ANELLOVECTORS FOR DELIVERY OF EFFECTORS TO THE CENTRAL NERVOUS SYSTEM

Abstract
This invention relates generally to Anelloviridae family vectors (e.g., anellovectors) and compositions and uses thereof.
Description
SEQUENCE LISTING

The instant application contains a Sequence Listing which has been submitted electronically in XML file format and is hereby incorporated by reference in its entirety. Said XML copy, created on May 31, 2024, is named V2057-703110_SL.xml and is 311,526 bytes in size.


BACKGROUND

There is an ongoing need to develop suitable vectors to deliver therapeutic genetic material to patients.


SUMMARY

The present disclosure provides an Anelloviridae family vector (e.g., anellovector), e.g., a synthetic Anelloviridae family vector (e.g., anellovector), that can be used as a delivery vehicle, e.g., for delivering genetic material, for delivering an effector, e.g., a payload, or for delivering a therapeutic agent or a therapeutic effector to a eukaryotic cell (e.g., a human cell or a human tissue).


Additional features of any of the aforesaid Anelloviridae family vectors (e.g., anellovectors), compositions or methods include one or more of the following enumerated embodiments.


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 enumerated embodiments.


ENUMERATED EMBODIMENTS

1. A method of delivering an exogenous effector to the central nervous system (CNS) of a subject, the method comprising administering to the CNS of the subject an Anelloviridae family vector (e.g., an anellovector).


2. A method of delivering a DNA capable of encoding (e.g., encoding) an exogenous effector to the CNS of a subject, the method comprising administering to the CNS of the subject an Anelloviridae family vector (e.g., an anellovector).


3. The method of embodiment 1 or 2, which comprises delivery of the Anelloviridae family vector to the brain of the subject.


4. The method of any of embodiments 1-3, which comprises production of the exogenous effector and/or DNA capable of encoding (e.g., encoding) the exogenous effector in the brain of the subject.


5. A method of modulating a biological function in the CNS of a subject, the method comprising administering to the CNS of the subject an Anelloviridae family vector (e.g., an anellovector).


6. The method of embodiment 5, which comprises delivery of the Anelloviridae family vector to the brain of the subject.


7. The method of embodiment 5 or 6, which comprises modulating a biological function in the brain of the subject.


8. A method of treating a CNS disease or disorder in a subject in need thereof, the method comprising administering to the subject an Anelloviridae family vector (e.g., an anellovector).


9. The method of embodiment 8, wherein the CNS disease or disorder is a brain disease or disorder.


10. The method of embodiment 8 or 9, wherein the CNS disease or disorder is chosen from: ataxia imbalance (e.g., Friedreich's ataxia), spinal cerebellar atrophy, frontotemporal dementia, Alzheimer's Disease, Parkinson's Disease, Lewy body dementia, a genetic seizure disorder, Huntington's disease, a psychiatric disorder (e.g., schizophrenia, bipolar disorder, or depression), spinal muscular atrophy (SMA), multiple sclerosis, traumatic brain injury, a brain tumor, a genetic disease (e.g., a rare genetic disease), Batten disease, Rett syndrome, Duchenne muscular dystrophy, Amyotrophic Lateral Sclerosis (ALS), a neural development disease (e.g., autism, Rett syndrome, or Fragile X syndrome), or a leukodystrophy (e.g., adrenal leukodystrophy or metachromatic leukodystrophy).


11. The method of any of embodiments 8-10, wherein the subject has an impairment of one or more of: memory, behavior, or language.


12. The method of any of embodiments 1-11, which results in delivery of anellovector DNA in one or more cell types selected from: neurons (e.g., GABAergic neurons, glutamatergic neurons, spinal motor neurons), glial cells, microglia, oligodenroglia, and astrocytes.


13. The method of any of embodiments 1-12, which results in expression of the exogenous effector in one or more cell types selected from: neurons (e.g., GABAergic neurons, glutamatergic neurons, spinal motor neurons), glial cells, microglia, oligodenroglia, and astrocytes.


14. The method of any of embodiments 1-13, which comprises contacting (e.g., directly or indirectly) the anellovector to one or more cell types selected from: neurons (e.g. GABAergic neurons, glutamatergic neurons, spinal motor neurons), glial cells, microglia, oligodenroglia, and astrocytes.


15. The method of any of embodiments 1-14, which results in delivery of anellovector DNA in one or more of: cerebellum, frontotemporal lobe (e.g., in the hippocampus), parietal lobe, brain stem, and spinal cord.


16. The method of any of embodiments 1-15, which results in expression of the exogenous effector in one or more of: cerebellum, frontotemporal lobe (e.g., in the hippocampus), parietal lobe, brain stem, and spinal cord.


17. The method of any of embodiments 1-16, which comprises contacting (e.g., directly or indirectly) the anellovector to one or more of: cerebellum, frontotemporal lobe (e.g., in the hippocampus), parietal lobe, brain stem, and spinal cord.


18. The method of any of embodiments 1-17, which results in greater delivery of the exogenous effector and/or the DNA encoding the exogenous effector to the brain than to the spinal cord.


19. The method of any of embodiments 1-18, wherein the Anelloviridae family vector (e.g., an anellovector) is administered according to a route of administration chosen from: intrathecal (IT), intracerebroventricular (ICV), intra cisterna magna (ICM), or intraparenchymal (IPa).


20. The method of embodiment 18, wherein the Anelloviridae family vector (e.g., an anellovector) is administered according to an intracerebroventricular (ICV) route of administration.


21. The method of any of the preceding embodiments, wherein the Anelloviridae family vector is an anellovector comprising a proteinaceous exterior comprising an ORF1 molecule and a genetic element enclosed by the proteinaceous exterior, wherein the genetic element comprises a promoter element operably linked to a nucleic acid sequence (e.g., a DNA sequence) encoding the exogenous effector.


22. The method of any of the preceding embodiments, wherein the Anelloviridae family vector is an Anellovector comprising a proteinaceous exterior comprising an ORF1 molecule having an ORF1 sequence as listed in any of Tables A1-A3, or a polypeptide comprising an amino acid sequence having at least about 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity thereto.


23. The method of any of the preceding embodiments, wherein the ORF1 molecule comprises at least one difference relative to the wild-type ORF1 protein of Table A1-A3.


24. The method any of the preceding embodiments, wherein the difference comprises a mutation (e.g., an insertion, a substitution, or a deletion), a chemical modification, or an enzymatic modification.


25. The method of any of the preceding embodiments, wherein the mutation comprises a deletion, e.g., a deletion of a domain (e.g., one or more of an arginine-rich region, jelly-roll domain, HVR, N22, or CTD, e.g., as described herein).


26. The method of any of the preceding embodiments, wherein the ORF1 molecule comprises an ORF1 domain and an exogenous moiety, e.g., an exogenous surface moiety.


27. The method of any of the preceding embodiments, wherein the Anelloviridae family vector is an Anellovector comprising a proteinaceous exterior comprising a polypeptide encoded by an Anellovirus ORF1 nucleic acid sequence as listed in Table N1-N3, or a polypeptide encoded by a nucleic acid sequence having at least about 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity to the Anellovirus ORF1 nucleic acid sequence.


28. The method of any of the preceding embodiments, wherein the genetic element comprises an Anellovirus 5′ UTR conserved domain having a sequence of the reverse complement of nucleotides 323-393 of SEQ ID NO: 54, or a nucleic acid sequence having at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity thereto, or a functional portion thereof.


29. The method of any of the preceding embodiments, wherein the genetic element comprises an Anellovirus GC-rich region having a sequence of the reverse complement of nucleotides 2868-2929 of SEQ ID NO: 54, or a nucleic acid sequence having at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity thereto, or a functional portion thereof.


30. The method of embodiments 1-27, wherein the genetic element comprises an Anellovirus 5′ UTR conserved domain having a sequence of the reverse complement of nucleotides 1-71 of SEQ ID NO: 1, or a nucleic acid sequence having at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity thereto, or a functional portion thereof.


31. The method of embodiments 1-27 or 30, wherein the genetic element comprises an Anellovirus GC-rich region having a sequence of the reverse complement of nucleotides 2515-2615 of SEQ ID NO: 1, or a nucleic acid sequence having at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity thereto, or a functional portion thereof.


32. The method of any of the preceding embodiments, wherein the portion of the Anellovirus GC-rich region is at least 20, 30, 40, 50, 60, 70, 80, 90, or 100 nucleotides.


33. The method of any of the preceding embodiments, wherein the genetic element is DNA.


34. The method of any of the preceding embodiments, wherein the genetic element is circular, single stranded DNA.


35. The method of any of embodiments 1-32, wherein the genetic element is RNA, e.g., mRNA.


36. The method of any of the preceding embodiments, wherein the exogenous effector comprises: an intracellular peptide or intracellular polypeptide, a secreted polypeptide, or a protein replacement therapeutic.


37. The method of any of the preceding embodiments, wherein the Anelloviridae family vector (e.g., an anellovector) is a Betatorquevirus.


38. The method of any of the preceding embodiments, which further comprises administering an additional dose of an Anelloviridae family vector (e.g., anellovector) to the CNS of the subject.


39. The method of embodiment 38, wherein the Anelloviridae family vector and the Anelloviridae family vector of the additional dose are the same.


40. The method of embodiment 38, wherein the Anelloviridae family vector and the Anelloviridae family vector of the additional dose are different.


41. The method of any of the preceding embodiments, which results in greater delivery of the DNA encoding the exogenous effector to the CNS than to muscle.


42. The method of any of the preceding embodiments, which results in greater delivery of the DNA encoding the exogenous effector to the CNS than to liver.


43. The method of any of the preceding embodiments, which results in greater delivery of the DNA encoding the exogenous effector to the spinal cord than to muscle.


44. The method of any of the preceding embodiments, which results in greater delivery of the DNA encoding the exogenous effector to the spinal cord than to liver.


45. The method of any of embodiments 41-44, wherein the greater delivery is 2 times, 5 times, or 10 times higher.


46. A delivery system suitable for delivery to the CNS, wherein the delivery system comprises an Anelloviridae family vector (e.g., an anellovector).


47. The Anelloviridae family vector (e.g., anellovector) of any of the preceding embodiments, wherein the proteinaceous exterior (e.g., ORF1 molecule of the proteinaceous exterior) comprises the amino acid sequence YNPX2DXGX2N (SEQ ID NO: 829), wherein X″ is a contiguous sequence of any n amino acids.


48. The Anelloviridae family vector (e.g., anellovector) of embodiment 47, wherein the amino acid sequence YNPX2DXGX2N (SEQ ID NO: 829) is comprised in an N22 domain of the ORF1 molecule.


49. The Anelloviridae family vector (e.g., anellovector) of any of the preceding embodiments, wherein the ORF1 molecule comprises one or more (e.g., 1, 2, 3, 4, or all 5) of the following Anellovirus ORF1 subdomains: an arginine-rich region, a jelly-roll region, a hypervariable region, an N22 domain, a C-terminal domain (CTD) (e.g., as described herein), e.g., of an Anellovirus ORF1 protein as listed in Table A1-A3 (or a sequence having at least about 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity thereto).


50. The Anelloviridae family vector (e.g., anellovector) of any of the preceding embodiments, which comprises at least one difference relative to a wild type Anellovirus genome sequence, wherein the at least one difference comprises a deletion (e.g., lacks one or more of: an Anellovirus 5′ UTR conserved domain, an ORF1 gene, ORF2 gene, an Anellovirus GC-rich region, an ORF3 gene, or a functional fragment thereof).


51. The Anelloviridae family vector (e.g., anellovector) of any of the preceding embodiments, wherein the genetic element further comprises a promoter.


52. The Anelloviridae family vector (e.g., anellovector) of any of the preceding embodiments, wherein the genetic element further comprises a poly A sequence.


53. A pharmaceutical composition comprising an Anelloviridae family vector (e.g., anellovector) of any of the preceding embodiments.


54. The pharmaceutical composition of embodiment 53, wherein the pharmaceutical composition has one or more of the following characteristics:

    • a) the pharmaceutical composition meets a pharmaceutical or good manufacturing practices (GMP) standard;
    • b) the pharmaceutical composition was made according to good manufacturing practices (GMP);
    • c) the pharmaceutical composition has a pathogen level below a predetermined reference value, e.g., is substantially free of pathogens;
    • d) the pharmaceutical composition has a contaminant level below a predetermined reference value, e.g., is substantially free of contaminants;
    • e) the pharmaceutical composition has a predetermined level of non-infectious particles or a predetermined ratio of particles:infectious units (e.g., <300:1, <200:1, <100:1, or <50:1), or
    • f) the pharmaceutical composition has low immunogenicity or is substantially non-immunogenic, e.g., as described herein.


55. The pharmaceutical composition of any one of embodiments 53-54, wherein the pharmaceutical composition has a contaminant level below a predetermined reference value, e.g., is substantially free of contaminants.


56. The pharmaceutical composition of embodiment 55, wherein the contaminant is selected from the group consisting of: mycoplasma, endotoxin, host cell nucleic acids (e.g., host cell DNA and/or host cell RNA), animal-derived process impurities (e.g., serum albumin or trypsin), replication-competent agents (RCA), e.g., replication-competent virus or unwanted Anelloviridae family vector (e.g., anellovector) (e.g., an Anelloviridae family vector other than the desired Anelloviridae family vector, e.g., a synthetic Anelloviridae family vector as described herein), free viral capsid protein, adventitious agents, and aggregates.


57. The pharmaceutical composition of embodiment 55, wherein the contaminant is host cell DNA and the threshold amount is about 10 ng of host cell DNA per dose of the pharmaceutical composition.


58. The pharmaceutical composition of any one of embodiments 53-57, wherein the pharmaceutical composition comprises less than 10% (e.g., less than about 10%, 5%, 4%, 3%, 2%, 1%, 0.5%, or 0.1%) contaminant by weight.


59. The Anelloviridae family vector (e.g., anellovector) or method of any of the preceding embodiments, wherein the genetic element is single-stranded.


60. The Anelloviridae family vector (e.g., anellovector or method of any of the preceding embodiments, wherein the genetic element is circular.


61. The Anelloviridae family vector (e.g., anellovector) or method of any of the preceding embodiments, wherein the genetic element comprises DNA.


62. The Anelloviridae family vector (e.g., anellovector) or method of any of the preceding embodiments, wherein the genetic element is a negative strand DNA.


63. The Anelloviridae family vector (e.g., anellovector) or method of any of the preceding embodiments, wherein the genetic element is double-stranded.


64. The Anelloviridae family vector (e.g., anellovector) or method of any of the preceding embodiments, wherein the genetic element is linear.


65. The Anelloviridae family vector (e.g., anellovector) or method of any of the preceding embodiments, wherein the genetic element comprises RNA.


66. The Anelloviridae family vector (e.g., anellovector) or method of any of the preceding embodiments, wherein the genetic element comprises a nucleic acid sequence encoding an Anelloviridae capsid protein, e.g., an Anellovirus ORF1 molecule (e.g., an ORF1 protein as listed in Table A1-A3 or an amino acid sequence having at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity thereto).


67. The Anelloviridae family vector (e.g., anellovector) or method of any of the preceding embodiments, wherein the genetic element does not comprise a nucleic acid sequence encoding an Anelloviridae capsid protein, e.g., an Anellovirus ORF1 molecule (e.g., an ORF1 protein as listed in Table A1-A3 or an amino acid sequence having at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity thereto).


68. The Anelloviridae family vector (e.g., anellovector) or method of any of the preceding embodiments, wherein the genetic element comprises a nucleic acid sequence encoding an Anellovirus ORF2 molecule (e.g., an ORF2 protein as listed in Table A1-A3, or an amino acid sequence having at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity thereto).


69. The Anelloviridae family vector (e.g., anellovector), nucleic acid molecule, or method of any of the preceding embodiments, wherein the genetic element does not comprise a nucleic acid sequence encoding an Anellovirus ORF2 molecule (e.g., an ORF2 protein as listed in Table A1-A3 or an amino acid sequence having at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity thereto).


70. The Anelloviridae family vector (e.g., anellovector) or method of any of the preceding embodiments, wherein the genetic element comprises at least 20, 25, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, or 40 consecutive nucleotides having a GC content of at least 70%, 75%, 80%, 85%, 90%, 95%, or 99%.


71. The Anelloviridae family vector (e.g., anellovector) or method of any of the preceding embodiments, wherein the proteinaceous exterior comprises the amino acid sequence YNPX2DXGX2N (SEQ ID NO: 829), wherein X″ is a contiguous sequence of any n amino acids.


72. The Anelloviridae family vector (e.g., anellovector), nucleic acid molecule, or method of embodiment 71, wherein the amino acid sequence YNPX2DXGX2N (SEQ ID NO: 829) is comprised in an N22 domain.


73. The Anelloviridae family vector (e.g., anellovector), nucleic acid molecule, or method of any of the preceding embodiments, wherein the ORF1 molecule comprises an arginine-rich region (e.g., having at least 70%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100% identity to an arginine-rich region sequence of an ORF1 protein listed in Table A1-A3).


74. The Anelloviridae family vector (e.g., anellovector) or method of any of the preceding embodiments, wherein the proteinaceous exterior comprises an amino acid sequence of at least 15, 20, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 45, or 50 consecutive nucleotides comprising at least 40% (e.g., at least 40%, 41%, 42%, 43%, 44%, 45%, 46%, 47%, 48%, 49%, 50%, 55%, 60%, 65%, 66%, 67%, 68%, 69%, 70%, 75%, 80%, 85%, 90%, or 95%) arginine residues.


75. The Anelloviridae family vector (e.g., anellovector), nucleic acid molecule, or method of embodiment 73 or 74, wherein the arginine-rich region is located at the N-terminal or C-terminal end of the ORF1 molecule.


76. The Anelloviridae family vector (e.g., anellovector) or method of any of the preceding embodiments, wherein the genetic element comprises one or more of: a TATA box, an initiator element, a cap site, a transcriptional start site, an ORF1/1-encoding sequence, an ORF1/2-encoding sequence, an ORF2/2-encoding sequence, an ORF2/3-encoding sequence, an ORF2/3t-encoding sequence, a three open-reading frame region, a poly(A) signal, and/or a GC-rich region from an Anellovirus described herein, or a sequence having at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity thereto.


77. The Anelloviridae family vector (e.g., anellovector) or method of any of the preceding embodiments, wherein the genetic element comprises at least 75% (e.g., at least 75, 76, 77, 78, 79, 80, 85, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, or 100%) sequence identity to an Anellovirus 5′ UTR conserved domain sequence as described herein.


78. The Anelloviridae family vector (e.g., anellovector), nucleic acid molecule, or method of any of the preceding embodiments, wherein the genetic element comprises at least 75% (e.g., at least 75, 76, 77, 78, 79, 80, 85, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, or 100%) sequence identity to an Anellovirus GC-rich region sequence as described herein.


79. The Anelloviridae family vector (e.g., anellovector) or method of any of the preceding embodiments, wherein the effector encodes a therapeutic agent, e.g., a therapeutic peptide or polypeptide or a therapeutic nucleic acid.


80. The Anelloviridae family vector (e.g., anellovector), nucleic acid molecule, or method of any of the preceding embodiments, wherein the effector is an exogenous effector.


81. The Anelloviridae family vector (e.g., anellovector), nucleic acid molecule, or method of any of the preceding embodiments, wherein the effector is an endogenous effector (e.g., wherein the anellovector overexpresses the endogenous effector in a target cell).


82. The Anelloviridae family vector (e.g., anellovector) or method of any of the preceding embodiments, wherein the effector modulates expression or activity of a gene or protein, e.g., increases or decreases expression or activity of the gene or protein.


83. The Anelloviridae family vector (e.g., anellovector), nucleic acid molecule, or method of any of the preceding embodiments, wherein the Anelloviridae family vector is capable of replicating autonomously.


84. The Anelloviridae family vector (e.g., anellovector), nucleic acid molecule, or method of any of the preceding embodiments, wherein the Anelloviridae family vector is replication-deficient (e.g., incapable of replicating autonomously).


85. The Anelloviridae family vector (e.g., anellovector), nucleic acid molecule, or method of any of the preceding embodiments, wherein the genetic element integrates into the genome of a eukaryotic cell at a frequency of less than about 0.001%, 0.005%, 0.01%, 0.05%, 0.1%, 0.5%, 1%, 1.5%, or 2% of the genetic element that enters the cell.


86. The Anelloviridae family vector (e.g., anellovector) or method of any of the preceding embodiments, wherein the Anelloviridae family vector is substantially non-pathogenic, e.g., does not induce a detectable deleterious symptom in a subject (e.g., elevated cell death or toxicity, e.g., relative to a subject not exposed to the anellovector).


87. The Anelloviridae family vector (e.g., anellovector), nucleic acid molecule, or method of any of the preceding embodiments, wherein the Anelloviridae family vector is substantially non-immunogenic, e.g., does not induce a detectable and/or unwanted immune response.


88. The Anelloviridae family vector (e.g., anellovector) or method of any of the preceding embodiments, wherein a population of at least 1000 of the Anelloviridae family vectors is capable of delivering at least about 100 copies (e.g., at least 1, 2, 3, 4, 5, 10, 20, 30, 40, 50, 100, 200, 300, 400, 500, 600, 700, 800, 900, or 1000 copies) of the genetic element into one or more eukaryotic cells (e.g., mammalian cells, e.g., human cells).


89. The Anelloviridae family vector (e.g., anellovector), nucleic acid molecule, or method of any of the preceding embodiments, wherein a population of the Anelloviridae family vectors (e.g., at least 1, 2, 3, 4, 5, 10, 20, 30, 40, 50, 100, 200, 300, 400, 500, 600, 700, 800, 900, or 1000 genome equivalents of the genetic element per cell) is capable of delivering the genetic element into at least 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, 99%, or more of a population of eukaryotic cells (e.g., mammalian cells, e.g., human cells).


90. The Anelloviridae family vector (e.g., anellovector) or method of any of the preceding embodiments, wherein a population of the Anelloviridae family vectors (e.g., at least 1, 2, 3, 4, 5, 10, 20, 30, 40, 50, 100, 200, 300, 400, 500, 600, 700, 800, 900, or 1000 genome equivalents of the genetic element per cell) is capable of delivering at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 20, 50, 100, 200, 500, 1000, 2000, 5000, 8,000, 1×104, 1×105, 1×106, 1×107 or greater copies of the genetic element per cell to a population of eukaryotic cells (e.g., mammalian cells, e.g., human cells).


91. The Anelloviridae family vector (e.g., anellovector) or method of any of the preceding embodiments, wherein a population of the Anelloviridae family vectors (e.g., at least 1, 2, 3, 4, 5, 10, 20, 30, 40, 50, 100, 200, 300, 400, 500, 600, 700, 800, 900, or 1000 genome equivalents of the genetic element per cell) is capable of delivering 1-3, 1-4, 1-5, 1-6, 1-7, 1-8, 1-9, 1-10, 5-10, 10-20, 20-50, 50-100, 100-1000, 1000-104, 1×104-1×105, 1×104-1×106, 1×104-1×107, 1×105-1×106, 1×105-1×107, or 1×106-1×107 copies of the genetic element per cell to a population of eukaryotic cells (e.g., mammalian cells, e.g., human cells).


92. The Anelloviridae family vector (e.g., anellovector) or method of any of the preceding embodiments, wherein the target cells into which the genetic element is delivered each receive at least 10, 50, 100, 500, 1000, 10,000, 50,000, 100,000, or more copies of the genetic element.


93. The Anelloviridae family vector (e.g., anellovector) or method of any of the preceding embodiments, wherein the Anelloviridae family vector is resistant to degradation by a detergent (e.g., a mild detergent, e.g., a biliary salt, e.g., sodium deoxycholate) relative to a viral particle comprising an external lipid bilayer, e.g., a retrovirus.


94. The Anelloviridae family vector (e.g., anellovector) or method of any of the preceding embodiments, wherein the genetic element enclosed by the proteinaceous exterior is resistant to degradation by a nuclease enzyme (e.g., a DNase).


95. The Anelloviridae family vector (e.g., anellovector) or method of any of the preceding embodiments, wherein the Anelloviridae family vector is capable of infecting mammalian cells, e.g., human cells, e.g., in vitro, in vivo, or ex vivo.


96. The Anelloviridae family vector (e.g., anellovector) or method of any of the preceding embodiments, wherein the Anelloviridae family vector selectively delivers the effector to, or is present at higher levels in (e.g., preferentially accumulates in), a desired cell type, tissue, or organ


97. The Anelloviridae family vector (e.g., anellovector) or method of any of the preceding embodiments, wherein the proteinaceous exterior is provided in cis relative to the genetic element.


98. The Anelloviridae family vector (e.g., anellovector) or method of any of the preceding embodiments, wherein the proteinaceous exterior is provided in trans relative to the genetic element.


99. The Anelloviridae family vector of any of the previous embodiments, which is substantially free of wild-type Anellovirus genomes.


Other features, objects, and advantages of the invention will be apparent from the description and drawings, and from the claims.


Unless otherwise defined, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs. All publications, patent applications, patents, and other references mentioned herein are incorporated by reference in their entirety. In addition, the materials, methods, and examples are illustrative only and not intended to be limiting.





BRIEF DESCRIPTION OF THE DRAWINGS

The following detailed description of the embodiments of the invention will be better understood when read in conjunction with the appended drawings. For the purpose of illustrating the invention, there are shown in the drawings embodiments that are presently exemplified. It should be understood, however, that the invention is not limited to the precise arrangement and instrumentalities of the embodiments shown in the drawings.



FIG. 1A and FIG. 1B are representative merged (GFP and brightfield) images of MOLT4 cells transfected with the pRTx-2847 (R19-eGFP vector, SEQ ID NO: 502), pRTx-2848 (iCre plasmid, SEQ ID NO: 503), and pRTx-3525 (R19 SRR, SEQ ID NO: 500) 72 hours post-electroporation. Images were taken at 10× for Study #1 producer cells (FIG. 1A) and Study #2 producer cells (FIG. 1B). White cells exhibit GFP expression.



FIG. 2A and FIG. 2B are plots depicting number of DNase-protected eGFP copies per fraction of cell lysate on post-iodixanol fractions for Study #1 Ring19-eGFP Anellovector material (FIG. 2A) and Study #2 Ring19-eGFP Anellovector material (FIG. 2B).



FIG. 3A and FIG. 3B are plots depicting the number of DNase-protected Ring19-eGFP copies pre- and post-concentration of viral material for Study #1 Ring19-eGFP Anellovector material (FIG. 3A) and Study #2 Ring19-eGFP Anellovector material (FIG. 3B).



FIG. 4A and FIG. 4B depict gels showing a Coomassie Stain on pre- and post-concentration viral material for Study #1 Ring19-eGFP Anellovector material (FIG. 4A) and Study #2 Ring19-eGFP Anellovector material (FIG. 4B).



FIG. 5A and FIG. 5B depict plots showing the quantitation of eGFP gDNA in mouse brain (FIG. 5A) and spinal cord (FIG. 5B) by qPCR. Each dot represents one brain hemisphere or spinal cord. n=5 per group.



FIG. 6 depicts a plot showing the quantitation of eGFP mRNA in the mouse brain by RT-ddPCR. Each dot represents one brain hemisphere. n=5 brain hemispheres per group.



FIG. 7A-7F depict fluorescence microscopy images of representative brain sections of ICV-injected mice stained for eGFP protein from the PBS control group (FIG. 7A, FIG. 7D), the R19-eGFP group (FIG. 7B, FIG. 7E), and the AAV9-eGFP group (FIG. 7C, FIG. 7F). Images were taken at 2× (FIG. 7A-C) or 20× (FIG. 7D-F) magnification. The white square in FIGS. 7A-C indicates the location of the 20× magnification in the respective 2× image. eGFP protein expression is detected in the R19-eGFP and AAV9-eGFP groups, while no staining is detected in the PBS control group. n=2 brains per group.



FIG. 8A and FIG. 8B depict plots showing the quantitation of eGFP gDNA by qPCR in the spinal cord (FIG. 8A) and brain (FIG. 8B) in mice administered with PBS, R19-fCMV-eGFP, or AAV9-fCMV-eGFP by intrathecal (IT) injection. n=5 per group.



FIGS. 9A-9D depict plots showing the quantitation of eGFP gDNA by qPCR in the spinal cord (FIG. 9A), brain (FIG. 9B), liver (FIG. 9C), and muscle (FIG. 9D) in mice from Group 1 (“PBS-D21”), Group 2 (“PBS×1 D42”), Group 3 (“PBS×2 D42”), Group 4 (“R19-eGFP—D21”), Group 5 (“R19-eGFP×1 D42”), Group 6 (“R19-eGFP×2 D42”), Group 7 (“AAV9-eGFP-D21”), Group 8 (“AAV-eGFP×1 D42”), and Group 9 (“AAV-eGFP×2 D42”).



FIGS. 10A-10D depict plots showing the quantitation of eGFP mRNA by RT-ddPCR in the spinal cord (FIG. 10A), liver (FIG. 10B), brain (FIG. 10C), and muscle (FIG. 10D) in mice from Group 1 (“PBS-D21”), Group 2 (“PBS×1 D42”), Group 3 (“PBS×2 D42”), Group 4 (“R19-eGFP—D21”), Group 5 (“R19-eGFP×1 D42”), Group 6 (“R19-eGFP×2 D42”), Group 7 (“AAV9-eGFP-D21”), Group 8 (“AAV-eGFP×1 D42”), or Group 9 (“AAV-eGFP×2 D42”). In FIGS. 10C and 10D, “PBS” corresponds to Group 1, “Ring19-eGFP” corresponds to Group 4, and “AAV9-eGFP” corresponds to Group 7.



FIGS. 11A-11J depict fluorescence microscopy images of representative spinal cord sections of IT-injected mice stained for eGFP protein from Group 1 (“PBS 21 day) (FIG. 11A), Group 2 (“PBS 42 day”) (FIG. 11B), Group 3 (“PBS redose”) (FIG. 11C), Group 4 (“R19 21 day”) (FIG. 11D), Group 5 (“R19 42 day”) (FIG. 11E), Group 6 (“R19 redose”) (FIG. 11F), Group 7 (“AAV 21 day”) (FIG. 11G), Group 8 (“AAV 42 day”) (FIG. 11H), and Group 9 (“AAV redose”) (FIGS. 11I and 11J).



FIGS. 11A-11I are shown at 20× magnification. The square in FIG. 11J (2× magnification) indicates the location of the tissue magnified to 20× in FIG. 11I.





DETAILED DESCRIPTION OF CERTAIN EMBODIMENTS
Definitions

The present invention will be described with respect to particular embodiments and with reference to certain figures, but the invention is not limited thereto but only by the claims. Terms as set forth hereinafter are generally to be understood in their common sense unless indicated otherwise.


Where the term “comprising” is used in the present description and claims, it does not exclude other elements. For the purposes of the present invention, the term “consisting of” is considered to be a preferred embodiment of the term “comprising of”. If hereinafter a group is defined to comprise at least a certain number of embodiments, this is to be understood to preferably also disclose a group which consists only of these embodiments.


Where an indefinite or definite article is used when referring to a singular noun, e.g. “a”, “an” or “the”, this includes a plural of that noun unless something else is specifically stated.


The wording “compound, composition, product, etc. for treating, modulating, etc.” is to be understood to refer a compound, composition, product, etc. per se which is suitable for the indicated purposes of treating, modulating, etc. The wording “compound, composition, product, etc. for treating, modulating, etc.” additionally discloses that, as an embodiment, such compound, composition, product, etc. is for use in treating, modulating, etc.


The wording “compound, composition, product, etc. for use in . . . ”, “use of a compound, composition, product, etc. in the manufacture of a medicament, pharmaceutical composition, veterinary composition, diagnostic composition, etc. for . . . ”, or “compound, composition, product, etc. for use as a medicament . . . ” indicates that such compounds, compositions, products, etc. are to be used in therapeutic methods which may be practiced on the human or animal body. They are considered as an equivalent disclosure of embodiments and claims pertaining to methods of treatment, etc. If an embodiment or a claim thus refers to “a compound for use in treating a human or animal being suspected to suffer from a disease”, this is considered to be also a disclosure of a “use of a compound in the manufacture of a medicament for treating a human or animal being suspected to suffer from a disease” or a “method of treatment by administering a compound to a human or animal being suspected to suffer from a disease”. The wording “compound, composition, product, etc. for treating, modulating, etc.” is to be understood to refer a compound, composition, product, etc. per se which is suitable for the indicated purposes of treating, modulating, etc.


If hereinafter examples of a term, value, number, etc. are provided in parentheses, this is to be understood as an indication that the examples mentioned in the parentheses can constitute an embodiment. For example, if it is stated that “in embodiments, the nucleic acid molecule comprises a nucleic acid sequence having at least about 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to the Anellovirus ORF1-encoding nucleotide sequence of Table 1 (e.g., nucleotides 571-2613 of the nucleic acid sequence of Table 1)”, then some embodiments relate to nucleic acid molecules comprising a nucleic acid sequence having at least about 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to nucleotides 571-2613 of the nucleic acid sequence of Table 1.


As used herein, the term “Anelloviridae family vector” refers to a vehicle derived from or similar to a virus of the Anelloviridae family (e.g., an Alphatorquevirus, Betatorquevirus, Gammatorquevirus, or chicken anemia virus), wherein the vehicle comprises a genetic element enclosed in a proteinaceous exterior (e.g., the genetic element is substantially protected from digestion with DNAse I by a proteinaceous exterior). In some embodiments, an Anelloviridae family vector comprises a genetic element derived from or highly similar to (e.g., at least 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100% identical to) that of an Alphatorquevirus, Betatorquevirus, or Gammatorquevirus. In some embodiments, an Anelloviridae family vector comprises a proteinaceous exterior comprising a protein derived from or similar to (e.g., at least 50%, 60%, 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100% identical to) a capsid protein of an Alphatorquevirus, Betatorquevirus, or Gammatorquevirus (e.g., an Alphatorquevirus ORF1, Betatorquevirus ORF1, or Gammatorquevirus ORF1). In some embodiments, enclosed within a proteinaceous exterior encompasses 100% coverage by a proteinaceous exterior, as well as less than 100% coverage, e.g., 95%, 90%, 85%, 80%, 70%, 60%, 50% or less. For example, gaps or discontinuities (e.g., that render the proteinaceous exterior permeable to water, ions, peptides, or small molecules) may be present in the proteinaceous exterior, so long as the genetic element is retained in the proteinaceous exterior or protected from digestion with DNAse I, e.g., prior to entry into a host cell. In some embodiments, the Anelloviridae family vector is purified, e.g., it is separated from its original source and/or substantially free (>50%, >60%, >70%, >80%, >90%) of other components. In some embodiments, the Anelloviridae family vector is capable of introducing the genetic element into a target cell (e.g., via infection). In some embodiments, the Anelloviridae family vector is an infective synthetic viral particle.


As used herein, the term “anellovector” refers to a vehicle comprising a genetic element, e.g., e.g., circular DNA, enclosed in a proteinaceous exterior. A “synthetic anellovector,” as used herein, generally refers to an anellovector that is not naturally occurring, e.g., has a sequence that is different relative to a wild-type virus (e.g., a wild-type Anellovirus as described herein). In some embodiments, the synthetic anellovector is engineered or recombinant, e.g., comprises a genetic element that comprises a difference or modification relative to a wild-type viral genome (e.g., a wild-type Anellovirus genome as described herein). In some embodiments, enclosed within a proteinaceous exterior encompasses 100% coverage by a proteinaceous exterior, as well as less than 100% coverage, e.g., 95%, 90%, 85%, 80%, 70%, 60%, 50% or less. For example, gaps or discontinuities (e.g., that render the proteinaceous exterior permeable to water, ions, peptides, or small molecules) may be present in the proteinaceous exterior, so long as the genetic element is retained in the proteinaceous exterior, e.g., prior to entry into a host cell. In some embodiments, the anellovector is purified, e.g., it is separated from its original source and/or substantially free (>50%, >60%, >70%, >80%, >90%) of other components.


An anellovector may, in some embodiments, comprise a nucleic acid vector that comprises sufficient nucleic acid sequence derived from or highly similar to (e.g., at least 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100% identical to) an Anellovirus genome sequence or a contiguous portion thereof to allow packaging into a proteinaceous exterior (e.g., a capsid), and further comprises a heterologous sequence. In some embodiments, the nucleic acid vector is a viral vector or a naked nucleic acid. In some embodiments, the nucleic acid vector comprises at least about 50, 60, 70, 71, 72, 73, 74, 75, 80, 90, 100, 150, 200, 300, 400, 500, 600, 700, 800, 900, 1000, 1100, 1200, 1300, 1400, 1500, 1600, 1700, 1800, 1900, 2000, 2500, 3000, or 3500 consecutive nucleotides of a native Anellovirus sequence or a sequence highly similar (e.g., at least 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100% identical) thereto. In some embodiments, the anellovector further comprises one or more of an Anellovirus ORF1, ORF2, or ORF3. In some embodiments, the heterologous sequence comprises a multiple cloning site, comprises a heterologous promoter, comprises a coding region for a therapeutic protein, or encodes a therapeutic nucleic acid. In some embodiments, the capsid is a wild-type Anellovirus capsid. In embodiments, an anellovector comprises a genetic element described herein, e.g., comprises a genetic element comprising a promoter, a sequence encoding a therapeutic effector, and a capsid binding sequence.


As used herein, the term “Anellovirus non-coding region (NCR)” refers to a sequence of an untranslated region of an Anellovirus genome sequence that extends from just upstream of the ORF2 start codon to the untranslated region of the Anellovirus genome sequence just downstream of the ORF3 stop codon in a circular genome. The Anellovirus NCR may comprise the “Anellovirus 5′ NCR sequence” and “Anellovirus 3′ NCR sequence.” In some embodiments, the Anellovirus NCR sequence is contiguous. In some embodiments, the Anellovirus NCR sequence is non-contiguous. In some embodiments, the portions of a non-contiguous Anellovirus NCR sequence are separated by a heterologous insertion (e.g., comprising a recombinase hybrid site, e.g., as described herein). In some embodiments, the Anellovirus NCR sequence comprises a sequence found in a wild-type Anellovirus, and in other embodiments, the Anellovirus NCR comprises one or more mutations relative to the closest Anellovirus sequence. In some embodiments, the Anellovirus NCR is comprised by a nucleic acid molecule that does not comprise Anellovirus ORF2 or ORF3 coding sequences. In some instances, the Anellovirus NCR refers to the sense strand, the antisense strand, or a double-stranded DNA comprising both the sense strand and antisense strands. In some instances, a listing of an Anellovirus NCR sequence herein can refer to the sequence listed and/or its reverse complement.


As used herein, the term “Anellovirus 5′ NCR” refers to a sequence of an untranslated region of an Anellovirus genome sequence that extends from just upstream of the ORF2 start codon through the 5′ UTR conserved domain to the Anellovirus 3′ NCR sequence, and sequences with homology thereto. In some embodiments, an Anellovirus 5′ NCR sequence comprises origin of replication activity. In some embodiments, the Anellovirus 5′ NCR sequence is contiguous. In some embodiments, the Anellovirus 5′ NCR sequence is non-contiguous. In some embodiments, the portions of a non-contiguous Anellovirus 5′ NCR sequence are separated by a heterologous insertion (e.g., comprising a recombinase hybrid site, e.g., as described herein). In some embodiments, the Anellovirus 5′ NCR sequence comprises a sequence found in a wild-type Anellovirus, and in other embodiments, the Anellovirus 5′ NCR comprises one or more mutations relative to the closest Anellovirus sequence. In some instances, the Anellovirus 5′ NCR refers to the sense strand, the antisense strand, or a double-stranded DNA comprising both the sense strand and antisense strands. In some instances, a listing of an Anellovirus 5′ NCR sequence herein can refer to the sequence listed and/or its reverse complement. In a circular genetic element, the Anellovirus 5′ NCR and Anellovirus 3′ NCR may be directly adjacent to each other (e.g., to form an Anellovirus NCR). Exemplary dividing points between the Anellovirus 5′ NCR and the Anellovirus 3′ NCR are shown, e.g., as described herein.


As used herein, the term “Anellovirus 3′ NCR” refer to a sequence of an untranslated region of an Anellovirus genome sequence that extends from just downstream of the ORF3 stop codon through the GC-rich region to the Anellovirus 5′ NCR sequence, and sequences with homology thereto. In some embodiments, the Anellovirus 3′ NCR sequence is contiguous. In some embodiments, the Anellovirus 3′ NCR sequence is non-contiguous. In some embodiments, the portions of a non-contiguous Anellovirus 3′ NCR sequence are separated by a heterologous insertion (e.g., comprising a recombinase hybrid site, e.g., as described herein). In some embodiments, the Anellovirus 3′ NCR sequence comprises a sequence found in a wild-type Anellovirus, and in other embodiments, the Anellovirus 3′ NCR comprises one or more mutations relative to the closest Anellovirus sequence. In some instances, the Anellovirus 3′ NCR refers to the sense strand, the antisense strand, or a double-stranded DNA comprising both the sense strand and antisense strands. In some instances, a listing of an Anellovirus 3′ NCR sequence herein can refer to the sequence listed and/or its reverse complement.


As used herein, the term “Anellovirus GC-rich region” refers to a wild-type or engineered sequence that has an activity and a structural feature of a GC-rich region of a wild-type Anellovirus, or a functional fragment thereof. In some embodiments, the functional fragment has a length of at least 20, 30, 40, 50, 60, 70, 80, 90, or 100 nucleotides. Typically, the negative strand comprising the Anellovirus GC-rich region is packaged into a particle (e.g., an Anelloviridae family vector) as described herein. In some embodiments, the Anellovirus GC-rich region is a wild-type Anellovirus GC-rich region. In some embodiments, the Anellovirus GC-rich region is an engineered Anellovirus GC-rich region having a nucleic acid sequence with at least one difference relative to the closest wild-type Anellovirus GC-rich region sequence.


