WO2024102739A2 - Adeno-associated virus (aav) production - Google Patents

Adeno-associated virus (aav) production Download PDF

Info

Publication number
WO2024102739A2
WO2024102739A2 PCT/US2023/078957 US2023078957W WO2024102739A2 WO 2024102739 A2 WO2024102739 A2 WO 2024102739A2 US 2023078957 W US2023078957 W US 2023078957W WO 2024102739 A2 WO2024102739 A2 WO 2024102739A2
Authority
WO
WIPO (PCT)
Prior art keywords
seq
mutations
enzyme
identity
nucleotide sequence
Prior art date
Application number
PCT/US2023/078957
Other languages
French (fr)
Inventor
Joseph J. HIGGINS
Ray TABIBIZAR
Feng Yao
Quan Karen ZHU
Original Assignee
Saliogen Therapeutics, Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Saliogen Therapeutics, Inc. filed Critical Saliogen Therapeutics, Inc.
Publication of WO2024102739A2 publication Critical patent/WO2024102739A2/en

Links

Definitions

  • the present invention relates, in part, to a method of making a viral particle packaging and producer stable cell line using transposon(s) encoding genes required for AAV production and an enzyme capable of performing targeted genomic integration.
  • CROSS-REFERENCE TO RELATED APPLICATIONS This application claims the benefit of priority to U.S. Provisional Patent Application No.63/432,220, filed November 7, 2022, which is incorporated by reference herein in its entirety.
  • SEQUENCE LISTING The instant application contains a Sequence Listing that has been submitted electronically in XML file format and is hereby incorporated by reference in its entirety.
  • AAV Adeno-associated virus
  • the genome includes three open reading frames (ORFs) encoding for replication (non-structural) proteins (rep), capsid (structural) proteins (cap), and the assembly activating protein (AAP). These three genes give rise to at least nine gene products through the use of three promoters, alternative translation start sites, and differential splicing. See Naso et al., BioDrugs.2017;31(4):317-334.
  • the coding sequences for the rep, cap, and AAP genes are flanked by inverted terminal repeats (ITRs). The 145-nt ITRs are partially paired, and they fold upon themselves to maximize base pairing and form a T-shaped hairpin structure.
  • the rep gene encodes four Rep proteins (Rep78, Rep68, Rep52, and Rep40), which are required for viral genome replication and packaging.
  • the cap gene expression results in viral capsid (Cap) proteins (VP; VP1/VP2/VP3), which form the outer capsid shell that protects the viral genome, as well as being actively involved in cell binding and internalization. Naso et al. (2017).
  • the AAV genome contains two viral promoters known as p5 and p19, which regulate the transcription of the four Rep proteins with molecular masses of 78, 68, 52, and 40 kDa (Rep68 and Rep40 being the splice variants of Rep78 and Rep52, respectively).
  • cap gene is driven by the P40 promoter and regulated by alternative splicing and different translation initiation sites, resulting in three cap proteins (VP1, VP2, and VP3) that form an icosahedral capsid of 3.9 kDa.
  • the molecular ratio of these proteins (VP1:VP2:VP3) is approximately 1:1:10.
  • the AAV genome also encodes for the AAP in an alternative ORF of the cap gene that plays a major role for capsid assembly. See Penaud-Budloo et al., Mol Ther Methods Clin Dev 2018;8:166-80.
  • AAV includes 12 different AAV serotypes.
  • AAV belongs to the genus Dependoparvovirus (the Parvoviridae family) because it needs the presence of a helper virus for replication and assembly, e.g., adeno-, herpes-, human papilloma- or vaccinia viruses.
  • a helper virus for replication and assembly e.g., adeno-, herpes-, human papilloma- or vaccinia viruses.
  • the virus gets internalized by endocytosis.
  • the viral capsid sheds to release the single-stranded AAV genome which is then converted to double-stranded DNA.
  • the free end of the ITR hairpin hereby acts as a primer for the DNA synthesis.
  • AAV can infect dividing or non-dividing cells.
  • AAV derived vectors have several advantages for viral-based gene therapy. AAV is believed to be non-pathogenic for humans, which makes it a suitable vehicle for delivery of genetic material to human cells. AAV also has low cytotoxicity and elicits a very mild immune response. Therefore, AAV vectors are well suitable for in vivo gene delivery. Furthermore, different AAV serotypes can target various tissues. Due to their properties, AAV derived vectors are becoming a more preferable tool as compared to adenovirus- and retrovirus-derived vectors.
  • the present application provides methods of making viral particle packaging and producer cell lines.
  • the methods comprise transfecting a cell with two or more nucleic acids.
  • the nucleic acids encode one or more of: (a) an enzyme capable of performing targeted genomic integration, (b) an inducible viral replication (Rep) gene, (c) an inducible viral Capsid (cap) gene, (d) one or more adenoviral auxiliary genes (e.g., selected from one or more of E1A, E1B, E4, E2A, and VA of an AAV), (e) an insulator (e.g., selected from HS4, D4Z4), (f) one or more terminal ends recognized by the enzyme, and (g) a transgene flanked by AAV inverted terminal repeats (ITRs).
  • an enzyme capable of performing targeted genomic integration e.g., an enzyme capable of performing targeted genomic integration
  • Rep inducible viral replication
  • Cap inducible viral Capsid
  • adenoviral auxiliary genes e.g., selected from one or more of E1A, E1B, E4, E2A, and VA of an AAV
  • the inducible viral replication (Rep) and Capsid (cap) genes are controlled by at least one of a tetO-containing P5 promoter, P19 promoter, SV40 promoter, P40 promoter, or a functional variant or fragment thereof.
  • the resulting transfected cell expresses the transgene in association with the viral particle.
  • the cell is transfected with two nucleic acids, and the first nucleic acid encodes (a) an enzyme capable of performing targeted genomic integration, and the second nucleic acid encodes an inducible viral Rep gene, (c) an inducible viral cap gene, (d) one or more adenoviral auxiliary genes, (e) an insulator, (f) one or more terminal ends recognized by the enzyme, and (g) a transgene flanked by AAV ITRs.
  • the first nucleic acid encodes (a) an enzyme capable of performing targeted genomic integration, and the second nucleic acid encodes an inducible viral Rep gene, (c) an inducible viral cap gene, (d) one or more adenoviral auxiliary genes, (e) an insulator, (f) one or more terminal ends recognized by the enzyme, and (g) a transgene flanked by AAV ITRs.
  • the cell is transfected with three nucleic acids, and the first nucleic acid encodes (a) an enzyme capable of performing targeted genomic integration; the second nucleic acid encodes (b) an inducible viral Rep gene, (c) an inducible viral cap gene, (d) one or more adenoviral auxiliary genes, (e) an insulator, and (f) one or more terminal ends recognized by the enzyme; and the third nucleic acid encodes (g) a transgene flanked by AAV ITRs.
  • the first nucleic acid encodes (a) an enzyme capable of performing targeted genomic integration
  • the second nucleic acid encodes (b) an inducible viral Rep gene, (c) an inducible viral cap gene, (d) one or more adenoviral auxiliary genes, (e) an insulator, and (f) one or more terminal ends recognized by the enzyme
  • the third nucleic acid encodes (g) a transgene flanked by AAV ITRs.
  • the nucleic acids encode one or more of: (a) an enzyme capable of performing targeted genomic integration, (b) an inducible viral replication (Rep) gene, (c) an inducible viral Capsid (cap) gene, (d) an insulator (e.g., selected from HS4, D4Z4), (e) one or more terminal ends recognized by the enzyme, and (f) a transgene flanked by AAV inverted terminal repeats (ITRs).
  • the inducible viral replication (Rep) and Capsid (cap) genes are controlled by at least one of a tetO-containing P5 promoter, P19 promoter, SV40 promoter, P40 promoter, or a functional variant or fragment thereof.
  • the resulting transfected cell expresses the transgene in association with the viral particle.
  • the E1A, E1B+ cell is transfected with two nucleic acids, and the first nucleic acid encodes (a) an enzyme capable of performing targeted genomic integration, and the second nucleic acid encodes an inducible viral Rep gene, (c) an inducible viral cap gene, (d) an insulator, (e) one or more terminal ends recognized by the enzyme, and (f) a transgene flanked by AAV ITRs.
  • the cell is transfected with three nucleic acids, and the first nucleic acid encodes (a) an enzyme capable of performing targeted genomic integration; the second nucleic acid encodes (b) an inducible viral Rep gene, (c) an inducible viral cap gene, (d) an insulator, and (e) one or more terminal ends recognized by the enzyme; and the third nucleic acid encodes (f) a transgene flanked by AAV ITRs.
  • the cell is transfected with four nucleic acids, and the first nucleic acid encodes (a) an enzyme capable of performing targeted genomic integration; the second nucleic acid encodes (b) an inducible viral Rep gene, (d) an insulator, and (e) one or more terminal ends recognized by the enzyme; the third nucleic acid encodes (c) an inducible viral cap gene; and the fourth nucleic acid encodes (f) a transgene flanked by AAV ITRs.
  • AAV adeno-associated virus
  • the methods make use of a technique to include, among other elements, the AAV viral replication (Rep) and Capsid (Cap) genes under a control of an inducible promoter, in an E1A, E1B+ cell.
  • the inducible Rep and Cap genes (under a control of an inducible promoter), AAV vector DNA sequences, and essential helper genes are included in a single genetic construct such as, e.g., a donor plasmid flanked by end sequences recognized by an enzyme capable of performing targeted genomic integration (e.g., without limitation, a mobile element enzyme in accordance with embodiments of the present disclosure).
  • a transgene of interest can be encoded by a separate nucleic acid, or it can be part of the genetic construct encoding the inducible Rep and Cap genes, AAV vector DNA sequences, and essential helper genes.
  • methods for making a packaging and producer cell line with a dual donor/helper system are provided.
  • inducible Rep and Cap genes (sometimes collectively referred to as “Rep/Cap genes”), and a transgene of interest are encoded by respective separate nucleic acids.
  • the dual donor/helper system includes, without limitation, a nucleic acid encoding inducible Rep and Cap genes, a nucleic acid (e.g., a donor expression vector, such as a helper AAV donor plasmid) encoding a transgene of interest, and a nucleic acid encoding an enzyme capable of performing targeted genomic integration (e.g., without limitation, a mobile element enzyme).
  • a nucleic acid e.g., a donor expression vector, such as a helper AAV donor plasmid
  • an enzyme capable of performing targeted genomic integration e.g., without limitation, a mobile element enzyme
  • the present application provides a method of making a viral particle packaging and producer cell line, the method comprising transfecting an E1A, E1B+ cell with a first nucleic acid encoding an enzyme capable of performing targeted genomic integration, a second nucleic acid encoding a donor DNA comprising inducible viral replication (Rep) and Capsid (cap) genes, adenoviral auxiliary genes selected from E1A and E1B, and insulators (optionally selected from HS4, D4Z4), and comprising terminal ends recognized by the enzyme, and a third nucleic acid encoding a donor DNA comprising a transgene flanked by AAV inverted terminal repeats (ITRs), to thereby result in a transfected cell that expresses the transgene in association with the viral particle.
  • ITRs inverted terminal repeats
  • a method of making a viral particle packaging and producer cell line comprises transfecting an E1A, E1B+ cell with: a first nucleic acid encoding an enzyme capable of performing targeted genomic integration; a second nucleic acid encoding a donor DNA comprising inducible viral replication (Rep) and Capsid (Cap) genes, adenoviral auxiliary genes selected from E4ORF6, E2A and VA RNA, and insulators (e.g., without limitation, HS4 and/or D4Z4), and comprising terminal ends recognized by the enzyme; wherein the inducible expression of AAV viral replication (Rep) and Capsid (cap) genes, as well as the adenoviral auxiliary genes E4ORF6 and E2A, are controlled by at least one of a tetO-containing P5 promoter, P19 promoter, SV40 promoter, P40 promoter, or a functional variant or fragment thereof, and a third nucleic acid encoding an enzyme capable of performing targeted genomic integration
  • inducible Rep/Cap genes and a nucleic acid e.g., a donor expression vector, such as a helper AAV donor plasmid
  • a nucleic acid e.g., a donor expression vector, such as a helper AAV donor plasmid
  • the single donor/helper system includes, without limitation, a nucleic acid (e.g., a donor expression vector) encoding inducible Rep and Cap genes and a transgene of interest, and a nucleic acid encoding an enzyme capable of performing targeted genomic integration (e.g., without limitation, a mobile element enzyme).
  • a method of making a viral particle packaging and producer cell line comprises transfecting an E1A, E1B+ cell with a first nucleic acid encoding an enzyme capable of performing targeted genomic integration; and a second nucleic acid encoding viral packaging genes, viral helper genes, insulators (e.g., without limitation, HS4 and D4Z4), and a transgene, and comprising terminal ends recognized by the enzyme, to thereby result in a transfected cell that expresses the transgene in association with a viral particle.
  • insulators e.g., without limitation, HS4 and D4Z4
  • the E1A, E1B+ cell is, without limitation, a HEK293, E1A, E1B+ engineered CHO-K1, or Sf9 cell line.
  • the E1A, E1B+ cell is, without limitation, a Chinese hamster ovary (CHO), baby hamster kidney (BHK), human embryonic kidney (HEK293T) cells, Vero cell, or Spodoptera frugiperda 9 (Sf9) cell
  • the viral helper genes comprise adenoviral auxiliary genes.
  • the adenoviral auxiliary genes are selected from one or more of E1A, E1B, E4ORF6, E2A, and VA of an AAV, optionally E1A and E1B.
  • the viral packaging genes comprise rep and cap genes of viral replication (Rep) and Capsid (Cap) proteins of an AAV.
  • the inducible viral replication (Rep) and Capsid (cap) genes are controlled by at least one of a tetO- containing P5 promoter, P19 promoter, SV40 promoter, P40 promoter, or a functional variant or fragment thereof.
  • the tetO-containing AAV2 P40 promoter comprises a nucleotide sequence of SEQ ID NO: 816, or a functional variant or fragment thereof, or sequence having at least about 70%, or at least about 80%, or at least about 85%, or at least about 90%, or at least about 95%, or at least about 97% identity thereto, or at least about 98% identity thereto, or at least about 99% identity thereto or at least about 10 mutations, or at least about 9 mutations, or at least about 8 mutations, or at least about 7 mutations, or at least about 6 mutations, or at least about 5 mutations, or at least about 4 mutations, or at least about 3 mutations, or at least about 2 mutations, or at least about 1 mutation.
  • the P40 intron is an AAV2 P40 intron.
  • the AAV2 P40 intron comprises a C276A substitution, or a substitution at position corresponding thereto relative to SEQ ID NO: 818.
  • the AAV2 P40 intron comprises one or more mutated translation start sites (ATGs), optionally wherein the translation start sites are mutated to one of CTG, ACG, and TTG.
  • the AAV2 P40 intron comprises substitutions at one or more positions A13, A32, T42, A61, A71, A89, A203, A246, A258, and T282, or one or more positions corresponding thereto, relative to SEQ ID NO: 818.
  • the AAV2 P40 intron comprises substitutions at one or more positions A13C, A32C, T42C, A61C, A71T, A89C, A203T, A246C, A258C, and T282C corresponding to SEQ ID NO: 818.
  • the AAV2 P40 intron comprises the nucleotide sequence of SEQ ID NO: 818, or a functional variant or fragment thereof, or sequence having at least about 70%, or at least about 80%, or at least about 85%, or at least about 90%, or at least about 95%, or at least about 97% identity thereto, or at least about 98% identity thereto, or at least about 99% identity thereto, or at least about 10 mutations, or at least about 9 mutations, or at least about 8 mutations, or at least about 7 mutations, or at least about 6 mutations, or at least about 5 mutations, or at least about 4 mutations, or at least about 3 mutations, or at least about 2 mutations, or at least about 1 mutation.
  • the AAV2 P40 intron comprises the nucleotide sequence of SEQ ID NO: 819, or a functional variant or fragment thereof, or sequence having at least about 70%, or at least about 80%, or at least about 85%, or at least about 90%, or at least about 95%, or at least about 97% identity thereto, or at least about 98% identity thereto, or at least about 99% identity thereto, or at least about 10 mutations, or at least about 9 mutations, or at least about 8 mutations, or at least about 7 mutations, or at least about 6 mutations, or at least about 5 mutations, or at least about 4 mutations, or at least about 3 mutations, or at least about 2 mutations, or at least about 1 mutation.
  • the AAV2 P40 intron comprises the nucleotide sequence of SEQ ID NO: 817, or a functional variant or fragment thereof, or sequence having at least about 70%, or at least about 80%, or at least about 85%, or at least about 90%, or at least about 95%, or at least about 97% identity thereto, or at least about 98% identity thereto, or at least about 99% identity thereto, or at least about 10 mutations, or at least about 9 mutations, or at least about 8 mutations, or at least about 7 mutations, or at least about 6 mutations, or at least about 5 mutations, or at least about 4 mutations, or at least about 3 mutations, or at least about 2 mutations, or at least about 1 mutation.
  • the modified TATA box element has the nucleotide sequence of TATATAA.
  • the tetO- containing AAV2 P40 promoter or a functional variant or fragment thereof, comprising a modified TATA box element and a modified AAV2 P40 intron comprises the nucleotide sequence of SEQ ID NO: 823, or a functional variant or fragment thereof, or sequence having at least about 70%, or at least about 80%, or at least about 85%, or at least about 90%, or at least about 95%, or at least about 97% identity thereto, or at least about 98% identity thereto, or at least about 99% identity thereto, or at least about 10 mutations, or at least about 9 mutations, or at least about 8 mutations, or at least about 7 mutations, or at least about 6 mutations, or at least about 5 mutations, or at least about 4 mutations, or at least about 3 mutations, or at least about 2 mutations, or at least about 1 mutation.
  • the tetO-containing P40 promoter is a tetO-containing AAV2 P40 promoter.
  • the AAV2 P40 promoter comprises the nucleotide sequence of SEQ ID NO: 820, or a functional variant or fragment thereof, or sequence having at least about 70%, or at least about 80%, or at least about 85%, or at least about 90%, or at least about 95%, or at least about 97% identity thereto, or at least about 98% identity thereto, or at least about 99% identity thereto, or at least about 10 mutations, or at least about 9 mutations, or at least about 8 mutations, or at least about 7 mutations, or at least about 6 mutations, or at least about 5 mutations, or at least about 4 mutations, or at least about 3 mutations, or at least about 2 mutations, or at least about 1 mutation.
  • the modified P40 intron is a AAV9 P40 intron.
  • the modified AAV9 P40 intron comprises the nucleotide sequence of SEQ ID NO: 821, or a functional variant or fragment thereof, or sequence having at least about 70%, or at least about 80%, or at least about 85%, or at least about 90%, or at least about 95%, or at least about 97% identity thereto, or at least about 98% identity thereto, or at least about 99% identity thereto, or at least about 10 mutations, or at least about 9 mutations, or at least about 8 mutations, or at least about 7 mutations, or at least about 6 mutations, or at least about 5 mutations, or at least about 4 mutations, or at least about 3 mutations, or at least about 2 mutations, or at least about 1 mutation.
  • the modified TATA box element has the nucleotide sequence of TATATAA.
  • the tetO-containing AAV2 P40 promoter or a functional variant or fragment thereof, comprising a modified TATA box element and a modified AAV9 P40 intron comprises the nucleotide sequence of SEQ ID NO: 822, or a functional variant or fragment thereof, or sequence having at least about 70%, or at least about 80%, or at least about 85%, or at least about 90%, or at least about 95%, or at least about 97% identity thereto, or at least about 98% identity thereto, or at least about 99% identity thereto, or at least about 10 mutations, or at least about 9 mutations, or at least about 8 mutations, or at least about 7 mutations, or at least about 6 mutations, or at least about 5 mutations, or at least about 4 mutations, or at least about 3 mutations, or at least about 2 mutations, or at least about 1 mutation.
  • the tetO-containing AAV2 P40 promoter or a functional variant or fragment thereof, comprising a modified TATA box element and a modified AAV9 P40 intron comprises a nucleotide sequence of SEQ ID NO: 824, or a functional variant or fragment thereof, or sequence having at least about 70%, or at least about 80%, or at least about 85%, or at least about 90%, or at least about 95%, or at least about 97% identity thereto, or at least about 98% identity thereto, or at least about 99% identity thereto, or at least about 10 mutations, or at least about 9 mutations, or at least about 8 mutations, or at least about 7 mutations, or at least about 6 mutations, or at least about 5 mutations, or at least about 4 mutations, or at least about 3 mutations, or at least about 2 mutations, or at least about 1 mutation.
  • the P40 promoter further comprises one or more enhancer elements.
  • the enhancer element comprises one or more cis-acting elements, optionally selected from an Sp1 binding site, GC rich sequence, GCGGAAC motif, TAATGARAT element, AP1 binding site, and CCAAT box element.
