 plasmid backbone, which is a plasmid that contains AAV2 ITRs. This plasmid is commercially available from the American Type Culture Collection (ATCC MBA-331).

[0071] In some embodiments, the method includes at least the step of administering to one or both eyes of the mammal, an amount of one or more of the rAAV particles described herein; for a time effective to provide the mammal with a therapeutically-effective amount of the selected therapeutic agent.

[0072] In some embodiments, the described admixing or administration of rAAV and HA is a co-administration. In some embodiments, administration of rAAV and HA is a sequential administration. In some embodiments, administration of rAAV and HA is following a pre incubation period of admixing.

[0073] In some embodiments, the method includes the step of intravitreally administering (a single time or multiple times) to either one or both eyes of the mammal, an amount of one or more rAAV particles described herein; and for a time effective to provide the mammal with a therapeutically-effective amount of the selected diagnostic or therapeutic agent.

[0074] In some embodiments, the therapeutic agent is a therapeutic protein. In some embodiments, the therapeutic protein is a neurotrophic factor or an optogenetic actuator. In some embodiments, the neurotrophic factor is BDNF, NGF, neurotrophin-3, ciliary neurotrophic factor (CNTF), ephrins and glial cell line-derived neurotrophic factors (GDNF), or a combination thereof.

[0075] In some embodiments, the optogenetic actuators of the described methods is a bacteriorhodopsin, a halorhodopsin, a channelrhodopsin, a microbial sensory rhodopsin, a mammalian rhodopsin, a cone opsin, a melanopsin; and a combination thereof. In some embodiments, the optogenetic actuator is channelrhodopsin-2 (ChR-2). In some

embodiments, the disclosure provides a method for treating or ameliorating one or more symptoms of a disease, a disorder, a dysfunction, an injury, an abnormal condition, or trauma in a mammal. In some embodiments, the method includes at least the step of administering to one or both eyes of the mammal in need thereof, one or more of the described rAAV particles herein, in an amount and for a time sufficient to treat or ameliorate the one or more symptoms of the disease, the disorder, the dysfunction, the injury, the abnormal condition, or the trauma in the mammal.

[0076] In some embodiments, the disclosure provides a method for expressing a heterologous nucleic acid segment in one or more photoreceptor cells or one or more RPE cells of a mammal (e.g., a human). In some embodiments, the method includes administering (e.g. directly administering intravitreally) to one or both eyes of the mammal one or more of the rAAV particles described herein, wherein the polynucleotide further comprises at least a first polynucleotide that comprises a PR- or an RPE-cell- specific promoter operably linked to at least a first heterologous nucleic acid segment that encodes a therapeutic agent or a biologically functional fragment thereof, for a time effective to produce the therapeutic agent in the one or more PR cells or RPE cells of the mammal. In some embodiments, the therapeutic agent or a biologically functional is stably expressed in a photoreceptor cell, retinal pigment epithelium cell, retinal ganglion cell, bipolar cell, Miiller glial cell or astrocyte cell, or combinations thereof.

[0077] In some embodiments, the mammal is a human. In some embodiments, the human is a neonate, a newborn, an infant, a juvenile, an adult, or a senior. In some embodiments, the subject is at least about 1 day old to more than about 100 years old. In some embodiments, the human has, is suspected of having, is at risk for developing, or has been diagnosed with one or more retinal disorders, diseases, or dystrophies. In some embodiments, the retinal disorders, diseases, and dystrophies are genetically linked, or inheritable.

[0078] In some embodiments, the production of the therapeutic agent or a biologically active fragment thereof in the cells targeted for administration of the therapeutic construct a) preserves one or more photoreceptor cells or one or more RPE cells, b) restores one or more rod- and/or cone-mediated functions, c) restores visual behavior in one or both eyes, or d) any combination thereof. In some embodiments, production of the therapeutic agent in the described methods preserves one or more PR cells, RPE cells, retinal ganglion cells, bipolar cells Miiller glial cells or astrocyte cells.

[0079] In some embodiments, production of the therapeutic agent or a biologically active fragment thereof persists in the one or more photoreceptor cells or the one or more RPE cells substantially for a period of at least about one week, at least about two weeks, at least about three weeks, at least about one month, at least about two months, at least about three months, at least about six months, at least about nine months, or at least a year or more, following an initial administration of the rAAV gene therapy construct into the one or both eyes of the mammal.

[0080] In some embodiments, the described methods comprise providing a mammal in need thereof with channelrhodopsin-2, cone-opsin or biologically active fragments thereof to the ON bipolar cells of the subject. In some embodiments, the described methods comprise providing a mammal in need thereof with channelrhodopsin-2, cone-opsin, or biologically active fragments thereof to the RGCs and the Muller glial cells on the subject.

[0081] In some embodiments, a rAAV vector construct disclosed herein can be administered via an intravitreal injection, subretinal injection, orally, parenterally, intraocularly, intravenously, intranasally, intra-articularly, intramuscularly, subcutaneously, or a combination thereof. In some embodiments, a rAAV vector construct disclosed herein is administered a single time to a subject. In some embodiments, the rAAV vector construct is administered to the subject in one or more administration periods, for example at least once a day, twice a day, three times per day, once a week, twice a week, once a month, twice a month or at least one a year. In some embodiments, the AAV vector-based therapeutics may be provided successively in one or more daily, weekly, monthly, or less-frequent periods, as may be necessary to achieve treatment, or amelioration of one or more symptoms of the disease or disorder being treated. In some embodiments, a pharmaceutical composition disclosed herein can be administered a single time or multiple times, for example daily, weekly, biweekly, or monthly, hourly, or is administered upon recurrence, relapse or progression of the disease, disorder or condition being treated.

Vectors

[0082] In some embodiments, the vector described herein is a self-complementary rAAV (scAAV) vector. In some embodiments, the vector is provided to the one or both eyes by one or more administrations of an infectious adeno-associated viral particle, an rAAV virion, or a plurality of infectious rAAV particles in an amount and for a time sufficient to treat or ameliorate one or more symptoms of the disease or condition being treated.

[0083] In some embodiments, the disclosure provides improved rAAV particles that have been derived from a number of different serotypes, including but not limited to AAV1, AAV2, AAV3, AAV4, AAV5, AAV6, AAV7, AAV8, AAV9, AAV10 and combinations thereof. In some embodiments, the capsid protein sequences are set forth in SEQ ID NO:l, SEQ ID NO:2, SEQ ID NO:3, SEQ ID NO:4, SEQ ID NO:5, SEQ ID NO:6, SEQ ID NO:7,

SEQ ID NO:8, SEQ ID NO:9, or SEQ ID NO: 10. In some embodiments, the capsid protein sequences comprises SEQ ID NO:l, SEQ ID NO:2, SEQ ID NO:3, SEQ ID NO:4,

SEQ ID NO:5, SEQ ID NO:6, SEQ ID NO:7, SEQ ID NO:8, SEQ ID NO:9, or

SEQ ID NO: 10. In some embodiments, the capsid protein sequences comprises at least about 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% sequence identity with SEQ ID NO:l, SEQ ID NO:2, SEQ ID NO:3, SEQ ID NO:4, SEQ ID NO:5, SEQ ID NO:6,

SEQ ID NO:7, SEQ ID NO:8, SEQ ID NO:9, or SEQ ID NO: 10.