As used herein, the term “Anellovirus 5′ UTR conserved domain” refers to a wild-type or engineered sequence that has an activity and a structural feature of an Anellovirus 5′ UTR conserved domain of a wild-type Anellovirus, or a functional fragment thereof. In some embodiments, the functional fragment has a length of at least 15, 20, 30, 40, 50, 60, or 70 nucleotides. Typically, the negative strand comprising the Anellovirus 5′ UTR conserved domain is packaged into a particle (e.g., an Anelloviridae family vector) as described herein. In some embodiments, the Anellovirus 5′ UTR conserved domain is a wild-type Anellovirus 5′ UTR conserved domain. In some embodiments, the Anellovirus 5′ UTR conserved domain is an engineered Anellovirus 5′ UTR conserved domain having a nucleic acid sequence with at least one difference relative to the closest wild-type Anellovirus 5′ UTR conserved domain sequence.


As used herein, the term “antibody molecule” refers to a protein, e.g., an immunoglobulin chain or fragment thereof, comprising at least one immunoglobulin variable domain sequence. The term “antibody molecule” encompasses full-length antibodies and antibody fragments (e.g., scFvs). In some embodiments, an antibody molecule is a multispecific antibody molecule, e.g., the antibody molecule comprises a plurality of immunoglobulin variable domain sequences, wherein a first immunoglobulin variable domain sequence of the plurality has binding specificity for a first epitope and a second immunoglobulin variable domain sequence of the plurality has binding specificity for a second epitope. In embodiments, the multispecific antibody molecule is a bispecific antibody molecule. A bispecific antibody molecule is generally characterized by a first immunoglobulin variable domain sequence which has binding specificity for a first epitope and a second immunoglobulin variable domain sequence that has binding specificity for a second epitope.


The term “in vitro assembly,” as used herein with respect to an Anelloviridae family vector or an anelloVLP, refers to the formation of a proteinaceous exterior comprising an ORF1 molecule, wherein the formation does not take place inside of a cell (e.g., takes place in a cell-free system such as a cell-free suspension, a lysate, or a supernatant). In some instances, in vitro assembly of an Anelloviridae family vector comprises enclosure, outside of a cell, of a genetic element (e.g., as described herein) within the proteinaceous exterior. In some instances, in vitro assembly of an anelloVLP comprises association, outside of a cell, of an effector (e.g., an exogenous effector, e.g., as described herein) with the proteinaceous exterior (e.g., enclosed within the proteinaceous exterior). In vitro assembly of a proteinaceous exterior may occur, in some instances, under conditions suitable for multimerization of a plurality of ORF1 molecules (e.g., nondenaturing conditions), e.g., to form a multimer of more than 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20 ORF1 molecules. In some instances, in vitro assembly results in the formation of a proteinaceous exterior comprising at least about 20, 30, 40, 50, or 60 ORF1 molecules, or about 20-30, 30-40, 40-50, 50-60, or 60-70 ORF1 molecules). In some instances, the proteinaceous exterior is formed from ORF1 molecules that were produced in a cell and then purified therefrom. In some instances, the in vitro assembly takes place in a solution free of cells or constituents thereof. In other instances, the in vitro assembly takes place in a solution comprising cell debris (e.g., from lysed cells). In some instances, the in vitro assembly takes place in a solution substantially free of cellular nucleic acid molecules (e.g., genomic DNA, mitochondrial DNA, mRNA, and/or noncoding RNA from a cell).


As used herein, a nucleic acid “encoding” refers to a nucleic acid sequence encoding an amino acid sequence or a functional polynucleotide (e.g., a non-coding RNA, e.g., an siRNA or miRNA).


An “exogenous” agent (e.g., an effector, a nucleic acid (e.g., RNA), a gene, payload, protein) as used herein refers to an agent that is either not comprised by, or not encoded by, a corresponding wild-type virus, e.g., an Anellovirus as described herein. In some embodiments, the exogenous agent does not naturally exist, such as a protein or nucleic acid that has a sequence that is altered (e.g., by insertion, deletion, or substitution) relative to a naturally occurring protein or nucleic acid. In some embodiments, the exogenous agent does not naturally exist in the host cell. In some embodiments, the exogenous agent exists naturally in the host cell but is exogenous to the virus. In some embodiments, the exogenous agent exists naturally in the host cell, but is not present at a desired level or at a desired time.


A “heterologous” agent or element (e.g., an effector, a nucleic acid sequence, an amino acid sequence), as used herein with respect to another agent or element (e.g., an effector, a nucleic acid sequence, an amino acid sequence), refers to agents or elements that are not naturally found together, e.g., in a wild-type virus, e.g., an Anellovirus. In some embodiments, a heterologous nucleic acid sequence may be present in the same nucleic acid as a naturally occurring nucleic acid sequence (e.g., a sequence that is naturally occurring in the Anellovirus). In some embodiments, a heterologous agent or element is exogenous relative to an Anellovirus from which other (e.g., the remainder of) elements of the anellovector are based.


As used herein, the term “genetic element” refers to a nucleic acid sequence, generally in an anellovector. It is understood that the genetic element can be produced as naked DNA and optionally further assembled into a proteinaceous exterior. It is also understood that an anellovector can insert its genetic element into a cell, resulting in the genetic element being present in the cell and the proteinaceous exterior not necessarily entering the cell.


As used herein, the term “ORF1 molecule” refers to a polypeptide having an activity and/or a structural feature of an Anellovirus ORF1 protein (e.g., an Anellovirus ORF1 protein as described herein, e.g., as listed in Table A1-A3), or a functional fragment thereof. An ORF1 molecule may, in some instances, comprise one or more of (e.g., 1, 2, 3 or 4 of): a first region comprising at least 60% basic residues (e.g., at least 60% arginine residues), a second region comprising at least about six beta strands (e.g., at least 4, 5, 6, 7, 8, 9, 10, 11, or 12 beta strands), a third region comprising a structure or an activity of an Anellovirus N22 domain (e.g., as described herein, e.g., an N22 domain from an Anellovirus ORF1 protein as described herein), and/or a fourth region comprising a structure or an activity of an Anellovirus C-terminal domain (CTD) (e.g., as described herein, e.g., a CTD from an Anellovirus ORF1 protein as described herein). In some instances, the ORF1 molecule comprises, in N-terminal to C-terminal order, the first, second, third, and fourth regions. In some instances, an anellovector comprises an ORF1 molecule comprising, in N-terminal to C-terminal order, the first, second, third, and fourth regions. An ORF1 molecule may, in some instances, comprise a polypeptide encoded by an Anellovirus ORF1 nucleic acid (e.g., as listed in any of Tables N1-N3). An ORF1 molecule may, in some instances, further comprise a heterologous sequence, e.g., a hypervariable region (HVR), e.g., an HVR from an Anellovirus ORF1 protein, e.g., as described herein. An “Anellovirus ORF1 protein,” as used herein, refers to an ORF1 protein encoded by an Anellovirus genome (e.g., a wild-type Anellovirus genome, e.g., as described herein), e.g., an ORF1 protein having the amino acid sequence as listed in Table A1-A3, or as encoded by the ORF1 gene as listed in any of Tables N1-N3.


The term “ORF1 domain,” as used herein with respect to an ORF1 molecule, refers to the portion of the ORF1 molecule having the structure or function of an Anellovirus ORF1 protein. The ORF1 domain is generally capable of forming a multimer with other copies of the ORF1 domain (e.g., in other ORF1 molecules), or with other ORF1 molecules, e.g., to form a proteinaceous exterior (e.g., of an anellovector or anelloVLP as described herein). In some instances, the ORF1 molecule may comprise one or more additional domains other than the ORF1 domain (for example, a domain comprising or attached to a surface effector). In some instances, the amino acid sequence of an ORF1 domain comprises an insertion (e.g., an insertion encoding a surface moiety or a domain capable of binding to a surface moiety), e.g., between the N-terminal end and C-terminal end of the ORF1 domain. In certain instances, the insertion does not substantially disrupt the structure and/or function of the ORF1 domain, e.g., such that the ORF1 domain remains capable of forming a multimer with other ORF1 domains or ORF1 molecules. The position within the ORF1 domain sequence into which the insertion is made is referred to herein as the “insertion point.” An insertion can be made into an ORF1 domain by any genetic or polypeptide engineering method known in the art. In some embodiments, an ORF1 molecule consists of an ORF1 domain. In other embodiments, an ORF1 molecule comprises an ORF1 domain and a heterologous domain (e.g., a surface moiety as described herein). In some embodiments, an ORF1 domain is connected to a surface moiety by a polypeptide linker region.


As used herein, the term “ORF2 molecule” refers to a polypeptide having an activity and/or a structural feature of an Anellovirus ORF2 protein (e.g., an Anellovirus ORF2 protein as described herein, e.g., as listed in Table A1-A3), or a functional fragment thereof. An “Anellovirus ORF2 protein,” as used herein, refers to an ORF2 protein encoded by an Anellovirus genome (e.g., a wild-type Anellovirus genome, e.g., as described herein), e.g., an ORF2 protein having the amino acid sequence as listed in Table A1-A3, or as encoded by the ORF2 gene as listed in any of Tables N1-N3.


As used herein, the term “particle” refers to a vehicle having a diameter of less than 100 nm (e.g., about 20-25, 25-30, 30-35, or 35-40 nm) comprising a proteinaceous exterior. In some instances, the particle comprises a plurality of ORF1 molecules. The proteinaceous exterior of the particle generally forms an enclosure capable of limiting or preventing movement of certain molecules between the inside and outside of the proteinaceous exterior. In some embodiments, gaps or discontinuities (e.g., that render the proteinaceous exterior permeable to water, ions, peptides, or small molecules) may be present in the proteinaceous exterior. In certain embodiments, the gaps or discontinuities are of a sufficiently small size (e.g., diameter) that the proteinaceous exterior limits or prevents one or more large macromolecules (e.g., peptides, polypeptides, polynucleotides, lipids, or polysaccharides) from passing through the proteinaceous exterior.


As used herein, the term “proteinaceous exterior” refers to an exterior component that is predominantly (e.g., >50%, >60%, >70%, >80%, >90%) protein.


As used herein, the term “regulatory nucleic acid” refers to a nucleic acid sequence that modifies expression, e.g., transcription and/or translation, of a DNA sequence that encodes an expression product. In embodiments, the expression product comprises RNA or protein.


As used herein, the term “regulatory sequence” refers to a nucleic acid sequence that modifies transcription of a target gene product. In some embodiments, the regulatory sequence is a promoter or an enhancer.


As used herein, the term “replication protein” refers to a protein, e.g., a viral protein, that is utilized during infection, viral genome replication/expression, viral protein synthesis, and/or assembly of the viral components.


As used herein, a “substantially non-pathogenic” organism, particle, or component, refers to an organism, particle (e.g., a virus or an anellovector, e.g., as described herein), or component thereof that does not cause or induce a detectable disease or pathogenic condition, e.g., in a host organism, e.g., a mammal, e.g., a human. In some embodiments, administration of an anellovector to a subject can result in minor reactions or side effects that are acceptable as part of standard of care.


As used herein, the term “non-pathogenic” refers to an organism or component thereof that does not cause or induce a detectable disease or pathogenic condition, e.g., in a host organism, e.g., a mammal, e.g., a human.


As used herein, a “substantially non-integrating” genetic element refers to a genetic element, e.g., a genetic element in a virus or anellovector, e.g., as described herein, wherein less than about 0.01%, 0.05%, 0.1%, 0.5%, or 1% of the genetic element that enter into a host cell (e.g., a eukaryotic cell) or organism (e.g., a mammal, e.g., a human) integrate into the genome. In some embodiments the genetic element does not detectably integrate into the genome of, e.g., a host cell. In some embodiments, integration of the genetic element into the genome can be detected using techniques as described herein, e.g., nucleic acid sequencing, PCR detection and/or nucleic acid hybridization.


As used herein, a “substantially non-immunogenic” organism, particle, or component, refers to an organism, particle (e.g., a virus or anellovector, e.g., as described herein), or component thereof, that does not cause or induce an undesired or untargeted immune response, e.g., in a host tissue or organism (e.g., a mammal, e.g., a human). In some embodiments, the substantially non-immunogenic organism, particle, or component does not produce a detectable immune response. In some embodiments, the substantially non-immunogenic anellovector does not produce a detectable immune response against a protein comprising an amino acid sequence or encoded by a nucleic acid sequence shown in any of Tables N1-N3. In some embodiments, an immune response (e.g., an undesired or untargeted immune response) is detected by assaying antibody presence or level (e.g., presence or level of an anti-anellovector antibody, e.g., presence or level of an antibody against an anellovector as described herein) in a subject, e.g., according to the anti-TTV antibody detection method described in Tsuda et al. (1999; J. Virol. Methods 77: 199-206; incorporated herein by reference) and/or the method for determining anti-TTV IgG levels described in Kakkola et al. (2008; Virology 382: 182-189; incorporated herein by reference). Antibodies against an Anellovirus or an anellovector based thereon can also be detected by methods in the art for detecting anti-viral antibodies, e.g., methods of detecting anti-AAV antibodies, e.g., as described in Calcedo et al. (2013; Front. Immunol. 4(341): 1-7; incorporated herein by reference).


A “subsequence” as used herein refers to a nucleic acid sequence or an amino acid sequence that is comprised in a larger nucleic acid sequence or amino acid sequence, respectively. In some instances, a subsequence may comprise a domain or functional fragment of the larger sequence. In some instances, the subsequence may comprise a fragment of the larger sequence capable of forming secondary and/or tertiary structures when isolated from the larger sequence similar to the secondary and/or tertiary structures formed by the subsequence when present with the remainder of the larger sequence. In some instances, a subsequence can be replaced by another sequence (e.g., a subsequence comprising an exogenous sequence or a sequence heterologous to the remainder of the larger sequence, e.g., a corresponding subsequence from a different Anellovirus).


As used herein, the term “surface moiety” refers to a moiety for which at least a portion is exposed on the exterior surface of a particle (e.g., exposed to the solution surrounding the particle). The surface moiety is generally attached, directly or indirectly, to a component of the proteinaceous exterior of the particle (e.g., an ORF1 molecule). In some instances, the surface moiety is covalently attached to the component of the proteinaceous exterior of the particle (e.g., the ORF1 molecule). In some instances, the surface moiety is noncovalently attached to the component of the proteinaceous exterior of the particle (e.g., the ORF1 molecule). In some instances, the surface moiety is bound to a binding moiety that is in turn attached (e.g., covalently or noncovalently) to the component of the proteinaceous exterior of the particle (e.g., the ORF1 molecule). In some instances, the surface moiety is comprised in an ORF1 molecule (e.g., is a heterologous domain of an ORF1 molecule). In some instances, a surface moiety is exogenous relative to an Anellovirus (e.g., the Anellovirus from which the ORF1 molecule was derived and/or an Anellovirus for which the ORF1 protein has at least 50%, 60%, 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to the ORF1 molecule). In some instances, a surface moiety is exogenous relative a target cell (e.g., a mammalian cell, e.g., a human cell) to be infected by the particle.


As used herein, “treatment”, “treating” and cognates thereof refer to the medical management of a subject with the intent to improve, ameliorate, stabilize, prevent or cure a disease, pathological condition, or disorder. This term includes active treatment (treatment directed to improve the disease, pathological condition, or disorder), causal treatment (treatment directed to the cause of the associated disease, pathological condition, or disorder), palliative treatment (treatment designed for the relief of symptoms), preventative treatment (treatment directed to preventing, minimizing or partially or completely inhibiting the development of the associated disease, pathological condition, or disorder); and supportive treatment (treatment employed to supplement another therapy).


This invention relates generally to Anelloviridae family vectors (e.g., anellovectors), e.g., synthetic Anelloviridae family vectors (e.g., anellovectors), and uses thereof. The present disclosure provides Anelloviridae family vectors (e.g., anellovectors), compositions comprising Anelloviridae family vectors (e.g., anellovectors), and methods of making or using Anelloviridae family vectors (e.g., anellovectors). Anelloviridae family vectors (e.g., anellovectors) are generally useful as delivery vehicles, e.g., for delivering a therapeutic agent to a eukaryotic cell. Generally, an Anelloviridae family vector (e.g., anellovector) will include a genetic element comprising a nucleic acid sequence (e.g., encoding an effector, e.g., an exogenous effector or an endogenous effector) enclosed within a proteinaceous exterior. An Anelloviridae family vector (e.g., anellovector) may include one or more deletions of sequences (e.g., regions or domains as described herein) relative to an Anellovirus sequence (e.g., as described herein). Anelloviridae family vectors (e.g., anellovectors) can be used as a substantially non-immunogenic vehicle for delivering the genetic element, or an effector encoded therein (e.g., a polypeptide or nucleic acid effector, e.g., as described herein), into eukaryotic cells, e.g., to treat a disease or disorder in a subject comprising the cells.


TABLE OF CONTENTS





    • I. Anelloviridae Family Vectors (e.g., Anellovectors)
      • A. Anelloviridae Family Viruses (e.g., Anelloviruses)
        • i. Nucleic acid sequences
        • ii. Amino acid sequences encoded by nucleic acid sequences
        • iii. Proteins comprising amino acid sequences
        • iv. Polypeptides comprising amino acid sequences
      • B. Capsid Proteins (e.g., ORF1 molecules)
        • i. Conserved ORF1 motif in N22 domain
        • ii. Exemplary ORF1 sequences
        • iii. Identification of ORF1 protein sequences
      • C. ORF2 molecules
        • i. Conserved ORF2 motif
      • D. Genetic elements
      • E. Protein binding sequence
      • F. 5′ UTR Conserved Domains
      • G. GC-rich regions
      • H. Effectors
      • I. Surface Moieties

    • II. Compositions and Methods for Making Anelloviridae Family Vectors
      • A. Genetic Element Constructs
        • i. Tandem constructs
        • ii. Cis/trans constructs
      • B. Recombinase-based production of genetic elements and Anellovectors
        • i. Self-replicating rescue (SRR) constructs (e.g., SRR plasmids)
        • ii. Exemplary site-specific recombinases and recombinase recognition sites
      • C. Host Cells and methods of using host cells for producing an Anellovector
        • i. Introduction of genetic elements into host cells
        • ii. Exemplary cell types
      • D. Culture Conditions
      • E. Harvest
      • F. In vitro assembly methods
      • G. Enrichment and Purification

    • III. Pharmaceutical Compositions

    • IV. Methods of use

    • V. Redosing





I. Anelloviridae Family Vectors (e.g., Anellovectors)

In some aspects, the invention described herein comprises compositions and methods of using and making an Anelloviridae family vector (e.g., anellovector), Anelloviridae family vector (e.g., anellovector) preparations, and therapeutic compositions. In some embodiments, the anellovector has a sequence, structure, and/or function that is based on an Anelloviridae virus (e.g., an Anellovirus as described herein). It is understood that applicable embodiments described herein with respect to anellovectors may also be applied to Anelloviridae family vectors (e.g., a vector based on or derived from a chicken anemia virus (CAV), e.g., as described herein). In some embodiments, the Anelloviridae family vector (e.g., anellovector) comprises a nucleic acid or polypeptide comprising a sequence as shown in Table A1-A3 (e.g., Table A1, A2, or A3); or Table N1-N3 (e.g., Table N1, N2, or N3), or fragments or portions thereof, or other substantially non-pathogenic virus, e.g., a symbiotic virus, commensal virus, native virus. In some embodiments, an Anelloviridae family virus-based vector comprises at least one element exogenous to that Anelloviridae family virus, e.g., an exogenous effector or a nucleic acid sequence encoding an exogenous effector disposed within a genetic element of the vector. In some embodiments, an Anelloviridae family virus-based vector comprises at least one element heterologous to another element from that Anelloviridae family virus, e.g., an effector-encoding nucleic acid sequence that is heterologous to another linked nucleic acid sequence, such as a promoter element. In some embodiments, an Anelloviridae family vector comprises a genetic element (e.g., circular DNA, e.g., single stranded DNA), which comprise at least one element that is heterologous relative to the remainder of the genetic element and/or the proteinaceous exterior (e.g., an exogenous element encoding an effector, e.g., as described herein). An Anelloviridae family vector may be a delivery vehicle (e.g., a substantially non-pathogenic delivery vehicle) for a payload into a host, e.g., a human. In some embodiments, the Anelloviridae family vector is capable of replicating in a eukaryotic cell, e.g., a mammalian cell, e.g., a human cell. In some embodiments, the Anelloviridae family vector is substantially non-pathogenic and/or substantially non-integrating in the mammalian (e.g., human) cell. In some embodiments, the Anelloviridae family vector is substantially non-immunogenic in a mammal, e.g., a human. In some embodiments, the Anelloviridae family vector is replication-deficient. In some embodiments, the Anelloviridae family vector is replication-competent.


In one aspect, the invention includes an Anelloviridae family vector comprising:

    • a) a genetic element comprising (i) a sequence encoding an exterior protein (e.g., a non-pathogenic exterior protein), (ii) an exterior protein binding sequence that binds the genetic element to the non-pathogenic exterior protein, and (iii) a sequence encoding an effector (e.g., an endogenous or exogenous effector); and
    • b) a proteinaceous exterior that is associated with, e.g., envelops or encloses, the genetic element.


In some embodiments, the Anelloviridae family vector (e.g. anellovector) includes sequences or expression products from (or having >70%, 75%, 80%, 85%, 90%, 95%, 97%, 98%, 99%, 100% homology to) a non-enveloped, circular, single-stranded DNA virus. Animal circular single-stranded DNA viruses generally refer to a subgroup of single strand DNA (ssDNA) viruses, which infect eukaryotic non-plant hosts, and have a circular genome. Thus, animal circular ssDNA viruses are distinguishable from ssDNA viruses that infect prokaryotes (i.e. Microviridae and Inoviridae) and from ssDNA viruses that infect plants (i.e. Geminiviridae and Nanoviridae). They are also distinguishable from linear ssDNA viruses that infect non-plant eukaryotes (i.e. Parvoviridiae).


In some embodiments, the genetic element comprises a promoter element. In some embodiments, the promoter element is selected from an RNA polymerase II-dependent promoter, an RNA polymerase III-dependent promoter, a PGK promoter, a CMV promoter, an EF-1α promoter, an SV40 promoter, a CAGG promoter, or a UBC promoter, TTV viral promoters, Tissue specific, U6 (pollIII), minimal CMV promoter with upstream DNA binding sites for activator proteins (TetR-VP16, Gal4-VP16, dCas9-VP16, etc.). In some embodiments, the promoter element comprises a TATA box. In some embodiments, the promoter element is endogenous to a wild-type Anelloviridae family virus (e.g., Anellovirus), e.g., as described herein.


In some embodiments, the genetic element comprises one or more of the following characteristics: single-stranded, circular, negative strand, and/or DNA. In some embodiments, the portions of the genetic element excluding the effector have a combined size of about 2.5-5 kb (e.g., about 2.8-4 kb, about 2.8-3.2 kb, about 3.6-3.9 kb, or about 2.8-2.9 kb), less than about 5 kb (e.g., less than about 2.9 kb, 3.2 kb, 3.6 kb, 3.9 kb, or 4 kb), or at least 100 nucleotides (e.g., at least 1 kb).


In some embodiments, a replication deficient, replication defective, or replication incompetent genetic element does not encode all of the necessary machinery or components required for replication of the genetic element. In some embodiments, a replication defective genetic element does not encode a replication factor. In some embodiments, a replication defective genetic element does not encode one or more ORFs (e.g., ORF1, ORF1/1, ORF1/2, ORF2, ORF2/2, ORF2/3, and/or ORF2t/3 e.g., as described herein). In some embodiments, the machinery or components not encoded by the genetic element may be provided in trans (e.g., using a helper, e.g., a helper virus or helper plasmid, or encoded in a nucleic acid comprised by the host cell, e.g., integrated into the genome of the host cell), e.g., such that the genetic element can undergo replication in the presence of the machinery or components provided in trans.


In some embodiments, a packaging deficient, packaging defective, or packaging incompetent genetic element cannot be packaged into a proteinaceous exterior (e.g., wherein the proteinaceous exterior comprises a capsid or a portion thereof, e.g., comprising a polypeptide encoded by an ORF1nucleic acid, e.g., as described herein). In some embodiments, a packaging deficient genetic element is packaged into a proteinaceous exterior at an efficiency less than 10% (e.g., less than 10%, 9%, 8%, 7%, 6%, 5%, 4%, 3%, 2%, 1%, 0.5%, 0.1%, 0.01%, or 0.001%) compared to a wild-type Anelloviridae family virus (e.g., Anellovirus) (e.g., as described herein). In some embodiments, the packaging defective genetic element cannot be packaged into a proteinaceous exterior even in the presence of factors (e.g., ORF1, ORF1/1, ORF1/2, ORF2, ORF2/2, ORF2/3, or ORF2t/3) that would permit packaging of the genetic element of a wild-type Anelloviridae family virus (e.g., Anellovirus) (e.g., as described herein). In some embodiments, a packaging deficient genetic element is packaged into a proteinaceous exterior at an efficiency less than 10% (e.g., less than 10%, 9%, 8%, 7%, 6%, 5%, 4%, 3%, 2%, 1%, 0.5%, 0.1%, 0.01%, or 0.001%) compared to a wild-type Anelloviridae family virus (e.g., Anellovirus) (e.g., as described herein), even in the presence of factors (e.g., ORF1, ORF1/1, ORF1/2, ORF2, ORF2/2, ORF2/3, or ORF2t/3) that would permit packaging of the genetic element of a wild-type Anelloviridae family virus (e.g., Anellovirus) (e.g., as described herein).


In some embodiments, a packaging competent genetic element can be packaged into a proteinaceous exterior (e.g., wherein the proteinaceous exterior comprises a capsid or a portion thereof, e.g., comprising a polypeptide encoded by an ORF1nucleic acid, e.g., as described herein). In some embodiments, a packaging competent genetic element is packaged into a proteinaceous exterior at an efficiency of at least 20% (e.g., at least 20%, 30%, 40%, 50%, 60%, 70%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, 100%, or higher) compared to a wild-type Anelloviridae family virus (e.g., Anellovirus) (e.g., as described herein). In some embodiments, the packaging competent genetic element can be packaged into a proteinaceous exterior in the presence of factors (e.g., ORF1, ORF1/1, ORF1/2, ORF2, ORF2/2, ORF2/3, or ORF2t/3) that would permit packaging of the genetic element of a wild-type Anelloviridae family virus (e.g., Anellovirus) (e.g., as described herein). In some embodiments, a packaging competent genetic element is packaged into a proteinaceous exterior at an efficiency of at least 20% (e.g., at least 20%, 30%, 40%, 50%, 60%, 70%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, 100%, or higher) compared to a wild-type Anelloviridae family virus (e.g., Anellovirus) (e.g., as described herein) in the presence of factors (e.g., ORF1, ORF1/1, ORF1/2, ORF2, ORF2/2, ORF2/3, or ORF2t/3) that would permit packaging of the genetic element of a wild-type Anelloviridae family virus (e.g., Anellovirus) (e.g., as described herein).


Anelloviridae Family Viruses (e.g., Anelloviruses)

In some embodiments, an Anelloviridae family vector, e.g., as described herein, comprises sequences or expression products derived from an Anellovirus. In some embodiments, an Anelloviridae family vector includes one or more sequences or expression products that are exogenous relative to the Anellovirus. In some embodiments, an Anelloviridae family vector includes one or more sequences or expression products that are endogenous relative to the Anellovirus. In some embodiments, an Anelloviridae family vector includes one or more sequences or expression products that are heterologous relative to one or more other sequences or expression products in the Anelloviridae family vector. Anelloviridae family viruses (e.g., Anellovirus) generally have single-stranded circular DNA genomes with negative polarity.


It is understood that applicable embodiments described herein with respect to anellovectors may also be applied to Anelloviridae family vectors (e.g., a vector based on or derived from a chicken anemia virus (CAV), e.g., as described herein). Examples of chicken anemia viruses, and compositions and uses thereof, are described in PCT Publication No. WO/2022/094238, incorporated herein by reference in its entirety, including the sequences of Tables 1A and 1B therein.


In some embodiments, the genetic element comprises a nucleotide sequence encoding an amino acid sequence or a functional fragment thereof or a sequence having at least about 60%, 70% 80%, 85%, 90% 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to any one of the amino acid sequences described herein, e.g., an Anellovirus amino acid sequence.


In some embodiments, an Anelloviridae family vector as described herein comprises one or more nucleic acid molecules (e.g., a genetic element as described herein) comprising a sequence having at least about 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to an Anellovirus sequence, e.g., as described herein, or a fragment thereof. In embodiments, the Anelloviridae family vector comprises a nucleic acid sequence selected from a sequence as shown in any of Tables N1-N3, or a sequence having at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity thereto. In embodiments, the Anelloviridae family vector comprises a polypeptide comprising a sequence as shown in Table A1-A3, or a sequence having at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity thereto.


In some embodiments, an Anelloviridae family vector as described herein comprises one or more nucleic acid molecules (e.g., a genetic element as described herein) comprising a sequence having at least about 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to one or more of a TATA box, cap site, initiator element, transcriptional start site, 5′ UTR conserved domain, ORF1, ORF1/1, ORF1/2, ORF2, ORF2/2, ORF2/3, ORF2t/3, three open-reading frame region, poly(A) signal, GC-rich region, or any combination thereof, of any of the Anelloviridae family viruses (e.g., Anellovirus) described herein (e.g., an Anelloviridae family virus (e.g., Anellovirus) sequence as annotated, or as encoded by a sequence listed, in any of Tables N1-N3. In some embodiments, the nucleic acid molecule comprises a sequence encoding a capsid protein, e.g., an ORF1, ORF1/1, ORF1/2, ORF2, ORF2/2, ORF2/3, or ORF2t/3sequence of any of the Anelloviruses described herein (e.g., an Anelloviridae family virus (e.g., Anellovirus) sequence as annotated, or as encoded by a sequence listed, in any of Tables N1-N3). In some embodiments, the nucleic acid molecule comprises a sequence encoding a capsid protein comprising an amino acid sequence having at least about 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to an Anelloviridae family virus (e.g., Anellovirus) ORF1 or ORF2 protein (e.g., an ORF1 or ORF2 amino acid sequence as shown in Table A1-A3, or an ORF1 or ORF2 amino acid sequence encoded by a nucleic acid sequence as shown in any of Tables N1-N3). In embodiments, the nucleic acid molecule comprises a sequence encoding a capsid protein comprising an amino acid sequence having at least about 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to an Anelloviridae family virus (e.g., Anellovirus) ORF1 protein (e.g., an ORF1 amino acid sequence as shown in Table A1-A3, or an ORF1 amino acid sequence encoded by a nucleic acid sequence as shown in any of Tables N1-N3).


Nucleic Acid Sequences

In some embodiments, the nucleic acid molecule comprises a nucleic acid sequence having at least about 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to the Anelloviridae family virus (e.g., Anellovirus) ORF1 nucleotide sequence of any of Tables N1-N3. In some embodiments, the nucleic acid molecule comprises a nucleic acid sequence having at least about 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to the Anelloviridae family virus (e.g., Anellovirus) ORF2 nucleotide sequence of any of Tables N1-N3. In some embodiments, the nucleic acid molecule comprises a nucleic acid sequence having at least about 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to the Anelloviridae family virus (e.g., Anellovirus) ORF3 nucleotide sequence of any of Tables N1-N3. In some embodiments, the nucleic acid molecule comprises a nucleic acid sequence having at least about 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to the Anelloviridae family virus (e.g., Anellovirus) GC-rich region nucleotide sequence of any of Tables N1-N3. In some embodiments, the nucleic acid molecule comprises a nucleic acid sequence having at least about 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to the Anelloviridae family virus (e.g., Anellovirus) 5′ UTR conserved domain nucleotide sequence of any of Tables N1-N3.


It is understood that Tables N1-N3 herein provide the positive strand sequence corresponding to a particular Anellovirus. However, as described herein, a genetic element is typically a negative strand (e.g., comprising the reverse complement of a nucleic acid sequence as listed in any of Tables N1-N3, or a portion thereof). Consequently, a 5′ UTR conserved domain of a genetic element as described herein may comprise the reverse complement of a sequence annotated as a 5′ UTR conserved domain (e.g., in any of Tables N1-N3). Consequently, a GC-rich region of a genetic element as described herein may comprise the reverse complement of a sequence annotated as a GC-rich region (e.g., in any of Tables N1-N3).


In some embodiments, the Anellovirus is an Alphatorquevirus. In some embodiments, the Anellovirus is a Betatorquevirus. In some embodiments, the Anellovirus is a Gammatorquevirus.


In some embodiments, the Anellovirus is a Ring1, Ring3.1, Ring4, Ring5.2, Ring6.0, Ring7, Ring9, Ring10, or Ring20 Anellovirus.


The Ring1 Anellovirus genomic sequence is disclosed as SEQ ID NO: 16 of International Application PCT/US2021/037076, and the corresponding amino acid sequences are disclosed in Table A2 of the same International Application, and the sequences are herein incorporated by reference in their entireties. In some embodiments, the genetic element comprises a nucleic acid sequence having at least about 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to the reverse complement of the sequence annotated as the GC-rich region of SEQ ID NO: 16 of International Application PCT/US2021/037076. In some embodiments, the genetic element comprises a nucleic acid sequence having at least about 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to the reverse complement of the sequence annotated as the 5′ UTR conserved domain nucleotide sequence of SEQ ID NO: 16 of International Application PCT/US2021/037076. The Ring3.1 Anellovirus genomic sequence is disclosed as SEQ ID NO: 878 of International Application PCT/US2022/015499, and the corresponding amino acid sequences are disclosed in Table B4 of the same International Application, and the sequences are herein incorporated by reference in their entireties. In some embodiments, the genetic element comprises a nucleic acid sequence having at least about 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to the reverse complement of the sequence annotated as the GC-rich region nucleotide sequence of SEQ ID NO: 878 of International Application PCT/US2022/015499. In some embodiments, the genetic element comprises a nucleic acid sequence having at least about 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to the reverse complement of the sequence annotated as the 5′ UTR conserved domain nucleotide sequence of SEQ ID NO: 878 of International Application PCT/US2022/015499. The Ring4 Anellovirus genomic sequence is disclosed as SEQ ID NO: 886 of International Application PCT/US2021/037076, and the corresponding amino acid sequences are disclosed in Table C2 of the same International Application, and the sequences are herein incorporated by reference in their entireties. In some embodiments, the genetic element comprises a nucleic acid sequence having at least about 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to the reverse complement of the sequence annotated as the GC-rich region nucleotide sequence of SEQ ID NO: 886 of International Application PCT/US2021/037076. In some embodiments, the genetic element comprises a nucleic acid sequence having at least about 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to the reverse complement of the sequence annotated as the 5′ UTR conserved domain nucleotide sequence of SEQ ID NO: 886 of International Application PCT/US2021/037076. The Ring5.2 Anellovirus genomic sequence is disclosed as SEQ ID NO: 894 of International Application PCT/US2022/015499, and the corresponding amino acid sequences are disclosed in Table D2 of the same International Application, and the sequences are herein incorporated by reference in their entireties. In some embodiments, the genetic element comprises a nucleic acid sequence having at least about 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to the reverse complement of the sequence annotated as the GC-rich region nucleotide sequence of SEQ ID NO: 894 of International Application PCT/US2022/015499. In some embodiments, the genetic element comprises a nucleic acid sequence having at least about 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to the reverse complement of the sequence annotated as the 5′ UTR conserved domain nucleotide sequence of SEQ ID NO: 894 of International Application PCT/US2022/015499. The Ring6.0 Anellovirus genomic sequence is disclosed as SEQ ID NO: 903 of International Application PCT/US2019/065995, and the corresponding amino acid sequences are disclosed in Table C4 of the same International Application, and the sequences are herein incorporated by reference in their entireties. In some embodiments, the genetic element comprises a nucleic acid sequence having at least about 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to the reverse complement of the sequence annotated as the GC-rich region nucleotide sequence of SEQ ID NO: 903 of International Application PCT/US2019/065995. In some embodiments, the genetic element comprises a nucleic acid sequence having at least about 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to the reverse complement of the sequence annotated as the 5′ UTR conserved domain nucleotide sequence of SEQ ID NO: 903 of International Application PCT/US2019/065995. The Ring7 Anellovirus genomic sequence is disclosed as SEQ ID NO: 911 of International Application PCT/US2019/065995, and the corresponding amino acid sequences are disclosed in Table C5 of the same International Application, and the sequences are herein incorporated by reference in their entireties. In some embodiments, the genetic element comprises a nucleic acid sequence having at least about 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to the reverse complement of the sequence annotated as the GC-rich region nucleotide sequence of SEQ ID NO: 911 of International Application PCT/US2019/065995. In some embodiments, the genetic element comprises a nucleic acid sequence having at least about 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to the reverse complement of the sequence annotated as the 5′ UTR conserved domain nucleotide sequence of SEQ ID NO: 911 of International Application PCT/US2019/065995. The Ring9 Anellovirus genomic sequence is disclosed as SEQ ID NO: 1001 of International Application PCT/US2022/015499, and the corresponding amino acid sequences are disclosed in Table F2 of the same International Application, and the sequences are herein incorporated by reference in their entireties. In some embodiments, the genetic element comprises a nucleic acid sequence having at least about 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to the reverse complement of the sequence annotated as the GC-rich region nucleotide sequence of SEQ ID NO: 1001 of International Application PCT/US2022/015499. In some embodiments, the genetic element comprises a nucleic acid sequence having at least about 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to the reverse complement of the sequence annotated as the 5′ UTR conserved domain nucleotide sequence of SEQ ID NO: 1001 of International Application PCT/US2022/015499. The Ring10 Anellovirus genomic sequence is disclosed as SEQ ID NO: 1008 of International Application PCT/US2022/015499, and the corresponding amino acid sequences are disclosed in Table F4 of the same International Application, and the sequences are herein incorporated by reference in their entireties. In some embodiments, the genetic element comprises a nucleic acid sequence having at least about 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to the reverse complement of the sequence annotated as the GC-rich region nucleotide sequence of SEQ ID NO: 1008 of International Application PCT/US2022/015499. In some embodiments, the genetic element comprises a nucleic acid sequence having at least about 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to the reverse complement of the sequence annotated as the 5′ UTR conserved domain nucleotide sequence of SEQ ID NO: 1008 of International Application PCT/US2022/015499. The Ring20 Anellovirus genomic sequence is disclosed as SEQ ID NO: 1014 of International Application PCT/US2022/015499, and the corresponding amino acid sequences are disclosed in Table F6 of the same International Application, and the sequences are herein incorporated by reference in their entireties. In some embodiments, the genetic element comprises a nucleic acid sequence having at least about 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to the reverse complement of the sequence annotated as the GC-rich region nucleotide sequence of SEQ ID NO: 1014 of International Application PCT/US2022/015499. In some embodiments, the genetic element comprises a nucleic acid sequence having at least about 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to the reverse complement of the sequence annotated as the 5′ UTR conserved domain nucleotide sequence of SEQ ID NO: 1014 of International Application PCT/US2022/015499.


Amino Acid Sequences Encoded by Nucleic Acid Sequences

In embodiments, the nucleic acid molecule comprises a nucleic acid sequence encoding an amino acid sequence having at least about 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to the Anelloviridae family virus (e.g., Anellovirus) ORF1 amino acid sequence of Table A1-A3. In embodiments, the nucleic acid molecule comprises a nucleic acid sequence encoding an amino acid sequence having at least about 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to the Anelloviridae family virus (e.g., Anellovirus) ORF2 amino acid sequence of Table A1-A3. In embodiments, the nucleic acid molecule comprises a nucleic acid sequence encoding an amino acid sequence having at least about 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to the Anelloviridae family virus (e.g., Anellovirus) ORF3 amino acid sequence of Table A1-A3. In some embodiments, the nucleic acid is a genetic element construct or a construct for providing the polypeptide (e.g., an ORF1 molecule and/or an ORF2 molecule) in trans.


Proteins Comprising Amino Acid Sequences

In some embodiments, the Anelloviridae family vector described herein comprises an Anellovirus ORF or ORF molecule (e.g., an Anellovirus ORF1, ORF2, ORF2/2, ORF2/3, ORF1/1, or ORF1/2) includes a polypeptide comprising an amino acid sequence having at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to a corresponding Anellovirus ORF sequence, e.g., as described herein). In embodiments, the Anelloviridae family vector described herein comprises a protein having an amino acid sequence having at least about 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to the Anelloviridae family virus (e.g., Anellovirus) ORF1 amino acid sequence of Table A1-A3. In embodiments, the Anelloviridae family vector described herein comprises a protein having an amino acid sequence having at least about 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to the Anelloviridae family virus (e.g., Anellovirus) ORF2 amino acid sequence of Table A1-A3. In embodiments, the Anelloviridae family vector described herein comprises a protein having an amino acid sequence having at least about 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to the Anelloviridae family virus (e.g., Anellovirus) ORF3 amino acid sequence of Table A1-A3.


In some embodiments, an ORF1 molecule has an amino acid sequence having at least about 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to the ORF1 molecule of Table A2 of International Application PCT/US2021/037076. In some embodiments, an ORF1 molecule has an amino acid sequence having at least about 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to the ORF1 molecule of Table B4 of International Application PCT/US2022/015499. In some embodiments, an ORF1 molecule has an amino acid sequence having at least about 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to the ORF1 molecule of Table C2 of International Application PCT/US2021/037076. In some embodiments, an ORF1 molecule has an amino acid sequence having at least about 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to the ORF1 molecule of Table D2 of International Application PCT/US2022/015499. In some embodiments, an ORF1 molecule has an amino acid sequence having at least about 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to the ORF1 molecule of Table C4 of International Application PCT/US2019/065995. In some embodiments, an ORF1 molecule has an amino acid sequence having at least about 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to the ORF1 molecule of Table C5 of International Application PCT/US2019/065995. In some embodiments, an ORF1 molecule has an amino acid sequence having at least about 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to the ORF1 molecule of Table F2 of International Application PCT/US2022/015499. In some embodiments, an ORF1 molecule has an amino acid sequence having at least about 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to the ORF1 molecule of Table F4 of International Application PCT/US2022/015499. In some embodiments, an ORF1 molecule has an amino acid sequence having at least about 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to the ORF1 molecule of Table F6 of International Application PCT/US2022/015499.