  • the enhancer element comprises about 1 to about 5 Sp1 binding sites, optionally about 1, or about 2, or about 3, or about 4, or about 5 Sp1 binding sites.
  • the enhancer element comprises about 1 or about 2 GC rich sequences.
  • the enhancer element is derived from an hCMV Enhancer Element-3.
  • the hCMV Enhancer Element-3 comprises the nucleotide sequence of SEQ ID NO: 830 or a functional variant or fragment thereof, or sequence having at least about 70%, or at least about 80%, or at least about 85%, or at least about 90%, or at least about 95%, or at least about 97% identity thereto, or at least about 98% identity thereto, or at least about 99% identity thereto, or at least about 10 mutations, or at least about 9 mutations, or at least about 8 mutations, or at least about 7 mutations, or at least about 6 mutations, or at least about 5 mutations, or at least about 4 mutations, or at least about 3 mutations, or at least about 2 mutations, or at least about 1 mutation.
  • the expression of the Rep and Cap proteins is controlled by an inducible promoter, optionally an antibiotic-dependent promoter.
  • the antibiotic-dependent promoter is tetracycline- or a variant thereof dependent promoter and a tetracycline-repressor-based (t-REx) system is used.
  • the antibiotic- dependent promoter is tetracycline- or a variant thereof dependent promoter and a tetracycline-controlled transactivator (rtTA) system is used.
  • the antibiotic-dependent promoter is a coumermycin/novobiocin promoter, or a variant thereof.
  • the viral particle is an AAV, and wherein the AAV is selected from AAV1, AAV2, AAV3, AAV4, AAV5, AAV6, AAV7, AAV8, and AAV9.
  • any one or more of the first, second, and third nucleic acids is encoded by a single nucleic acid.
  • the second nucleic acid and/or the third nucleic acid are included in a single expression vector.
  • the first and second nucleic acids are included in a single expression vector
  • the third nucleic acid is included in an expression vector that is different from the expression vector including the first and second nucleic acids.
  • the single expression vector comprises a plasmid.
  • the plasmid comprises the nucleotide sequence of SEQ ID NO: 825 or a functional variant or fragment thereof, or sequence having at least about 70%, or at least about 80%, or at least about 85%, or at least about 90%, or at least about 95%, or at least about 97% identity thereto, or at least about 98% identity thereto, or at least about 99% identity thereto, or at least about 10 mutations, or at least about 9 mutations, or at least about 8 mutations, or at least about 7 mutations, or at least about 6 mutations, or at least about 5 mutations, or at least about 4 mutations, or at least about 3 mutations, or at least about 2 mutations, or at least about 1 mutation.
  • the plasmid comprises the nucleotide sequence of SEQ ID NO: 826 or a functional variant or fragment thereof, or sequence having at least about 70%, or at least about 80%, or at least about 85%, or at least about 90%, or at least about 95%, or at least about 97% identity thereto, or at least about 98% identity thereto, or at least about 99% identity thereto, or at least about 10 mutations, or at least about 9 mutations, or at least about 8 mutations, or at least about 7 mutations, or at least about 6 mutations, or at least about 5 mutations, or at least about 4 mutations, or at least about 3 mutations, or at least about 2 mutations, or at least about 1 mutation.
  • the plasmid comprises the nucleotide sequence of SEQ ID NO: 841 or a functional variant or fragment thereof, or sequence having at least about 70%, or at least about 80%, or at least about 85%, or at least about 90%, or at least about 95%, or at least about 97% identity thereto, or at least about 98% identity thereto, or at least about 99% identity thereto, or at least about 10 mutations, or at least about 9 mutations, or at least about 8 mutations, or at least about 7 mutations, or at least about 6 mutations, or at least about 5 mutations, or at least about 4 mutations, or at least about 3 mutations, or at least about 2 mutations, or at least about 1 mutation.
  • the plasmid comprises the nucleotide sequence of SEQ ID NO: 842, or a functional variant or fragment thereof, or sequence having at least about 70%, or at least about 80%, or at least about 85%, or at least about 90%, or at least about 95%, or at least about 97% identity thereto, or at least about 98% identity thereto, or at least about 99% identity thereto, or at least about 10 mutations, or at least about 9 mutations, or at least about 8 mutations, or at least about 7 mutations, or at least about 6 mutations, or at least about 5 mutations, or at least about 4 mutations, or at least about 3 mutations, or at least about 2 mutations, or at least about 1 mutation.
  • the plasmid comprises the nucleotide sequence of SEQ ID NO: 843, or a functional variant or fragment thereof, or sequence having at least about 70%, or at least about 80%, or at least about 85%, or at least about 90%, or at least about 95%, or at least about 97% identity thereto, or at least about 98% identity thereto, or at least about 99% identity thereto, or at least about 10 mutations, or at least about 9 mutations, or at least about 8 mutations, or at least about 7 mutations, or at least about 6 mutations, or at least about 5 mutations, or at least about 4 mutations, or at least about 3 mutations, or at least about 2 mutations, or at least about 1 mutation.
  • one or more of the first, second, and third nucleic acids is RNA, optionally mRNA, optionally synthetic mRNA. In embodiments, one or more of the first, second, and third nucleic acids is DNA, optionally plasmid DNA. In embodiments, one or more of the first, second, and third nucleic acids is an expression vector, wherein the expression vector is optionally a plasmid.
  • the transfected cell generates using the method of making a viral particle packaging and producer cell line in accordance with embodiments of the present disclosure comprises a donor DNA comprising the transgene flanked by AAV inverted terminal repeats (ITRs).
  • the terminal ends or ITRs comprise the nucleotide sequence of SEQ ID NO: 831 and/or SEQ ID NO: 832, or a functional variant or fragment thereof, or sequence having at least about 70%, or at least about 80%, or at least about 85%, or at least about 90%, or at least about 95%, or at least about 97% identity thereto, or at least about 98% identity thereto, or at least about 99% identity thereto, or at least about 10 mutations, or at least about 9 mutations, or at least about 8 mutations, or at least about 7 mutations, or at least about 6 mutations, or at least about 5 mutations, or at least about 4 mutations, or at least about 3 mutations, or at least about 2 mutations, or at least about 1 mutation.
  • the transgene has a size of about 200,000 base pairs or less.
  • the methods in accordance with the present disclosure can be used to produce viral particles carrying a transgene of a large size.
  • the transgene has a size of at least 200,000 base pairs.
  • the transgene has a size of about 200,000 base pairs.
  • the method further comprises culturing the transfected cell in a medium that expands a population of the transfected cells to create a stably transfected packaging and producer cell line.
  • the stably transfected producer cell line is capable of producing replication-deficient viral particles in association with the transgene.
  • the transfection comprises electroporation, nucleofection, lipofection, or calcium phosphate transfection.
  • the method is helper virus-free.
  • an enzyme capable of performing targeted genomic integration causes the transgene to be inserted in a certain genomic locus and/or site (e.g., at a TA dinucleotide site or a TTAA tetranucleotide site in a genomic safe harbor site (GSHS)) of a nucleic acid molecule.
  • the enzyme capable of performing targeted genomic integration is a recombinase.
  • the enzyme has one or more mutations which confer hyperactivity.
  • the recombinase is an integrase.
  • the recombinase is an integrase or a mobile element enzyme.
  • the integrase is a mobile element enzyme.
  • the mobile element enzyme is an engineered mammalian mobile element enzyme.
  • the mobile element enzyme is a mammal-derived RNA mobile element enzyme (e.g., a helper RNA mobile element enzyme).
  • the mobile element enzyme is a mammal-derived DNA mobile element enzyme.
  • the mobile element enzyme is a chimeric mobile element enzyme.
  • the enzyme has gene cleavage activity (Exc+) and/or gene integration activity (Int+).
  • enzyme has gene cleavage activity (Exc+) and/or a lack of gene integration activity (Int-).
  • the mobile element enzyme comprises: (a) a gene-editing system, and (b) a mobile element enzyme that is capable of inserting the donor DNA comprising a transgene at a TA dinucleotide site or a TTAA tetranucleotide site in a genomic safe harbor site (GSHS).
  • the transgene encodes a complete polypeptide.
  • the transgene is defective or substantially absent in a disease state.
  • the gene-editing system comprises a Cas9 enzyme guide RNA complex.
  • the Cas9 enzyme guide RNA complex comprises a nuclease-deficient (or inactive, or dead) dCas9 guide RNA complex, also referred to as dCas9 guide RNA complex.
  • the nuclease-deficient dCas9 guide RNA complex comprises a guide RNA selected from: GTTTAGCTCACCCGTGAGCC (SEQ ID NO: 91), CCCAATATTATTGTTCTCTG (SEQ ID NO: 92), GGGGTGGGATAGGGGATACG (SEQ ID NO: 93), GGATCCCCCTCTACATTTAA (SEQ ID NO: 94), GTGATCTTGTACAAATCATT (SEQ ID NO: 95), CTACACAGAATCTGTTAGAA (SEQ ID NO: 96), TAAGCTAGAGAATAGATCTC (SEQ ID NO: 97), and TCAATACACTTAATGATTTA (SEQ ID NO: 98), or a variant thereof.
  • the enzyme is derived from Bombyx mori, Xenopus tropicalis, Trichoplusia ni, Myotis lucifugus, Rhinolophus ferrumequinum, Rousettus aegyptiacus, Phyllostomus discolor, Myotis myotis, Pteropus vampyrus, Pipistrellus kuhlii, Molossus molossus, Pan troglodytes, or Homo sapiens.
  • the enzyme is an engineered version, including but not limited to hyperactive forms, of an enzyme (e.g., without limitation, a mobile element enzyme) derived from Bombyx mori, Xenopus tropicalis, Trichoplusia ni, Myotis lucifugus, Rhinolophus ferrumequinum, Rousettus aegyptiacus, Phyllostomus discolor, Myotis myotis, Pteropus vampyrus, Pipistrellus kuhlii, Molossus molossus, Pan troglodytes, or Homo sapiens.
  • an enzyme e.g., without limitation, a mobile element enzyme derived from Bombyx mori, Xenopus tropicalis, Trichoplusia ni, Myotis lucifugus, Rhinolophus ferrumequinum, Rousettus aegyptiacus, Phyllostomus discolor, Myotis
  • the mobile element enzyme is from one or more of the Sleeping beauty, Tn1, Tn2, Tn3, Tn5, Tn7, Tn9, Tn10, Tn552, Tn903, Tn1000/Gamma-delta, Tn/O, tnsA, tnsB, tnsC, tniQ, IS10, ISS, IS911, Minos, piggyBac, Tol2, Mos1, Himar1, Hermes, Tol2, Minos, Tel, P-element, MuA, Ty1, Chapaev, transib, Tc1/mariner, or Tc3 donor DNA system, or biologically active fragments variants thereof, inclusive of hyperactive variants.
  • the enzyme is a mobile element enzyme such as a Myotis lucifugus mobile element enzyme (MLT mobile element enzyme), also referred to herein as an MLT mobile element enzyme.
  • MLT mobile element enzyme is a wild-type MLT mobile element enzyme.
  • the MLT mobile element enzyme is a modified MLT mobile element enzyme, also referred to herein as a corrected MLT mobile element enzyme.
  • the MLT mobile element enzyme e.g., the wild-type MLT mobile element enzyme, the corrected MLT mobile element enzyme, or a variant thereof
  • has one or more mutations such as hyperactive mutations.
  • the MLT mobile element enzyme is a modified MLT mobile element enzyme.
  • the MLT mobile element enzyme has an amino acid sequence of SEQ ID NO: 1, or a variant having at least about 90%, or at least about 93%, or at least about 95%, or at least about 97%, or at least about 98%, or at least about 99% identity thereto, and one or more mutations selected from L573X, E574X, and S2X, wherein X is any amino acid or no amino acid, optionally X is A, G, or a deletion.
  • the mutations are L573del E574del, and S2A (SEQ ID NO: 1).
  • the MLT mobile element enzyme has an amino acid sequence of SEQ ID NO: 1 and S8P and C13R mutations (SEQ ID NO: 11).
  • the MLT mobile element enzyme has an amino acid sequence having mutations at positions which correspond to at least one of S8P and C13R mutations relative to the amino acid of SEQ ID NO: 1 or a functional equivalent thereof. In embodiments, the MLT mobile element enzyme has an amino acid sequence having mutations at positions which correspond to S8P and C13R mutations relative to the amino acid of SEQ ID NO: 1 or a functional equivalent thereof. In embodiments, the MLT mobile element enzyme has an amino acid sequence having mutations at positions which correspond to S8P, C13R, and N125K mutations relative to the amino acid of SEQ ID NO: 1 or a functional equivalent thereof.
  • the MLT mobile element enzyme has a nucleotide sequence of SEQ ID NO: 2 (which is codon- optimized) and an amino acid sequence SEQ ID NO: 1.
  • the MLT mobile element enzyme has a nucleotide sequence of SEQ ID NO: 2, or a nucleotide sequence having at least about 90%, or at least about 93%, or at least about 95%, or at least about 97%, or at least about 98%, or at least about 99% identity thereto, or a codon- optimized form thereof.
  • the MLT mobile element enzyme has an amino acid sequence SEQ ID NO: 1, or an amino acid sequence having at least about 90%, or at least about 93%, or at least about 95%, or at least about 97%, or at least about 98%, or at least about 99% identity thereto.
  • the MLT mobile element enzyme includes a hyperactive mutation selected from TABLE 14A and TABLE 14B, e.g., about 1, or about 2, or about 3, or about 4, or about 5 hyperactive mutations selected from TABLE 14A or TABLE 14B, or combinations thereof.
  • the MLT mobile element enzyme is a wild-type mobile element enzyme that has an amino acid sequence of SEQ ID NO: 10.
  • the MLT mobile element enzyme is a modified mobile element enzyme having the amino acid sequence of SEQ ID NO: 11.
  • at least one of the first, second, and third nucleic acids is in the form of a lipid nanoparticle (LNP).
  • LNP lipid nanoparticle
  • a nucleic acid encoding an enzyme capable of performing targeted genomic integration (e.g., without limitation, a mobile element enzyme) and a nucleic acid encoding a donor DNA are in the form of the same LNP, optionally in a co-formulation.
  • a nucleic acid encoding an enzyme capable of performing targeted genomic integration e.g., without limitation, a mobile element enzyme
  • a nucleic acid encoding a donor DNA are in the same mixture with an LNP.
  • a method of producing an AAV bearing a gene of interest is provided, to produce the AAV bearing the gene of interest.
  • the method of making a viral packaging or producer cell line of the present disclosure provides a stable cell with substantially reduced or ablated CAP expression.
  • the method of making a viral packaging or producer cell line of the present disclosure provides a stable cell with REP expression or substantially enhanced REP expression.
  • the method of making a viral packaging or producer cell line of the present disclosure provides a stable cell with substantially reduced or ablated CAP expression and REP expression or substantially enhanced REP expression. In embodiments, the method of making a viral packaging or producer cell line of the present disclosure provides a stable cell with REP expression or substantially enhanced VP1 expression. In embodiments, the method of making a viral packaging or producer cell line of the present disclosure provides a stable cell with substantially reduced or ablated CAP expression. In embodiments, a cell for gene therapy is provided that is generated by a method in accordance with embodiments of the present disclosure. In some embodiments, a pharmaceutical composition comprising the cell is provided.
  • a method for treating a disease or condition using gene therapy comprising administering to a subject in need thereof a transfected cell generated using a method in accordance with embodiments of the present disclosure.
  • the disease or condition comprises cancer.
  • the disease or condition comprises an infectious disease.
  • the infectious disease is a coronavirus infection, optionally selected from infection with SAR-CoV, MERS-CoV, and SARS-CoV-2, or variants thereof.
  • FIG.1A is a non-limiting representation of an AAV production strategy in accordance with embodiments of the present disclosure, using a donor vector to create a HEK293 producer cell line or another cell line expressing the AdV E1a and E1b genes (e.g., engineered CHO-K1 or Vero cell line with E1a, E1b) that forms a replication deficient AAV particle containing a transgene of interest.
  • the single or dual donor comprising a transgene of interest is incorporated (e.g., by transfection such as electroporation) into a HEK293 cell line or another cell line expressing the E1a and E1b genes.
  • FIG.1B is a representation of an example of an inducible Rep/Cap and helper AAV donor plasmid construct (Kana r ), encoding E2A, E4ORF6, and VA RNA helper genes flanked by insulators and mobile element enzyme recognition ends, and used with a helper RNA or DNA to create an AAV producer cell line, in accordance with embodiments of the present disclosure.
  • FIG.1C is a representation of an example of a nucleic acid (plasmid) encoding a transgene (gene of interest (GOI)) included between AAV inverted terminal repeats (ITRs), in accordance with embodiments of the present disclosure.
  • FIGs.2A-E depict non-limiting representations of chimeric, monomer or head-to-tail dimer mobile element enzymes that are designed to target human GSHS using TALE and Cas9/guide RNA DNA binders.
  • TALEs include nuclear localization signals (NLS) and an activation domain (AD) to function as transcriptional activators.
  • the DNA binding domain has approximately 16.5 repeats of 33-34 amino acids with a residual variable di-residue (RVD) at position 12-13.
  • RVD residual variable di-residue
  • FIG.2B RVDs are shown that have specificity for one or several nucleotides. Only bases of the DNA leading strand are shown.
  • FIG.2C A chimeric mobile element enzyme construct comprising a TALE DNA-binding protein fused thereto by a linker that is greater than 23 amino acids in length (top) and a chimeric mobile element enzyme construct comprising dCas9 linked to one or more guide RNAs (bottom).
  • 2D is a non-limiting representation of a system in accordance with embodiments of the present disclosure comprising a nucleic acid (e.g., helper RNA) encoding an enzyme capable of performing targeted genomic integration and a nucleic acid encoding a mobile element enzyme (donor DNA).
  • the helper RNA is translated into a bioengineered enzyme (e.g., integrase, recombinase, or mobile element enzyme) that recognizes specific ends and seamlessly inserts the donor DNA into the human genome in a site-specific manner without a footprint.
  • Chimeric mobile element enzymes form dimers or tetramers at open chromatin to insert donor DNA at TTAA recognition sites near DNA binding regions targeted by dCas9/gRNA or TALEs.
  • FIG.2E is a non-limiting representation of a system in accordance with embodiments of the present disclosure comprising a nucleic acid (e.g., helper RNA) encoding an enzyme capable of performing targeted genomic integration and a nucleic acid encoding a mobile element enzyme (donor DNA).
  • a nucleic acid e.g., helper RNA
  • the helper RNA is translated into a bioengineered enzyme (e.g., integrase, recombinase, or mobile element enzyme) that recognizes specific ends and seamlessly inserts the donor DNA into the human genome in a site-specific manner without a footprint.
  • FIG.3B depicts a donor DNA construct template with the transgene VLDLR.
  • FIG.3C depicts a chimeric mobile element enzyme construct template with a TALE DNA binder. Other TALEs and mobile element enzymes can be substituted.
  • FIG.3D depicts a chimeric mobile element enzyme construct template with a dCas9/gRNA DNA binder.
  • FIG.3E depicts a system comprising donor DNA (panel A) and helper RNA (panel B): panel (A)
  • the donor DNA can be any gene of interest (GOI) including a gene that replaces, inactivates, or provides suicide or helper functions.
  • the GOI can be driven by a predetermined promoter and flanked by insulators to prevent gene silencing.
  • the internal terminal repeats (ITRs) can be specific for a mammal- derived mobile element enzyme.
  • the helper RNA is 5’-m7G capped (cap 0, or cap1, or cap 2) with flanking globin 5’- and 3’-UTRs, an N-terminus nuclear localization signal (SV40 or nucleoplasmin), a 34 polyalanine tail region, and pseudouridine modification.
  • the mobile element enzyme e.g., hyperactive mobile element enzyme
  • FIG.4 depicts a non-limiting representation of a conventional rAAV production system.
  • Production in adenovirus complementation systems is usually performed as plasmid transfection processes, where AAV Rep/Cap genes, the ITR-flanked gene of interest (GOI), as well as Ad-helper genes are provided as three separate plasmids, respectively, to a E1a/E1b containing HEK293 cell line or other engineered cell line that contains E1a/E1b (e.g., engineered CHO- K1 or Vero cells).
  • AAV Rep/Cap genes the ITR-flanked gene of interest (GOI)
  • Ad-helper genes are provided as three separate plasmids, respectively, to a E1a/E1b containing HEK293 cell line or other engineered cell line that contains E1a/E1b (e.g., engineered CHO- K1 or Vero cells).
  • FIG.5 depicts a non-limiting schematic of an engineered mobile element enzyme ITR containing plasmid that encodes 1) AAV2_REP78/68 ORFs under the control of P5TO1 promote with AAV2_REP52/40 ORFs and CAPs (VP1/2/3) under their native P19 or P40 promoter, respectively, 2) Ad5 E2A_IRES_Ad5 E4 ORF6 under the control of the tetO- containing SV40 promoter, 3) VA RNAs under the control of HSV-2 ICP4 promoter, and 4) hygro-B resistant gene under the control of TK promoter.
