The contents of the electronic sequence listing submitted herewith as file 38013_0027P1Updated.xml; Size: 135,000 bytes; and Date of Creation: Oct. 7, 2022, is herein incorporated by reference in its entirety.
The present invention relates to recombinant adeno-associated viruses (rAAVs) having capsid proteins with one or more amino acid substitutions and/or peptide insertions that confer and/or enhance desired properties, including tissue tropisms. In particular, the invention provides engineered capsid proteins comprising one or more amino acid substitutions that reduce transduction by the recombinant AAV of one or more tissue types, such as lung, liver, pancreas, nervous tissue and/or kidney as compared to the transduction of the tissue by AAV9 or another parental capsid. Such engineered capsids with reduced tissue tropism may be further engineered to display a targeting domain, such as a peptide or antibody comprising a binding domain, to enhance transduction of one or more tissue types as compared to the engineered capsid that does not comprise the targeting domain and/or as compared to AAV9 or AAV with another parental capsid and, in embodiments, preferentially transducer the target tissue and does not transduce other tissue types which are not targeted. rAAVs having the capsid proteins disclosed herein are useful for delivering a transgene encoding a therapeutic protein or nucleic acid for treatment of disease associated with the tissue in which transduction of the engineered rAAV is enhanced.
The use of adeno-associated viruses (AAV) as gene delivery vectors is a promising avenue for the treatment of many unmet patient needs. Dozens of naturally occurring AAV capsids have been reported, and mining the natural diversity of AAV sequences in primate tissues has identified over a hundred variants, distributed in clades. AAVs belong to the parvovirus family and are single-stranded DNA viruses with relatively small genomes and simple genetic components. Without a helper virus, AAV establishes a latent infection. An AAV genome generally has a Rep gene and a Cap gene, flanked by inverted terminal repeats (ITRs), which serve as replication and packaging signals for vector production. The capsid proteins form capsids that carry genome DNA and can determine tissue tropism to deliver DNA into target cells.
Due to low pathogenicity and the promise of long-term, targeted gene expression, recombinant AAVs (rAAVs) have been used as gene transfer vectors, in which therapeutic sequences are packaged into various capsids. Such vectors have been used in preclinical gene therapy studies and over twenty gene therapy products are currently in clinical development. It may be useful to design AAV capsids which have enhanced transduction of specific tissue types such as, but not limited to, CNS, muscle and/or heart tissue while also exhibiting reduced transduction of tissue types such as, liver and/or dorsal root ganglion cells and/or kidney may also be desirable to reduce toxicity.
There remains a need for rAAV vectors with enhanced tropism for specific tissue types, such as muscle and/or heart and/or CNS for use, e.g., in treating disorders associated with the central nervous system or where expression in the heart and/or muscle are desirable, with minimal transduction in other tissue types, such as, for example, liver and/or dorsal root ganglion cells and/or peripheral nerve cells and/or kidney cells to minimize adverse effects. There also is a need for rAAV vectors with enhanced tissue-specific targeting and/or enhanced tissue-specific transduction to deliver therapies.
Provided are recombinant adeno-associated viruses (rAAVs) having capsid proteins engineered to have one or more amino acid substitutions that reduce or obviate targeting, transduction and/or integration of the rAAV genome in mammalian, including human, tissues, including all tissues of the subject or a subset of tissues, such as, but not limited to, one or more of liver, heart, skeletal muscle (including biceps, transverse abdominal muscle, gastrocnemius muscle, or quadriceps), cardiac muscle, brain, kidney, lung, pancreas, meniscus, and/or peripheral nervous system relative to a reference capsid, for example, the parent capsid, including an AAV8 or AAV9 capsid. Such “atropic” or limited tropic capsids may also be termed “detargeted” for one or more tissue types. The atropic capsids may then be further engineered to incorporate or insert a targeting moiety, such as a peptide, antibody, DARPin, or other molecule, which has a targeting and/or binding domain that enhances transduction of the rAAV in one or more specific tissue types, such as, for example but not limited to, CNS, muscle, or heart, relative to the parent atropic (or limited tropic) capsid (having the one or more amino acid substitutions) and/or the ultimate patent capsid, including AAV8 or AAV9. Biodistribution studies in mice and non-human primates permit assessment of relative transduction and transgene transcription and expression in various tissue types of capsids, including engineered capsids (see, Examples 13-20, infra).
In particular, provided are AAV9 capsid proteins or AAV8 capsid proteins (SEQ ID NO:74 or 73, respectively, and as numbered in
These atropic (or limited tropic) capsids may be further engineered to include a heterologous molecule, including a polypeptide, such as a peptide, antibody or antigen binding domain thereof (including single domain antibodies) or other binding or targeting domain, such as a DARPin, such that the polypeptide or other molecule is displayed on the surface of the rAAV when the engineered capsid protein is incorporated into the capsid, and confers tissue tropism onto the AAV particles having the engineered capsid relative to the atropic or reduced tropic capsid (or even the parental capsid engineered to make the atropic capsid). The peptide, antibody, antigen binding domain thereof, DARPin or other binding or targeting domain may be inserted at an appropriate position in the VP1 capsid protein, including at or near the VP2 initiation codon, or within the VR-1 region, VR-IV region, or VR-VIII region. For example, the peptide, antibody, antigen binding domain thereof, DARPin or other binding or targeting domain may be inserted at one of position 138, 262-273, 452-461, or 585-593 for AAV9 or corresponding position for a different AAV capsid. Also provided are capsids, particularly AAV9 capsids but including other capsid types, that have one of G266A, N272A, W503A or W503R amino acid substitution and 496-NNN/AAA-498 amino acid substitutions (or corresponding substitutions in a different AAV serotype) and also have a peptide TLAAPFK (SEQ ID NO: 1) inserted between Q588 and A589 (herein PHP.hDYN) or alternatively at an appropriate position, including between S268 and S269 or between S454 and G455 or inserted in another AAV capsid at a corresponding position (see, e.g.,
Accordingly, provided herein are rAAVs with enhanced or increased biodistribution, including transduction, genome integration, transgene transcription and expression, in CNS tissues (including frontal cortex, hippocampus, cerebellum, midbrain) relative to a reference capsid (for example the parental capsid that has the amino acid substitution reducing tissue targeting or transduction, or AAV8 or AAV9), with reduced distribution, including transduction, genome integration, transgene transcription and expression in one or more of the heart, liver, lung, kidney, pancreas, meniscus, muscle, and/or dorsal root ganglion cells (cervical, thoracic, and/or lumbar) compared to the biodistribution of a reference capsid, such as the parental capsid or AAV8 or AAV9, that do not have the amino acid substitutions and insertion of the targeting domain. Such rAAVs may be useful to deliver therapeutic proteins or nucleic acids for the treatment of CNS disease or other disease associated with tissues for which the engineered capsid AAV has increased transduction, for example transgenes provided in Table 1A or 1B.
In addition, provided herein are rAAVs with enhanced or increased biodistribution, including transduction, genome integration, transgene transcription and expression, in skeletal muscle and/or cardiac muscle tissues relative to a reference capsid (for example the unengineered, parental capsid or AAV8 or AAV9), with reduced distribution, including transduction, genome integration, transgene transcription and expression in the liver, lung, kidney, pancreas, meniscus, brain, and/or dorsal root ganglion cells (cervical, thoracic, and/or lumbar) compared to the biodistribution in skeletal and/or cardiac muscle tissue and/or relative to an AAV with a reference capsid, such as the parental capsid or AAV8 or AAV9. Such rAAVs may be useful to deliver therapeutic proteins or nucleic acids for the treatment of muscle disease, including transgenes provided in Table 1A and 1B.
In other embodiments, the targeting domain increases targeting of the rAAV to other tissues such as lung, kidney, pancreas, peripheral nervous system.
In certain embodiments, transduction is measured by detection of transgene, such as GFP fluorescence.
The capsid protein to be engineered may be an AAV9 capsid protein but may also be any AAV capsid protein, such as AAV serotype 1 (SEQ ID NO:63); AAV serotype 2 (SEQ ID NO:64); AAV serotype 3 (SEQ ID NO:65), AAV serotype 3-3 (SEQ ID NO: 66); AAV serotype 3B (SEQ ID NO:67); AAV serotype 4 (SEQ ID NO:68); AAV serotype 4-4 (SEQ ID NO:69); AAV serotype 5 (SEQ ID NO:70); AAV serotype 6 (SEQ ID NO:71); AAV serotype 7 (SEQ ID NO:72); AAV serotype 8 (SEQ ID NO:73); AAV serotype 9 (SEQ ID NO:74); AAV serotype 9e (SEQ ID NO:75); AAV serotype rh10 (SEQ ID NO:76); AAV serotype rh20 (SEQ ID NO:77); and AAV serotype hu.37 (SEQ ID NO:85), AAV serotype rh39 (SEQ ID NO:78), and AAV serotype rh74 (SEQ ID NO:80 or SEQ ID NO:81), AAV serotype rh.34 (SEQ ID NO:88), AAV serotype hu.60, AAV serotype rh.21 (SEQ ID NO:93), AAV serotype rh.15, AAV serotype rh.24, AAV serotype hu.5, AAV serotype hu.10, AAV serotype rh64R1 (SEQ ID NO:48), AAV serotype rh46 (SEQ ID NO:97), and AAV serotype rh73 (SEQ ID NO:79) (see
In certain embodiments, provided are rAAVs incorporating the engineered capsids described herein, including rAAVs with genomes comprising a transgene of therapeutic interest, including a transgene encoding a therapeutic protein or nucleic acid for treatment of a muscle, heart or CNS disease or other disease associated with tissue for which the engineered AAV has increased tropism (see, for example, the transgenes in Tables 1A and 1B). Packaging cells for producing the rAAVs described herein are provided which comprise nucleic acids encoding an engineered capsid described herein under the control of appropriate regulatory elements. Method of treatment by delivery of, and pharmaceutical compositions comprising, the engineered rAAVs described herein are also provided. Also provided are methods of manufacturing the rAAVs with the engineered capsids described herein. Provided are nucleic acids encoding the engineered capsid proteins, plasmid vectors, such as “RepCap” constructs in which the Cap gene encodes an engineered capsid described herein as well as host cells, such as bacterial host cells, for replication and production of these plasmid vectors.
The invention is illustrated by way of examples infra describing the construction of rAAV9 capsids engineered with amino acid substitutions and assaying of tissue distribution when administered to mice or non-human primates.
1. A recombinant AAV capsid protein comprising one or more amino acid substitutions relative to the wild type or unengineered capsid protein, in which the rAAV capsid protein is an AAV9 capsid protein (SEQ ID NO:74) with (1) a G266A substitution, a G272A substitution, a W503R substitution, or a W503A substitution and (2) 496-NNN/AAA-498 substitutions, or a capsid protein of a type other than AAV9 with corresponding substitutions in a capsid protein of another AAV type capsid, which when the capsid is incorporated into an rAAV vector, the rAAV vector exhibits reduced transduction of at least one tissue type relative to a rAAV vector incorporating the corresponding wild type capsid protein without the amino acid substitutions.
2. The recombinant AAV capsid protein of embodiment 1 which is an AAV9.496-NNN/AAA-498.W503R capsid (SEQ ID NO:32), an AAV9.N272A.496-NNN/AAA-498 capsid (SEQ ID NO:49), an AAV9.G266A.496-NNN/AAA-498 capsid (SEQ ID NO:50) or an AAV9.496-NNN/AAA-498.W503A capsid (SEQ ID NO:51) or having corresponding amino acid substitutions in a capsid protein of another AAV type capsid.
3. The recombinant AAV capsid protein of embodiments 1 or 2 in which the amino acid substitutions or insertions are in an AAV9 capsid, including an AAV.PHP.eB capsid, protein, or an AAV8 capsid.
4. The recombinant AAV capsid protein of embodiment 1 or 2 wherein the AAV type capsid is AAV rh.34, AAV4, AAV5, AAV hu.26, AAV rh.31, AAV hu.13, AAV hu.26, AAV hu.56, AAV hu.53, AAV7, AAV rh.10, AAV rh.64.R1, AAV rh.46 or AAV rh.73.
5. The recombinant AAV capsid protein of any of embodiments 1 to 4, which when incorporated into a rAAV vector, the rAAV vector has decreased targeting, transduction or genome integration into one or more of heart, lung, kidney, pancreas, meniscus, muscle, brain or liver, relative to a rAAV vector incorporating the corresponding wild type capsid protein without the amino acid substitutions.
6. The recombinant AAV capsid protein of embodiment 5, which when incorporated into a rAAV vector, the rAAV vector has decreased targeting, transduction or genome integration into heart, lung, kidney, pancreas, meniscus, muscle, brain and liver tissues relative to an rAAV vector incorporating the corresponding wild type capsid protein without the amino acid substitutions.
7. The recombinant AAV capsid protein of any of embodiments 1 to 6 where the transduction is reduced by 2 fold, 5 fold, 10 fold, 20 fold, 50 fold, 100 fold, 1000 fold, 10,000 fold relative to that of an rAAV vector incorporating the corresponding wild type capsid.
8. A recombinant AAV capsid protein in which the recombinant capsid protein of any of embodiments 1 to 7 further comprises an insertion of a targeting domain and which, when incorporated into an rAAV vector, the rAAV vector has increased targeting, transduction or genome integration into one or more tissue types relative to an rAAV vector incorporating the capsid protein that is identical to the recombinant capsid protein except for the insertion of the targeting domain.
9. The recombinant AAV capsid of embodiment 8 wherein the targeting domain is a peptide of 7 to 20 amino acids, an antibody or antigen-binding domain thereof, or a DARPin.
10. The recombinant AAV capsid of embodiment 9 wherein the targeting domain is the peptide TLAVPFK (SEQ ID NO:20), TLAAPFK (SEQ ID NO:1), TILSRSTQTG (SEQ ID NO:15), LPVAS (SEQ ID NO:6), CLPVASC (SEQ IN NO:5), or RTIGPSV (SEQ ID NO: 12) or any peptide in Tables 5A-5C.
11. The recombinant AAV capsid of embodiment 8 wherein the targeting domain is an scFv, single domain antibody, a minibody, a diabody or an scFv-Fc.
12. The recombinant AAV capsid of any of embodiments 8 to 11 wherein the targeting domain targets skeletal muscle, cardiac muscle, CNS tissue, lung, kidney, pancreas, meniscus, or liver.
13. The recombinant AAV capsid protein of any of embodiments 8 to 12 wherein the insertion is at or near the VP2 initiation codon, or within the VR-1 region, VR-IV region, or VR-VIII region.
14. The recombinant capsid protein of any of embodiments 8 to 13, wherein the insertion is at one of position 138, 262-273, 452-461, or 585-593 for AAV9 or corresponding position for a different AAV capsid.
15. The recombinant capsid protein of any of embodiments 8 to 14, wherein the insertion is between Q588 and A589, S268 and S269, or S454 and G455 of AAV9 or corresponding position of a different AAV capsid.
16. The recombinant AAV capsid protein of any of embodiments 8 to 15, which when incorporated into a rAAV vector, the rAAV vector has increased targeting, transduction or genome integration into CNS cells, relative to a rAAV vector incorporating the corresponding capsid protein without the targeting domain insertion.
17. The recombinant AAV capsid protein of any of embodiments 8 to 15, which when incorporated into a rAAV vector, the rAAV vector has increased targeting, transduction or genome integration into skeletal and/or cardiac muscle cells, relative to a rAAV vector incorporating the corresponding capsid protein without the the targeting domain insertion.
18. The recombinant capsid protein of embodiment 8 to 17, which when incorporated into a rAAV vector, the rAAV vector has decreased targeting, transduction or genome integration into liver cells, relative to a rAAV vector incorporating the corresponding capsid protein without the targeting domain insertion.
19. The recombinant capsid protein of embodiment 18, which when incorporated into a rAAV vector, the rAAV vector has decreased targeting, transduction or genome integration into CNS cells, relative to a rAAV vector incorporating the corresponding capsid protein without the targeting domain insertion.
20. The recombinant capsid protein of any of embodiments 8 to 16 or 18, which when incorporated into a rAAV vector, the rAAV vector has decreased targeting, transduction or genome integration into dorsal root ganglion cells, relative to an rAAV vector incorporating the corresponding capsid protein without the targeting domain insertion.
21. A nucleic acid comprising a nucleotide sequence encoding the rAAV capsid protein of any of embodiments 1 to 20, or encoding an amino acid sequence sharing at least 80% identity therewith and retaining the biological activity of the capsid.
22. The nucleic acid of embodiment 18 encoding the rAAV capsid protein of any of embodiments 1 to 20.
23. A plasmid vector comprising the nucleic acid of embodiment 21 or 22 which is replicable in a bacterial cell.
24. A bacterial host cell comprising the plasmid vector of embodiment 23.
25. A packaging cell which expresses the nucleic acid of embodiment 21 or 22 to produce AAV vectors comprising the capsid protein encoded by said nucleotide sequence.