In some embodiments, an ORF1 molecule (e.g., comprised in the Anelloviridae family vector) comprises a polypeptide encoded by the Anelloviridae family virus (e.g., Anellovirus) ORF1 nucleic acid sequence of any of Tables N1-N3. In some embodiments, the ORF1 molecule (e.g., comprised in the Anelloviridae family vector) comprises an Anelloviridae family virus (e.g., Anellovirus) ORF1 protein of Table A1-A3 or a splice variant or post-translationally processed (e.g., proteolytically processed) variant thereof. In some embodiments, an ORF2 molecule (e.g., comprised in the Anelloviridae family vector) comprises a polypeptide encoded by the Anelloviridae family virus (e.g., Anellovirus) ORF2 nucleic acid sequence of any of Tables N1-N3. In some embodiments, the ORF2 molecule (e.g., comprised in the Anelloviridae family vector) comprises an Anelloviridae family virus (e.g., Anellovirus) ORF2 protein of Table A1-A3 or a splice variant or post-translationally processed (e.g., proteolytically processed) variant thereof. In some embodiments, an ORF3 molecule (e.g., comprised in the Anelloviridae family vector) comprises a polypeptide encoded by the Anelloviridae family virus (e.g., Anellovirus) ORF3 nucleic acid sequence of any of Tables N1-N3. In some embodiments, the ORF3 molecule (e.g., comprised in the Anelloviridae family vector) comprises an Anelloviridae family virus (e.g., Anellovirus) ORF3 protein of Table A1-A3 or a splice variant or post-translationally processed (e.g., proteolytically processed) variant thereof.


Polypeptides Comprising Amino Acid Sequences

In some embodiments, the polypeptide described herein comprises an amino acid sequence having at least about 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to an Anelloviridae family virus (e.g., Anellovirus) ORF1 amino acid sequence described herein. In embodiments, the polypeptide described herein comprises an amino acid sequence having at least about 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to the Anelloviridae family virus (e.g., Anellovirus) ORF1 amino acid sequence of Table A1-A3.


In some embodiments, the polypeptide described herein comprises an amino acid sequence having at least about 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to an ORF1 molecule encoded by an Anelloviridae family virus (e.g., Anellovirus) ORF1 nucleic acid described herein. In some embodiments, the polypeptide described herein comprises an amino acid sequence having at least about 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to an ORF1 molecule encoded by an Anelloviridae family virus (e.g., Anellovirus) ORF1 nucleic acid as listed in Table N1-N3.


In some embodiments, the polypeptide described herein comprises an amino acid sequence having at least about 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to an Anelloviridae family virus (e.g., Anellovirus) ORF2 amino acid sequence described herein. In embodiments, the polypeptide described herein comprises an amino acid sequence having at least about 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to the Anelloviridae family virus (e.g., Anellovirus) ORF2 amino acid sequence of Table A1-A3.


In some embodiments, the polypeptide described herein comprises an amino acid sequence having at least about 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to an ORF2 molecule encoded by an Anelloviridae family virus (e.g., Anellovirus) ORF2 nucleic acid described herein. In some embodiments, the polypeptide described herein comprises an amino acid sequence having at least about 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to an ORF2 molecule encoded by an Anelloviridae family virus (e.g., Anellovirus) ORF2 nucleic acid as listed in Table N1-N3.


In some embodiments, the polypeptide described herein comprises an amino acid sequence having at least about 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to an Anelloviridae family virus (e.g., Anellovirus) ORF3 amino acid sequence described herein. In embodiments, the polypeptide described herein comprises an amino acid sequence having at least about 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to the Anelloviridae family virus (e.g., Anellovirus) ORF3 amino acid sequence of Table A1-A3.


In some embodiments, the polypeptide described herein comprises an amino acid sequence having at least about 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to an ORF3 molecule encoded by an Anelloviridae family virus (e.g., Anellovirus) ORF3 nucleic acid described herein. In some embodiments, the polypeptide described herein comprises an amino acid sequence having at least about 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to an ORF3 molecule encoded by an Anelloviridae family virus (e.g., Anellovirus) ORF3 nucleic acid as listed in Table N1-N3.


In some embodiments, the polypeptide comprises an amino acid sequence (e.g., an ORF1, ORF1/1, ORF1/2, ORF2, ORF2/2, ORF2/3, or ORF2t/3 sequence) as shown in Table A1-A3, or a sequence having at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity thereto.


Ring19 is an Anellovirus that was isolated from RPE cells. In some embodiments, a method described herein comprises delivering an Anellovector (e.g., an Anellovector having sequence similarity to Ring19) to eye tissue (e.g., to the eye of a subject), for example, to retinal tissue and/or RPE cells.









TABLE N1





Exemplary Anellovirus nucleic acid sequence (Betatorquevirus).
















Name
RING 19





Genus/Clade

Betatorquevirus






Source tissue
Retinal pigment epithelium





Accession
N/A










Full Sequence: 2876 bp





1         10       20        30         40       50


|         |        |         |          |        |


CGGGAGCCGAAGGTGAGTGCAACCACCGTAGTCTAGGGGCAATTCGGGCT


AGTTCAGTATGGCGGAACGGGCAAGAAACTTAAATATTATTATTTTACAG


ATGCAAATACAACCACCTATTAGAACCTTCAAACAAACAATTTCAGATTG


GAAAAACTTAATTGTCCACGTTCACGACAACATTTGCAACTGCAATAAAC


CATTAGAACACACTATTGATACCTGTATCACCAATCCAGATGAATTAAGA


TTAAACAAATCTACTAAACAACAACTACAAAAATGCCTTGGTACCCCAGA


AGAAGATACCCAAGAAGACGTTATCGATGGCTTCGCAGATGGAGAGCTAG


ACGCCCTTTTCGCCCAAGATACAGAAGAAGATACTGGGTAAGAAACTATT


CTCGAAAGAGAAAACTATTTAAAATAACAACCAAAGAATGGCAACCAAAA


GTTATAAGAAAGACTCATGTAAAGGGCACCTATCCTTTGTTTCTTTGTAC


AAAGCACAGAATTAACAATAATATGATACAATATTTAGACTCTATAGCTC


CAGAACACTATTACGGAGGAGGAGGATTTTCAATAATGCAATTTTCCTTA


CAAGCCTTATATGAAGAATTTATAAAAGCAAAAAACTGGTGGACTAATAC


AAACTGCTTTTTACCACTTGTAAGATATATGGGTTGCTCATTCAAATTTT


ATAAAACTGAATTTTATGATTATATTGTACTAATTGAAAGATGTTATCCA


CTTGCTTGTACTGATGAAATGTACTTATCTACTCAACCTAGTATTATGAT


GCTTACAAGAAAATGTATTTTTGTACCATGCAAACAAAACAGCAAAGGTA


AAAAACCTTACAAAAAAGTTAGAGTAAGACCACCTTCACAAATGACTACA


GGATGGCATTTCTCACAAGACTTAGCAAACATGCCACTTGTAGTACTAAA


AACTTCAGTATGCAGCTTTGACAGATATTACACAGACAGTACAGCTAAAT


CAACCACAATAGGCTTTAAAACACTTAACACACAAACATTTAGATATCAT


GACTGGCAGGAACCACCTACAACAGGATACAAACCACAAAACCTACTATG


GTTTTATGGAGCAGAAAACGGATCACCAGTAGACCCCAACAACACAATAG


TATCAAACCTAATATACTTAGGAGGCACAGGACCTTATGAAAAAGGCACA


CCAATAAAAACAAACATAAGCAATTACTTTTCAGAGCCTAAACTGTGGGG


AAATATATTTCACGATGATTATACATCAGGAACATCACCCGTGTTTGTTA


CAAACAAATCACCATCAGAAATTAAAACCGCATGGAACACTATAAAAGAC


TTAACTGTTAAAGCTAGCGGTGTATTTACATTAAGAACAATTCCACTATG


GCTACCTTGCAGATACAACCCATTTGCAGACAAAGCAACCAACAACAAAA


TATGGCTAGTTTCTATACATTCAGACCACACAGAATGGAAACCAATAGAC


AATCCATTACTACAACGAACAGACCTTCCTTTATGGTTACTTGTATGGGG


TTGGCAAGATTGGCAGAAAAAAAACCAACAAACTTCACAACCTGATATTA


ATTATTTAACAGTAATATCTTCACCATATATATCATGCTACCCAAAATTA


GATTACTATGTGTTACTAGATGAAGGATTTTGGGAGGGTCACTCAACATA


CATAGAGTCAATTACAGACTCAGACAAAAAACACTGGTACCCTAAAAATA


GATTTCAAATAGAAACACTTAATCTAATAGCTAACACAGGTCCAGGAACT


GTAAAACTAAGAGAAAACCAAGCAGCAGAAGGTCACATGGTATATCGCTT


TAATTTTAAGCTTGGAGGATGTCCCGCACCGATGGAAAAAATATGTGACC


CTAGCAAACAATCCAAATATCCTATTCCCAATAACCAGCAACAAACAACT


TCGTTGCAGAGTCCAGAAAACCCAATTCAAACCTATCTCTACGACTTCGA


CGAAAGGAGGGGCCTACTTACAGAAAGAGCTACAAAAAGAATCAAACAAG


ATCACACATCTGAAAAAACTGTTTTGCCATTTACAGGAGCAGCAACAGAC


CTCCCCATACTCCAAACAACATCACAGGAGGAAAGCTCCTCGGAAGAAGA


AGAAGAGCAACAAGCGGAGAAGAAACTACTCCAGCTCCGAAGAAAGCAGC


ACCGACTCCGGGAGCGAATCCTCCAGCTATTAGACATACAAAATACATAA


TAAAACAAAGTACTGTAAAAATTGATATGTTTGGAGATACTCATGTACCT


AACCGTAGAATGACCCCAGAAGAATTTGAACAAGAACTAATTGTCGCTGG


TGTTTTTCGCAGACCTCCTTGTTACTATATAAAAGATAGACCTACTTATC


CTTATGTACCAAAACCTACTGATGAAAAATGTATGGTAAACTTTGACTTA


AACTTTCCTTAATAAACTACGCCTGCAAACTTTCACTCTCGGTGTCCATT


TATATAAGATAAAACTTAAATAAACATCCACCACTCTCCCAAATACGCAG


GCGCACAAGGGGGCTCCGCCCCCTTAAACCCCCAAGGGGGCTCCGCCCCC


TTAAACCCCCAAGGGGGCTCCGCCCCCTTACACCCCCTAATAAATATTCA


ACAGGAAAACCACCTAATTAGAATTGCCGACCACAAACCGTCACTTACTT


CTCCTTTTTGCACTTACTTCCTCTTTTACTTATTATTATTCATTACATTA


ATTAATAATCACTGTAATTCCGGGGAGGAGCTAACAATCTATATAACTAA


CTACACTTCCGAATGGCTGAGTTTATGCCGCCAGACGGAGACGGGATCAC


TTCAGTGACTCCAGGCTGAACTTGGG (SEQ ID NO: 1)





Annotations:








Putative Domain
Base range





ORF1
283-2250





ORF2
 101-391





ORF3
2277-2462





GC-rich region
2515-2615





5′ UTR Conserved Domain, or a portion
   1-71


thereof
















TABLE A1





Exemplary Anellovirus amino acid sequence (Betatorquevirus)


RING 19 (Betatorquevirus)
















ORF1
MPWYPRRRYPRRRYRWLRRWRARRPFRPRYRRRYWVRNYSRKRKLFKITT



KEWQPKVIRKTHVKGTYPLFLCTKHRINNNMIQYLDSIAPEHYYGGGGFS



IMQFSLQALYEEFIKAKNWWTNTNCFLPLVRYMGCSFKFYKTEFYDYIVL



IERCYPLACTDEMYLSTQPSIMMLTRKCIFVPCKQNSKGKKPYKKVRVRP



PSQMTTGWHFSQDLANMPLVVLKTSVCSFDRYYTDSTAKSTTIGFKTLNT



QTFRYHDWQEPPTTGYKPQNLLWFYGAENGSPVDPNNTIVSNLIYLGGTG



PYEKGTPIKTNISNYFSEPKLWGNIFHDDYTSGTSPVFVINKSPSEIKTA



WNTIKDLTVKASGVFTLRTIPLWLPCRYNPFADKATNNKIWLVSIHSDHT



EWKPIDNPLLQRTDLPLWLLVWGWQDWQKKNQQTSQPDINYLTVISSPYI



SCYPKLDYYVLLDEGFWEGHSTYIESITDSDKKHWYPKNRFQIETLNLIA



NTGPGTVKLRENQAAEGHMVYRFNFKLGGCPAPMEKICDPSKQSKYPIPN



NQQQTTSLQSPENPIQTYLYDFDERRGLLTERATKRIKQDHTSEKTVLPF



TGAATDLPILQTTSQEESSSEEEEEQQAEKKLLQLRRKQHRLRERILQLL



DIQNT (SEQ ID NO: 2)





ORF2
MQIQPPIRTFKQTISDWKNLIVHVHDNICNCNKPLEHTIDTCITNPDELR



LNKSTKQQLQKCLGTPEEDTQEDVIDGFADGELDALFAQDTEEDTG



(SEQ ID NO: 173)





ORF3
MFGDTHVPNRRMTPEEFEQELIVAGVFRRPPCYYIKDRPTYPYVPKPTDE



KCMVNFDLNFP (SEQ ID NO: 4)
















TABLE N2





Exemplary Anellovirus nucleic acid sequence (Betatorquevirus).
















Name
Ring2





Genus/Clade

Betatorquevirus






Accession Number
JX134045.1










Full Sequence: 2797 bp





1         10       20        30         40       50


|         |        |         |          |        |


TAATAAATATTCAACAGGAAAACCACCTAATTTAAATTGCCGACCACAAA


CCGTCACTTAGTTCCCCTTTTTGCAACAACTTCTGCTTTTTTCCAACTGC


CGGAAAACCACATAATTTGCATGGCTAACCACAAACTGATATGCTAATTA


ACTTCCACAAAACAACTTCCCCTTTTAAAACCACACCTACAAATTAATTA


TTAAACACAGTCACATCCTGGGAGGTACTACCACACTATAATACCAAGTG


CACTTCCGAATGGCTGAGTTTATGCCGCTAGACGGAGAACGCATCAGTTA


CTGACTGCGGACTGAACTTGGGCGGGTGCCGAAGGTGAGTGAAACCACCG


AAGTCAAGGGGCAATTCGGGCTAGTTCAGTCTAGCGGAACGGGCAAGAAA


CTTAAAATTATTTTATTTTTCAGATGAGCGACTGCTTTAAACCAACATGC


TACAACAACAAAACAAAGCAAACTCACTGGATTAATAACCTGCATTTAAC


CCACGACCTGATCTGCTTCTGCCCAACACCAACTAGACACTTATTACTAG


CTTTAGCAGAACAACAAGAAACAATTGAAGTGTCTAAACAAGAAAAAGAA


AAAATAACAAGATGCCTTATTACTACAGAAGAAGACGGTACAACTACAGA


CGTCCTAGATGGTATGGACGAGGTTGGATTAGACGCCCTTTTCGCAGAAG


ATTTCGAAGAAAAAGAAGGGTAAGACCTACTTATACTACTATTCCTCTAA


AGCAATGGCAACCGCCATATAAAAGAACATGCTATATAAAAGGACAAGAC


TGTTTAATATACTATAGCAACTTAAGACTGGGAATGAATAGTACAATGTA


TGAAAAAAGTATTGTACCTGTACATTGGCCGGGAGGGGGTTCTTTTTCTG


TAAGCATGTTAACTTTAGATGCCTTGTATGATATACATAAACTTTGTAGA


AACTGGTGGACATCCACAAACCAAGACTTACCACTAGTAAGATATAAAGG


ATGCAAAATAACATTTTATCAAAGCACATTTACAGACTACATAGTAAGAA


TACATACAGAACTACCAGCTAACAGTAACAAACTAACATACCCAAACACA


CATCCACTAATGATGATGATGTCTAAGTACAAACACATTATACCTAGTAG


ACAAACAAGAAGAAAAAAGAAACCATACACAAAAATATTTGTAAAACCAC


CTCCGCAATTTGAAAACAAATGGTACTTTGCTACAGACCTCTACAAAATT


CCATTACTACAAATACACTGCACAGCATGCAACTTACAAAACCCATTTGT


AAAACCAGACAAATTATCAAACAATGTTACATTATGGTCACTAAACACCA


TAAGCATACAAAATAGAAACATGTCAGTGGATCAAGGACAATCATGGCCA


TTTAAAATACTAGGAACACAAAGCTTTTATTTTTACTTTTACACCGGAGC


AAACCTACCAGGTGACACAACACAAATACCAGTAGCAGACCTATTACCAC


TAACAAACCCAAGAATAAACAGACCAGGACAATCACTAAATGAGGCAAAA


ATTACAGACCATATTACTTTCACAGAATACAAAAACAAATTTACAAATTA


TTGGGGTAACCCATTTAATAAACACATTCAAGAACACCTAGATATGATAC


TATACTCACTAAAAAGTCCAGAAGCAATAAAAAACGAATGGACAACAGAA


AACATGAAATGGAACCAATTAAACAATGCAGGAACAATGGCATTAACACC


ATTTAACGAGCCAATATTCACACAAATACAATATAACCCAGATAGAGACA


CAGGAGAAGACACTCAATTATACCTACTCTCTAACGCTACAGGAACAGGA


TGGGACCCACCAGGAATTCCAGAATTAATACTAGAAGGATTTCCACTATG


GTTAATATATTGGGGATTTGCAGACTTTCAAAAAAACCTAAAAAAAGTAA


CAAACATAGACACAAATTACATGTTAGTAGCAAAAACAAAATTTACACAA


AAACCTGGCACATTCTACTTAGTAATACTAAATGACACCTTTGTAGAAGG


CAATAGCCCATATGAAAAACAACCTTTACCTGAAGACAACATTAAATGGT


ACCCACAAGTACAATACCAATTAGAAGCACAAAACAAACTACTACAAACT


GGGCCATTTACACCAAACATACAAGGACAACTATCAGACAATATATCAAT


GTTTTATAAATTTTACTTTAAATGGGGAGGAAGCCCACCAAAAGCAATTA


ATGTTGAAAATCCTGCCCACCAGATTCAATATCCCATACCCCGTAACGAG


CATGAAACAACTTCGTTACAGAGTCCAGGGGAAGCCCCAGAATCCATCTT


ATACTCCTTCGACTATAGACACGGGAACTACACAACAACAGCTTTGTCAC


GAATTAGCCAAGACTGGGCACTTAAAGACACTGTTTCTAAAATTACAGAG


CCAGATCGACAGCAACTGCTCAAACAAGCCCTCGAATGCCTGCAAATCTC


GGAAGAAACGCAGGAGAAAAAAGAAAAAGAAGTACAGCAGCTCATCAGCA


ACCTCAGACAGCAGCAGCAGCTGTACAGAGAGCGAATAATATCATTATTA


AAGGACCAATAACTTTTAACTGTGTAAAAAAGGTGAAATTGTTTGATGAT


AAACCAAAAAACCGTAGATTTACACCTGAGGAATTTGAAACTGAGTTACA


AATAGCAAAATGGTTAAAGAGACCCCCAAGATCCTTTGTAAATGATCCTC


CCTTTTACCCATGGTTACCACCTGAACCTGTTGTAAACTTTAAGCTTAAT


TTTACTGAATAAAGGCCAGCATTAATTCACTTAAGGAGTCTGTTTATTTA


AGTTAAACCTTAATAAACGGTCACCGCCTCCCTAATACGCAGGCGCAGAA


AGGGGGCTCCGCCCCCTTTAACCCCCAGGGGGCTCCGCCCCCTGAAACCC


CCAAGGGGGCTACGCCCCCTTACACCCCC (SEQ ID NO: 54)





Annotations:








Putative Domain
Base range





TATA Box
 237-243





Cap Site
 260-267





Transcriptional Start Site
 267





5′ UTR Conserved Domain
 323-393





ORF2
 424-723





ORF2/2
 424-719; 2274-2589





ORF2/3
 424-719; 2449-2812





ORF1
 612-2612





ORF1/1
 612-719; 2274-2612





ORF1/2
 612-719; 2449-2589





Three open-reading frame region
2441-2586





Poly(A) Signal
2808-2813





GC-rich region
2868-2929
















TABLE A2





Exemplary Anellovirus amino acid sequences (Betatorquevirus)


Ring2 (Betatorquevirus)
















ORF2
MSDCFKPTCYNNKTKQTHWINNLHLTHDLICFCPTPTRHLLLALAEQQETIEVSKQE



KEKITRCLITTEEDGTTTDVLDGMDEVGLDALFAEDFEEKEG (SEQ ID NO: 55)





ORF2/2
MSDCFKPTCYNNKTKQTHWINNLHLTHDLICFCPTPTRHLLLALAEQQETIEVSKQE



KEKITRCLITTEEDGTTTDVLDGMDEVGLDALFAEDFEEKEGFNIPYPVTSMKQLRY



RVQGKPQNPSYTPSTIDTGTTQQQLCHELAKTGHLKTLFLKLQSQIDSNCSNKPSNA



CKSRKKRRRKKKKKYSSSSATSDSSSSCTESE (SEQ ID NO: 56)





ORF2/3
MSDCFKPTCYNNKTKQTHWINNLHLTHDLICFCPTPTRHLLLALAEQQETIEVSKQE



KEKITRCLITTEEDGTTTDVLDGMDEVGLDALFAEDFEEKEGARSTATAQTSPRMP



ANLGRNAGEKRKRSTAAHQQPQTAAAAVQRANNIIIKGPITFNCVKKVKLFDDKPK



NRRFTPEEFETELQIAKWLKRPPRSFVNDPPFYPWLPPEPVVNFKLNFTE (SEQ ID



NO: 57)





ORF1
MPYYYRRRRYNYRRPRWYGRGWIRRPFRRRFRRKRRVRPTYTTIPLKQWQPPYKR



TCYIKGQDCLIYYSNLRLGMNSTMYEKSIVPVHWPGGGSFSVSMLTLDALYDIHKL



CRNWWTSTNQDLPLVRYKGCKITFYQSTFTDYIVRIHTELPANSNKLTYPNTHPLM



MMMSKYKHIIPSRQTRRKKKPYTKIFVKPPPQFENKWYFATDLYKIPLLQIHCTACN



LQNPFVKPDKLSNNVTLWSLNTISIQNRNMSVDQGQSWPFKILGTQSFYFYFYTGA



NLPGDTTQIPVADLLPLTNPRINRPGQSLNEAKITDHITFTEYKNKFTNYWGNPENK



HIQEHLDMILYSLKSPEAIKNEWTTENMKWNQLNNAGTMALTPFNEPIFTQIQYNP



DRDTGEDTQLYLLSNATGTGWDPPGIPELILEGFPLWLIYWGFADFQKNLKKVTNID



TNYMLVAKTKFTQKPGTFYLVILNDTFVEGNSPYEKQPLPEDNIKWYPQVQYQLEA



QNKLLQTGPFTPNIQGQLSDNISMFYKFYFKWGGSPPKAINVENPAHQIQYPIPRNE



HETTSLQSPGEAPESILYSFDYRHGNYTTTALSRISQDWALKDTVSKITEPDRQQLLK



QALECLQISEETQEKKEKEVQQLISNLRQQQQLYRERIISLLKDQ (SEQ ID NO: 58)





ORF1/1
MPYYYRRRRYNYRRPRWYGRGWIRRPFRRRFRRKRRIQYPIPRNEHETTSLQSPGE



APESILYSFDYRHGNYTTTALSRISQDWALKDTVSKITEPDRQQLLKQALECLQISEE



TQEKKEKEVQQLISNLRQQQQLYRERIISLLKDQ (SEQ ID NO: 59)





ORF1/2
MPYYYRRRRYNYRRPRWYGRGWIRRPFRRRFRRKRRSQIDSNCSNKPSNACKSRK



KRRRKKKKKYSSSSATSDSSSSCTESE (SEQ ID NO: 60)
















TABLE N3





Exemplary Anellovirus nucleic acid sequence (Alphatorquevirus).
















Name
Ring18





Genus/Clade

Alphatorquevirus






Source tissue






Accession Number
N/A










Full Sequence: 3733 bp











1
CACGTGACTC CCGCAGGCCA ACCAGAGTCT ACGTCGTGCA CTTCCTGGGC ATGGTCTACA





61
TCATAATATA AGAAGGCGCA CTTCCGAATG GCTGAGTTTT CCACGCCCGT CCGCAGCGAG





121
AACGCCACGG AGGGAGATCC TCGCGTCCCG AGGGCGGGTG CCGGAGGTGA GTTTACACAC





181
CGCAGTCAAG GGGCAATTCG GGCTCGGGAC TGGCCGGGCC CCGGGCAAGG CTCTTAAAAA





241
ATGCGCTTTC GCAGGGTTGC TGAGAAAAGG AAAGTGCTTC TGCAAACTGT GCGAGCTGCA





301
GAGAAGACTA GGCGGCTTCT AGGTATGTGG CAGCCCCCCG CGCACAATGT CCCCGGCATC





361
GAGAGAAACT GGTACGAGAG CTGTTTTCGA TCCCATGCTG CTGTTTGCGG CTGTGGCGAC





421
TTTGTTGGCC ATCTTAGTTA TCTGGCAACT ACTCTGGGTC GTCCTCCGCG TCCTGGGCCC





481
CCAGGCGGAC CCCGCACACC GCAGATAAGA AACCTGCCAG CGCTCCCGGC GCCCCAGGGC





541
GAGCCCGGTG ACAGAGCGCC ATGGCGTGGG GCTTCTGGGG CCGACGCCGC CGGTGGAGAC





601
GGTGGAGACC ACGGCGCAGA CGGTGGAGAC CCCGCAGACG TAGGAGACGA CGCCCTGCTC





661
GCCGCTTTCG AGCTCGTCGA AGAGTAAGGA GGCGCGGGGG GCGGTGGCGC AGACGCTACA





721
GAAAATGGCG ACGGGGCAGA CGCAGACGAA CTCACAGAAA AAAGATAGTC ATAAAACAGT





781
GGCAACCAAA CTTTATAAGA CGCTGCTACA TCATAGGGTA CCTACCTTTA ATATTCTGTG





841
GCGAAAACAC AACCGCCCAG AACTATGCCA CTCACTCAGA CGACATGATA AGCAAAGGAC





901
CATACGGGGG GGGCATGACT ACCACAAAAT TTACTCTGAG AATACTGTAC GACGAGTTTA





961
CCAGGTTTAT GAACTTTTGG ACTGTTAGTA ACGAAGACCT AGACCTGTGT AGATACGTGG





1021
GCTGCAAACT CATATTTTTT AAACATCCCA CAGTGGACTT TATAGTACAG ATAAACACTC





1081
AGCCTCCTTT CTTAGACACG CACCTTACCG CGGCCAGCAT ACACCCGGGC ATCATGATGC





1141
TCAGCAAGAG ACGCATACTA ATACCCTCTC TAAAAACCCG GCCAAGCAGA AAACACAGGG





1201
TGGTTGTTAG GGTGGGCGCC CCAAGACTTT TTCAGGACAA GTGGTACCCC CAGTCAGACC





1261
TGTGTGACAC AGTTCTGCTT TCCATATTTG CAACCGCCTG TGACTTGCAA TATCCGTTCG





1321
GCTCACCACT AACTGACAAC CCTTGCGTCA ACTTCCAGAT TCTGGGGCCC CAGTACAAAA





1381
AACACCTTAG TATTAGCTCT ACTATGGATA CAACTAACAA ACAGCACTAT GACAGCAATT





1441
TGTTTAACCA AACTCAGCTA TACAACACCT TTCAAACTAT AGCTCAGCTT AAAGAGACAG





1501
GACAAACTGC AAACATATCT CCTAGTTGGA GTGCAGTGCA AAATAATATG GCCCTTAGTA





1561
ATACCGGTGA AAATGCAACC CAAAGCAAAG ACACTTGGTA CAAAGGAAAC ACATACAACA





1621
ACCACATTAC AACGTTAGCA CAAAAAACCA GAGAAAGATT TAAAGGTGCA ACAAAAGCAG





1681
CACTACAAAA CTACCCCACC ATAATGTCCA CAGACTTATA TGAATACCAC TCAGGCATAT





1741
ACTCCAGCAT ATTTCTATCA GCTGGCAGGA GCTACTTTGA AACCACCGGG GCCTACTCTG





1801
ACATTATATA CAACCCTTTC ACAGACAAAG GCACAGGCAA CATAATCTGG ATAGACTACC





1861
TCACAAAAGA AGACACCATT TTTGTAAAAA ACAAAAGCAA ATGCGAAATA ATGGACATGC





1921
CCCTGTGGGC GGCCTGCACA GGATATACAG AGTTCTGTGC AAAGTATACA GGAGACTCTG





1981
CCATTATTTA CAATGCCAGA GTACTAATAA GATGCCCGTA CACTGAACCC ATGCTAATAG





2041
ACCACTCAGA CCCGAACAAA GGCTTCGTAC CCTACTCATT TAACTTTGGC AACGGAAAAA





2101
TGCCCGGAGG CAGCTCCAAC GTACCCATAA GAATGAGAGC CAAATGGTAC GTGAACATAT





2161
TCCACCAAAA AGAGGTTCTA GAGACTATAG TACAGAGCGG ACCGTTCGGG TACAAGGGCG





2221
ACATAAAATC AGCTGTACTG GCCATGAAAT ACAGATTTCA CTGGAAATGG GGTGGAAACC





2281
CTATATCCAA ACAGGTCGTC AGGAATCCCT GCTCCAACTC CAGCTCCTCC GCGGCCCATA





2341
GAGGACCTCG CAGCGTACAA GCAGTTGACC CGAAATACAA TACCCCAGAG GTCACGTGGC





2401
ACTCGTGGGA CATCAGACGA GGACTCTTTG GCAAAGCAGG TATTAAAAGA ATGCAACAAG





2461
AATCAGATGC TCTTTACATT CCTCCAGGAC CATTCAAGAG ACCTCGCAGA GACACGAACG





2521
CCCAAGACCC AGAAGAGCAA AACGAAAGCT CAGGTTTCAG AGTCCAGCAG CGACTCCCGT





2581
GGGTCCACTC CAGCCAAGAG ACGCAAAGCT CCCAAGAAGA AACGGAGGCG CAGGGGTCGG





2641
TACAAGACCA ACTACTCCTC CAGCTCCGAG AGCAGCGAGT ACTCCGACTC CAGCTCCAGC





2701
AACTCGCAAC CCAAGTCCTC AAAGTCCAAG CAGGGCACGG CATACACCCC CTATTATCTT





2761
CCCAAGCGTA AACAAAGTCT TTATGTTTGA GCCCCACGGT CCTAAACCCA TACAGGGCTA





2821
CAACGATTGG CTAGAGGAGT ACACTGCTTG TAAATTCTGG GACAGACCCC CAAGAAAGCT





2881
ACACACAGAC TTACCCTTTT ACCCCTGGGC ACCAAAACCC CAAGACCAAG TCAGGGTAAG





2941
CTTTAAACTC AACTTTCAAT AAAAATTCTA GGCCGTGGGA GTTTCACTTG TCGGTGTCTG





3001
CTTCTTAAGG TCGCCAAGCA CTCCGAGCGC CAGCGAGGAG TGCGACCCCC CCTCCGGTAG





3061
CAACGCCTTC GGAGCCGCGC GCTACGCCTT CGGCTGCGCG CGGCACCTCA GACCCCCCCT





3121
CCACCCGAAA CGCTTGCGCG TTTCGGACCT TCGGCGTCGG GGGGGTCGGG AGCTTTATTA





3181
AACAGACTCC GAGTTGCCAT TGGACACTGG AGCTGTGAAT CAGTAACGAA AGTGAGTGGG





3241
GCCAGACTTC GCCATAGGGC CTTTATCTTC TCGCCATTGG ATAGTGTCCG GGGTCGCCGT





3301
AGGCTTCGGC CTCGTTTTTA GGCCTTCCGG ACTACAAAAA TGGCGAACTT GGTGACGTCA





3361
CGGCCGCCAT TTTAAGTAAG GCGGAAGCAG CTCCACTTTC TCACAAAATG GCGGCGGAGC





3421
ACTTCCGGCT TGCCCAAAAT AGCGGGCAAG CTCTTCCGGG TCAAAGGTCA GCAGCTACGT





3481
CACAAGTCAC CTGACTGAGG AGGAGCTAAA ACCCGGAAGT CCTCCTCGGT CACGTGGCTA





3541
GTCACGTGAC TACTACGTCA TCGGCGCCAT CTTGTGTGAC AAAATGGCGG ACAACTTCCG





3601
CTTTTTTGAA AAAAGGCGCG AAAAAACGGC GGCGGCGGCG CGCGCGCTGC GCGCGCGCGC





3661
CGAGGGGGCG CCAGCGCCCC CACTGTGCGG TCCCCCGCGG GGCTCCGGCC CCCCCCCGAA





3721
GTCCGTCACT AAC (SEQ ID NO: 213)










Annotations








Putative Domain
Base range





TATA Box
  67-71





Cap Site
  88-95





Transcriptional Start Site
  95





5′ UTR Conserved Domain
 155-225





ORF2
 325-687





TAIP
 347-508





ORF2/2
 325-683, 2295-2790





ORF2/3
 325-683, 2488-2962





ORF1
 561-2771





ORF1/1
 561-683, 2295-2771





ORF1/2
 561-683, 2488-2790





Three open-reading frame region
2447-2771





Poly(A) Signal
2958-2963





GC-rich region
3627-3718
















TABLE A3





Exemplary Anellovirus amino acid sequences for Ring 18 (Alphatorquevirus)
















ORF1
MAWGFWGRRRRWRRWRPRRRRWRPRRRRRRRPARRFRARRRVRRRGGR



WRRRYRKWRRGRRRRTHRKKIVIKQWQPNFIRRCYIIGYLPLIFCGENTTA



QNYATHSDDMISKGPYGGGMTTTKFTLRILYDEFTRFMNFWTVSNEDLDL



CRYVGCKLIFFKHPTVDFIVQINTQPPFLDTHLTAASIHPGIMMLSKRRILIPS



LKTRPSRKHRVVVRVGAPRLFQDKWYPQSDLCDTVLLSIFATACDLQYPF



GSPLTDNPCVNFQILGPQYKKHLSISSTMDTTNKQHYDSNLFNQTQLYNTF



QTIAQLKETGQTANISPSWSAVQNNMALSNTGENATQSKDTWYKGNTYN



NHITTLAQKTRERFKGATKAALQNYPTIMSTDLYEYHSGIYSSIFLSAGRSY



FETTGAYSDIIYNPFTDKGTGNIIWIDYLTKEDTIFVKNKSKCEIMDMPLWA



ACTGYTEFCAKYTGDSAIIYNARVLIRCPYTEPMLIDHSDPNKGFVPYSFNF



GNGKMPGGSSNVPIRMRAKWYVNIFHQKEVLETIVQSGPFGYKGDIKSAV



LAMKYRFHWKWGGNPISKQVVRNPCSNSSSSAAHRGPRSVQAVDPKYNT



PEVTWHSWDIRRGLFGKAGIKRMQQESDALYIPPGPFKRPRRDTNAQDPEE



QNESSGFRVQQRLPWVHSSQETQSSQEETEAQGSVQDQLLLQLREQRVLR



LQLQQLATQVLKVQAGHGIHPLLSSQA (SEQ ID NO: 1100)





ORF1/1
MAWGFWGRRRRWRRWRSRRRRWRPRRRRRRRPARRFRARRRVVRNPCS



NSSSSAAHRGPRSVQAVDPKYNTPEVTWHSWDIRRGLFGKAGIKRMQQES



DALYIPPGPFKRPRRDTNAQDPEEQNESSGFRVQQRLPWVHSSQETQSSQE



ETEAQGSVQDQLLLQLREQRVLRLQLQQLATQVLKVQAGHGIHPLLSSQA



(SEQ ID NO: 214)





ORF1/2
MAWGFWGRRRRWRRWRPRRRRWRPRRRRRRRPARRFRARRRDHSRDLA



ETRTPKTQKSKTKAQVSESSSDSRGSTPAKRRKAPKKKRRRRGRYKTNYSS



SSESSEYSDSSSSNSQPKSSKSKQGTAYTPYYLPKRKQSLYV (SEQ ID NO:



215)





ORF2
MWQPPAHNVPGIERNWYESCFRSHAAVCGCGDFVGHLSYLATTLGRPPRP



GPPGGPRTPQIRNLPALPAPQGEPGDRAPWRGASGADAAGGDGGDHGAD



GGDPADVGDDALLAAFELVEE (SEQ ID NO: 216)





ORF2/2
MWQPPAHNVPGIERNWYESCFRSHAAVCGCGDFVGHLSYLATTLGRPPRP



GPPGGPRTPQIRNLPALPAPQGEPGDRAPWRGASGADAAGGDGGDHGAD



GGDPADVGDDALLAAFELVEESSGIPAPTPAPPRPIEDLAAYKQLTRNTIPQ



RSRGTRGTSDEDSLAKQVLKECNKNQMLFTFLQDHSRDLAETRTPKTQKS



KTKAQVSESSSDSRGSTPAKRRKAPKKKRRRRGRYKTNYSSSSESSEYSDS



SSSNSQPKSSKSKQGTAYTPYYLPKRKQSLYV (SEQ ID NO: 217)





ORF2/3
MWQPPAHNVPGIERNWYESCFRSHAAVCGCGDFVGHLSYLATTLGRPPRP



GPPGGPRTPQIRNLPALPAPQGEPGDRAPWRGASGADAAGGDGGDHGAD



GGDPADVGDDALLAAFELVEETIQETSQRHERPRPRRAKRKLRFQSPAATP



VGPLQPRDAKLPRRNGGAGVGTRPTTPPAPRAASTPTPAPATRNPSPQSPSR



ARHTPPIIFPSVNKVFMFEPHGPKPIQGYNDWLEEYTACKFWDRPPRKLHT



DLPFYPWAPKPQDQVRVSFKLNFQ (SEQ ID NO: 218)









Capsid Proteins (e.g., ORF1 Molecules)

In some embodiments, the anellovector comprises an ORF1 molecule and/or a nucleic acid encoding an ORF1 molecule.


Generally, an ORF1 molecule comprises a polypeptide having the structural features and/or activity of an Anellovirus ORF1 protein (e.g., an Anellovirus ORF1 protein as described herein, e.g., as listed in Table A1-A3), or a functional fragment thereof. In some embodiments, the ORF1 molecule comprises a truncation relative to an Anellovirus ORF1 protein (e.g., an Anellovirus ORF1 protein as described herein, e.g., as listed in Table A1-A3). In some embodiments, the ORF1 molecule is truncated by at least 10, 20, 30, 40, 50, 60, 70, 80, 90, 100, 150, 200, 250, 300, 350, 400, 450, 500, 550, 600, 650, or 700 amino acids of the Anellovirus ORF1 protein. In some embodiments, an ORF1 molecule comprises an amino acid sequence having at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to an Anellovirus ORF1 protein sequence as shown in Table A1-A3. In some embodiments, an ORF1 molecule comprises an amino acid sequence having at least 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity to an Betatorquevirus ORF1 protein, e.g., as described herein. An ORF1 molecule can generally bind to a nucleic acid molecule, such as DNA (e.g., a genetic element, e.g., as described herein). In some embodiments, an ORF1 molecule localizes to the nucleus of a cell. In certain embodiments, an ORF1 molecule localizes to the nucleolus of a cell. In some embodiments, an ORF1 molecule is encoded by an ORF1 nucleic acid. In some embodiments, the ORF1 nucleic acid comprises an antisense strand, which can be directly transcribed to produce mRNA encoding the ORF1 molecule. In some embodiments, the ORF1 nucleic acid comprises a sense strand.


In some embodiments, an ORF1 molecule as described herein comprises an amino acid sequence (e.g., an ORF1 sequence, or an arginine-rich region, jelly-roll domain, HVR, N22, or C-terminal domain sequence) as listed in any of Tables A2, A4, A6, A8, A10, A12, C1-C5, 2, 4, 6, 8, 10, 12, 14, 16, 18, 20-37, or D1-D10 of PCT Publication No. WO2020/123816 (incorporated herein by reference in its entirety), or a sequence having at least 70% 80%, 85%, 90% 95%, 96%, 97%, 98% and 99% nucleotide sequence identity thereto.


Without wishing to be bound by theory, an ORF1 molecule may be capable of binding to other ORF1 molecules, e.g., to form a proteinaceous exterior (e.g., as described herein). Such an ORF1 molecule may be described as having the capacity to form a capsid. In some embodiments, the proteinaceous exterior may encapsidate a nucleic acid molecule (e.g., a genetic element as described herein). In some embodiments, a plurality of ORF1 molecules may form a multimer, e.g., to produce a proteinaceous exterior. In some embodiments, the multimer may be a homomultimer. In other embodiments, the multimer may be a heteromultimer (e.g., comprising a plurality of distinct ORF1 molecules). It is also contemplated that an ORF1 molecule may have replicase activity.