[0084] In some embodiments, the intravitreal administration methods described herein are suitable for delivery of, and enhance the transduction capacity of, any capsid having cationic patches of amino acid residues on its surface. In some embodiments, the capsid comprises AAV2, AAV6 and capsids derived from AAV2 and AAV6. In some embodiments, the capsid comprises AAV7m8, AAV-DJ, AAV2/2-MAX, AAVSHhlO, AAVSHhlOY, AAV3,

AAV3b, AAVLK03, AAV7BP2, AAV1(E531K), AAV6(D532N), AAV6-3pmut, AAV2G9 or elements thereof.

[0085] In some embodiments, AAV-DJ comprises the insertion of 7 amino acids into the HSPG binding domain of the AAV2 capsid and has high expression efficiency in Muller cells following intravitreal injection. In some embodiments, the AAV7m8 capsid is closely related to AAV-DJ. In some embodiments, the AAV2/2-MAX capsid comprises five point mutations, Y272F, Y444F, Y500F, Y730F, T491V. In some embodiments, the AAVSHhlO and AAV6(D532N) capsids are derivatives of AAV6. In some embodiments, the AAV6- 3pmut is (also known as AAV6(TM6) and AAV6(Y705+Y731F+T492V)).

[0086] In some embodiments, the capsid comprises capsids comprising non-native amino acid substitutions at amino acid residues of a wild-type AAV2 capsid as set forth in SEQ ID NO: 2. In some embodiments, the non-native amino acid substitutions comprise one or more of Y272F, Y444F, T491V, Y500F, Y700F, Y704F Y730F or a combination thereof. In some embodiments, the capsids comprises non-native amino acid substitutions at amino acid residues of a wild-type AAV6 capsid as set forth in SEQ ID NO: 6. In some embodiments, the non-native amino acid substitutions comprise one or more of Y445F, Y705F, Y731F, T492V, S663V or a combination thereof.

[0087] In some embodiments, the capsid comprises AAV2G9, a variant of AAV2.

[0088] In some embodiments, the capsid comprises a non-native amino acid substitution at amino acid residue 533 of a wild-type AAV8 capsid as set forth in SEQ ID NO: 8. In some embodiments, the non-native amino acid substitution is E533K, Y733F, or a combination thereof. In some embodiments, the capsid comprises AAV7BP2, a variant of AAV8.

[0089] In some embodiments, the capsid comprises non-native amino acid substitutions of a wild-type AAV2 capsid as set forth in SEQ ID NO: 2. In some embodiments, the capsid comprises one or more of:

(a) Y444F;

(b) Y444F+Y500F+Y730F;

(c) Y272F+Y444F+Y500F+Y730F;

(d) Y 444F+ Y 500F+ Y730F+T491 V ; or

(e) Y272F+Y444F+Y500F+Y730F+T491V, or at equivalent amino acid positions corresponding thereto in any one of the wild-type AAV1, AAV3, AAV4, AAV5, AAV7, AAV9, or AAV10 capsid proteins, as set forth, respectively, in SEQ ID NO: 1, SEQ ID NO: 3, SEQ ID NO: 4, SEQ ID NO: 5, SEQ ID NO: 7, SEQ ID NO: 9, or SEQ ID NO: 10.

[0090] In some embodiments, the capsid comprises non-native amino acid substitutions of a wild-type AAV6 capsid as set forth in SEQ ID NO: 6. In some embodiments, the capsid comprises one or more of:

(a) Y445F;

(b) Y705F+Y731F;

(c) T492V;

(d) Y705F+Y731F+T492V;

(e) S663V; or

(f) S663V+T492V.

[0091] In various embodiments, the rAAV particles comprise one of the following capsids, i.e., capsid variants of AAV2: DGE-DF (also known as‘V1V4 VR-V’), P2-V2, P2-V3, and ME-B(Y-F-i-T-V). The DGE-DF capsid variant contains aspartic acid, glycine, glutamic acid, aspartic acid, and phenylalanine at amino acid positions 492, 493, 494, 499, and 500 of wild- type AAV2 VP1. The P2-V2 capsid variant contains alanine, threonine, proline, aspartic acid, phenylalanine, and aspartic acid at positions 263, 490, 492, 499, 500, and 530 of AAV2 VP1. The P2-V3 capsid variant contains asparagine, alanine, phenylalanine, alanine, asparagine, valine, threonine, arginine, aspartic acid, and aspartic acid at positions 263, 264, 444, 451, 454, 455, 459, 527, 530, and 531 of AAV2 VP1. The ME-B(Y-F+T-V) capsid variant contains aspartic acid, glycine, glutamic acid, aspartic acid, and phenylalanine at positions 492, 493, 494, 499, and 500 of AAV2 VP1, respectively, SAAGADXAXDS (SEQ ID NO: 5) at positions 546-556 of AAV2 VP1, and the following substitutions: Y272F, Y444F, and T491V.

[0092] In other embodiments, the rAAV particles comprise a capsid selected from

AAV6(3pMut), AAV2(quadYF+T-V), or AAV2(trpYF). In some embodiments, the rAAV particles comprise any of the capsid variants described in International Patent Publication No. WO 2018/156654.

[0093] In some embodiments, described herein are methods of administering HA with rAAV particles comprising a DGE-DF capsid, P2-V2 capsid, P2-V3 capsid, or ME-B(Y-F-i-T-V) capsid for the enhanced transduction of said rAAV particles in retinal cells. In some embodiments, the described methods comprise the administration of HA with rAAV particles comprising a capsid selected from AAV2(Y444F), AAV2(Y444F+Y500F+Y730F),

AAV2(Y272F+Y444F+Y500F+Y730F), AAV2(Y444F+Y500F+Y730F+T491V) and AAV2(Y272F+Y444F+Y500F+Y730F+T491V), AAV6(Y445F), AAV6(Y705F+Y73 IF), AAV6(Y705F+Y731F+T492V), AAV6(S663V), AAV6(T492V) or AAV6(S663V+T492V).

[0094] In some embodiments, the rAAV polynucleotide or nucleic acid vectors of the present disclosure may be comprised within a virion having a serotype that is selected from the group consisting of AAV serotype 1, AAV serotype 2, AAV serotype 3, AAV serotype 4, AAV serotype 5, AAV serotype 6, AAV serotype 7, AAV serotype 8, AAV serotype 9, or AAV serotype 10, or any other serotype as known to one of ordinary skill in the viral arts.

Cargo

[0095] In some embodiments, the disclosure further provides populations and pluralities of rAAV polynucleotide or nucleic acid vectors, virions, infectious viral particles, or host cells that comprise a multi-mutated capsid protein and one or more nucleic acid segments that include an RPE- or a PR-cell-specific promoter operably linked to a selected polynucleotide encoding at least a first diagnostic and/or a first therapeutic molecule.

[0096] In some embodiments, the disclosure provides composition and formulations that include one or more of the proteins or biological active fragments thereof, nucleic acid segments or biological active fragments thereof, viral polynucleotide or biological active fragments thereof, nucleic acid vectors, host cells, or viral particles of the present disclosure together with one or more pharmaceutically-acceptable buffers, diluents, or excipients. In some embodiments, the compositions are included in one or more diagnostic or therapeutic kits, for diagnosing, preventing, treating or ameliorating one or more symptoms of a mammalian disease, injury, disorder, trauma or dysfunction. In some embodiments, the diagnostic or therapeutic kit comprises a kit for delivery of a therapeutic agent to

photoreceptors and/or RPE cells of the mammalian eye.

[0097] In some embodiments, described herein is a method for providing a mammal in need thereof with a diagnostically- or therapeutically-effective amount of a selected therapeutic agent, the method comprising providing to a cell, tissue or organ of a mammal in need thereof, an amount of one or more of the described rAAV multi-capsid mutant particles or nucleic acid vectors; and for a time effective to provide the mammal with a diagnostically- or a therapeutically-effective amount of the selected therapeutic agent.