26. A rAAV vector comprising the capsid protein of any of embodiments 1 to 20.
27. The rAAV vector of embodiment 26 further comprising a nucleic acid comprising a transgene encoding a therapeutic protein or nucleic acid operably linked to a regulatory sequence for expression in the target cells and flanked by AAV ITR sequences.
28. A pharmaceutical composition comprising the rAAV vector of embodiment 26 or 27 and a pharmaceutically acceptable carrier.
29. A method of delivering a transgene to a cell, said method comprising contacting said cell with the rAAV vector of embodiment 26 or 27 wherein said transgene is delivered to said cell.
30. The method of embodiment 29 in which the cell is a CNS cell, cardiac muscle cell or skeletal muscle cell.
31. A method of delivering a transgene to a target tissue associated with a disease and treatable by expression of said transgene in said target tissue in a subject in need thereof, said method comprising administering to said subject the rAAV vector of embodiment 26 or 27, wherein the transgene is delivered to and expressed in said target tissue.
32. The method of embodiment 31 wherein the transgene is a muscle disease or heart disease therapeutic and said target tissue is cardiac muscle or skeletal muscle.
33. The method of embodiment 32, wherein the rAAV is administered systemically, including intravenously or intramuscularly.
34. The method of embodiment 31 wherein the transgene is a CNS disease therapeutic and said target tissue is CNS.
35. The method of embodiment 34 wherein the rAAV is administered intrathecally, intracerebroventricularly or intravenously.
36. A pharmaceutical composition for use in delivering a transgene to a cell, said pharmaceutical composition comprising the rAAV vector of embodiment 26 or 27, wherein said transgene is delivered to said cell.
37. A pharmaceutical composition for use in delivering a transgene encoding a therapeutic protein or nucleic acid to a target tissue associated with a disease treatable by expression of the transgene in said target tissue of a subject in need thereof, said pharmaceutical composition comprising the rAAV vector of embodiment 26 or 27, wherein the transgene is delivered to and expressed in said target tissue.
38. The pharmaceutical composition of embodiment 36 or 37 wherein said therapeutic protein or nucleic acid is a muscle disease therapeutic or a heart disease therapeutic and said target tissue is cardiac muscle or skeletal muscle.
39. The pharmaceutical composition of embodiment 36 to 38 wherein the rAAV exhibits at least 1.1-fold, 1.5-fold, 2-fold, 3-fold, 4-fold, 5-fold, 6-fold, 7-fold, 8-fold, 9-fold, or 10-fold greater transduction in cardiac muscle or skeletal muscle cells compared to a reference AAV capsid.
40. The pharmaceutical composition of embodiment 36 or 37 wherein said therapeutic protein is a CNS disease therapeutic and said target tissue is CNS.
41. The pharmaceutical composition of embodiment 36, 37 or 40 wherein the rAAV exhibits at least 1.1-fold, 1.5-fold, 2-fold, 3-fold, 4-fold, 5-fold, 6-fold, 7-fold, 8-fold, 9-fold, or 10-fold greater transduction in CNS cells compared to a reference AAV capsid.
42. The pharmaceutical composition of embodiment 36 to 41 wherein the rAAV exhibits at least 50%, 60%, 70%, 80%, 90%, 95% or 99% less transduction in liver compared to the reference AAV capsid.
43. The pharmaceutical composition of embodiment 36 to 42 wherein the rAAV exhibits at least 50%, 60%, 70%, 80%, 90%, 95% or 99% less transduction in dorsal root ganglion cells compared to the reference AAV capsid.
44. The pharmaceutical composition of embodiments 36 to 43, wherein the AAV reference capsid is AAV8 or AAV9.
45. A method of treating a CNS disorder in a subject in need thereof, said method comprising administering a therapeutically effective amount of pharmaceutical composition of any of embodiments 36, 37, or 40 to 44.
46. A method of treating a muscle disorder in a subject in need thereof, said method comprising administering a therapeutically effective amount of the pharmaceutical composition of any of embodiments 36, 37-39, or 39-41.
47. A method of or pharmaceutical composition for use in delivering a transgene to a target cell or target tissue associated with a disease treatable by expression of said transgene in said target tissue, said method or use comprising administering an rAAV vector comprising an AAV9 capsid protein (SEQ ID NO:74) with (1) a G266A substitution, (2) 496-NNN/AAA-498 substitutions and (3) insertion of a targeting domain which targets the target tissue, said rAAV vector comprising an artificial genome comprising the transgene operably linked to a regulatory sequence for expression in said target tissue, wherein the rAAV vector transduces the target cell or target tissue upon binding of the targeting domain to the target cell or target tissue, thus delivering the transgene to the target cell or target tissue.
48. A method of or pharmaceutical composition for use in delivering a transgene to a target cell or target tissue associated with a disease treatable by expression of said transgene in said target tissue, said method or use comprising administering an rAAV vector comprising an AAV9 capsid protein (SEQ ID NO:74) with (1) a G272A substitution, (2) 496-NNN/AAA-498 substitutions and (3) insertion of a targeting domain which targets the target tissue, said rAAV vector comprising an artificial genome comprising the transgene operably linked to a regulatory sequence for expression in said target tissue, wherein the rAAV vector transduces the target cell or target tissue upon binding of the targeting domain to the target cell or target tissue, thus delivering the transgene to the target cell or target tissue.
49. A method of or pharmaceutical composition for use in delivering a transgene to a target cell or target tissue associated with a disease treatable by expression of said transgene in said target tissue, said method or use comprising administering an rAAV vector comprising an AAV9 capsid protein (SEQ ID NO:74) with (1) a W503R substitution, (2) 496-NNN/AAA-498 substitutions and (3) insertion of a targeting domain which targets the target tissue, said rAAV vector comprising an artificial genome comprising the transgene operably linked to a regulatory sequence for expression in said target tissue, wherein the rAAV vector transduces the target cell or target tissue upon binding of the targeting domain to the target cell or target tissue, thus delivering the transgene to the target cell or target tissue.
50. A method of or pharmaceutical composition for use in delivering a transgene to a target cell or target tissue associated with a disease treatable by expression of said transgene in said target tissue, said method or use comprising administering an rAAV vector comprising an AAV9 capsid protein (SEQ ID NO:74) with (1) a W503A substitution, (2) 496-NNN/AAA-498 substitutions and (3) insertion of a targeting domain which targets the target tissue, said rAAV vector comprising an artificial genome comprising the transgene operably linked to a regulatory sequence for expression in said target tissue, wherein the rAAV vector transduces the target cell or target tissue upon binding of the targeting domain to the target cell or target tissue, thus delivering the transgene to the target cell or target tissue.
51. The method or composition of any of embodiments 47-50, wherein the insertion is at or near the VP2 initiation codon, or within the VR-1 region, VR-IV region, or VR-VIII region.
52. The method or composition of any of embodiments 47 to 51, wherein the insertion is at one of position 138, 262-273, 452-461, or 585-593 for AAV9 or corresponding position for a different AAV capsid.
53. The method or composition of any of embodiments 47 to 52, wherein the insertion is between Q588 and A589, S268 and S269, or S454 and G455 of AAV9 or corresponding position of a different AAV capsid
54. The method or composition or rAAV of any of the foregoing, wherein the rAAV comprises a nucleic acid comprising a nucleic acid sequence encoding a therapeutic protein or nucleic acid operably linked to a regulatory sequence that promotes expression of the therapeutic protein in the target cell or tissue flanked by AAV ITR sequence, such that upon transduction of the rAAV into the target cell or target tissue, the therapeutic protein or nucleic acid is expressed.
Provided are recombinant adeno-associated viruses (rAAVs) having capsid proteins engineered relative to a reference capsid protein, such that the rAAV has enhanced desired properties, such as altered tissue targeting, including transduction, genome integration and transgene expression, particularly, preferentially, relative to the reference capsid protein (e.g., the unengineered or wild type capsid), to CNS or to heart and/or skeletal muscle tissue or other tissue. In embodiments, the engineered capsid has one or more amino acid substitutions resulting in reduced tropism or atropisms (i.e., tissue targeting, transduction and integration of the rAAV genome) relative to the reference capsid (e.g., AAV9 or AAV8) for all or a subset of tissues, including one or more of heart, lung, kidney, pancreas, meniscus, liver, muscle (including biceps, transabdominal muscle, gastrocnemius muscle, and quadriceps), dorsal root ganglion and/or peripheral nervous tissue. The reduction may be a one fold, 2 fold, 5 fold, 10 fold, 20 fold, 50 fold, 100 fold, 1000 fold, 10,000 fold or even greater reduction relative to a reference capsid. The modifications include amino acid substitutions (including 1, 2, 3, 4, 5, 6, 7 or 8 amino acid substitutions), including, for AAV9, the amino acid substitutions G266A, N272A, W503R or W503A, or the corresponding amino acid substitutions for a different AAV capsid (see alignment
The engineered capsids with reduced or obviated transduction for all tissue types or one or more of heart, lung, kidney, pancreas, meniscus, liver, skeletal muscle, brain or peripheral nervous system may be further engineered to comprise a peptide insertion or attachment of another moiety, which peptide or moiety targets the rAAV to one or more tissue types, conferring tropism on the engineered capsid of the rAAV, where, for example, the peptide or moiety includes a binding domain for a receptor or other cell surface moiety characteristic of one or more tissue types. The increased targeting or transduction of tissue may be any tissue or combination of tissues, for example, skeletal muscle, heart, central nervous system, etc. In embodiments, the peptide insertion is 4 to 20, or 7 contiguous amino acids of a heterologous (not an AAV) peptide, and in embodiments no more than 12 contiguous amino acids from a heterologous protein as described herein. The targeting domain may also be an antibody or antigen binding domain thereof or other form of binding domain, such as a DARPin. The peptide, antibody, antigen binding domain thereof, DARPin or other binding or targeting domain may be inserted at an appropriate position in the VP1 capsid protein, including at or near the VP2 initiation codon, or within the VR-1 region, VR-IV region, or VR-VIII region. For example, the peptide, antibody, antigen binding domain thereof, DARPin or other binding or targeting domain may be inserted at one of position 138, 262-273, 452-461, or 585-593 for AAV9 or corresponding position for a different AAV capsid.
rAAV having an a tropic (or limited tropic) capsid comprising an insert with a targeting domain may exhibit increased transduction of one or more tissues (including skeletal muscle, heart or CNS) that is 1 fold, 2 fold, 5 fold, 10 fold, 20 fold, 50 fold, 100 fold, 500 fold, 1000 fold, 10,000 fold or 100,000 fold greater than an rAAV having the parental atropic (or limited tropic) capsid without the insert of the targeting domain.
Also provided are engineered capsids, particularly AAV9 capsids having amino acid substitutions of one of G266A, N272A, W503A or W503R and the amino acid substitutions 496-NNN/AAA-498 of AAV9 or corresponding substitutions in a different capsid type and to enhance tissue specific transduction, further comprising a peptide insert of TLAAPFK (SEQ ID NO: 1) inserted between Q588 and A589 (herein PHP.hDYN) or alternatively between S268 and S269 or between S454 and G455) or inserted in another AAV capsid at a corresponding position (see, e.g.,
In another embodiment, provided is a recombinant capsid protein, including an engineered AAV9 capsid protein (having the amino acid substitutions which reduce transduction of one or more tissues), and an rAAV comprising the capsid protein, in which the peptide TLAVPFK (SEQ ID NO:20) is or further comprising the peptide TLAVPFK (SEQ ID NO:20) inserted between G588 and A589 of AAV9, and, in particular, the capsid protein also has amino acid substitutions A587D/Q588G (PHP.eB) and further has the peptide TILSRSTQTG (SEQ ID NO:15) inserted after position 138 of AAV9 (collectively, AAVPHPeB.VP2Herp; see Table 17), or in the corresponding positions of another AAV. Provided are recombinant AAV capsids comprising an amino acid substitution of G266A, W503A or W503R of AAV9 (or corresponding amino acid substitution in another capsid) and further comprising insertion of the peptide RTIGPSV (SEQ ID NO:12), including inserted between positions 138 and 139, positions S454 and G455, or positions Q588 and A589 of AAV9, or corresponding position of another AAV capsid, or at any other position that displays the peptide on the capsid surface to promote tissue specific binding and transduction, for example, including at or near the VP2 initiation codon, or within the VR-1 region, VR-IV region, or VR-VIII region. Additional capsids further comprise a Kidney1 peptide LPVAS (SEQ ID NO:6) (or alternatively CLPVASC (SEQ ID NO:5)) inserted into the capsid, for example between S454 and G455 of AAV9 (see Table 17), or alternatively between S268 and S269 or between Q588 and A589, or the corresponding position of a different capsid or at any other position that displays the peptide on the capsid surface to promote tissue specific binding and transduction, for example, including at or near the VP2 initiation codon, or within the VR-1 region, VR-IV region, or VR-VIII region. Such a capsid comprising a targeting domain may exhibit preferential targeting for heart and skeletal muscle, and reduced targeting (as compared to an AAV having the unengineered capsid) for liver and/or dorsal root ganglion cells and may particularly useful for delivery of a transgene encoding a therapeutic protein or nucleic acid for treatment of a muscle disease (such as, but not limited to a muscular dystrophy).
In embodiments the engineered rAAV exhibits at least 1.1-fold, 1.5-fold, 2-fold, 3-fold, 4-fold, 5-fold, 6-fold, 7-fold, 8-fold, 9-fold, or 10-fold greater transduction in cardiac muscle and/or skeletal muscle cells compared to a reference AAV capsid, including an AAV9 capsid or an AAV8 capsid, or the atropic parental capsid having the amino acid substitutions which reduce or obviate rAAV transduction of one or more tissues. In particular embodiments, the muscle is gastrocnemius muscle, bicep, tricep and/or heart muscle. In further embodiments, the engineered rAAV exhibits of 50%, 60%, 70%, 80%, 90%, 95% or 99% less transduction in liver compared to the reference AAV capsid compared to a reference AAV capsid, including an AAV9 capsid or an AAV8 capsid, or the parental capsid. In further embodiments, the rAAV exhibits of 50%, 60%, 70%, 80%, 90%, 95% or 99% less transduction in dorsal root ganglion cells (including in cervical, thoracic or lumbar DRG cells) compared to the reference AAV capsid. The enhanced and/or reduce transduction may be with any mode of administration, by intravenous administration, intramuscular administration, or any type of systemic administration, intrathecal administration or ICV administration.
Also provided are engineered capsids having one or more amino acid substitutions that alter transduction and/or tissue tropism, for example, promote transduction and/or tissue tropism, particularly for enhanced, relative to an unengineered capsid (or capsid having only the amino acid substitutions which detarget the capsid from one or more tissues), targeting for CNS and, in embodiments, reduced, relative to an unengineered capsid or an atropic or reduced tropic capsid, targeting for liver, heart, skeletal muscle, lung, kidney, pancreas, meniscus, dorsal root ganglion, and/or peripheral nervous tissue. In embodiments, the amino acid substitutions are A269S of AAV8 (or at a corresponding position in a different AAV serotype capsid), S263G/S269T/A273T of AAV9 (or at a corresponding position in a different AAV serotype capsid), N272A or G266A of AAV9 (or at a corresponding position in a different AAV serotype capsid), Q474A of AAV9 (or at a corresponding position in a different AAV serotype capsid), or W503R of AAV9 (or at a corresponding position in a different AAV serotype capsid), or R697W of rh64R1 (or at a corresponding position in a different AAV serotype capsid). The capsids having these amino acid substitutions and insertions may further have or alternatively have substitutions of the NNN (asparagines) at 496 to 498 with AAA (alanines) of the AAV9 capsid (SEQ ID NO:74) or have substitutions of the NNN (asparagines) at 498 to 500 with AAA (alanines) of the AAV8 capsid (SEQ ID NO:37), or corresponding substitutions in other AAV type capsids. Capsids comprising a targeting domain may exhibit preferential targeting for CNS, and reduced targeting (compared to an AAV bearing the unengineered capsid) for liver, heart, muscle, lung, kidney, pancreas, meniscus, and/or dorsal root ganglion cells and/or peripheral nervous system tissue, and may particularly useful for delivery of a transgene encoding a therapeutic protein or nucleic acid for treatment of a CNS disease.