  • ITR engineered mobile element enzyme
  • FIG.6 depicts a non-limiting schematic of an engineered mobile element enzyme ITR containing plasmid that encodes 1) AAV2_REP78/68 ORFs under the control of P5TO1 promoter and AAV2_REP52/40 ORFs under the native P19 promoter, 2) AAV2 CAPs under the tetO-containing P40 promoter with AAV9 P40 intron, 3) Ad5 E2A_IRES_Ad5 E4 ORF6 under the control of the tetO-containing SV40 promoter, 4) VA RNAs under the control of HSV-2 ICP4 promoter, and 5) hygro-B resistant gene under the control of TK promoter.
  • ITR containing plasmid that encodes 1) AAV2_REP78/68 ORFs under the control of P5TO1 promoter and AAV2_REP52/40 ORFs under the native P19 promoter, 2) AAV2 CAPs under the tetO-containing P40 promote
  • FIG.7 depicts a non-limiting schematic of an engineered mobile element enzyme ITR containing plasmid that encodes 1) AAV2_REP 78/68 ORFs under the control of P5TO1 promoter and AAV2_REP52/40 ORFs under the native P19 promoter, 2) Ad5 E2A_IRES_Ad5 E4 ORF6 under the control of the tetO-containing SV40 promoter, 3) VA RNAs under the control of HSV-2 ICP4 promoter, and 4) hygro-B resistant gene under the control of TK promoter.
  • FIG.8 depicts a non-limiting schematic of a construct encoding AAV2 CAP under the control of the tetO-containing hCMV-derived enhancer element 3/AAV2 P40 promoter with a modified AAV2 P40 TATA element and modified AAV2 P40 intron_1 (a at position 276 substituted for C) and WPRE/modified ICP27 poly A, followed by ICP0 promoter-Zeocin resistant gene-ICP0 poly A cassette flanked with ITRs for Sleeping Beauty (SB) transposase.
  • SB Sleeping Beauty
  • FIG.9 depicts a non-limiting schematic of a construct encoding AAV2 CAP under the control of the tetO-containing hCMV enhancer element 3/AAV2 P40 promoter with a modified AAV2 P40 TATA element and MODIFIED AAV2 P40 intron_2 and WPRE/modified ICP27 poly A, followed by ICP0 promoter-Zeocin resistant gene-ICP0 poly A cassette flanked with SB ITRs.
  • FIG.10A-10B depicts GFP images 120 hours post-transduction of AAVH clone 17+ (FIG.10A) and ddPCR analyses for quantification of the rAAV2-EGFP genome copy number per nanogram (ng) of total genomic DNA (FIG.10B).
  • FIG.11A – 11B depicts GFP images 120 hours post-transduction of AAVH clone 13 + (FIG.10A) and ddPCR analyses for quantification of the rAAV2-EGFP genome copy number per nanogram (ng) of total genomic DNA (FIG.10B).
  • FIG.12A – 12B depicts expression of functional AAV2 Rep proteins in AAV2-HB (Plasmid shown in FIG.6) clones as determined by western blot and ddPCR.
  • FIG.12A depicts a western blot analysis illustrating cells from selected AAV2- H B cell clones HB-36, HB-42, HB-118, and HB-120 were treated with10ng/ml doxycycline for 72 hours followed by extraction with RIPA buffer in the presence of proteinase inhibitor.
  • FIG.12B shows the results of a ddPCR analysis for AAV Genome Amplification / Rep function Assay.
  • FIG.13A – 13B depicts expression of functional AAV2 Rep proteins in AAV-CDH (Plasmid shown in FIG.7) clones as determined by western blot (FIG.13A) and ddPCR (FIG.13B).
  • FIG.14A – 14B depicts establishment of AAV2 full producer stable cell pools, CDH-24/Cap-SB, CDH-48/Cap-SB, and CDH-129/Cap-SB, with sleeping beauty transposase-mediated integration of AAV2 CAPs-expressing cassettes (Plasmid shown in FIG.7) into AAV-CDH Clone 24, 48, 129 serotype adaptable (Cap minus or Cap-) stable cell lines.
  • FIG.14A shows ddPCR analyses of SB-mediated AAV2 Cap transposon integration in each Zeocin-selected stable pools.
  • FIG.14B shows western blot analysis of SB-mediated AAV2 Cap transposon integration in each Zeocin-selected stable pools.
  • FIG.15A – 15B depicts expression of functional AAV2 Rep proteins in CDH-24/Cap-SB, CDH-48/Cap-SB, and CDH- 129/Cap-SB pools as determined by western blot and ddPCR.
  • FIG.15A shows ddPCR results of rAAV Rep proteins being expressed and
  • FIG.15B shows expressed AAV2 Rep proteins are functionally active in supporting AAV genome amplification.
  • FIG.16 depicts expression of AAV2 Cap and Rep in CDH-48/Cap-SB stable pool increased with doxycycline dosages and over time.
  • Cells from CDH-48/Cap-SB stable pools were incubated either in the presence of 0, 5, 10, 25, 50, or 100ng/ml doxycycline and harvest at 72 hours post induction; or incubated with 50ng/ml doxycycline and harvested at 24, 48, 72, or 96 hours post induction (FIG.16).
  • Cell extraction with RIPA buffer containing proteinase inhibitors, western blot, and ddPCR analyses of the CDH-48/Cap-SB (AAV2-capsid minus stable lines Clone CDH-48) samples were caried out as described previously.
  • FIG.17A – 17B depicts production of competent rAAV2 particles from stably engineered CDH-48/Cap-SB cell pools in response to doxycycline.
  • FIG.17A show a graph depicting rAAV2 physical titers from CDH-48/CAP-SB stable pool harvests.
  • FIG.17B shows a graph depicting transduction of CDH-48 cells (AAV2-CAPSID MINUS STABLE LINES CLONE CDH-48) with unconcentrated rAAV supernatents.
  • FIG.18A – 18B depicts characterization of Zeocin-resistant single clones selected from the CDH-48/Cap-SB stable pool.
  • FIG.18A depicts ddPCR analysis for SB-mediated AAV2-Cap cassette integration.
  • FIG.18B depicts western blot analysis with antibodies specific for AAV2 CAP.
  • FIG.19A – 19B depicts characterization of Zeocin-resistant single clones selected from the CDH-48/Cap-SB stable pool.
  • FIG.19A depicts western blot analysis with antibodies specific for AAV2 REP.
  • FIG.19B shows ddPCR assays of AAV genome amplification following transduction with low VGC/cell of AAV2-EGFP viral particles.
  • the present invention is based, in part, on the discovery that a mobile element enzyme-mediated site-specific genomic integration can be used to produce a stable AAV-based cell line.
  • the cell line is produced by insertion of inducible viral replication (rep) and Capsid (cap) genes and helper genes (E4ORF6, E2A, and VA) in specific genomic locations of a cell such as, e.g., a HEK293 (E1+) cell or a genetically engineered CHO-K1 or Vero cell line.
  • An advantage of the approach in accordance with embodiments of the present disclosure is that it does not require DNA homology for recombination, and it is independent of the size of the donor or recipient DNA molecules.
  • the described method is performed in a brief enzyme-catalyzed reaction using an enzyme (e.g., without limitation, a mobile element enzyme) delivered as either DNA or RNA.
  • an enzyme e.g., without limitation, a mobile element enzyme
  • this protocol allows for the production of stable expression human cell pools in about 4 weeks or in less than 4 weeks.
  • the present application provides methods of making viral particle packaging and producer cell lines.
  • the methods comprise transfecting a cell with two or more nucleic acids.
  • the nucleic acids encode one or more of: (a) an enzyme capable of performing targeted genomic integration, (b) an inducible viral replication (Rep) gene, (c) an inducible viral Capsid (cap) gene, (d) one or more adenoviral auxiliary genes (e.g., selected from one or more of E1A, E1B, E4, E2A, and VA of an AdV), (e) an insulator (e.g., selected from HS4, D4Z4), (f) one or more terminal ends recognized by the enzyme, and (g) a transgene flanked by AAV inverted terminal repeats (ITRs).
  • an enzyme capable of performing targeted genomic integration e.g., an enzyme capable of performing targeted genomic integration
  • Rep inducible viral replication
  • Cap inducible viral Capsid
  • adenoviral auxiliary genes e.g., selected from one or more of E1A, E1B, E4, E2A, and VA of an AdV
  • the inducible viral replication (Rep) and Capsid (cap) genes are controlled by at least one of a tetO-containing P5 promoter, P19 promoter, SV40 promoter, P40 promoter, or a functional variant or fragment thereof.
  • the resulting transfected cell expresses the transgene in association with the viral particle.
  • the cell is transfected with two nucleic acids, and the first nucleic acid encodes (a) an enzyme capable of performing targeted genomic integration, and the second nucleic acid encodes an inducible viral Rep gene, (c) an inducible viral cap gene, (d) one or more adenoviral auxiliary genes, (e) an insulator, (f) one or more terminal ends recognized by the enzyme, and (g) a transgene flanked by AAV ITRs.
  • the first nucleic acid encodes (a) an enzyme capable of performing targeted genomic integration, and the second nucleic acid encodes an inducible viral Rep gene, (c) an inducible viral cap gene, (d) one or more adenoviral auxiliary genes, (e) an insulator, (f) one or more terminal ends recognized by the enzyme, and (g) a transgene flanked by AAV ITRs.
  • the cell is transfected with three nucleic acids, and the first nucleic acid encodes (a) an enzyme capable of performing targeted genomic integration; the second nucleic acid encodes (b) an inducible viral Rep gene, (c) an inducible viral cap gene, (d) one or more adenoviral auxiliary genes, (e) an insulator, and (f) one or more terminal ends recognized by the enzyme; and the third nucleic acid encodes (g) a transgene flanked by AAV ITRs.
  • the first nucleic acid encodes (a) an enzyme capable of performing targeted genomic integration
  • the second nucleic acid encodes (b) an inducible viral Rep gene, (c) an inducible viral cap gene, (d) one or more adenoviral auxiliary genes, (e) an insulator, and (f) one or more terminal ends recognized by the enzyme
  • the third nucleic acid encodes (g) a transgene flanked by AAV ITRs.
  • the cell is transfected with four nucleic acids, and the first nucleic acid encodes (a) an enzyme capable of performing targeted genomic integration; the second nucleic acid encodes (b) an inducible viral Rep gene, (d) one or more adenoviral auxiliary genes, (e) an insulator, and (f) one or more terminal ends recognized by the enzyme; the third nucleic acid encodes (c) an inducible viral cap gene; and the fourth nucleic acid encodes (g) a transgene flanked by AAV ITRs.
  • the present application provides methods of transfecting an E1A, E1B+ cell with two or more nucleic acids.
  • the nucleic acids encode one or more of: (a) an enzyme capable of performing targeted genomic integration, (b) an inducible viral replication (Rep) gene, (c) an inducible viral Capsid (cap) gene, (d) an insulator (e.g., selected from HS4, D4Z4), (e) one or more terminal ends recognized by the enzyme, and (f) a transgene flanked by AAV inverted terminal repeats (ITRs).
  • the inducible viral replication (Rep) and Capsid (cap) genes are controlled by at least one of a tetO-containing P5 promoter, P19 promoter, SV40 promoter, P40 promoter, or a functional variant or fragment thereof.
  • the resulting transfected cell expresses the transgene in association with the viral particle.
  • the E1A, E1B+ cell is transfected with two nucleic acids, and the first nucleic acid encodes (a) an enzyme capable of performing targeted genomic integration, and the second nucleic acid encodes an inducible viral Rep gene, (c) an inducible viral cap gene, (d) an insulator, (e) one or more terminal ends recognized by the enzyme, and (f) a transgene flanked by AAV ITRs.
  • the cell is transfected with three nucleic acids, and the first nucleic acid encodes (a) an enzyme capable of performing targeted genomic integration; the second nucleic acid encodes (b) an inducible viral Rep gene, (c) an inducible viral cap gene, (d) an insulator, and (e) one or more terminal ends recognized by the enzyme; and the third nucleic acid encodes (f) a transgene flanked by AAV ITRs.
  • the cell is transfected with four nucleic acids, and the first nucleic acid encodes (a) an enzyme capable of performing targeted genomic integration; the second nucleic acid encodes (b) an inducible viral Rep gene, (d) an insulator, and (e) one or more terminal ends recognized by the enzyme; the third nucleic acid encodes (c) an inducible viral cap gene; and the fourth nucleic acid encodes (f) a transgene flanked by AAV ITRs.
  • the first nucleic acid encodes (a) an enzyme capable of performing targeted genomic integration
  • the second nucleic acid encodes (b) an inducible viral Rep gene, (d) an insulator, and (e) one or more terminal ends recognized by the enzyme
  • the third nucleic acid encodes (c) an inducible viral cap gene
  • the fourth nucleic acid encodes (f) a transgene flanked by AAV ITRs.
  • the present application provides a method of making a viral particle packaging and producer cell line, the method comprising transfecting an E1A, E1B+ cell with a first nucleic acid encoding an enzyme capable of performing targeted genomic integration, a second nucleic acid encoding a donor DNA comprising inducible viral replication (Rep) and Capsid (cap) genes, adenoviral auxiliary genes selected from E1A and E1B, and insulators (optionally selected from HS4, D4Z4), and comprising terminal ends recognized by the enzyme, and a third nucleic acid encoding a donor DNA comprising a transgene flanked by AAV inverted terminal repeats (ITRs), to thereby result in a transfected cell that expresses the transgene in association with the viral particle.
  • ITRs inverted terminal repeats
  • a method of making a viral particle packaging and producer cell line comprises: transfecting an E1A, E1B+ cell with a first nucleic acid encoding an enzyme capable of performing targeted genomic integration; a second nucleic acid encoding a donor DNA comprising inducible viral replication (rep) and Capsid (cap) genes, adenoviral auxiliary genes selected from E2A, E4ORF6, and VA, and insulators (optionally selected from HS4, D4Z4), and comprising terminal ends recognized by the enzyme, wherein the inducible viral replication (Rep) and Capsid (cap) genes are controlled by at least one of a tetO-containing P5 promoter, P19 promoter, SV40 promoter, P40 promoter, or a functional variant or fragment thereof; and a third nucleic acid encoding a donor DNA comprising a transgene flanked by AAV inverted terminal repeats (ITRs), to thereby result in
  • a method of making a viral particle packaging and producer cell line comprises: transfecting an E1A, E1B+ cell with a first nucleic acid encoding an enzyme capable of performing targeted genomic integration, and a second nucleic acid encoding viral packaging genes, viral helper genes, insulators (optionally selected from HS4 and D4Z4), and a transgene, and comprising terminal ends recognized by the enzyme to thereby result in a transfected cell that expresses the transgene in association with a viral particle.
  • the inducible viral replication (Rep) and Capsid (cap) genes are controlled by at least one of a tetO- containing P5 promoter, P19 promoter, SV40 promoter, P40 promoter, or a functional variant or fragment thereof.
  • a mammal-derived, helper RNA mobile element enzyme and donor DNA system is used to produce purified recombinant adeno-associated virus (rAAV) vector stocks.
  • rAAV stocks comprising an ITR-flanked therapeutic transgene or a gene of interest (GOI), are generated by a one plasmid or two-plasmid system, which creates a single producer cell line or separate packaging and producer cell lines.
  • the assembly of rAAV vectors typically requires (1) a recombinant vector genome comprising a gene of interest (GOI) and the regulation elements for the GOI expression in target cells (e.g., a promoter, poly A, introns, etc.) flanked by AAV ITRs, (2) the AAV rep and cap genes provided in trans, and (3) Helper functions from adeno-, herpes-, human papilloma- or vaccinia viruses, for replication and rescue of the recombinant genome.
  • GOI gene of interest
  • target cells e.g., a promoter, poly A, introns, etc.
  • helper functions from adeno-, herpes-, human papilloma- or vaccinia viruses for replication and rescue of the recombinant genome.
  • a conventional method for production of a vector is co-transfection of a vector plasmid containing the vector genome and a helper plasmid encoding the rep and cap genes into E1A-transformed human embryonic kidney cells (HEK293 cells) infected with Ad (see FIG.4).
  • HEK293 cells E1A-transformed human embryonic kidney cells
  • Ad Ad
  • AAV vector packaging strategies that rely on overexpression of the viral gene products face challenges such as issues associated with the cellular toxicity of Rep proteins, and the requirement that rep and cap expression levels be tightly regulated for cell viability hence maximal virion production.
  • An inducible promoter e.g., tetracycline (Tet), cumate, coumermycin/novobiocin, etc.
  • HEK293 cells are simultaneously transfected by a vector plasmid (containing the GOI) and one or two helper plasmids.
  • the helper plasmid(s) allow the expression of the four Rep proteins, the three AAV structural proteins VP1, VP2, and VP3, the AAP, and the adenoviral auxiliary functions E2A, E4ORF6, and VA RNA.
  • adenoviral E1A/E1B co-factors necessary for rAAV replication are expressed in HEK293 producer cells. See Qiao et al., J Virol 2002;76:1904-13; Yuan et al., Hum Gene Ther 2011;22:613-24; Lock et al., Hum Gene Ther 2010;21:1259- 71.
  • Rep/cap genes and adenoviral helper sequences are either cloned on two separate plasmids or combined on one plasmid, hence evolving from a triple plasmid system to transfection with only two plasmids.
  • the triple plasmid protocol provides versatility with a cap gene that can be switched from one serotype to another.
  • the plasmids are usually produced by conventional techniques in E. coli using bacterial origin and antibiotic-resistance gene or by minicircle (MC) technology.
  • Schnodt et al. Mol Ther Nucleic Acids 2016;5:e355.
  • transient transfection in adherent HEK293 cells has been used for large-scale manufacturing of rAAV vectors, it typically requires multiple production batches to fulfill the needs of clinical trials, resulting in lengthy and costly production campaigns.
  • HEK293 cells have been adapted to suspension conditions to be economically viable in the long term.
  • Grieger et al. Mol Ther 2016;24:287-97.
  • a method of making a viral particle packaging and producer cell line comprises transfecting an E1A, E1B+ cell with (1) a first nucleic acid encoding an enzyme capable of performing targeted genomic integration, (2) a second nucleic acid encoding a donor DNA comprising inducible viral replication (rep) and Capsid (cap) genes, adenoviral auxiliary genes selected from E2A, E4ORF6, and VA, and insulators (optionally selected from HS4 and D4Z4), and comprising terminal ends recognized by the enzyme, wherein the inducible viral replication (Rep) and Capsid (cap) genes are controlled by at least one of a tetO-containing P5 promoter, P19 promoter, SV40 promoter, P40 promoter, or a functional variant or fragment thereof, and (3) a third nucleic acid encoding a donor DNA comprising a transgene flank
  • a method of making a viral particle packaging and producer cell line comprises transfecting an E1A, E1B+ cell with (1) a first nucleic acid encoding an enzyme capable of performing targeted genomic integration, and (2) a second nucleic acid encoding viral packaging genes, viral helper genes, insulators (optionally selected from HS4 and D4Z4), and a transgene, and comprising terminal ends recognized by the enzyme, to thereby result in a transfected cell that expresses the transgene in association with a viral particle.
  • FIG.1A illustrates a non-limiting example of a method for generating AAV, using a single or dual donor DNA to create a producer cell line from a E1+ cell line (e.g., without limitation, an HEK293 cell line) that forms a replication deficient (rep-) AAV particle containing a transgene of interest.
  • a E1+ cell line e.g., without limitation, an HEK293 cell line
  • rep- replication deficient
  • a donor DNA construct encodes inducible Rep and Cap (“Capsid”) proteins (VP1, VP2, and VP3) and helper genes (E4, E2A, and VA) having end sequences recognized by an enzyme capable of performing targeted genomic integration (e.g., without limitation, a mobile element enzyme), as well as a transgene (or gene) of interest flanked by AAV ITRs.
  • the donor DNA construct is a dual donor DNA comprising a nucleic acid encoding the inducible Rep and Cap proteins and helper genes that is separate from a nucleic acid encoding transgene.
  • the donor DNA construct is a single donor DNA comprising a nucleic acid encoding the inducible Rep and Cap proteins, helper genes, and a transgene.
  • FIG.1B shows an example of an inducible Rep/Cap and helper AAV donor plasmid construct (Kana r ), encoding helper E2A, E4ORF6 and VA genes flanked by insulators and mobile element enzyme recognition ends, and used with a helper RNA or DNA (e.g., a mobile element enzyme) to create an AAV producer cell line.
  • FIG.1C shows an example of a plasmid encoding a transgene included between AAV ITRs.
  • the plasmids shown in FIGs.1B and 1C can be combined or can be used separately for transfection into an E1+ cell line to produce an AAV particle comprising the transgene (or gene of interest (GOI)).
  • the method depicted in FIG.1A is performed using a system in accordance with embodiments of the present disclosure shown in FIG.2D.