An ORF1 molecule may, in some embodiments, comprise one or more of: a first region comprising an arginine rich region, e.g., a region having at least 60% basic residues (e.g., at least 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 100% basic residues; e.g., between 60%-90%, 60%-80%, 70%-90%, or 70-80% basic residues), and a second region comprising jelly-roll domain, e.g., at least six beta strands (e.g., 4, 5, 6, 7, 8, 9, 10, 11, or 12 beta strands).


Arginine-Rich Region

An arginine rich region (e.g., comprised an ORF1 molecule as described herein) has at least 70% (e.g., at least about 70, 80, 90, 95, 96, 97, 98, 99, or 100%) sequence identity to an arginine-rich region sequence described herein or a sequence of at least about 40 amino acids comprising at least 60%, 70%, or 80% basic residues (e.g., arginine, lysine, or a combination thereof).


In some embodiments, an ORF1 molecule as described herein comprises a deletion or truncation of an arginine-rich region. In some embodiments, the entire arginine-rich region is deleted. In some embodiments, a portion of the arginine-rich region (e.g., a N-terminal portion of the structural arginine-rich region) is deleted. In embodiments, the ORF1 molecule does not comprise an Anellovirus ORF1 arginine-rich region, or an amino acid sequence having at least 70%, 75%, 80%, 8%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity thereto.


In some embodiments, an ORF1 molecule having a deletion or truncation of the arginine-rich region further comprises a deletion or truncation of at least a portion of a C-terminal domain (e.g., as described herein).


Jelly Roll Domain

A jelly-roll domain or region (e.g., comprised an ORF1 molecule as described herein) comprises (e.g., consists of) a polypeptide (e.g., a domain or region comprised in a larger polypeptide) comprising one or more (e.g., 1, 2, or 3) of the following characteristics:

    • (i) at least 30% (e.g., at least 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 90%, or more) of the amino acids of the jelly-roll domain are part of one or more β-sheets;
    • (ii) the secondary structure of the jelly-roll domain comprises at least four (e.g., at least 4, 5, 6, 7, 8, 9, 10, 11, or 12) β-strands; and/or
    • (iii) the tertiary structure of the jelly-roll domain comprises at least two (e.g., at least 2, 3, or 4) (3-sheets; and/or
    • (iv) the jelly-roll domain comprises a ratio of β-sheets to α-helices of at least 2:1, 3:1, 4:1, 5:1, 6:1, 7:1, 8:1, 9:1, or 10:1.


In certain embodiments, a jelly-roll domain comprises two β-sheets.


In certain embodiments, one or more (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10) of the β-sheets comprises about eight (e.g., 4, 5, 6, 7, 8, 9, 10, 11, or 12) β-strands. In certain embodiments, one or more (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10) of the β-sheets comprises eight β-strands. In certain embodiments, one or more (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10) of the β-sheets comprises seven β-strands. In certain embodiments, one or more (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10) of the β-sheets comprises six β-strands. In certain embodiments, one or more (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10) of the β-sheets comprises five β-strands. In certain embodiments, one or more (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10) of the β-sheets comprises four β-strands.


In some embodiments, the jelly-roll domain comprises a first β-sheet in antiparallel orientation to a second β-sheet. In certain embodiments, the first β-sheet comprises about four (e.g., 3, 4, 5, or 6) 0-strands. In certain embodiments, the second β-sheet comprises about four (e.g., 3, 4, 5, or 6) β-strands. In embodiments, the first and second β-sheet comprise, in total, about eight (e.g., 6, 7, 8, 9, 10, 11, or 12) β-strands.


In certain embodiments, a jelly-roll domain is a component of a capsid protein (e.g., an ORF1 molecule as described herein). In certain embodiments, a jelly-roll domain has self-assembly activity. In some embodiments, a polypeptide comprising a jelly-roll domain binds to another copy of the polypeptide comprising the jelly-roll domain. In some embodiments, a jelly-roll domain of a first polypeptide binds to a jelly-roll domain of a second copy of the polypeptide.


Other Subdomains

An ORF1 molecule may also include a third region comprising the structure or activity of an Anellovirus N22 domain (e.g., as described herein, e.g., an N22 domain from an Anellovirus ORF1 protein as described herein).


An ORF1 molecule may also include a fourth region comprising the structure or activity of an Anellovirus C-terminal domain (CTD) (e.g., as described herein, e.g., a CTD from an Anellovirus ORF1 protein as described herein).


In some embodiments, an ORF1 molecule as described herein comprises a deletion or truncation of a C-terminal domain (CTD). In some embodiments, the entire CTD is deleted. In some embodiments, a portion of the CTD (e.g., a C-terminal portion of the CTD) is deleted. In embodiments, the ORF1 molecule does not comprise an Anellovirus ORF1 CTD, or an amino acid sequence having at least 70%, 75%, 80%, 8%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity thereto. In some embodiments, an ORF1 molecule having a deletion or truncation of the CTD further comprises a deletion or truncation of at least a portion of an arginine-rich region (e.g., as described herein).


In some embodiments, the ORF1 molecule comprises, in N-terminal to C-terminal order, the first, second, third, and fourth regions.


The ORF1 molecule may, in some embodiments, further comprise a hypervariable region (HVR), e.g., an HVR from an Anellovirus ORF1 protein, e.g., as described herein. In some embodiments, the HVR is positioned between the second region and the third region. In some embodiments, the HVR comprises at least about 55 (e.g., at least about 45, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, or 65) amino acids (e.g., about 45-160, 50-160, 55-160, 60-160, 45-150, 50-150, 55-150, 60-150, 45-140, 50-140, 55-140, or 60-140 amino acids).


In some embodiments, the first region can bind to a nucleic acid molecule (e.g., DNA). In some embodiments, the basic residues are selected from arginine, histidine, or lysine, or a combination thereof. In some embodiments, the first region comprises at least 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 100% arginine residues (e.g., between 60%-90%, 60%-80%, 70%-90%, or 70-80% arginine residues). In some embodiments, the first region comprises about 30-120 amino acids (e.g., about 40-120, 40-100, 40-90, 40-80, 40-70, 50-100, 50-90, 50-80, 50-70, 60-100, 60-90, or 60-80 amino acids). In some embodiments, the first region comprises the structure or activity of a viral ORF1 arginine-rich region (e.g., an arginine-rich region from an Anellovirus ORF1 protein, e.g., as described herein). In some embodiments, the first region comprises a nuclear localization signal.


In some embodiments, the second region comprises a jelly-roll domain, e.g., the structure or activity of a viral ORF1 jelly-roll domain (e.g., a jelly-roll domain from an Anellovirus ORF1 protein, e.g., as described herein). In some embodiments, the second region is capable of binding to the second region of another ORF1 molecule, e.g., to form a proteinaceous exterior (e.g., capsid) or a portion thereof.


In some embodiments, the fourth region is exposed on the surface of a proteinaceous exterior (e.g., a proteinaceous exterior comprising a multimer of ORF1 molecules, e.g., as described herein).


In some embodiments, the first region, second region, third region, fourth region, and/or HVR each comprise fewer than four (e.g., 0, 1, 2, or 3) beta sheets.


In some embodiments, one or more of the first region, second region, third region, fourth region, and/or HVR may be replaced by a heterologous amino acid sequence (e.g., the corresponding region from a heterologous ORF1 molecule). In some embodiments, the heterologous amino acid sequence has a desired functionality, e.g., as described herein.


In some embodiments, the ORF1 molecule comprises a plurality of conserved motifs (e.g., motifs comprising about 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 25, 30, 35, 40, 45, 50, 60, 70, 80, 90, 100, or more amino acids). In some embodiments, the conserved motifs may show 60, 70, 80, 85, 90, 95, or 100% sequence identity to an ORF1 protein of one or more wild-type Anellovirus clades (e.g., Betatorquevirus). In some embodiments, the conserved motifs each have a length between 1-1000 (e.g., between 5-10, 5-15, 5-20, 10-15, 10-20, 15-20, 5-50, 5-100, 10-50, 10-100, 10-1000, 50-100, 50-1000, or 100-1000) amino acids. In certain embodiments, the conserved motifs consist of about 2-4% (e.g., about 1-8%, 1-6%, 1-5%, 1-4%, 2-8%, 2-6%, 2-5%, or 2-4%) of the sequence of the ORF1 molecule, and each show 100% sequence identity to the corresponding motifs in an ORF1 protein of the wild-type Anellovirus clade. In certain embodiments, the conserved motifs consist of about 5-10% (e.g., about 1-20%, 1-10%, 5-20%, or 5-10%) of the sequence of the ORF1 molecule, and each show 80% sequence identity to the corresponding motifs in an ORF1 protein of the wild-type Anellovirus clade. In certain embodiments, the conserved motifs consist of about 10-50% (e.g., about 10-20%, 10-30%, 10-40%, 10-50%, 20-40%, 20-50%, or 30-50%) of the sequence of the ORF1 molecule, and each show 60% sequence identity to the corresponding motifs in an ORF1 protein of the wild-type Anellovirus clade.


In some embodiments, an ORF1 molecule comprises at least one difference (e.g., a mutation, chemical modification, or epigenetic alteration) relative to a wild-type ORF1 protein, e.g., as described herein (e.g., as shown in Table A1-A3).


Conserved ORF1 Motif in N22 Domain

In some embodiments, a polypeptide (e.g., an ORF1 molecule) described herein comprises the amino acid sequence YNPX2DXGX2N (SEQ ID NO: 829), wherein X″ is a contiguous sequence of any n amino acids. For example, X2 indicates a contiguous sequence of any two amino acids. In some embodiments, the YNPX2DXGX2N (SEQ ID NO: 829) is comprised within the N22 domain of an ORF1 molecule, e.g., as described herein. In some embodiments, a genetic element described herein comprises a nucleic acid sequence (e.g., a nucleic acid sequence encoding an ORF1 molecule, e.g., as described herein) encoding the amino acid sequence YNPX2DXGX2N (SEQ ID NO: 829), wherein X″ is a contiguous sequence of any n amino acids.


In some embodiments, a polypeptide (e.g., an ORF1 molecule) comprises a conserved secondary structure, e.g., flanking and/or comprising a portion of the YNPX2DXGX2N (SEQ ID NO: 829) motif, e.g., in an N22 domain. In some embodiments, the conserved secondary structure comprises a first beta strand and/or a second beta strand. In some embodiments, the first beta strand is about 5-6 (e.g., 3, 4, 5, 6, 7, or 8) amino acids in length. In some embodiments, the first beta strand comprises the tyrosine (Y) residue at the N-terminal end of the YNPX2DXGX2N (SEQ ID NO: 829) motif. In some embodiments, the YNPX2DXGX2N (SEQ ID NO: 829) motif comprises a random coil (e.g., about 8-9 amino acids of random coil). In some embodiments, the second beta strand is about 7-8 (e.g., 5, 6, 7, 8, 9, or 10) amino acids in length. In some embodiments, the second beta strand comprises the asparagine (N) residue at the C-terminal end of the YNPX2DXGX2N (SEQ ID NO: 829) motif.


Exemplary ORF1 Sequences

In some embodiments, a polypeptide (e.g., an ORF1 molecule) described herein comprises an amino acid sequence having at least about 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to one or more Anellovirus ORF1 subsequences, e.g., as described herein). In some embodiments, an Anelloviridae family vector (e.g., anellovector) described herein comprises an ORF1 molecule comprising an amino acid sequence having at least about 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to one or more Anellovirus ORF1 subsequences, e.g., as described herein. In some embodiments, an anellovector described herein comprises a nucleic acid molecule (e.g., a genetic element) encoding an ORF1 molecule comprising an amino acid sequence having at least about 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to one or more Anellovirus ORF1 subsequences, e.g., as described herein.


In some embodiments, the one or more Anellovirus ORF1 subsequences comprises one or more of an arginine (Arg)-rich domain, a jelly-roll domain, a hypervariable region (HVR), an N22 domain, or a C-terminal domain (CTD) (e.g., as listed herein), or sequences having at least about 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity thereto. In some embodiments, the ORF1 molecule comprises a plurality of subsequences from different Anelloviruses. In some embodiments, the ORF1 molecule comprises one or more of an Arg-rich domain, a jelly-roll domain, an N22 domain, and a CTD from one Anelloviridae family virus (e.g., Anellovirus), and an HVR from another. In some embodiments, the ORF1 molecule comprises one or more of a jelly-roll domain, an HVR, an N22 domain, and a CTD from one Anelloviridae family virus (e.g., Anellovirus), and an Arg-rich domain from another. In some embodiments, the ORF1 molecule comprises one or more of an Arg-rich domain, an HVR, an N22 domain, and a CTD from one Anelloviridae family virus (e.g., Anellovirus), and a jelly-roll domain from another. In some embodiments, the ORF1 molecule comprises one or more of an Arg-rich domain, a jelly-roll domain, an HVR, and a CTD from one Anelloviridae family virus (e.g., Anellovirus), and an N22 domain from another. In some embodiments, the ORF1 molecule comprises one or more of an Arg-rich domain, a jelly-roll domain, an HVR, and an N22 domain from one Anelloviridae family virus (e.g., Anellovirus), and a CTD from another.


Identification of ORF1 Protein Sequences

In some embodiments, an ORF1 protein sequence, or a nucleic acid sequence encoding an ORF1 protein, can be identified from the genome of an Anelloviridae family virus, e.g., an Anellovirus (e.g., a putative Anelloviridae family virus genome identified, for example, by nucleic acid sequencing techniques, e.g., deep sequencing techniques). In some embodiments, an ORF1 protein sequence is identified by one or more (e.g., 1, 2, or all 3) of the following selection criteria:

    • (i) Length Selection: Protein sequences (e.g., putative ORF1 sequences passing the criteria described in (ii) or (iii) below) may be size-selected for those greater than about 600 amino acid residues to identify putative ORF1 proteins. In some embodiments, an ORF1 protein sequence is at least about 600, 650, 700, 750, 800, 850, 900, 950, or 1000 amino acid residues in length. In some embodiments, an Alphatorquevirus ORF1 protein sequence is at least about 700, 710, 720, 730, 740, 750, 760, 770, 780, 790, 800, 900, or 1000 amino acid residues in length. In some embodiments, a Betatorquevirus ORF1 protein sequence is at least about 650, 660, 670, 680, 690, 700, 750, 800, 900, or 1000 amino acid residues in length. In some embodiments, a Gammatorquevirus ORF1 protein sequence is at least about 650, 660, 670, 680, 690, 700, 750, 800, 900, or 1000 amino acid residues in length. In some embodiments, a nucleic acid sequence encoding an ORF1 protein is at least about 1800, 1900, 2000, 2100, 2200, 2300, 2400, or 2500 nucleotides in length. In some embodiments, a nucleic acid sequence encoding an Alphatorquevirus ORF1 protein sequence is at least about 2100, 2150, 2200, 2250, 2300, 2400, or 2500 nucleotides in length. In some embodiments, a nucleic acid sequence encoding a Betatorquevirus ORF1 protein sequence is at least about 1900, 1950, 2000, 2500, 2100, 2150, 2200, 2250, 2300, 2400, or 2500 or 1000 nucleotides in length. In some embodiments, a nucleic acid sequence encoding a Gammatorquevirus ORF1 protein sequence is at least about 1900, 1950, 2000, 2500, 2100, 2150, 2200, 2250, 2300, 2400, or 2500 or 1000 nucleotides in length.
    • (ii) Presence of ORF1 motif Protein sequences (e.g., putative ORF1 sequences passing the criteria described in (i) above or (iii) below) may be filtered to identify those that contain the conserved ORF1 motif in the N22 domain described above. In some embodiments, a putative Anellovirus ORF1 sequence comprises the sequence YNPXXDXGXXN (SEQ ID NO: 829). In some embodiments, a putative Anellovirus ORF1 sequence comprises the sequence Y[NCS]PXXDX[GASKR]XX[NTSVAK].
    • (iii) Presence of arginine-rich region: Protein sequences (e.g., putative ORF1 sequences passing the criteria described in (i) and/or (ii) above) may be filtered for those that include an arginine-rich region (e.g., as described herein). In some embodiments, a putative ORF1 sequence comprises a contiguous sequence of at least about 30, 35, 40, 45, 50, 55, 60, 65, or 70 amino acids that comprises at least 30% (e.g., at least about 20%, 25%, 30%, 35%, 40%, 45%, or 50%) arginine residues. In some embodiments, a putative ORF1 sequence comprises a contiguous sequence of about 35-40, 40-45, 45-50, 50-55, 55-60, 60-65, or 65-70 amino acids that comprises at least 30% (e.g., at least about 20%, 25%, 30%, 35%, 40%, 45%, or 50%) arginine residues. In some embodiments, the arginine-rich region is positioned at least about 30, 40, 50, 60, 70, or 80 amino acids downstream of the start codon of the putative ORF1 protein. In some embodiments, the arginine-rich region is positioned at least about 50 amino acids downstream of the start codon of the putative ORF1 protein.


In some embodiments, an ORF1 protein is identified in an Anellovirus genome sequence as described in Example 36 of PCT Publication No. WO2020/123816 (incorporated herein by reference in its entirety).


ORF2 Molecules

In some embodiments, the anellovector comprises an ORF2 molecule and/or a nucleic acid encoding an ORF2 molecule. Generally, an ORF2 molecule comprises a polypeptide having the structural features and/or activity of an Anellovirus ORF2 protein (e.g., an Anellovirus ORF2 protein as described herein, e.g., as listed in Table A1-A3)-, or a functional fragment thereof. In some embodiments, an ORF2 molecule comprises an amino acid sequence having at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to an Anellovirus ORF2 protein sequence as shown in Table A1-A3. In some embodiments, an ORF2 molecule is encoded by an ORF2 nucleic acid. In some embodiments, the ORF2 nucleic acid comprises an antisense strand, which can be directly transcribed to produce mRNA encoding the ORF2 molecule. In some embodiments, the ORF2 nucleic acid comprises a sense strand.


In some embodiments, an ORF2 molecule comprises an amino acid sequence having at least 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity to an Alphatorquevirus, Betatorquevirus, or Gammatorquevirus ORF2 protein. In some embodiments, an ORF2 molecule (e.g., an ORF2 molecule having at least 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity to an Alphatorquevirus ORF2 protein) has a length of 250 or fewer amino acids (e.g., about 150-200 amino acids). In some embodiments, an ORF2 molecule (e.g., an ORF2 molecule having at least 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity to a Betatorquevirus ORF2 protein) has a length of about 50-150 amino acids. In some embodiments, an ORF2 molecule (e.g., an ORF2 molecule having at least 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity to a Gammatorquevirus ORF2 protein) has a length of about 100-200 amino acids (e.g., about 100-150 amino acids). In some embodiments, the ORF2 molecule comprises a helix-turn-helix motif (e.g., a helix-turn-helix motif comprising two alpha helices flanking a turn region). In some embodiments, the ORF2 molecule does not comprise the amino acid sequence of the ORF2 protein of TTV isolate TA278 or TTV isolate SANBAN. In some embodiments, an ORF2 molecule has protein phosphatase activity. In some embodiments, an ORF2 molecule comprises at least one difference (e.g., a mutation, chemical modification, or epigenetic alteration) relative to a wild-type ORF2 protein, e.g., as described herein (e.g., as shown in Table A1-A3).


Conserved ORF2 Motif

In some embodiments, a polypeptide (e.g., an ORF2 molecule) described herein comprises the amino acid sequence [W/F]X7HX3CX1CX5H (SEQ ID NO: 949), wherein X″ is a contiguous sequence of any n amino acids. In embodiments, X7 indicates a contiguous sequence of any seven amino acids. In some embodiments, X3 indicates a contiguous sequence of any three amino acids. In some embodiments, X1 indicates any single amino acid. In some embodiments, X5 indicates a contiguous sequence of any five amino acids. In some embodiments, the [W/F] can be either tryptophan or phenylalanine. In some embodiments, the [W/F]X7HX3CX1CX5H (SEQ ID NO: 949) is comprised within the N22 domain of an ORF2 molecule, e.g., as described herein. In some embodiments, a genetic element described herein comprises a nucleic acid sequence (e.g., a nucleic acid sequence encoding an ORF2 molecule, e.g., as described herein) encoding the amino acid sequence [W/F]X7HX3CX1CX5H (SEQ ID NO: 949), wherein X″ is a contiguous sequence of any n amino acids.


Genetic Elements

In some embodiments, the Anelloviridae family vector (e.g., anellovector) comprises a genetic element. In some embodiments, the genetic element has one or more of the following characteristics: is substantially non-integrating with a host cell's genome, is an episomal nucleic acid, is a single stranded DNA, is circular, is about 1 to 10 kb, exists within the nucleus of the cell, can be bound by endogenous proteins, produces an effector, such as a polypeptide or nucleic acid (e.g., an RNA, iRNA, microRNA) that targets a gene, activity, or function of a host or target cell. In one embodiment, the genetic element is a substantially non-integrating DNA. In some embodiments, the genetic element comprises a packaging signal, e.g., a sequence that binds a capsid protein. In some embodiments, outside of the packaging or capsid-binding sequence, the genetic element has less than 70%, 60%, 50%, 40%, 30%, 20%, 10%, 5% sequence identity to a wild type Anellovirus nucleic acid sequence, e.g., has less than 70%, 60%, 50%, 40%, 30%, 20%, 10%, 5% sequence identity to an Anellovirus nucleic acid sequence, e.g., as described herein. In some embodiments, outside of the packaging or capsid-binding sequence, the genetic element has less than 500, 450, 400, 350, 300, 250, 200, 150, or 100 contiguous nucleotides that are at least 70%, 75%, 80%, 8%, 90%, 95%, 96%, 97%, 98%, 99%, or 100% identical to an Anellovirus nucleic acid sequence. In certain embodiments, the genetic element is a circular, single stranded DNA that comprises a promoter sequence, a sequence encoding a therapeutic effector, and a capsid binding protein. In some embodiments, the genetic element may comprise other sequences that include DNA, RNA, or artificial nucleic acids.


In some embodiments, the genetic element has at least about 70%, 75%, 80%, 8%, 90%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to an Anellovirus nucleic acid sequence, e.g., as described herein (e.g., as described in any of Tables N1-N3), or a fragment thereof, or encodes an amino acid sequence having at least about 70%, 75%, 80%, 8%, 90%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to an Anellovirus amino acid sequence (e.g., as described in any of Tables A1-A3), or a fragment thereof. In some embodiments, the genetic element comprises a sequence encoding an effector (e.g., an endogenous effector or an exogenous effector, e.g., a payload), e.g., a polypeptide effector (e.g., a protein) or nucleic acid effector (e.g., a non-coding RNA, e.g., a miRNA, siRNA, mRNA, lncRNA, RNA, DNA, an antisense RNA, gRNA).


In some embodiments, the genetic element has a length less than 20 kb (e.g., less than about 19 kb, 18 kb, 17 kb, 16 kb, 15 kb, 14 kb, 13 kb, 12 kb, 11 kb, 10 kb, 9 kb, 8 kb, 7 kb, 6 kb, 5 kb, 4 kb, 3 kb, 2 kb, 1 kb, or less). In some embodiments, the genetic element has, independently or in addition to, a length greater than 1000b (e.g., at least about 1.1 kb, 1.2 kb, 1.3 kb, 1.4 kb, 1.5 kb, 1.6 kb, 1.7 kb, 1.8 kb, 1.9 kb, 2 kb, 2.1 kb, 2.2 kb, 2.3 kb, 2.4 kb, 2.5 kb, 2.6 kb, 2.7 kb, 2.8 kb, 2.9 kb, 3 kb, 3.1 kb, 3.2 kb, 3.3 kb, 3.4 kb, 3.5 kb, 3.6 kb, 3.7 kb, 3.8 kb, 3.9 kb, 4 kb, 4.1 kb, 4.2 kb, 4.3 kb, 4.4 kb, 4.5 kb, 4.6 kb, 4.7 kb, 4.8 kb, 4.9 kb, 5 kb, or greater). In some embodiments, the genetic element has a length of about 2.5-4.6, 2.8-4.0, 3.0-3.8, or 3.2-3.7 kb. In some embodiments, the genetic element has a length of about 1.5-2.0, 1.5-2.5, 1.5-3.0, 1.5-3.5, 1.5-3.8, 1.5-3.9, 1.5-4.0, 1.5-4.5, or 1.5-5.0 kb. In some embodiments, the genetic element has a length of about 2.0-2.5, 2.0-3.0, 2.0-3.5, 2.0-3.8, 2.0-3.9, 2.0-4.0, 2.0-4.5, or 2.0-5.0 kb. In some embodiments, the genetic element has a length of about 2.5-3.0, 2.5-3.5, 2.5-3.8, 2.5-3.9, 2.5-4.0, 2.5-4.5, or 2.5-5.0 kb. In some embodiments, the genetic element has a length of about 3.0-5.0, 3.5-5.0, 4.0-5.0, or 4.5-5.0 kb. In some embodiments, the genetic element has a length of about 1.5-2.0, 2.0-2.5, 2.5-3.0, 3.0-3.5, 3.1-3.6, 3.2-3.7, 3.3-3.8, 3.4-3.9, 3.5-4.0, 4.0-4.5, or 4.5-5.0 kb.


In some embodiments, the genetic element comprises one or more of the features described herein, e.g., a sequence encoding a substantially non-pathogenic protein, a protein binding sequence, one or more sequences encoding a regulatory nucleic acid, one or more regulatory sequences, one or more sequences encoding a replication protein, and other sequences. In some embodiments, the substantially non-pathogenic protein comprises an amino acid sequence or a functional fragment thereof or a sequence having at least about 60%, 65%, 70%, 75%, 80%, 85%, 90% 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to any one of the amino acid sequences described herein, an Anellovirus amino acid sequence, e.g., as listed in any of Tables A1-A3.


In some embodiments, the double-stranded circular DNA and/or the genetic element does not comprise one or more plasmid elements (e.g., an origin of replication or a selectable marker, e.g., a resistance gene). In some embodiments, the double-stranded circular DNA and/or the genetic element does not comprise a plasmid backbone. In some embodiments, the double-stranded circular DNA and/or the genetic element does not comprise one or more bacterial plasmid elements (e.g., a bacterial origin of replication or a selectable marker, e.g., a bacterial resistance gene). In some embodiments, the double-stranded circular DNA and/or the genetic element does not comprise a bacterial plasmid backbone. In some embodiments, the double-stranded circular DNA and/or the genetic element does not comprise one or more mammalian plasmid elements (e.g., a mammalian origin of replication or a selectable marker, e.g., a mammalian resistance gene). In some embodiments, the double-stranded circular DNA and/or the genetic element does not comprise a mammalian plasmid backbone. In some embodiments, the double-stranded circular DNA and/or the genetic element does not comprise one or more insect plasmid elements (e.g., an insect origin of replication or a selectable marker, e.g., an insect resistance gene). In some embodiments, the double-stranded circular DNA and/or the genetic element does not comprise an insect plasmid backbone.


In some embodiments, a genetic element as described herein comprises a sequence (e.g., a TATA box, cap site, transcriptional start site, 5′ UTR, open reading frame (ORF), poly(A) signal, or GC-rich region sequence) as listed in any of Tables A1, A3, A5, A7, A9, A11, B1-B5, 1, 3, 5, 7, 9, 11, 13, 15, or 17 of PCT Publication No. WO2020/123816 (incorporated herein by reference in its entirety), or a sequence having at least 70% 80%, 85%, 90% 95%, 96%, 97%, 98% and 99% nucleotide sequence identity thereto.


In some embodiments, a genetic element comprises a sequence encoding an effector (e.g., an exogenous effector). In some embodiments, the effector-encoding sequence is inserted into an Anellovirus genome sequence (e.g., as described herein). In some embodiments, the effector-encoding sequence replaces a contiguous sequence (e.g., of at least 5, 10, 15, 20, 25, 30, 40, 50, 60, 70, 80, 90, 100, 200, 300, 400, 500, 600, 700, 800, 900, 1000, or more nucleotides) from the Anellovirus genome sequence. In some embodiments, the effector-encoding sequence replaces a TATA box, cap site, transcriptional start site, 5′ UTR, open reading frame (ORF), poly(A) signal, or GC-rich region sequence, or a portion thereof (e.g., a portion consisting of at least 5, 10, 15, 20, 25, 30, 40, 50, 60, 70, 80, 90, 100, 200, 300, 400, 500, 600, 700, 800, 900, 1000, or more nucleotides), e.g., as described herein, or a sequence having at least 70% 80%, 85%, 90% 95%, 96%, 97%, 98% and 99% nucleotide sequence identity thereto.


In some embodiments, the sequence of a first nucleic acid element comprised in a genetic element (e.g., a TATA box, cap site, transcriptional start site, 5′ UTR, open reading frame (ORF), poly(A) signal, or GC-rich region) overlaps with the sequence of a second nucleic acid element (e.g., a TATA box, cap site, transcriptional start site, 5′ UTR, open reading frame (ORF), poly(A) signal, or GC-rich region), e.g., by at least 5, 10, 15, 20, 25, 30, 40, 50, 60, 70, 80, 90, 100, 150, 200, 250, 300, 400, or 500 nucleotides. In some embodiments, the sequence of a first nucleic acid element comprised in a genetic element (e.g., a TATA box, cap site, transcriptional start site, 5′ UTR, open reading frame (ORF), poly(A) signal, or GC-rich region) does not overlap with the sequence of a second nucleic acid element (e.g., a TATA box, cap site, transcriptional start site, 5′ UTR, open reading frame (ORF), poly(A) signal, or GC-rich region).


Protein Binding Sequence

In some embodiments, the genetic element encodes a protein binding sequence that binds to the substantially non-pathogenic protein. In some embodiments, the protein binding sequence facilitates packaging the genetic element into the proteinaceous exterior. In some embodiments, the protein binding sequence specifically binds an arginine-rich region of the substantially non-pathogenic protein. In some embodiments, the genetic element comprises a protein binding sequence having at least 70%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to a 5′ UTR conserved domain or GC-rich domain of an Anellovirus sequence (e.g., to the reverse complement of the sequence annotated in any of Tables N1-N3).


In embodiments, the protein binding sequence has at least about 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to the reverse complement of the sequence annotated as 5′ UTR conserved domain nucleotide sequence of any of Tables N1-N3. In embodiments, the protein binding sequence has at least about 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to the reverse complement of the sequence annotated as the GC-rich domain nucleotide sequence of any of Tables N1-N3.


5′ UTR Conserved Domains

A genetic element may include an Anellovirus 5′ UTR conserved domain. Typically, the negative strand comprising the Anellovirus 5′ UTR conserved domain is packaged into a particle (e.g., an Anelloviridae family vector as described herein. In some embodiments, the Anellovirus 5′ UTR conserved domain is a wild-type Anellovirus 5′ UTR conserved domain. In some embodiments, the Anellovirus 5′ UTR conserved domain is an engineered Anellovirus 5′ UTR conserved domain having a nucleic acid sequence with at least one difference relative to the closest wild-type Anellovirus 5′ UTR conserved domain sequence. In some embodiments, the genetic element comprises a nucleic acid sequence having at least about 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to the reverse complement of the sequence annotated as 5′ UTR conserved domain nucleotide sequence of any of Tables N1-N3 or Table 38.









TABLE 38







Exemplary 5′ UTR sequences from Anelloviruses.









Source
Sequence
SEQ ID NO:





Consensus
CGGGTGCCGX1AGGTGAGTTTACACACCGX2AGT
105



CAAGGGGCAATTCGGGCTCX3GGACTGGCCGGG




CX4X5TGGG




X1 = G or T




X2 = C or A




X3 = G or A




X4 = T or C




X5 = A, C, or T






Alphatorquevirus
CGGGTGCCGGAGGTGAGTTTACACACCGCAGTC
112


Consensus 5′ UTR
AAGGGGCAATTCGGGCTCGGGACTGGCCGGGC




X1X2TGGG; wherein X1 comprises T or C, and




wherein X2 comprises A, C, or T.









Identification of 5′ UTR Sequences

In some embodiments, an Anelloviridae family virus (e.g., Anellovirus) 5′ UTR sequence can be identified within the genome of an Anelloviridae family virus (e.g., Anellovirus) (e.g., a putative Anelloviridae family virus genome identified, for example, by nucleic acid sequencing techniques, e.g., deep sequencing techniques). In some embodiments, an Anelloviridae family virus (e.g., Anellovirus) 5′ UTR sequence is identified by one or both of the following steps:

    • (i) Identification of circularization junction point: In some embodiments, a 5′ UTR will be positioned near a circularization junction point of a full-length, circularized Anelloviridae family virus (e.g., Anellovirus) genome. A circularization junction point can be identified, for example, by identifying overlapping regions of the sequence. In some embodiments, an overlapping region of the sequence can be trimmed from the sequence to produce a full-length Anelloviridae family virus (e.g., Anellovirus) genome sequence that has been circularized. In some embodiments, a genome sequence is circularized in this manner using software. Without wishing to be bound by theory, computationally circularizing a genome may result in the start position for the sequence being oriented in a non-biological. Landmarks within the sequence can be used to re-orient sequences in the proper direction. For example, landmark sequence may include sequences having substantial homology to one or more elements within an Anelloviridae family virus (e.g., Anellovirus) genome as described herein (e.g., one or more of a TATA box, cap site, initiator element, transcriptional start site, 5′ UTR conserved domain, ORF1, ORF1/1, ORF1/2, ORF2, ORF2/2, ORF2/3, ORF2t/3, three open-reading frame region, poly(A) signal, or GC-rich region of an Anelloviridae family virus (e.g., Anellovirus), e.g., as described herein).
    • (ii) Identification of 5′ UTR sequence: Once a putative Anelloviridae family virus (e.g., Anellovirus) genome sequence has been obtained, the sequence (or portions thereof, e.g., having a length between about 40-50, 50-60, 60-70, 70-80, 80-90, or 90-100 nucleotides) can be compared to one or more Anelloviridae family virus (e.g., Anellovirus) 5′ UTR sequences (e.g., as described herein) to identify sequences having substantial homology thereto. In some embodiments, a putative Anelloviridae family virus (e.g., Anellovirus) 5′ UTR region has at least 50%, 60%, 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to an Anelloviridae family virus (e.g., Anellovirus) 5′ UTR sequence as described herein.


GC-Rich Regions

A genetic element may include an Anellovirus GC-rich region. Typically, the negative strand comprising the Anellovirus GC-rich region is packaged into a particle (e.g., an Anelloviridae family vector as described herein. In some embodiments, the Anellovirus GC-rich region is a wild-type Anellovirus GC-rich region. In some embodiments, the Anellovirus GC-rich region is an engineered Anellovirus GC-rich region having a nucleic acid sequence with at least one difference relative to the closest wild-type Anellovirus GC-rich region sequence. In some embodiments, the Anellovirus GC-rich region comprises a contiguous sequence of at least 20, 25, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, or 40 consecutive nucleotides having a GC content of at least 70%, 75%, 80%, 85%, 90%, 95%, or 99%. In some embodiments, the genetic element comprises a nucleic acid sequence having at least about 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to the reverse complement of the sequence annotated as GC-rich region nucleotide sequence of any of Tables N1-N3 or Table 39.









TABLE 39







Exemplary GC-rich sequences from Anelloviruses.











SEQ




ID


Source
Sequence
NO:





Consensus
CGGCGGX1GGX2GX3X4X5CGCGCTX6CGCGC
120



GCX7X8X9X10CX11X12X13X14GGGGX15X16X17X18




X19X20X21GCX22X23X24X25CCCCCCCX26CGCGC




ATX27X28GCX29CGGGX30CCCCCCCCCX31X32




X33GGGGGGCTCCGX34CCCCCCGGCCCCCC




X1 = G or C




X2 = G, C, or absent




X3 = C or absent




X4 = G or C




X5 = G or C




X6 = T, G, or A




X7 = G or C




X8 = G or absent




X9 = C or absent




X10 = C or absent




X11 = G, A, or absent




X12 = G or C




X13 = C or T




X14 = G or A




X15 = G or A




X16 = A, G, T, or absent




X17 = G, C, or absent




X18 = G, C, or absent




X19 = C, A, or absent




X20 = C or A




X21 = T or A




X22 = G or C




X23 = G, T, or absent




X24 = C or absent




X25 = G, C, or absent




X26 = G or C




X27 = G or absent




X28 = C or absent




X29 = G or A




X30 = G or T




X31 = C, T, or absent




X32 = G, C, A, or absent




X33 = G or C




X34 = C or absent









Effectors

In some embodiments, the genetic element encodes an effector, e.g., an exogenous effector. In some embodiments, the genetic element comprises a therapeutic expression sequence, e.g., a sequence that encodes an effector such as a therapeutic peptide or polypeptide, e.g., an intracellular peptide or intracellular polypeptide, a secreted polypeptide, or a protein replacement therapeutic. In some embodiments, the intracellular polypeptide is a cytosolic polypeptide, a regulatory intracellular peptide, or an anti-apoptotic agent. In some embodiments, the secreted polypeptide is a cytokine, a hormone, a growth factor, an antibody molecule that binds a hormone or a growth factor, a polypeptide that specifically binds to a VEGF, or a clotting-associated factor. In some embodiments, an effector described herein comprises an anti-VEGF antibody molecule. In some embodiments, the protein replacement therapeutic is an enzymatic effector, a non-enzymatic effector, erythropoietin, a micro-dystrophin, or a functional variant of a wild-type protein.


In some embodiments, the genetic element includes a sequence encoding a protein e.g., a therapeutic protein. Some examples of therapeutic proteins may include, but are not limited to, a hormone, a cytokine, an enzyme, an antibody (e.g., one or a plurality of polypeptides encoding at least a heavy chain or a light chain), a transcription factor, a receptor (e.g., a membrane receptor), a ligand, a membrane transporter, a secreted protein, a peptide, a carrier protein, a structural protein, a nuclease, or a component thereof.


Some examples of peptides include, but are not limited to, fluorescent tag or marker, antigen, peptide therapeutic, synthetic or analog peptide from naturally-bioactive peptide, agonist or antagonist peptide, anti-microbial peptide, a targeting or cytotoxic peptide, a degradation or self-destruction peptide, and degradation or self-destruction peptides.


In some embodiments, the effector comprises a regeneration, repair, and fibrosis factor (e.g., a growth factor, an antibody or fragment thereof against such growth factors, or miRNAs that promote regeneration and repair). In some embodiments, the effector comprises a transformation factor (e.g., protein factors or miRNAs that transform fibroblasts into differentiated cells). In some embodiments, the effector comprises a protein that stimulates cellular regeneration. In some embodiments, the effector comprises a secreted STING modulator (e.g., a STING inhibitor or a STING activator).


In some embodiments, the genetic element comprises an effector-encoding sequence. In some embodiments, the effector comprises a regulatory nucleic acid (e.g., miRNA, siRNA, mRNA, lncRNA, dsRNA, shRNA, RNA, DNA, an antisense RNA, or a gRNA).


In some embodiments, the genetic element comprises a sequence that encodes small peptides, peptidomimetics (e.g., peptoids), amino acids, and amino acid analogs.


In some embodiments, the effector mentioned in this section may also be a functional variant, homologue, or fragment thereof.


In some embodiments, the genetic element comprises a sequence encoding an exogenous effector, e.g., as described in PCT publication No. WO/2020/123753 pp. 337-366, PCT publication No. WO/2020/123773 pp. 334-361, and PCT publication No. WO/2020/123795 pp. 333-358, each of which is incorporated by reference herein in its entirety.


Regulatory Sequences

In some embodiments, the genetic element comprises a regulatory sequence, e.g., a promoter or an enhancer, operably linked to the sequence encoding the effector.


In some embodiments, a promoter includes a DNA sequence that is located adjacent to a DNA sequence that encodes an expression product. A promoter may be linked operatively to the adjacent DNA sequence. A promoter typically increases an amount of product expressed from the DNA sequence as compared to an amount of the expressed product when no promoter exists. A promoter from one organism can be utilized to enhance product expression from the DNA sequence that originates from another organism. For example, a vertebrate promoter may be used for the expression of jellyfish GFP in vertebrates. In addition, one promoter element can increase an amount of products expressed for multiple DNA sequences attached in tandem. Hence, one promoter element can enhance the expression of one or more products. Multiple promoter elements are well-known to persons of ordinary skill in the art.


In some embodiments, a native promoter for a gene or nucleic acid sequence of interest is used. The native promoter may be used when it is desired that expression of the gene or the nucleic acid sequence should mimic the native expression. The native promoter may be used when expression of the gene or other nucleic acid sequence must be regulated temporally or developmentally, or in a tissue-specific manner, or in response to specific transcriptional stimuli. In a further embodiment, other native expression control elements, such as enhancer elements, polyadenylation sites or Kozak consensus sequences may also be used to mimic the native expression.


In one embodiment, high-level constitutive expression is desired. Examples of such promoters include, without limitation, the retroviral Rous sarcoma virus (RSV) long terminal repeat (LTR) promoter/enhancer, the cytomegalovirus (CMV) immediate early promoter/enhancer (see, e.g., Boshart et al, Cell, 41:521-530 (1985)), the SV40 promoter, the dihydrofolate reductase promoter, the cytoplasmic .beta.-actin promoter and the phosphoglycerol kinase (PGK) promoter.


In another embodiment, inducible promoters may be desired. Inducible promoters are those which are regulated by exogenously supplied compounds, either in cis or in trans. Other types of inducible promoters which may be useful in this context are those which are regulated by a specific physiological state, e.g., temperature, acute phase, or in replicating cells only.


In some embodiments, the genetic element comprises a gene operably linked to a tissue-specific promoter.