Also provided are recombinant capsid proteins, and rAAVs comprising them, that have inserted peptides that target and/or promote rAAV cellular uptake, transduction and/or genome integration in CNS tissue and, in embodiments, reduced, relative to an unengineered capsid (or atropic capsid), targeting for liver, heart, muscle, lung, kidney, pancreas, meniscus, dorsal root ganglion, and/or peripheral nervous tissue, for example, the peptide TILSRSTQTG (SEQ ID NO:15); TLAVPFK (SEQ ID NO:20); or TLAAPFK (SEQ ID NO:1). In particular embodiments the peptide TLAAPFK (SEQ ID NO:1) is inserted between Q588 and A589 of AAV9 (AAV9.hDyn; see Table 17), or the corresponding position of another AAV (see FIG. 7). Alternatively, the capsid is rh.34, rh.10, rh.46, rh.73, or rh64.R1 (
In embodiments the engineered rAAV exhibits at least 1.1-fold, 1.5-fold, 2-fold, 3-fold, 4-fold, 5-fold, 6-fold, 7-fold, 8-fold, 9-fold, or 10-fold greater transduction in CNS tissue compared to a reference AAV capsid, such as the parental capsid (including an atropic or reduced tropic capsid) or AAV8 or AAV9. The CNS tissue may be one or more of the frontal cortex, hippocampus, cerebellum, midbrain and/or hindbrain. In further embodiments, the engineered rAAV exhibits of 50%, 60%, 70%, 80%, 90%, 95% or 99% less transduction in liver compared to the reference AAV capsid such as the parental capsid or AAV8 or AAV9. In further embodiments, the rAAV exhibits of 50%, 60%, 70%, 80%, 90%, 95% or 99% less transduction in dorsal root ganglion cells (including in cervical, thoracic or lumbar DRG cells) compared to the reference AAV capsid such as the parental capsid or AAV8 or AAV9. The enhanced and/or reduce transduction may be with any mode of administration, by intravenous administration, intramuscular administration, or any type of systemic administration, intrathecal administration or ICV administration.
Recombinant vectors comprising the capsid proteins also are provided, along with pharmaceutical compositions thereof, nucleic acids encoding the capsid proteins, and methods of making and using the capsid proteins and rAAV vectors having the engineered capsids for targeted delivery, improved transduction and/or treatment of disorders associated with the target tissue.
As used throughout, AAV “serotype” refers to an AAV having an immunologically distinct capsid, a naturally-occurring capsid, or an engineered capsid.
The term “AAV” or “adeno-associated virus” refers to a Dependoparvovirus within the Parvoviridae genus of viruses. The AAV can be an AAV derived from a naturally occurring “wild-type” virus, an AAV derived from a rAAV genome packaged into a capsid comprising capsid proteins encoded by a naturally occurring cap gene and/or from a rAAV genome packaged into a capsid comprising capsid proteins encoded by a non-naturally occurring capsid cap gene. An example of the latter includes a rAAV having a capsid protein comprising a peptide insertion into the amino acid sequence of the naturally-occurring capsid.
The term “rAAV” refers to a “recombinant AAV.” In some embodiments, a recombinant AAV has an AAV genome in which part or all of the rep and cap genes have been replaced with heterologous sequences.
The term “rep-cap helper plasmid” refers to a plasmid that provides the viral rep and cap gene function and aids the production of AAVs from rAAV genomes lacking functional rep and/or the cap gene sequences.
The term “cap gene” refers to the nucleic acid sequences that encode capsid proteins that form or help form the capsid coat of the virus. For AAV, the capsid protein may be VP1, VP2, or VP3.
The term “rep gene” refers to the nucleic acid sequences that encode the non structural protein needed for replication and production of virus.
As used herein, the terms “nucleic acids” and “nucleotide sequences” include DNA molecules (e.g., cDNA or genomic DNA), RNA molecules (e.g., mRNA), combinations of DNA and RNA molecules or hybrid DNA/RNA molecules, and analogs of DNA or RNA molecules. Such analogs can be generated using, for example, nucleotide analogs, which include, but are not limited to, inosine or tritylated bases. Such analogs can also comprise DNA or RNA molecules comprising modified backbones that lend beneficial attributes to the molecules such as, for example, nuclease resistance or an increased ability to cross cellular membranes. The nucleic acids or nucleotide sequences can be single-stranded, double-stranded, may contain both single-stranded and double-stranded portions, and may contain triple-stranded portions, but preferably is double-stranded DNA.
As used herein, the terms “subject”, “host”, and “patient” are used interchangeably. As used herein, a subject is a mammal such as a non-primate (e.g., cows, pigs, horses, cats, dogs, rats etc.) or a primate (e.g., monkey and human), or, in certain embodiments, a human.
As used herein, the terms “therapeutic agent” refers to any agent which can be used in treating, managing, or ameliorating symptoms associated with a disease or disorder, where the disease or disorder is associated with a function to be provided by a transgene. As used herein, a “therapeutically effective amount” refers to the amount of agent, (e.g., an amount of product expressed by the transgene) that provides at least one therapeutic benefit in the treatment or management of the target disease or disorder, when administered to a subject suffering therefrom. Further, a therapeutically effective amount with respect to an agent of the invention means that amount of agent alone, or when in combination with other therapies, that provides at least one therapeutic benefit in the treatment or management of the disease or disorder.
As used herein, the term “prophylactic agent” refers to any agent which can be used in the prevention, delay, or slowing down of the progression of a disease or disorder, where the disease or disorder is associated with a function to be provided by a transgene. As used herein, a “prophylactically effective amount” refers to the amount of the prophylactic agent (e.g., an amount of product expressed by the transgene) that provides at least one prophylactic benefit in the prevention or delay of the target disease or disorder, when administered to a subject predisposed thereto. A prophylactically effective amount also may refer to the amount of agent sufficient to prevent or delay the occurrence of the target disease or disorder; or slow the progression of the target disease or disorder; the amount sufficient to delay or minimize the onset of the target disease or disorder; or the amount sufficient to prevent or delay the recurrence or spread thereof. A prophylactically effective amount also may refer to the amount of agent sufficient to prevent or delay the exacerbation of symptoms of a target disease or disorder. Further, a prophylactically effective amount with respect to a prophylactic agent of the invention means that amount of prophylactic agent alone, or when in combination with other agents, that provides at least one prophylactic benefit in the prevention or delay of the disease or disorder.
A prophylactic agent of the invention can be administered to a subject “pre-disposed” to a target disease or disorder. A subject that is “pre-disposed” to a disease or disorder is one that shows symptoms associated with the development of the disease or disorder, or that has a genetic makeup, environmental exposure, or other risk factor for such a disease or disorder, but where the symptoms are not yet at the level to be diagnosed as the disease or disorder. For example, a patient with a family history of a disease associated with a missing gene (to be provided by a transgene) may qualify as one predisposed thereto. Further, a patient with a dormant tumor that persists after removal of a primary tumor may qualify as one predisposed to recurrence of a tumor.
The “central nervous system” (“CNS”) as used herein refers to neural tissue reaches by a circulating agent after crossing a blood-brain barrier, and includes, for example, the brain, optic nerves, cranial nerves, and spinal cord. The CNS also includes the cerebrospinal fluid, which fills the central canal of the spinal cord as well as the ventricles of the brain.
Provided are recombinant adeno-associated viruses (rAAVs) having capsid proteins engineered relative to a reference capsid protein, such that the rAAV has enhanced desired properties, such as altered tissue targeting, including transduction, genome integration and transgene expression, particularly, preferentially, relative to the reference capsid protein (e.g., the unengineered or wild type capsid), to CNS or to heart and/or skeletal muscle tissue or any other type of tissue. In embodiments, provided are atropic or reduced tropic engineered capsids which comprise or consist of one or more amino acid substitutions resulting in reduced tropism or atropisms (i.e., tissue targeting, transduction and integration of the rAAV genome) relative to the reference capsid (e.g., AAV9 or AAV8) for all or a subset of tissues, including one or more of heart, lung, kidney, pancreas, meniscus, liver, muscle (including biceps, transabdominal muscle, gastrocnemius muscle, and quadriceps), brain, dorsal root ganglion and/or peripheral nervous tissue. The reduction may be a one fold, 2 fold, 5 fold, 10 fold, 20 fold, 50 fold, 100 fold, 1000 fold, 10,000 fold or even greater reduction relative to a reference capsid. The modifications include amino acid substitutions (including 1, 2, 3, 4, 5, 6, 7 or 8 amino acid substitutions), including, for AAV9, the amino acid substitutions G266A, N272A, W503R or W503A, or the corresponding amino acid substitutions for a different AAV capsid (see alignment
5.2.1 Engineered Capsids with Amino Acid Substitutions
In some embodiments, AAV capsids were modified by introducing selected single to multiple amino acid substitutions which reduce the transduction of vectors incorporating the AAV capsids to one or more tissue types, including liver, heart, muscle, brain, lung, kidney, pancreas, meniscus, and/or muscle to generate atropic or reduced tropic capsids and/or increase effective gene delivery to the CNS or to cardiac or skeletal muscle or other target tissue (including when the atropic or limited tropic capsids incorporate a targeting domain to enhance tropism to CNS or to cardiac or skeletal muscle or other target tissue), detarget the liver and/or dorsal root ganglion to reduce toxicity, and/or reduce immune responses of neutralizing antibodies.
In particular embodiments the capsids have one or more amino acid substitutions including a W503A or W503R substitution, a Q474 substitution, a N272A or N266A substitution in AAV9 or the corresponding substitution in another AAV serotype or an A269S substitution in AAV8 or the corresponding substitution in another AAV serotype. rAAV having a capsid with the Q474A substitution may be particularly useful for delivery to skeletal and/or cardiac muscle or CNS tissue and rAAV having a capsid with the W503R substitution may be particularly useful for delivery to CNS tissue, particularly with reduced, compared to reference capsid containing rAAVs, transduction in the liver and/or DRGs. Other substitutions include S263G/S269R/A273T substitutions in AAV9 or A587D/Q588G in AAV9 or corresponding substitutions in other AAV serotypes. In some embodiments, the rAAV capsid can have a R697W substitution. The capsids having these amino acid substitutions and insertions may further have substitutions of the NNN (asparagines) at 496 to 498 with AAA (alanines) of the AAV9 capsid, or of the NNN (asparagines) at 498 to 500 with AAA (alanines) of the AAV8 capsid corresponding substitutions in other AAV type capsids. Other AAV serotypes that may be used for the amino acid substitutions and that may be the reference capsid include AAV8, AAV rh.34, AAV4, AAV5, AAV hu.26, AAV rh.31, AAV hu. 13, AAV hu.26, AAV hu.56, AAV hu.53, AAV7, rh64R1, rh46 or rh73. In particular embodiments for CNS delivery, the capsid is rh34, either unmodified or serving as the parental capsid to be modified as detailed herein.
Provided are atropic capsids which may be further be modified to confer specific tissue tropism and/or enhanced tissue-specific transduction by inserting a targeting domain (or introducing one or more amino acid substitutions which create a tissue specific binding domain within the capsid). Such atropic capsids include AAV9 and other capsids comprising or consisting of an amino acid substitution of G266A, N272A, W503R or W503A for AAV9, or the corresponding amino acid substitutions for a different AAV capsid (see alignment
Effective gene delivery to the CNS by intravenously administered rAAV vectors requires crossing the blood brain barrier. Key clusters of residues on the AAVrh. 10 capsid that enabled transport across the brain vasculature and widespread neuronal transduction in mice have recently been reported. Specifically, AAVrh.10-derived amino acids N262, G263, T264, S265, G267, S268, T269, and T273 were identified as key residues that promote crossing the BBB (Albright et al, 2018, Mapping the Structural Determinants Required for AAVrh.10 Transport across the Blood-Brain Barrier). Amino acid substitutions in capsids, such as AAV8 and AAV9 capsids that promote rAAV crossing of the blood brain barrier, transduction, detargeting of the liver and/or reduction in immune responses have been identified.
In some embodiments, provided are capsids having one or more amino acid substitutions that promote transduction and/or tissue tropism of the rAAV having the modified capsid. In particular embodiments, provided are capsids having a single mutation at amino acid 269 of the AAV8 capsid replacing alanine with serine (A269S) (see, Tables 5a-5c, herein referred to as AAV8.BBB) and amino acid substitutions at corresponding positions in other AAV types. In some embodiments, provided are capsids having multiple substitutions at amino acids 263, 269, and 273 of the AAV9 capsid resulting in the following substitutions: S263G, S269T, and A273T (herein referred to as AAV9.BBB) or substitutions corresponding to these positions in other AAV types. These amino acid substitutions may be incorporated into the atropic capsids described herein (including, for example, AAV9.G266A.496NNN/AAA498 (SEQ ID NO:50), AAV9.N272A.496NNN/AAA498 (SEQ ID NO:49), AAV9.496NNN/AAA498.W503R (SEQ ID NO:32), and AAV9.496NNN/AAA498.W503A (SEQ ID NO:51)).
Exposure to the AAV capsid can generate an immune response of neutralizing antibodies. One approach to overcome this response is to map the AAV-specific neutralizing epitopes and rationally design an AAV capsid able to evade neutralization. A monoclonal antibody, specific for intact AAV9 capsids, with high neutralizing titer has recently been described (Giles et al, 2018, Mapping an Adeno-associated Virus 9-Specific Neutralizing Epitope To Develop Next-Generation Gene Delivery Vectors). The epitope was mapped to the 3-fold axis of symmetry on the capsid, specifically to residues 496-NNN-498 and 588-QAQAQT-592 of AAV9 (SEQ ID NO:8). Capsid mutagenesis demonstrated that single amino acid substitution within this epitope markedly reduced binding and neutralization. In addition, in vivo studies showed that mutations in the epitope conferred a “liver-detargeting” phenotype to the mutant vectors, suggesting that the same residues are also responsible for AAV9 tropism. Liver detargeting has also been associated with substitution of amino acid 503 replacing tryptophan with arginine. Presence of the W503R mutation in the AAV9 capsid was associated with low glycan binding avidity (Shen et al, 2012, Glycan Binding Avidity Determines the Systemic Fate of Adeno-Associated Virus Type 9).
In some embodiments, provided are capsids in which the AAV8.BBB and AAV9.BBB capsids were further modified by substituting asparagines at amino acid positions 498, 499, and 500 of AAV8 (herein referred to as AAV8.BBB.LD) or 496, 497, and 498 of AAV9 (herein referred to as AAV9.BBB.LD) with alanines. In some embodiments, the AAVrh10 capsid was modified by substituting three asparagines at amino acid positions 498, 499, and 500 to alanines (AAVrh10.LD) (Tables 5a-5c).
In some embodiments, provided are capsids having three asparagines at amino acid positions 496, 497, and 498 of the AAV9 capsid replaced with alanines and also tryptophan at amino acid 503 of the AAV9 capsid with alanine or arginine or capsids with substitutions corresponding to these positions in other AAV types. In some embodiments, provided are capsids having glutamine at amino acid position 474 of the AAV9 capsid substituted with alanine or capsids with substitutions corresponding to this position in other AAV types.
In some embodiments, the capsid is an AAV8.BB.LD capsid (A269S,498-NNN/AAA-500 substitutions in the amino acid sequence of AAV8, SEQ ID NO:73), an AAV9.BBB.LD capsid (S263G/S269T/A273T, 496-NNN/AAA-498 substitutions in the amino acid sequence of AAV9, SEQ ID NO:74), an AAV9.496-NNN/AAA-498 capsid (SEQ ID NO:31), an AAV9.496-NNN/AAA-498.W503R capsid (SEQ ID NO:32), an AAV9.W503R capsid (SEQ ID NO:33), or an AAV9.Q474A capsid (SEQ ID NO:34). In other examples, the capsid can be an AAV9.N272A.496-NNN/AAA-498 capsid (SEQ ID NO:49) or an AAV9.G266A.496NNN/AAA498 capsid (SEQ ID NO:50), or AAV9.496NNN/AAA498.W503A (SEQ ID NO:51).
In some embodiments, the rAAVs described herein (including those with atropic or limited tropic capsids having a tissue targeting domain inserted therein) increase tissue-specific (such as, but not limited to, CNS or skeletal and/or cardiac muscle) cell transduction in a subject (a human, non-human-primate, or mouse subject) or in cell culture, compared to the rAAV not comprising the amino acid substitution and/or targeting domain insertion (including relative to parental atropic capsids). In some embodiments, the increase in tissue specific cell transduction is at least 2, 10, 20, 30, 40, 50, 60, 70, 80, 90, or 100 fold more than that without the modification, i.e., relative to the parental capsid, including an atropic or limited tropic capsid. For example, in some embodiments, there is a 50-80 fold increase in tissue specific cell transduction compared to transduction with the same AAV type without the modification. The increase in transduction may be assessed using methods described in the Examples herein and known in the art.