  • the system of FIG.2D is an integrative, non-viral donor DNA system for site- specific, stable genomic integration.
  • the system allows producing any recombinant AAV serotype by the insertion of inducible AAV rep/cap genes and helper genes (E4, E2A, and VA) in specific genomic locations in a cell, such as, e.g., HEK293 (E1+) cells, a genetically engineered CHO-K1 cell line, or in other cells.
  • the system of FIG.2D comprises comprising a nucleic acid (e.g., helper RNA [which is different from helper genes]) encoding an enzyme capable of performing targeted genomic integration (e.g., without limitation, a mobile element enzyme) and a nucleic acid encoding a transgene of interest (donor DNA).
  • the helper RNA is translated into a bioengineered enzyme (e.g., integrase, recombinase, or mobile element enzyme) that recognizes specific ends and seamlessly inserts the transgene of interest into the human genome in a site-specific manner without a footprint.
  • a bioengineered enzyme e.g., integrase, recombinase, or mobile element enzyme
  • the present disclosure provides, in embodiments, methods for producing AAV packaging cell lines capable of regulated gene amplification.
  • the methods of the present disclosure employ tetracycline repressor-controlled (tetR) tetO containing promoters (Yao F et al., Hum Gene Ther., 9:1939-1950, 1998) or reverse tetracycline-controlled transactivator (rtTA)-controlled gene expression and SV40 origin replication to regulate the expression of the AAV rep and cap genes, and the AdV5 E2A and E4ORF6.
  • tetR tetracycline repressor-controlled
  • rtTA reverse tetracycline-controlled transactivator
  • binding of doxycycline to t-REx or rtTA will initiate transcription from the tetO-containing promoters or SV40 T-antigen gene, leading to expression of the AAV rep and cap genes, and AdV5, E2A, and E4ORF6 genes (FIG. 1B).
  • the construct shown in FIG.1B is permanently integrated into a E1a/E1b containing cell line (e.g., without limitations, an HEK293 cell line stably expressing tetR gene).
  • Transfection in media with an inducer (e.g., without limitation, doxycycline) of inducible rep and cap genes results in high-level production of Rep and Cap proteins and AAV virion assembly.
  • producer cells can be generated from these packaging lines by the addition of a vector construct comprising a transgene (or a GOI) flanked by AAV ITRs that can excise, replicate, and be packaged into virions once rep and cap are expressed.
  • the transgene construct is included in the same plasmid as the donor DNA, in a “single plasmid” protocol.
  • methods for an AAV packaging and producer cell line are provided.
  • the AAV rep and cap genes are under control of an inducible promoter (e.g., t-Rex or rtTA), such that the rep and cap genes, the AAV vector DNA sequences, and the essential helper genes are encoded on a single donor plasmid flanked by mobile element enzyme recognition sequences (FIG.1B).
  • a transgene is encoded separately from the donor plasmid (see, e.g., FIG.1C).
  • the donor plasmid comprises a transgene such that a one-plasmid (e.g., without limitation, a minicircle) is created and used to generate a transgene-specific cell line (e.g., an E1A, E1B+ cell such as, without limitation, a HEK293 cell line) that is able to produce a clinical grade product for human gene therapy.
  • a transgene-specific cell line e.g., an E1A, E1B+ cell such as, without limitation, a HEK293 cell line
  • methods for making a packaging and producer cell line with a dual donor/helper system are provided.
  • inducible Rep and Cap genes (sometimes collectively referred to as “Rep/Cap genes”), and a transgene of interest are encoded by respective separate nucleic acids.
  • the dual donor/helper system includes, without limitation, a nucleic acid encoding inducible Rep and Cap genes, a nucleic acid (e.g., a donor expression vector, such as a helper AAV donor plasmid) encoding a transgene of interest, and a nucleic acid encoding an enzyme capable of performing targeted genomic integration (e.g., without limitation, a mobile element enzyme).
  • a method of making a viral particle packaging and producer cell line is provided.
  • the method comprises transfecting an E1A, E1B+ cell with: a first nucleic acid encoding an enzyme capable of performing targeted genomic integration; a second nucleic acid encoding a donor DNA comprising inducible viral replication (Rep) and Capsid (Cap) genes, adenoviral auxiliary genes selected from E2A, E4ORF6, and VA, and insulators (e.g., without limitation, HS4 or D4Z4), and comprising terminal ends recognized by the enzyme; and a third nucleic acid encoding a donor DNA comprising a transgene flanked by AAV inverted terminal repeats (ITRs), to thereby result in a transfected cell that expresses the transgene in association with the viral particle.
  • ITRs inverted terminal repeats
  • inducible Rep/Cap genes and a nucleic acid e.g., a donor expression vector, such as a helper AAV donor plasmid
  • a nucleic acid e.g., a donor expression vector, such as a helper AAV donor plasmid
  • the single donor/helper system includes, without limitation, a nucleic acid (e.g., a donor expression vector) encoding inducible Rep and Cap genes and a transgene of interest, and a nucleic acid encoding an enzyme capable of performing targeted genomic integration (e.g., without limitation, a mobile element enzyme).
  • a method of making a viral particle packaging and producer cell line comprises transfecting an E1A, E1B+ cell with a first nucleic acid encoding an enzyme capable of performing targeted genomic integration; and a second nucleic acid encoding viral packaging genes, viral helper genes, insulators (e.g., without limitation, HS4, D4Z4), and a transgene, and comprising terminal ends recognized by the enzyme, to thereby result in a transfected cell that expresses the transgene in association with a viral particle.
  • insulators e.g., without limitation, HS4, D4Z4
  • the E1A, E1B+ cell is, without limitation, HEK293, E1A, E1B+ engineered CHO-K1, or Sf9 cell line.
  • the viral helper genes comprise adenoviral auxiliary genes.
  • the adenoviral auxiliary genes are selected from one or more of E1A, E1B, E4, E2A, and VA of an AAV, optionally E1A and E1B.
  • the viral packaging genes comprise rep and cap genes of viral replication (Rep) and Capsid (Cap) proteins of an AAV.
  • the expression of the Rep and Cap proteins is controlled by an inducible promoter, optionally an antibiotic-dependent promoter.
  • the antibiotic-dependent promoter is tetracycline- or a variant thereof dependent promoter and a tetracycline repressor-controlled (tetR) or tetracycline-controlled transactivator (rtTA) system is used.
  • the antibiotic-dependent promoter is a coumermycin/novobiocin promoter, or a variant thereof.
  • the inducible viral replication (Rep) and Capsid (cap) genes are controlled by at least one of a tetO- containing P5 promoter, P19 promoter, SV40 promoter, P40 promoter, or a functional variant or fragment thereof.
  • the tetO-containing AAV2 P40 promoter comprises a nucleotide sequence of SEQ ID NO: 816, or a functional variant or fragment thereof, or sequence having at least about 70%, or at least about 80%, or at least about 85%, or at least about 90%, or at least about 95%, or at least about 97% identity thereto, or at least about 98% identity thereto, or at least about 99% identity thereto or at least about 10 mutations, or at least about 9 mutations, or at least about 8 mutations, or at least about 7 mutations, or at least about 6 mutations, or at least about 5 mutations, or at least about 4 mutations, or at least about 3 mutations, or at least about 2 mutations, or at least about 1 mutation.
  • the P40 intron is an AAV2 P40 intron.
  • the AAV2 P40 intron comprises a C276A substitution, or a substitution at position corresponding thereto relative to SEQ ID NO: 818.
  • the AAV2 P40 intron comprises one or more mutated translation start sites (ATGs), optionally wherein the translation start sites are mutated to one of CTG, ACG, and TTG.
  • the AAV2 P40 intron comprises substitutions at one or more positions A13, A32, T42, A61, A71, A89, A203, A246, A258, and T282, or one or more positions corresponding thereto, relative to SEQ ID NO: 818.
  • the AAV2 P40 intron comprises substitutions at one or more positions A13C, A32C, T42C, A61C, A71T, A89C, A203T, A246C, A258C, and T282C corresponding to SEQ ID NO: 818.
  • the AAV2 P40 intron comprises the nucleotide sequence of SEQ ID NO: 818, or a functional variant or fragment thereof, or sequence having at least about 70%, or at least about 80%, or at least about 85%, or at least about 90%, or at least about 95%, or at least about 97% identity thereto, or at least about 98% identity thereto, or at least about 99% identity thereto, or at least about 10 mutations, or at least about 9 mutations, or at least about 8 mutations, or at least about 7 mutations, or at least about 6 mutations, or at least about 5 mutations, or at least about 4 mutations, or at least about 3 mutations, or at least about 2 mutations, or at least about 1 mutation.
  • the AAV2 P40 intron comprises the nucleotide sequence of SEQ ID NO: 819, or a functional variant or fragment thereof, or sequence having at least about 70%, or at least about 80%, or at least about 85%, or at least about 90%, or at least about 95%, or at least about 97% identity thereto, or at least about 98% identity thereto, or at least about 99% identity thereto, or at least about 10 mutations, or at least about 9 mutations, or at least about 8 mutations, or at least about 7 mutations, or at least about 6 mutations, or at least about 5 mutations, or at least about 4 mutations, or at least about 3 mutations, or at least about 2 mutations, or at least about 1 mutation.
  • the AAV2 P40 intron comprises the nucleotide sequence of SEQ ID NO: 817, or a functional variant or fragment thereof, or sequence having at least about 70%, or at least about 80%, or at least about 85%, or at least about 90%, or at least about 95%, or at least about 97% identity thereto, or at least about 98% identity thereto, or at least about 99% identity thereto, or at least about 10 mutations, or at least about 9 mutations, or at least about 8 mutations, or at least about 7 mutations, or at least about 6 mutations, or at least about 5 mutations, or at least about 4 mutations, or at least about 3 mutations, or at least about 2 mutations, or at least about 1 mutation.
  • the modified TATA box element has the nucleotide sequence of TATATAA.
  • the tetO- containing AAV2 P40 promoter or a functional variant or fragment thereof, comprising a modified TATA box element and a modified AAV2 P40 intron comprises the nucleotide sequence of SEQ ID NO: 823, or a functional variant or fragment thereof, or sequence having at least about 70%, or at least about 80%, or at least about 85%, or at least about 90%, or at least about 95%, or at least about 97% identity thereto, or at least about 98% identity thereto, or at least about 99% identity thereto, or at least about 10 mutations, or at least about 9 mutations, or at least about 8 mutations, or at least about 7 mutations, or at least about 6 mutations, or at least about 5 mutations, or at least about 4 mutations, or at least about 3 mutations, or at least about 2 mutations, or at least about 1 mutation.
  • the tetO-containing P40 promoter is a tetO-containing AAV2 P40 promoter.
  • the AAV2 P40 promoter comprises the nucleotide sequence of SEQ ID NO: 820, or a functional variant or fragment thereof, or sequence having at least about 70%, or at least about 80%, or at least about 85%, or at least about 90%, or at least about 95%, or at least about 97% identity thereto, or at least about 98% identity thereto, or at least about 99% identity thereto, or at least about 10 mutations, or at least about 9 mutations, or at least about 8 mutations, or at least about 7 mutations, or at least about 6 mutations, or at least about 5 mutations, or at least about 4 mutations, or at least about 3 mutations, or at least about 2 mutations, or at least about 1 mutation.
  • the modified P40 intron is a AAV9 P40 intron.
  • the modified AAV9 P40 intron comprises the nucleotide sequence of SEQ ID NO: 821, or a functional variant or fragment thereof, or sequence having at least about 70%, or at least about 80%, or at least about 85%, or at least about 90%, or at least about 95%, or at least about 97% identity thereto, or at least about 98% identity thereto, or at least about 99% identity thereto, or at least about 10 mutations, or at least about 9 mutations, or at least about 8 mutations, or at least about 7 mutations, or at least about 6 mutations, or at least about 5 mutations, or at least about 4 mutations, or at least about 3 mutations, or at least about 2 mutations, or at least about 1 mutation.
  • the modified TATA box element has the nucleotide sequence of TATATAA.
  • the tetO-containing AAV2 P40 promoter or a functional variant or fragment thereof, comprising a modified TATA box element and a modified AAV9 P40 intron comprises the nucleotide sequence of SEQ ID NO: 822, or a functional variant or fragment thereof, or sequence having at least about 70%, or at least about 80%, or at least about 85%, or at least about 90%, or at least about 95%, or at least about 97% identity thereto, or at least about 98% identity thereto, or at least about 99% identity thereto, or at least about 10 mutations, or at least about 9 mutations, or at least about 8 mutations, or at least about 7 mutations, or at least about 6 mutations, or at least about 5 mutations, or at least about 4 mutations, or at least about 3 mutations, or at least about 2 mutations, or at least about 1 mutation.
  • the tetO-containing AAV2 P40 promoter or a functional variant or fragment thereof, comprising a modified TATA box element and a modified AAV9 P40 intron comprises a nucleotide sequence of SEQ ID NO: 824, or a functional variant or fragment thereof, or sequence having at least about 70%, or at least about 80%, or at least about 85%, or at least about 90%, or at least about 95%, or at least about 97% identity thereto, or at least about 98% identity thereto, or at least about 99% identity thereto, or at least about 10 mutations, or at least about 9 mutations, or at least about 8 mutations, or at least about 7 mutations, or at least about 6 mutations, or at least about 5 mutations, or at least about 4 mutations, or at least about 3 mutations, or at least about 2 mutations, or at least about 1 mutation.
  • the P40 promoter further comprises one or more enhancer elements.
  • the enhancer element comprises one or more cis-acting elements, optionally selected from an Sp1 binding site, GC rich sequence, GCGGAAC motif, TAATGARAT element, AP1 binding site, and CCAAT box element.
  • the enhancer element comprises about 1 to about 5 Sp1 binding sites, optionally about 1, or about 2, or about 3, or about 4, or about 5 Sp1 binding sites.
  • the enhancer element comprises about 1 or about 2 GC rich sequences.
  • the enhancer element is derived from an hCMV Enhancer Element-3.
  • the hCMV Enhancer Element-3 comprises the nucleotide sequence of SEQ ID NO: 830 or a functional variant or fragment thereof, or sequence having at least about 70%, or at least about 80%, or at least about 85%, or at least about 90%, or at least about 95%, or at least about 97% identity thereto, or at least about 98% identity thereto, or at least about 99% identity thereto, or at least about 10 mutations, or at least about 9 mutations, or at least about 8 mutations, or at least about 7 mutations, or at least about 6 mutations, or at least about 5 mutations, or at least about 4 mutations, or at least about 3 mutations, or at least about 2 mutations, or at least about 1 mutation.
  • the viral particle is an AAV of a suitable serotype.
  • the AAV is selected from AAV1, AAV2, AAV5, AAV6, AAV7, AAV8, and AAV9.
  • the AAV serotype is a natural serotype or a synthetically engineered serotype.
  • the AAV is AAV2.
  • the AAV is AAV9.
  • the viral particle is an AAV of any of the AAV serotypes. Today, 12 AAV serotypes are known, and more than 100 variants have been identified.
  • any one or more of the first, second, and third nucleic acids is encoded by a single nucleic acid.
  • the second nucleic acid and the third nucleic acid are included in a single expression vector.
  • the first and second nucleic acids are included in a single expression vector, and the third nucleic acid is included in an expression vector that is different from the expression vector including the first and second nucleic acids.
  • the single expression vector comprises a plasmid.
  • the plasmid comprises an AAV2 CAP under the control of the tetO-containing hCMV-derived enhancer element 3/AAV2 P40 promoter with a modified AAV2 P40 TATA element and modified AAV2 P40 intron_1 (a at position 276 substituted for C) and WPRE/modified ICP27 poly A followed by ICP0 promoter-Zeocin resistant gene-ICP0 poly A cassette flanked with SB ITRs.
  • the plasmid comprises the nucleotide sequence of SEQ ID NO: 825 or a functional variant or fragment thereof, or sequence having at least about 70%, or at least about 80%, or at least about 85%, or at least about 90%, or at least about 95%, or at least about 97% identity thereto, or at least about 98% identity thereto, or at least about 99% identity thereto, or at least about 10 mutations, or at least about 9 mutations, or at least about 8 mutations, or at least about 7 mutations, or at least about 6 mutations, or at least about 5 mutations, or at least about 4 mutations, or at least about 3 mutations, or at least about 2 mutations, or at least about 1 mutation.
  • the plasmid comprises an AAV2 CAP under the control of the tetO-containing hCMV enhancer element 3/AAV2 P40 promoter with a modified AAV2 P40 TATA element and MODIFIED AAV2 P40 intron_2 and WPRE/modified ICP27 poly A followed by ICP0 promoter-Zeocin resistant gene-ICP0 poly A cassette flanked with SB ITRs.
  • the plasmid comprises the nucleotide sequence of SEQ ID NO: 826 or a functional variant or fragment thereof, or sequence having at least about 70%, or at least about 80%, or at least about 85%, or at least about 90%, or at least about 95%, or at least about 97% identity thereto, or at least about 98% identity thereto, or at least about 99% identity thereto, or at least about 10 mutations, or at least about 9 mutations, or at least about 8 mutations, or at least about 7 mutations, or at least about 6 mutations, or at least about 5 mutations, or at least about 4 mutations, or at least about 3 mutations, or at least about 2 mutations, or at least about 1 mutation.
  • the plasmid comprises an engineered mobile element enzyme ITR containing plasmid that encodes 1) AAV2_REP 78/68 ORFs under the control of P5TO1 promoter, REP 52/40 under the control of P19 promoter, 2)Ad5 E2A_IRES_Ad5 E4 OR F6 under the control of the tetO-containing SV40 promoter, 3) VA RNAs under the control of HSV-2 ICP4 promoter, and 4) hygro-B resistant gene under the control of TK promoter.
  • ITR containing plasmid that encodes 1) AAV2_REP 78/68 ORFs under the control of P5TO1 promoter, REP 52/40 under the control of P19 promoter, 2)Ad5 E2A_IRES_Ad5 E4 OR F6 under the control of the tetO-containing SV40 promoter, 3) VA RNAs under the control of HSV-2 ICP4 promoter, and 4) hygro-
  • the plasmid comprises the nucleotide sequence of SEQ ID NO: 841 or a functional variant or fragment thereof, or sequence having at least about 70%, or at least about 80%, or at least about 85%, or at least about 90%, or at least about 95%, or at least about 97% identity thereto, or at least about 98% identity thereto, or at least about 99% identity thereto, or at least about 10 mutations, or at least about 9 mutations, or at least about 8 mutations, or at least about 7 mutations, or at least about 6 mutations, or at least about 5 mutations, or at least about 4 mutations, or at least about 3 mutations, or at least about 2 mutations, or at least about 1 mutation.
  • the plasmid comprises an engineered mobile element enzyme ITR containing plasmid that encodes 1) AAV2_REP78/68 ORFs under the control of P5TO1 promoter, REP 52/40 under the control of P19 promoter, and CAPs under the native P40 promoter, 2) Ad5 E2A_IRES_Ad5 E4 ORF6 under the control of the tetO-containing SV40 promoter, 3) VA RNAs under the control of HSV-1 ICP4 promoter, and 4) hygro-B resistant gene under the control of TK promoter.
  • ITR containing plasmid that encodes 1) AAV2_REP78/68 ORFs under the control of P5TO1 promoter, REP 52/40 under the control of P19 promoter, and CAPs under the native P40 promoter, 2) Ad5 E2A_IRES_Ad5 E4 ORF6 under the control of the tetO-containing SV40 promoter, 3) VA RNAs under
  • the plasmid comprises the nucleotide sequence of SEQ ID NO: 842, or a functional variant or fragment thereof, or sequence having at least about 70%, or at least about 80%, or at least about 85%, or at least about 90%, or at least about 95%, or at least about 97% identity thereto, or at least about 98% identity thereto, or at least about 99% identity thereto, or at least about 10 mutations, or at least about 9 mutations, or at least about 8 mutations, or at least about 7 mutations, or at least about 6 mutations, or at least about 5 mutations, or at least about 4 mutations, or at least about 3 mutations, or at least about 2 mutations, or at least about 1 mutation.
  • the plasmid comprises an engineered mobile element enzyme ITR containing plasmid that encodes 1) AAV2_REP78/68 ORFs under the control of P5TO1 promoter, REP 52/40 under the control of P19 promoter, 2) AAV2 CAPs under the tetO-containing P40 promoter with AAV9 P40 intron, 3) Ad5 E2A_IRES_Ad5 E4 ORF6 under the control of the tetO-containing SV40 promoter, 4) VA RNAs under the control of HSV-1 ICP4 promoter, and 5) hygro- B resistant gene under the control of TK promoter.