[0098] In some embodiments, the disclosure provides a method for diagnosing, preventing, treating, or ameliorating at least one or more symptoms of a disease, a disorder, a

dysfunction, an injury, an abnormal condition, or trauma in a mammal. In some

embodiments, the method includes at least the step of administering to a mammal in need thereof one or more of the described rAAV particles or nucleic acid vectors, in an amount and for a time sufficient to diagnose, prevent, treat or ameliorate the one or more symptoms of the disease, disorder, dysfunction, injury, abnormal condition, or trauma in the mammal.

[0099] In some embodiments, the disclosure provides a method of transducing a population of mammalian cells, and particular one or more ocular cells in the human eye. In some embodiments, the method includes at least the step of introducing into one or more cells of the population, a composition that comprises an effective amount of one or more of the rAAV particles or nucleic acid vectors described herein. In some embodiments, delivery of the described gene therapy constructs to one or more cells subretinally, permitted the high- efficiency transduction of photoreceptors and RPE cells.

[00100] In some embodiments, the disclosure provides isolated nucleic acid segments that encode one or more of the AAV mutant capsid proteins as described herein, and provides recombinant nucleic acid vectors comprising said segments.

[00101] In some embodiments, the present disclosure provides compositions, as well as therapeutic and/or diagnostic kits that include one or more of the described AAV particle or nucleic acid vector compositions, formulated with one or more additional ingredients, or prepared with one or more instructions for their use.

[00102] In some embodiments, the disclosure provides compositions comprising recombinant adeno-associated viral (AAV) vectors, virions, viral particles, and

pharmaceutical formulations thereof, useful in methods for delivering genetic material encoding one or more beneficial or therapeutic product(s) to mammalian cells and tissues. In some embodiments, the compositions and methods of the disclosure provide a significant advancement in the art through their use in the treatment, prevention, and/or amelioration of symptoms of one or more mammalian diseases.

[00103] In some embodiments, the rAAV particles of the described methods additionally includes a vector comprising a heterologous nucleic acid sequence that encodes at least a first diagnostic or biologically active fragment thereof, or therapeutic agent or a biologically active fragment thereof operably linked to an RPE- or a PR-cell-specific promoter capable of expressing the segment in one or more cells that have been transformed with the vector. I n some embodiments, the PR-cell-specific promoter is human rhodopsin kinase (hGRKl), IRBP, rod opsin, NRL, GNAT2e-IRBP, L/M opsin, cone arrestin promoter, a biologically active fragment of any of these or combination thereof. In some embodiments, a RPE-cell- specific promoter comprises VMD2 (BEST1) or RPE65 promoter.

[00104] In some embodiments, the surface-exposed amino acid-modified rAAV particles or nucleic acid vectors of the present disclosure comprises one or more enhancer sequences that are each operably linked to the nucleic acid segment that encodes the diagnostic or therapeutic molecule of interest or biologically active fragments thereof. The surface-exposed amino acid-modified rAAV particles or nucleic acid vectors of the present disclosure comprises one or more enhancer sequences. In some embodiments, the enhancer sequence is CMV enhancer, a synthetic enhancer, a photoreceptor- specific- specific enhancer, a retinal pigment epithelial cell-specific enhancer, a vascular-specific enhancer, an ocular- specific enhancer, a neural cell-specific enhancer, a retinal cell-specific enhancer, biologically active fragments of these, and any combination thereof.

[00105] In some embodiments, the promoter is a tissue- specific promoter. In some embodiments, the tissue-specific promoter is a photoreceptor specific- and/or a RPE cell- specific promoter.

[00106] In some embodiments, the nucleic acid vector comprises a post-transcriptional regulatory sequences or a polyadenylation signals. In some embodiments, the nucleic acid vector comprises a woodchuck hepatitis virus post-transcription regulatory element (WPRE), a polyadenylation signal sequence, an intron/exon junctions/splicing signals, or any combination thereof.

[00107] In some embodiments, a cargo described herein is a nucleic acid sequence, a gene, an encoded protein, or an encoded active fragment of a protein disclosed herein.

[00108] In some embodiments, the improved rAAV particle comprise a sequence that encodes a diagnostic or therapeutic protein, polypeptide or a biologically active fragment of a molecular marker, a photosensitive opsin, an adrenergic agonist, an anti-apoptosis factor, an apoptosis inhibitor, a cytokine receptor, a cytokine, a cytotoxin, an erythropoietic agent, a glutamic acid decarboxylase, a glycoprotein, a growth factor, a growth factor receptor, a hormone, a hormone receptor, an interferon, an interleukin, an interleukin receptor, a kinase, a kinase inhibitor, a nerve growth factor, a netrin, a neuroactive peptide, a neuroactive peptide receptor, a neurogenic factor, a neurogenic factor receptor, a neuropilin, a neurotrophic factor, a neurotrophin, a neurotrophin receptor, an N-methyl-D-aspartate antagonist, a plexin, a protease, a protease inhibitor, a protein decarboxylase, a protein kinase, a protein kinase inhibitor, a proteolytic protein, a proteolytic protein inhibitor, a semaphorin, a semaphorin receptor, a serotonin transport protein, a serotonin uptake inhibitor, a serotonin receptor, a serpin, a serpin receptor, a tumor suppressor, or any combination thereof. In some embodiments, a photosensitive opsin comprises a rhodopsin, a melanopsin, a cone opsin, a channel rhodopsin, or a bacterial, archea-associated opsin, biologically active fragments of any of these or combinations thereof.

[00109] In some embodiments, the rAAV particles of the present disclosure comprises a nucleic acid segment that encodes the polypeptide RPE65, Bestrophin (BEST1), REP1, MERTK, SOD2, MY06A, MFRP, LRAT, KCNJ13, ornithine aminotransferase (OAT), a biologically active fragment of any of these or any combination thereof.

[00110] In some embodiments, the rAAV particle comprises a nucleic acid segment that encodes the polypeptide CNTF, GDNF, BDNF, IL6, LIF, XIAP, STAT3, a biologically active fragment of any of these or any combination thereof.

[00111] In certain embodiments, the rAAV particle comprises a nucleic acid segment that encodes nyctalopin (nyx), metabotropic glutamate receptor 6-mGluR6 ( Grm6 ), transient receptor potential melastatin 1 ( TRPM1 ), G protein coupled receptor 179 ( GPR179 ), and G proteins , G ?5, G[33, GaO , G/13, RGS7, RGS11, R8AP, and any combination or peptide fragment thereof.

[00112] In some embodiments, described herein is a rAAV nucleic acid vector that comprises at least a first nucleic acid segment that encodes one or more diagnostic, therapeutic agents or biologically active fragments thereof that alter, inhibit, reduce, prevent, eliminate, or impair the activity of one or more endogenous biological processes in a mammalian cell suitably transformed with the vector of interest. In some embodiments, the diagnostic, therapeutic agent or biologically active fragment thereof comprises a molecule that selectively inhibits or reduces the effects of one or more metabolic processes, dysfunctions, disorders, or diseases. In some embodiments, the defect is caused by injury or trauma to the mammal for which treatment is desired. In some embodiments, the defect is caused by the over-expression of an endogenous biological compound. In some

embodiments; the defect is caused by the under-expression or lack of one or more endogenous biological compounds.