The genetic element may include an enhancer, e.g., a DNA sequence that is located adjacent to the DNA sequence that encodes a gene. Enhancer elements are typically located upstream of a promoter element or can be located downstream of or within a coding DNA sequence (e.g., a DNA sequence transcribed or translated into a product or products). Hence, an enhancer element can be located 100 base pairs, 200 base pairs, or 300 or more base pairs upstream or downstream of a DNA sequence that encodes the product. Enhancer elements can increase an amount of recombinant product expressed from a DNA sequence above increased expression afforded by a promoter element. Multiple enhancer elements are readily available to persons of ordinary skill in the art.


Surface Moieties

An Anelloviridae family vector as described herein may, in some instances, include one or more moieties attached to its surface (e.g., a surface moiety that can act as an effector and/or a targeting agent). In some instances, an Anelloviridae family vector comprises more than one distinct surface moiety (e.g., a first surface moiety having an effector function as described herein and a second surface moiety that targets the Anelloviridae family vector to a cell or tissue of interest). In some instances, the surface moiety is covalently attached to the surface of the Anelloviridae family vector. For example, the surface moiety may be covalently attached to the proteinaceous exterior or a component thereof (e.g., covalently attached to an ORF1 molecule of the proteinaceous exterior). In certain embodiments, the surface moiety is fused to an ORF1 molecule. In some instances, the surface moiety is noncovalently attached to the surface of the Anelloviridae family vector. For example, the surface moiety may be noncovalently bound to the proteinaceous exterior or a component thereof (e.g., noncovalently bound to an ORF1 molecule of the proteinaceous exterior). In certain embodiments, the surface moiety comprises a region that specifically binds to a cognate moiety on or attached to the ORF1 molecule. In an embodiment, the ORF1 molecule comprises a binding moiety (e.g., an antibody molecule) that specifically recognizes an epitope on the region on the surface moiety. In an embodiment, the surface moiety comprises a binding moiety (e.g., an antibody molecule) that specifically recognizes an epitope on the ORF1 molecule.


The surface moiety can, in some instances, comprise a polypeptide. The surface moiety may, in some instances, comprise a nucleic acid molecule (e.g., DNA and/or RNA). The surface moiety may, in some instances, comprise a small molecule. In some instances, a surface moiety comprises an antigen (e.g., an antigen recognized by the immune system of a subject to be delivered the Anelloviridae family vector). In some instances, a surface moiety as described herein comprises a ligand (e.g., a ligand that binds specifically to a receptor on a target cell).


In some instances, the surface moiety comprises an effector function (e.g., as described herein). For example, the surface moiety may modulate a biological activity, e.g., of a target cell or organ. In some instances, the surface moiety induces modulation of the biological activity via binding to a cognate moiety on a target cell. For example, the surface moiety may comprise a ligand that binds to a receptor on the surface of the target cell, e.g., wherein binding of the surface moiety to the receptor initiates a downstream signaling cascade of interest. In some instances, the effector activity comprises increasing or decreasing enzymatic activity, gene expression, cell signaling, and/or cellular or organ function within a target cell or organ. Effector activities may also include binding regulatory proteins to modulate activity of the regulator, such as transcription or translation. Effector activities also may include activator or inhibitor functions.


In some instances, the surface moiety can target the Anelloviridae family vector to a target cell. For example, the surface moiety may specifically bind to a cognate moiety on the surface of the target cell. The cognate moiety on the surface of the target cell may be, for example, a molecule specifically expressed or preferentially expressed by the target cell. The cognate moiety may be, for example, a polypeptide, lipid, sugar, or small molecule. In certain embodiments, the cognate moiety is a transmembrane protein (e.g., comprising an extracellular domain that binds to the surface moiety of the Anelloviridae family vector). In certain embodiments, the cognate moiety is tethered to the surface of the cell (e.g., via a GPI anchor). In some instances, the surface moiety provides a tropism (e.g., to a target tissue or target cell type) for the Anelloviridae family vector.


In an aspect, the disclosure provides an ORF1 molecule comprising: (i) the amino acid sequence of an Anellovirus ORF1 protein, or an amino acid sequence having at least 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity thereto; and (ii) a click handle (e.g., an NHS click handle or a maleimide click handle, e.g., as described herein). In certain embodiments, the click handle is covalently attached to the ORF1 molecule. In certain embodiments, the click handle is noncovalently attached to the ORF1 molecule. In certain embodiments, the click handle is used to attach the ORF1 molecule to a surface moiety, e.g., via a click reaction, e.g., as described herein.


A “click handle,” as that term is used herein, refers to a chemical moiety that is capable of reacting with a second click handle in a click reaction. In some embodiments, a click handle comprises an NHS moiety and/or a maleimide moiety. In certain embodiments, a click handle comprises a DBCO moiety. In certain embodiments, a click handle comprises an azide moiety. In some embodiments, a click handle is attached to a polypeptide (e.g., an ORF1 molecule). In other embodiments, a click handle comprises a reactive group capable of forming a covalent bond with a polypeptide (e.g., an ORF1 molecule). A “click reaction”, as that term is used herein, refers to a range of reactions used to covalently link a first and a second moiety, for convenient production of linked products. It typically has one or more of the following characteristics: it is fast, is specific, is high-yield, is efficient, is spontaneous, does not significantly alter biocompatibility of the linked entities, has a high reaction rate, produces a stable product, favors production of a single reaction product, has high atom economy, is chemoselective, is modular, is stereoselective, is insensitive to oxygen, is insensitive to water, is high purity, generates only inoffensive or relatively non-toxic byproducts that can be removed by nonchromatographic methods (e.g., crystallization or distillation), needs no solvent or can be performed in a solvent that is benign or physiologically compatible, e.g., water, stable under physiological conditions. Examples include an alkyne/azide reaction, a diene/dienophile reaction, or a thiol/alkene reaction. Other reactions can be used.


II. Compositions and Methods for Making Anelloviridae Family Vectors

The present disclosure provides, in some aspects, Anelloviridae family vectors (e.g., anellovectors) and methods thereof for delivering effectors. In some embodiments, the Anelloviridae family vectors (e.g., anellovectors) or components thereof can be made as described below. In some embodiments, the compositions and methods described herein can be used to produce a genetic element or a genetic element construct. In some embodiments, the compositions and methods described herein can be used to produce one or more Anelloviridae family virus capsid proteins (e.g., Anellovirus ORF) molecules (e.g., an ORF1, ORF2, ORF2/2, ORF2/3, ORF1/1, or ORF1/2molecule, or a functional fragment or splice variant thereof). In some embodiments, the compositions and methods described herein can be used to produce a proteinaceous exterior or a component thereof (e.g., an ORF1 molecule), e.g., in a host cell. In some embodiments, the Anelloviridae family vector (e.g., anellovector) or components thereof can be made using a tandem construct, e.g., as described in PCT Publication No. WO 2021252955, which is incorporated herein by reference in its entirety. In some embodiments, the Anelloviridae family vector (e.g., anellovector) or components thereof (e.g., an Anelloviridae family polypeptide, e.g., an ORF1, ORF2, ORF2/2, ORF2/3, ORF1/1, and/or ORF1/2 molecule, or a functional fragment or splice variant thereof) can be made using a bacmid/insect cell system, e.g., as described as described in PCT Publication No. WO 2021/252943, which is incorporated herein by reference in its entirety. Methods of producing an anellovector using a host cell are described, for example, below in the section entitled “Host Cells and Methods of Using Host Cells for Producing an Anellovector”. Method of producing an anellovector in a cell-free system are described, for example, below in the section entitled “In vitro assembly methods”.


Without wishing to be bound by theory, rolling circle amplification may occur via Rep protein binding to a Rep binding site (e.g., comprising a 5′ UTR, e.g., comprising a hairpin loop and/or an origin of replication, e.g., as described herein) positioned 5′ relative to (or within the 5′ region of) the genetic element region. The Rep protein may then proceed through the genetic element region, resulting in the synthesis of the genetic element. The genetic element may then be circularized and then enclosed within a proteinaceous exterior to form an Anelloviridae family vector (e.g., anellovector).


Genetic Element Constructs, e.g., for Assembly of Anelloviridae Family Vectors

In some methods of making as described herein, the genetic element is made using a genetic element construct, wherein the genetic element construct can act as a template for the production of the genetic element. In some embodiments, a genetic element construct comprises a genetic element region and optionally other sequence such as vector backbone. A genetic element construct may be any nucleic acid construct suitable for delivery of the sequence of the genetic element into a host cell in which the genetic element can be enclosed within a proteinaceous exterior. In some embodiments, the genetic element construct comprises a promoter. In some embodiments, the genetic element construct is a linear nucleic acid molecule. In some embodiments, the genetic element construct is a circular nucleic acid molecule (e.g., a plasmid, viral nucleic acid, bacmid, artificial chromosome, or a minicircle, e.g., as described herein). In some embodiments, a double-stranded circular nucleic acid (e.g., a minicircle) can be excised from a plasmid (e.g., by in vitro circularization). In some embodiments, in vitro circularized DNA constructs can be produced by digesting a genetic element construct (e.g., a plasmid comprising the sequence of a genetic element) to be packaged, such that the genetic element sequence is excised as a linear DNA molecule. The resultant linear DNA can then be ligated, e.g., using a DNA ligase, to form a double-stranded circular DNA (e.g., a minicircle). In embodiments, the double-stranded circular nucleic acid construct (e.g., minicircle) can be introduced into a host cell, in which it can be converted into or used as a template for generating single-stranded circular genetic elements, e.g., as described herein. In some embodiments, the genetic element is generated by a polymerase based on a template sequence in the nucleic acid construct. In some embodiments, the polymerase produces a single-stranded copy of the genetic element sequence, which can optionally be circularized to form a genetic element as described herein.


The genetic element construct may, in some embodiments, be double-stranded. In some embodiments, the genetic element construct comprises RNA. In some embodiments, the genetic element construct comprises one or more modified nucleotides.


Tandem Constructs

In some embodiments, a genetic element construct comprises a first copy of a genetic element sequence (e.g., the nucleic acid sequence of a genetic element, e.g., as described herein) and at least a portion of a second copy of a genetic element sequence (e.g., the nucleic acid sequence of the same genetic element, or the nucleic acid sequence of a different genetic element), arranged in tandem. Genetic element constructs having such a structure are generally referred to herein as tandem constructs. Such tandem constructs are used for producing an Anelloviridae family vector (e.g., anellovector) genetic element. The first copy of the genetic element sequence and the second copy of the genetic element sequence may, in some instances, be immediately adjacent to each other on the genetic acid construct. In other instances, the first copy of the genetic element sequence and the second copy of the genetic element sequence may be separated, e.g., by a spacer sequence. Without being bound by theory, a tandem construct described herein may, in some embodiments, replicate by rolling circle replication. In some embodiments, a tandem construct is a plasmid. In some embodiments, a tandem construct is circular. In some embodiments, a tandem construct is linear. In some embodiments, a tandem construct is single-stranded. In some embodiments, a tandem construct is double-stranded. In some embodiments, a tandem construct is DNA.


Additional descriptions of tandem constructs that can be used with the invention are described, for example, PCT Publication No. WO 2021252955, incorporated herein by reference in its entirety.


Cis/Trans Constructs

In some embodiments, a genetic element construct as described herein comprises one or more sequences encoding one or more Anelloviridae family virus ORFs, e.g., proteinaceous exterior components (e.g., polypeptides encoded by an Anellovirus ORF1 nucleic acid, e.g., as described herein). For example, the genetic element construct may comprise a nucleic acid sequence encoding an Anellovirus ORF1 molecule. Such genetic element constructs can be suitable for introducing the genetic element and the Anelloviridae family virus ORF(s) into a host cell in cis. In other embodiments, a genetic element construct as described herein does not comprise sequences encoding one or more Anelloviridae family virus ORFs, e.g., proteinaceous exterior components (e.g., polypeptides encoded by an Anellovirus ORF1 nucleic acid, e.g., as described herein). For example, the genetic element construct may not comprise a nucleic acid sequence encoding an Anellovirus ORF1 molecule. Such genetic element constructs can be suitable for introducing the genetic element into a host cell, with the one or more Anelloviridae family virus ORFs to be provided in trans (e.g., via introduction of a second nucleic acid construct encoding one or more of the Anelloviridae family virus ORFs, or via an Anelloviridae family virus ORF cassette integrated into the genome of the host cell). In some embodiments, an ORF1 molecule is provided in trans, e.g., as described herein. In some embodiments, an ORF2 molecule is provided in trans, e.g., as described herein. In some embodiments, an ORF1 molecule and an ORF2 molecule are both provided in trans, e.g., as described herein.


In some embodiments, the genetic element construct comprises a sequence encoding an Anellovirus ORF1 molecule, or a splice variant or functional fragment thereof (e.g., a jelly-roll region, e.g., as described herein). In embodiments, the portion of the genetic element that does not comprise the sequence of the genetic element comprises the sequence encoding the Anellovirus ORF1 molecule, or splice variant or functional fragment thereof (e.g., in a cassette comprising a promoter and the sequence encoding the Anellovirus ORF1 molecule, or splice variant or functional fragment thereof). In further embodiments, the portion of the construct comprising the sequence of the genetic element comprises a sequence encoding an Anellovirus ORF1 molecule, or a splice variant or functional fragment thereof (e.g., a jelly-roll region, e.g., as described herein). In embodiments, enclosure of such a genetic element in a proteinaceous exterior (e.g., as described herein) produces a replication-component Anelloviridae family vector (e.g., anellovector) (e.g., an Anelloviridae family vector that upon infecting a cell, enables the cell to produce additional copies of the anellovector without introducing further nucleic acid constructs, e.g., encoding one or more Anelloviridae family virus ORFs as described herein, into the cell).


In other embodiments, the genetic element does not comprise a sequence encoding an Anellovirus ORF1 molecule, or a splice variant or functional fragment thereof (e.g., a jelly-roll region, e.g., as described herein). In embodiments, enclosure of such a genetic element in a proteinaceous exterior (e.g., as described herein) produces a replication-incompetent Anelloviridae family vector (e.g., anellovector) (e.g., an Anelloviridae family vector that, upon infecting a cell, does not enable the infected cell to produce additional Anelloviridae family vector, e.g., in the absence of one or more additional constructs, e.g., encoding one or more Anellovirus ORFs as described herein).


Recombinase-Based Production of Genetic Elements and Anellovectors

A genetic element for an Anelloviridae family vector (e.g., an anellovector) may be produced via site-specific recombination of a genetic element construct to produce a circular nucleic acid molecule comprising the genetic element sequence. In some embodiments, the circular nucleic acid molecule is a double-stranded DNA minicircle. In some embodiments, the circular nucleic acid molecule is in turn converted to a circular single-stranded DNA molecule, which can in turn serve as the genetic element of an Anelloviridae family vector (e.g., an anellovector) as described herein. Generally, the genetic element construct comprises a set of recombinase recognition sequences flanking the sequence of a genetic element of an Anelloviridae family vector. The recombinase recognition sites may be recognized by a site-specific recombinase. The remainder of the genetic element construct may, in some instances, comprise a vector backbone comprising elements for replication of the construct in a cell, such as a mammalian cell.


Contacting the genetic element construct with the site-specific recombinase (e.g., in a host cell) may result in excision of the genetic element sequence from the remainder of the construct and the formation of two circular nucleic acid molecules, one comprising the genetic element sequence and the other comprising the remainder of the vector backbone. In some embodiments, the circular nucleic acid molecule (e.g., a minicircle) comprising the genetic element sequence is converted to cssDNA in the host cell (e.g., a mammalian host cell), and is then encapsulated in a proteinaceous exterior comprising ORF1 molecules (e.g., as described herein) to produce an Anelloviridae family vector.


In some embodiments, a site specific recombinase-based system for producing genetic elements comprises three plasmids: (1) a first plasmid (e.g., a vector plasmid) comprising the sequence of the genetic element, flanked by a pair of recombinase recognition sites; (2) an expression plasmid comprising a cassette encoding a site-specific recombinase (e.g., as described herein) capable of recognizing the recombinase recognition sites; and (3) a plasmid (e.g., a self-replicating rescue (SRR) plasmid) providing one or more Anelloviridae family viral proteins (e.g., Anellovirus ORF1, ORF3, and/or ORF3 molecules), including a capsid protein (e.g., an ORF1 molecule, e.g., as described herein).


In some embodiments, a site specific recombinase-based system for producing genetic elements comprises two plasmids: (1) a first plasmid (e.g., a vector plasmid) comprising the sequence of the genetic element, flanked by a pair of recombinase recognition sites; and (2) a second plasmid (e.g., a self-replicating rescue (SRR) plasmid) providing one or more Anelloviridae family viral proteins (e.g., Anellovirus ORF1, ORF3, and/or ORF3 molecules), including a capsid protein (e.g., an ORF1 molecule, e.g., as described herein).


Self-Replicating Rescue (SRR) Constructs (e.g., SRR Plasmids)

In some embodiments, a rescue construct can be used to provide one or more Anelloviridae family viral proteins, or functional fragments or variants thereof (e.g., one or more of Anellovirus ORF1, ORF2, and/or ORF3 molecules). In some embodiments, the rescue construct is capable of self-replicating in a host cell (e.g., a self-replicating rescue (SRR) plasmid). In some embodiments, the SRR plasmid can be used in the site-specific recombinase based systems.


In some embodiments, the rescue construct includes an expression cassette comprising the protein coding sequence of an Anelloviridae family virus (e.g., an Anellovirus as described herein), or a sequence having at least 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% identity thereto. In some embodiments, the rescue construct includes a sequence encoding a replication protein (e.g., a large T antigen, a PCV Rep, or a PCV Rep′), or a sequence having at least 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% identity thereto. In some embodiments, the rescue construct includes an exogenous origin of replication (e.g., a viral origin, e.g., an SV40, PCV, or AAV origin). In certain embodiments, the replication protein coding sequence is downstream of an internal ribosome entry site (IRES) positioned downstream of the expression cassette comprising the protein coding sequence of the Anelloviridae family virus. In certain embodiments, the replication protein coding sequence is comprised in a separate cassette from the expression cassette comprising the protein coding sequence of the Anelloviridae family virus. In some embodiments, the rescue construct further comprises one or more additional expression cassettes, e.g., encoding a site-specific recombinase, an Anelloviridae family viral protein (e.g., an Anellovirus ORF1 molecule), a replication protein and/or viral origin (e.g., as described herein), or another transgene of interest.


In some embodiments, the rescue construct comprises a single expression cassette (e.g., as described above). In some embodiments, the rescue construct comprises two expression cassettes. In some embodiments, the rescue construct comprises three expression cassettes. In some embodiments, the rescue construct comprises four expression cassettes. In some embodiments, the rescue construct comprises five or more expression cassettes. The exemplary expression cassettes described herein can be positioned in any order within the rescue construct. In some embodiments, one or more of the expression cassettes is in the opposite orientation relative to one or more of the other expression cassettes. In other embodiments, all of the expression cassettes in a rescue construct are in the same orientation relative to each other.


In some embodiments, the rescue construct does not contain an Anellovirus NCR sequence (e.g. does not contain an Anellovirus 5′ NCR sequence and/or an Anellovirus 3′ NCR sequence). In some embodiments, the rescue construct does not comprise an Anellovirus 5′ UTR conserved domain. In some embodiments, the rescue construct does not comprise an Anellovirus GC-rich region. In some embodiments, the rescue construct does not contain Anellovirus sequences homologous to the vector plasmid (e.g., a contiguous sequence of at least 10, 20, 30, 40, 50, 60, 70, 80, 90, 100, 125, 150, 175, 200, 250, 300, 400, or 500 nucleotides having sequence identity to any Anellovirus sequence of the same length in the vector plasmid).


Exemplary SRR plasmids that can, in some embodiments, be used in a site-specific recombinase system as described herein (e.g., a two-plasmid system or three-plasmid system as described herein) are provided in Table WI.









TABLE W1





Exemplary Ring19 SRR plasmid (pRTx-3525)
















Name
pRTx-3525





Type
Plasmid





Description
phEF1a_Ring19-UTR-FullORF_SVLT_SV40ori.





Length
9391 bp











1
TCGCGCGTTT CGGTGATGAC GGTGAAAACC TCTGACACAT GCAGCTCCCG GAGACGGTCA





61
CAGCTTGTCT GTAAGCGGAT GCCGGGAGCA GACAAGCCCG TCAGGGCGCG TCAGCGGGTG





121
TTGGCGGGTG TCGGGGCTGG CTTAACTATG CGGCATCAGA GCAGATTGTA CTGAGAGTGC





181
ACCATATGCG GTGTGAAATA CCGCACAGAT GCGTAAGGAG AAAATACCGC ATCAGGCGCC





241
ATTCGCCATT CAGGCTGCGC AACTGTTGGG AAGGGCGATC GGTGCGGGCC TCTTCGCTAT





301
TACGCCAGCT GGCGAAAGGG GGATGTGCTG CAAGGCGATT AAGTTGGGTA ACGCCAGGGT





361
TTTCCCAGTC ACGACGTTGT AAAACGACGG CCAGAGAATT CGAGCTCGGT ACCTCGCGAA





421
TACATCTAGA TATGGTTGGC TCCGGTGCCC GTCAGTGGGC AGAGCGCACA TCGCCCACAG





481
TCCCCGAGAA GTTGTGGGGA GGGGTCGGCA ATTGAACCGG TGCCTAGAGA AGGTGGCGCG





541
GGGTAAACTG GGAAAGTGAT GTCGTGTACT GGCTCCGCCT TTTTCCCGAG GGTGGGGGAG





601
AACCGTATAT AAGTGCAGTA GTCGCCGTGA ACGTTCTTTT TCGCAACGGG TTTGCCGCCA





661
GAACACAGGT AAGTGCCGTG TGTGGTTCCC GCGGGCCTGG CCTCTTTACG GGTTATGGCC





721
CTTGCGTGCC TTGAATTACT TCCACCTGGC TGCAGTACGT GATTCTTGAT CCCGAGCTTC





781
GGGTTGGAAG TGGGTGGGAG AGTTCGAGGC CTTGCGCTTA AGGAGCCCCT TCGCCTCGTG





841
CTTGAGTTGA GGCCTGGCCT GGGCGCTGGG GCCACCGCGT GCGAATCTGG TGGCACCTTC





901
GCGCCTGTCT CGCTGCTTTC GATAAGTCTC TAGCCATTTA AAATTTTTGA TGACCTGCTG





961
CGACGCTTTT TTTCTGGCAA GATAGTCTTG TAAATGCGGG CCAAGATCTG CACACTGGTA





1021
TTTCGGTTTT TGGGGCCGCG GGCGGCGACG GGGCCCGTGC GTCCCAGCGC ACATGTTCGG





1081
CGAGGCGGGG CCTGCGAGCG CGGCCACCGA GAATCGGACG GGGGTAGTCT CAAGCTGGCC





1141
GGCCTGCTCT GGTGCCTGGC CTCGCGCCGC CGTGTATCGC CCCGCCCTGG GGGCAAGGC





1201
TGGCCCGGTC GGCACCAGTT GCGTGAGCGG AAAGATGGCC GCTTCCCGGC CCTGCTGCAG





1261
GGAGCTCAAA ATGGAGGACG CGGCGCTCGG GAGAGCGGGC GGGTGAGTCA CCCACACAAA





1321
GGAAAAGGGC CTTTCCGTCC TCAGCCGTCG CTTCATGTGA CTCCACGGAG TACCGGGCGC





1381
CGTCCAGGCA CCTCGATTAG TTCTCGAGCT TTTGGAGTAC GTCGTCTTTA GGTTGGGGGG





1441
AGGGGTTTTA TGCGATGGAG TTTCCCCACA CTGAGTGGGT GGAGACTGAA GTTAGGCCAG





1501
CTTGGCACTT GATGTAATTC TCCTTGGAAT TTGCCCTTTT TGAGTTTGGA TCTTGGTTCA





1561
TTCTCAAGCC TCAGACAGTG GTTCAAAGTT TTTTTCTTCC ATTTCAGGTG GATGTTTATG





1621
CCGCCAGACG GAGACGGGAT CACTTCAGTG ACTCCAGGCT GAACTTGGGC GGGAGCCGAA





1681
GGTGAGTGCA ACCACCGTAG TCTAGGGGCA ATTCGGGCTA GTTCAGTATG GCGGAACGGG





1741
CAAGAAACTT AAATATTATT ATTTTACAGA TGCAAATACA ACCACCTATT AGAACCTTCA





1801
AACAAACAAT TTCAGATTGG AAAAACTTAA TTGTCCACGT TCACGACAAC ATTTGCAACT





1861
GCAATAAACC ATTAGAACAC ACTATTGATA CCTGTATCAC CAATCCAGAT GAATTAAGAT





1921
TAAACAAATC TACTAAACAA CAACTACAAA AATGCCTTGG TACCCCAGAA GAAGATACCC





1981
AAGAAGACGT TATCGATGGC TTCGCAGATG GAGAGCTAGA CGCCCTTTTC GCCCAAGATA





2041
CAGAAGAAGA TACTGGGTAA GAAACTATTC TCGAAAGAGA AAACTATTTA AAATAACAAC





2101
CAAAGAATGG CAACCAAAAG TTATAAGAAA GACTCATGTA AAGGGCACCT ATCCTTTGTT





2161
TCTTTGTACA AAGCACAGAA TTAACAATAA TATGATACAA TATTTAGACT CTATAGCTCC





2221
AGAACACTAT TACGGAGGAG GAGGATTTTC AATAATGCAA TTTTCCTTAC AAGCCTTATA





2281
TGAAGAATTT ATAAAAGCAA AAAACTGGTG GACTAATACA AACTGCTTTT TACCACTTGT





2341
AAGATATATG GGTTGCTCAT TCAAATTTTA TAAAACTGAA TTTTATGATT ATATTGTACT





2401
AATTGAAAGA TGTTATCCAC TTGCTTGTAC TGATGAAATG TACTTATCTA CTCAACCTAG





2461
TATTATGATG CTTACAAGAA AATGTATTTT TGTACCATGC AAACAAAACA GCAAAGGTAA





2521
AAAACCTTAC AAAAAAGTTA GAGTAAGACC ACCTTCACAA ATGACTACAG GATGGCATTT





2581
CTCACAAGAC TTAGCAAACA TGCCACTTGT AGTACTAAAA ACTTCAGTAT GCAGCTTTGA





2641
CAGATATTAC ACAGACAGTA CAGCTAAATC AACCACAATA GGCTTTAAAA CACTTAACAC





2701
ACAAACATTT AGATATCATG ACTGGCAGGA ACCACCTACA ACAGGATACA AACCACAAAA





2761
CCTACTATGG TTTTATGGAG CAGAAAACGG ATCACCAGTA GACCCCAACA ACACAATAGT





2821
ATCAAACCTA ATATACTTAG GAGGCACAGG ACCTTATGAA AAAGGCACAC CAATAAAAAC





2881
AAACATAAGC AATTACTTTT CAGAGCCTAA ACTGTGGGGA AATATATTTC ACGATGATTA





2941
TACATCAGGA ACATCACCCG TGTTTGTTAC AAACAAATCA CCATCAGAAA TTAAAACCGC





3001
ATGGAACACT ATAAAAGACT TAACTGTTAA AGCTAGCGGT GTATTTACAT TAAGAACAAT





3061
TCCACTATGG CTACCTTGCA GATACAACCC ATTTGCAGAC AAAGCAACCA ACAACAAAAT





3121
ATGGCTAGTT TCTATACATT CAGACCACAC AGAATGGAAA CCAATAGACA ATCCATTACT





3181
ACAACGAACA GACCTTCCTT TATGGTTACT TGTATGGGGT TGGCAAGATT GGCAGAAAAA





3241
AAACCAACAA ACTTCACAAC CTGATATTAA TTATTTAACA GTAATATCTT CACCATATAT





3301
ATCATGCTAC CCAAAATTAG ATTACTATGT GTTACTAGAT GAAGGATTTT GGGAGGGTCA





3361
CTCAACATAC ATAGAGTCAA TTACAGACTC AGACAAAAAA CACTGGTACC CTAAAAATAG





3421
ATTTCAAATA GAAACACTTA ATCTAATAGC TAACACAGGT CCAGGAACTG TAAAACTAAG





3481
AGAAAACCAA GCAGCAGAAG GTCACATGGT ATATCGCTTT AATTTTAAGC TTGGAGGATG





3541
TCCCGCACCG ATGGAAAAAA TATGTGACCC TAGCAAACAA TCCAAATATC CTATTCCCAA





3601
TAACCAGCAA CAAACAACTT CGTTGCAGAG TCCAGAAAAC CCAATTCAAA CCTATCTCTA





3661
CGACTTCGAC GAAAGGAGGG GCCTACTTAC AGAAAGAGCT ACAAAAAGAA TCAAACAAGA





3721
TCACACATCT GAAAAAACTG TTTTGCCATT TACAGGAGCA GCAACAGACC TCCCCATACT





3781
CCAAACAACA TCACAGGAGG AAAGCTCCTC GGAAGAAGAA GAAGAGCAAC AAGCGGAGAA





3841
GAAACTACTC CAGCTCCGAA GAAAGCAGCA CCGACTCCGG GAGCGAATCC TCCAGCTATT





3901
AGACATACAA AATACATAAT AAAACAAAGT ACTGTAAAAA TTGATATGTT TGGAGATACT





3961
CATGTACCTA ACCGTAGAAT GACCCCAGAA GAATTTGAAC AAGAACTAAT TGTCGCTGGT





4021
GTTTTTCGCA GACCTCCTTG TTACTATATA AAAGATAGAC CTACTTATCC TTATGTACCA





4081
AAACCTACTG ATGAAAAATG TATGGTAAAC TTTGACTTAA ACTTTCCTTA ATAAAATGAA





4141
TGCAATTGTT GTTGTTAACG GGGATCCTCA TCGCGGCCGC TACGTAAATT CCGCCCCCCC





4201
CCCCCCTCTC CCTCCCCCCC CCCTAACGTT ACTGGCCGAA GCCGCTTGGA ATAAGGCCGG





4261
TGTGCGTTTG TCTATATGTT ATTTTCCACC ATATTGCCGT CTTTTGGCAA TGTGAGGGCC





4321
CGGAAACCTG GCCCTGTCTT CTTGACGAGC ATTCCTAGGG GTCTTTCCCC TCTCGCCAAA





4381
GGAATGCAAG GTCTGTTGAA TGTCGTGAAG GAAGCAGTTC CTCTGGAAGC TTCTTGAAGA





4441
CAAACAACGT CTGTAGCGAC CCTTTGCAGG CAGCGGAACC CCCCACCTGG CGACAGGTGC





4501
CTCTGCGGCC AAAAGCCACG TGTATAAGAT ACACCTGCAA AGGCGGCACA ACCCCAGTGC





4561
CACGTTGTGA GTTGGATAGT TGTGGAAAGA GTCAAATGGC TCTCCTCAAG CGTATTCAAC





4621
AAGGGGCTGA AGGATGCCCA GAAGGTACCC CATTGTATGG GATCTGATCT GGGGCCTCGG





4681
TGCACATGCT TTACATGTGT TTAGTCGAGG TTAAAAAAAC GTCTAGGCCC CCCGAACCAC





4741
GGGGACGTGG TTTTCCTTTG AAAAACACGA TGATAATATG GCCACAACCA TGGATAAAGT





4801
TTTAAACAGA GAGGAATCTT TGCAGCTAAT GGACCTTCTA GGTCTTGAAA GGAGTGCCTG





4861
GGGGAATATT CCTCTGATGA GAAAGGCATA TTTAAAAAAA TGCAAGGAGT TTCATCCTGA





4921
TAAAGGAGGA GATGAAGAAA AAATGAAGAA AATGAATACT CTGTACAAGA AAATGGAAGA





4981
TGGAGTAAAA TATGCTCATC AACCTGACTT TGGAGGCTTC TGGGATGCAA CTGAGATTCC





5041
AACCTATGGA ACTGATGAAT GGGAGCAGTG GTGGAATGCC TTTAATGAGG AAAACCTGTT





5101
TTGCTCAGAA GAAATGCCAT CTAGTGATGA TGAGGCTACT GCTGACTCTC AACATTCTAC





5161
TCCTCCAAAA AAGAAGAGAA AGGTAGAAGA CCCCAAGGAC TTTCCTTCAG AATTGCTAAG





5221
TTTTTTGAGT CATGCTGTGT TTAGTAATAG AACTCTTGCT TGCTTTGCTA TTTACACCAC





5281
AAAGGAAAAA GCTGCACTGC TATACAAGAA AATTATGGAA AAATATTCTG TAACCTTTAT





5341
AAGTAGGCAT AACAGTTATA ATCATAACAT ACTGTTTTTT CTTACTCCAC ACAGGCATAG





5401
AGTGTCTGCT ATTAATAACT ATGCTCAAAA ATTGTGTACC TTTAGCTTTT TAATTTGTAA





5461
AGGGGTTAAT AAGGAATATT TGATGTATAG TGCCTTGACT AGAGATCCAT TTTCTGTTAT





5521
TGAGGAAAGT TTGCCAGGTG GGTTAAAGGA GCATGATTTT AATCCAGAAG AAGCAGAGGA





5581
AACTAAACAA GTGTCCTGGA AGCTTGTAAC AGAGTATGCA ATGGAAACAA AATGTGATGA





5641
TGTGTTGTTA TTGCTTGGGA TGTACTTGGA ATTTCAGTAC AGTTTTGAAA TGTGTTTAAA





5701
ATGTATTAAA AAAGAACAGC CCAGCCACTA TAAGTACCAT GAAAAGCATT ATGCAAATGC





5761
TGCTATATTT GCTGACAGCA AAAACCAAAA AACCATATGC CAACAGGCTG TTGATACTGT





5821
TTTAGCTAAA AAGCGGGTTG ATAGCCTACA ATTAACTAGA GAACAAATGT TAACAAACAG





5881
ATTTAATGAT CTTTTGGATA GGATGGATAT AATGTTTGGT TCTACAGGCT CTGCTGACAT





5941
AGAAGAATGG ATGGCTGGAG TTGCTTGGCT ACACTGTTTG TTGCCCAAAA TGGATTCAGT





6001
GGTGTATGAC TTTTTAAAAT GCATGGTGTA CAACATTCCT AAAAAAAGAT ACTGGCTGTT





6061
TAAAGGACCA ATTGATAGTG GTAAAACTAC ATTAGCAGCT GCTTTGCTTG AATTATGTGG





6121
GGGGAAAGCT TTAAATGTTA ATTTGCCCTT GGACAGGCTG AACTTTGAGC TAGGAGTAGC





6181
TATTGACCAG TTTTTAGTAG TTTTTGAGGA TGTAAAGGGC ACTGGAGGGG AGTCCAGAGA





6241
TTTGCCTTCA GGTCAGGGAA TTAATAACCT GGACAATTTA AGGGATTATT TGGATGGCAG





6301
TGTTAAGGTA AACTTAGAAA AGAAACACCT AAATAAAAGA ACTCAAATAT TTCCCCCTGG





6361
AATAGTCACC ATGAATGAGT ACAGTGTGCC TAAAACACTG CAGGCCAGAT TTGTAAAACA





6421
AATAGATTTT AGGCCCAAAG ATTATTTAAA GCATTGCCTG GAACGCAGTG AGTTTTTGTT





6481
AGAAAAGAGA ATAATTCAAA GTGGCATTGC TTTGCTTCTT ATGTTAATTT GGTACAGACC





6541
TGTGGCTGAG TTTGCTCAAA GTATTCAGAG CAGAATTGTG GAGTGGAAAG AGAGATTGGA





6601
CAAAGAGTTT AGTTTGTCAG TGTATCAAAA AATGAAGTTT AATGTGGCTA TGGGAATTGG





6661
AGTTTTAGAT TGGCTAAGAA ACAGTGATGA TGATGATGAA GACAGCCAGG AAAATGCTGA





6721
TAAAAATGAA GATGGTGGGG AGAAGAACAT GGAAGACTCA GGGCATGAAA CAGGCATTGA





6781
TTCACAGTCC CAAGGCTCAT TTCAGGCCCC TCAGTCCTCA CAGTCTGTTC ATGATCATAA





6841
TCAGCCATAC CACATTTGTA GAGGTTTTAC TTGCTTTAAA AAACCTCCCA CACCTCCCCC





6901
TGAACCTGAA ACATAATAAG CTTGCGGCCG CTTCGAGCAG ACATGATAAG ATACATTGAT





6961
GAGTTTGGAC AAACCACAAC TAGAATGCAG TGAAAAAAAT GCTTTATTTG TGAAATTTGT





7021
GATGCTATTG CTTTATTTGT AACCATTATA AGCTGCAATA AACAAGTTTC ATGTCTGGCT





7081
CTAGCTATCC CGCCCCTAAC TCCGCCCATC CCGCCCCTAA CTCCGCCCAG TTCCGCCCAT





7141
TCTCCGCCCC ATGGCTGACT AATTTTTTTT ATTTATGCAG AGGCCGAGGC CGCCTCGGCC





7201
TCTGAGCTAT TCCAGAAGTA GTGAGGAGGC TTTTTTGGAG GCCATCGGAT CCCGGGCCCG





7261
TCGACTGCAG AGGCCTGCAT GCAAGCTTGG TGTAATCATG GTCATAGCTG TTTCCTGTGT





7321
GAAATTGTTA TCCGCTCACA ATTCCACACA ACATACGAGC CGGAAGCATA AAGTGTAAAG





7381
CCTGGGGTGC CTAATGAGTG AGCTAACTCA CATTAATTGC GTTGCGCTCA CTGCCCGCTT





7441
TCCAGTCGGG AAACCTGTCG TGCCAGCTGC ATTAATGAAT CGGCCAACGC GCGGGGAGAG





7501
GCGGTTTGCG TATTGGGCGC TCTTCCGCTT CCTCGCTCAC TGACTCGCTG CGCTCGGTCG





7561
TTCGGCTGCG GCGAGCGGTA TCAGCTCACT CAAAGGCGGT AATACGGTTA TCCACAGAAT





7621
CAGGGGATAA CGCAGGAAAG AACATGTGAG CAAAAGGCCA GCAAAAGGCC AGGAACCGTA





7681
AAAAGGCCGC GTTGCTGGCG TTTTTCCATA GGCTCCGCCC CCCTGACGAG CATCACAAAA





7741
ATCGACGCTC AAGTCAGAGG TGGCGAAACC CGACAGGACT ATAAAGATAC CAGGCGTTTC





7801
CCCCTGGAAG CTCCCTCGTG CGCTCTCCTG TTCCGACCCT GCCGCTTACC GGATACCTGT





7861
CCGCCTTTCT CCCTTCGGGA AGCGTGGCGC TTTCTCATAG CTCACGCTGT AGGTATCTCA





7921
GTTCGGTGTA GGTCGTTCGC TCCAAGCTGG GCTGTGTGCA CGAACCCCCC GTTCAGCCCG





7981
ACCGCTGCGC CTTATCCGGT AACTATCGTC TTGAGTCCAA CCCGGTAAGA CACGACTTAT





8041
CGCCACTGGC AGCAGCCACT GGTAACAGGA TTAGCAGAGC GAGGTATGTA GGCGGTGCTA





8101
CAGAGTTCTT GAAGTGGTGG CCTAACTACG GCTACACTAG AAGAACAGTA TTTGGTATCT





8161
GCGCTCTGCT GAAGCCAGTT ACCTTCGGAA AAAGAGTTGG TAGCTCTTGA TCCGGCAAAC





8221
AAACCACCGC TGGTAGCGGT GGTTTTTTTG TTTGCAAGCA GCAGATTACG CGCAGAAAAA





8281
AAGGATCTCA AGAAGATCCT TTGATCTTTT CTACGGGGTC TGACGCTCAG TGGAACGAAA





8341
ACTCACGTTA AGGGATTTTG GTCATGAGAT TATCAAAAAG GATCTTCACC TAGATCCTTT





8401
TAAATTAAAA ATGAAGTTTT AAATCAAGCC CAATCTGAAT AATGTTACAA CCAATTAACC





8461
AATTCTGATT AGAAAAACTC ATCGAGCATC AAATGAAACT GCAATTTATT CATATCAGGA





8521
TTATCAATAC CATATTTTTG AAAAAGCCGT TTCTGTAATG AAGGAGAAAA CTCACCGAGG





8581
CAGTTCCATA GGATGGCAAG ATCCTGGTAT CGGTCTGCGA TTCCGACTCG TCCAACATCA





8641
ATACAACCTA TTAATTTCCC CTCGTCAAAA ATAAGGTTAT CAAGTGAGAA ATCACCATGA





8701
GTGACGACTG AATCCGGTGA GAATGGCAAA AGTTTATGCA TTTCTTTCCA GACTTGTTCA





8761
ACAGGCCAGC CATTACGCTC GTCATCAAAA TCACTCGCAT CAACCAAACC GTTATTCATT





8821
CGTGATTGCG CCTGAGCGAG ACGAAATACG CGATCGCTGT TAAAAGGACA ATTACAAACA





8881
GGAATCGAAT GCAACCGGCG CAGGAACACT GCCAGCGCAT CAACAATATT TTCACCTGAA





8941
TCAGGATATT CTTCTAATAC CTGGAATGCT GTTTTTCCGG GGATCGCAGT GGTGAGTAAC





9001
CATGCATCAT CAGGAGTACG GATAAAATGC TTGATGGTCG GAAGAGGCAT AAATTCCGTC





9061
AGCCAGTTTA GTCTGACCAT CTCATCTGTA ACATCATTGG CAACGCTACC TTTGCCATGT





9121
TTCAGAAACA ACTCTGGCGC ATCGGGCTTC CCATACAAGC GATAGATTGT CGCACCTGAT





9181
TGCCCGACAT TATCGCGAGC CCATTTATAC CCATATAAAT CAGCATCCAT GTTGGAATTT





9241
AATCGCGGCC TCGACGTTTC CCGTTGAATA TGGCTCATAA CACCCCTTGT ATTACTGTTT





9301
ATGTAAGCAG ACAGTTTTAT TGTTCATGAT GATATATTTT TATCTTGTGC AATGTAACAT





9361
CAGAGATTTT GAGACACGGG CCAGAGCTGC A (SEQ ID NO: 500)










Annotations:








Region/Element
Base range





pHEf1A promoter
 436-1609





EF-1-alpha core promoter
 457-668





EF-1-alpha intron A
 669-1607





Initiator element
1614-1618





5′ UTR conserved domain
1670-1740





Intron 1
1682-1769





ORF2/3 coding sequence
1770-2056, 3756-4131





ORF2/2 coding sequence
1770-2056, 3629-3902





ORF2 coding sequence
1770-2060





ORF1 coding sequence
1952-3919





ORF1/1 coding sequence
1952-2056, 3629-3919





ORF1/2 coding sequence
1952-2056, 3756-3902





IRES-SV Large T Antigen-SV40 ori-pUC57-Kan
4131-9391, 1-435


concatenated sequence 1






EMCV internal ribosome entry site (IRES)
4203-4789





SV40 Large T Antigen coding sequence
4790-6916





SV40 polyA sequence (complement)
6947-7068





SV40 origin of replication
7108-7243





LacI repressor protein binding site (complement)
7325-7341





Lac operon promoter (complement)
7349-7379





CAP binding site (complement)
7394-7415





pUC origin of replication (complement)
7644-8317





ColE1/pMB1/pBR322/pUC origin of replication
7703-8291





Aminoglycoside phosphotransferase (Kan/G418
8469-9278


resistance protein) coding sequence (complement)









Exemplary SV40 Large T Antigen Amino Acid Sequence:









(SEQ ID NO: 504)



MDKVLNREESLQLMDLLGLERSAWGNIPLMRKAYLKKCKEFHPDKGGDEEKMKKMNTLYKK






MEDGVKYAHQPDFGGFWDATEIPTYGTDEWEQWWNAFNEENLFCSEEMPSSDDEATADSQHS





TPPKKKRKVEDPKDFPSELLSFLSHAVESNRTLACFAIYTTKEKAALLYKKIMEKYSVTFISRHNS





YNHNILFFLTPHRHRVSAINNYAQKLCTFSFLICKGVNKEYLMYSALTRDPFSVIEESLPGGLKEH





DFNPEEAEETKQVSWKLVTEYAMETKCDDVLLLLGMYLEFQYSFEMCLKCIKKEQPSHYKYHE





KHYANAAIFADSKNQKTICQQAVDTVLAKKRVDSLQLTREQMLTNRFNDLLDRMDIMFGSTGS





ADIEEWMAGVAWLHCLLPKMDSVVYDFLKCMVYNIPKKRYWLFKGPIDSGKTTLAAALLELC





GGKALNVNLPLDRLNFELGVAIDQFLVVFEDVKGTGGESRDLPSGQGINNLDNLRDYLDGSVKV





NLEKKHLNKRTQIFPPGIVTMNEYSVPKTLQARFVKQIDFRPKDYLKHCLERSEFLLEKRIIQSGIA





LLLMLIWYRPVAEFAQSIQSRIVEWKERLDKEFSLSVYQKMKFNVAMGIGVLDWLRNSDDDDE





DSQENADKNEDGGEKNMEDSGHETGIDSQSQGSFQAPQSSQSVHDHNQPYHICRGFTCFKKPPT





PPPEPET






Exemplary Site-Specific Recombinases and Recombinase Recognition Sites

The recombinase-based systems described herein utilize site-specific recombinases to induce recombination of vector plasmids at recombinase recognition sites, thereby producing double-stranded DNA molecules (e.g., minicircles) comprising the sequence between the recombinase recognition sites (e.g., comprising the sequence of a genetic element for an Anelloviridae family vector as described herein).