  • ITR containing plasmid that encodes 1) AAV2_REP78/68 ORFs under the control of P5TO1 promoter, REP 52/40 under the control of P19 promoter, 2) AAV2 CAPs under the tetO-containing P40 promoter with AAV9 P40 intron, 3) Ad5 E2A_
  • the plasmid comprises the nucleotide sequence of SEQ ID NO: 843, or a functional variant or fragment thereof, or sequence having at least about 70%, or at least about 80%, or at least about 85%, or at least about 90%, or at least about 95%, or at least about 97% identity thereto, or at least about 98% identity thereto, or at least about 99% identity thereto, or at least about 10 mutations, or at least about 9 mutations, or at least about 8 mutations, or at least about 7 mutations, or at least about 6 mutations, or at least about 5 mutations, or at least about 4 mutations, or at least about 3 mutations, or at least about 2 mutations, or at least about 1 mutation.
  • any one of the first, second, and third nucleic acids is RNA, optionally mRNA, optionally synthetic mRNA. In embodiments, any one of the first, second, and third nucleic acids is DNA, optionally plasmid DNA. In embodiments, any one of the first, second, and third nucleic acids is an expression vector, wherein the expression vector is optionally a plasmid.
  • the transfected cell generates using the method of making a viral particle packaging and producer cell line in accordance with embodiments of the present disclosure comprises a donor DNA comprising the transgene flanked by AAV inverted terminal repeats (ITRs).
  • the method further comprises culturing the transfected cell in a medium that expands a population of the transfected cells to create a stably transfected packaging and producer cell line.
  • the stably transfected producer cell line is capable of producing replication-deficient viral particles in association with the transgene.
  • the transfection comprises electroporation, nucleofection, lipofection, or calcium phosphate transfection.
  • the method is helper virus-free.
  • the cell is human embryonic kidney (HEK293), Chinese hamster ovary (CHO) E1A, E1B+ engineered CHO-K1, or Spodoptera frugiperda (Sf9) cell line, baby hamster kidney (BHK), vero cell.
  • the viral helper genes comprise adenoviral auxiliary genes.
  • the adenoviral auxiliary genes are selected from one or more of E1A, E1B, E4, E2A, and VA of an AAV, optionally E1A and E1B.
  • the viral packaging genes comprise rep and cap genes of viral replication (Rep) and Capsid (Cap) proteins of an AAV.
  • the expression of the Rep and Cap proteins is controlled by an inducible promoter.
  • the inducible promoter is an antibiotic-dependent promoter, optionally a tetracycline-dependent promoter or a variant thereof, or a cumate or coumermycin/novobiocin promoter or a variant thereof.
  • the viral particle is an AAV, and optionally wherein the AAV is selected from AAV1, AAV5, AAV2, AAV6, AAV7, AAV8, and AAV9.
  • any one or more of the first, second, and third nucleic acids is encoded by a single nucleic acid.
  • the second nucleic acid and the third nucleic acid are included in a single expression vector.
  • the first and second nucleic acids are included in a single expression vector, and the third nucleic acid is included in an expression vector that is different from the expression vector including the first and second nucleic acids.
  • the expression vector is or comprises a plasmid.
  • the plasmid comprises an AAV2 CAP under the control of the tetO-containing hCMV-derived enhancer element 3/AAV2 P40 promoter with a modified AAV2 P40 TATA element and modified AAV2 P40 intron_1 (a at position 276 substituted for C) and WPRE/modified ICP27 poly A followed by ICP0 promoter-Zeocin resistant gene-ICP0 poly A cassette flanked with SB ITRs.
  • the plasmid comprises the nucleotide sequence of SEQ ID NO: 825 or a functional variant or fragment thereof, or sequence having at least about 70%, or at least about 80%, or at least about 85%, or at least about 90%, or at least about 95%, or at least about 97% identity thereto, or at least about 98% identity thereto, or at least about 99% identity thereto, or at least about 10 mutations, or at least about 9 mutations, or at least about 8 mutations, or at least about 7 mutations, or at least about 6 mutations, or at least about 5 mutations, or at least about 4 mutations, or at least about 3 mutations, or at least about 2 mutations, or at least about 1 mutation.
  • the plasmid comprises an AAV2 CAP under the control of the tetO-containing hCMV enhancer element 3/AAV2 P40 promoter with a modified AAV2 P40 TATA element and MODIFIED AAV2 P40 intron_2 and WPRE/modified ICP27 poly A followed by ICP0 promoter-Zeocin resistant gene-ICP0 poly A cassette flanked with SB ITRs.
  • the plasmid comprises the nucleotide sequence of SEQ ID NO: 826 or a functional variant or fragment thereof, or sequence having at least about 70%, or at least about 80%, or at least about 85%, or at least about 90%, or at least about 95%, or at least about 97% identity thereto, or at least about 98% identity thereto, or at least about 99% identity thereto, or at least about 10 mutations, or at least about 9 mutations, or at least about 8 mutations, or at least about 7 mutations, or at least about 6 mutations, or at least about 5 mutations, or at least about 4 mutations, or at least about 3 mutations, or at least about 2 mutations, or at least about 1 mutation.
  • the plasmid comprises an engineered mobile element enzyme ITR containing plasmid that encodes 1) AAV2_REP 78/68 ORFs under the control of P5TO1 promoter, REP 52/40 under the control of P19 promoter, 2)Ad5 E2A_IRES_Ad5 E4 OR F6 under the control of the tetO-containing SV40 promoter, 3) VA RNAs under the control of HSV-1 ICP4 promoter, and 4) hygro-B resistant gene under the control of TK promoter.
  • ITR containing plasmid that encodes 1) AAV2_REP 78/68 ORFs under the control of P5TO1 promoter, REP 52/40 under the control of P19 promoter, 2)Ad5 E2A_IRES_Ad5 E4 OR F6 under the control of the tetO-containing SV40 promoter, 3) VA RNAs under the control of HSV-1 ICP4 promoter, and 4) hygro-
  • the plasmid comprises the nucleotide sequence of SEQ ID NO: 841 or a functional variant or fragment thereof, or sequence having at least about 70%, or at least about 80%, or at least about 85%, or at least about 90%, or at least about 95%, or at least about 97% identity thereto, or at least about 98% identity thereto, or at least about 99% identity thereto, or at least about 10 mutations, or at least about 9 mutations, or at least about 8 mutations, or at least about 7 mutations, or at least about 6 mutations, or at least about 5 mutations, or at least about 4 mutations, or at least about 3 mutations, or at least about 2 mutations, or at least about 1 mutation.
  • the plasmid comprises an engineered mobile element enzyme ITR containing plasmid that encodes 1) AAV2_REP78/68 ORFs under the control of P5TO1 promoter, REP 52/40 under the control of P19 promoter, and CAPs under the native P40 promoter, 2) Ad5 E2A_IRES_Ad5 E4 ORF6 under the control of the tetO-containing SV40 promoter, 3) VA RNAs under the control of HSV-1 ICP4 promoter, and 4) hygro-B resistant gene under the control of TK promoter.
  • ITR containing plasmid that encodes 1) AAV2_REP78/68 ORFs under the control of P5TO1 promoter, REP 52/40 under the control of P19 promoter, and CAPs under the native P40 promoter, 2) Ad5 E2A_IRES_Ad5 E4 ORF6 under the control of the tetO-containing SV40 promoter, 3) VA RNAs under
  • the plasmid comprises the nucleotide sequence of SEQ ID NO: 842, or a functional variant or fragment thereof, or sequence having at least about 70%, or at least about 80%, or at least about 85%, or at least about 90%, or at least about 95%, or at least about 97% identity thereto, or at least about 98% identity thereto, or at least about 99% identity thereto, or at least about 10 mutations, or at least about 9 mutations, or at least about 8 mutations, or at least about 7 mutations, or at least about 6 mutations, or at least about 5 mutations, or at least about 4 mutations, or at least about 3 mutations, or at least about 2 mutations, or at least about 1 mutation.
  • the plasmid comprises an engineered mobile element enzyme ITR containing plasmid that encodes 1) AAV2_REP78/68 ORFs under the control of P5TO1 promoter and REP 52/40 under the control of P19 promoter, 2) AAV2 CAPs under the tetO-containing P40 promoter with AAV9 P40 intron, 3) Ad5 E2A_IRES_Ad5 E4 ORF6 under the control of the tetO-containing SV40 promoter, 4) VA RNAs under the control of HSV-1 ICP4 promoter, and 5) hygro- B resistant gene under the control of TK promoter.
  • ITR containing plasmid that encodes 1) AAV2_REP78/68 ORFs under the control of P5TO1 promoter and REP 52/40 under the control of P19 promoter, 2) AAV2 CAPs under the tetO-containing P40 promoter with AAV9 P40 intron, 3) Ad5 E2A_
  • the plasmid comprises the nucleotide sequence of SEQ ID NO: 843, or a functional variant or fragment thereof, or sequence having at least about 70%, or at least about 80%, or at least about 85%, or at least about 90%, or at least about 95%, or at least about 97% identity thereto, or at least about 98% identity thereto, or at least about 99% identity thereto, or at least about 10 mutations, or at least about 9 mutations, or at least about 8 mutations, or at least about 7 mutations, or at least about 6 mutations, or at least about 5 mutations, or at least about 4 mutations, or at least about 3 mutations, or at least about 2 mutations, or at least about 1 mutation.
  • any one of the first, second, and third nucleic acids is or comprises RNA, optionally mRNA, optionally synthetic mRNA or modified mRNA. In embodiments, any one of the first, second, and third nucleic acids is DNA, optionally plasmid DNA. In embodiments, any one of the first, second, and third nucleic acids is an expression vector, wherein the expression vector is optionally a plasmid.
  • the transgene is flanked by AAV inverted terminal repeats (ITRs). In embodiments, the transgene is flanked by Sleeping beauty ITRs.
  • SEQ ID NO: 831 Sleeping Beauty LE ITR Sequence (IR/DR(L) Lmut44) (231 bp including TATA): 5’- tatacagttgaagtcggaagtttacatacacttaagttggagtcattaaaactcgtttttcaactactccacaaatttcttgttaacaaacaatagttttggcaagtcagttag gacatctactttgtgcatgacacaagtcatttttccaacaattgtttacagacagattatttcacttataattcactgtatcacaattccagtgggtcagaagtttacatacact aa– 3’
  • the transgene or transposon has a size of about 200,000 base pairs or less. In embodiments, the transgene or transposon has a size of at least 200,000 base pairs. In embodiments, the transgene or transposon has a size of about 200,000 base pairs. In embodiments, the transgene or transposon has a size of about 150,000 base pairs or less. In embodiments, the transgene or transposon has a size of at least 150,000 base pairs. In embodiments, the transgene or transposon has a size of about 150,000 base pairs. In embodiments, the transgene or transposon has a size of about 100,000 base pairs or less. In embodiments, the transgene or transposon has a size of at least 100,000 base pairs.
  • the transgene or transposon has a size of about 100,000 base pairs. In embodiments, the transgene or transposon has a size of about 75,000 base pairs or less. In embodiments, the transgene or transposon has a size of at least 75,000 base pairs. In embodiments, the transgene or transposon has a size of about 75,000 base pairs. In embodiments, the transgene or transposon has a size of about 50,000 base pairs or less. In embodiments, the transgene or transposon has a size of at least 50,000 base pairs. In embodiments, the transgene or transposon has a size of about 50,000 base pairs.
  • the method of the present disclosure further comprises culturing the transfected cell in a medium that expands a population of the transfected cells to create a stably transfected packaging and producer cell line.
  • the stably transfected producer cell line is capable of producing replication-deficient viral particles in association with the transgene.
  • the transfection comprises electroporation, nucleofection, lipofection, or calcium phosphate transfection.
  • the method is helper virus-free.
  • the enzyme capable of performing targeted genomic integration is a recombinase.
  • the recombinase is an integrase or a mobile element enzyme.
  • the enzyme is a mobile element enzyme.
  • the enzyme is derived from Bombyx mori, Xenopus tropicalis, Trichoplusia ni, Myotis lucifugus, Rhinolophus ferrumequinum, Rousettus aegyptiacus, Phyllostomus discolor, Myotis myotis, Pteropus vampyrus, Pipistrellus kuhlii, Molossus molossus, Pan troglodytes, or Homo sapiens.
  • the enzyme is an engineered version, including but not limited to hyperactive forms, of an enzyme derived from Bombyx mori, Xenopus tropicalis, Trichoplusia ni, Myotis lucifugus, Rhinolophus ferrumequinum, Rousettus aegyptiacus, Phyllostomus discolor, Myotis myotis, Pteropus vampyrus, Pipistrellus kuhlii, Molossus molossus, Pan troglodytes, or Homo sapiens.
  • an enzyme derived from Bombyx mori, Xenopus tropicalis, Trichoplusia ni, Myotis lucifugus, Rhinolophus ferrumequinum, Rousettus aegyptiacus, Phyllostomus discolor, Myotis myotis, Pteropus vampyrus, Pipistrellus kuhlii, Molos
  • the mobile element enzyme is from one or more of the Sleeping beauty, Tn1, Tn2, Tn3, Tn5, Tn7, Tn9, Tn10, Tn552, Tn903, Tn1000/Gamma-delta, Tn/O, tnsA, tnsB, tnsC, tniQ, IS10, ISS, IS911, Minos, piggyBac, Tol2, Mos1, Himar1, Hermes, Tol2, Minos, Tel, P-element, MuA, Ty1, Chapaev, transib, Tc1/mariner, or Tc3 donor DNA system, or biologically active fragments variants thereof, inclusive of hyperactive variants.
  • the mobile element enzyme has the amino acid sequence of SEQ ID NO: 1, or an amino acid sequence having at least about 80%, or an amino acid sequence having at least about 90%, or at least about 93%, or at least about 95%, or at least about 97%, or at least about 98%, or at least about 99% identity thereto.
  • SEQ ID NO: 1 MLT mobile element enzyme protein (amino acid sequence of a variant of the hyperactive mobile element enzyme with S at position 8 and C at position 13 (572 amino acids) MAQHSDYSDDEFCADKLSNYSCDSDLENASTSDEDSSDDEVMVRPRTLRRRRISSSSSDSESDIEGGREEWSHV DNPPVLEDFLGHQGLNTDAVINNIEDAVKLFIGDDFFEFLVEESNRYYNQNRNNFKLSKKSLKWKDITPQEMKK FLGLIVLMGQVRKDRRDDYWTTEPWTETPYFGKTMTRDRFRQIWKAWHFNNNADIVNESDRLCKVRPVLDYFVP KFINIYKPHQQLSLDEGIVPWRGRLFFRVYNAGKIVKYGILVRLLCESDTGYICNMEIYCGEGKRLLETIQTVV SPYTDSWYHIYMDNYYNSVANCEALMKNKFRICGTIRKNRGIPKDFQTISLKKGETKFIRKNDILL
  • the amino acid is a non-polar aliphatic amino acid, optionally a non-polar aliphatic amino acid optionally selected from G, A, V, L, I and P, optionally A.
  • the mobile element enzyme does not have additional residues at the C terminus relative to SEQ ID NO: 1.
  • the enzyme has one or more mutations which confer hyperactivity.
  • the enzyme has one or more amino acid substitutions selected from S8X 1 and/or C13X 2 , or positions corresponding thereto relative to SEQ ID NO: 1.
  • the enzyme has S8X 1 and/or C13X 2 substitutions, at positions corresponding thereto relative to SEQ ID NO: 1.
  • the enzyme has S8X 1 and C13X 2 substitutions, at positions corresponding thereto relative to SEQ ID NO: 1. In embodiments, the enzyme has S8X 1 substitution, at position corresponding thereto relative to SEQ ID NO: 1. In embodiments, the enzyme has C13X 2 substitution, at positions corresponding thereto relative to SEQ ID NO: 1. In embodiments, X 1 is selected from G, A, V, L, I, and P and X 2 is selected from K, R, and H. In embodiments, X 1 is P and X 2 is R. In embodiments, the enzyme of the present disclosure comprises an amino acid sequence of SEQ ID NO: 11.
  • the enzyme comprises at least one, at least five, at least seven, at least nine, or ten substitutions at positions corresponding to: 5, 8, 9, 10, 11, 14, 22, 36, 37, 54, 130, 239, 281, 282, 283, 284, 285, 294, 300, 310, 330, 331, 332, 333, 334, 335, 336, 337, 338, 339, 345, 375, 416, 427, 475, 481, 491, 520, and/or 561 of SEQ ID NO: 11.
  • the enzyme comprises one, two, three, four, five, six, seven, eight, nine, or ten substitutions at positions corresponding to: 5, 8, 9, 10, 11, 14, 22, 36, 37, 54, 130, 239, 281, 282, 283, 284, 285, 294, 300, 310, 330, 331, 332, 333, 334, 335, 336, 337, 338, 339, 345, 375, 416, 427, 475, 481, 491, 520, and/or 561 of SEQ ID NO: 11.
  • the enzyme comprises at least one, at least five, at least seven, at least nine, or ten substitutions selected from S5P, S8F, D9G, D10G, E11G, A14V, T22C, S36G, T37C, S54N, K130T, G239R, Y281A, C282A, G283A, E284A, G285A, T294A, T300A, N310A, G330A, T331A, I332A, R333A, K334A, N335A, R336A, G337A, I338A, P339A, I345V, T375G, D416A, R427H, D475G, M481V, P491Q, A520T, and A561T, wherein the positions are corresponding to positions of SEQ ID NO: 11.
  • the enzyme comprises one, two, three, four, five, six, seven, eight, nine, or ten substitutions selected from S5P, S8F, D9G, D10G, E11G, A14V, T22C, S36G, T37C, S54N, K130T, G239R, Y281A, C282A, G283A, E284A, G285A, T294A, T300A, N310A, G330A, T331A, I332A, R333A, K334A, N335A, R336A, G337A, I338A, P339A, I345V, T375G, D416A, R427H, D475G, M481V, P491Q, A520T, and A561T, wherein the positions are corresponding to positions of SEQ ID NO: 11.
  • the mobile element enzyme is an engineered mammalian mobile element enzyme. In embodiments, the mobile element enzyme is a mammal-derived, helper RNA mobile element enzyme. In embodiments, the mobile element enzyme is a mammal-derived, helper DNA mobile element enzyme. In embodiments, the enzyme is capable of inserting a donor DNA at a TA dinucleotide site. In embodiments, the enzyme is capable of inserting a donor DNA at a TTAA (SEQ ID NO: 440) tetranucleotide site.
  • TTAA SEQ ID NO: 440
  • the mobile element enzyme has gene cleavage activity (Exc+) and/or gene integration activity (Int+), and the mobile element enzyme having at least about 90% identity to the amino acid sequence of SEQ ID NO: 1, SEQ ID NO: 3, SEQ ID NO: 4, SEQ ID NO: 6, SEQ ID NO: 7, SEQ ID NO: 8, SEQ ID NO: 9, SEQ ID NO: 10, SEQ ID NO: 11 or SEQ ID NO: 430, or a nucleotide sequence encoding the same.
  • the mobile element enzyme has one or more mutations which confer hyperactivity.
  • the mobile element enzyme has an amino acid sequence of SEQ ID NO: 1, SEQ ID NO: 10, or SEQ ID NO: 11 or a functional equivalent thereof.
  • the mobile element enzyme has the nucleotide sequence having at least about 90% identity to SEQ ID NO: 5 or a codon-optimized form thereof.
  • the mobile element enzyme has an amino acid sequence having I83P and/or V118R mutation relative to the amino acid sequence of SEQ ID NO: 6 or a functional equivalent thereof. In embodiments, the mobile element enzyme has an amino acid sequence having S20P and/or A29R mutation relative to the amino acid sequence of SEQ ID NO: 7 or a functional equivalent thereof. In embodiments, the mobile element enzyme has an amino acid sequence having A12P and/or I28R mutation and/or R152K mutation relative to the amino acid sequence of SEQ ID NO: 9 or a functional equivalent thereof. In embodiments, the mobile element enzyme has an amino acid sequence having T4P and/or L13R mutation relative to the amino acid sequence of SEQ ID NO: 8 or a functional equivalent thereof.
  • the donor DNA is included in a vector comprising left and right end sequences recognized by the mobile element enzyme.
  • the end sequences are selected from MER, MER75A, MER75B, and MER85.
  • the end sequences are selected from nucleotide sequences of SEQ ID NO: 12, SEQ ID NO: 13, SEQ ID NO: 14, SEQ ID NO: 15, SEQ ID NO: 16, SEQ ID NO: 17, SEQ ID NO: 18, SEQ ID NO: 19, SEQ ID NO: 20, SEQ ID NO: 21, SEQ ID NO: 441, and SEQ ID NO: 22, or a nucleotide sequence having at least about 90% identity thereto.
  • SEQ ID NO: 12 Pteropus vampyrus Left End Sequence Sequence 381 bp.
  • the end sequences include at least one repeat from a nucleotide sequence having at least about 90% identity to the nucleotide sequence of SEQ ID NO: 12, and wherein the at least one repeat from the nucleotide sequence having at least about 90% identity to the nucleotide sequence of SEQ ID NO: 12 is positioned at the 5’ end of the donor DNA.