[00113] In some embodiments, the rAAV nucleic acid vectors and expression systems of the present disclosure comprises a second nucleic acid segment that comprises, consists essentially of, or consists of, a enhancer, a regulatory element, one or more transcriptional elements, or any combination thereof, that alter, improve, regulate, and/or affect the transcription of the nucleotide sequence of interest expressed by the rAAV particles.

[00114] In some embodiments, the rAAV nucleic acid vectors of the present disclosure comprises a second nucleic acid segment that comprises, consists essentially of, or consists of, a CMV enhancer, a synthetic enhancer, a cell-specific enhancer, a tissue-specific enhancer, or any combination thereof. In some embodiments, the second nucleic acid segment further comprises, consists essentially of, or consists of, one or more intron sequences, one or more post-transcriptional regulatory elements, or one or more enhancers from rhodopsin, melanopsin, cone opsins, channel rhodopsins, bacterial or archea-associated opsins, an adrenergic agonist, an anti-apoptosis factor, an apoptosis inhibitor, a cytokine receptor, a cytokine, a cytotoxin, an erythropoietic agent, a glutamic acid decarboxylase, a glycoprotein, a growth factor, a growth factor receptor, a hormone, a hormone receptor, an interferon, an interleukin, an interleukin receptor, a kinase, a kinase inhibitor, a nerve growth factor, a netrin, a neuroactive peptide, a neuroactive peptide receptor, a neurogenic factor, a neurogenic factor receptor, a neuropilin, a neurotrophic factor, a neurotrophin, a neurotrophin receptor, an N-methyl-D-aspartate antagonist, a plexin, a protease, a protease inhibitor, a protein decarboxylase, a protein kinase, a protein kinase inhibitor, a proteolytic protein, a proteolytic protein inhibitor, a semaphorin, a semaphorin receptor, a serotonin transport protein, a serotonin uptake inhibitor, a serotonin receptor, a serpin, a serpin receptor, or a tumor suppressor. In some embodiments, , the second nucleic acid segment further comprises, consists essentially of, or consists of, one or more intron sequences, one or more post-transcriptional regulatory elements, or one or more enhancers from RPE65, Bestrophin (BEST1), REP1, MERTK, SOD2, MY06A, MFRP, LRAT, KCNJ13, or ornithine aminotransferase (OAT). In some embodiments, the second nucleic acid segment comprises, consists essentially of, or consists of, one or more intron sequences, one or more post- transcriptional regulatory elements, or one or more enhancers from RPE65, Bestrophin (BEST1), REP1, MERTK, SOD2, MY06A, MFRP, LRAT, KCNJ13, ornithine

aminotransferase (OAT), CNTF, GDNF, BDNF, IL6, LIF, XIAP, or STAT3.

[00115] In some embodiments, the rAAV particles comprise a polynucleotide that comprises, consists essentially of, or consists of, one or more polylinkers, restriction sites, and/or multiple cloning region(s) to facilitate insertion (cloning) of one or more selected genetic elements, genes of interest, and/or one or more therapeutic or diagnostic molecules into the rAAV particle at a selected site within the vector.

[00116] In some embodiments, the exogenous polynucleotide(s) is delivered into suitable host cells by the rAAV particles comprising nucleic acid vectors described herein are of mammalian origin, with polynucleotides encoding one or more polypeptides or peptides of, e.g., human, non-human primate, porcine, bovine, ovine, feline, canine, equine, epine, caprine, or lupine origin.

[00117] In some embodiments, the exogenous polynucleotide(s) that is delivered into host cells by the described particles or viral vectors in some embodiments encodes one or more proteins, one or more polypeptides, one or more peptides, one or more enzymes, or one or more antibodies (or antigen-binding fragments thereof), a biologically active fragment of any of these or any combination thereof. In some embodiments, the exogenous polynucleotides express one or more siRNAs, ribozymes, antisense oligonucleotides, PNA molecules, or any combination thereof. In some embodiments, two or more different molecules is produced from a single rAAV expression system, or alternatively, a selected host cell may be transfected with two or more unique rAAV expression systems, each of which may comprise one or more distinct polynucleotides that encode a therapeutic agent.

[00118] In some embodiments, provided herein are rAAV nucleic acid vectors that are comprised within an infectious adeno-associated viral particle or a virion, as well as pluralities of such virions or infectious particles. In some embodiments, the vectors and virions disclosed herein is in a composition with one or more diluents, buffers, physiological solutions or pharmaceutical vehicles, formulated for administration to a mammal in one or more diagnostic, therapeutic, and/or prophylactic regimens. In some embodiments, the vectors, virus particles, virions, and pluralities thereof of the present disclosure are provided in excipient formulations that are acceptable for veterinary administration to selected livestock, exotics, domesticated animals, and companion animals (including pets and such like), as well as to non-human primates, zoological or otherwise captive specimens.

[00119] In embodiments, described herein are recombinant adeno-associated virus virion particles (e.g., , improved transduction efficiency particles), compositions, and host cells that comprise, consist essentially of, or consist of, one or more of the rAAV particles described herein, such as for example pharmaceutical formulations of the vectors intended for intravitreal or subretinal administration to a mammalian eye.

Kits

[00120] In some embodiments, described herein are kits comprising one or more of the described rAAV particles or nucleic acid vectors (as well as one or more virions, viral particles, transformed host cells or pharmaceutical compositions comprising such vectors); and instructions for using such kits in one or more therapeutic, diagnostic, and/or

prophylactic clinical embodiments are also provided by the present disclosure. In some embodiments, the kits further comprise one or more reagents, restriction enzymes, peptides, therapeutics, pharmaceutical compounds, or means for delivery of the composition(s) to host cells, or to an animal (e.g., syringes, injectables, and the like). In some embodiments, the kits disclosed herein is treating, preventing, or ameliorating the symptoms of a disease, deficiency, dysfunction, and/or injury. In some embodiments, the kit includes components for the large-scale production of the viral vectors themselves, such as for commercial sale, or for use by others, including e.g., virologists, medical professionals, and the like.

Use

[00121] In some embodiments, described herein are methods of use of the described rAAV particles or vectors, virions, expression systems, compositions, and host cells described herein in the preparation of medicaments for diagnosing, preventing, treating or ameliorating at least one or more symptoms of a disease, a dysfunction, a disorder, an abnormal condition, a deficiency, injury, or trauma in an animal, and in particular, in the eye. In some

embodiments, the methods comprise direct administration to the vitreous of one or both eyes of a mammal in need thereof, one or more of the described vectors, virions, viral particles, host cells, compositions, or pluralities thereof, in an amount and for a time sufficient to diagnose, prevent, treat, or lessen one or more symptoms of such a disease, dysfunction, disorder, abnormal condition, deficiency, injury, or trauma in one or both eyes of the affected animal.

[00122] In some embodiments, described herein are compositions comprising one or more of the described rAAV particles, expression systems, infectious AAV particles, and host cells. In some embodiments, the compositions further comprise at least a first

pharmaceutically-acceptable excipient for use in the manufacture of medicaments and methods involving therapeutic administration of such rAAV particles or nucleic acid vectors. In some embodiments, pharmaceutical formulations are suitable for intravitreal

administration into one or both eyes of a human or other mammal.