In some embodiments, the rAAVs described herein increase the incorporation of rAAV genomes into a cell or tissue type in a subject (a human, non-human primate or mouse subject) or in cell culture compared to the rAAV (e.g., the parental atropic or limited tropic AAV capsid) not comprising the peptide insertion. In some embodiments, the increase in genome integration is at least 2, 10, 20, 30, 40, 50, 60, 70, 80, 90, or 100 fold more than an AAV having a capsid without the modification (i.e., the parental capsid). For example, in some embodiments, there is a 50-80 fold increase in genome integration compared to genome integration with the same AAV type without the modification.
5.2.2 rAAV Vectors with Peptide Insertions
Provided are rAAVs having capsid proteins with one or more (generally one or two) peptide insertions wherein the peptide insertion increase effective gene delivery to the CNS or to cardiac or skeletal muscle and to detarget the liver and/or dorsal root ganglion to reduce toxicity relative to the parental capsid protein. In particular embodiments, the peptides include TLAVPFK (SEQ ID NO:20), TLAAPFK (SEQ ID NO: 1), or TILSRSTQTG (SEQ ID NO: 15) (or an at least 4, 5, 6, 7 amino acid portion thereof). The peptides may be inserted into the AAV9 capsid, for example after the positions 138; 262-273; 452-461; 585-593 of AAV9 cap, particularly after position 138, 454 or 588 of AAV9 or a corresponding position in another AAV as detailed herein. In particular embodiments, the capsid has the peptide TLAVPFK (SEQ ID NO:20) is inserted between G588 and A589 of AAV9, and, in particular, the capsid protein also has amino acid substitutions A587D/Q588G (PHP.eB) and further has the peptide TILSRSTQTG (SEQ ID NO:15) inserted after position 138 of AAV9 (collectively, AAVPHPeB.VP2Herp; see Table 17), or in the corresponding positions of another AAV. Additional capsids have a Kidney1 peptide LPVAS (SEQ ID NO:6) inserted into the capsid, for example between 454 and 455 of AAV9 (see Table 17), or alternatively or alternatively between S268 and S269 or between Q588 and A589 of AAV9 or the corresponding position of another AAV serotype. Such an engineered capsid may exhibit preferential targeting for heart and skeletal muscle, and reduced targeting (as compared to an AAV having the unengineered capsid) for liver and/or dorsal root ganglion cells and may particularly useful for delivery of a transgene encoding a therapeutic protein or nucleic acid for treatment of a muscle disease (such as, but not limited to a muscular dystrophy).
In some embodiments, the peptide insertion comprises at least 4, 5, 6, 7, 8, 9, or all 10 consecutive amino acids of sequence TILSRSTQTG (SEQ ID NO: 15), preferably which contains the TQT or STQT (SEQ ID NO:9) motif. In some embodiments, the peptide insertion consists of at least 4, 5, 6, 7, 8, 9, or all 10 consecutive amino acids of sequence TILSRSTQTG (SEQ ID NO: 15), preferably which contains the TQT or STQT (SEQ ID NO:9) motif.
In certain embodiments, the peptide insertion may be a sequence of consecutive amino acids from a domain that targets kidney tissue, or a conformation analog designed to mimic the three-dimensional structure of said domain. In some embodiments, the kidney-homing domain comprises the sequence CLPVASC (SEQ ID NO:5) (see, e.g., U.S. Pat. No. 5,622,699). In some embodiments, the peptide insertion from said kidney-homing domain comprises at least 4, 5, 6, or all 7 amino acids from sequence CLPVASC (SEQ ID NO:5). In some embodiments, the peptide insertion comprises or consists of the sequence CLPVASC (SEQ ID NO:5).
It has been found that both of the cysteine residues in certain homing peptides can be deleted without significantly affecting the organ homing activity of the peptide. For example, a peptide having the sequence LPVAS (SEQ ID NO:6) also can be a kidney-homing peptide. Methods for determining the necessity of a cysteine residue or of amino acid residues N-terminal or C-terminal to a cysteine residue for organ homing activity of a peptide are routine and well known in the art. Thus, in some embodiments, the peptide insertion comprises at least 4 or all 5 amino acids from sequence LPVAS (SEQ ID NO:6). In some embodiments, the peptide insertion comprises or consists of the sequence LPVAS (SEQ ID NO:6).
In particular embodiments, provided are rAAVs having a capsid that has the peptide TLAAPFK (SEQ ID NO:1) is inserted between Q588 and A589 of AAV9 (AAV9.hDyn; see Table 4a), or the corresponding position of another AAV (see, e.g.,
Provided are capsids with peptide insertions at positions amenable to peptide insertions within and near the AAV9 capsid VR-IV loop (see
Accordingly, provided are rAAV vectors carrying peptide insertions at these points, in particular, within surface-exposed variable regions in the capsid coat, particularly within or near the variable region IV of the capsid protein. In some embodiments, the rAAV capsid protein comprises a peptide insertion immediately after (i.e., connected by a peptide bond C-terminal to) an amino acid residue corresponding to one of amino acids 451 to 461 of AAV9 capsid protein (amino acid sequence SEQ ID NO:74 and see
A peptide insertion described as inserted “at” a given site refers to insertion immediately after, that is having a peptide bond to the carboxy group of, the residue normally found at that site in the wild type virus. For example, insertion at Q588 in AAV9 means that the peptide insertion appears between Q588 and the consecutive amino acid (A589) in the AAV9 wildtype capsid protein sequence (SEQ ID NO:74). In embodiments, there is no deletion of amino acid residues at or near (within 5, 10, 15 residues or within the structural loop that is the site of the insertion) the point of insertion.
In particular embodiments, the capsid protein is an AAV9 capsid protein (including modified atropic or reduced tropic AAV9 capsids) and the insertion occurs immediately after at least one of the amino acid residues 451 to 461. In particular embodiments, the peptide insertion occurs immediately after amino acid I451, N452, G453, S454, G455, Q456, N457, Q458, Q459, T460, or L461 of the AAV9 capsid (amino acid sequence SEQ ID NO:74). In certain embodiments, the peptide is inserted between residues S454 and G455 of AAV9 capsid protein or between the residues corresponding to S454 and G455 of an AAV capsid protein other than an AAV9 capsid protein (amino acid sequence SEQ ID NO:74).
In other embodiments, provided are engineered capsid proteins comprising targeting peptides heterologous to the capsid protein that are inserted into the AAV capsid protein such that, when incorporated into the AAV vector the heterologous peptide is surface exposed.
In other embodiments, the capsid protein is from at least one AAV type selected from AAV serotype 1 (AAV1), serotype 2 (AAV2), serotype 3 (AAV3), serotype 4 (AAV4), serotype 5 (AAV5), serotype 6 (AAV6), serotype 7 (AAV7), serotype 8 (AAV8), serotype rh8 (AAVrh8), serotype 9e (AAV9e), serotype rh10 (AAVrh10), serotype rh20 (AAVrh20), serotype rh39 (AAVrh39), serotype hu.37 (AAVhu.37), serotype rh74 (AAVrh74, versions 1 and 2), serotype rh34 (AAVrh34), serotype hu26 (AAVhu26), serotype rh31 (AAVrh31), serotype hu56 (AAVhu56), serotype hu53 (AAVhu53), serotype rh64R1 (AAVrh64R1), serotype rh46 (AAVrh46), and serotype rh73 (AAVrh73) (see
In specific embodiments, the peptide is inserted after 138; 262-272; 450-459; or 585-593 of AAV1 capsid (SEQ ID NO:63); 138; 262-272; 449-458; or 584-592 of AAV2 capsid (SEQ ID NO:64); 138; 262-272; 449-459; or 585-593 of AAV3 capsid (SEQ ID NO:65); 137; 256-262; 443-453; or 583-591 of AAV4 capsid (SEQ ID NO:68); 137; 252-262; 442-445; or 574-582 of AAV5 capsid (SEQ ID NO:70); 138; 262-272; 450-459; 585-593 of AAV6 capsid (SEQ ID NO:71); 138; 263-273; 451-461; 586-594 of AAV7 capsid (SEQ ID NO:72); 138; 263-274; 452-461; 587-595 of AAV8 capsid (SEQ ID NO:73); 138; 262-273; 452-461; 585-593 of AAV9 capsid (SEQ ID NO:74); 138; 262-273; 452-461; 585-593 of AAV9e capsid (SEQ ID NO:75); 138; 263-274; 452-461; 587-595 of AAVrh10 capsid (SEQ ID NO:76); 138; 263-274; 452-461; 587-595 of AAVrh20 capsid (SEQ ID NO:77); 138; 263-274; 452-461; 587-595 of AAVrh74 capsid (SEQ ID NO:80 or SEQ ID NO:81), 138; 263-274; 452-461; 587-595 of AAVhu37 capsid (SEQ ID NO:85); or 138; 263-274; 452-461; 587-595 of AAVrh39 capsid (SEQ ID NO:78) (as numbered in
Generally, the peptide insertion is sequence of contiguous amino acids from a heterologous protein or domain thereof. The peptide to be inserted typically is long enough to retain a particular biological function, characteristic, or feature of the protein or domain from which it is derived. The peptide to be inserted typically is short enough to allow the capsid protein to form a coat, similarly or substantially similarly to the native capsid protein without the insertion. In preferred embodiments, the peptide insertion is from about 4 to about 30 amino acid residues in length, about 4 to about 20, about 4 to about 15, about 5 to about 10, or about 7 amino acids in length. The peptide sequences for insertion are at least 4 amino acids in length and may be 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, or 15 amino acids in length. In some embodiments, the peptide sequences are 16, 17, 18, 19, or 20 amino acids in length. In embodiments, the peptide is no more than 7 amino acids, 10 amino acids or 12 amino acids in length.
A “peptide insertion from a heterologous protein” in an AAV capsid protein refers to an amino acid sequence that has been introduced into the capsid protein and that is not native to any AAV serotype capsid. Non-limiting examples include a peptide of a human protein in an AAV capsid protein.
In some embodiments, the rAAVs described herein increase tissue-specific (such as, but not limited to, CNS or skeletal and/or cardiac muscle) cell transduction in a subject (a human, non-human-primate, or mouse subject) or in cell culture, compared to the rAAV not comprising the amino acid substitution. In some embodiments, the increase in tissue specific cell transduction is at least 2, 10, 20, 30, 40, 50, 60, 70, 80, 90, or 100 fold more than that without the peptide insertion. For example, in some embodiments, there is a 50-80 fold increase in tissue specific cell transduction compared to transduction with the same AAV type without the modification. The increase in transduction may be assessed using methods described in the Examples herein and known in the art.
In some embodiments, the rAAVs described herein increase the incorporation of rAAV genomes into a cell or tissue type, particularly CNS or heart and/or skeletal muscle in a subject (a human, non-human primate or mouse subject) or in cell culture to the rAAV not comprising the peptide insertion. In some embodiments, the increase in genome integration is at least 2, 10, 20, 30, 40, 50, 60, 70, 80, 90, or 100 fold more than an AAV having a capsid without the peptide insertion. For example, in some embodiments, there is a 50-80 fold increase in genome integration compared to genome integration with the same AAV type without a peptide insert.
In another aspect, the polypeptide insertion is an antigen binding domain, for example, an scFv, scFv-Fc, single domain antibody, minibody, diabody or other single chain form of an antigen binding domain. Alternatively, the polypeptide insertion is a DARPin as a single or multiple domain binding protein. These targeting or binding domains may be directed to tissue specific cell surface markers, for example, markers, such as cell surface proteins, specific for CNS tissue, muscle tissue, cardiac tissue, peripheral nervous system tissue, etc.
In another aspect, provided are libraries of capsids, including heterologous peptide insertion libraries or libraries of capsids having one or more amino acid substitutions. A heterologous peptide insertion library refers to a collection of rAAV vectors that carry the same peptide insertion at different insertion sites in the virus capsid, e.g., at different positions within a given variable region of the capsid or different variant peptides or even one or more amino acid substitutions. Provided are methods of screening the rAAVs having capsids from the library for enhance of improved properties such as tissue tropism, including enhanced transduction in CNS or cardiac and/or skeletal muscle tissue and, including, reduced transduction in liver and/or DRG cells. Generally, the capsid proteins used comprise AAV genomes that contain modified rep and cap sequences to prevent the replication of the virus under conditions in which it could normally replicate (co-infection of a mammalian cell along with a helper virus such as adenovirus). The members of the peptide insertion libraries may then be assayed for functional display of the peptide on the rAAV surface, tissue targeting and/or gene transduction.
The follow summarizes insertion sites for the peptides or polypeptides described herein immediately after amino acid residues of AAV capsids as set forth below (see also, FIG. 7):
In embodiments, the peptide insertion occurs between amino acid residues 588-589 of the AAV9 capsid, or between corresponding residues of another AAV type capsid as determined by an amino acid sequence alignment (for example, as in
In some embodiments, one or more peptide insertions can be used in a single system. In some embodiments, the capsid is chosen and/or further modified to reduce recognition of the AAV particles by the subject's immune system, such as avoiding pre-existing antibodies in the subject. In some embodiments. In some embodiments, the capsid is chosen and/or further modified to enhance desired tropism/targeting.
Also provided are AAV vectors comprising the engineered capsids. In some embodiments, the AAV vectors are non-replicating and do not include the nucleotide sequences encoding the rep or cap proteins (these are supplied by the packaging cells in the manufacture of the rAAV vectors). In some embodiments, AAV-based vectors comprise components from one or more serotypes of AAV. In some embodiments, AAV based vectors provided herein comprise capsid components from one or more of AAV1, AAV2, AAV3, AAV4, AAV5, AAV6, AAV7, AAV8, AAV9, AAV10, AAV11, AAV12, AAV13, AAV14, AAV15, AAV16, AAV.rh8, AAV.rh10, AAV.rh20, AAV.rh39, AAV.Rh74, AAV.RHM4-1, AAV.hu37, AAV.Anc80, AAV.Anc80L65, AAV.7m8, AAV.PHP.B, AAV.PHP.eB, AAV2.5, AAV2tYF, AAV3B, AAV.LK03, AAV.HSC1, AAV.HSC2, AAV.HSC3, AAV.HSC4, AAV.HSC5, AAV.HSC6, AAV.HSC7, AAV.HSC8, AAV.HSC9, AAV.HSC10, AAV.HSC11, AAV.HSC12, AAV.HSC13, AAV.HSC14, AAV.HSC15, AAV.HSC16, AAVrh34, AAVhu26, AAVrh31, AAVhu56, AAVhu53, AAVrh64R1, AAVrh46, and AAVrh73, or other rAAV particles, or combinations of two or more thereof. In some embodiments, AAV based vectors provided herein comprise components from one or more of AAV1, AAV2, AAV3, AAV4, AAV5, AAV6, AAV7, AAV8, AAV9, AAV10, AAV11, AAV12, AAV13, AAV14, AAV15, AAV16, AAV.rh8, AAV.rh10, AAV.rh20, AAV.rh39, AAV.Rh74, AAV.RHM4-1, AAV.hu37, AAV.Anc80, AAV.Anc80L65, AAV.7m8, AAV.PHP.B, AAV.PHP.eB, AAV2.5, AAV2tYF, AAV3B, AAV.LK03, AAV.HSC1, AAV.HSC2, AAV.HSC3, AAV.HSC4, AAV.HSC5, AAV.HSC6, AAV.HSC7, AAV.HSC8, AAV.HSC9, AAV.HSC10, AAV.HSC11, AAV.HSC12, AAV.HSC13, AAV.HSC14, AAV.HSC15, or AAV.HSC16, AAVrh34, AAVhu26, AAVrh31, AAVhu56, AAVhu53, AAVrh64R1, AAVrh46, and AAVrh73, or other rAAV particles, or combinations of two or more thereof serotypes. In some embodiments, rAAV particles comprise a capsid protein at least 80% or more identical, e.g., 85%, 85%, 87%, 88%, 89%, 90% 91% 92%, 93%, 94%, 95%, 96%, 97%, 98% 99%, 99.5%, etc., i.e. up to 100% identical, to e.g., VP1, VP2 and/or VP3 sequence of an AAV capsid serotype selected from AAV1, AAV2, AAV3, AAV4, AAV5, AAV6, AAV7, AAV8, AAV9, AAV10, AAV11, AAV12, AAV13, AAV14, AAV15, AAV16, AAV.rh8, AAV.rh10, AAV.rh20, AAV.rh39, AAV.Rh74, AAV.RHM4-1, AAV.hu37, AAV.Anc80, rAAV.Anc80L65, AAV.7m8, AAV.PHP.B, AAV.PHP.eB, AAV2.5, AAV2tYF, AAV3B, AAV.LK03, AAV.HSC1, AAV.HSC2, AAV.HSC3, AAV.HSC4, AAV.HSC5, AAV.HSC6, AAV.HSC7, AAV.HSC8, AAV.HSC9, AAV.HSC10, AAV.HSC11, AAV.HSC12, AAV.HSC13, AAV.HSC14, AAV.HSC15, or AAV.HSC16, AAVrh34, AAVhu26, AAVrh31, AAVhu56, AAVhu53, AAVrh64R1, AAVrh46, and AAVrh73, or a derivative, modification, or pseudotype thereof. These engineered AAV vectors may comprise a genome comprising a transgene encoding a therapeutic protein or nucleic acid.