  • the end sequences include at least one repeat from a nucleotide sequence having at least about 90% identity to the nucleotide sequence of SEQ ID NO: 17, and wherein the at least one repeat from the nucleotide sequence having at least about 90% identity to the nucleotide sequence of SEQ ID NO: 17 is positioned at the 3’ end of the donor DNA.
  • the end sequences are optionally flanked by a TTAA (SEQ ID NO: 440) sequence.
  • the end sequences include at least one repeat from a nucleotide sequence having at least about 90% identity to the nucleotide sequence of SEQ ID NO: 13, and wherein the at least one repeat from the nucleotide sequence having at least about 90% identity to the nucleotide sequence of SEQ ID NO: 13 is positioned at the 5’ end of the donor DNA.
  • the end sequences include at least one repeat from a nucleotide sequence having at least about 90% identity to the nucleotide sequence of SEQ ID NO: 18, and wherein the at least one repeat from the nucleotide sequence having at least about 90% identity to the nucleotide sequence of SEQ ID NO: 18 is positioned at the 3’ end of the donor DNA.
  • the end sequences are optionally flanked by a TTAA (SEQ ID NO: 440) sequence.
  • the end sequences include at least one repeat from a nucleotide sequence having at least about 90% identity to the nucleotide sequence of SEQ ID NO: 14, wherein the at least one repeat from the nucleotide sequence having at least about 90% identity to the nucleotide sequence of SEQ ID NO: 14 is positioned at the 5’ end of the donor DNA.
  • the end sequences include at least one repeat from a nucleotide sequence having at least about 90% identity to the nucleotide sequence of SEQ ID NO: 19, wherein the at least one repeat from the nucleotide sequence having at least about 90% identity to the nucleotide sequence of SEQ ID NO: 19 is positioned at the 3’ end of the donor DNA.
  • the end sequences are optionally flanked by a TTAA (SEQ ID NO: 440) sequence.
  • the end sequences include at least one repeat from a nucleotide sequence having at least about 90% identity to the nucleotide sequence of SEQ ID NO: 15, wherein the at least one repeat from the nucleotide sequence having at least about 90% identity to the nucleotide sequence of SEQ ID NO: 15 is positioned at the 5’ end of the donor DNA.
  • end sequences include at least one repeat from a nucleotide sequence having at least about 90% identity to the nucleotide sequence of SEQ ID NO: 20, wherein the at least one repeat from the nucleotide sequence having at least about 90% identity to the nucleotide sequence of SEQ ID NO: 20 is positioned at the 3’ end of the donor DNA.
  • the end sequences are optionally flanked by a TTAA (SEQ ID NO: 440) sequence.
  • the end sequences include at least one repeat from a nucleotide sequence having at least about 90% identity to the nucleotide sequence of SEQ ID NO: 16, wherein the at least one repeat from the nucleotide sequence having at least about 90% identity to the nucleotide sequence of SEQ ID NO: 16 is positioned at the 5’ end of the donor DNA.
  • the end sequences include at least one repeat from a nucleotide sequence having at least about 90% identity to the nucleotide sequence of SEQ ID NO: 21 or SEQ ID NO: 441, wherein the at least one repeat from the nucleotide sequence having at least about 90% identity to the nucleotide sequence of SEQ ID NO: 21 or SEQ ID NO: 441 is positioned at the 3’ end of the donor DNA.
  • the mobile element enzyme is an engineered form of a mobile element enzyme reconstructed from Homo sapiens or a predecessor thereof.
  • the enzyme is in a monomeric or dimeric form.
  • the enzyme is in a multimeric form.
  • the method of the present disclosure provides an enzyme comprising (a) a targeting element, and (b) an enzyme that is capable of inserting the donor DNA comprising a transgene, optionally at a TA dinucleotide site or a TTAA (SEQ ID NO: 440) tetranucleotide site in a genomic safe harbor site (GSHS).
  • the donor DNA comprises a transgene encoding a complete polypeptide.
  • the donor DNA comprises a transgene which is defective or substantially absent in a disease state.
  • the enzyme has one or more mutations which confer hyperactivity.
  • the enzyme has gene cleavage (Exc) and/or gene integration activity (Int+).
  • the enzyme has gene cleavage (Exc) and/or a lack of gene integration activity (Int-).
  • the mobile element enzyme is a chimeric mobile element enzyme.
  • the targeting element comprises one or more of a gRNA, optionally associated with a Cas enzyme, which is optionally catalytically inactive, transcription activator-like effector (TALE), catalytically inactive Zinc finger, catalytically inactive transcription factor, nickase, a transcriptional activator, a transcriptional repressor, a recombinase, a DNA methyltransferase, a histone methyltransferase, a paternally expressed gene 10 (PEG10), and a TnsD.
  • TALE transcription activator-like effector
  • the targeting element comprises a transcription activator-like effector (TALE) DNA binding domain (DBD).
  • TALE DBD comprises one or more repeat sequences.
  • the TALE DBD comprises about 14, or about 15, or about, 16, or about 17, or about 18, or about 18.5 repeat sequences.
  • the TALE DBD repeat sequences comprise 33 or 34 amino acids.
  • the one or more of the TALE DBD repeat sequences comprise a repeat variable di-residue (RVD) at residue 12 or 13 of the 33 or 34 amino acids.
  • RVD recognizes one base pair in the nucleic acid molecule.
  • the RVD recognizes a C residue in the nucleic acid molecule and is selected from HD, N(gap), HA, ND, and HI. In embodiments, the RVD recognizes a G residue in the nucleic acid molecule and is selected from NN, NH, NK, HN, and NA. In embodiments, the RVD recognizes an A residue in the nucleic acid molecule and is selected from NI and NS. In embodiments, the RVD recognizes a T residue in the nucleic acid molecule and is selected from NG, HG, H(gap), and IG. In embodiments, the GSHS is in an open chromatin location in a chromosome.
  • the GSHS is selected from adeno-associated virus site 1 (AAVS1), chemokine (C-C motif) receptor 5 (CCR5) gene, HIV-1 coreceptor, and human Rosa26 locus.
  • AAVS1 adeno-associated virus site 1
  • CCR5 chemokine receptor 5
  • HIV-1 coreceptor HIV-1 coreceptor
  • Rosa26 locus human Rosa26 locus.
  • the GSHS is located on human chromosome 2, 4, 6, 10, 11, 17, 22, or X.
  • the GSHS is selected from TALC1, TALC2, TALC3, TALC4, TALC5, TALC7, TALC8, AVS1, AVS2, AVS3, ROSA1, ROSA2, TALER1, TALER2, TALER3, TALER4, TALER5, SHCHR2-1, SHCHR2-2, SHCHR2-3, SHCHR2-4, SHCHR4-1, SHCHR4-2, SHCHR4-3, SHCHR6-1, SHCHR6-2, SHCHR6-3, SHCHR6-4, SHCHR10-1, SHCHR10-2, SHCHR10-3, SHCHR10-4, SHCHR10-5, SHCHR11-1, SHCHR11-2, SHCHR11-3, SHCHR17-1, SHCHR17-2, SHCHR17-3, and SHCHR17-4.
  • the targeting element comprises a Cas9 enzyme guide RNA complex.
  • the Cas9 enzyme guide RNA complex comprises a nuclease-deficient dCas9 guide RNA complex.
  • the targeting element comprises a Cas12 enzyme guide RNA complex or wherein the targeting element comprises a nuclease- deficient dCas12 guide RNA complex, optionally dCas12j guide RNA complex or dCas12a guide RNA complex.
  • the targeting element comprises: a gRNA of or comprising a sequence of TABLE 3A-3F, or a variant thereof; or a TALE DBD of or comprising a sequence of TABLE 4A-4F, or a variant thereof; or a ZNF of or comprising a sequence of TABLE 5A-5E, or a variant thereof.
  • the targeting element is or comprises a nucleic acid binding component of the gene-editing system.
  • the enzyme and the targeting element are connected.
  • the enzyme and the targeting element are fused to one another or linked via a linker to one another.
  • the linker is a flexible linker.
  • the flexible linker is substantially comprised of glycine and serine residues, optionally wherein the flexible linker comprises (Gly 4 Ser) n , where n is from about 1 to about 12. In embodiments, the flexible linker is of about 20, or about 30, or about 40, or about 50, or about 60 amino acid residues.
  • the donor DNA comprises a gene encoding a complete polypeptide. In embodiments, the donor DNA comprises a gene which is defective or substantially absent in a disease state. In embodiments, the donor DNA is flanked by one or more inverted terminal ends. In embodiments, at least one of the first nucleic acid and the second nucleic acid is in the form of a lipid nanoparticle (LNP).
  • LNP lipid nanoparticle
  • the first nucleic acid encoding the enzyme and the second nucleic acid encoding the donor DNA are in the form of the same LNP, optionally in a co-formulation.
  • the LNP comprises one or more lipids selected from 1,2-dioleoyl-3-trimethylammonium propane (DOTAP), a cationic cholesterol derivative mixed with dimethylaminoethane-carbamoyl (DC-Chol), phosphatidylcholine (PC), triolein (glyceryl trioleate), and 1,2-distearoyl- sn-glycero-3-phosphoethanolamine-N-[carboxy(polyethylene glycol)-2000] (DSPE-PEG), 1,2-dimyristoyl-rac-glycero- 3-methoxypolyethyleneglycol – 2000 (DMG-PEG 2K), and 1,2 distearol -sn-glycerol-3phosphocholine (DSPC) and/or comprising of one or
  • DOTAP
  • the enzyme is encoded by a recombinant or synthetic nucleic acid.
  • the nucleic acid is mRNA or a helper RNA.
  • the nucleic acid is RNA that has a 5’-m7G cap (cap0, cap1, or cap2) with pseudouridinesubstitution, and a poly-A tail of about 30, or about 50, or about 100, of about 150 nucleotides in length.
  • the enzyme is incorporated into a vector or a vector-like particle.
  • the vector is a non- viral vector.
  • the enzyme and the donor DNA are included in the same vector. In embodiments, the enzyme and the donor DNA are included in different vectors.
  • the enzyme and the donor DNA are included in a single pharmaceutical composition. In embodiments, the enzyme and the donor DNA are included in different pharmaceutical compositions. In embodiments, the enzyme and the donor DNA are co- administered. In embodiments, the enzyme and the donor DNA are administered separately.
  • the present disclosure provides a method of producing an AAV bearing a gene of interest, comprising employing a method of the present disclosure to produce the AAV bearing the gene of interest. In embodiments, the present disclosure provides a cell for gene therapy, generated by a method of the present disclosure. In embodiments, the present disclosure provides a method of delivering a cell therapy, comprising administering to a patient in need thereof the transfected cell generated by a method of the present disclosure.
  • the present disclosure provides a method of treating a disease or condition using a cell therapy, comprising administering to a patient in need thereof the transfected cell generated by a method of the present disclosure.
  • the present disclosure provides a method of treating a disease or condition using a biologic, e.g., antibody, therapy, comprising administering to a patient in need thereof the transfected cell generated by a method of the present disclosure.
  • a biologic e.g., antibody
  • the enzyme capable of targeted genomic integration is any type of an enzyme that cause a transgene to be inserted from one location (e.g., without limitation, donor DNA) to a specific site and/or locus in a subject’s genome.
  • the enzyme capable of targeted genomic integration is a recombinase.
  • the recombinase is an integrase.
  • the enzyme is a mobile element enzyme.
  • the recombinase is an integrase or a mobile element enzyme.
  • the mobile element enzyme is an engineered mammalian mobile element enzyme.
  • the mobile element enzyme is a mammal-derived, helper RNA mobile element enzyme.
  • Messenger RNA (mRNA) is an effective alternative to DNA as a source of a mobile element enzyme for targeting somatic cells and tissues, given that RNA is a safer alternative to DNA as a source of a mobile element enzyme for somatic gene therapy applications.
  • the mobile element enzyme is a mammal-derived, DNA mobile element enzyme. In embodiments, the mobile element enzyme is a chimeric mobile element enzyme.
  • the enzyme capable of targeted genomic integration is a mobile element enzyme
  • the mobile element enzyme comprises (a) a targeting element that is or comprises a gene-editing system, and (b) a mobile element enzyme that is capable of inserting the donor DNA (e.g., mobile element) comprising a transgene at a TA dinucleotide site or a TTAA (SEQ ID NO: 440) tetranucleotide site in a GSHS, as described elsewhere herein.
  • the enzyme is derived from Bombyx mori, Xenopus tropicalis, Trichoplusia ni, Myotis lucifugus, Rhinolophus ferrumequinum, Rousettus aegyptiacus, Phyllostomus discolor, Myotis myotis, Pteropus vampyrus, Pipistrellus kuhlii, Molossus molossus, Pan troglodytes, or Homo sapiens.
  • the enzyme is an engineered version, including but not limited to hyperactive forms, of an enzyme derived from Bombyx mori, Xenopus tropicalis, Trichoplusia ni, Myotis lucifugus, Rhinolophus ferrumequinum, Rousettus aegyptiacus, Phyllostomus discolor, Myotis myotis, Pteropus vampyrus, Pipistrellus kuhlii, Molossus molossus, Pan troglodytes, or Homo sapiens.
  • an enzyme derived from Bombyx mori, Xenopus tropicalis, Trichoplusia ni, Myotis lucifugus, Rhinolophus ferrumequinum, Rousettus aegyptiacus, Phyllostomus discolor, Myotis myotis, Pteropus vampyrus, Pipistrellus kuhlii, Molos
  • the enzyme is a mobile element enzyme derived from Bombyx mori, Xenopus tropicalis, Trichoplusia ni, Myotis lucifugus, Rhinolophus ferrumequinum, Rousettus aegyptiacus, Phyllostomus discolor, Myotis myotis, Pteropus vampyrus, Pipistrellus kuhlii, Molossus molossus, Pan troglodytes, or Homo sapiens.
  • the enzyme is an engineered version, including but not limited to hyperactive forms, of a mobile element enzyme derived from Bombyx mori, Xenopus tropicalis, Trichoplusia ni, Myotis lucifugus, Rhinolophus ferrumequinum, Rousettus aegyptiacus, Phyllostomus discolor, Myotis myotis, Pteropus vampyrus, Pipistrellus kuhlii, Molossus molossus, Pan troglodytes, or Homo sapiens.
  • a mobile element enzyme derived from Bombyx mori, Xenopus tropicalis, Trichoplusia ni, Myotis lucifugus, Rhinolophus ferrumequinum, Rousettus aegyptiacus, Phyllostomus discolor, Myotis myotis, Pteropus vampyrus, Pipistrellus kuh
  • the mobile element enzyme is from one or more of the Tn1, Tn2, Tn3, Tn5, Tn7, Tn9, Tn10, Tn552, Tn903, Tn1000/Gamma-delta, Tn/O, tnsA, tnsB, tnsC, tniQ, IS10, ISS, IS911, Minos, Sleeping beauty, piggyBac, Tol2, Mos1, Himar1, Hermes, Tol2, Minos, Tel, P-element, MuA, Ty1, Chapaev, transib, Tc1/mariner, or Tc3 donor DNA system, or biologically active fragments variants thereof, inclusive of hyperactive mutants (e.g., without limitation selected from TABLE 1, or equivalents thereof).
  • the mobile element enzyme is from a MLT donor DNA system that is based on a cut-and-paste MLT element obtained from the little brown bat (Myotis lucifugus) or other bat mobile element enzymes, such as Rhinolophus ferrumequinum, Rousettus aegyptiacus, Phyllostomus discolor, Myotis myotis, Pipistrellus kuhlii, and Molossus molossus.
  • MLT donor DNA system that is based on a cut-and-paste MLT element obtained from the little brown bat (Myotis lucifugus) or other bat mobile element enzymes, such as Rhinolophus ferrumequinum, Rousettus aegyptiacus, Phyllostomus discolor, Myotis myotis, Pipistrellus kuhlii, and Molossus molossus.
  • hyperactive forms of a bat mobile element enzyme are used.
  • the MLT mobile element enzyme has been shown to be capable of transposition in bat, human, mammalian, and yeast cells.
  • the hyperactive forms of the MLT mobile element enzyme enhance the transposition process.
  • chimeric MLT mobile element enzymes are capable of site-specific excision without genomic integration.
  • the mobile element enzyme is a Myotis lucifugus mobile element enzyme (MLT), which is either the wild type, monomer, dimer, tetramer (or another multimer), hyperactive, an Int-mutant, or of any other form.
  • MMT Myotis lucifugus mobile element enzyme
  • the MLT mobile element enzyme has an amino acid sequence of SEQ ID NO: 1, or a variant having at least about 80%, at least about 90%, or at least about 93%, or at least about 95%, or at least about 97%, or at least about 98%, or at least about 99% identity thereto, and one or more mutations selected from L573X, E574X, and S2X, wherein X is any amino acid or no amino acid, optionally X is A, G, or a deletion, optionally the mutations are L573del E574del, and S2A).
  • the MLT mobile element enzyme has the nucleotide sequence of SEQ ID NO: 2 (which is a codon-optimized form of MLT), or a nucleotide sequence having at least about 90%, or at least about 93%, or at least about 95%, or at least about 97%, or at least about 98%, or at least about 99% identity thereto.
  • SEQ ID NO: 1 is: MAQHSDYSDDEFCADKLSNYSCDSDLENASTSDEDSSDDEVMVRPRTLRRRRISSSSSDSESDIEGGREEWSHV DNPPVLEDFLGHQGLNTDAVINNIEDAVKLFIGDDFFEFLVEESNRYYNQNRNNFKLSKKSLKWKDITPQEMKK FLGLIVLMGQVRKDRRDDYWTTEPWTETPYFGKTMTRDRFRQIWKAWHFNNNADIVNESDRLCKVRPVLDYFVP KFINIYKPHQQLSLDEGIVPWRGRLFFRVYNAGKIVKYGILVRLLCESDTGYICNMEIYCGEGKRLLETIQTVV SPYTDSWYHIYMDNYYNSVANCEALMKNKFRICGTIRKNRGIPKDFQTISLKKGETKFIRKNDILLQVWQSKKP VYLISSIHSAEMEESQNIDRTSKKKIVKPNALIDYNKHMKGVDRADQYLS
  • the amino acid is a non-polar aliphatic amino acid, optionally a non-polar aliphatic amino acid optionally selected from G, A, V, L, I and P, optionally A.
  • the mobile element enzyme does not have additional residues at the C terminus relative to SEQ ID NO: 1.
  • the MLT mobile element enzyme has an amino acid sequence of SEQ ID NO: 1 or a variant having at least about 80%, at least about 90%, or at least about 93%, or at least about 95%, or at least about 97%, or at least about 98%, or at least about 99% identity thereto and comprises an alanine at the position corresponding to position 2 of SEQ ID NO: 1.
  • the MLT mobile element enzyme has an amino acid sequence of SEQ ID NO: 1 or a variant having at least about 80%, at least about 90%, or at least about 93%, or at least about 95%, or at least about 97%, or at least about 98%, or at least about 99% identity thereto and comprises an alanine at the position corresponding to position 2 of SEQ ID NO: 1 and no additional amino acids at the C terminal end.
  • the MLT mobile element enzyme has a nucleotide sequence of SEQ ID NO: 2 (which is codon- optimized) and an amino acid sequence SEQ ID NO: 1, respectively.
  • the MLT mobile element enzyme has a nucleotide sequence of SEQ ID NO: 2, or a nucleotide sequence having at least about 90%, or at least about 93%, or at least about 95%, or at least about 97%, or at least about 98%, or at least about 99% identity thereto, or a codon-optimized form thereof.
  • the MLT mobile element enzyme has an amino acid sequence SEQ ID NO: 1, or an amino acid sequence having at least about 90%, or at least about 93%, or at least about 95%, or at least about 97%, or at least about 98%, or at least about 99% identity thereto.
  • the mobile element enzyme can act on an MLT left terminal end, or a sequence having at least about 90%, or at least about 93%, or at least about 95%, or at least about 97%, or at least about 98%, or at least about 99% identity thereto, wherein the nucleotide sequence of the MLT left terminal end (5’ to 3’) is as follows: ttaacacttggattgcgggaaacgagttaagtcggctcgcgtgaattgcgcgcgtactccgcgggagccgtcttaactcggttcatatag atttgcggtggagtgcgggaaacgtgtaaactcgggccgattgtaactgcgtattaccaaatatttgtt (SEQ ID NO: 21)
  • the mobile element enzyme can act on an MLT right terminal end, or a sequence having at least about 90%, or at least about
  • the donor DNA is or comprises a gene encoding a compete polypeptide. In embodiments, the donor DNA is or comprises a gene which is defective or substantially absent in a disease state.
  • the enzyme e.g., without limitation, a mobile element enzyme, e.g., without limitation, MLT mobile element enzyme
  • the enzyme has one or more mutations which confer hyperactivity.