[00123] In some embodiments, described herein are methods of use of the particles, vectors, virions, expression systems, compositions, and host cells described herein in a method for treating or ameliorating the symptoms or in the preparation of medicaments for treating or ameliorating the symptoms of various deficiencies in an eye of a mammal, and in particular one or more deficiencies in human photoreceptors or RPE cells. In some embodiments, the diseases and disorders of the eye (e.g., caused by one or more genetic deficiencies in a PR or PRE cell) for treatment or amelioration of symptoms include Retinitis pigmentosa, Leber Congenital Amaurosis, Age Related Macular Degeneration (AMD) wet AMD, dry AMD, uveitis, Best disease, Stargardts disease, Usher Syndrome, Geographic Atrophy, Diabetic Retinopathy, Retinoschisis, Achromatopsia, Choroideremia, Bardet Biedl Syndrome, glycogen storage diseases (ocular manifestation) or a combination thereof. In some embodiments, the methods comprise intravitreal or subretinal administration to one or both eyes of a subject in need thereof, one or more of the described particles vectors, virions, host cells, or compositions, in an amount and for a time sufficient to treat or ameliorate the symptoms of such a deficiency in the affected mammal. In some embodiments, the methods comprise prophylactic treatment of an animals suspected of having such conditions, or administration of such compositions to those animals at risk for developing such conditions either following diagnosis, or prior to the onset of symptoms. In some embodiments, the rAAV particle is not comprised in a chimeric viral/non-viral nanoparticle.

Storage and Manufacturing Buffers

[00124] In some embodiments, provided herein are buffers for storage and manufacturing of rAAV particles disclosed herein. In some embodiments, a pharmaceutical composition disclosed herein comprises a buffered solution.

[00125] In some embodiments, described herein are buffers for storage of a mixture of rAAV vectors or capsids and HA. In some embodiments, the described buffers comprise HA in a concentration of at least about 0.05%, 0.1%, 0.2%, 0.25%, 0.3%, 0.35%, 0.4%, 0.45%, 0.5%, 0.6%, 0.75%, 1.0%, 2%, 3%, 4%, 5%, 6%, 7%, 8%, 9%, 10%, or 15% weight by volume (w/v) and BSS. In some embodiments, the described buffers comprise HA in a concentration of at least about 0.1%, 0.2%, 0.25%, 0.3%, 0.35%, 0.4%, 0.45%, 0.5%, 0.6%, 0.75%, 1.0%, 2%, 3%, 4%, 5%, 6%, 7%, 8%, 9%, 10%, or 15% weight by volume (w/v) and BSS, and one or more of the following excipients: artificial cerebrospinal fluid (CSF), PBS, Ringer’s lactate solution, TMN200 solution, polysorbate 20, and poloxamer 188 (known commercially as Pluronic F-68 ® ), and/or additional excipients or combinations thereof.

[00126] In some embodiments, the buffer comprises HA in a concentration of about 0.4% w/v, and one or more of BSS, artificial CSF, or PBS. In some embodiments, the buffer comprises HA in a concentration of about 0.4% w/v, and one or more of BSS, artificial CSF, PBS, Ringer’s lactate solution; and optionally TMN200 solution, polysorbate 20 (Tween 20), and poloxamer 188. In some embodiments, the described buffers comprise Tween 20 in a concentration of about at least 0.005%, 0.009%, 0.01% 0.014%, 0.02%, 0.1%, 0.2% 0.5% or 1% and poloxamer 100 in a concentration of about 0.005%, 0.009%, 0.01%, 0.02%, 0.1%, 0.2% 0.5% or 1%.

[00127] In some embodiments, the described buffers consist essentially of HA in a concentration of about HA in a concentration of 0.1%, 0.2%, 0.25%, 0.3%, 0.35%, 0.4%, 0.45%, 0.5%, 0.6%, 0.75%, or 1.0% w/v, BSS, artificial CSF, PBS and Ringer’s lactate solution.

[00128] In some embodiments, described herein are elution buffers for manufacturing rAAV particles that comprise HA. In some embodiments, the buffers may be used to elute AAV capsids from an affinity column immediately prior to packaging and formulation to a final product. In some embodiments, the buffers prevent aggregation of AAV capsids having surface-exposed cationic patches from aggregating during manufacturing. In some embodiments, the buffers comprise a pH lower than 7. In some embodiments, the buffers comprise a pH lower than 5. In some embodiments, the buffers are acidic. In some embodiments, the buffers are basic. In some embodiments, a pharmaceutical composition disclosed herein is acidic. In some embodiments, a pharmaceutical composition disclosed herein s basic. In some embodiments, a pharmaceutical composition disclosed herein is neutral. In some embodiments, a composition described herein can have a pH of 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, or 14.

[00129] In some embodiments, the buffers comprise HA in a concentration of 0.1%, 0.2%, 0.25%, 0.3%, 0.35%, 0.4%, 0.45%, 0.5%, 0.6%, 0.75%, or 1.0% w/v.

[00130] In some embodiments, described herein are methods of manufacturing an rAAV particle comprising purifying AAV capsids wherein capsids are eluted from an affinity column containing a resin selected from AAV-X or AVB affinity resins, or another resin known in the art. rAAV particles

[00131] Aspects of the disclosure relate to recombinant adeno-associated virus (rAAV) particles or preparations of such particles for delivery of one or more nucleic acid vectors comprising a sequence encoding a Rep protein, and/or a protein or polypeptide of interest, into various tissues, organs, and/or cells. In some embodiments, the rAAV particle is delivered to a host cell in the presence of a Rep protein as described herein.

[00132] Recombinant adeno-associated virus (rAAV) vectors have been used successfully for in vivo gene transfer in numerous pre-clinical animal models of human disease, and have been used successfully for long-term expression of a wide variety of therapeutic. AAV vectors have also generated long-term clinical benefit in humans when targeted to immune- privileged sites, e.g., ocular delivery for Leber’s congenital amaurosis. An advantage of this vector is its comparatively low immune profile, eliciting only limited inflammatory responses and, in some cases, even directing immune tolerance to transgene products. Nonetheless, the therapeutic efficiency, when targeted to non-immune privileged organs, has been limited in humans due to antibody and CD8 + T cell responses against the viral capsid, while in animal models, adaptive responses to the transgene product have also been reported.

[00133] Adeno-associated vims (AAV) is used for ocular gene therapy due to its efficiency, persistence and low immunogenicity. Identifying vectors capable of transducing PRs via the vitreous has historically relied on identifying which serotypes have native tropism for this cell type following local delivery. Several serotypes have been used to successfully target transgene to PRs following subretinal injection (including, e.g., AAV2, AAV5 and AAV8) with all three demonstrating efficacy in experiments performed across multiple mammalian species (e.g., mouse, rat, dog, pig and non-human primate).

[00134] In some embodiments, AAV2 and AAV8 vectors comprises point mutations of surface-exposed tyrosine residues (tyrosine to phenylalanine, Y-F) display to increase transgene expression in retinal cell types relative to unmodified vectors following both subretinal and intravitreal injection. In some embodiments, AAV2 comprises a triple mutant (designated“triple Y— >F”). In some embodiments, an AAV2 comprises a quadruple mutant (“quad Y— >F”). In some embodiments, an AAV comprises directed mutagenesis of surface-exposed threonine (T) or serine (S) residues to non-native amino acids at one of more of those amino acids. In some embodiments, an AAV comprises an Y— >F and/or T— >V/T— >A mutations.

[00135] In some embodiments, the rAAV is an rAAV described herein.

[00136] In some embodiments, the rAAV nucleic acid vector comprises a single-stranded (ss) or self-complementary (sc) AAV nucleic acid vectors, such as single-stranded or self complementary recombinant viral genomes. In some embodiments, the rAAV particle is not comprised in a chimeric viral/non-viral nanoparticle.