In some embodiments, the site-specific recombinase comprises a Cre recombinase (e.g., as described herein, e.g., in Table V2), or an amino acid sequence having at least 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% identity thereto. In some embodiments, the site-specific recombinase comprises the amino acid sequence of Cre as listed in Table V2, or an amino acid sequence having at least 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% identity thereto. In some embodiments, the site-specific recombinase comprises the amino acid sequence of SV40-NLS-iCre as listed in Table V2, or an amino acid sequence having at least 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% identity thereto.


In certain embodiments, at least one (e.g., one or both) of the recombinase recognition sites comprises a lox66 site as listed in Table V3, or a nucleic acid sequence having at least 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% identity thereto. In certain embodiments, at least one (e.g., one or both) of the recombinase recognition sites comprises a lox71 site as listed in Table V3, or a nucleic acid sequence having at least 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% identity thereto. In certain embodiments, a circular double-stranded nucleic acid molecule (e.g., minicircle) produced after a Cre recombination event (e.g., as described herein) comprises a loxP site as listed in Table V3, or a nucleic acid sequence having at least 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% identity thereto. In certain embodiments, a circular double-stranded nucleic acid molecule (e.g., minicircle) produced after a Cre recombination event (e.g., as described herein) comprises a lox72 site as listed in Table V3, or a nucleic acid sequence having at least 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% identity thereto.


In some embodiments, the site-specific recombinase comprises a Bxb1 recombinase (e.g., as described herein, e.g., in Table V2), or an amino acid sequence having at least 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% identity thereto. In some embodiments, the site-specific recombinase comprises the amino acid sequence of Bxb1 as listed in Table V2, or an amino acid sequence having at least 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% identity thereto. In some embodiments, the site-specific recombinase comprises the amino acid sequence of SV40-NLS-HA_Bxb1 as listed in Table V2, or an amino acid sequence having at least 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% identity thereto.


In certain embodiments, at least one of the recombinase recognition sites comprises an attB site as listed in Table V4, or a nucleic acid sequence having at least 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% identity thereto. In certain embodiments, at least one of the recombinase recognition sites comprises an attP site as listed in Table V4, or a nucleic acid sequence having at least 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% identity thereto. In certain embodiments, one recombinase recognition site comprises an attB site as listed in Table V4, or a nucleic acid sequence having at least 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% identity thereto, and the other recombinase recognition site comprises an attP site as listed in Table V4, or a nucleic acid sequence having at least 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% identity thereto. In certain embodiments, a circular double-stranded nucleic acid molecule (e.g., minicircle) produced after a Bxb1 recombination event (e.g., as described herein) comprises an attL site as listed in Table V4, or a nucleic acid sequence having at least 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% identity thereto. In certain embodiments, a circular double-stranded nucleic acid molecule (e.g., minicircle) produced after a Bxb1 recombination event (e.g., as described herein) comprises an attR site as listed in Table V4, or a nucleic acid sequence having at least 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% identity thereto.









TABLE V2







Exemplary site-specific recombinase polypeptides








Site-specific
Amino Acid Sequence


recombinase
(Underline = SV40 NLS sequence; bolded italics = HA tag sequence)





SV40-
MVPKKKRKVSNLLTVHQNLPALPVDATSDEVRKNLMDMFRDRQAFSEHTWKMLLSVCRS


NLS_iCre
WAAWCKLNNRKWFPAEPEDVRDYLLYLQARGLAVKTIQQHLGQLNMLHRRSGLPRPSDS



NAVSLVMRRIRKENVDAGERAKQALAFERTDFDQVRSLMENSDRCQDIRNLAFLGIAYN



TLLRIAEIARIRVKDISRTDGGRMLIHIGRTKTLVSTAGVEKALSLGVTKLVERWISVS



GVADDPNNYLFCRVRKNGVAAPSATSQLSTRALEGIFEATHRLIYGAKDDSGQRYLAWS



GHSARVGAARDMARAGVSIPEIMQAGGWINVNIVMNYIRNLDSETGAMVRLLEDGD*



(SEQ ID NO: 1101)





Cre (same as
MSNLLTVHQNLPALPVDATSDEVRKNLMDMFRDRQAFSEHTWKMLLSVCRSWAAWCKLN


iCre)
NRKWFPAEPEDVRDYLLYLQARGLAVKTIQQHLGQLNMLHRRSGLPRPSDSNAVSLVMR



RIRKENVDAGERAKQALAFERTDFDQVRSLMENSDRCQDIRNLAFLGIAYNTLLRIAEI



ARIRVKDISRTDGGRMLIHIGRTKTLVSTAGVEKALSLGVTKLVERWISVSGVADDPNN



YLFCRVRKNGVAAPSATSQLSTRALEGIFEATHRLIYGAKDDSGQRYLAWSGHSARVGA



ARDMARAGVSIPEIMQAGGWTNVNIVMNYIRNLDSETGAMVRLLEDGD (SEQ ID



NO: 1102)





SV40-NLS-
MPKKKRKVYPYDVPDYAGSRALVVIRLSRVTDATTSPERQLESCQQLCAQRGWDVVGVA


HA_Bxb1
EDLDVSGAVDPFDRKRRPNLARWLAFEEQPFDVIVAYRVDRLTRSIRHLQQLVHWAEDH



KKLVVSATEAHFDTTTPFAAVVIALMGTVAQMELEAIKERNRSAAHFNIRAGKYRGSLP



PWGYLPTRVDGEWRLVPDPVQRERILEVYHRVVDNHEPLHLVAHDLNRRGVLSPKDYFA



QLQGREPQGREWSATALKRSMISEAMLGYATLNGKTVRDDDGAPLVRAEPILTREQLEA



LRAELVKTSRAKPAVSTPSLLLRVLFCAVCGEPAYKFAGGGRKHPRYRCRSMGFPKHCG



NGTVAMAEWDAFCEEQVLDLLGDAERLEKVWVAGSDSAVELAEVNAELVDLTSLIGSPA



YRAGSPQREALDARIAALAARQEELEGLEARPSGWEWRETGQRFGDWWREQDTAAKNTW



LRSMNVRLTFDVRGGLTRTIDFGDLQEYEQHLRLGSVVERLHTGMS* (SEQ ID NO:



1103)





Bxb1
MRALVVIRLSRVTDATTSPERQLESCQQLCAQRGWDVVGVAEDLDVSGAVDPFDRKRRP



NLARWLAFEEQPFDVIVAYRVDRLTRSIRHLQQLVHWAEDHKKLVVSATEAHFDTTTPF



AAVVIALMGTVAQMELEAIKERNRSAAHFNIRAGKYRGSLPPWGYLPTRVDGEWRLVPD



PVQRERILEVYHRVVDNHEPLHLVAHDLNRRGVLSPKDYFAQLQGREPQGREWSATALK



RSMISEAMLGYATLNGKTVRDDDGAPLVRAEPILTREQLEALRAELVKTSRAKPAVSTP



SLLLRVLFCAVCGEPAYKFAGGGRKHPRYRCRSMGFPKHCGNGTVAMAEWDAFCEEQVL



DLLGDAERLEKVWVAGSDSAVELAEVNAELVDLTSLIGSPAYRAGSPQREALDARIAAL



AARQEELEGLEARPSGWEWRETGQRFGDWWREQDTAAKNTWLRSMNVRLTFDVRGGLTR



TIDFGDLQEYEQHLRLGSVVERLHTGMS (SEQ ID NO: 1104)
















TABLE V3







Exemplary recombinase recognition sequences and recombinase hybrid sites


for Cre recombinases








Recombinase



Recognition Site Name
Nucleic acid sequence





loxP
ATAACTTCGTATAGCATACATTATACGAAGTTAT (SEQ



ID NO: 1105)





lox66
Ataacttcgtatagcatacattatacgaacggta (SEQ



ID NO: 1106)





lox71
Taccgttcgtatagcatacattatacgaagttat (SEQ



ID NO: 1107)





lox72 (hybrid)
TACCGTTCGTATAGCATACATTATACGAACGGTA (SEQ



ID NO: 1108)
















TABLE V4







Exemplary recombinase recognition sequences for Bxb1 recombinases








Recombinase



Recognition



Site Name
Nucleic acid sequence





attB
TCGGCCGGCTTGTCGACGACGGCGGTCTCCGTCGTCAGGATCATCCGGGC (SEQ ID



NO: 1109)





attP
GTCGTGGTTTGTCTGGTCAACCACCGCGGTCTCAGTGGTGTACGGTACAAACCCCGAC



(SEQ ID NO: 1110)





attL
TCGGCCGGCTTGTCGACGACGGCGGTCTCAGTGGTGTACGGTACAAACCCCGAC (SEQ



ID NO: 1111)





attR
GTCGTGGTTTGTCTGGTCAACCACCGCGGTCTCCGTCGTCAGGATCATCCGGGC (SEQ



ID NO: 1112)









As used herein, the term “recombinase hybrid site” refers to a DNA site having a sequence that, when in double stranded form, is capable of being produced by a site-specific recombinase that recombines two recombinase recognition sites. No particular process of making is implied: a recombinase hybrid site can be produced by a site specific recombinase or another method, such as DNA replication of an existing sequence. In some embodiments, a single stranded DNA that comprises a recombinase hybrid site was produced by a method wherein a site-specific recombinase generated a double stranded DNA comprising a recombinase hybrid site, followed by conversion of the double stranded DNA to a single stranded DNA. In some embodiments (e.g., with Cre recombinase) the recombinase hybrid site has the same sequence as one of the corresponding recombinase recognition sites. In some embodiments (e.g., with Bxb1 recombinase), the recombinase hybrid site has a different site from either of the two corresponding recombinase recognition sites. In some embodiments, the recombinase hybrid site is a loxP site, an attL site, or an attR site.


As used herein, the term “recombinase recognition site” refers to a DNA site having a sequence that is capable of being recognized by a site-specific recombinase and recombined with a second recombinase recognition site, thereby producing a recombinase hybrid site. In some embodiments, the two recombinase recognition sites recognized by the recombinase have the same sequence, and in other embodiments, they have different sequences. In some embodiments, the recombinase recognition site is a loxP site, an attB site, or an attP site.


Host Cells and Methods of Using Host Cells for Producing an Anellovector

The Anelloviridae family vector (e.g., anellovector) described herein can be produced, for example, in a host cell. Generally, a host cell is provided that comprises an Anelloviridae family vector (e.g., anellovector) genetic element and the components of an Anelloviridae family vector (e.g., anellovector) proteinaceous exterior (e.g., a polypeptide encoded by an Anellovirus ORF1 nucleic acid, or an Anellovirus ORF1 molecule). For example, in some embodiments, the host cell comprises a nucleic acid sequence encoding an Anellovirus ORF1 molecule, e.g., a splice variant or a functional fragment of an Anellovirus ORF1 polypeptide (e.g., a wild-type Anellovirus ORF1 protein or a polypeptide encoded by a wild-type Anellovirus ORF1 nucleic acid, e.g., as described herein). In embodiments, the nucleic acid sequence encoding the Anellovirus ORF1 molecule is comprised in a nucleic acid construct (e.g., a plasmid, viral vector, virus, minicircle, bacmid, or artificial chromosome) comprised in the host cell. In embodiments, the nucleic acid sequence encoding the Anellovirus ORF1 molecule is integrated into the genome of the host cell.


Producing an Anelloviridae family vector (e.g. anellovector) using the compositions or methods described herein may also involve expression of an Anellovirus ORF2 molecule (e.g., as described herein), or a splice variant or functional fragment thereof. In some embodiments, the anellovector does not comprise an ORF2 molecule, or a splice variant or functional fragment thereof, and/or a nucleic acid encoding an ORF2 molecule, or a splice variant or functional fragment thereof. In some embodiments, producing the anellovector comprises expression of an ORF2 molecule, or a splice variant or functional fragment thereof, but the ORF2 molecule is expressed from a nucleic acid other than Anelloviridae family vector.


The host cell is then incubated under conditions suitable for enclosure of the genetic element within the proteinaceous exterior (e.g., culture conditions as described herein). In some embodiments, the host cell is further incubated under conditions suitable for release of the Anelloviridae family vector (e.g., anellovector) from the host cell, e.g., into the surrounding supernatant. In some embodiments, the host cell is lysed for harvest of Anelloviridae family vector (e.g., anellovector) from the cell lysate. In some embodiments, an Anelloviridae family vector (e.g., anellovector) may be introduced to a host cell line grown to a high cell density. In some embodiments, a host cell is an Expi-293 cell.


In an aspect, the present disclosure provides a host cell (e.g., as described herein). In some embodiments, the host or host cell is a plant, insect, bacteria, fungus, vertebrate, mammal (e.g., human), or other organism or cell.


Introduction of Genetic Elements into Host Cells


The genetic element, or a nucleic acid construct comprising the sequence of a genetic element, may be introduced into a host cell. In some embodiments, the genetic element itself is introduced into the host cell. In some embodiments, a genetic element construct comprising the sequence of the genetic element (e.g., as described herein) is introduced into the host cell. A genetic element or genetic element construct can be introduced into a host cell, for example, using methods known in the art. For example, a genetic element or genetic element construct can be introduced into a host cell by transfection (e.g., stable transfection or transient transfection). In embodiments, the genetic element or genetic element construct is introduced into the host cell by lipofectamine transfection. In embodiments, the genetic element or genetic element construct is introduced into the host cell by calcium phosphate transfection. In some embodiments, the genetic element or genetic element construct is introduced into the host cell by electroporation. In some embodiments, the genetic element or genetic element construct is introduced into the host cell using a gene gun. In some embodiments, the genetic element or genetic element construct is introduced into the host cell by nucleofection. In some embodiments, the genetic element or genetic element construct is introduced into the host cell by PEI transfection. In some embodiments, the genetic element is introduced into the host cell by contacting the host cell with an Anelloviridae family vector (e.g., anellovector) comprising the genetic element. In some embodiments, cells are suspended in 2S Chica buffers.


In embodiments, the genetic element construct is capable of replication once introduced into the host cell. In embodiments, the genetic element can be produced from the genetic element construct once introduced into the host cell. In some embodiments, the genetic element is produced in the host cell by a polymerase, e.g., using the genetic element construct as a template.


In some embodiments, the genetic elements or vectors comprising the genetic elements are introduced (e.g., transfected) into cell lines that express a viral polymerase protein in order to achieve expression of the Anelloviridae family vector (e.g., anellovector). To this end, cell lines that express an Anelloviridae family vector (e.g., anellovector) polymerase protein may be utilized as appropriate host cells. Host cells may be similarly engineered to provide other viral functions or additional functions.


To prepare the Anelloviridae family vector (e.g., anellovector) disclosed herein, a genetic element construct may be used to transfect cells that provide Anelloviridae family vector (e.g., anellovector) proteins and functions required for replication and production. Alternatively, cells may be transfected with a second construct (e.g., a virus) providing Anelloviridae family vector (e.g., anellovector) proteins and functions before, during, or after transfection by the genetic element or vector comprising the genetic element disclosed herein. In some embodiments, the second construct may be useful to complement production of an incomplete viral particle. The second construct (e.g., virus) may have a conditional growth defect, such as host range restriction or temperature sensitivity, e.g., which allows the subsequent selection of transfectant viruses. In some embodiments, the second construct may provide one or more replication proteins utilized by the host cells to achieve expression of the Anelloviridae family vector (e.g., anellovector). In some embodiments, the host cells may be transfected with vectors encoding viral proteins such as the one or more replication proteins. In some embodiments, the second construct comprises an antiviral sensitivity.


The genetic element or vector comprising the genetic element disclosed herein can, in some instances, be replicated and produced into Anelloviridae family vectors (e.g., anellovectors) using techniques known in the art. For example, various viral culture methods are described, e.g., in U.S. Pat. Nos. 4,650,764; 5,166,057; 5,854,037; European Patent Publication EP 0702085A1; U.S. patent application Ser. No. 09/152,845; International Patent Publications PCT WO97/12032; WO96/34625; European Patent Publication EP-A780475; WO 99/02657; WO 98/53078; WO 98/02530; WO 99/15672; WO 98/13501; WO 97/06270; and EPO 780 47SA1, each of which is incorporated by reference herein in its entirety.


Exemplary Cell Types

Exemplary host cells suitable for production of Anelloviridae family vector (e.g., anellovector) include, without limitation, mammalian cells, e.g., human cells and insect cells. In some embodiments, the host cell is a human cell or cell line. In some embodiments, the cell is an immune cell or cell line, e.g., a T cell or cell line, a cancer cell line, a hepatic cell or cell line, a neuron, a glial cell, a skin cell, an epithelial cell, a mesenchymal cell, a blood cell, an endothelial cell, an eye cell (e.g., a photoreceptor cell, a retinal cell, a cell of the posterior eye cup (PEC), retinal ganglion cell, a cell of the optic nerve, a cell of the optic nerve head, or a retinal pigmented epithelium (RPE) cell), a gastrointestinal cell, a progenitor cell, a precursor cell, a stem cell, a lung cell, a cardiac cell, or a muscle cell. In some embodiments, the host cell is an animal cell (e.g., a mouse cell, rat cell, rabbit cell, or hamster cell, or insect cell).


In some embodiments, the host cell is a human cell. In embodiments, the host cell is a HEK293T cell, HEK293F cell, A549 cell, Jurkat cell, Raji cell, Chang cell, HeLa cell Phoenix cell, MRC-5 cell, NCI-H292 cell, or Wi38 cell. In some embodiments, the host cell is a non-human primate cell (e.g., a Vero cell, CV-1 cell, or LLCMK2 cell). In some embodiments, the host cell is a murine cell (e.g., a McCoy cell). In some embodiments, the host cell is a hamster cell (e.g., a CHO cell or BHK 21 cell). In some embodiments, the host cell is a MARC-145, MDBK, RK-13, or EEL cell. In some embodiments, the host cell is an epithelial cell (e.g., a cell line of epithelial lineage).


In some embodiments, the host cell is a lymphoid cell. In some embodiments, the host cell is a T cell or an immortalized T cell. In embodiments, the host cell is a Jurkat cell. In embodiments, the host cell is a MOLT cell (e.g., a MOLT-4 or a MOLT-3 cell). In embodiments, the host cell is a MOLT-4 cell. In embodiments, the host cell is a MOLT-3 cell. In some embodiments, the host cell is an acute lymphoblastic leukemia (ALL) cell, e.g., a MOLT cell, e.g., a MOLT-4 or MOLT-3 cell. In some embodiments, the host cell is a B cell or an immortalized B cell. In some embodiments, the host cell comprises a genetic element construct (e.g., as described herein).


In some embodiments, the host cell is a MOLT cell (e.g., a MOLT-4 or a MOLT-3 cell).


In some embodiments, the host cell is an acute lymphoblastic leukemia (ALL) cell, e.g., a MOLT cell, e.g., a MOLT-4 or MOLT-3 cell.


In some embodiments, the host cell is a 293 cell (e.g., a HEK293 cell, a HEK293T cell, or an Expi-293 cell). In some embodiments, the host cell is an Expi-293F cell.


In an aspect, the present disclosure provides a method of manufacturing an Anelloviridae family vector (e.g., anellovector) comprising a genetic element enclosed in a proteinaceous exterior, the method comprising providing a Expi-293 cell comprising an Anelloviridae family vector (e.g., anellovector) genetic element, and incubating the Expi-293 cell under conditions that allow the Anelloviridae family vector (e.g., anellovector) genetic element to become enclosed in a proteinaceous exterior in the Expi-293 cell. In some embodiments, the Expi-293 cell further comprises one or more Anellovirus proteins (e.g., an Anellovirus ORF1 molecule) that form part or all of the proteinaceous exterior. In some embodiments, the Anelloviridae family vector (e.g., anellovector) genetic element is produced in the Expi-293 cell, e.g., from a genetic element construct (e.g., as described herein). In some embodiments, the method further comprises introducing the Anelloviridae family vector (e.g., anellovector) genetic element construct into the Expi-293 cell.


In an aspect, the present disclosure provides a method of manufacturing an Anelloviridae family vector (e.g., anellovector) comprising a genetic element enclosed in a proteinaceous exterior, the method comprising providing a MOLT-4 cell comprising an Anelloviridae family vector (e.g., anellovector) genetic element, and incubating the MOLT-4 cell under conditions that allow the Anelloviridae family vector (e.g., anellovector) genetic element to become enclosed in a proteinaceous exterior in the MOLT-4 cell. In some embodiments, the MOLT-4 cell further comprises one or more Anellovirus proteins (e.g., an Anellovirus ORF1 molecule) that form part or all of the proteinaceous exterior. In some embodiments, the Anelloviridae family vector (e.g., anellovector) genetic element is produced in the MOLT-4 cell, e.g., from a genetic element construct (e.g., as described herein). In some embodiments, the method further comprises introducing the Anelloviridae family vector (e.g., anellovector) genetic element construct into the MOLT-4 cell.


In an aspect, the present disclosure provides a method of manufacturing an Anelloviridae family vector (e.g., anellovector) comprising a genetic element enclosed in a proteinaceous exterior, the method comprising providing a MOLT-3 cell comprising an Anelloviridae family vector (e.g., anellovector) genetic element, and incubating the MOLT-3 cell under conditions that allow the Anelloviridae family vector (e.g., anellovector) genetic element to become enclosed in a proteinaceous exterior in the MOLT-3 cell. In some embodiments, the MOLT-3 cell further comprises one or more Anellovirus proteins (e.g., an Anellovirus ORF1 molecule) that form part or all of the proteinaceous exterior. In some embodiments, the Anelloviridae family vector (e.g., anellovector) genetic element is produced in the MOLT-3 cell, e.g., from a genetic element construct (e.g., as described herein). In some embodiments, the method further comprises introducing the Anelloviridae family vector (e.g., anellovector) genetic element construct into the MOLT-3 cell.


In some embodiments, the Anelloviridae family vector (e.g., anellovector) is cultivated in continuous animal cell line (e.g., immortalized cell lines that can be serially propagated).


Culture Conditions

Host cells comprising a genetic element and components of a proteinaceous exterior can be incubated under conditions suitable for enclosure of the genetic element within the proteinaceous exterior, thereby producing an Anelloviridae family vector (e.g., anellovector). In some embodiments, the host cells are incubated in liquid media (e.g., Grace's Supplemented (TNM-FH), IPL-41, TC-100, Schneider's Drosophila, SF-900 II SFM, or and EXPRESS-FIVE™ SFM). In some embodiments, the host cells are incubated in adherent culture. In some embodiments, the host cells are incubated in suspension culture. In some embodiments, the host cells are incubated in a tube, bottle, microcarrier, or flask. In some embodiments, the host cells are incubated in a dish or well (e.g., a well on a plate). In some embodiments, the host cells are incubated under conditions suitable for proliferation of the host cells. In some embodiments, the host cells are incubated under conditions suitable for the host cells to release Anelloviridae family vectors (e.g., anellovectors) produced therein into the surrounding supernatant.


The production of Anelloviridae family vector (e.g., anellovector)-containing cell cultures according to the present invention can be carried out in different scales (e.g., in flasks, roller bottles or bioreactors). The media used for the cultivation of the cells to be infected generally comprise the standard nutrients required for cell viability, but may also comprise additional nutrients dependent on the cell type. Optionally, the medium can be protein-free and/or serum-free. Depending on the cell type the cells can be cultured in suspension or on a substrate. In some embodiments, different media is used for growth of the host cells and for production of Anelloviridae family vectors (e.g., anellovectors).


Harvest

Anelloviridae family vectors (e.g., anellovectors) produced by host cells can be harvested, e.g., according to methods known in the art. For example, Anelloviridae family vectors (e.g., anellovectors) released into the surrounding supernatant by host cells in culture can be harvested from the supernatant. In some embodiments, the supernatant is separated from the host cells to obtain the Anelloviridae family vectors (e.g., anellovectors). In some embodiments, the host cells are lysed before or during harvest. In some embodiments, the Anelloviridae family vectors (e.g., anellovectors) are harvested from the host cell lysates. In some embodiments, the Anelloviridae family vectors (e.g., anellovectors) are harvested from both the host cell lysates and the supernatant. In some embodiments, the purification and isolation of Anelloviridae family vectors (e.g., anellovectors) is performed according to known methods in virus production, for example, as described in Rinaldi, et al., DNA Vaccines: Methods and Protocols (Methods in Molecular Biology), 3rd ed. 2014, Humana Press (incorporated herein by reference in its entirety). In some embodiments, the Anelloviridae family vector (e.g., anellovector) may be harvested and/or purified by separation of solutes based on biophysical properties, e.g., ion exchange chromatography or tangential flow filtration, prior to formulation with a pharmaceutical excipient.


In Vitro Assembly Methods

An Anelloviridae family vector (e.g., anellovector) may be produced, e.g., by in vitro assembly, e.g., in a cell-free suspension or in a supernatant. In some embodiments, the genetic element is contacted to an ORF1 molecule in vitro, e.g., under conditions that allow for assembly.


In some embodiments, baculovirus constructs are used to produce Anelloviridae family virus (e.g., Anellovirus) proteins. These proteins may then be used, e.g., for in vitro assembly to encapsidate a genetic element, e.g., a genetic element comprising RNA. In some embodiments, a polynucleotide encoding one or more Anelloviridae family virus (e.g., Anellovirus) protein is fused to a promoter for expression in a host cell, e.g., an insect or animal cell. In some embodiments, the polynucleotide is cloned into a baculovirus expression system. In some embodiments, a host cell, e.g., an insect cell is infected with the baculovirus expression system and incubated for a period of time. In some embodiments, an infected cell is incubated for about 1, 2, 3, 4, 5, 10, 15, or 20 days. In some embodiments, an infected cell is lysed to recover the Anelloviridae family virus (e.g., Anellovirus) protein.


In some embodiments, an isolated Anelloviridae family virus (e.g., Anellovirus) protein is purified. In some embodiments, an Anellovirus protein is purified using purification techniques including but not limited to chelating purification, heparin purification, gradient sedimentation purification, and/or SEC purification. In some embodiments, a purified Anelloviridae family virus (e.g., Anellovirus) protein is mixed with a genetic element to encapsidate the genetic element, e.g., a genetic element comprising RNA. In some embodiments, a genetic element is encapsidated using an ORF1 protein, ORF2 protein, or modified version thereof. In some embodiments two nucleic acids are encapsidated. For instance, the first nucleic acid may be an mRNA e.g., chemically modified mRNA, and the second nucleic acid may be DNA.


In some embodiments, DNA encoding Anellovirus (AV) ORF1 (e.g., wildtype ORF1 protein, ORF1 proteins harboring mutations, e.g., to improve assembly efficiency, yield or stability, chimeric ORF1 protein, or fragments thereof) are expressed in insect cell lines (e.g., Sf9 and/or HighFive), animal cell lines (e.g., chicken cell lines (MDCC)), bacterial cells (e.g., E. coli) and/or mammalian cell lines (e.g., 293expi and/or MOLT4). In some embodiments, DNA encoding AV ORF1 may be untagged. In some embodiments, DNA encoding AV ORF1 may contain tags fused N-terminally and/or C-terminally. In some embodiments, DNA encoding AV ORF1 may harbor mutations, insertions or deletions within the ORF1 protein to introduce a tag, e.g., to aid in purification and/or identity determination, e.g., through immunostaining assays (including but not limited to ELISA or Western Blot). In some embodiments, DNA encoding AV ORF1 may be expressed alone or in combination with any number of helper proteins. In some embodiments, DNA encoding AV ORF1 is expressed in combination with AV ORF2 and/or ORF3 proteins.


In some embodiments, ORF1 proteins harboring mutations to improve assembly efficiency may include, but are not limited to, ORF1 proteins that harbor mutations introduced into the N-terminal Arginine Arm (ARG arm) to alter the pI of the ARG arm permitting pH sensitive nucleic acid binding to trigger particle assembly. In some embodiments, ORF1 proteins harboring mutations that improve stability may include mutations to an interprotomer contacting beta strands F and G of the canonical jellyroll beta-barrel to alter hydrophobic state of the protomer surface and improve thermodynamic favorability of capsid formation.


In some embodiments, the present disclosure describes a method of making an anellovector, the method comprising: (a) providing a mixture comprising: (i) a genetic element, and (ii) an ORF1 molecule and (b) incubating the mixture under conditions suitable for enclosing the genetic element within a proteinaceous exterior comprising the ORF1 molecule, thereby making an anellovector; optionally wherein the mixture is not comprised in a cell. In some embodiments, the method further comprises, prior to the providing of (a), expressing the ORF1 molecule, e.g., in a host cell (e.g., an insect cell or a mammalian cell). In some embodiments, the expressing comprises incubating a host cell (e.g., an insect cell or a mammalian cell) comprising a nucleic acid molecule (e.g., a baculovirus expression vector) encoding the ORF1 molecule under conditions suitable for producing the ORF1 molecule. In some embodiments, the method further comprises, prior to the providing of (a), purifying the ORF1 molecule expressed by the host cell. In some embodiments, the method is performed in a cell-free system. In some embodiments, the present disclosure describes a method of manufacturing an anellovector composition, comprising: (a) providing a plurality of anellovectors or compositions according to any of the preceding embodiments; (b) optionally evaluating the plurality for one or more of: a contaminant described herein, an optical density measurement (e.g., OD 260), particle number (e.g., by HPLC), infectivity (e.g., particle:infectious unit ratio, e.g., as determined by fluorescence and/or ELISA); and (c) formulating the plurality of anellovectors, e.g., as a pharmaceutical composition suitable for administration to a subject, e.g., if one or more of the parameters of (b) meet a specified threshold.


Enrichment and Purification

Harvested Anelloviridae family vectors can be purified and/or enriched, e.g., to produce an anellovector preparation. In some embodiments, the harvested anellovectors are isolated from other constituents or contaminants present in the harvest solution, e.g., using methods known in the art for purifying viral particles (e.g., purification by sedimentation, chromatography, and/or ultrafiltration). In some embodiments, the purification steps comprise removing one or more of serum, host cell DNA, host cell proteins, particles lacking the genetic element, and/or phenol red from the preparation. In some embodiments, the harvested Anelloviridae family vectors are enriched relative to other constituents or contaminants present in the harvest solution, e.g., using methods known in the art for enriching viral particles.


In some embodiments, the resultant preparation or a pharmaceutical composition comprising the preparation will be stable over an acceptable period of time and temperature, and/or be compatible with the desired route of administration and/or any devices this route of administration will require, e.g., needles or syringes.


III. Pharmaceutical Compositions

The Anelloviridae family vector, anellovector, or other vector described herein may also be included in pharmaceutical compositions with a pharmaceutical excipient, e.g., as described herein. In some embodiments, the pharmaceutical composition comprises at least 105, 106, 107, 108, 109, 1010, 1011, 1012, 1013, 1014, or 1015 Anelloviridae family vectors. In some embodiments, the pharmaceutical composition comprises about 105-1015, 105-1010, or 1010-1015 Anelloviridae family vectors. In some embodiments, the pharmaceutical composition comprises about 108 (e.g., about 105, 106, 107, 108, 109, or 1010) genomic equivalents/mL of the Anelloviridae family vector. In some embodiments, the pharmaceutical composition comprises 105-1010, 106-1010, 107-1010, 108-1010, 109-1010, 105-106, 105-107, 105-108, 105-109, 105-1011, 105-1012, 105-1011, 105-1014, 105-1015, or 1010-1015 genomic equivalents/mL of the Anelloviridae family vector. In some embodiments, the pharmaceutical composition comprises sufficient Anelloviridae family vectors to deliver at least 1, 2, 5, or 10, 100, 500, 1000, 2000, 5000, 8,000, 1×104, 1×105, 1×106, 1×107 or greater copies of a genetic element comprised in the Anelloviridae family vectors per cell to a population of the eukaryotic cells. In some embodiments, the pharmaceutical composition comprises sufficient Anelloviridae family vectors to deliver at least about 1×104, 1×105, 1×106, 1× or 107, or about 1×104-1×105, 1×104-1×106, 1×104-1×107, 1×105-1×106, 1×105-1×107, or 1×106-1×107 copies of a genetic element comprised in the Anelloviridae family vectors per cell to a population of the eukaryotic cells.


In some embodiments, the pharmaceutical composition has one or more of the following characteristics: the pharmaceutical composition meets a pharmaceutical or good manufacturing practices (GMP) standard; the pharmaceutical composition was made according to good manufacturing practices (GMP); the pharmaceutical composition has a pathogen level below a predetermined reference value, e.g., is substantially free of pathogens; the pharmaceutical composition has a contaminant level below a predetermined reference value, e.g., is substantially free of contaminants; or the pharmaceutical composition has low immunogenicity or is substantially non-immunogenic, e.g., as described herein.


In some embodiments, the pharmaceutical composition comprises below a threshold amount of one or more contaminants. Exemplary contaminants that are desirably excluded or minimized in the pharmaceutical composition include, without limitation, host cell nucleic acids (e.g., host cell DNA and/or host cell RNA), animal-derived components (e.g., serum albumin or trypsin), replication-competent viruses, non-infectious particles, free viral capsid protein, adventitious agents, and aggregates. In embodiments, the contaminant is host cell DNA. In embodiments, the composition comprises less than about 10 ng of host cell DNA per dose. In embodiments, the level of host cell DNA in the composition is reduced by filtration and/or enzymatic degradation of host cell DNA. In embodiments, the pharmaceutical composition consists of less than 10% (e.g., less than about 10%, 5%, 4%, 3%, 2%, 1%, 0.5%, or 0.1%) contaminant by weight.


In one aspect, the invention described herein includes a pharmaceutical composition comprising:

    • a) an Anelloviridae family vector (e.g., anellovector) comprising a genetic element comprising (i) a sequence encoding a non-pathogenic exterior protein, (ii) an exterior protein binding sequence that binds the genetic element to the non-pathogenic exterior protein, and (iii) a sequence encoding a regulatory nucleic acid; and a proteinaceous exterior that is associated with, e.g., envelops or encloses, the genetic element; and
    • b) a pharmaceutical excipient.


IV. Methods of Use

The Anelloviridae family vectors, e.g., anellovectors, and compositions comprising Anelloviridae family vectors, e.g., anellovectors, described herein may be used in methods of treating a disease, disorder, or condition, e.g., in a subject (e.g., a mammalian subject, e.g., a human subject) in need thereof. Administration of a pharmaceutical composition described herein may be, for example, by way of parenteral administration. In some embodiments, an Anelloviridae family vector, e.g., anellovector, or pharmaceutical composition as described herein is administered subretinally. In some embodiments, an Anelloviridae family vector, e.g., anellovector, or pharmaceutical composition as described herein is administered intravitreally. In some embodiments, an Anelloviridae family vector, e.g., anellovector, or pharmaceutical composition as described herein is administered suprachoroidally. The anellovectors may be administered alone or formulated as a pharmaceutical composition.


The Anelloviridae family vector (e.g., anellovector) may be administered in the form of a unit-dose composition, such as a unit dose parenteral composition. Such compositions are generally prepared by admixture and can be suitably adapted for parenteral administration. Such compositions may be, for example, in the form of injectable and infusable solutions or suspensions or suppositories or aerosols.


In some embodiments, administration of an Anelloviridae family vector (e.g., anellovector) or composition comprising same, e.g., as described herein, may result in delivery of a genetic element comprised by the Anelloviridae family vector (e.g., anellovector) to a target cell, e.g., in a subject.


An Anelloviridae family vector (e.g., anellovector) or composition thereof described herein, e.g., comprising an effector (e.g., an endogenous or exogenous effector), may be used to deliver the effector to a cell, tissue, or subject. In some embodiments, the effector is a therapeutic effector. In some embodiments, the Anelloviridae family vector (e.g., anellovector) or composition thereof is used to deliver the effector to the eye of a subject, e.g., a mammalian subject, e.g., a human subject. In some embodiments, the Anelloviridae family vector (e.g., anellovector) or composition thereof is used to deliver the effector to a cell of the eye of a subject, e.g., a mammalian subject, e.g., a human subject. In certain embodiments, the cell of the eye is a photoreceptor cell, a retinal cell, a cell of the posterior eye cup (PEC), retinal ganglion cell, a cell of the optic nerve, a cell of the optic nerve head, or a retinal pigmented epithelium (RPE) cell. In some embodiments, the Anelloviridae family vector (e.g., anellovector) or composition thereof is used to deliver the effector to bone marrow, blood, heart, GI or skin. Delivery of an effector by administration of an Anelloviridae family vector (e.g., anellovector) composition described herein may modulate (e.g., increase or decrease) expression levels of a noncoding RNA or polypeptide in the cell, tissue, or subject. Modulation of expression level in this fashion may result in alteration of a functional activity in the cell to which the effector is delivered. In some embodiments, the modulated functional activity may be enzymatic, structural, or regulatory in nature.


In some embodiments, the Anelloviridae family vector (e.g., anellovector), or copies thereof, are detectable in a cell 24 hours (e.g., 1 day, 2 days, 3 days, 4 days, 5 days, 6 days, 1 week, 2 weeks, 3 weeks, 4 weeks, 30 days, or 1 month) after delivery into a cell. In embodiments, an Anelloviridae family vector (e.g., anellovector) or composition thereof mediates an effect on a target cell, and the effect lasts for at least 1, 2, 3, 4, 5, 6, or 7 days, 2, 3, or 4 weeks, or 1, 2, 3, 6, or 12 months. In some embodiments (e.g., wherein the Anelloviridae family vector (e.g., anellovector) or composition thereof comprises a genetic element encoding an exogenous protein), the effect lasts for less than 1, 2, 3, 4, 5, 6, or 7 days, 2, 3, or 4 weeks, or 1, 2, 3, 6, or 12 months.