  • the enzyme e.g., without limitation, a mobile element enzyme, e.g., without limitation, MLT mobile element enzyme
  • the enzyme e.g., without limitation, a mobile element enzyme, e.g., without limitation, MLT mobile element enzyme
  • the mobile element enzyme e.g., without limitation, MLT mobile element enzyme includes a hyperactive mutation, e.g., about 1, or about 2, or about 3, or about 4, or about 5 hyperactive mutations or combinations thereof.
  • the mobile element enzyme can include any number of any of the hyperactive mutations, or equivalents thereof, described herein.
  • the MLT mobile element enzyme includes a hyperactive mutation, e.g., about 1, or about 2, or about 3, or about 4, or about 5 hyperactive mutations, or combinations thereof.
  • the mobile element enzyme can include any number of any of the hyperactive mutations, or equivalents thereof, described herein.
  • the enzyme comprises one or more mutations corresponding to TABLE 1, which, without being bound by theory, provides hyperactive mutations. Numbering relative to the amino acid sequence of protein of SEQ ID NO: 1, and nucleic acid sequence of SEQ ID NO: 2.
  • the MLT mobile element enzyme has one or more amino acid substitutions selected from S8X1, C13X2 and/or N125X3, or positions corresponding thereto, relative to SEQ ID NO: 1, wherein X1 is selected from G, A, V, L, I and P, X2 is selected from K, R, and H, and X3 is selected from K, R, and H, or wherein: X1 is P, X2 is R, and/or X3 is K.
  • the MLT mobile element enzyme has S8X1, C13X2 and N125X3 substitutions, at positions corresponding to SEQ ID NO: 1, wherein X1 is selected from G, A, V, L, I and P, X2 is selected from K, R, and H, and X3 is selected from K, R, and H, or wherein: X1 is P, X2 is R, and/or X3 is K.
  • the MLT mobile element enzyme has S8X1 and C13X2 substitutions, at positions corresponding to SEQ ID NO: 1, wherein X1 is selected from G, A, V, L, I and P, X2 is selected from K, R, and H, and X3 is selected from K, R, and H, or wherein: X1 is P, X2 is R, and/or X3 is K.
  • the MLT mobile element enzyme has S8X1 and N125X3 substitutions, at positions corresponding to SEQ ID NO: 1, wherein X1 is selected from G, A, V, L, I and P, X2 is selected from K, R, and H, and X3 is selected from K, R, and H, or wherein: X1 is P, X2 is R, and/or X3 is K.
  • the MLT mobile element enzyme has C13X2 and N125X3 substitutions, at positions corresponding to SEQ ID NO: 1, wherein X1 is selected from G, A, V, L, I and P, X2 is selected from K, R, and H, and X3 is selected from K, R, and H, or wherein: X1 is P, X2 is R, and/or X3 is K.
  • the MLT mobile element enzyme has an amino acid sequence of SEQ ID NO: 1, or a variant thereof, and S8P and C13R mutations (SEQ ID NO: 11).
  • the MLT mobile element enzyme has an amino acid sequence having mutations at positions which correspond to at least one of S8P and C13R mutations relative to the amino acid of SEQ ID NO: 1 or a functional equivalent thereof. In embodiments, the MLT mobile element enzyme has an amino acid sequence having mutations at positions which correspond to S8P and C13R mutations relative to the amino acid of SEQ ID NO: 1 or a functional equivalent thereof. In embodiments, the MLT mobile element enzyme has an amino acid sequence of SEQ ID NO: 1, or a variant thereof, and S8P, C13R, and N125K mutations (SEQ ID NO: 10).
  • a MLT mobile element enzyme comprising the amino acid sequence of SEQ ID NO: 1, or a variant thereof, and includes one or more hyperactive mutations selected from a substitution or deletion at one or more of positions S5, S8, D9, D10, E11, C13, A14, S36, S54, N125, K130, G239, T294, T300, I345, R427, D475, M481, P491, A520, and A561, or positions corresponding thereto.
  • a MLT mobile element enzyme comprising the amino acid sequence of SEQ ID NO: 1, or a variant thereof, and includes one or more hyperactive mutations selected from S5P, S8P, S8P/C13R, D9G, D10G, E11G, C13R, A14V, S36G, S54N, N125K, K130T, G239S, T294A, T300A, I345V, R427H, D475G, M481V, P491Q, A520T, and A561T, or positions corresponding thereto.
  • the MLT mobile element enzyme comprises one or more of hyperactive mutants selected from S8X 1 , C13X 2 and/or N125X 3 (e.g., all of S8X 1 , C13X 2 and N125X 3 , S8X 1 and C13X 2 , S8X 1 and N125X 3 , and C13X 2 and N125X 3 ), where X 1 , X 2 , and X 3 is each independently any amino acid, or X 1 is a non-polar aliphatic amino acid, selected from G, A, V, L, I and P, X 2 is a positively charged amino acid selected from K, R, and H, and/or X 3 is a positively charged amino acid selected from K, R, and H.
  • S8X 1 , C13X 2 and/or N125X 3 e.g., all of S8X 1 , C13X 2 and N125X 3 , S8X 1 and C13X 2 , S8X 1 and N125X 3 , and
  • X 1 is P
  • X 2 is R
  • X 3 is K
  • the enzyme e.g., without limitation, a mobile element enzyme, e.g., without limitation, MLT mobile element enzyme
  • the enzyme has gene cleavage activity (Exc+) and/or gene integration activity (Int+).
  • the enzyme e.g., without limitation, a mobile element enzyme
  • the MLT mobile element enzyme has gene cleavage activity (Exc+) and/or gene integration activity (Int+).
  • the MLT mobile element enzyme has gene cleavage activity (Exc+) and/or a lack of gene integration activity (Int-).
  • the mobile element enzyme e.g., without limitation, MLT mobile element enzyme includes an integration reduced or deficient mutation, e.g., about 1, or about 2, or about 3, or about 4, or about 5 integration reduced or deficient mutations or combinations thereof.
  • the mobile element enzyme can include any number of any of the integration reduced or deficient mutations, or equivalents thereof, described herein.
  • the MLT mobile element enzyme includes a integration reduced or deficient mutations, e.g. about 1, or about 2, or about 3, or about 4, or about 5 integration reduced or deficient mutations, or combinations thereof.
  • the mobile element enzyme can include any number of any of the integration reduced or deficient mutations, or equivalents thereof, described herein.
  • the enzyme comprises one or more mutations corresponding to TABLE 2A, or positions corresponding thereto, which, without being bound by theory, provides integration reduced or deficient mutations. Numbering relative to the amino acid sequence of protein of SEQ ID NO: 1.
  • TABLE 2A In embodiments, the enzyme comprises one or more mutations corresponding to TABLE 2B, or positions corresponding thereto, which, without being bound by theory, provides excision positive and integration deficient mutations. Numbering relative to the amino acid sequence of protein of SEQ ID NO: 1, and nucleic acid sequence of SEQ ID NO: 2.
  • a MLT mobile element enzyme comprising the amino acid sequence of SEQ ID NO: 1, or a variant thereof, and includes one or more mutations selected from S8P and/or C13R and one of R164N, W168V, M278A, K286A, R287A, R333A, K334A, N335A, K349A, K350A, K368A, K369A, and D416N, or positions corresponding thereto.
  • a MLT mobile element enzyme comprising the amino acid sequence of SEQ ID NO: 1, or a variant thereof, and includes one or more mutations selected from S8P and/or C13R and one of R164N, W168V, M278A, K286A, R287A, R333A, K334A, N335A, K349A, K350A, K368A, K369A, and D416N and/or one or more of E284A, K286A, R287A, N310A, R333A, K334A, R336A, K349A, K350A, K368A, and K369A, or positions corresponding thereto.
  • a MLT mobile element enzyme comprising the amino acid sequence of SEQ ID NO: 1, or a variant thereof, and includes one or more mutations selected from S8P and/or C13R and one of R164N, W168V, M278A, K286A, R287A, R333A, K334A, N335A, K349A, K350A, K368A, K369A, and D416N and/or one or more of E284A, K286A, R287A, N310A, R333A, K334A, R336A, K349A, K350A, K368A, and K369A and/or one R336A, or positions corresponding thereto.
  • the mobile element enzyme is or is derived from any of Bombyx mori, Xenopus tropicalis, Trichoplusia ni, Rhinolophus ferrumequinum, Rousettus aegyptiacus, Phyllostomus discolor, Myotis myotis, Myotis lucifugus, Pipistrellus kuhlii, Pteropus vampyrus, and Molossus molossus.
  • the mobile element enzyme is or is derived from any of Trichoplusia ni (SEQ ID NO: 433), Myotis myotis (SEQ ID NO: 435, SEQ ID NO: 436, SEQ ID NO: 438, or SEQ ID NO: 439), or Pteropus vampyrus (SEQ ID NO: 434).
  • the mobile element enzymes have one or more hyperactive and/or integration deficient mutations selected from TABLE 1, TABLE 2A, and/or TABLE 2B, or equivalents thereof.
  • Trichoplusia ni SEQ ID NO: 433
  • Myotis lucifugus SEQ ID NO: 437
  • Myotis myotis SEQ ID NO: 435
  • SEQ ID NO: 436 SEQ ID NO: 438
  • SEQ ID NO: 439 Pteropus vampyrus
  • Pteropus vampyrus SEQ ID NO: 434
  • Trichnoplusia ni 1 MGSSLDDEHI LSALLQSDDE LVGEDSDSEI SDHVSEDDVQ SDTEEAFIDE VHEVQPTSSG 61
  • the mobile element enzyme is an engineered version of a mobile element enzyme, including but not limited to monomers, dimers, tetramers, hyperactive, or Int-forms, derived from Bombyx mori, Xenopus tropicalis, or Trichoplusia ni. In embodiments, the mobile element enzyme is derived from Bombyx mori, Xenopus tropicalis, Trichoplusia ni, or Myotis lucifugus.
  • the mobile element enzyme is an engineered version, including but not limited to a mobile element enzyme that is a monomer, dimer, tetramer (or another multimer), hyperactive, or has a reduced interaction with non-TTAA (SEQ ID NO: 440) recognitions sites (Int-), derived from Bombyx mori, Xenopus tropicalis, Trichoplusia ni or Myotis lucifugus.
  • the mobile element enzymes have one or more hyperactive and/or integration deficient mutations selected from TABLE 1, TABLE 2A, and TABLE 2B, or equivalents thereof.
  • one skilled in the art can correspond such mutants to mobile element enzymes from any of Bombyx mori, Xenopus tropicalis, Trichoplusia ni, Rhinolophus ferrumequinum, Rousettus aegyptiacus, Phyllostomus discolor, Myotis myotis, Myotis lucifugus, Pipistrellus kuhlii, Pteropus vampyrus, Pan troglodytes, and Molossus molossus.
  • the mobile element enzyme has a nucleotide sequence having at least about 90%, or at least about 93%, or at least about 95%, or at least about 97%, or at least about 98%, or at least about 99% identity to a nucleotide sequence of any of Rhinolophus ferrumequinum, Rousettus aegyptiacus, Phyllostomus discolor, Myotis myotis, Myotis lucifugus, Pteropus vampyrus, Pipistrellus kuhliim, Pan troglodytes, and Molossus molossus.
  • the mobile element enzyme has an amino acid sequence having at least about 90%, or at least about 93%, or at least about 95%, or at least about 97%, or at least about 98%, or at least about 99% identity to an amino acid sequence of any of Rhinolophus ferrumequinum, Rousettus aegyptiacus, Phyllostomus discolor, Myotis myotis, Myotis lucifugus, Pteropus vampyrus, Pipistrellus kuhlii, and Molossus molossus. See Jebb, et al. (2020).
  • the enzyme e.g., without limitation, a mobile element enzyme
  • the enzyme is derived from Bombyx mori, Xenopus tropicalis, Trichoplusia ni, Myotis lucifugus, Rhinolophus ferrumequinum, Rousettus aegyptiacus, Phyllostomus discolor, Myotis myotis, Pteropus vampyrus, Pipistrellus kuhlii, Molossus molossus, Pan troglodytes, or Homo sapiens.
  • the enzyme e.g., without limitation, a mobile element enzyme
  • the enzyme is an engineered version, including but not limited to hyperactive forms, of a mobile element enzyme derived from Bombyx mori, Xenopus tropicalis, Trichoplusia ni, Myotis lucifugus, Rhinolophus ferrumequinum, Rousettus aegyptiacus, Phyllostomus discolor, Myotis myotis, Pteropus vampyrus, Pipistrellus kuhlii, Molossus molossus, Pan troglodytes, or Homo sapiens.
  • the enzyme is either the wild type, monomer, dimer, tetramer, hyperactive, or an Int-mutant.
  • the mobile element enzymes have one or more hyperactive and/or integration deficient mutations selected from TABLE 1, TABLE 2A, and/or TABLE 2B, or equivalents thereof.
  • the mobile element enzyme has a nucleotide sequence having at least about 90%, or at least about 93%, or at least about 95%, or at least about 97%, or at least about 98%, or at least about 99% identity to a nucleotide sequence of any of Rhinolophus ferrumequinum, Rousettus aegyptiacus, Phyllostomus discolor, Myotis myotis, Pteropus vampyrus, Pipistrellus kuhlii, Molossus molossus, and Pan troglodytes.
  • the mobile element enzyme has an amino acid sequence having at least about 90%, or at least about 93%, or at least about 95%, or at least about 97%, or at least about 98%, or at least about 99% identity to an amino acid sequence of any of Rhinolophus ferrumequinum, Rousettus aegyptiacus, Phyllostomus discolor, Myotis myotis, Pteropus vampyrus, Pipistrellus kuhlii, Molossus molossus, Pan troglodytes, and Homo sapiens.
  • the mobile element enzyme is an engineered version, including but not limited to a mobile element enzyme that is a monomer, dimer, tetramer, hyperactive, or has a reduced interaction with non-TTAA (SEQ ID NO: 440) recognitions sites (Int-), derived from any of Bombyx mori, Xenopus tropicalis, Trichoplusia ni, Rhinolophus ferrumequinum, Rousettus aegyptiacus, Phyllostomus discolor, Myotis myotis, Myotis lucifugus, Pipistrellus kuhlii, Pteropus vampyrus, and Molossus molossus Bombyx mori, Xenopus tropicalis, Trichoplusia ni, Pan troglodytes, Myotis lucifugus, and Homo sapiens.
  • a mobile element enzyme that is a monomer, dimer, tetramer, hyperactive, or
  • the mobile element enzyme is either the wild type, monomer, dimer, tetramer or another multimer, hyperactive, or a an Int-mutant.
  • the mobile element enzyme is from a Tc1/mariner donor DNA system. See, e.g., Plasterk et al. Trends in Genetics.1999; 15(8):326–32.
  • the mobile element enzyme is from a Sleeping Beauty donor DNA system (see, e.g., Cell.
  • a hyperactive form of Sleeping Beauty e.g., SB100X (see Gene Therapy volume 18, pages 849–856(2011), or a piggyBac (PB) donor DNA system (see, e.g., Trends Biotechnol.2015 Sep;33(9):525-33, which is incorporated herein by reference in its entirety)
  • PB piggyBac
  • a hyperactive form of PB mobile element enzyme e.g., with seven amino acid substitutions (e.g., I30V, S103P, G165S, M282V, S509G, N570S, N538K on mPB, or functional equivalents in non-mPB, see Mol Ther Nucleic Acids.2012 Oct; 1(10): e50, which is incorporated herein by reference in its entirety); see also Yusa et al., PNAS January 25, 2011108 (4) 1531-1536; Voigt et al., PNAS January 25, 2011108 (4) 1531-1536; Voig
  • the piggyBac mobile element enzymes belong to the IS4 mobile element enzyme family. De Palmenaer et al., BMC Evolutionary Biology.2008;8:18. doi: 10.1186/1471-2148-8-18.
  • the piggyBac family includes a large diversity of donor DNAs, and any of these donor DNAs can be used in embodiments of the present disclosure. See, e.g., Bouallègue et al., Genome Biol Evol.2017;9(2):323-339.
  • the founding member of the piggyBac (super)family, insect piggyBac was originally identified in the cabbage looper moth (Trichoplusiani ni) and studied both in vivo and in vitro.
  • Insect piggyBac is known to transpose by a canonical cut-and-paste mechanism promoted by an element-encoded mobile element enzyme with a catalytic site resembling the RNase H fold shared by many recombinases.
  • the insect piggyBac donor DNA system has been shown to be highly active in a wide range of animals, including Drosophila and mice, where it has been developed as a powerful tool for gene tagging and genome engineering.
  • Other donor DNAs affiliated to the piggyBac superfamily are common in arthropods and vertebrates including Xenopus and Bombyx.
  • Mammalian piggyBac donor DNAs and mobile element enzymes including hyperactive mammalian piggyBac variants, which can be used in embodiments of the present disclosure, are described, e.g., in International Application WO2010085699, which is incorporated herein by reference in its entirety.
  • the mobile element enzyme is from a LEAP-IN 1 type or LEAP-IN donor DNA system (Biotechnol J. 2018 Oct;13(10):e1700748. doi: 10.1002/biot.201700748. Epub 2018 Jun 11).
  • the LEAPIN mobile element enzyme system includes a mobile element enzyme (e.g., without limitation, a mobile element enzyme mRNA) and a vector containing one or more genes of interest (donor DNAs), selection markers, regulatory elements, insulators, etc., flanked by the donor DNA cognate inverted terminal ends and the transposition recognition motif (TTAT).
  • a mobile element enzyme e.g., without limitation, a mobile element enzyme mRNA
  • donor DNAs genes of interest
  • selection markers e.g., selection markers, regulatory elements, insulators, etc.
  • TTAT transposition recognition motif
  • the LEAPIN mobile element enzyme generates stable transgene integrants with various advantageous characteristics, including single copy integrations at multiple genomic loci, primarily in open chromatin segments; no payload limit, so multiple independent transcriptional units may be expressed from a single construct; the integrated transgenes maintain their structural and functional integrity; and maintenance of transgene integrity ensures the desired chain ratio in every recombinant cell.
  • the mobile element enzyme is an engineered form of a mobile element enzyme reconstructed from Homo sapiens or a predecessor thereof.
  • PGBD1 and PGBD2 may resemble the PGBD3 donor DNA in which the mobile element enzyme ORF is flanked upstream by a 3' splice site and downstream by a polyadenylation site. See Newman et al., PLoS Genet 2008;4:e1000031. PLoS Genet 4(3): e1000031.
  • the PGBD5 inactive mobile element enzyme sequence belongs to the RNase H clan of Pfam structures, while PGBD3 has sustained only a single D to N mutation in the essential catalytic triad DDD(D) and retains the ability to bind the upstream piggyBac terminal inverted repeat. Bailey et al., DNA Repair (Amst) 2012;11:488-501.
  • the PGBD5 mobile element enzyme does not retain the catalytic DDD (D) motif found in active elements, and the mobile element enzyme is not only inactive but fails to associate with either DNA or chromatin in vivo.
  • DDD catalytic DDD
  • PGBD1 and PGBD2 are thought to be present in the common ancestor of mammals, while PGBD3 and PGBD4 are restricted to primates. See Sarkar et al., Mol Genet Genomics 2003;270:173-80.
  • the Pteropus vampyrus mobile element enzyme is closely related to PGBD4 and shares DDD catalytic domain and the C-terminal region that are involved in excision mechanisms. See Mitra et al., EMBO J 2008;27:1097-109.
  • a mammalian mobile element enzyme which has gene cleavage and/or gene integration activity, can be constructed based on alignment of the amino acid sequence of Pteropus vampyrus mobile element enzyme to PGBD1, PGBD2, PGBD3, PGBD4, and PGBD5 sequences. Also, in embodiments. the mammalian mobile element enzyme has mutations that confers hyperactivity to a recombinant mammalian mobile element enzyme.
  • the mobile element enzyme has gene cleavage activity (Exc+) and/or gene integration activity (Int+).
  • the mobile element enzyme has gene cleavage activity (Exc+) and/or lacks gene integration activity (Int-).
  • an enzyme capable of targeted genomic integration is a recombinant mammalian mobile element enzyme that was derived by, in part, aligning several inactive mobile element enzyme sequences from a human genome to Pteropus vampyrus mobile element enzyme sequence.
  • the Pteropus vampyrus mobile element enzyme has an amino acid sequence having at least 90% identity (e.g., a sequence having at least about 90%, or at least about 93%, or at least about 95%, or at least about 97%, or at least about 98%, or at least about 99% identity) to SEQ ID NO: 430 (or a functional equivalent thereof.
  • the Pteropus vampyrus mobile element enzyme has an amino acid sequence of SEQ ID NO: 430, or a functional equivalent thereof.
  • the Pteropus vampyrus mobile element enzyme has a nucleotide sequence having at least 90% identity (e.g., a sequence having at least about 90%, or at least about 93%, or at least about 95%, or at least about 97%, or at least about 98%, or at least about 99% identity) to SEQ ID NO: 429 or a codon-optimized variant thereof.