[00137] The wild-type AAV genome is a single-stranded deoxyribonucleic acid (ssDNA), either positive- or negative-sensed. The genome comprises two inverted terminal repeats (ITRs), one at each end of the DNA strand, two open reading frames (ORFs): rep and cap between the ITRs, and an insert nucleic acid positioned between the ITRs and optionally comprising a transgene. The rep ORF comprises four overlapping genes encoding Rep proteins required for the AAV life cycle. The cap ORF comprises overlapping genes encoding capsid proteins: VP1, VP2 and VP3, which interact together to form the viral capsid. VP1, VP2 and VP3 are translated from one mRNA transcript, which can be spliced in two different manners: either a longer or shorter intron can be excised resulting in the formation of two isoforms of mRNAs: a -2.3 kb- and a -2.6 kb-long mRNA isoform. The capsid forms a supramolecular assembly of approximately 60 individual capsid protein subunits into a non-enveloped, T-l icosahedral lattice capable of protecting the AAV genome. The mature capsid is composed of VP1, VP2, and VP3 (molecular masses of approximately 87, 73, and 62 kDa respectively) in a ratio of about 1: 1: 10.

[00138] Recombinant AAV (rAAV) particles may comprise a nucleic acid vector, which may comprise at a minimum: (a) one or more transgenes comprising a sequence encoding a protein or polypeptide of interest or an RNA of interest (e.g., a siRNA or microRNA), or one or more nucleic acid regions comprising a sequence encoding a Rep protein; and (b) one or more regions comprising inverted terminal repeat (ITR) sequences) flanking the one or more heterologous nucleic acid regions (e.g., transgenes). In some embodiments, the nucleic acid vector is between 4kb and 5kb in size (e.g., 4.2 to 4.7 kb in size). In some embodiments, the nucleic acid vector further comprises a region encoding a Rep protein as described herein. Any nucleic acid vector described herein may be encapsidated by a viral capsid, such as an AAV6 capsid or another serotype (e.g., a serotype that is of the same serotype as the ITR sequences), which may comprises a modified capsid protein as described herein. In some embodiments, the nucleic acid vector is circular. In some embodiments, the nucleic acid vector is single- stranded. In some embodiments, the nucleic acid vector is double- stranded.

In some embodiments, a double- stranded nucleic acid vector may be, for example, a self complimentary vector that contains a region of the nucleic acid vector that is complementary to another region of the nucleic acid vector, initiating the formation of the double

strandedness of the nucleic acid vector.

[00139] Accordingly, in some embodiments, an rAAV particle or rAAV preparation containing such particles comprises a viral capsid and a nucleic acid vector as described herein, which is encapsidated by the viral capsid. In some embodiments, the insert nucleic acid of the nucleic acid vector comprises (1) one or more transgenes comprising a sequence encoding a protein or polypeptide of interest, (2) one or more nucleic acid regions comprising a sequence that facilitates expression of the transgene (e.g., a promoter), and (3) one or more nucleic acid regions comprising a sequence that facilitate integration of the transgene (optionally with the one or more nucleic acid regions comprising a sequence that facilitates expression) into the genome of the subject.

Helper plasmids

[00140] In some embodiments a plasmid containing the nucleic acid vector sequence disclosed herein is combined with one or more helper plasmids, e.g., that contain a rep gene (e.g., encoding Rep78, Rep68, Rep52 and Rep40) and a cap gene (encoding VP1, VP2, and VP3, including a modified VP3 region as described herein). In some embodiments, a helper plasmid is transfected into a producer cell line such that the rAAV particle can be packaged and subsequently purified.

[00141] In some embodiments, the one or more helper plasmids comprises a first helper plasmid comprising a rep gene and a cap gene and/or a second helper plasmid comprising a Ela gene, a Elb gene, a E4 gene, a E2a gene, a VA gene or a combination thereof. In some embodiments, the rep gene is a rep gene derived from AAV2. In some embodiments the cap gene is derived from AAV2. In some embodiments, the cap gene includes modifications to the gene in order to produce a modified capsid protein described herein. In certain embodiments, the rep gene comprises Rep78, Rep68, Rep52 or Rep40. In certain

embodiments, the cap gene comprises VP1, VP2, VP3 or variants thereof. In some embodiments, the helper plasmid comprises pDM, pDG, pDPlrs, pDP2rs, pDP3rs, pDP4rs, pDP5rs, pDP6rs, pDG(R484E/R585E), or pDP8.ape plasmids.

Production

[00142] In some embodiments, described herein is a method of rAAV particle production. In some embodiments, one or more helper plasmids are produced or obtained. In some embodiments, the one or more helper plasmids comprise rep and cap ORFs for the desired AAV serotype and the adenoviral VA, E2A (DBP), and E4 genes. In some embodiments, the rep and cap ORFs for the desired AAV serotype and the adenoviral VA, E2A (DBP), and E4 genes are under the transcriptional control of their native promoters. In some embodiments, the cap ORF comprises one or more modifications to produce a modified capsid protein as described herein. In some embodiments, HEK293 cells (available from ATCC®) are transfected via CaP04-mediated transfection, lipids or polymeric molecules such as

Polyethylenimine (PEI) with the helper plasmid(s) and a plasmid containing a nucleic acid vector described herein. In some embodiments, the HEK293 cells are incubated for at least about 60 hours to allow for rAAV particle production. In some embodiments, a Sf9-based producer stable cell line is infected with a single recombinant baculovirus containing the nucleic acid vector. In some embodiments, HEK293 or BHK cell lines are infected with a HSV containing the nucleic acid vector and optionally one or more helper HSVs. Ins some embodiments, the helper HSVs contain rep and cap ORFs as described herein and the adenoviral VA, E2A (DBP), and E4 genes under the transcriptional control of their native promoters. In some embodiments, the HEK293, BHK, or Sf9 cells are then incubated for at least 60 hours to allow for rAAV particle production. In some embodiments, the rAAV particles are purified. In some embodiments, the rAAV particles are purified by iodixanol step gradient, CsCl gradient, chromatography, or polyethylene glycol (PEG) precipitation.

Kits and Uses for Improved rAAV Delivery

[00143] In some embodiments, described herein is a mixture of rAAV particles and hyaluronic acid comprised within a kit for diagnosing, preventing, treating or ameliorating one or more symptoms of a mammalian disease, injury, disorder, trauma or dysfunction. In some embodiments, kits are useful in diagnosis, prophylaxis, and/or therapy, and in the treatment, prevention, and/or amelioration of one or more defects in the mammalian eye as discussed herein. In some embodiments, a kit can comprise a pharmaceutical composition disclosed herein. In some embodiments, a kit can comprise a pharmaceutical composition in unit dose form. In some embodiments, a method can comprise making a kit disclosed herein. In some embodiments, a kit can comprise one or more container, bottle, or ampoule. In some embodiments, a kit comprises instructions for use.

[00144] In some embodiments, described herein are methods comprising the use of the buffers and compositions described herein in the manufacture of a medicament for treating, preventing or ameliorating the symptoms of a disease, disorder, dysfunction, injury or trauma comprising the treatment, prevention, and/or prophylaxis of a disease, disorder or dysfunction, and/or the amelioration of one or more symptoms of such a disease, disorder or dysfunction.