V. Redosing

The Anelloviridae family vector (e.g., anellovector) described herein can, in some instances, be used as a delivery vehicle that can be administered in multiple doses (e.g., doses administered separately). While not wishing to be bound by theory, in some embodiments, an Anelloviridae family vector (e.g., anellovector) (e.g., as described herein) induces a relatively low immune response (as measured, for example, as 50% GMT values), e.g., allowing for repeated dosing of a subject with one or more Anelloviridae family vectors (e.g., anellovectors) (e.g., multiple doses of the same Anelloviridae family vector (e.g., anellovector) or different Anelloviridae family vectors (e.g., anellovectors)). In an aspect, the invention provides a method of delivering an effector, comprising administering to a subject a first plurality of Anelloviridae family vectors (e.g., anellovectors) and then a second plurality of Anelloviridae family vectors (e.g., anellovectors). In some embodiments, the second plurality of Anelloviridae family vectors (e.g., anellovectors) comprise the same proteinaceous exterior as the Anelloviridae family vectors (e.g., anellovectors) of the first plurality. In another aspect, the invention provides a method of selecting a subject (e.g., a human subject) to receive an effector, wherein the subject previously received, or was identified as having received, a first plurality of Anelloviridae family vectors (e.g., anellovectors) comprising a genetic element encoding an effector, in which the method involves selecting the subject to receive a second plurality of Anelloviridae family vectors (e.g., anellovectors) comprising a genetic element encoding an effector (e.g., the same effector as that encoded by the genetic element of the first plurality of Anelloviridae family vectors (e.g., anellovectors), or a different effector as that encoded by the genetic element of the first plurality of Anelloviridae family vectors (e.g., anellovectors)). In another aspect, the invention provides a method of identifying a subject (e.g., a human subject) as suitable to receive a second plurality of Anelloviridae family vectors (e.g., anellovectors), the method comprising identifying the subject has having previously received a first plurality of Anelloviridae family vectors (e.g., anellovectors) comprising a genetic element encoding an effector, wherein the subject being identified as having received the first plurality of Anelloviridae family vectors (e.g., anellovectors) is indicative that the subject is suitable to receive the second plurality of Anelloviridae family vectors (e.g., anellovectors).


All references and publications cited herein are hereby incorporated by reference.


The following examples are provided to further illustrate some embodiments of the present invention, but are not intended to limit the scope of the invention; it will be understood by their exemplary nature that other procedures, methodologies, or techniques known to those skilled in the art may alternatively be used.


EXAMPLES
Table of Contents





    • Example 1: Anellovector production for in vivo transduction of the CNS

    • Example 2: In vivo transduction of the brain via intracerebroventricular (ICV) administration with an Anellovector (Study #1)

    • Example 3: In vivo delivery of an Anellovector to the spinal cord via intrathecal (IT) administration (Study #2)

    • Example 4: In vivo delivery of an Anellovector to the spinal cord and redosing via IT administration

    • Example 5: In vivo delivery of an Anellovector to brain and redosing via ICV administration





Example 1: Anellovector Production for In Vivo Transduction of the CNS

This example describes the production of the Anellovector and AAV control vector used for Study #1 (Example 2) and Study #2 (Example 3) below for in vivo transduction of the CNS.


AAV9-fCMV-eGFP Control

AAV9-fCMV-eGFP is an adeno-associated virus based on the plasmid pRTx-2770 (SEQ ID NO: 501) with a payload comprising from the 5′ to 3′ direction an AAV2 ITR, Ring2 5′ NCR, CMV promoter, eGFP, SV40pA, Ring2 3′ NCR, and AAV2 ITR (Table Z6). The payload was packaged into AAV9 by Packgene Biotech Inc (Houston, Texas) and generated at a titer of 1×1013 viral genomes (vgs)/ml. Prior to injection, AAV9-fCMV-eGFP was diluted in sterile 1×PBS to the desired titer.









TABLE Z6





pRTx-2770
















Name
pRTx-2770





Type
Plasmid





Length
5128 bp











1
GTCAGTGAGC GAGGAAGCGG AAGAGCGCCC AATACGCAAA CCGCCTCTCC CCGCGCGTTG





61
GCCGATTCAT TAATGCAGCT GGCACGACAG GTTTCCCGAC TGGAAAGCGG GCAGTGAGCG





121
CAACGCAATT AATGTGAGTT AGCTCACTCA TTAGGCACCC CAGGCTTTAC ACTTTATGCT





181
TCCGGCTCGT ATGTTGTGTG GAATTGTGAG CGGATAACAA TTTCACACAG GAAACAGCTA





241
TGACCATGAT TACGCCAAGC TTGCATGCCC TGCAGGCAGC TGCGCGCTCG CTCGCTCACT





301
GAGGCCGCCC GGGCAAAGCC CGGGCGTCGG GCGACCTTTG GTCGCCCGGC CTCAGTGAGC





361
GAGCGAGCGC GCAGAGAGGG AGTGGCCAAC TCCATCACTA GGGGTTCCTG CGAAAGATAT





421
CTAATAAATA TTCAACAGGA AAACCACCTA ATTTAAATTG CCGACCACAA ACCGTCACTT





481
AGTTCCCCTT TTTGCAACAA CTTCTGCTTT TTTCCAACTG CCGGAAAACC ACATAATTTG





541
CATGGCTAAC CACAAACTGA TATGCTAATT AACTTCCACA AAACAACTTC CCCTTTTAAA





601
ACCACACCTA CAAATTAATT ATTAAACACA GTCACATCCT GGGAGGTACT ACCACACTAT





661
AATACCAAGT GCTAATCCGA ATGGCTGAGT TTATGCCGCT AGACGGAGAA CGCATCAGTT





721
ACTGACTGCG GACTGAACTT GGGCGGGTGC CGAAGGTGAG TGAAACCACC GAAGTCAAGG





781
GGCAATTCGG GCTAGTTCAG TCTAGCGGAA CGGGCAAGAA ACTTAAAATT ATTTTATTTT





841
TCAGATGGAT GGCTGATCGA GTGTAGCCAG ATCTGCGATC GACATTGATT ATTGACTAGT





901
TATTAATAGT AATCAATTAC GGGGTCATTA GTTCATAGCC CATATATGGA GTTCCGCGTT





961
ACATAACTTA CGGTAAATGG CCCGCCTGGC TGACCGCCCA ACGACCCCCG CCCATTGACG





1021
TCAATAATGA CGTATGTTCC CATAGTAACG CCAATAGGGA CTTTCCATTG ACGTCAATGG





1081
GTGGAGTATT TACGGTAAAC TGCCCACTTG GCAGTACATC AAGTGTATCA TATGCCAAGT





1141
ACGCCCCCTA TTGACGTCAA TGACGGTAAA TGGCCCGCCT GGCATTATGC CCAGTACATG





1201
ACCTTATGGG ACTTTCCTAC TTGGCAGTAC ATCTACGTAT TAGTCATCGC TATTACCATG





1261
GTGATGCGGT TTTGGCAGTA CATCAATGGG CGTGGATAGC GGTTTGACTC ACGGGGATTT





1321
CCAAGTCTCC ACCCCATTGA CGTCAATGGG AGTTTGTTTT GGCACCAAAA TCAACGGGAC





1381
TTTCCAAAAT GTCGTAACAA CTCCGCCCCA TTGACGCAAA TGGGCGGTAG GCGTGTACGG





1441
TGGGAGGTCT ATATAAGCAG AGCTCTCTGG CTAACTGGAT CTACAAAAAA GCAGATCCAC





1501
CGGTCGCCAC CATGGTGAGC AAGGGCGAGG AGCTGTTCAC CGGGGTGGTG CCCATCCTGG





1561
TCGAGCTGGA CGGCGACGTA AACGGCCACA AGTTCAGCGT GTCCGGCGAG GGCGAGGGCG





1621
ATGCCACCTA CGGCAAGCTG ACCCTGAAGT TCATCTGCAC CACCGGCAAG CTGCCCGTGC





1681
CCTGGCCCAC CCTCGTGACC ACCCTGACCT ACGGCGTGCA GTGCTTCAGC CGCTACCCCG





1741
ACCACATGAA GCAGCACGAC TTCTTCAAGT CCGCCATGCC CGAAGGCTAC GTCCAGGAGC





1801
GCACCATCTT CTTCAAGGAC GACGGCAACT ACAAGACCCG CGCCGAGGTG AAGTTCGAGG





1861
GCGACACCCT GGTGAACCGC ATCGAGCTGA AGGGCATCGA CTTCAAGGAG GACGGCAACA





1921
TCCTGGGGCA CAAGCTGGAG TACAACTACA ACAGCCACAA CGTCTATATC ATGGCCGACA





1981
AGCAGAAGAA CGGCATCAAG GTGAACTTCA AGATCCGCCA CAACATCGAG GACGGCAGCG





2041
TGCAGCTCGC CGACCACTAC CAGCAGAACA CCCCCATCGG CGACGGCCCC GTGCTGCTGC





2101
CCGACAACCA CTACCTGAGC ACCCAGTCCG CCCTGAGCAA AGACCCCAAC GAGAAGCGCG





2161
ATCACATGGT CCTGCTGGAG TTCGTGACCG CCGCCGGGAT CACTCTCGGC ATGGACGAGC





2221
TGTACAAGTA ATAAGCTTGC GGCCGCTTCG AGCAGACATG ATAAGATACA TTGATGAGTT





2281
TGGACAAACC ACAACTAGAA TGCAGTGAAA AAAATGCTTT ATTTGTGAAA TTTGTGATGC





2341
TATTGCTTTA TTTGTAACCA TTATAAGCTG CAATAAACAA GTTAACAACA ACAATTGCAT





2401
GAATGAATAA AGGCCAGCAT TAATTCACTT AAGGAGTCTG TTTATTTAAG TTAAACCTTA





2461
ATAAACGGTC ACCGCCTCCC TAATACGCAG GCGCAGAAAG GGGGCTCCGC CCCCTTTAAC





2521
CCCCAGGGGG CTCCGCCCCC TGAAACCCCC AAGGGGGCTA CGCCCCCTTA CACCCCCGAT





2581
ATCCCCGCAG GAACCCCTAG TGATGGAGTT GGCCACTCCC TCTCTGCGCG CTCGCTCGCT





2641
CACTGAGGCC GGGCGACCAA AGGTCGCCCG ACGCCCGGGC TTTGCCCGGG CGGCCTCAGT





2701
GAGCGAGCGA GCGCGCAGCT GCCTGCAGGG GCGCCTGGGT ACCGAGCTCG AATTCACTGG





2761
CCGTCGTTTT ACAACGTCGT GACTGGGAAA ACCCTGGCGT TACCCAACTT AATCGCCTTG





2821
CAGCACATCC CCCTTTCGCC AGCTGGCGTA ATAGCGAAGA GGCCCGCACC GATCGCCCTT





2881
CCCAACAGTT GCGCAGCCTG AATGGCGAAT GGCGCCTGAT GCGGTATTTT CTCCTTACGC





2941
ATCTGTGCGG TATTTCACAC CGCATATGGT GCACTCTCAG TACAATCTGC TCTGATGCCG





3001
CATAGTTAAG CCAGCCCCGA CACCCGCCAA CACCCGCTGA CGCGCCCTGA CGGGCTTGTC





3061
TGCTCCCGGC ATCCGCTTAC AGACAAGCTG TGACCGTCTC CGGGAGCTGC ATGTGTCAGA





3121
GGTTTTCACC GTCATCACCG AAACGCGCGA GACGAAAGGG CCTCGTGATA CGCCTATTTT





3181
TATAGGTTAA TGTCATGATA ATAATGGTTT CTTAGACGTC AGGTGGCACT TTTCGGGGAA





3241
ATGTGCGCGG AACCCCTATT TGTTTATTTT TCTAAATACA TTCAAATATG TATCCGCTCA





3301
TGAGACAATA ACCCTGATAA ATGCTTCAAT AATATTGAAA AAGGAAGAGT ATGAGTATTC





3361
AACATTTCCG TGTCGCCCTT ATTCCCTTTT TTGCGGCATT TTGCCTTCCT GTTTTTGCTC





3421
ACCCAGAAAC GCTGGTGAAA GTAAAAGATG CTGAAGATCA GTTGGGTGCA CGAGTGGGTT





3481
ACATCGAACT GGATCTCAAC AGCGGTAAGA TCCTTGAGAG TTTTCGCCCC GAAGAACGTT





3541
TTCCAATGAT GAGCACTTTT AAAGTTCTGC TATGTGGCGC GGTATTATCC CGTATTGACG





3601
CCGGGCAAGA GCAACTCGGT CGCCGCATAC ACTATTCTCA GAATGACTTG GTTGAGTACT





3661
CACCAGTCAC AGAAAAGCAT CTTACGGATG GCATGACAGT AAGAGAATTA TGCAGTGCTG





3721
CCATAACCAT GAGTGATAAC ACTGCGGCCA ACTTACTTCT GACAACGATC GGAGGACCGA





3781
AGGAGCTAAC CGCTTTTTTG CACAACATGG GGGATCATGT AACTCGCCTT GATCGTTGGG





3841
AACCGGAGCT GAATGAAGCC ATACCAAACG ACGAGCGTGA CACCACGATG CCTGTAGCAA





3901
TGGCAACAAC GTTGCGCAAA CTATTAACTG GCGAACTACT TACTCTAGCT TCCCGGCAAC





3961
AATTAATAGA CTGGATGGAG GCGGATAAAG TTGCAGGACC ACTTCTGCGC TCGGCCCTTC





4021
CGGCTGGCTG GTTTATTGCT GATAAATCTG GAGCCGGTGA GCGTGGGTCT CGCGGTATCA





4081
TTGCAGCACT GGGGCCAGAT GGTAAGCCCT CCCGTATCGT AGTTATCTAC ACGACGGGGA





4141
GTCAGGCAAC TATGGATGAA CGAAATAGAC AGATCGCTGA GATAGGTGCC TCACTGATTA





4201
AGCATTGGTA ACTGTCAGAC CAAGTTTACT CATATATACT TTAGATTGAT TTAAAACTTC





4261
ATTTTTAATT TAAAAGGATC TAGGTGAAGA TCCTTTTTGA TAATCTCATG ACCAAAATCC





4321
CTTAACGTGA GTTTTCGTTC CACTGAGCGT CAGACCCCGT AGAAAAGATC AAAGGATCTT





4381
CTTGAGATCC TTTTTTTCTG CGCGTAATCT GCTGCTTGCA AACAAAAAAA CCACCGCTAC





4441
CAGCGGTGGT TTGTTTGCCG GATCAAGAGC TACCAACTCT TTTTCCGAAG GTAACTGGCT





4501
TCAGCAGAGC GCAGATACCA AATACTGTTC TTCTAGTGTA GCCGTAGTTA GGCCACCACT





4561
TCAAGAACTC TGTAGCACCG CCTACATACC TCGCTCTGCT AATCCTGTTA CCAGTGGCTG





4621
CTGCCAGTGG CGATAAGTCG TGTCTTACCG GGTTGGACTC AAGACGATAG TTACCGGATA





4681
AGGCGCAGCG GTCGGGCTGA ACGGGGGGTT CGTGCACACA GCCCAGCTTG GAGCGAACGA





4741
CCTACACCGA ACTGAGATAC CTACAGCGTG AGCTATGAGA AAGCGCCACG CTTCCCGAAG





4801
GGAGAAAGGC GGACAGGTAT CCGGTAAGCG GCAGGGTCGG AACAGGAGAG CGCACGAGGG





4861
AGCTTCCAGG GGGAAACGCC TGGTATCTTT ATAGTCCTGT CGGGTTTCGC CACCTCTGAC





4921
TTGAGCGTCG ATTTTTGTGA TGCTCGTCAG GGGGGCGGAG CCTATGGAAA AACGCCAGCA





4981
ACGCGGCCTT TTTACGGTTC CTGGCCTTTT GCTGGCCTTT TGCTCACATG TTCTTTCCTG





5041
CGTTATCCCC TGATTCTGTG GATAACCGTA TTACCGCCTT TGAGTGAGCT GATACCGCTC





5101
GCCGCAGCCG AACGACCGAG CGCAGCGA (SEQ ID NO: 501)










Annotations:








Region/Element
Base range






E. coli CAP protein binding site

 130..151





Lac operon promoter
 166..196





LacI repressor protein binding site
 204..220





AAV2 inverted terminal repeat
 269..409





Ring2 5′NCR
 421..844





CMV enhancer
 881..1260





Full CMV promoter
 896..1465





CMV promoter
1261..1464





Inert 5′ UTR
1477..1507





Kozak sequence
1506..1515





eGFP coding sequence
1512..2231





SV40 poly(A) sequence
2262..2383





Ring2 3′NCR
2411..2575





AAV2 inverted terminal repeat
2589..2729





AmpR promoter
3246..3350





Beta lactamase ampicillin resistance
3351..4211


coding sequence






High-copy-number
4382..4970


ColE1/pMB1/pBR322/pUC origin of



replication










Exemplary eGFP Amino Acid Sequence (e.g., Encoded by the Nucleotides 1512-2231 of Table Z6):









(SEQ ID NO: 1113)


MVSKGEELFTGVVPILVELDGDVNGHKFSVSGEGEGDATYGKLTLKFIC





TTGKLPVPWPTLVTTLTYGVQCFSRYPDHMKQHDFFKSAMPEGYVQERT





IFFKDDGNYKTRAEVKFEGDTLVNRIELKGIDFKEDGNILGHKLEYNYN





SHNVYIMADKQKNGIKVNFKIRHNIEDGSVQLADHYQQNTPIGDGPVLL





PDNHYLSTQSALSKDPNEKRDHMVLLEFVTAAGITLGMDELYK






Exemplary Beta Lactamase Amino Acid Sequence (e.g., Encoded by of Nucleotides 3351-4211 of Table Z6):








(SEQ ID NO: 1114)


MSIQHFRVALIPFFAAFCLPVFAHPETLVKVKDAEDQLGARVGYIELDL





NSGKILESFRPEERFPMMSTFKVLLCGAVLSRIDAGQEQLGRRIHYSQN





DLVEYSPVTEKHLTDGMTVRELCSAAITMSDNTAANLLLTTIGGPKELT





AFLHNMGDHVTRLDRWEPELNEAIPNDERDTTMPVAMATTLRKLLTGEL





LTLASRQQLIDWMEADKVAGPLLRSALPAGWFIADKSGAGERGSRGIIA





ALGPDGKPSRIVVIYTTGSQATMDERNRQIAEIGASLIKHW






Ring19-fCMV-eGFP Anellovector
Anellovirus Vector Production: Parental Cell Culture

MOLT-4 cells were obtained from the National Cancer Institute. Cells were scaled-up and maintained in suspension culture in complete growth medium (Gibco's RPMI 1640 with 10% fetal bovine serum (FBS), supplemented with 1 mM sodium pyruvate, Pluronic F-68 [0.1%], and 2 mM L-glutamine) at 37° C. with 5% CO2. Cells were seeded into shake flasks (2-L, flat-bottomed, Erlenmeyer flask), each with a working volume of 800 mL, at a density of 0.1E+06 viable cells/mL and cultured in an orbital shaker (New Brunswick Innova 2100, 19-mm circular orbit) at 37° C. and 90 rpm with >85% relative humidity (RH) for 4 days.


Anellovirus Vector Production: Transfection of MOLT-4 Cells

Ring19-fCMV-eGFP Anellovector was produced using a distinct combination of three plasmids listed in Table E0. MOLT-4 cells were transfected with the three plasmids via electroporation.


For each of the viral vector production culture preps, 5E+08 MOLT-4 cells were pelleted and resuspended in Opti-MEM I Reduced Serum Medium. 800 μg of the plasmids were added to each of the resuspended cells and electroporated using a MaxCyte STx electroporator and CL1.1 electroporator assemblies (MaxCyte catalog #SCL1). Each batch of electroporated cells were then transferred to separate flasks containing pre-warmed complete growth medium. Transfected cells were allowed to incubate at 37° C. with 5% CO2 and harvested via centrifugation 72 hours post-electroporation. 10× images were captured with an EVOS (M7000, Invitrogen by ThermoFisher Scientific) (FIG. 1A and FIG. 1B).









TABLE E0







Plasmids transfected to produce


Ring19-fCMV-eGFP Anellovector










Anellovector
Plasmids







Ring19-fCMV-eGFP
pRTx-2847 (R19-eGFP vector)



(“R19-eGFP”)
(SEQ ID NO: 502)




pRTx-2848 (iCre plasmid)




(SEQ ID NO: 503)




pRTx-3525 (R19 SRR)




(SEQ ID NO: 500)










Genetic Element Construct Plasmid

The Ring19 genetic element construct, pRTX-2847 (SEQ ID NO: 502), was designed with lox66 and lox 71 sites flanking a RING19 non-coding region (NCR) and a CMV::egfp::WPRE::bGH-pA payload cassette. The sequence for the construct is provided below.









TABLE Z1





Exemplary floxed Ring19 vector plasmid for three-plasmid systems (pRTx-2847)
















Name
pRTx-2847





Type
Plasmid





Description
pLox-Ring19AORF::fCMV_EGFP_WPRE_bGH-pA-Rand100.





Length
5452 bp











1
TCGCGCGTTT CGGTGATGAC GGTGAAAACC TCTGACACAT GCAGCTCCCG GAGACGGTCA





61
CAGCTTGTCT GTAAGCGGAT GCCGGGAGCA GACAAGCCCG TCAGGGCGCG TCAGCGGGTG





121
TTGGCGGGTG TCGGGGCTGG CTTAACTATG CGGCATCAGA GCAGATTGTA CTGAGAGTGC





181
ACCATATGCG GTGTGAAATA CCGCACAGAT GCGTAAGGAG AAAATACCGC ATCAGGCGCC





241
ATTCGCCATT CAGGCTGCGC AACTGTTGGG AAGGGCGATC GGTGCGGGCC TCTTCGCTAT





301
TACGCCAGCT GGCGAAAGGG GGATGTGCTG CAAGGCGATT AAGTTGGGTA ACGCCAGGGT





361
TTTCCCAGTC ACGACGTTGT AAAACGACGG CCAGAGAATT CGAGCTCGGT ACCTCGCGAA





421
TACATCTAGA TTACCGTTCG TATAGCATAC ATTATACGAA GTTATTAAAC TACGCCTGCA





481
AACTTTCACT CTCGGTGTCC ATTTATATAA GATAAAACTT AAATAAACAT CCACCACTCT





541
CCCAAATACG CAGGCGCACA AGGGGGCTCC GCCCCCTTAA ACCCCCAAGG GGGCTCCGCC





601
CCCTTAAACC CCCAAGGGGG CTCCGCCCCC TTACACCCCC TAATAAATAT TCAACAGGAA





661
AACCACCTAA TTAGAATTGC CGACCACAAA CCGTCACTTA CTTCTCCTTT TTGCACTTAC





721
TTCCTCTTTT ACTTATTATT ATTCATTACA TTAATTAATA ATCACTGTAA TTCCGGGGAG





781
GAGCTAACAA TCTATATAAC TAACTACACT TCCGAATGGC TGAGTTTATG CCGCCAGACG





841
GAGACGGGAT CACTTCAGTG ACTCCAGGCT GAACTTGGGC GGGAGCCGAA GGTGAGTGCA





901
ACCACCGTAG TCTAGGGGCA ATTCGGGCTA GTTCAGTATG GCGGAACGGG CAAGAAACTT





961
AAATATTATT ATTTTACAGA TGGGCGTTGA CATTGATTAT TGACTAGTTA TTAATAGTAA





1021
TCAATTACGG GGTCATTAGT TCATAGCCCA TATATGGAGT TCCGCGTTAC ATAACTTACG





1081
GTAAATGGCC CGCCTGGCTG ACCGCCCAAC GACCCCCGCC CATTGACGTC AATAATGACG





1141
TATGTTCCCA TAGTAACGCC AATAGGGACT TTCCATTGAC GTCAATGGGT GGAGTATTTA





1201
CGGTAAACTG CCCACTTGGC AGTACATCAA GTGTATCATA TGCCAAGTAC GCCCCCTATT





1261
GACGTCAATG ACGGTAAATG GCCCGCCTGG CATTATGCCC AGTACATGAC CTTATGGGAC





1321
TTTCCTACTT GGCAGTACAT CTACGTATTA GTCATCGCTA TTACCATGGT GATGCGGTTT





1381
TGGCAGTACA TCAATGGGCG TGGATAGCGG TTTGACTCAC GGGGATTTCC AAGTCTCCAC





1441
CCCATTGACG TCAATGGGAG TTTGTTTTGG CACCAAAATC AACGGGACTT TCCAAAATGT





1501
CGTAACAACT CCGCCCCATT GACGCAAATG GGCGGTAGGC GTGTACGGTG GGAGGTCTAT





1561
ATAAGCAGAG CTCTCTGGCT AACTGGATCT ACAAAAAAGC AGATCCACCG GTCGCCACCA





1621
TGGTGAGCAA GGGCGAGGAG CTGTTCACCG GGGTGGTGCC CATCCTGGTC GAGCTGGACG





1681
GCGACGTAAA CGGCCACAAG TTCAGCGTGT CCGGCGAGGG CGAGGGCGAT GCCACCTACG





1741
GCAAGCTGAC CCTGAAGTTC ATCTGCACCA CCGGCAAGCT GCCCGTGCCC TGGCCCACCC





1801
TCGTGACCAC CCTGACCTAC GGCGTGCAGT GCTTCAGCCG CTACCCCGAC CACATGAAGC





1861
AGCACGACTT CTTCAAGTCC GCCATGCCCG AAGGCTACGT CCAGGAGCGC ACCATCTTCT





1921
TCAAGGACGA CGGCAACTAC AAGACCCGCG CCGAGGTGAA GTTCGAGGGC GACACCCTGG





1981
TGAACCGCAT CGAGCTGAAG GGCATCGACT TCAAGGAGGA CGGCAACATC CTGGGGCACA





2041
AGCTGGAGTA CAACTACAAC AGCCACAACG TCTATATCAT GGCCGACAAG CAGAAGAACG





2101
GCATCAAGGT GAACTTCAAG ATCCGCCACA ACATCGAGGA CGGCAGCGTG CAGCTCGCCG





2161
ACCACTACCA GCAGAACACC CCCATCGGCG ACGGCCCCGT GCTGCTGCCC GACAACCACT





2221
ACCTGAGCAC CCAGTCCGCC CTGAGCAAAG ACCCCAACGA GAAGCGCGAT CACATGGTCC





2281
TGCTGGAGTT CGTGACCGCC GCCGGGATCA CTCTCGGCAT GGACGAGCTG TACAAGTAAT





2341
AAAATCAACC TCTGGATTAC AAAATTTGTG AAAGATTGAC TGGTATTCTT AACTATGTTG





2401
CTCCTTTTAC GCTATGTGGA TACGCTGCTT TAATGCCTTT GTATCATGCT ATTGCTTCCC





2461
GTATGGCTTT CATTTTCTCC TCCTTGTATA AATCCTGGTT GCTGTCTCTT TATGAGGAGT





2521
TGTGGCCCGT TGTCAGGCAA CGTGGCGTGG TGTGCACTGT GTTTGCTGAC GCAACCCCCA





2581
CTGGTTGGGG CATTGCCACC ACCTGTCAGC TCCTTTCCGG GACTTTCGCT TTCCCCCTCC





2641
CTATTGCCAC GGCGGAACTC ATCGCCGCCT GCCTTGCCCG CTGCTGGACA GGGGCTCGGC





2701
TGTTGGGCAC TGACAATTCC GTGGTGTTGT CGGGGAAGCT GACGTCCTTT CCATGGCTGC





2761
TCGCCTGTGT TGCCACCTGG ATTCTGCGCG GGACGTCCTT CTGCTACGTC CCTTCGGCCC





2821
TCAATCCAGC GGACCTTCCT TCCCGCGGCC TGCTGCCGGC TCTGCGGCCT CTTCCGCGTC





2881
TTCGCCTTCG CCCTCAGACG AGTCGGATCT CCCTTTGGGC CGCCTCCCCG CCTGGTTCCG





2941
ACTGTGCCTT CTAGTTGCCA GCCATCTGTT GTTTGCCCCT CCCCCGTGCC TTCCTTGACC





3001
CTGGAAGGTG CCACTCCCAC TGTCCTTTCC TAATAAAATG AGGAAATTGC ATCGCATTGT





3061
CTGAGTAGGT GTCATTCTAT TCTGGGGGGT GGGGTGGGGC AGGACAGCAA GGGGGAGGAT





3121
TGGGTAGACA ATAGCAGGCA TGCTGGGGAT GCGGTGGGCT CTATGGTCGA GTTAGTTTGC





3181
TCCAGTAAAG TTGTTTATAA TAACTACTAA ATCCGCATGT TACGGAATTT CTTATTAATT





3241
TTTTTTTCGT AAGGAACAAC GGATCTTGAA ATAACTTCGT ATAGCATACA TTATACGAAC





3301
GGTAATCGGA TCCCGGGCCC GTCGACTGCA GAGGCCTGCA TGCAAGCTTG GTGTAATCAT





3361
GGTCATAGCT GTTTCCTGTG TGAAATTGTT ATCCGCTCAC AATTCCACAC AACATACGAG





3421
CCGGAAGCAT AAAGTGTAAA GCCTGGGGTG CCTAATGAGT GAGCTAACTC ACATTAATTG





3481
CGTTGCGCTC ACTGCCCGCT TTCCAGTCGG GAAACCTGTC GTGCCAGCTG CATTAATGAA





3541
TCGGCCAACG CGCGGGGAGA GGCGGTTTGC GTATTGGGCG CTCTTCCGCT TCCTCGCTCA





3601
CTGACTCGCT GCGCTCGGTC GTTCGGCTGC GGCGAGCGGT ATCAGCTCAC TCAAAGGCGG





3661
TAATACGGTT ATCCACAGAA TCAGGGGATA ACGCAGGAAA GAACATGTGA GCAAAAGGCC





3721
AGCAAAAGGC CAGGAACCGT AAAAAGGCCG CGTTGCTGGC GTTTTTCCAT AGGCTCCGCC





3781
CCCCTGACGA GCATCACAAA AATCGACGCT CAAGTCAGAG GTGGCGAAAC CCGACAGGAC





3841
TATAAAGATA CCAGGCGTTT CCCCCTGGAA GCTCCCTCGT GCGCTCTCCT GTTCCGACCC





3901
TGCCGCTTAC CGGATACCTG TCCGCCTTTC TCCCTTCGGG AAGCGTGGCG CTTTCTCATA





3961
GCTCACGCTG TAGGTATCTC AGTTCGGTGT AGGTCGTTCG CTCCAAGCTG GGCTGTGTGC





4021
ACGAACCCCC CGTTCAGCCC GACCGCTGCG CCTTATCCGG TAACTATCGT CTTGAGTCCA





4081
ACCCGGTAAG ACACGACTTA TCGCCACTGG CAGCAGCCAC TGGTAACAGG ATTAGCAGAG





4141
CGAGGTATGT AGGCGGTGCT ACAGAGTTCT TGAAGTGGTG GCCTAACTAC GGCTACACTA





4201
GAAGAACAGT ATTTGGTATC TGCGCTCTGC TGAAGCCAGT TACCTTCGGA AAAAGAGTTG





4261
GTAGCTCTTG ATCCGGCAAA CAAACCACCG CTGGTAGCGG TGGTTTTTTT GTTTGCAAGC





4321
AGCAGATTAC GCGCAGAAAA AAAGGATCTC AAGAAGATCC TTTGATCTTT TCTACGGGGT





4381
CTGACGCTCA GTGGAACGAA AACTCACGTT AAGGGATTTT GGTCATGAGA TTATCAAAAA





4441
GGATCTTCAC CTAGATCCTT TTAAATTAAA AATGAAGTTT TAAATCAAGC CCAATCTGAA





4501
TAATGTTACA ACCAATTAAC CAATTCTGAT TAGAAAAACT CATCGAGCAT CAAATGAAAC





4561
TGCAATTTAT TCATATCAGG ATTATCAATA CCATATTTTT GAAAAAGCCG TTTCTGTAAT





4621
GAAGGAGAAA ACTCACCGAG GCAGTTCCAT AGGATGGCAA GATCCTGGTA TCGGTCTGCG





4681
ATTCCGACTC GTCCAACATC AATACAACCT ATTAATTTCC CCTCGTCAAA AATAAGGTTA





4741
TCAAGTGAGA AATCACCATG AGTGACGACT GAATCCGGTG AGAATGGCAA AAGTTTATGC





4801
ATTTCTTTCC AGACTTGTTC AACAGGCCAG CCATTACGCT CGTCATCAAA ATCACTCGCA





4861
TCAACCAAAC CGTTATTCAT TCGTGATTGC GCCTGAGCGA GACGAAATAC GCGATCGCTG





4921
TTAAAAGGAC AATTACAAAC AGGAATCGAA TGCAACCGGC GCAGGAACAC TGCCAGCGCA





4981
TCAACAATAT TTTCACCTGA ATCAGGATAT TCTTCTAATA CCTGGAATGC TGTTTTTCCG





5041
GGGATCGCAG TGGTGAGTAA CCATGCATCA TCAGGAGTAC GGATAAAATG CTTGATGGTC





5101
GGAAGAGGCA TAAATTCCGT CAGCCAGTTT AGTCTGACCA TCTCATCTGT AACATCATTG





5161
GCAACGCTAC CTTTGCCATG TTTCAGAAAC AACTCTGGCG CATCGGGCTT CCCATACAAG





5221
CGATAGATTG TCGCACCTGA TTGCCCGACA TTATCGCGAG CCCATTTATA CCCATATAAA





5281
TCAGCATCCA TGTTGGAATT TAATCGCGGC CTCGACGTTT CCCGTTGAAT ATGGCTCATA





5341
ACACCCCTTG TATTACTGTT TATGTAAGCA GACAGTTTTA TTGTTCATGA TGATATATTT





5401
TTATCTTGTG CAATGTAACA TCAGAGATTT TGAGACACGG GCCAGAGCTG CA (SEQ ID NO: 502)










Annotations:








Region/Element
Base range





Lox71 (loxP site)
 432-465





GC-rich region
 529-640





Initiator element
 813-828





5′ UTR conserved domain
 880-950





Intron 1
 892-979





Full CMV promoter
1004-1573





Inert 5′ UTR
1585-1619





eGFP coding sequence
1620-2339





WPRE
2343-2934





bGHpA terminator sequence
2939-3166





100 bp random stuffer sequence
3167-3266





Lox66 (loxP site)
3271-3304





LacI repressor binding site (complement)
3384-3406





Lac operon operator (lacO)
3386-3402





Lac operon promoter (complement)
3410-3440





C-tag
3418-3429






E coli catabolite activator protein binding site

3455-3476


(complement)






Origin of replication (pUC origin) (complement)
3705-4378





Kanamycin resistance (KanR) CDS (complement)
4530-5339









Cre Expression Plasmid

pRTX-2848 (SEQ ID NO: 503) expresses iCre from the CMV promoter. The sequence of this construct is provided below.









TABLE Z5





Exemplary Cre recombinase expression plasmid (pRTx-2848)
















Name
pRTx-2848





Type
Plasmid





Description
CMV_iCre_pcDNA3.1(+).