In particular embodiments, the recombinant AAV for use in compositions and methods herein is Anc80 or Anc80L65 (see, e.g., Zinn et al., 2015, Cell Rep. 12(6): 1056-1068, which is incorporated by reference in its entirety). In particular embodiments, the recombinant AAV for use in compositions and methods herein is AAV.7m8 (including variants thereof) (see, e.g., U.S. Pat. Nos. 9,193,956; 9,458,517; 9,587,282; US 2016/0376323, and WO 2018/075798, each of which is incorporated herein by reference in its entirety). In particular embodiments, the AAV for use in compositions and methods herein is any AAV disclosed in U.S. Pat. No. 9,585,971, such as AAV-PHP.B. In particular embodiments, the AAV for use in compositions and methods herein is an AAV2/Rec2 or AAV2/Rec3 vector, which has hybrid capsid sequences derived from AAV8 and serotypes cy5, rh20 or rh39 (see, e.g., Issa et al., 2013, PLoS One 8(4): e60361, which is incorporated by reference herein for these vectors). In particular embodiments, the AAV for use in compositions and methods herein is an AAV disclosed in any of the following, each of which is incorporated herein by reference in its entirety: U.S. Pat. Nos. 7,282,199; 7,906,111; 8,524,446; 8,999,678; 8,628,966; 8,927,514; 8,734,809; 9,284,357; 9,409,953; 9,169,299; 9,193,956; 9,458,517; 9,587,282; US 2015/0374803; US 2015/0126588; US 2017/0067908; US 2013/0224836; US 2016/0215024; US 2017/0051257; PCT/US2015/034799; and PCT/EP2015/053335. In some embodiments, rAAV particles have a capsid protein at least 80% or more identical, e.g., 85%, 85%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.5%, etc., i.e. up to 100% identical, to the VP1, VP2 and/or VP3 sequence of an AAV capsid disclosed in any of the following patents and patent applications, each of which is incorporated herein by reference in its entirety: U.S. Pat. Nos. 7,282,199; 7,906,111; 8,524,446; 8,999,678; 8,628,966; 8,927,514; 8,734,809; 9,284,357; 9,409,953; 9,169,299; 9,193,956; 9,458,517; and 9,587,282; US patent application publication nos. 2015/0374803; 2015/0126588; 2017/0067908; 2013/0224836; 2016/0215024; 2017/0051257; and International Patent Application Nos. PCT/US2015/034799; PCT/EP2015/053335.
In some embodiments, rAAV particles comprise any AAV capsid disclosed in U.S. Pat. No. 9,840,719 and WO 2015/013313, such as AAV.Rh74 and RHM4-1, each of which is incorporated herein by reference in its entirety. In some embodiments, rAAV particles comprise any AAV capsid disclosed in WO 2014/172669, such as AAV rh.74, which is incorporated herein by reference in its entirety. In some embodiments, rAAV particles comprise the capsid of AAV2/5, as described in Georgiadis et al., 2016, Gene Therapy 23: 857-862 and Georgiadis et al., 2018, Gene Therapy 25: 450, each of which is incorporated by reference in its entirety. In some embodiments, rAAV particles comprise any AAV capsid disclosed in WO 2017/070491, such as AAV2tYF, which is incorporated herein by reference in its entirety. In some embodiments, rAAV particles comprise the capsids of AAVLK03 or AAV3B, as described in Puzzo et al., 2017, Sci. Transl. Med. 29(9): 418, which is incorporated by reference in its entirety. In some embodiments, rAAV particles comprise any AAV capsid disclosed in U.S. Pat. Nos. 8,628,966; 8,927,514; 9,923,120 and WO 2016/049230, such as HSC1, HSC2, HSC3, HSC4, HSC5, HSC6, HSC7, HSC8, HSC9, HSC10, HSC11, HSC12, HSC13, HSC14, HSC15, or HSC16, each of which is incorporated by reference in its entirety.
In some embodiments, rAAV particles have a capsid protein disclosed in Intl. Appl. Publ. No. WO 2003/052051 (see, e.g., SEQ ID NO:2 of '051 publication), WO 2005/033321 (see, e.g., SEQ ID NOs: 123 and 88 of '321 publication), WO 03/042397 (see, e.g., SEQ ID NOs: 2, 81, 85, and 97 of '397 publication), WO 2006/068888 (see, e.g., SEQ ID NOs: 1 and 3-6 of '888 publication), WO 2006/110689, (see, e.g., SEQ ID NOs: 5-38 of '689 publication) WO2009/104964 (see, e.g., SEQ ID NOs: 1-5, 7, 9, 20, 22, 24 and 31 of '964 publication), WO 2010/127097 (see, e.g., SEQ ID NOs: 5-38 of '097 publication), and WO 2015/191508 (see, e.g., SEQ ID NOs: 80-294 of '508 publication), and U.S. Appl. Publ. No. 20150023924 (see, e.g., SEQ ID NOs: 1, 5-10 of '924 publication), the contents of each of which is herein incorporated by reference in its entirety. In some embodiments, rAAV particles have a capsid protein at least 80% or more identical, e.g., 85%, 85%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.5%, etc., i.e. up to 100% identical, to the VP1, VP2 and/or VP3 sequence of an AAV capsid disclosed in Intl. Appl. Publ. No. WO 2003/052051 (see, e.g., SEQ ID NO: 2 of '051 publication), WO 2005/033321 (see, e.g., SEQ ID NOs: 123 and 88 of '321 publication), WO 03/042397 (see, e.g., SEQ ID NOs: 2, 81, 85, and 97 of '397 publication), WO 2006/068888 (see, e.g., SEQ ID NOs: 1 and 3-6 of '888 publication), WO 2006/110689 (see, e.g., SEQ ID NOs: 5-38 of '689 publication) WO2009/104964 (see, e.g., SEQ ID NOs: 1-5, 7, 9, 20, 22, 24 and 31 of 964 publication), WO 2010/127097 (see, e.g., SEQ ID NOs: 5-38 of '097 publication), and WO 2015/191508 (see, e.g., SEQ ID NOs: 80-294 of '508 publication), and U.S. Appl. Publ. No. 20150023924 (see, e.g., SEQ ID NOs: 1, 5-10 of '924 publication).
In additional embodiments, rAAV particles comprise a pseudotyped AAV capsid. In some embodiments, the pseudotyped AAV capsids are rAAV2/8 or rAAV2/9 pseudotyped AAV capsids. Methods for producing and using pseudotyped rAAV particles are known in the art (see, e.g., Duan et al., J. Virol., 75:7662-7671 (2001); Halbert et al., J. Virol., 74:1524-1532 (2000); Zolotukhin et al., Methods 28:158-167 (2002); and Auricchio et al., Hum. Molec. Genet. 10:3075-3081, (2001).
In certain embodiments, a single-stranded AAV (ssAAV) may be used. In certain embodiments, a self-complementary vector, e.g., scAAV, may be used (see, e.g., Wu, 2007, Human Gene Therapy, 18(2):171-82; McCarty et al, 2001, Gene Therapy, 8(16):1248-1254; U.S. Pat. Nos. 6,596,535; 7,125,717; and 7,456,683, each of which is incorporated herein by reference in its entirety).
Another aspect of the present invention involves making rAAV particles having the capsids disclosed herein. In some embodiments, an rAAV particle is made by providing a nucleotide comprising the nucleic acid sequence encoding any of the capsid proteins described herein; and using a packaging cell system to prepare corresponding rAAV particles with capsid coats made up of the capsid protein. In some embodiments, the nucleic acid sequence encodes a sequence having at least 60% 70% 80% 85% 90% 91% 92% 93% 94% 95% 96% 97%, 98%, 99% or 99.9%, identity to the sequence of a capsid protein molecule described herein, and retains (or substantially retains) biological function of the capsid protein. In some embodiments, the nucleic acid encodes a sequence having at least 60%, 70%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 99.9%, identity to the sequence of the one of the capsid proteins described herein, for example, those with sequences in Table 17 or otherwise described herein (see also
The capsid protein, coat, and rAAV particles may be produced by techniques known in the art. In some embodiments, the viral genome comprises at least one inverted terminal repeat to allow packaging into a vector. In some embodiments, the viral genome further comprises a cap gene and/or a rep gene for expression and splicing of the cap gene. In other embodiments, the cap and rep genes are provided by a packaging cell and not present in the viral genome.
In some embodiments, the nucleic acid encoding the engineered capsid protein is cloned into an AAV Rep-Cap helper plasmid in place of the existing capsid gene. When introduced together into host cells, this plasmid helps package an rAAV genome into the engineered capsid protein as the capsid coat. Packaging cells can be any cell type possessing the genes necessary to promote AAV genome replication, capsid assembly, and packaging. Nonlimiting examples include 293 cells or derivatives thereof, HELA cells, or insect cells.
Accordingly, provided are nucleic acids encoding the modified capsids described herein, including plasmid vectors, specifically AAV Rep-Cap helper plasmids which comprise the nucleotide sequence encoding the modified capsid described herein (including the modified capsids AAV9.G266A.496NNN/AAA498 (SEQ ID NO:50), AAV9.N272A.496NNN/AAA498 (SEQ ID NO:49), AAV9.496NNN/AAA498.W503R (SEQ ID NO:32), and AAV9.496NNN/AAA498.W503A (SEQ ID NO:51) and these modified capsids further modified with a targeting domain insertion). Provided also are bacterial host cells comprising the AAV Rep-Cap helper plasmid and methods of amplifying and producing the AAV Rep-Cap helper plasmid comprising the nucleotide sequence encoding the modified capsid. Further provided are packaging cells (including insect or mammalian cells) which comprise a nucleotide sequence encoding the modified capsid and which packaging cells produce rAAV vectors having a modified capsid as described herein.
Standard techniques can be used for recombinant DNA, oligonucleotide synthesis, and tissue culture and transformation (e.g., electroporation, lipofection). Enzymatic reactions and purification techniques can be performed according to manufacturer's specifications or as commonly accomplished in the art or as described herein. The foregoing techniques and procedures can be generally performed according to conventional methods well known in the art and as described in various general and more specific references that are cited and discussed throughout the present specification. See, e.g., Sambrook et al., Molecular Cloning: A Laboratory Manual (2d ed., Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y. (1989)), which is incorporated herein by reference for any purpose. Unless specific definitions are provided, the nomenclatures utilized in connection with, and the laboratory procedures and techniques of, analytical chemistry, synthetic organic chemistry, and medicinal and pharmaceutical chemistry described herein are those well-known and commonly used in the art. Standard techniques can be used for chemical syntheses, chemical analyses, pharmaceutical preparation, formulation, and delivery, and treatment of patients. Nucleic acid sequences of AAV-based viral vectors, and methods of making recombinant AAV and AAV capsids, are taught, e.g., in U.S. Pat. Nos. 7,282,199; 7,790,449; 8,318,480; 8,962,332; and PCT/EP2014/076466, each of which is incorporated herein by reference in its entirety.
In some embodiments, the rAAVs provide transgene delivery vectors that can be used in therapeutic and prophylactic applications, as discussed in more detail below. In some embodiments, the rAAV vector also includes regulatory control elements known to one skilled in the art to influence the expression of the RNA and/or protein products encoded by nucleic acids (transgenes) within target cells of the subject. Regulatory control elements and may be tissue-specific, that is, active (or substantially more active or significantly more active) only in the target cell/tissue. In specific embodiments, the AAV vector comprises a regulatory sequence, such as a promoter, operably linked to the transgene that allows for expression in target tissues. The promoter may be a constitutive promoter, for example, the CB7 promoter. Additional promoters include: cytomegalovirus (CMV) promoter, Rous sarcoma virus (RSV) promoter, MMT promoter, EF-1 alpha promoter, UB6 promoter, chicken beta-actin promoter, CAG promoter, RPE65 promoter, opsin promoter, the TBG (Thyroxine-binding Globulin) promoter, the APOA2 promoter, SERPINAI (hAAT) promoter, or MIR122 promoter. In some embodiments, particularly where it may be desirable to turn off transgene expression, an inducible promoter is used, e.g., hypoxia-inducible or rapamycin-inducible promoter.
Provided in particular embodiments are AAV vectors comprising a viral genome comprising an expression cassette for expression of the transgene, under the control of regulatory elements, and flanked by ITRs and an engineered viral capsid as described herein or is at least 95%, 96%, 97%, 98%, 99% or 99.9% identical to the amino acid sequence of the a capsid protein described herein (see Table 17, e.g.), while retaining the biological function of the engineered capsid. In certain embodiments, the encoded engineered capsid has the sequence of an AAV8.BBB.LD capsid (SEQ ID NO:27), an AAV9.BBB.LD capsid (SEQ ID NO:29), an AAV9.496-NNN/AAA-498 capsid (SEQ ID NO:31), AAV9.496-NNN/AAA-498.503R capsid (SEQ ID NO:32), AAV9.W503R capsid (SEQ ID NO:33), AAV9.Q474A capsid (SEQ ID NO:34), AAV9.N272A.496-NNN/AAA-498 capsid (SEQ ID NO:49) or AAV9.G266A.496-NNN/AAA-498 capsid (SEQ ID NO:50), and AAV9.496NNN/AAA498.W503A (SEQ ID NO:51). Also provided are engineered AAV vectors other than AAV9 vectors, such as engineered AAV1, AAV2, AAV3, AAV4, AAV5, AAV6, AAV7, AAV8, AAV9, AAV9e, AAVrh10, AAVrh20, AAVhu.37, AAVrh39, AAVrh74, AAVrh34, AAVhu26, AAVrh3l, AAVhu56, AAVhu53, AAVrh.46, AAVrh.64.R1, AAV.rh.73 vectors, including with the amino acid substitutions and/or peptide insert as described herein and 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, or 30 amino acid substitutions relative to the wild type or unengineered sequence for that AAV type and that retains its biological function.
The recombinant adenovirus can be a first-generation vector, with an E1 deletion, with or without an E3 deletion, and with the expression cassette inserted into either deleted region. The recombinant adenovirus can be a second-generation vector, which contains full or partial deletions of the E2 and E4 regions. A helper-dependent adenovirus retains only the adenovirus inverted terminal repeats and the packaging signal (phi). The transgene generally is inserted between the packaging signal and the 3′ITR, with or without stuffer sequences to keep the genome close to wild-type size of approximately 36 kb. An exemplary protocol for production of adenoviral vectors may be found in Alba et al., 2005, “Gutless adenovirus: last generation adenovirus for gene therapy,” Gene Therapy 12:S18-S27, which is incorporated by reference herein in its entirety
The rAAV vector for delivering the transgene to target tissues, cells, or organs, has a tropism for that particular target tissue, cell, or organ. Tissue-specific promoters may also be used. The construct comprising the transgene, within AAV ITR sequences further can include expression control elements that enhance expression of the transgene driven by the vector (e.g., introns such as the chicken β-actin intron, minute virus of mice (MVM) intron, human factor IX intron (e.g., FIX truncated intron 1), β-globin splice donor/immunoglobulin heavy chain spice acceptor intron, adenovirus splice donor/immunoglobulin splice acceptor intron, SV40 late splice donor/splice acceptor (19S/16S) intron, and hybrid adenovirus splice donor/IgG splice acceptor intron and polyA signals such as the rabbit β-globin polyA signal, human growth hormone (hGH) polyA signal, SV40 late polyA signal, synthetic polyA (SPA) signal, and bovine growth hormone (bGH) polyA signal. See, e.g., Powell and Rivera-Soto, 2015, Discov. Med., 19(102):49-57.
In certain embodiments, nucleic acids sequences disclosed herein may be codon-optimized, for example, via any codon-optimization technique known to one of skill in the art (see, e.g., review by Quax et al., 2015, Mol Cell 59:149-161).
In a specific embodiment, the recombinant AAVs described herein comprise an artificial genome comprising the following components: (1) AAV2 inverted terminal repeats that flank the expression cassette; (2) control elements, which include a) a promoter and, optionally, enhancer elements to promote expression of the transgene in CNS and/or muscle cells, b) optionally an intron sequence, such as a chicken β-actin intron, and c) a polyadenylation sequence, such as an SV40 polyA or rabbit β-globin poly A signal; and (3) transgene providing (e.g., coding for) a nucleic acid or protein product of interest, including a therapeutic nucleic acid or protein.