[00145] In some embodiments, described herein are methods for treating or ameliorating the symptoms of such a disease, injury, disorder, or dysfunction in one or both eyes of a mammal, and of a human. In some embodiments, the methods comprise at least the step of administering to a mammal in need thereof, one or more of the rAAV particles as described herein, in an amount and for a time sufficient to treat or ameliorate the symptoms of such a disease, injury, disorder, or dysfunction in one or both eyes of the mammal. In some embodiments,

Pharmaceutical Compositions Comprising rAAV particles

[00146] In some embodiments, the improved rAAV delivery methods described herein permit the delivery of smaller titers of viral particles in order to achieve the same

transduction efficiency as that obtained using higher levels of conventional, rAAV methods. In some embodiments, the method comprises the administration of therapeutically-effective amounts of the described compositions comprising a single administration. In some embodiments, the method comprises a single injection of sufficient numbers of infectious particles to provide therapeutic benefit to the patient undergoing such treatment. In some embodiments, the method comprises multiple, or successive administrations of the AAV vector compositions, either over a relatively short, or over a relatively prolonged period. In some embodiments, the number of infectious particles administered to a subject is at least about 10 1 , 10 2 , 10 3 , 10 4 , 10 5 , 10 6 , 10 7 , 10 8 , 10 9 , 10 10 , 10 11 , 10 12 , 10 13 , or even higher, infectious particles/mL. In some embodiments, the infectious particles are given either as a single dose (or divided into two or more administrations, etc.,) as may be required to achieve therapy of the particular disease or disorder being treated. In some embodiments, the method comprises administering two or more different rAAV particle- or vector-based compositions, either alone, or in combination with one or more other diagnostic agents, drugs, bioactives, to achieve the desired effects of a particular regimen or therapy. In some embodiments, lower titers of infectious particles will be required when practicing the described methods of pre treating and co-administering AAV capsids with HA when compared to the titers of infectious particles required when AAV capsids are not pre-treated or co-administered with HA. In some embodiments, the capsid sequence comprises at least about 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% sequence identity with SEQ ID NO: l,

SEQ ID NO:2, SEQ ID NO:3, SEQ ID NO:4, SEQ ID NO:5, SEQ ID NO:6, SEQ ID NO:7, SEQ ID NO:8, SEQ ID NO:9, or SEQ ID NO: 10. In some embodiments, the capsid sequence comprises SEQ ID NO: l, SEQ ID NO:2, SEQ ID NO:3, SEQ ID NO:4, SEQ ID NO:5, SEQ ID NO:6, SEQ ID NO:7, SEQ ID NO:8, SEQ ID NO:9, or SEQ ID NO: 10. In some embodiment, the rAAV particle is not comprised in a chimeric viral/non- viral nanoparticle.

[00147] In some embodiments, the rAAV particle comprises a therapeutic agent-encoding a nucleic acid segment under the control of one or more promoters. In some embodiments, to bring a sequence“under the control of’ a promoter, one positions the 5' end of the transcription initiation site of the transcriptional reading frame generally between about 1 and about 50 nucleotides“downstream” of ( i.e ., 3' of) the chosen promoter. In some

embodiments, the“upstream” promoter stimulates transcription of the DNA and promotes expression of the encoded polypeptide. In some embodiments, recombinant vector constructs are those that include a capsid-protein modified rAAV vector that contains an RPE cell- or a photoreceptor cell-specific promoter, operably linked to at least one nucleic acid segment encoding one or more diagnostic, and/or therapeutic agents.

[00148] In some embodiments, the method comprises introducing one or more exogenous proteins, polypeptides, peptides, ribozymes, and/or antisense oligonucleotides, to a particular cell transfected with the vector. In some embodiments, the rAAV particles described herein are used to deliver one or more exogenous polynucleotides to a selected host cell, e.g., to one or more selected cells within the mammalian eye.

[00149] In some embodiments, the number of viral particles administered to a subject may be on the order ranging from at least about 10 1 to 10 20 particles/ml or 10 3 to 10 15 particles/ml, or any values therebetween for either range, such as for example, about 10 1 , 10 2 , 10 3 , 10 4 ,

10 5 , 10 6 , 10 7 , 10 8 , 10 9 , 10 10 , 10 11 , 10 12 , 10 13 , 10 14 10 15 , 10 16 , 10 17 , 10 18 , 10 19 or 10 20 particles/ml. In some embodiments, viral particles of higher than 10 13 particles/ml may be administered. In some embodiments, the number of viral particles administered to a subject may be on the order ranging from 10 6 to 10 14 vector genomes(vgs)/ml or 10 3 to 10 15 vgs/ml, or any values therebetween for either range. In some embodiments, the number of viral particles administered to a subject may be on the order of about 10 1 , 10 2 , 10 3 , 10 4 , 10 5 , 10 6 , 10 7 , 10 8 , 10 9 , 10 10 , 10 11 , 10 12 , 10 13 , or 10 14 vgs/ml. In some embodiments, viral particles of higher than 10 13 vgs/ml are be administered. In some embodiments, the viral particles are administered as a single dose. In some embodiments, the viral particles are divided into two or more administrations as may be required to achieve therapy of the particular disease or disorder being treated. In some embodiments, 0.0001 ml to 10 ml, e.g., 0.001 ml, 0.01 ml, 0.1 ml, 1 ml, 2 ml, 5 ml or 10 ml, are delivered to a subject.

[00150] In some embodiments, described herein are formulations of one or more viral- based compositions described herein in pharmaceutically acceptable solutions for

administration to a cell or an animal, either alone or in combination with one or more other modalities of therapy, and in particular, for therapy of human cells, tissues, and diseases affecting the subject.

[00151] In some embodiments, rAAV particles described herein are administered in combination with other agents as well, such as, e.g., proteins or polypeptides or various pharmaceutically-active agents, including one or more systemic or topical administrations of therapeutic polypeptides, biologically active fragments, or variants thereof. In some embodiments, the compositions may be purified from host cells or other biological sources, or alternatively may be chemically synthesized as described herein.

[00152] In some embodiment, the composition comprises a pharmaceutically-acceptable buffer, excipients or carrier solutions. In some embodiments, the compositions are formulated for oral, parenteral, intravitreal, intraocular, intravenous, intranasal, topical, intra- articular, intramuscular administration or a combination thereof..

[00153] In some embodiments, a composition disclosed herein comprises a

pharmaceutically acceptable carrier. In some embodiments, pharmaceutically acceptable carriers include, but are not limited to, buffers such as neutral buffered saline, phosphate buffered saline and the like; carbohydrates such as glucose, mannose, sucrose or dextrans, mannitol; proteins; polypeptides or amino acids such as glycine; antioxidants; chelating agents such as EDTA or glutathione; adjuvants (e.g., aluminum hydroxide); or preservatives.

[00154] In some embodiments, these formulations comprise at least about 0.1% of the therapeutic agent (e.g., rAAV particle) or more. In some embodiments, the percentage of the active ingredient(s) is between at least about 0.01%, 0.02%, 0.05%, 0.1%, 0.5% 1% or 2% and about 70% or 80% or more of the weight or volume of the total formulation.

[00155] In some embodiments, term“excipient” refers to a diluent, adjuvant, carrier, or vehicle with which the rAAV particle is administered. In some embodiments, the

pharmaceutical excipients comprises sterile liquids, such as water or oils, including those of petroleum oil such as mineral oil, vegetable oil such as peanut oil, soybean oil, or sesame oil, animal oil, or oil of synthetic origin. In some embodiments, liquid carriers comprise saline solutions, aqueous dextrose, glycerol solutions or combinations thereof. In some

embodiments, excipients and vehicles comprise HA, BSS, artificial CSF, PBS, Ringer’s lactate solution, TMN200 solution, polysorbate 20, poloxamer 100 or a combination thereof.

[00156] In some embodiments, the administration of therapeutically-effective amounts of the composition comprise a single administration, such as for example, a single injection of sufficient numbers of viral particles to provide therapeutic benefit to the patient undergoing such treatment. In some embodiments, administration of a therapeutically effective about of the composition comprises multiple, or successive administrations of the compositions, either over a relatively short, or a relatively prolonged period of time, as may be determined by the medical practitioner overseeing the administration of such compositions.