Length
6473 bp











1
GACGGATCGG GAGATCTCCC GATCCCCTAT GGTGCACTCT CAGTACAATC TGCTCTGATG





61
CCGCATAGTT AAGCCAGTAT CTGCTCCCTG CTTGTGTGTT GGAGGTCGCT GAGTAGTGCG





121
CGAGCAAAAT TTAAGCTACA ACAAGGCAAG GCTTGACCGA CAATTGCATG AAGAATCTGC





181
TTAGGGTTAG GCGTTTTGCG CTGCTTCGCG ATGTACGGGC CAGATATACG CGTTGACATT





241
GATTATTGAC TAGTTATTAA TAGTAATCAA TTACGGGGTC ATTAGTTCAT AGCCCATATA





301
TGGAGTTCCG CGTTACATAA CTTACGGTAA ATGGCCCGCC TGGCTGACCG CCCAACGACC





361
CCCGCCCATT GACGTCAATA ATGACGTATG TTCCCATAGT AACGCCAATA GGGACTTTCC





421
ATTGACGTCA ATGGGTGGAG TATTTACGGT AAACTGCCCA CTTGGCAGTA CATCAAGTGT





481
ATCATATGCC AAGTACGCCC CCTATTGACG TCAATGACGG TAAATGGCCC GCCTGGCATT





541
ATGCCCAGTA CATGACCTTA TGGGACTTTC CTACTTGGCA GTACATCTAC GTATTAGTCA





601
TCGCTATTAC CATGGTGATG CGGTTTTGGC AGTACATCAA TGGGCGTGGA TAGCGGTTTG





661
ACTCACGGGG ATTTCCAAGT CTCCACCCCA TTGACGTCAA TGGGAGTTTG TTTTGGCACC





721
AAAATCAACG GGACTTTCCA AAATGTCGTA ACAACTCCGC CCCATTGACG CAAATGGGCG





781
GTAGGCGTGT ACGGTGGGAG GTCTATATAA GCAGAGCTCT CTGGCTAACT AGAGAACCCA





841
CTGCTTACTG GCTTATCGAA ATTAATACGA CTCACTATAG GGAGACCCAA GCTGGCTAGC





901
GTTTAAACTT AAGCTTGGTA CCGAGCTCGG ATCCACTAGT CCAGTGTGGT GGAATTCGCC





961
ACCATGGTGC CCAAGAAGAA GAGGAAAGTC TCCAACCTGC TGACTGTGCA CCAAAACCTG





1021
CCTGCCCTCC CTGTGGATGC CACCTCTGAT GAAGTCAGGA AGAACCTGAT GGACATGTTC





1081
AGGGACAGGC AGGCCTTCTC TGAACACACC TGGAAGATGC TCCTGTCTGT GTGCAGATCC





1141
TGGGCTGCCT GGTGCAAGCT GAACAACAGG AAATGGTTCC CTGCTGAACC TGAGGATGTG





1201
AGGGACTACC TCCTGTACCT GCAAGCCAGA GGCCTGGCTG TGAAAACCAT CCAACAGCAC





1261
CTGGGCCAGC TCAACATGCT GCACAGGAGA TCTGGCCTGC CTCGCCCTTC TGACTCCAAT





1321
GCTGTGTCCC TGGTGATGAG GAGAATCAGA AAGGAGAATG TGGATGCTGG GGAGAGAGCC





1381
AAGCAGGCCC TGGCCTTTGA ACGCACTGAC TTTGACCAAG TCAGATCCCT GATGGAGAAC





1441
TCTGACAGAT GCCAGGACAT CAGGAACCTG GCCTTCCTGG GCATTGCCTA CAACACCCTG





1501
CTGCGCATTG CCGAAATTGC CAGAATCAGA GTGAAGGACA TCTCCCGCAC CGATGGTGGG





1561
AGAATGCTGA TCCACATTGG CAGGACCAAG ACCCTGGTGT CCACAGCTGG TGTGGAGAAG





1621
GCCCTGTCCC TGGGGGTTAC CAAGCTGGTG GAGAGATGGA TCTCTGTGTC TGGTGTGGCT





1681
GATGACCCCA ACAACTACCT GTTCTGCCGG GTCAGAAAGA ATGGTGTGGC TGCCCCTTCT





1741
GCCACCTCCC AACTGTCCAC CCGCGCCCTG GAAGGGATCT TTGAGGCCAC CCACCGCCTG





1801
ATCTATGGTG CCAAGGATGA CTCTGGGCAG AGATACCTGG CCTGGTCTGG CCACTCTGCC





1861
AGAGTGGGTG CTGCCAGGGA CATGGCCAGG GCTGGTGTGT CCATCCCTGA AATCATGCAG





1921
GCTGGTGGCT GGACCAATGT GAACATAGTG ATGAACTACA TCAGAAACCT GGACTCTGAG





1981
ACTGGGGCCA TGGTGAGGCT GCTCGAGGAT GGGGACTGAG CGGCCGCTCG AGTCTAGAGG





2041
GCCCGTTTAA ACCCGCTGAT CAGCCTCGAC TGTGCCTTCT AGTTGCCAGC CATCTGTTGT





2101
TTGCCCCTCC CCCGTGCCTT CCTTGACCCT GGAAGGTGCC ACTCCCACTG TCCTTTCCTA





2161
ATAAAATGAG GAAATTGCAT CGCATTGTCT GAGTAGGTGT CATTCTATTC TGGGGGGTGG





2221
GGTGGGGCAG GACAGCAAGG GGGAGGATTG GGAAGACAAT AGCAGGCATG CTGGGGATGC





2281
GGTGGGCTCT ATGGCTTCTG AGGCGGAAAG AACCAGCTGG GGCTCTAGGG GGTATCCCCA





2341
CGCGCCCTGT AGCGGCGCAT TAAGCGCGGC GGGTGTGGTG GTTACGCGCA GCGTGACCGC





2401
TACACTTGCC AGCGCCCTAG CGCCCGCTCC TTTCGCTTTC TTCCCTTCCT TTCTCGCCAC





2461
GTTCGCCGGC TTTCCCCGTC AAGCTCTAAA TCGGGGGCTC CCTTTAGGGT TCCGATTTAG





2521
TGCTTTACGG CACCTCGACC CCAAAAAACT TGATTAGGGT GATGGTTCAC GTAGTGGGCC





2581
ATCGCCCTGA TAGACGGTTT TTCGCCCTTT GACGTTGGAG TCCACGTTCT TTAATAGTGG





2641
ACTCTTGTTC CAAACTGGAA CAACACTCAA CCCTATCTCG GTCTATTCTT TTGATTTATA





2701
AGGGATTTTG CCGATTTCGG CCTATTGGTT AAAAAATGAG CTGATTTAAC AAAAATTTAA





2761
CGCGAATTAA TTCTGTGGAA TGTGTGTCAG TTAGGGTGTG GAAAGTCCCC AGGCTCCCCA





2821
GCAGGCAGAA GTATGCAAAG CATGCATCTC AATTAGTCAG CAACCAGGTG TGGAAAGTCC





2881
CCAGGCTCCC CAGCAGGCAG AAGTATGCAA AGCATGCATC TCAATTAGTC AGCAACCATA





2941
GTCCCGCCCC TAACTCCGCC CATCCCGCCC CTAACTCCGC CCAGTTCCGC CCATTCTCCG





3001
CCCCATGGCT GACTAATTTT TTTTATTTAT GCAGAGGCCG AGGCCGCCTC TGCCTCTGAG





3061
CTATTCCAGA AGTAGTGAGG AGGCTTTTTT GGAGGCCTAG GCTTTTGCAA AAAGCTCCCG





3121
GGAGCTTGTA TATCCATTTT CGGATCTGAT CAAGAGACAG GATGAGGATC GTTTCGCATG





3181
ATTGAACAAG ATGGATTGCA CGCAGGTTCT CCGGCCGCTT GGGTGGAGAG GCTATTCGGC





3241
TATGACTGGG CACAACAGAC AATCGGCTGC TCTGATGCCG CCGTGTTCCG GCTGTCAGCG





3301
CAGGGGCGCC CGGTTCTTTT TGTCAAGACC GACCTGTCCG GTGCCCTGAA TGAACTGCAG





3361
GACGAGGCAG CGCGGCTATC GTGGCTGGCC ACGACGGGCG TTCCTTGCGC AGCTGTGCTC





3421
GACGTTGTCA CTGAAGCGGG AAGGGACTGG CTGCTATTGG GCGAAGTGCC GGGGCAGGAT





3481
CTCCTGTCAT CTCACCTTGC TCCTGCCGAG AAAGTATCCA TCATGGCTGA TGCAATGCGG





3541
CGGCTGCATA CGCTTGATCC GGCTACCTGC CCATTCGACC ACCAAGCGAA ACATCGCATC





3601
GAGCGAGCAC GTACTCGGAT GGAAGCCGGT CTTGTCGATC AGGATGATCT GGACGAAGAG





3661
CATCAGGGGC TCGCGCCAGC CGAACTGTTC GCCAGGCTCA AGGCGCGCAT GCCCGACGGC





3721
GAGGATCTCG TCGTGACCCA TGGCGATGCC TGCTTGCCGA ATATCATGGT GGAAAATGGC





3781
CGCTTTTCTG GATTCATCGA CTGTGGCCGG CTGGGTGTGG CGGACCGCTA TCAGGACATA





3841
GCGTTGGCTA CCCGTGATAT TGCTGAAGAG CTTGGCGGCG AATGGGCTGA CCGCTTCCTC





3901
GTGCTTTACG GTATCGCCGC TCCCGATTCG CAGCGCATCG CCTTCTATCG CCTTCTTGAC





3961
GAGTTCTTCT GAGCGGGACT CTGGGGTTCG AAATGACCGA CCAAGCGACG CCCAACCTGC





4021
CATCACGAGA TTTCGATTCC ACCGCCGCCT TCTATGAAAG GTTGGGCTTC GGAATCGTTT





4081
TCCGGGACGC CGGCTGGATG ATCCTCCAGC GCGGGGATCT CATGCTGGAG TTCTTCGCCC





4141
ACCCCAACTT GTTTATTGCA GCTTATAATG GTTACAAATA AAGCAATAGC ATCACAAATT





4201
TCACAAATAA AGCATTTTTT TCACTGCATT CTAGTTGTGG TTTGTCCAAA CTCATCAATG





4261
TATCTTATCA TGTCTGTATA CCGTCGACCT CTAGCTAGAG CTTGGCGTAA TCATGGTCAT





4321
AGCTGTTTCC TGTGTGAAAT TGTTATCCGC TCACAATTCC ACACAACATA CGAGCCGGAA





4381
GCATAAAGTG TAAAGCCTGG GGTGCCTAAT GAGTGAGCTA ACTCACATTA ATTGCGTTGC





4441
GCTCACTGCC CGCTTTCCAG TCGGGAAACC TGTCGTGCCA GCTGCATTAA TGAATCGGCC





4501
AACGCGCGGG GAGAGGCGGT TTGCGTATTG GGCGCTCTTC CGCTTCCTCG CTCACTGACT





4561
CGCTGCGCTC GGTCGTTCGG CTGCGGCGAG CGGTATCAGC TCACTCAAAG GCGGTAATAC





4621
GGTTATCCAC AGAATCAGGG GATAACGCAG GAAAGAACAT GTGAGCAAAA GGCCAGCAAA





4681
AGGCCAGGAA CCGTAAAAAG GCCGCGTTGC TGGCGTTTTT CCATAGGCTC CGCCCCCCTG





4741
ACGAGCATCA CAAAAATCGA CGCTCAAGTC AGAGGTGGCG AAACCCGACA GGACTATAAA





4801
GATACCAGGC GTTTCCCCCT GGAAGCTCCC TCGTGCGCTC TCCTGTTCCG ACCCTGCCGC





4861
TTACCGGATA CCTGTCCGCC TTTCTCCCTT CGGGAAGCGT GGCGCTTTCT CATAGCTCAC





4921
GCTGTAGGTA TCTCAGTTCG GTGTAGGTCG TTCGCTCCAA GCTGGGCTGT GTGCACGAAC





4981
CCCCCGTTCA GCCCGACCGC TGCGCCTTAT CCGGTAACTA TCGTCTTGAG TCCAACCCGG





5041
TAAGACACGA CTTATCGCCA CTGGCAGCAG CCACTGGTAA CAGGATTAGC AGAGCGAGGT





5101
ATGTAGGCGG TGCTACAGAG TTCTTGAAGT GGTGGCCTAA CTACGGCTAC ACTAGAAGAA





5161
CAGTATTTGG TATCTGCGCT CTGCTGAAGC CAGTTACCTT CGGAAAAAGA GTTGGTAGCT





5221
CTTGATCCGG CAAACAAACC ACCGCTGGTA GCGGTGGTTT TTTTGTTTGC AAGCAGCAGA





5281
TTACGCGCAG AAAAAAAGGA TCTCAAGAAG ATCCTTTGAT CTTTTCTACG GGGTCTGACG





5341
CTCAGTGGAA CGAAAACTCA CGTTAAGGGA TTTTGGTCAT GAGATTATCA AAAAGGATCT





5401
TCACCTAGAT CCTTTTAAAT TAAAAATGAA GTTTTAAATC AATCTAAAGT ATATATGAGT





5461
AAACTTGGTC TGACAGTTAC CAATGCTTAA TCAGTGAGGC ACCTATCTCA GCGATCTGTC





5521
TATTTCGTTC ATCCATAGTT GCCTGACTCC CCGTCGTGTA GATAACTACG ATACGGGAGG





5581
GCTTACCATC TGGCCCCAGT GCTGCAATGA TACCGCGAGA CCCACGCTCA CCGGCTCCAG





5641
ATTTATCAGC AATAAACCAG CCAGCCGGAA GGGCCGAGCG CAGAAGTGGT CCTGCAACTT





5701
TATCCGCCTC CATCCAGTCT ATTAATTGTT GCCGGGAAGC TAGAGTAAGT AGTTCGCCAG





5761
TTAATAGTTT GCGCAACGTT GTTGCCATTG CTACAGGCAT CGTGGTGTCA CGCTCGTCGT





5821
TTGGTATGGC TTCATTCAGC TCCGGTTCCC AACGATCAAG GCGAGTTACA TGATCCCCCA





5881
TGTTGTGCAA AAAAGCGGTT AGCTCCTTCG GTCCTCCGAT CGTTGTCAGA AGTAAGTTGG





5941
CCGCAGTGTT ATCACTCATG GTTATGGCAG CACTGCATAA TTCTCTTACT GTCATGCCAT





6001
CCGTAAGATG CTTTTCTGTG ACTGGTGAGT ACTCAACCAA GTCATTCTGA GAATAGTGTA





6061
TGCGGCGACC GAGTTGCTCT TGCCCGGCGT CAATACGGGA TAATACCGCG CCACATAGCA





6121
GAACTTTAAA AGTGCTCATC ATTGGAAAAC GTTCTTCGGG GCGAAAACTC TCAAGGATCT





6181
TACCGCTGTT GAGATCCAGT TCGATGTAAC CCACTCGTGC ACCCAACTGA TCTTCAGCAT





6241
CTTTTACTTT CACCAGCGTT TCTGGGTGAG CAAAAACAGG AAGGCAAAAT GCCGCAAAAA





6301
AGGGAATAAG GGCGACACGG AAATGTTGAA TACTCATACT CTTCCTTTTT CAATATTATT





6361
GAAGCATTTA TCAGGGTTAT TGTCTCATGA GCGGATACAT ATTTGAATGT ATTTAGAAAA





6421
ATAAACAAAT AGGGGTTCCG CGCACATTTC CCCGAAAAGT GCCACCTGAC GTC (SEQ ID NO: 503)










Annotations:








Region/ Element
Base range





CMV enhancer
 235-614





CMV promoter
 615-818





T7 RNA polymerase promoter
 863-880





iCre coding sequence
 964-2019





bGHpA terminator sequence
2070-2293





Origin of replication
2962-3097





SV40 polyA sequence
4146-4267





Lac operon operator (lacO)
4340-4356





Lac operon promoter (complement)
4364-4394





C-tag
4372-4383





Catabolite activator protein binding site
4409-4430





Origin of replication (complement)
4718-5306





Ampicillin resistance gene promoter
6338-6442


(complement)









Anellovirus Vector Production: Cell Lysis

Cell pellets were resuspended in lysis buffer containing 50 mM Tris pH 8.0, 0.5% Triton-X100, 100 mM NaCl, 100× Halt protease inhibitor cocktail (Thermo Fisher Scientific catalog #78439), and 200 U of mSAN nuclease (ArcticZyme catolog #NC1920045). The cell lysates were clarified by centrifugation at 12,500×g for 30 minutes at 4° C.


Anellovirus Vector Production: Isopycnic Centrifugation

To prepare iodixanol linear gradients, 13 mL of 60% OptiPrep (Sigma-Aldrich catalog #D1556) was overlayed with 13 mL of 20% OptiPrep in 26.3-mL polycarbonate tubes, which were then spun at a 46-degree angle and a speed of 20 rpm for 16 minutes using Gradient Master (BioComp). Following the generation of the iodixanol linear gradient, 2 mL of iodixanol were removed from the top of each gradient, and 2 mL of clarified lysate was added on top of the gradient. The sample-containing tubes were spun at 347,000×g and 20° C. for 3 hours using Type 70 Ti rotor (Beckman Coulter). 1-mL fractions were collected from the top of the tubes and transferred to a 96-well 2.2 ml capacity plate. Each fraction was then subjected to DNase-protected qPCR assay as described below.


Anellovirus Vector Production: Concentration of Material

Fractions of interest were determined based on the viral titer (FIG. 2A and FIG. 2B). The pooled iodixanol fractions were then diluted 1:50 in formulation buffer (1×DPBS, 0.001% Pluronic F-68) and concentrated using Vivaspin 20 100K MWCO centrifugal filter units (Fisher Scientific catalog #1455810).


Anellovirus Vector Production: DNase-Protection Assay on Final Material

5 μl of the sample to be titered was incubated with 20 U of DNAse I endonuclease (Thermo Fisher Scientific catalog #18047019) in a 20-μl reaction. The reaction was incubated at 37° C. for 30 minutes. Following DNase-treatment, each sample was subjected to Proteinase K (Fisher Scientific catalog #FERE00491) and proteinase K buffer (1% SDS, 0.1M EDTA, 0.1M Tris pH 8.0, 0.1% Pluronic F-68). The reaction was incubated at 37° C. for 30 minutes, followed by proteinase K inactivation at 95° C. for 15 minutes. 4 μl of the 1:10 diluted DNase reaction was subjected to qPCR analysis in a 20-μl reaction using TaqMan Fast Universal PCR Master Mix (Thermo Fisher Scientific catalog #44-449-63) according to the manufacturer's protocol (FIG. 3A and FIG. 3B). Primer and probe sequences are listed in Table E1.









TABLE E1







Primer and probes designed to quantify eGFP









Target
Label
Sequence (5′→3′)





eGFP
Forward Primer
GAACCGCATCGAGCTGAA




(SEQ ID NO: 1115)



Reverse Primer
TGCTTGTCGGCCATGATATAG




(SEQ ID NO: 1116)



Probe (FAM)
ATCGACTTCAAGGAGGACGGCAAC




(SEQ ID NO: 1117)









Anellovirus Vector Production: Endotoxin Test on Final Material

2.241 of the sample was diluted 1:50 in formulation buffer (1×DPBS, 0.001% Pluronic F-68) and the sample was subjected to the LAL detection test (Charles River) according to the manufacturer's protocol.


Anellovirus Vector Production: SDS-PAGE and Coomassie Stain on Final Material

2 μl of the sample was diluted 1:10 in formulation buffer (1×DPBS, 0.001% Pluronic F-68) and the sample was mixed 1:1 with loading dye and Bolt sample reducing agent (Thermo Fisher Scientific catalog #B0009), followed by boiling at 95° C. for 10 minutes. Proteins were separated on Bolt 4-12% Bis-Tris gel in 1×Bolt MOPS SDS running buffer (Thermo Fisher Scientific catalog #B0001). Separated proteins were stained using InstantBlue Coomassie Protein Stain (ABCAM catalog #ab119211) according to the manufacturers protocol. Following the stain, the gel was washed three times with diH2O. The stained gel was visualized using Chemidoc Imaging System (BioRad) (FIGS. 4A and 4B).


Anellovirus Vector Production: Bicinchonic Acid Assay (BCA)

5 μl of the sample was diluted 1:10 in formulation buffer (1×DPBS, 0.001% Pluronic F-68) and the sample was subjected to the Pierce BCA Protein Assay Kit (ThermoScientific catalogue #23227) according to the manufacturer's protocol.


Example 2: In Vivo Transduction of the Brain Via Intracerebroventricular (ICV) Administration with an Anellovector (Study #1)

This example demonstrates that an Anellovector transduces the brain in vivo via ICV administration.


Materials and Methods

The production of the AAV9-fCMV-eGFP control and Ring19-fCMV-eGFP Anellovector used in this Study #1 are described in Example 1.


Care and Use of Animals

All mouse studies were approved and governed by the Ring Therapeutics Institutional Animal Care and Use Committee. Female FVB/NJ 7-8 weeks of age were obtained from Jackson Laboratories for use in these studies.


Intracerebroventricular Injections

Mice were anesthetized with 2%-3% isoflurane and placed with a heat pad on the stereotaxic frame (Neurotar GmbH). Eye ointment was applied to both eyes. After applying aseptic betadine and 70% alcohol, an incision was made to expose the skull. A craniotomy was made by a micro-driller. A 5-10 μL Hamilton syringe was slowly moved to the target coordinates (lateral ventricle, AP+0.25 mm, ML, ±0.7 mm, DV, −2 mm. 0.5-3 μL of test article was injected with the speed approximately 0.02 μL/min. Test articles were injected bilaterally or unilaterally into the specific brain regions using established mouse brain coordinates. The incision was sutured with absorbable sutures (Suture CT-13.0 coated Vicryl 27”) or by applying surgical glue on skin incision or sutures. Each animal received ICV injection once and SC injection of buprenorphine once after the ICV injection for analgesic purpose during the week of the procedure.


Tissue Harvest for DNA and RNA Extraction

Brains were dissected out and then cut in half in sagittal orientation. Half of the brain was used for DNA processing and the other half of the brain from the same animal was used for RNA processing. Brains were frozen and stored in 2 ml reinforced bead homogenizer tubes for both DNA and RNA extraction. Spinal cord samples were also collected and stored in All-prep reagent for All-prep DNA and RNA extraction (Qiagen).


DNA Isolation

Frozen tissue samples were lysed with an automated tissue homogenizer (Geno/Grinder SPEX Sample Prep) in Buffer ATL (Qiagen, USA) and proteinase K (Qiagen, USA) at 1250 rpm for (2) 30-second rounds. Homogenized tissues were digested on heat block at 56 C for about 4 hours. Genomic DNA was precipitated with Buffer AL (Qiagen, USA) and ethanol, then isolated with Qiagen DNeasy 96 Blood & Tissue Kit. Isolated DNA was quantified using a NanoDrop 8000 Spectrophotometer (Thermofisher, USA).


qPCR


Genomic DNA was assayed by qPCR on the QuantStudio 5—Real-Time PCR System (Thermo Fisher, USA) using TaqMan Gene Expression Master Mix (Thermofisher, USA). The sequence detection primers and FAM custom probes that were used in this study were synthesized by Integrated DNA Technologies, USA. eGFP primer/probe sequences are in Table EL.


All reactions including the DNA samples and different dilutions of a known quantity of the linearized eGFP and Ring19 plasmid standards were run in triplicate on the same plate. The standard curve method was used to calculate the amount of viral/vector DNA, which was normalized with the total amount of genomic DNA for each sample (quantified using nanodrop as described above).


RNA Isolation

Frozen tissue samples were lysed with an automated tissue homogenizer (Geno/Grinder SPEX Sample Prep, USA) in QIAzol lysis reagent (Qiagen, USA) at 1250 rpm for (2) 30-second rounds. RNA was isolated in aqueous phase by addition of phenol chloroform (Thermofisher, USA) and centrifuged at 6000 rpm for 15 minutes at 4 C. The upper aqueous phase was transferred into a fresh S-block (Qiagen, USA). RNA was then precipitated with the addition of 1 volume of 70% Ethanol and isolated with the Qiagen RNeasy 96 kit. RNA concentration was quantified via the Qubit RNA High Sensitivity Assay Kit (Thermofisher, USA)


All-Prep DNA and RNA Extraction

To extract DNA and RNA from the spinal cord samples, 10 uL BME per 1 mL Buffer RLT Plus were added to the sample. Tissue was homogenized with 350 uL RLT Plus/BME. Spex tubes were spun down and lysates were transferred into clean Eppendorf tubes. Lysate were spun at 6000 rpm for 4 minutes at RT. 150 μL of supernatant was transferred into an All-Prep DNA Mini Column (Qiagen) and the remaining supernatant lysate was transferred into a freshly labelled Eppendorf for long-term storage at −80 C. DNA columns were spun at max speed for 30 seconds. DNA columns were then placed into new collection tubes and stored at 4 C for later DNA purification. 14 uL Proteinase K was added to the flowthrough from the last step. 58 uL 100% ethanol was added to flow through and samples were mixed well and incubated at RT for 10 min. 115 μL of 100% ethanol was added and mixed well. All contents were transferred into RNeasy Mini Spin columns. After centrifugation, 500 uL Buffer RPE was added to the RNeasy Mini Spin Column. 80 μL of DNase I was transferred and mixed into the RNeasy column with incubation at RT for 15 min. FRN, buffer RPE, and 100% ethanol were added in order to wash the columns. 30 uL RNase-free water was added directly to the column and incubated for 2-5 minutes at RT. Samples were spun for 1 minute at max speed to elute the RNA.


One-Step RT-ddPCR

RNA was diluted in nuclease-free water and combined with the reagents from the One-Step RT-ddPCR Advanced Kit for Probes (Bio-Rad, USA; Catalog #1864022) and eGFP primer/probe set with final primer concentrations of 900 nM and probe concentrations of 250 nM to measure transgene expression. After the RT-ddPCR reaction setup, each reaction was converted to droplets using the Automated Droplet Generator (Bio-Rad, USA) according to the manufacturer's instructions. After droplet generation, the droplets were subjected to endpoint PCR thermocycling with the following cycling conditions: 1 cycle of 48 C for 1 hour for reverse transcription followed by 1 cycle of 95 C for 10 mins; 40 cycles of 95 C for 30 sec, 60 C for 1 min; and 1 cycle of 98 C for 10 min and finally a 4 C hold. The cycled plate was then transferred to the QX200 Droplet Reader (Bio-Rad, USA) and analyzed using QX Manager Software (Bio-Rad, USA).


Tissue Harvest for Immunohistochemistry

Brains were collected in 4% PFA and fixed for 24 hours. Brain samples were changed from PFA to 30% sucrose and stayed in sucrose for at least 2 nights until the tissue sunk to the bottom. Brains were embedded in OCT compound for sectioning. Brains were sectioned at 30 m in a Leica cryostat and mounted immediately on poly-L-Lysine coated slides. Sections were stored at −80° C. prior to processing.


Immunohistochemistry

Slides were air-dried for 20 minutes and then washed with 1×PBS for 5 minutes three times. Slides were incubated at room temperature with 10% goat serum in TBST for 2 hours followed by the primary antibody at 1:500 (GFP recombinant rabbit monoclonal antibody, ThermoFisher G10362, TBST with 10% goat serum) for 48 hours at 4 C. Slides were washed with PBS for 5 minutes three times and incubated in 1:2000 secondary antibody (goat anti-rabbit IgG (H+L) cross-absorbed secondary antibody Alexa-fluor 488 (ThermoFisher A11008) TBST with 10% goat serum) for 2 hours at room temperature, avoiding light. Slides were washed with PBS for 5 minutes three times. DAPI mounting media (ThermoFisher P36966) was added to the slides and covered with a coverslip. 2× images were captured with an EVOS (M7000, Invitrogen by ThermoFisher Scientific), and 20× images were captured with a confocal microscope (Zeiss LSM900).


Results
Infectivity of Anellovector R19-fCMV-eGFP (“R19-eGFP”) in the Brain

The prepared virus preparations were administered to mice by intracerebroventricular (ICV) injection as shown in Table E2. Mice were injected with PBS, R19-eGFP, or dose-matched AAV9-eGFP. 21 days after injection, the brain and spinal cord were collected and processed.









TABLE E2







Study design #1 for ICV administration.













Treatment
Dose/eye
Route
N
Terminal


Group
Day 0
(vg)
(volume)
(mice)
Day





1
PBS
0
ICV (2 × 2 ul)
7
21


2
R19-fCMV-eGFP (“R19-eGFP”)
3.6e+8
ICV (2 × 2 ul)
7
21


3
AAV9-fCMV-eGFP (“AAV9-eGFP”)
3.6e+8
ICV (2 × 2 ul)
7
21









DNA was collected from the left brain hemisphere. eGFP genomes were detected by qPCR in the brain transduced with R19-eGFP Anellovector and AAV9-eGFP, whereas no eGFP genomes were detected in the PBS control group (FIG. 5A). In contrast, the level of eGFP genomes were below the limit of detection by qPCR in the spinal cord in the PBS, R19-eGFP Anellovector, and AAV9-eGFP groups at the doses administered (FIG. 5B).


Administration of Ring19-eGFP Anellovector by ICV Injection Induces eGFP mRNA Expression in the Brain


To determine whether administration of R19-eGFP Anellovector can induce eGFP mRNA expression, RNA was collected from the right brain hemisphere at 21 days after infection (n=5 brain hemispheres per group). eGFP mRNA was then quantified by RT-ddPCR. eGFP mRNA was detected in the brain 21 days after infection with R19-eGFP and AAV9-eGFP, while no eGFP mRNA was detected in the PBS control group (FIG. 6).


Administration of R19-eGFP Anellovector by ICV Injection Induces eGFP Protein Expression in the Brain


To determine whether administration of R19-eGFP Anellovector can produce eGFP protein in vivo in the brain, brain tissue was collected at day 21 after infection for fixation and immunohistochemistry (n=2 brains per group). Representative brain sections of ICV-injected mice were stained for eGFP. FIGS. 7A, 7B, and 7C show images of brain sections stained for eGFP from the PBS control, R19-eGFP, and AAV9-eGFP groups, respectively, at 2× magnification. The area in the red squares was magnified to 20× (FIGS. 7D, 7E, and 7F). While the PBS control showed no eGFP staining (FIG. 7D), eGFP-positive neurons were detected in the hippocampus of both R19-eGFP (FIG. 7E) and AAV9-eGFP (FIG. 7F)-treated brains. These data show that administration of a R19-eGFP Anellovector resulted in expression of eGFP protein in the brain.


Example 3: In Vivo Delivery of an Anellovector to the Spinal Cord Via Intrathecal (IT) Administration (Study #2)

This example describes the in vivo delivery of a Ring19-eGFP Anellovector to the spinal cord.


Materials and Methods

The production of the AAV9-fCMV-eGFP control and Ring19-fCMV-eGFP Anellovector used in this Study #2 are described in Example 1.


All other methods are according to Example 2 above, except that intrathecal injections were performed as described below.


Intrathecal Injections

The mice were anesthetized with 3% isoflurane, until they showed no signs of righting reflex. In addition, tail and/or paw pinch reflex were checked to further ensure the state of anesthesia. The posterior end of the animal, near the base of the tail, was shaved in an area around 2 cm2 to facilitate better visualization during needle insertion. The mouse was placed in a nose cone for continued isoflurane administration during the procedure. During the procedure, the isoflurane was reduced to 1.5% and the eyes of the mouse were covered with eye lubricant to prevent eye damage. A 10-50 μl Hamilton with a 26 gauge needle was used to locate the L5-L6 region of the spinal cord. The needle was inserted between the groove of the L5 and L6 vertebrae. A tail flick indicated successful entry of the needle into the intradural space. Once a tail flick was observed, immediately, but carefully, the needle position was secured with one hand and the desired volume of the substance was injected with the other hand slowly. No more than 5 μl was injected per mouse. Once the injection was performed, the mouse was moved back to the cage to recover from anesthesia. Each animal received once weekly IT injections for 2 weeks followed by one time subcutaneous injection of an analgesic drug during the procedure week as needed.


The virus preparations were administered into mice by intrathecal (IT) injection as shown in Table E3. Mice were injected with PBS, R19-eGFP, or dose-matched AAV9-eGFP. 21 days after injection, the spinal cords and brain were collected and processed.









TABLE E3







Study design #2 for IT administration.













Treatment
Dose/eye
Route
N
Terminal


Group
Day 0
(vg)
(volume)
(mice)
Day





1
PBS
0
IT (50 uL)
7
21


2
R19-fCMV-eGFP
9.6e+9
IT (50 uL)
7
21


3
AAV9-fCMV-eGFP
9.6e+9
IT (50 uL)
7
21









Results

DNA was collected from the spinal cord and the brain as described in Example 2 and eGFP genomes were detected by qPCR in the spinal cord (FIG. 8A) and in the brain (FIG. 8B). eGFP genomes were transduced with R19-eGFP Anellovector at similar levels as the dose-matched AAV9-eGFP, while no eGFP genomes were detected in the PBS control.


Example 4: In Vivo Delivery of an Anellovector to the Spinal Cord and Redosing Via IT Administration

This example describes the in vivo redosing of a Ring19-eGFP Anellovector to the spinal cord.


Materials and Methods

The AAV9-fCMV-eGFP control was produced as described above.


The R19-fCMV-eGFP anellovector was produced by transfecting MOLT-4 cells with the three plasmids shown in Table E0 (pRTx-3525; pRTx-2847; pRTx-2848) via electroporation. The transfected cells were harvested 72 hours post-electroporation and centrifuged. The supernatant was discarded, and the pellet was frozen at −80°. The cells were thawed and resuspended in buffer. The lysate was initially clarified by centrifugation, then sterile filtered. The clarified harvest pool was buffer exchanged using tangential flow filtration (TFF). After buffer exchange, the pool was sterile filtered.


The buffer-exchanged clarified harvest pool was divided into 2 equal volume pools to perform two chromatography cycles on a CIMultus DEAE Monolith (Sartorius AG). The monolith was equilibrated and the first pool was loaded onto the monolith. After elution of the vector, the column was stripped and the chromatography process was then repeated with the second half of the harvest pool, and each resulting elution was pooled for further processing.


Triton phase separation was performed on the above elution pool to remove endotoxin from the pool using a Triton buffer. The pool was centrifuged and the top aqueous phase was collected from each tube via pipetting and pooled.


The post-triton phase separation pool was further purified via heparin affinity chromatography. Vector was eluted and the resulting 50 mL eluate pool was collected for further processing.


The heparin eluate pool was loaded onto a 100 kDa TFF cartridge (Formulatrix) and buffer exchange was performed. A final 2.5× concentration was then performed to a 4 mL final volume. The buffer exchanged TFF pool was sterile filtered and aliquoted for storage at −80° C.


Experimental Design

PBS control, AAV9-fCMV-eGFP (“AAV9-eGFP” or “AAV-eGFP”), and Ring19-fCMV-eGFP (“Ring19-eGFP”) were administered into FVB/NJ mice by intrathecal (IT) injection in accordance with Table E4. DNA, RNA, and immunohistochemistry for eGFP were analyzed as described in Example 2.









TABLE E4







Study Design















Treatment
Treatment
Dose Vector
Dose/animal

N
Terminal


Group
Day 0
Day 21
(vg/ml)
(vg)
Route
(animals)
Day





1
PBS
Takedown
0
0
IT
5
21







(50 ul)




2
PBS
NA
0
0
IT
5
42







(50 ul)




3
PBS
PBS
0/0
0/0
IT
5
42







(50 ul)




4
RING19-fCMV-
Takedown
1.55E+10
7.75E+8
IT
5
21



eGFP



(50 ul)




5
RING19-fCMV-
NA
1.55E+10
7.75E+8
IT
5
42



eGFP



(50 ul)




6
RING19-fCMV-
RING19-fCMV-
2.4E+10/2.4E+10
7.75E+8/7.75E+8
IT
5
42



eGFP
eGFP


(50 ul)




7
AAV9-fCMV-
Takedown
2.4E+10
1.2E+9
IT
5
21



eGFP



(50 ul)




8
AAV9-fCMV-
NA
2.4E+10
1.2E+9
IT
5
42



eGFP



(50 ul)




9
AAV9-fCMV-
AAV9-fCMV-
2.4E+10/2.4E+10
1.2E+9/1.2E+9
IT
5
42



eGFP
eGFP


(50 ul)









Results

DNA was collected from the spinal cord, brain, liver, and muscle on either day 21 or day 42 after treatment as outlined in Table E4 and eGFP genomes were detected by qPCR. As shown in FIG. 9A, no eGFP genomes were detected in the spinal cord of any of the animals that received PBS controls, while eGFP genomes were detected at day 21 (“R19-eGFP-D21”) and day 42 (“R19-eGFP×1 D42”) in the spinal cords in animals administered with a single dose of Ring19-eGFP anellovector, with an increase in genome levels detected on day 42. In the animals that received a second dose of Ring19-eGFP 21 days after the initial treatment with Ring19-eGFP (“R19-eGFP×2 D42”), the eGFP genome levels were greater in the spinal cord than in the animals that received only a single dose. While the animals received a slightly lower dose of Ring19-eGFP anellovector compared to AAV9-eGFP, the eGFP genome levels were higher in the spinal cords of animals on day 42 that received a single dose of R19-eGFP anellovector (“R19-eGFP×1 D42”) and animals that received two doses of Ring19-eGFP (“R19-eGFP×2 D42”) compared to animals that received a single dose of AAV9-eGP (“AAV9-eGFP×1 D42”) and animals that received two doses of AAV9-eGFP (“AAV9 eGFP×2 D42”).


As shown in FIG. 9B, no eGFP genomes were detected in the brain in any of the animals that received PBS controls or in the brain of the animals at day 21 (“R19-eGFP-D21”) and day 42 (“R19-eGFP×1 D42”) administered with a single dose of Ring19-eGFP anellovector. However, some eGFP genomes were detected in the brain of one animal that received a second dose of Ring19-eGFP 21 days after the initial treatment with Ring19-eGFP (“R19-eGFP×2 D42”), similar to eGFP genome levels in animals that received a second dose of AAV9-eGFP (“AAV-eGFP×2 D42”).


As shown in FIG. 9C, no eGFP genomes were detected in the liver of any of the animals that received PBS controls. eGFP genomes were detected in the liver at day 21 (“R19-eGFP-D21”) and day 42 (“R19-eGFP×1 D42”) in the liver of animals administered with a single dose of Ring19-eGFP anellovector. The level of eGFP genomes detected in the liver in the animals that received a second dose of Ring19-eGFP 21 days after the initial treatment with Ring19-eGFP (“R19-eGFP×2 D42”) was higher than in the animals that received a single dose of Ring19-eGFP anellovector.


As shown in FIG. 9D, no eGFP genomes were detected in the muscle of any of the animals that received PBS controls or in the muscle of the animals at day 21 (“R19-eGFP-D21”) and day 42 (“R19-eGFP×1 D42”) administered with a single dose of Ring9-eGFP anellovector. However, some eGFP genomes were detected in the muscle of animals that received a second dose of Ring19-eGFP 21 days after the initial treatment with Ring19-eGFP (“R19-eGFP×2 D42”). While eGFP genomes were detected in the muscle of animals that received a single dose (“AAV9-eGFP-D21” and “AAV-eGFP×1 D42”) of AAv9-eGFP, the eGFP genome levels did not increase with administration of a second dose of AAV9-eGFP (“AAV-eGFP×2 D42”).


RNA was collected from the spinal cord and liver on either day 21 or day 42 after treatment for eGFP mRNA detection by RT-ddPCR. As shown in FIG. 10A, the level of eGFP mRNA was below the limit of detection in both the PBS controls and in the spinal cord of mice treated with Ring19-eGFP at the doses administered, while eGFP mRNA was detected in the spinal cord of mice treated with AAV9-eGFP. Similarly, as shown in FIG. 10B, the level of eGFP mRNA was below the limit of detection in both the PBS controls and in the liver of mice treated with Ring19-eGFP, while eGFP mRNA was detected in the liver of mice treated with AAV9-eGFP.


RNA was collected from the brain and muscle from mice in Groups 4 and 7 on day 21 after treatment for eGFP mRNA detection by RT-ddPCR. As shown in FIG. 10C, eGFP mRNA was below the limit of detection in both the PBS controls and in the brain of mice treated with Ring19-eGFP at the doses administered, while eGFP mRNA was detected in the brain of mice treated with AAV9-eGFP. Similarly, as shown in FIG. 10D, eGFP mRNA was below the limit of detection in both PBS controls and in the muscle of mice treated with Ring19-eGFP at the doses administered, while eGFP mRNA was detected in the muscle of mice treated with AAV9-eGFP.


The spinal cord was also collected at day 21 or day 42 after treatment for fixation and immunohistochemistry. Representative spinal cord sections of treated mice were stained for eGFP. FIGS. 11A, 11B, and 11C show images of spinal cord sections stained for eGFP from the PBS control (Groups 1, 2, and 3, respectively) at 20× magnification. FIGS. 11D, 11E, and 11F show images of spinal cord stained for eGFP from mice treated with Ring19-eGRP in Groups 4, 5, and 6, respectively, at 20× magnification. FIGS. 11G, 11H, and 11I show images of spinal cord stained for eGFP from mice treated with AAV9-eGFP in Groups 7, 8, and 9, respectively, at 20× magnification. The area in the red square in FIG. 11J (2× magnification) shows the area that was magnified to 20× in FIG. 11I. No eGFP staining was detected in any of the PBS controls (FIGS. 11A-C). eGFP staining was not detected in the Ring19-eGFP-treated mice after a single dose at the doses administered (FIGS. 11D and 11E). eGFP protein was detected in the spinal cord of AAV9-eGFP-treated mice (FIGS. 11G-11I).


Example 5: In Vivo Delivery of an Anellovector to Brain and Redosing Via ICV Administration

This example describes the in vivo redosing of a Ring19-eGFP Anellovector to the brain.


Materials and Methods

AAV9-fCMV-eGFP is an adeno-associated virus based on the plasmid pRTx-2770 as described above and is packaged into AAV9. Ring19-fCMV-eGFP is produced by transfecting MOLT-4 cells with the three plasmids (pRTx-3525; pRTx-2847; pRTx-2848) shown in Table E0.


PBS control, AAV9-fCMV-eGFP (“AAV9-eGFP”), and Ring19-fCMV-eGFP (“Ring19-eGFP”) are administered into FVB/NJ mice by intracerebroventricular (ICV) injection in accordance with Table E5. DNA, RNA, and immunohistochemistry for eGFP in the brain are analyzed as similarly described in Example 2.









TABLE E5







Study Design













Group
Treatment Day 0
Treatment Day 21
Dose/animal (vg)
Route
N (animals)
Terminal Day
















1
PBS
Takedown
0
ICV
~6
21


2
PBS
NA
0
ICV
~5
42


3
PBS
PBS
0/0
ICV
~5
42


4
RING19-fCMV-eGFP
Takedown
E7-E10
ICV
~6
21


5
RING19-fCMV-eGFP
NA
E7-E10
ICV
~5
42


6
RING19-fCMV-eGFP
RING19-fCMV-eGFP
E7-E10/E7-E10
ICV
~5
42


7
AAV9-fCMV-eGFP
Takedown
E7-E10
ICV
~6
21


8
AAV9-fCMV-eGFP
NA
E7-E10
ICV
~5
42


9
AAV9-fCMV-eGFP
AAV9-fCMV-eGFP
E7-E10/E7-E10
ICV
~5
42









In accordance with the schedule shown in Table E5, on day 21 or 42 after initial administration, the animals are taken down and DNA and RNA from the brains are collected and processed. qPCR is used to detect eGFP genomes in the DNA samples and RT-ddPCR is used to detect eGFP RNA in the RNA samples. Immunohistochemistry may also be performed to detect eGFP protein.

Claims
  • 1. A method of delivering an exogenous effector to the central nervous system (CNS) of a subject, the method comprising administering to the CNS of the subject an Anelloviridae family vector.
  • 2. (canceled)
  • 3. The method of claim 1, which comprises delivery of the Anelloviridae family vector to the brain of the subject.
  • 4. The method of claim 1, which comprises production of the exogenous effector and/or DNA capable of encoding the exogenous effector in the brain of the subject.
  • 5.-7. (canceled)
  • 8. A method of treating a CNS disease or disorder in a subject in need thereof, the method comprising administering to the subject an Anelloviridae family vector.
  • 9.-11. (canceled)
  • 12. The method of claim 1, which results in delivery of anellovector DNA in one or more cell types selected from: neurons, glial cells, microglia, oligodendroglia, and astrocytes.
  • 13.-14. (canceled)
  • 15. The method of claim 1, which results in delivery of anellovector DNA in one or more of: cerebellum, frontotemporal lobe, parietal lobe, brain stem, and spinal cord.
  • 16.-17. (canceled)
  • 18. The method of claim 1, which results in greater delivery of the exogenous effector and/or the DNA encoding the exogenous effector to the brain than to the spinal cord.
  • 19. The method of claim 1, wherein the Anelloviridae family vector is administered according to a route of administration chosen from: intrathecal (IT), intracerebroventricular (ICV), intra cisterna magna (ICM), or intraparenchymal (IPa).
  • 20. (canceled)
  • 21. The method of claim 1, wherein the Anelloviridae family vector is an anellovector comprising a proteinaceous exterior comprising an ORF1 molecule and a genetic element enclosed by the proteinaceous exterior, wherein the genetic element comprises a promoter element operably linked to a nucleic acid sequence encoding the exogenous effector.
  • 22. The method of claim 21, wherein the ORF1 molecule: (i) has an ORF1 sequence as listed in any of Tables A1-A3, or a polypeptide comprising an amino acid sequence having at least about 70% sequence identity thereto; and/or(ii) comprises a polypeptide encoded by an Anellovirus ORF1 nucleic acid sequence as listed in Table N1-N3, or a polypeptide encoded by a nucleic acid sequence having at least about 70% sequence identity to the Anellovirus ORF1 nucleic acid sequence.
  • 23. The method of claim 22, wherein the ORF1 molecule comprises at least one difference relative to the wild-type ORF1 protein of Table A1-A3.
  • 24.-27. (canceled)
  • 28. The method of claim 1, wherein the genetic element comprises; (A) (i) an Anellovirus 5′ UTR conserved domain having a sequence of the reverse complement of nucleotides 323-393 of SEQ ID NO: 54, or a nucleic acid sequence having at least 70% sequence identity thereto, or a functional portion thereof; and/or(ii) an Anellovirus GC-rich region having a sequence of the reverse complement of nucleotides 2868-2929 of SEQ ID NO: 54, or a nucleic acid sequence having at least 70% sequence identity thereto, or a functional portion thereof; or(B) (i) an Anellovirus 5′ UTR conserved domain having a sequence of the reverse complement of nucleotides 1-71 of SEQ ID NO: 1, or a nucleic acid sequence having at least 70% sequence identity thereto, or a functional portion thereof; and/or(ii) an Anellovirus GC-rich region having a sequence of the reverse complement of nucleotides 2515-2615 of SEQ ID NO: 1, or a nucleic acid sequence having at least 70% sequence identity thereto, or a functional portion thereof.
  • 29.-32. (canceled)
  • 33. The method of claim 1, wherein the genetic element is: (i) DNA;(ii) circular, single stranded DNA; or(iii) mRNA.
  • 34.-35. (canceled)
  • 36. The method of claim 1, wherein the exogenous effector comprises: an intracellular peptide or intracellular polypeptide, a secreted polypeptide, or a protein replacement therapeutic.
  • 37. The method of claim 1, wherein the Anelloviridae family vector is a Betatorquevirus.
  • 38. The method of claim 1, which further comprises administering an additional dose of an Anelloviridae family vector to the CNS of the subject.
  • 39. The method of claim 38, wherein the Anelloviridae family vector and the Anelloviridae family vector of the additional dose are the same.
  • 40. (canceled)
  • 41. The method of claim 1, which results in greater delivery of the DNA encoding the exogenous effector to: (i) the CNS than to muscle;(ii) the CNS than to liver;(iii) the spinal cord than to muscle; and/or(iv) the spinal cord than to liver.
  • 42.-44. (canceled)
  • 45. The method of claim 41, wherein the greater delivery is 2 times, 5 times, or 10 times higher.
  • 46. A delivery system suitable for delivery to the CNS, wherein the delivery system comprises an Anelloviridae family vector.
CROSS REFERENCE TO RELATED APPLICATIONS

This application claims the benefit of U.S. Provisional Application No. 63/508,149, filed Jun. 14, 2023. The contents of the aforementioned application are hereby incorporated by reference in their entirety.

Provisional Applications (1)
Number Date Country
63508149 Jun 2023 US