The viral vectors provided herein may be manufactured using host cells, e.g., mammalian host cells, including host cells from humans, monkeys, mice, rats, rabbits, or hamsters. Nonlimiting examples include: A549, WEHI, 10T1/2, BHK, MDCK, COS1, COS7, BSC 1, BSC 40, BMT 10, VERO, W138, HeLa, 293, Saos, C2C12, L, HT1080, HepG2, primary fibroblast, hepatocyte, and myoblast cells. Typically, the host cells are stably transformed with the sequences encoding the transgene and associated elements (i.e., the vector genome), and genetic components for producing viruses in the host cells, such as the replication and capsid genes (e.g., the rep and cap genes of AAV). For a method of producing recombinant AAV vectors with AAV8 capsids, see Section IV of the Detailed Description of U.S. Pat. No. 7,282,199 B2, which is incorporated herein by reference in its entirety. Genome copy titers of said vectors may be determined, for example, by TAQMAN® analysis. Virions may be recovered, for example, by CsCl2 sedimentation. Alternatively, baculovirus expression systems in insect cells may be used to produce AAV vectors. For a review, see Aponte-Ubillus et al., 2018, App. Microbiol. Biotechnol. 102:1045-1054, which is incorporated by reference herein in its entirety for manufacturing techniques.
In vitro assays, e.g., cell culture assays, can be used to measure transgene expression from a vector described herein, thus indicating, e.g., potency of the vector. For example, the PER.C6® cell Line (Lonza), a cell line derived from human embryonic retinal cells, or retinal pigment epithelial cells, e.g., the retinal pigment epithelial cell line hTERT RPE-1 (available from ATCC®), can be used to assess transgene expression. Alternatively, cell lines derived from liver or other cell types may be used, for example, but not limited, to HuH-7, HEK293, fibrosarcoma HT-1080, HKB-11, and CAP cells. Once expressed, characteristics of the expressed product (i.e., transgene product) can be determined, including determination of the glycosylation and tyrosine sulfation patterns, using assays known in the art.
Another aspect relates to therapies which involve administering a transgene via a rAAV vector according to the invention to a subject in need thereof, for delaying, preventing, treating, and/or managing a disease or disorder, and/or ameliorating one or more symptoms associated therewith. A subject in need thereof includes a subject suffering from the disease or disorder, or a subject pre-disposed thereto, e.g., a subject at risk of developing or having a recurrence of the disease or disorder. Generally, a rAAV carrying a particular transgene will find use with respect to a given disease or disorder in a subject where the subject's native gene, corresponding to the transgene, is defective in providing the correct gene product, or correct amounts of the gene product. The transgene then can provide a copy of a gene that is defective in the subject.
Generally, the transgene comprises cDNA that restores protein function to a subject having a genetic mutation(s) in the corresponding native gene. In some embodiments, the cDNA comprises associated RNA for performing genomic engineering, such as genome editing via homologous recombination. In some embodiments, the transgene encodes a therapeutic RNA, such as a shRNA, artificial miRNA, or element that influences splicing.
Tables 1A-1B below provides a list of transgenes that may be used in any of the rAAV vectors described herein, in particular, in the novel insertion sites described herein, to treat or prevent the disease with which the transgene is associated, also listed in Tables 1A-1B. As described herein, the AAV vector may be engineered as described herein to target the appropriate tissue for delivery of the transgene to effect the therapeutic or prophylactic use. The appropriate AAV serotype may be chosen to engineer to optimize the tissue tropism and transduction of the vector.
For example, a rAAV vector comprising a transgene encoding glial derived growth factor (GDGF) finds use treating/preventing/managing Parkinson's disease. Generally, the rAAV vector is administered systemically. For example, the rAAV vector may be provided by intravenous, intrathecal, intra-nasal, and/or intra-peritoneal administration.
In certain embodiments, the transgene encodes a microdystrophin (for example, as disclosed in WO WO2021/108755, WO2002/029056, WO2016/115543, WO2015/197232, WO2016/177911, U.S. Pat. No. 7,892,824B2, U.S. Pat. No. 9,624,282B2, and WO2017221145, which are hereby incorporated by reference in their entirety) and is useful for treatment of dystrophinopathies, such as muscular dystrophy. Example 18 herein shows the relative abundance of capsids AAV7, AAV8, AAV9, AAVrh. 10, AAVrh.46, AAVrh.64.R1, and AAVrh.73 after intravenous administration to wild-type mice compared to mdx mice (animal model for muscular dystrophy). rAAV particles having these capsids, or an engineered forms thereof, may be useful for delivery of transgenes encoding microdystrophins or other dystrophinopathy therapeutic proteins to muscle cells, including skeletal and/or cardiac muscle, while having reduced delivery to liver cells, for treatment of muscular dystrophies, such as, Duchenne Muscular Dystrophy.
In particular aspects, the rAAVs of the present invention find use in delivery to target tissues, or target cell types, including cell matrix associated with the target cell types, associated with the disorder or disease to be treated/prevented. A disease or disorder associated with a particular tissue or cell type is one that largely affects the particular tissue or cell type, in comparison to other tissue of cell types of the body, or one where the effects or symptoms of the disorder appear in the particular tissue or cell type. Methods of delivering a transgene to a target tissue of a subject in need thereof involve administering to the subject an rAAV where the peptide insertion is a homing peptide. In the case of Parkinson's, for example, a rAAV vector comprising a peptide insertion that directs the rAAV to neural tissue can be used, in particular, where the peptide insertion facilitates the rAAV in crossing the blood brain barrier to the CNS.
For a disease or disorder associated with neural tissue, an rAAV vector can be used that comprises a peptide insertion from a neural tissue-homing domain, such as any described herein. Diseases/disorders associated with neural tissue include Alzheimer's disease, amyotrophic lateral sclerosis (ALS), amyotrophic lateral sclerosis (ALS), Battens disease, Batten's Juvenile NCL form, Canavan disease, chronic pain, Friedreich's ataxia, glioblastoma multiforme, Huntington's disease, Late Infantile neuronal ceroid lipofuscinosis (LINCL), lysosomal storage disorders, Leber's congenital amaurosis, multiple sclerosis, Parkinson's disease, Pompe disease, Rett syndrome, spinal cord injury, spinal muscular atrophy (SMA), stroke, and traumatic brain injury. The vector further can contain a transgene for therapeutic/prophylactic benefit to a subject suffering from, or at risk of developing, the disease or disorder (see Tables 1A-1B).
The rAAV vectors of the invention also can facilitate delivery, in particular, targeted delivery, of oligonucleotides, drugs, imaging agents, inorganic nanoparticles, liposomes, antibodies to target cells or tissues. The rAAV vectors also can facilitate delivery, in particular, targeted delivery, of non-coding DNA, RNA, or oligonucleotides to target tissues.
The agents may be provided as pharmaceutically acceptable compositions as known in the art and/or as described herein. Also, the rAAV molecule of the invention may be administered alone or in combination with other prophylactic and/or therapeutic agents.
The dosage amounts and frequencies of administration provided herein are encompassed by the terms therapeutically effective and prophylactically effective. The dosage and frequency will typically vary according to factors specific for each patient depending on the specific therapeutic or prophylactic agents administered, the severity and type of disease, the route of administration, as well as age, body weight, response, and the past medical history of the patient, and should be decided according to the judgment of the practitioner and each patient's circumstances. Suitable regimens can be selected by one skilled in the art by considering such factors and by following, for example, dosages reported in the literature and recommended in the Physician's Desk Reference (56th ed., 2002). Prophylactic and/or therapeutic agents can be administered repeatedly. Several aspects of the procedure may vary such as the temporal regimen of administering the prophylactic or therapeutic agents, and whether such agents are administered separately or as an admixture.
The amount of an agent of the invention that will be effective can be determined by standard clinical techniques. Effective doses may be extrapolated from dose-response curves derived from in vitro or animal model test systems. For any agent used in the method of the invention, the therapeutically effective dose can be estimated initially from cell culture assays. A dose may be formulated in animal models to achieve a circulating plasma concentration range that includes the IC50 (i.e., the concentration of the test compound that achieves a half-maximal inhibition of symptoms) as determined in cell culture. Such information can be used to more accurately determine useful doses in humans. Levels in plasma may be measured, for example, by high performance liquid chromatography.
Prophylactic and/or therapeutic agents, as well as combinations thereof, can be tested in suitable animal model systems prior to use in humans. Such animal model systems include, but are not limited to, rats, mice, chicken, cows, monkeys, pigs, dogs, rabbits, etc. Any animal system well-known in the art may be used. Such model systems are widely used and well known to the skilled artisan. In some embodiments, animal model systems for a CNS condition are used that are based on rats, mice, or other small mammal other than a primate.
Once the prophylactic and/or therapeutic agents of the invention have been tested in an animal model, they can be tested in clinical trials to establish their efficacy. Establishing clinical trials will be done in accordance with common methodologies known to one skilled in the art, and the optimal dosages and routes of administration as well as toxicity profiles of agents of the invention can be established. For example, a clinical trial can be designed to test a rAAV molecule of the invention for efficacy and toxicity in human patients.
Toxicity and efficacy of the prophylactic and/or therapeutic agents of the instant invention can be determined by standard pharmaceutical procedures in cell cultures or experimental animals, e.g., for determining the LD50 (the dose lethal to 50% of the population) and the ED50 (the dose therapeutically effective in 50% of the population). The dose ratio between toxic and therapeutic effects is the therapeutic index and it can be expressed as the ratio LD50/ED50. Prophylactic and/or therapeutic agents that exhibit large therapeutic indices are preferred. While prophylactic and/or therapeutic agents that exhibit toxic side effects may be used, care should be taken to design a delivery system that targets such agents to the site of affected tissue in order to minimize potential damage to uninfected cells and, thereby, reduce side effects.
A rAAV molecule of the invention generally will be administered for a time and in an amount effective for obtain a desired therapeutic and/or prophylactic benefit. The data obtained from the cell culture assays and animal studies can be used in formulating a range and/or schedule for dosage of the prophylactic and/or therapeutic agents for use in humans. The dosage of such agents lies within a range of circulating concentrations that include the ED50 with little or no toxicity. The dosage may vary within this range depending upon the dosage form employed and the route of administration utilized.
A therapeutically effective dosage of an rAAV vector for patients is generally from about 0.1 ml to about 100 ml of solution containing concentrations of from about 1×109 to about 1×1016 genomes rAAV vector, or about 1×1010 to about 1×1015, about 1×1012 to about 1×1016, or about 1×1014 to about 1×1016 AAV genomes. Levels of expression of the transgene can be monitored to determine/adjust dosage amounts, frequency, scheduling, and the like.
Treatment of a subject with a therapeutically or prophylactically effective amount of the agents of the invention can include a single treatment or can include a series of treatments. For example, pharmaceutical compositions comprising an agent of the invention may be administered once, or may be administered in a series of 2, 3 or 4 or more times, for example, weekly, monthly or every two months, 3 months, 6 months or one year until the series of doses has been administered.
The rAAV molecules of the invention may be administered alone or in combination with other prophylactic and/or therapeutic agents. Each prophylactic or therapeutic agent may be administered at the same time or sequentially in any order at different points in time; however, if not administered at the same time, they should be administered sufficiently close in time so as to provide the desired therapeutic or prophylactic effect. Each therapeutic agent can be administered separately, in any appropriate form and by any suitable route.
In various embodiments, the different prophylactic and/or therapeutic agents are administered less than 1 hour apart, at about 1 hour apart, at about 1 hour to about 2 hours apart, at about 2 hours to about 3 hours apart, at about 3 hours to about 4 hours apart, at about 4 hours to about 5 hours apart, at about 5 hours to about 6 hours apart, at about 6 hours to about 7 hours apart, at about 7 hours to about 8 hours apart, at about 8 hours to about 9 hours apart, at about 9 hours to about 10 hours apart, at about 10 hours to about 11 hours apart, at about 11 hours to about 12 hours apart, no more than 24 hours apart, or no more than 48 hours apart. In certain embodiments, two or more agents are administered within the same patient visit.
Methods of administering agents of the invention include, but are not limited to, parenteral administration (e.g., intradermal, intramuscular, intraperitoneal, intravenous, and subcutaneous, including infusion or bolus injection), epidural, and by absorption through epithelial or mucocutaneous or mucosal linings (e.g., intranasal, oral mucosa, rectal, and intestinal mucosa, etc.). In particular embodiments, such as where the transgene is intended to be expressed in the CNS, the vector is administered via lumbar puncture or via cisterna magna.
In certain embodiments, the agents of the invention are administered intravenously and may be administered together with other biologically active agents.
In another specific embodiment, agents of the invention may be delivered in a sustained release formulation, e.g., where the formulations provide extended release and thus extended half-life of the administered agent. Controlled release systems suitable for use include, without limitation, diffusion-controlled, solvent-controlled, and chemically-controlled systems. Diffusion controlled systems include, for example reservoir devices, in which the molecules of the invention are enclosed within a device such that release of the molecules is controlled by permeation through a diffusion barrier. Common reservoir devices include, for example, membranes, capsules, microcapsules, liposomes, and hollow fibers. Monolithic (matrix) device are a second type of diffusion controlled system, wherein the dual antigen-binding molecules are dispersed or dissolved in an rate-controlling matrix (e.g., a polymer matrix). Agents of the invention can be homogeneously dispersed throughout a rate-controlling matrix and the rate of release is controlled by diffusion through the matrix. Polymers suitable for use in the monolithic matrix device include naturally occurring polymers, synthetic polymers and synthetically modified natural polymers, as well as polymer derivatives.
Any technique known to one of skill in the art can be used to produce sustained release formulations comprising one or more agents described herein. See, e.g. U.S. Pat. No. 4,526,938; PCT publication WO 91/05548; PCT publication WO 96/20698; Ning et al., “Intratumoral Radioimmunotheraphy of a Human Colon Cancer Xenograft Using a Sustained-Release Gel,” Radiotherapy & Oncology, 39:179 189, 1996; Song et al., “Antibody Mediated Lung Targeting of Long-Circulating Emulsions,” PDA Journal of Pharmaceutical Science & Technology, 50:372 397, 1995; Cleek et al., “Biodegradable Polymeric Carriers for a bFGF Antibody for Cardiovascular Application,” Pro. Intl. Symp. Control. Rel. Bioact. Mater., 24:853 854, 1997; and Lam et al., “Microencapsulation of Recombinant Humanized Monoclonal Antibody for Local Delivery,” Proc. Int'l. Symp. Control Rel. Bioact. Mater., 24:759 760, 1997, each of which is incorporated herein by reference in its entirety. In one embodiment, a pump may be used in a controlled release system (see Langer, supra; Sefton, CRC Crit. Ref. Biomed. Eng., 14:20, 1987; Buchwald et al., Surgery, 88:507, 1980; and Saudek et al., N. Engl. J. Med., 321:574, 1989). In another embodiment, polymeric materials can be used to achieve controlled release of agents comprising dual antigen-binding molecule, or antigen-binding fragments thereof (see e.g., Medical Applications of Controlled Release, Langer and Wise (eds.), CRC Pres., Boca Raton, Fla. (1974); Controlled Drug Bioavailability, Drug Product Design and Performance, Smolen and Ball (eds.), Wiley, N.Y. (1984); Ranger and Peppas, J., Macromol. Sci. Rev. Macromol. Chem., 23:61, 1983; see also Levy et al., Science, 228:190, 1985; During et al., Ann. Neurol., 25:351, 1989; Howard et al., J. Neurosurg., 7 1:105, 1989); U.S. Pat. Nos. 5,679,377; 5,916,597; 5,912,015; 5,989,463; 5,128,326; PCT Publication No. WO 99/15154; and PCT Publication No. WO 99/20253). In yet another embodiment, a controlled release system can be placed in proximity of the therapeutic target (e.g., an affected joint), thus requiring only a fraction of the systemic dose (see, e.g., Goodson, in Medical Applications of Controlled Release, supra, vol. 2, pp. 115 138 (1984)). Other controlled release systems are discussed in the review by Langer, Science, 249:1527 1533, 1990.
In addition, rAAVs can be used for in vivo delivery of transgenes for scientific studies such as optogenetics, gene knock-down with miRNAs, recombinase delivery for conditional gene deletion, gene editing with CRISPRs, and the like.