[00157] In some embodiments, the composition comprises rAAV particles or nucleic acid vectors either alone, or in combination with one or more additional active ingredients, which may be obtained from natural or recombinant sources or chemically synthesized.

EXAMPLES

Example 1 - In Vitro Experiments

[00158] It was sought to determine whether AAV capsids containing cationic patches of surface-exposed capsid residues (i.e. AAV2 and AAV6) exhibited enhanced transduction of HEK293T (human) and 661W cells (a murine cone-derived photoreceptor cell line) following pre-incubation with HA.

[00159] AAV2 capsids were preincubated with Healon ® in a ratio of 3:1 (AAV:Healon ® ) and then injected into cells at a multiplicity of infection (MOI) of 2000. Controls included uninfected cells and cells infected with AAV vector alone. AAV-mediated mCherry reporter expression data demonstrated that HA increased transduction of 661W cells after

administration in vitro of rAAV2 particles following pre-incubation with HA (at 5 minutes, 15 minutes, and 1 hour prior to infection) in Fig. 2A.

[00160] AAV6 capsid harbors patches of positively-charged residues, similar to AAV2. Both serotypes show improvements in transduction after pre-incubation with HA. As with AAV2, HA pre-treatment and co-administration increased transduction of mCherry reporter (under the control of a ubiquitous CBA promoter) in HEK293T cells in vitro after

administration of self-complementary rAAV6-based vectors injected at multiplicities of infection (MOIs) of 5000 to 10,000, as shown in Fig. 3. For this experiment, transduction efficiencies of capsid variants AAV6(D532N) and AAV6-3pmut, in the presence and absence of HA pre-incubation, were evaluated.

[00161] By contrast, AAV5, which has a uniformly negative charge on the capsid surface, failed to exhibit enhanced transduction (see Figs. 2 and 3).

[00162] Next, it was established that HA-mediated enhancement of transduction was not dependent on binding to the CD44 cell surface receptor. First, it was confirmed via immunocytochemistry that HEK293T and 661W did not exhibition expression of CD44 (see Fig. 4), but that ARPE19 cells, a human RPE cell line that is known to express CD44, exhibited CD44 expression.

[00163] When a similar experiment as that performed with AAV6-based capsids in

HEK293 was performed in ARPE19 cells, the pre-treatment of AAV with HA was shown to reduce transduction. This inhibition was overcome by digesting the HA with hyaluronidase, which reduces the side-chain length of HA such that it is no longer recognized by CD44.

[00164] As noted, pre-incubation of AAV2 with HA inhibits transduction of ARPE19 cells, and ARPE19 cells express CD44. However, pre-incubation of AAV2 with HA that has been digested with hyaluronidase enhances transduction of ARPE19 cells. When hyaluronidase cleaves HA, the resulting shortened concatemer is generally 50-100 monomers long (having a molecular weight of 20-40kDa). The affinity of HA for CD44 substantially decreases with decreasing molecular weight, particularly around the 20kDa molecular weight mark

(monovalent interaction vs. divalent interaction).

[00165] Next, it was asked whether HA-coated AAV2 still recognizes the heparan sulfate proteoglycan (HSPG) receptor footprint required for the cell recognition and internalization events necessary for intravitreal transduction of retina. To assess this, HA was incubated with AAV2, and then binding to heparin, which is the established proxy material for HSPG, was evaluated. As illustrated in the elution profile depicted in Fig. 5, HA treatment did not alter binding to heparin. The heparin affinity of HA-coated AAV2 differed insignificantly from AAV2 alone, indicating that the HA-mediated transduction enhancements have little to no impact on the canonical HSPG binding of AAV capsids.

[00166] Taken together, the results indicate that improvements in transduction mediated by HA are not CD44-dependent.

Example 2 - In Vivo Experiments

[00167] It was then determined whether pre-incubation of several AAV2-based capsid variants with HA facilitated enhanced retinal transduction in intravitreally-injected mice relative to virus alone (see Figs. 6A-6B). 10pL of vims capsid (2xl0 12 vector genome copies (vg)/mL) was mixed with 10pL Healon ® (molecular weight of 5 million kDa) within 30 minutes of injection. The virus-only control was composed of 10pL of virus (2 xlO 12 vg/mL) mixed with 10pL Balanced Salt solution (BSS) + Tween20. The final vector concentration was 1 xlO 12 vg/mL. Intravitreal injections were performed on 4 week old Nrl-GFP mice and fundus images were taken at two weeks, and four weeks post injection. Four capsids— all variants of AAV2 that retain cationic surface-exposed patches— were tested. These capsids are known in the art as‘DGE-DF’,‘P2-V2’,‘P2-V3’, and‘ME-B(Y-F+T-V)’ (see

International Patent Publication No. WO 2018/156654). Gain and integration settings were consistent throughout all images (see Figs. 7A-7B, 8A-8B, 9A-9B, and 10A-10B).

[00168] Five weeks post-injection, electroretinogram (ERG) and FACS analyses were performed. Flow cytometry was used to analyze transduction of retinal cells by measuring mCherry expression, and gating was done using an un-injected eye’s whole retina. Fundus images at 2 weeks and 4 weeks post-injection, along with aggregated flow cytometry quantifications of the signals in these images, are shown for DGE-DF in Figs. 7A-7C, P2-V2 in Figs. 8A-8C, P2-V3 in Figs. 9A-9C, and ME-B(Y-F+T-V) in Figs. 10A-10C. Flow cytometry data for all four capsids overall was aggregated and is shown in Fig. 11.

[00169] Each of the four capsid variants exhibited improved retinal transduction after pre incubation with HA. It was also confirmed that pre-treatment and co-administration of capsid with HA had no significant impact on retinal function after intravitreal delivery, as assessed by ERG data (see Figs. 12A-12E). The amplitudes of A- and B-waves in rod and cone cells did not vary to a significant degree following treatment with HA.

[00170] Taken together, the results indicate that pre-treatment and co-administration with HA enhances transduction efficiencies in vivo across several different capsid types having surface-exposed cationic patches. Pre-treatment with HA represents a powerful novel method for improving AAV transduction, and lowering these AAV titer necessary to achieve a desired transduction, without modifying the residues of the capsid itself.

[00171] It should be understood that the examples and embodiments described herein are for illustrative purposes only and that various modifications or changes in light thereof will be suggested to persons skilled in the art and are to be included within the spirit and purview of this application and the scope of the appended claims. All references, including publications, patent applications and patents, cited herein are hereby incorporated by reference to the same extent as if each reference was individually and specifically indicated to be incorporated by reference and was set forth in its entirety herein. Recitation of ranges of values herein are merely intended to serve as a shorthand method of referring individually to each separate value falling within the range, unless otherwise indicated herein, and each separate value is incorporated into the specification as if it were individually recited herein.

[00172] All of the compositions and methods described and claimed herein can be made and executed without undue experimentation in light of the present disclosure. While the compositions and methods have been described in terms of preferred embodiments, it will be apparent to those of skill in the art that variations may be applied to the compositions and methods and in the steps or in the sequence of steps of the method described herein without departing from the concept, spirit and scope of the invention. More specifically, it will be apparent that certain agents that are chemically and/or physiologically related may be substituted for the agents described herein while the same or similar results would be achieved. All such similar substitutes and modifications apparent to those skilled in the art are deemed to be within the spirit, scope and concept of the invention as defined by the appended claims.