The invention further provides a pharmaceutical composition comprising a pharmaceutically acceptable carrier and an agent of the invention, said agent comprising a rAAV molecule of the invention. In some embodiments, the pharmaceutical composition comprises rAAV combined with a pharmaceutically acceptable carrier for administration to a subject. In one embodiment, the term “pharmaceutically acceptable” means approved by a regulatory agency of the Federal or a state government or listed in the U.S. Pharmacopeia or other generally recognized pharmacopeia for use in animals, and more particularly in humans. The term “carrier” refers to a diluent, adjuvant (e.g., Freund's complete and incomplete adjuvant), excipient, or vehicle with which the agent is administered. Such pharmaceutical carriers can be sterile liquids, such as water and oils, including those of petroleum, animal, vegetable, or synthetic origin, including, e.g., peanut oil, soybean oil, mineral oil, sesame oil and the like. Water is a common carrier when the pharmaceutical composition is administered intravenously. Saline solutions and aqueous dextrose and glycerol solutions can also be employed as liquid carriers, particularly for injectable solutions. Suitable pharmaceutical excipients include starch, glucose, lactose, sucrose, gelatin, malt, rice, flour, chalk, silica gel, sodium stearate, glycerol monostearate, talc, sodium chloride, dried skim milk, glycerol, propylene, glycol, water, ethanol and the like. Additional examples of pharmaceutically acceptable carriers, excipients, and stabilizers include, but are not limited to, buffers such as phosphate, citrate, and other organic acids; antioxidants including ascorbic acid; low molecular weight polypeptides; proteins, such as serum albumin and gelatin; hydrophilic polymers such as polyvinylpyrrolidone; amino acids such as glycine, glutamine, asparagine, arginine or lysine; monosaccharides, disaccharides, and other carbohydrates including glucose, mannose, or dextrins; chelating agents such as EDTA; sugar alcohols such as mannitol or sorbitol; salt-forming counterions such as sodium; and/or nonionic surfactants such as TWEEN™, polyethylene glycol (PEG), and PLURONICS as known in the art. The pharmaceutical composition of the present invention can also include a lubricant, a wetting agent, a sweetener, a flavoring agent, an emulsifier, a suspending agent, and a preservative, in addition to the above ingredients. These compositions can take the form of solutions, suspensions, emulsion, tablets, pills, capsules, powders, sustained-release formulations and the like.
In certain embodiments of the invention, pharmaceutical compositions are provided for use in accordance with the methods of the invention, said pharmaceutical compositions comprising a therapeutically and/or prophylactically effective amount of an agent of the invention along with a pharmaceutically acceptable carrier.
In certain embodiments, the agent of the invention is substantially purified (i.e., substantially free from substances that limit its effect or produce undesired side-effects). In a specific embodiment, the host or subject is an animal, e.g., a mammal such as non-primate (e.g., cows, pigs, horses, cats, dogs, rats etc.) and a primate (e.g., monkey such as, a cynomolgus monkey and a human). In a certain embodiment, the host is a human.
The invention provides further kits that can be used in the above methods. In one embodiment, a kit comprises one or more agents of the invention, e.g., in one or more containers. In another embodiment, a kit further comprises one or more other prophylactic or therapeutic agents useful for the treatment of a condition, in one or more containers.
The invention also provides agents of the invention packaged in a hermetically sealed container such as an ampoule or sachette indicating the quantity of the agent or active agent. In one embodiment, the agent is supplied as a dry sterilized lyophilized powder or water free concentrate in a hermetically sealed container and can be reconstituted, e.g., with water or saline, to the appropriate concentration for administration to a subject. Typically, the agent is supplied as a dry sterile lyophilized powder in a hermetically sealed container at a unit dosage of at least 5 mg, more often at least 10 mg, at least 15 mg, at least 25 mg, at least 35 mg, at least 45 mg, at least 50 mg, or at least 75 mg. The lyophilized agent should be stored at between 2 and 8° C. in its original container and the agent should be administered within 12 hours, usually within 6 hours, within 5 hours, within 3 hours, or within 1 hour after being reconstituted. In an alternative embodiment, an agent of the invention is supplied in liquid form in a hermetically sealed container indicating the quantity and concentration of agent or active agent. Typically, the liquid form of the agent is supplied in a hermetically sealed container at least 1 mg/ml, at least 2.5 mg/ml, at least 5 mg/ml, at least 8 mg/ml, at least 10 mg/ml, at least 15 mg/kg, or at least 25 mg/ml.
The compositions of the invention include bulk drug compositions useful in the manufacture of pharmaceutical compositions (e.g., impure or non-sterile compositions) as well as pharmaceutical compositions (i.e., compositions that are suitable for administration to a subject or patient). Bulk drug compositions can be used in the preparation of unit dosage forms, e.g., comprising a prophylactically or therapeutically effective amount of an agent disclosed herein or a combination of those agents and a pharmaceutically acceptable carrier.
The invention further provides a pharmaceutical pack or kit comprising one or more containers filled with one or more of the agents of the invention. Additionally, one or more other prophylactic or therapeutic agents useful for the treatment of the target disease or disorder can also be included in the pharmaceutical pack or kit. The invention also provides a pharmaceutical pack or kit comprising one or more containers filled with one or more of the ingredients of the pharmaceutical compositions of the invention. Optionally associated with such container(s) can be a notice in the form prescribed by a governmental agency regulating the manufacture, use or sale of pharmaceuticals or biological products, which notice reflects approval by the agency of manufacture, use, or sale for human administration.
Generally, the ingredients of compositions of the invention are supplied either separately or mixed together in unit dosage form, for example, as a dry lyophilized powder or water-free concentrate in a hermetically sealed container such as an ampoule or sachette indicating the quantity of agent or active agent. Where the composition is to be administered by infusion, it can be dispensed with an infusion bottle containing sterile pharmaceutical grade water or saline. Where the composition is administered by injection, an ampoule of sterile water for injection or saline can be provided so that the ingredients may be mixed prior to administration.
The following examples report an analysis of surface-exposed loops on the AAV9 capsid to identify candidates for capsid engineering via insertional mutagenesis. The invention is illustrated by way of examples, describing the construction of rAAV9 capsids engineered to contain 7-mer peptides designed on the basis of the human axonemal dynein heavy chain tail. Briefly, three criteria were used for selecting surface loops that might be amenable to short peptide insertions: 1) minimal side chain interactions with adjacent loops; 2) variable sequence and structure between serotypes (lack of conserved sequences); and 3) the potential for interrupting commonly targeted neutralizing antibody epitopes. A panel of peptide insertion mutants was constructed and the individual mutants were screened for viable capsid assembly, peptide surface exposure, and potency. The top candidates were then used as templates for insertion of homing peptides to test if these peptide insertion points could be used to re-target rAAV vectors to tissues of interest. Further examples, demonstrate the increased transduction and tissue tropism for certain of the modified AAV capsids described herein. Also provided are examples demonstrating reduced transduction and tissue tropism for certain of the modified AAV capsids described herein.
Eight AAV9 mutants were constructed, to each include a heterologous peptide but at different insertion points in the VR-IV loop. The heterologous peptide was a FLAG tag that was inserted immediately following the following residues in vectors identified as pRGNX1090-1097, as shown in Table 2.
As seen, all candidates package with high efficiency.
Transduced cells were lysed and centrifuged. 500 μL of cell culture supernatant was loaded on 20 μL agarose-FLAG beads and eluted with SDS-PAGE loading buffer also loaded directly on the gel. For a negative control, 293-ssc supernatant was used that contained no FLAG inserts.
As seen, 1090 had the lowest titer of the candidate vectors, indicating the least protein pulled down. Very low titers also were seen with the positive control. It is likely that not a sufficient amount of positive control had been loaded for visualization on SDS-PAGE.
CHO-derived Lec2 cells were grown in αMEM and 10% FBS. The Lec2 cells were transduced at a MOI of about 2×108 GC vector (a MOI of about 10,000) and were treated with ViraDuctin reagent (similar results were observed on transducing Lec2 cells at a MOI of about 10,000 GC/cell but treated with 40 μg/mL zinc chloride (ZnCl2); results not shown). Lec2 cells are proline auxotrophs from CHO.
As seen, transduction efficiency in vitro is lower than that obtained using wild type AAV9 (9-luc). Nonetheless, previous studies have shown that introduction of a homing peptide can decrease in vitro gene transfer in non-target cells (such as 293, Lec2, or HeLa), while significantly increasing in vitro gene transfer in target cells (see, e.g., Nicklin et al. 2001; and Grifman et al. 2001).
AAV9 vectors having a capsid protein containing a homing peptide of the following peptide sequences (Table 3) at the S454 insertion site were studied. Suspension-adapted HEK293 cells were seeded at 1×106 cells/mL one day before transduction in 10 mL of media. Triple plasmid DNA transfections were done with PEIpro® (Polypus transfection) at a DNA:PEI ratio of 1:1.75. Cells were spun down and supernatant harvested five days post-transfection and stored at −80° C.
qPCR was performed on harvested supernatant of transfected suspension HEK293 cells five days post-transfection. Samples were subjected to DNase I treatment to remove residual plasmid or cellular DNA and then heat treated to inactivate DNase I and denature capsids. Samples were titered via qPCR using TaqMan Universal PCR Master Mix, No AmpEraseUNG (ThermoFisherScientific) and primer/probe against the polyA sequence packaged in the transgene construct. Standard curves were established using RGX-501 vector BDS.
Peptide insertions directly after S454 ranging from 5 to 10 amino acids in length produced AAV particles having adequate titer, whereas an upper size limit is possible, with significant packaging deficiencies observed for the peptide insertion having a length of 12 amino acids.
Cell lines were plated at 5-20×103 cells/well (depending on the cell line) in 96-well 24 hours before transduction. Cells were transduced with AAV9-GFP vectors (with or without insertions) at 1×1010 particles/well and analyzed via Cytation5 (BioTek) 48-96 hours after transduction, depending on the difference in expression rate in each cell line. Lec2 cells were cultured as in Example 5, blood-brain barrier hCMEC/D3 (EMD Millipore) cells were cultured according to manufacturer's protocol, HT-22 and HUH7 cells were cultured in DMEM and 10% FBS, and C2C12 myoblasts were plated in DMEM and 10% FBS and differentiated for three days pre-transfection in DMEM supplemented with 2% horse serum and 0.1% insulin. AAV9.S454.FLAG showed low transduction levels in every cell type tested.
Images show that homing peptides can alter the transduction properties of AAV9 in vitro when inserted after S454 in the AAV9 capsid protein, as compared to unmodified AAV9 capsid. P7 (TfR1 peptide, HAIYPRH (SEQ ID NO: 10)) for all cell lines show the highest rate of transduction followed by P9 (TfR3 peptide, RTIGPSV (SEQ ID NO:12)). P4 (Kidney1 peptide, LPVAS (SEQ ID NO:6)) showed a slightly higher rate of transduction than that of AAV9 wildtype for all cell types. Higher transduction rates were observed for P6 (Muscle1 peptide, ASSLNIA (SEQ ID NO:7)) in the brain endothelial hCMEC/D3 cell line and the C2C12 muscle cell line cultures as compared to the Lec2 and HT-22 cell line cultures. P1 vector was not included in images due to extremely low transduction efficiency, and P8 vector was not included due to low titer.
AAV.PHP.B is a capsid having a TLAVPFK (SEQ ID NO:20) insertion in AAV9 capsid, with no other amino acid modifications to the capsid sequence. AAV.PHP.eB is a capsid having a TLAVPFK (SEQ ID NO:20) insertion in AAV9 capsid, with two amino acid modifications of the capsid sequence upstream of the PHP.B insertion (see also Table 17). Table 4A summarizes the capsids utilized in the study.
Constructs of AAV9, AAV.PHPeB, AAV.hDyn, AAV.PHP.S and AAV.PHP.SH encoding GFP transgene were prepared and formulated in 1×PBS+0.001% Pluronic. Female C57BL/6 mice were randomized into treatment groups base on Day 1 bodyweight. Five groups of female C57BL/6 mice were each intravenously administered AAV9.GFP, AAV.PHPeB.GFP, AAV.hDyn.GFP, AAV.PHP.S.GFP or AAV.PHP.SH.GFP in accordance with Table 4B, below. The dosing volume was 10 mL/kg (0.200 mL/20 g mouse). The mice were 8-12 weeks of age at the start date. At day 15 post administration, the animals were euthanized, and peripheral tissues were collected, including brain tissue, liver, forelimb biceps, heart, kidney, lung, ovaries, and the sciatic nerve.
Quantitiative PCR (qPCR) was used to determine the number of vector genomes per μg of brain genomic DNA. Brain samples from injected mice were processed and genomic DNA was isolated using Blood and Tissue Genomic DNA kit from Qiagen. The qPCR assay was run on a QuantStudio 5 instrument (Life Technologies Inc) using primer-probe combination specific for eGFP following a standard curve method.
The AAV vector genome copies per μg of brain genomic DNA was at least a log higher in mice that were administered AAV.hDyn compared to all other AAV serotypes: AAV9, AAV.PHPeB, PHP.S, and PHP.SH (see
AAV capsid sequences were modified either by peptide insertions or guided mutagenesis and pooled to give a bar-coded library packaged with a GFP expression cassette. The modified vectors were then evaluated in an in vitro assay, as well as for in vivo bio-distribution in mice using next generation sequencing (NGS) and quantitative PCR. AAV.hDyn was identified as a high brain transduction vector from this pool and was further evaluated in individual delivery studies in mice to characterize its transduction profile. Additionally, immunohistochemistry analysis of brain sections was performed to understand the cellular tropism of this vector.
The ability of the modified capsids to cross the blood brain barrier was tested in an in vitro transwell assay using hCMEC/D3 BBB cells (SCC066, Millipore-Sigma) (see
Capsid modifications were performed on widely used AAV capsids including AAV8, AAV9, and AAVrh.10 by inserting various peptide sequences after the position S454 of the VR-IV (Tables 5a-5c) or after position Q588 of the VR-VIII surface exposed loop of the AAV capsid, as well as insertions after the initiation codon of VP2, which begins at amino acid 137 (AAV4, AAV4-4, and AAV5) or at amino acid 138 (AAV1, AAV2, AAV3, AAV3-3, AAV6, AAV7, AAV8, AAV9, AAV9e, rh. 10, rh.20, rh.39, rh.74, and hu.37) (
rAAVs with certain modified capsids were tested for transduction in vitro in Lec2 cells as described above in Example 5. Modified AAVs tested for transduction in Lec2 cells as follows: eB 588 Ad, eB 588 Hep, eB 588 p79, eB 588 Rab, AAV9 588 Ad, AAV9 588 Hep, AAV9 588 p79, AAV9 588 Rab, eB VP2 Ad, eB VP2 Hep, eB VP2 p79, eB VP2 Rab, AAV9 VP2 Ad, AAV9 VP2 Hep, AAV9 VP2 p79, AAV9 VP2 Rab as compared to AAV9. See Table 5B below for identity of AAV capsids.
To test biodistribution, modified AAVs were packaged with an eGFP transgene cassette containing specific barcodes corresponding to each individual capsid. Novel barcoded vectors were pooled and injected into mice in order to increase the efficiency of screening.
To analyse the bio-distribution of genetically altered AAV vectors, various vectors encoding GFP were prepared and formulated in 1×PBS+0.0001% Pluronic acid. All vectors were made with cis plasmids containing a ten (10) bp barcode to enable next-generation sequencing (NGS) library (pool) preparation. Three (3) vector pools (Study 1, Study 2 and Study 3 vectors) were injected intravenously into a cohort of 5 female C57Bl/6 mice in accordance with Tables 5A-C. The dosing volume was 10 mL/kg (0.2 mL/20 g mouse) for each.
The mice were randomized into treatment groups based on Day 1 bodyweight and their age at start date was 8-12 weeks. At day 15 post administration, the animals were euthanized and peripheral tissues were collected, including brain, kidney, liver, sciatic nerve, lung, heart, and muscle tissue. In the studies where selected capsids from the pool were injected individually, the same protocol was followed
Genomic DNA (gDNA) was isolated from tissue samples using DNeasy Blood and Tissue kit (69506) from Qiagen. Each vector's barcode region was amplified with primers containing overlaps for NGS and unique dual indexing (UDI) and multiplex sequencing strategies, as recommended by the manufacturer (Illumina). Illumina MiSeq using reagent nano and micro kits v2 (MS-103-1001/1002) were used to determine the relative abundance of each barcoded AAV vector per sample collected from the mice. Accordingly, each vector sample in Tables 5A-C below was barcoded as noted above to allow for each read to be identified and sorted before the final data analysis. The data was normalized based on the composition of AAVs in the originally injected pool and quantified using the total genome copy number obtained from qPCR analysis with a primer-probe combination specific to the barcoded sample.
In the studies where selected capsids from the pool were injected individually, qPCR was used to determine the number of vector genomes per μg of tissue genomic DNA. qPCR was done on a QuantStudio 5 (Life Technologies, Inc.) using primer-probe combination specific for eGFP following a standard curve method (
From the study where individual vectors were injected into mice for characterization, formalin fixed mouse brains were sectioned at 40 μm thickness on a vibrating blade microtome (VT1000S, Leica) and the floating sections were probed with antibodies against GFP to look at the cellular distribution of the delivered vectors.
More specifically, fixed brains from the mice injected with AAV.hDyn were sectioned using a Vibratome (Leica, VT-1000) and the GFP expression was evaluated using an anti-GFP antibody (AB3080, Millipore Sigma), Vectastain ABC kit (PK-6100, Vector Labs) and DAB Peroxidase kit (SK-4100, Vector Labs). Broad distribution of GFP expressing cells were present throughout the brain in mice injected with AAV.hDyn, including distribution in the cortex, striatum, and hippocampus of the brain.
Results are shown in
Data for the Lec2 cell transduction assay not shown. The AAV9 588 Hep (AAV9 with the peptide TILSRSTQTG (SEQ ID NO:15) 5 inserted after position 588) exhibited significantly greater transduction (4-fold) than wild type AAV9, and AAV9 VP2 Ad (AAV9 with the peptide SITLVKSTQTV (SEQ ID NO: 14) inserted after position 138), AAV9 VP2 Hep (AAV9 with the peptide TILSRSTQTG (SEQ ID NO: 15) inserted after position 138), and AAV9 VP2 Rab (AAV9 with the peptide RSSEEDKSTQTT (SEQ ID NO: 19) inserted after position 138) exhibited slightly greater transduction of the Lec2 cells relative to AAV9. The other AAVs assayed exhibited lower levels of transduction than AAV9.
AAV capsid modifications performed either by insertions in surface exposed loops of VR-IV and VR-VIII or by specific amino acid mutations did not affect their packaging efficiency and were able to produce similar titers in the production system described herein.
Intravenous administration of AAV.hDyn to mice resulted in higher relative abundance of the viral genome and greater brain cell transduction than other modified AAV vectors and AAV9 tested.
AAV capsid modifications performed by insertions of different homing peptides in surface exposed loop VR-IV did not affect their packaging efficiency and were able to produce similar titers in the production system described herein.
Intravenous administration of AAV9 S454 Kidney1 and AAV9 S454 Kidney1C to mice resulted in higher relative abundance of the viral genome and greater kidney cell transduction than other modified AAV9 vectors and the parental AAV9 vector tested. Intravenous administration of the AAV9 S454 Kidney1 or AAV9 S454 Muscle1 vector to mice resulted also in lower liver cell transduction.
The administration, in vivo and post-mortem observations, and biodistribution of a pool of recombinant AAVs having engineered capsids and a GFP transgene will be evaluated following a single intravenous, intracerebroventricular or intravitreal injection in cynomolgus monkeys (Table 7). The pool contains multiple capsids each of which contains a unique barcode identification allowing identification using next generation sequencing (NGS) analysis following administration to cynomolgus monkeys. The cynomolgus monkey is chosen as the test system because of its established usefulness and acceptance as a model for AAV biodistribution studies in a large animal species and for further translation to human. All animals on this study are naïve with respect to prior treatment. The pool may comprise at least the following recombinant AAVs having the engineered capsids listed in Table 7.
Nine female cynomolgus animals will be used. Animals judged suitable for experimentation based on clinical sign data and prescreening antibody titers will be placed in study groups by body weight using computer-generated random numbers. Three different routes of administration will be used and relevant tissues collected to evaluate the biodistribution (measured by NGS and PCR) associated with the different routes. Three animals will be implanted with a catheter in the left lateral ventricle for intracerebroventricular (ICV) dose administration (Group 1), three animals will receive a single intravenous infusion (Group 2) and three animals will receive a single intravitreal injection (Group 3). Two animals will serve as replacement animals and will be implanted if required. Animals in Group 1 will have an MRI scan to determine coordinates for proper ICV catheter placement.
The IV infusion will be administered at a rate of 3 mL/min followed by 0.2 mL of vehicle to flush the dose from the IV catheter. The three intravenous animals will receive a single dose of the pooled recombinant AAVs at a volume of 4 mL/kg. The total dose (vg) and dose volume (mL/kg) will be recorded in the raw data. Based on literature review and previous studies in non-human primates, the IV dose of 1×1013 GC/kg body weight was determined to be required to have the desired distribution in the CNS from a systemic delivery as well as the peripheral tissues including skeletal muscle.
The ICV implanted animals will receive a single bolus dose at a volume of 1 mL of AAV-NAV-GFPbc (by slow infusion, approximately 0.1 mL/min) followed by 0.1 mL of vehicle to flush the dose from the catheter system. The ICV dose is based on distribution data from a previous non-human primate study to support current clinical programs.
The intravitreal (IVT) injection will be administered bilateral as a bolus injection at a dose volume of 50 μL.
Clinical signs will be recorded at least once daily beginning approximately two weeks prior to initiation of dosing and continuing throughout the study period. The animals will be observed for signs of clinical effects, illness, and/or death. Additional observations may be recorded based upon the condition of the animal at the discretion of the Study Director and/or technicians.
Ophthalmological examinations will be performed on Group 3 animals prior to dose administration, and on Days 2, 8, 15 and 22. All animals will be sedated with ketamine hydrochloride IM for the ophthalmologic examinations performed following Day 1. For the examinations on Day 1, the animals will be sedated with injectable anesthesia (refer to Section 15.3.3). The eyes will be dilated with 1% tropicamide prior to the examination. The examination will include slit-lamp biomicroscopy and indirect ophthalmoscopy. Additionally, applanation tonometry will be performed on Group 3 animals prior to dosing, immediately following dose administration (˜10 to 15 minutes) and on Days 2 and 22.
Blood samples (˜3 mL) will be collected from a peripheral vein for neutralizing antibodies analysis approximately 2 to 3 weeks prior to dose administration.
Whole blood samples (˜0.5 mL) will be collected from a peripheral vein for bioanalytical analysis (AAV capsid clearance) prior to dose administration, 3 (±10 minutes), 6 (±10 minutes) and 24 (±0.5 hour) hours following dose administration from animals in Group 2 (IV) only. The samples will be collected using a syringe and needle, transferred to two K2 EDTA tubes and the times recorded.
Blood samples (˜5 mL) will be collected from fasted animals from a peripheral vein for PBMC analysis prior to dose administration (Day 1), on Days 8 and 15 and prior to necropsy (Day 22). The samples will be obtained using lithium heparin tubes and the times recorded.
Blood samples will be collected from a peripheral vein for bioanalytical analysis prior to dose administration (Day 1, 2 mL) and necropsy (Day 22, 5 mL). The samples will be collected in clot tubes and the times recorded. The tubes will be maintained at room temperature until fully clotted, then centrifuged at approximately 2400 rpm at room temperature for 15 minutes. The serum will be harvested, placed in labeled vials (necropsy sample split into 1 mL aliquots), frozen in liquid nitrogen, and stored at −60° C. or below.
CSF (˜1.5 mL) will be collected prior to dose administration from a cisterna magna spinal tap from animals in Group 1 only. CSF (˜2 mL) will be collected immediately prior to necropsy from a cisterna magna spinal tap from all animals (Groups 1 to 3). An attempt to collect CSF will be made but due to unsuccessful spinal taps, samples may not be collected at all intervals from an animal(s). Upon collection, the samples will be stored on ice until processing.
A gross necropsy will be performed on any animal found dead or sacrificed moribund, and at the scheduled necropsy, following at least 21 days of treatment (Day 22). All animals, except those found dead, will be sedated with 8 mg/kg of ketamine HCl IM, maintained on an isoflurane/oxygen mixture and provided with an intravenous bolus of heparin sodium, 200 IU/kg. The animals will be perfused via the left cardiac ventricle with 0.001% sodium nitrite in saline. Animals found dead will be necropsied but will not be perfused.
The following tissues will be saved from all animals (including those found dead): Bone marrow, brain, cecum, colon, dorsal nerve roots and ganglion, duodenum, esophagus, eyes with optic nerves, gross lesions, heart, ileum, jejunum, kidneys, knee joint, liver, lungs with bronchi, lymph nodes, ovaries, pancreas, sciatic nerve, skeletal muscle, spinal cord, spleen, thyroids, trachea, and vagus nerve.
The whole blood collected from animals in Group 2 (IV) will be evaluated by qPCR and Next-Generation Sequencing (NGS).
PBMC samples collected from all animals will be evaluated by flow cytometry and enzyme-linked immune absorbent spot (ELISpot), if required.
The presence of circulating neutralizing antibodies as well as free vector in the serum and/or CSF will be evaluated by ELISA and cell based assays, as needed.
The vector copy number and number of transcripts in tissues will be examined by quantitative PCR and NGS methods.
A procedure like that described in Example 13 was used to study the biodistribution of a pool of rAAV capsids administered intravenously to cynomolgus monkey model. Several capsids exhibited good spread in CNS with high relative abundance (RA, compared to AAV9 reference capsid) in most brain regions, notably AAV4, AAV5, rh 34, hu 26, rh3l, and hu13. Favorable capsids exhibiting CNS-tropism have DNA RA values resulting in greater than 1.1-fold increase in DNA values in at least one CNS region, except dorsal root ganglion (DRG).
AAV.rh34, displayed a favorable profile with respect to CNS toxicity as well. The rh34 capsid displayed decreased transduction in dorsal root ganglion (DRG) while exhibiting a high frontal cortex tropism (transduction efficiency). AAVrh34 exhibits an increased RA to AAV9 in CNS regions as follows: 1.8-fold in Hippocampus, 7.4-fold in frontal cortex, 1.9-fold in amygdala, 6.0-fold in medulla, 3.1-fold in midbrain, 1.2-fold in hypothalamus, 8.8-fold in thalamus, 13-fold in globus pallidus, 5.7-fold in SNc, 3.5-fold in dorsal raphe, 2.0-fold in claustrum, 13-fold in putamen, 9-fold in occipital cortex, and 9.6-fold in cerebellum. Additionally, AAVrh34 exhibits a decreased RA in: DRGs: 90-99.5%,Liver: ˜99%, Biceps: ˜30%, Sciatic nerve: 83%, and Optic nerve: 17%.
A procedure like that described in Example 13 with ICV administration was used to study the biodistribution of a pool of rAAV capsids to cynomolgus monkey model. Several capsids exhibited good spread in CNS with high relative abundance (RA, compared to AAV9 reference capsid) in most brain regions, notably AAV4, AAV5, rh 34, hu 26, rh31, and hu13. Favorable capsids exhibiting CNS-tropism have DNA relative abundance values resulting in greater than 1.1-fold increase in DNA values in at least one CNS region, except dorsal root ganglion (DRG).
A procedure like that described in Example 13 with IV administration was used to study the muscle and liver biodistribution of a pool of rAAV capsids to cynomolgus monkey model.
Table 16 provides the rank of each capsid by RA values for the cynomolgus monkey model and the MDX mouse model. Capsids were ranked relative to one another in each animal to decrease variability across animals. Gastrocnemius, TA, heart, bicep, and triceps contributed 70% to the ranking for the MDX Mouse Model and the gastrocnemius, heart, and biceps contributed 70% to the ranking for the cynomolgus monkey model. Liver RA contributed 30% to rankings for each animal. The overall ranking was determined by weighting the ranking for each animal 50%.
Pooled barcoded vectors were administered to NHPs by IV injection. The pooled mixture consists of 118 different AAV capsids, including natural isolates and engineered AAVs, as described herein, expressing the GFP reporter gene from the universal CAG promoter. The intravenous study followed the protocol described in Examples 13 and 14, infra. Several capsids exhibited tropism that “detargeted” the liver, as such, mutated capsids exhibited lower abundance in liver tissue than the parental capsid (AAV9), e.g. AAV8.BBB.LD (A269S, 498-NNN/AAA-500), AAV9.BBB.LD (S263G/S269T/A273T, 496-NNN/AAA-498), AAV9.496-NNN-498, AAV9.496-NNN-498.W503R, AAV9.W503R, and AAV9.Q474A. AAV8 capsids having the NNN/AAA mutation exhibit overall approximately an 11-fold reduction in transduction in liver, and 42-fold reduction in expression of transcript in liver. AAV9 capsids having the NNN/AAA and W503R mutation exhibits approximately a 400-fold reduction in transduction in liver, and results in zero expression of transcript in liver. In some instances, brain distribution of these modified vectors was also diminished. AAV8.BBB.LD additionally exhibits a high level of transduction in gastrocnemius muscle.
Studies also show the change in relative abundance (adjusted for input, normalized to 1) between the abundance for each barcode (and therefore capsid) at 3 hr post and 24 hr post IV capsid library administration (RA at 3 hr/RA at 24 hr). Individual animals are indicated by different shape data points (3 animals total) (
A fold change >1 indicates that the capsid makes up a lower percentage of the total capsid “pool” present in the blood at 24 hr compared to 3 hr after dosing (i.e. faster blood clearance). A fold change <1 indicates that the capsid makes up a greater percentage of the total capsid “pool” present in the blood at 24 hr compared to 3 hr after dosing (i.e. slower clearance). Historically, slower clearance correlates with lower liver transduction/liver detargeting.
As represented by increase in blood retention, a depiction of the change in abundance for a given capsid in a given animal was plotted. Allowing for the calculation of the fold increase in blood retention over the baseline retention of AAV9, for example, the representations (
Pooled barcoded vectors were administered to mdx mice by IV (tail vein) injection. The pooled mixture consists of 118 different AAV capsids, including natural isolates and engineered AAVs, as described herein, expressing the GFP reporter gene from the universal CAG promoter. The IV study followed a protocol analogous to that described in Examples 12 and 16, infra.
At 3 week sacrifice, tissues were harvested and samples were collected in tubes with RNAlater (per manufacturer's instructions) and flash frozen at −80° C. until DNA and RNA analysis (biodistribution of each vector in the pool) were performed by NGS (see
Wild-type AAV9 was tested against three mutant capsids AAV9.G266A.496NNN/AAA498 (SEQ ID NO:50), AAV9.N272A.496NNN/AAA498 (SEQ ID NO:49), and AAV9.496NNN/AAA498.W503A (SEQ ID NO:51) for biodistribution in 8-10 week old C57BL/6 mice (5 mice per group). Each cohort was intravenously administered 1e13 GC/kg rAAV vector carrying a CAG.dtTomato transgene with unique barcode on day 0, and mice were sacrificed, and tissues collected on day 21. As seen in
Transgene RNA copies were significantly reduced from administration of mutant AAV9 vectors compared to wild-type AAV9, and transgene RNA copies from vector AAV9.NNN.W503A were undetectable in murine liver (
Pooled barcoded vectors were administered to NHPs by IV injection at a total dose of 2e13 GC/kg. The IV study followed a protocol analogous to that described in Examples 12 and 16, infra. The pooled mixture consists of 44 capsids in total, with each capsid gene construct expressing a CAG-tdTomato barcode. Studies were analyzed for the change in relative abundance (RNA copies, adjusted for input, and normalized to 1) between the abundance for each barcode (and therefore capsid) at 3 hr post and 24 hr post IV capsid library administration. Reduced RNA copies were observed for capsids AAV9.G266A.496NNN/AAA498 and AAV9.496NNN/AAA498.W503A in various muscle and peripheral tissues in NHPs (
Table 17 provides the amino acid sequences of certain engineered capsid proteins described and/or used in studies described herein. Heterologous peptides and amino acid substitutions are indicated in gray shading.
Although the invention is described in detail with reference to specific embodiments thereof, it will be understood that variations which are functionally equivalent are within the scope of this invention. Indeed, various modifications of the invention in addition to those shown and described herein will become apparent to those skilled in the art from the foregoing description and accompanying drawings. Such modifications are intended to fall within the scope of the appended claims. Those skilled in the art will recognize, or be able to ascertain using no more than routine experimentation, many equivalents to the specific embodiments of the invention described herein. Such equivalents are intended to be encompassed by the following claims.
All publications, patents and patent applications mentioned in this specification are herein incorporated by reference into the specification to the same extent as if each individual publication, patent or patent application was specifically and individually indicated to be incorporated herein by reference in their entireties.
The discussion herein provides a better understanding of the nature of the problems confronting the art and should not be construed in any way as an admission as to prior art nor should the citation of any reference herein be construed as an admission that such reference constitutes “prior art” to the instant application.
All references including patent applications and publications cited herein are incorporated herein by reference in their entirety and for all purposes to the same extent as if each individual publication or patent or patent application was specifically and individually indicated to be incorporated by reference in its entirety for all purposes. Many modifications and variations of this invention can be made without departing from its spirit and scope, as will be apparent to those skilled in the art. The specific embodiments described herein are offered by way of example only, and the invention is to be limited only by the terms of the appended claims, along with the full scope of equivalents to which such claims are entitled.
Number | Date | Country | Kind |
---|---|---|---|
PCT/US2021/054058 | Oct 2021 | WO | international |
Filing Document | Filing Date | Country | Kind |
---|---|---|---|
PCT/US2022/077816 | 10/7/2022 | WO |
Number | Date | Country | |
---|---|---|---|
63331161 | Apr 2022 | US |