  • the mobile element enzyme is a mammalian mobile element enzyme, such as a mobile element enzyme from a bat, e.g., without limitation, Pteropus vampyrus.
  • the mobile element enzyme is an engineered form that is based on a mobile element enzyme reconstructed from Homo sapiens or a predecessor thereof.
  • the mobile element enzyme includes but is not limited to an engineered version that is a monomer, dimer, tetramer (or another multimer), hyperactive, or has a reduced interaction with non-TTAA (SEQ ID NO: 440) recognitions sites (Int-), of an engineered version of a mobile element enzyme reconstructed from Homo sapiens or a predecessor thereof.
  • the mobile element enzyme is an engineered form that is based on a mobile element enzyme reconstructed from mammalian species.
  • the mobile element enzyme includes but is not limited to an engineered that is a monomer, dimer, tetramer (or another multimer), hyperactive, or has a reduced interaction with non-TTAA (SEQ ID NO: 440) recognitions sites (Int-), of a mobile element enzyme reconstructed from mammalian species.
  • the donor DNA is included in a vector comprising left and right end sequences recognized by the mobile element enzyme.
  • the end sequences are selected from MER, MER75A, MER75B, and MER85.
  • the end sequences are selected from nucleotide sequences of SEQ ID NO: 12, SEQ ID NO: 13 , SEQ ID NO: 14, SEQ ID NO: 15, SEQ ID NO: 16, SEQ ID NO: 17, SEQ ID NO: 18, SEQ ID NO: 19, SEQ ID NO: 20, SEQ ID NO: 21, SEQ ID NO: 441, and SEQ ID NO: 22, or a nucleotide sequence having at least about 90% identity (e.g., a sequence having at least about 90%, or at least about 93%, or at least about 95%, or at least about 97%, or at least about 98%, or at least about 99% identity) thereto.
  • a nucleotide sequence having at least about 90% identity e.g., a sequence having at least about 90%, or at least about 93%, or at least about 95%, or at least about 97%, or at least about 98%, or at least about 99% identity
  • one or more of the end sequences are optionally flanked by a TTAA (SEQ ID NO: 440) sequence.
  • the end sequences include at least one repeat from a nucleotide sequence having at least about 90% (e.g., a sequence having at least about 90%, or at least about 93%, or at least about 95%, or at least about 97%, or at least about 98%, or at least about 99% identity) identity to the nucleotide sequence of SEQ ID NO: 12, and wherein the at least one repeat from the nucleotide sequence having at least about 90% identity (e.g., a sequence having at least about 90%, or at least about 93%, or at least about 95%, or at least about 97%, or at least about 98%, or at least about 99% identity) to the nucleotide sequence of SEQ ID NO: 12 is positioned at the 5’ end of the donor DNA.
  • the end sequences can further include at least one repeat from a nucleotide sequence having at least about 90% identity (e.g., a sequence having at least about 90%, or at least about 93%, or at least about 95%, or at least about 97%, or at least about 98%, or at least about 99% identity to the nucleotide sequence of SEQ ID NO: 17, and wherein the at least one repeat from the nucleotide sequence having at least about 90% identity (e.g., a sequence having at least about 90%, or at least about 93%, or at least about 95%, or at least about 97%, or at least about 98%, or at least about 99% identity) to the nucleotide sequence of SEQ ID NO: 17 is positioned at the 3’ end of the donor DNA.
  • a nucleotide sequence having at least about 90% identity e.g., a sequence having at least about 90%, or at least about 93%, or at least about 95%, or at least about 97%, or at least about 98%, or at least
  • the end sequences which can be from, e.g., Pteropus vampyrus, are optionally flanked by a TTAA (SEQ ID NO: 440) sequence.
  • the end sequences include at least one repeat from a nucleotide sequence having at least about 90% identity (e.g., a sequence having at least about 90%, or at least about 93%, or at least about 95%, or at least about 97%, or at least about 98%, or at least about 99% identity) to the nucleotide sequence of SEQ ID NO: 13, and wherein the at least one repeat from the nucleotide sequence having at least about 90% identity (e.g., a sequence having at least about 90%, or at least about 93%, or at least about 95%, or at least about 97%, or at least about 98%, or at least about 99% identity) to the nucleotide sequence of SEQ ID NO: 13 is positioned at the 5’ end of the donor DNA.
  • the end sequences can further include at least one repeat from a nucleotide sequence having at least about 90% identity (e.g., a sequence having at least about 90%, or at least about 93%, or at least about 95%, or at least about 97%, or at least about 98%, or at least about 99% identity) to the nucleotide sequence of SEQ ID NO: 18, and wherein the at least one repeat from the nucleotide sequence having at least about 90% identity (e.g., a sequence having at least about 90%, or at least about 93%, or at least about 95%, or at least about 97%, or at least about 98%, or at least about 99% identity) to the nucleotide sequence of SEQ ID NO: 18 is positioned at the 3’ end of the donor DNA.
  • a nucleotide sequence having at least about 90% identity e.g., a sequence having at least about 90%, or at least about 93%, or at least about 95%, or at least about 97%, or at least about 98%, or at
  • the end sequences which can be, e.g., PGBD4, are optionally flanked by a TTAA (SEQ ID NO: 440) sequence.
  • the end sequences include at least one repeat from a nucleotide sequence having at least about 90% identity (e.g., a sequence having at least about 90%, or at least about 93%, or at least about 95%, or at least about 97%, or at least about 98%, or at least about 99% identity) to the nucleotide sequence of SEQ ID NO: 14, wherein the at least one repeat from the nucleotide sequence having at least about 90% identity (e.g., a sequence having at least about 90%, or at least about 93%, or at least about 95%, or at least about 97%, or at least about 98%, or at least about 99% identity) to the nucleotide sequence of SEQ ID NO: 14 is positioned at the 5’ end of the donor DNA.
  • the end sequences include at least one repeat from a nucleotide sequence having at least about 90% identity (e.g., a sequence having at least about 90%, or at least about 93%, or at least about 95%, or at least about 97%, or at least about 98%, or at least about 99% identity) to the nucleotide sequence of SEQ ID NO: 18, wherein the at least one repeat from the nucleotide sequence having at least about 90% identity (e.g., a sequence having at least about 90%, or at least about 93%, or at least about 95%, or at least about 97%, or at least about 98%, or at least about 99% identity) to the nucleotide sequence of SEQ ID NO: 19 is positioned at the 3’ end of the donor DNA.
  • a nucleotide sequence having at least about 90% identity e.g., a sequence having at least about 90%, or at least about 93%, or at least about 95%, or at least about 97%, or at least about 98%, or at least about
  • the end sequences which can be, e.g., MER75, are optionally flanked by a TTAA (SEQ ID NO: 440) sequence.
  • the end sequences include at least one repeat from a nucleotide sequence having at least about 90% identity (e.g., a sequence having at least about 90%, or at least about 93%, or at least about 95%, or at least about 97%, or at least about 98%, or at least about 99% identity) to the nucleotide sequence of SEQ ID NO: 15, wherein the at least one repeat from the nucleotide sequence having at least about 90% identity (e.g., a sequence having at least about 90%, or at least about 93%, or at least about 95%, or at least about 97%, or at least about 98%, or at least about 99% identity) to the nucleotide sequence of SEQ ID NO: 15 is positioned at the 5’ end of the donor DNA.
  • the end sequences include at least one repeat from a nucleotide sequence having at least about 90% identity (e.g., a sequence having at least about 90%, or at least about 93%, or at least about 95%, or at least about 97%, or at least about 98%, or at least about 99% identity) to the nucleotide sequence of SEQ ID NO: 20, wherein the at least one repeat from the nucleotide sequence having at least about 90% identity(e.g. a sequence having at least about 90%, or at least about 93%, or at least about 95%, or at least about 97%, or at least about 98%, or at least about 99% identity) to the nucleotide sequence of SEQ ID NO: 20 is positioned at the 3’ end of the donor DNA.
  • a nucleotide sequence having at least about 90% identity e.g., a sequence having at least about 90%, or at least about 93%, or at least about 95%, or at least about 97%, or at least about 98%, or at least about 99%
  • the end sequences which can be, e.g., MER75B, are optionally flanked by a TTAA (SEQ ID NO: 440) sequence.
  • the end sequences include at least one repeat from a nucleotide sequence having at least about 90% (e.g., a sequence having at least about 90%, or at least about 93%, or at least about 95%, or at least about 97%, or at least about 98%, or at least about 99% identity) identity to the nucleotide sequence of SEQ ID NO: 16, wherein the at least one repeat from the nucleotide sequence having at least about 90% identity (e.g., a sequence having at least about 90%, or at least about 93%, or at least about 95%, or at least about 97%, or at least about 98%, or at least about 99% identity) to the nucleotide sequence of SEQ ID NO: 16 is positioned at the 5’ end of the donor DNA.
  • the end sequences include at least one repeat from a nucleotide sequence having at least about 90% identity (e.g., a sequence having at least about 90%, or at least about 93%, or at least about 95%, or at least about 97%, or at least about 98%, or at least about 99% identity) to the nucleotide sequence of SEQ ID NO: 21 or SEQ ID NO: 441, wherein the at least one repeat from the nucleotide sequence having at least about 90% identity (e.g., a sequence having at least about 90%, or at least about 93%, or at least about 95%, or at least about 97%, or at least about 98%, or at least about 99% identity) to the nucleotide sequence of SEQ ID NO: 21 or SEQ ID NO: 441 is positioned at the 3’ end of the donor DNA.
  • a nucleotide sequence having at least about 90% identity e.g., a sequence having at least about 90%, or at least about 93%, or at least about 95%, or at least
  • a donor DNA is or comprises a vector comprising a donor DNA comprising one or more end sequences recognized by an enzyme such as, for example a mobile element enzyme.
  • the end sequences are selected from Pteropus vampyrus, MER75, MER75A, and MER75B. MERs contain end sequences with similarity to piggyBac-like mobile elements and exhibit duplications of their presumed TTAA (SEQ ID NO: 440) target sites.
  • the end sequences are selected from nucleotide sequences of SEQ ID NO: 12, SEQ ID NO: 13, SEQ ID NO: 14, SEQ ID NO: 15, SEQ ID NO: 16, SEQ ID NO: 17, SEQ ID NO: 18, SEQ ID NO: 19, SEQ ID NO: 20, SEQ ID NO: 21, SEQ ID NO: 441, and SEQ ID NO: 22, or a nucleotide sequence having at least about 90% identity (e.g. a sequence having at least about 90%, or at least about 93%, or at least about 95%, or at least about 97%, or at least about 98%, or at least about 99% identity) thereto.
  • a nucleotide sequence having at least about 90% identity e.g. a sequence having at least about 90%, or at least about 93%, or at least about 95%, or at least about 97%, or at least about 98%, or at least about 99% identity
  • the mobile element enzyme has an amino acid sequence of SEQ ID NO: 3, SEQ ID NO: 4, SEQ ID NO: 6, SEQ ID NO: 7, SEQ ID NO: 8, or SEQ ID NO: 9, or a variant sequence having at least about 90%, or at least about 93%, or at least about 95%, or at least about 97%, or at least about 98%, or at least about 99% identity thereto, or at least about 10 mutations, or at least about 9 mutations, or at least about 8 mutations, or at least about 7 mutations, or at least about 6 mutations, or at least about 5 mutations, or at least about 4 mutations, or at least about 3 mutations, or at least about 2 mutations, or at least about 1 mutation.
  • the mobile element enzyme has an amino acid sequence having S8P, G17R, and/or K134K mutation relative to the amino acid sequence of SEQ ID NO: 4 or a functional equivalent thereof. In embodiments, the mobile element enzyme has an amino acid sequence having S8P, G17R, and/or K134K mutation relative to the amino acid sequence of SEQ ID NO: 5 or a functional equivalent thereof. In embodiments, the mobile element enzyme has an amino acid sequence having I83P and/or V118R mutation relative to the amino acid sequence of SEQ ID NO: 6 or a functional equivalent thereof. In embodiments, the mobile element enzyme has an amino acid sequence having S20P and/or A29R mutation relative to the amino acid sequence of SEQ ID NO: 7 or a functional equivalent thereof.
  • the mobile element enzyme has an amino acid sequence having T4P and/or L13R mutation relative to the amino acid sequence of SEQ ID NO: 8 or a functional equivalent thereof. In embodiments, the mobile element enzyme has an amino acid sequence having A12P and/or I28R mutation and/or R152K mutation relative to the amino acid sequence of SEQ ID NO: 9 or a functional equivalent thereof.
  • the enzyme capable of targeted genomic integration e.g., without limitations, a mobile element enzyme
  • the enzyme capable of targeted genomic integration e.g., without limitations, a mobile element enzyme
  • the enzyme e.g., without limitation, a mobile element enzyme
  • the enzyme is an engineered version, including but not limited to a mobile element enzyme that is a monomer, dimer, tetramer, hyperactive, or has a reduced interaction with non-TTAA (SEQ ID NO: 440) recognitions sites (Int-), and is derived from any of Bombyx mori, Xenopus tropicalis, Trichoplusia ni, Myotis lucifugus, Rhinolophus ferrumequinum, Rousettus aegyptiacus, Phyllostomus discolor, Myotis myotis, Pteropus vampyrus, Pipistrellus kuhlii, Pan troglodytes, Molossus molossus, or Homo sapiens.
  • a mobile element enzyme that is a monomer, dimer, tetramer, hyperactive, or has a reduced interaction with non-TTAA (SEQ ID
  • the mobile element enzyme is either the wild type, monomer, dimer, tetramer or another multimer, hyperactive, or an Int-mutant.
  • Targeting Chimeric Constructs e.g., in embodiments, the enzyme, without limitation, a mobile element enzyme, comprises a targeting element.
  • the targeting element is or comprises a gene-editing system, e.g., that is catalytically inactive (or “dead”).
  • the enzyme without limitation, a mobile element enzyme, associated with the targeting element, is capable of inserting the donor DNA (e.g., mobile element) comprising a transgene, optionally at a TA dinucleotide site or a TTAA (SEQ ID NO: 440) tetranucleotide site in a GSHS.
  • the enzyme without limitation, a mobile element enzyme, associated with the targeting element has one or more mutations which confer hyperactivity.
  • the enzyme, without limitation, a mobile element enzyme, associated with the targeting element has gene cleavage activity (Exc+) and/or gene integration activity (Int+).
  • the enzyme without limitation, a mobile element enzyme, associated with the targeting element has gene cleavage activity (Exc+) and/or a lack of gene integration activity (Int-).
  • the targeting element comprises one or more proteins or nucleic acids that are capable of binding to a nucleic acid.
  • the targeting element comprises one or more of a of a gRNA, optionally associated with a Cas enzyme, which is optionally catalytically inactive, transcription activator-like effector (TALE), catalytically inactive Zinc finger, catalytically inactive transcription factor, nickase, a transcriptional activator, a transcriptional repressor, a recombinase, a DNA methyltransferase, a histone methyltransferase, paternally expressed gene 10 (PEG10), and TnsD.
  • the targeting element comprises a transcription activator-like effector (TALE) DNA binding domain (DBD).
  • the TALE DBD comprises one or more repeat sequences.
  • the TALE DBD comprises about 14, or about 15, or about, 16, or about 17, or about 18, or about 18.5 repeat sequences.
  • the TALE DBD repeat sequences comprise 33 or 34 amino acids.
  • the TALE DBD repeat sequences comprise a repeat variable di-residue (RVD) at residue 12 or 13 of the 33 or 34 amino acids.
  • RVD recognizes one base pair in the nucleic acid molecule.
  • the RVD recognizes a C residue in the nucleic acid molecule and is selected from HD, N(gap), HA, ND, and HI.
  • the RVD recognizes a G residue in the nucleic acid molecule and is selected from NN, NH, NK, HN, and NA. In embodiments, the RVD recognizes an A residue in the nucleic acid molecule and is selected from NI and NS. In embodiments, the RVD recognizes a T residue in the nucleic acid molecule and is selected from NG, HG, H(gap), and IG. In embodiments, the GSHS is in an open chromatin location in a chromosome. In embodiments, the GSHS is selected from adeno-associated virus site 1 (AAVS1), chemokine (C-C motif) receptor 5 (CCR5) gene, HIV-1 coreceptor, and human Rosa26 locus.
  • AAVS1 adeno-associated virus site 1
  • C-C motif chemokine receptor 5
  • the GSHS is located on human chromosome 2, 4, 6, 10, 11, 17, 22, or X.
  • the GSHS is selected from TALC1, TALC2, TALC3, TALC4, TALC5, TALC7, TALC8, AVS1, AVS2, AVS3, ROSA1, ROSA2, TALER1, TALER2, TALER3, TALER4, TALER5, SHCHR2-1, SHCHR2-2, SHCHR2-3, SHCHR2-4, SHCHR4-1, SHCHR4-2, SHCHR4-3, SHCHR6-1, SHCHR6-2, SHCHR6-3, SHCHR6-4, SHCHR10-1, SHCHR10-2, SHCHR10-3, SHCHR10-4, SHCHR10-5, SHCHR11-1, SHCHR11-2, SHCHR11-3, SHCHR17-1, SHCHR17-2, SHCHR17-3, and SHCHR17-4.
  • the targeting element comprises a Cas9 enzyme guide RNA complex.
  • the Cas9 enzyme guide RNA complex comprises a nuclease-deficient dCas9 guide RNA complex.
  • the targeting element comprises a Cas12 enzyme guide RNA complex.
  • the targeting element comprises a nuclease-deficient dCas12 guide RNA complex, optionally dCas12j guide RNA complex or dCas12a guide RNA complex.
  • the targeting element comprises a Cas12k enzyme guide RNA complex.
  • the targeting element comprises a nuclease-deficient dCas12 guide RNA complex, optionally dCas12k guide RNA complex.
  • a targeting chimeric system or construct having a DBD fused to a mobile element enzyme, directs binding of an enzyme capable of targeted genomic integration (e.g., without limitation, a mobile element enzyme) to a specific sequence (e.g., transcription activator-like effector proteins (TALE) repeat variable di-residues (RVD) or gRNA) near an enzyme recognition site.
  • TALE transcription activator-like effector proteins
  • RVD repeat variable di-residues
  • gRNA binds to human GSHS.
  • dCas9 i.e., deficient for nuclease activity
  • gRNAs directed to bind at a desired sequence of DNA in GSHS.
  • TALEs described herein can physically sequester the enzyme such as, e.g., a mobile element enzyme, to GSHS and promote transposition to nearby TTAA (SEQ ID NO: 440) sequences in close proximity to the RVD TALE nucleotide sequences.
  • GSHS in open chromatin sites are specifically targeted based on the predilection for mobile element enzymes to insert into open chromatin.
  • an enzyme capable of targeted genomic integration e.g., without limitation, a recombinase, integrase, or a mobile element enzyme such as, without limitation, a mammalian mobile element enzyme
  • a TALE DNA binding domain DBD
  • a Cas-based gene-editing system such as, e.g., Cas9 or a variant thereof.
  • the targeting element targets the enzyme to a locus of interest.
  • the targeting element comprises CRISPR (Clustered Regularly Interspaced Short Palindromic Repeat) associated protein 9 (Cas9), or a variant thereof.
  • a CRISPR/Cas9 tool only requires Cas9 nuclease for DNA cleavage and a single-guide RNA (sgRNA) for target specificity.
  • sgRNA single-guide RNA
  • the inactivated form of Cas9 which is a nuclease-deficient (or inactive, or “catalytically dead” Cas9, is typically denoted as “dCas9,” has no substantial nuclease activity.
  • dCas9 has no substantial nuclease activity.
  • CRISPR/dCas9 binds precisely to specific genomic sequences through targeting of guide RNA (gRNA) sequences.
  • gRNA guide RNA
  • dCas9 is utilized to edit gene expression when applied to the transcription binding site of a desired site and/or locus in a genome.
  • gRNA guide RNA
  • dCas9 prevents the proliferation of repeating codons and DNA sequences that might be harmful to an organism's genome.
  • the targeting element comprises a nuclease-deficient Cas enzyme guide RNA complex.
  • the targeting element comprises a nuclease-deficient (or inactive, or “catalytically dead” Cas, e.g., Cas9, typically denoted as “dCas” or “dCas9” ) guide RNA complex.
  • the dCas9/gRNA complex comprises a guide RNA selected from: GTTTAGCTCACCCGTGAGCC (SEQ ID NO: 91), CCCAATATTATTGTTCTCTG (SEQ ID NO: 92), GGGGTGGGATAGGGGATACG (SEQ ID NO: 93), GGATCCCCCTCTACATTTAA (SEQ ID NO: 94), GTGATCTTGTACAAATCATT (SEQ ID NO: 95), CTACACAGAATCTGTTAGAA (SEQ ID NO: 96), TAAGCTAGAGAATAGATCTC (SEQ ID NO: 97), and TCAATACACTTAATGATTTA (SEQ ID NO: 98), wherein the guide RNA directs the enzyme to a chemokine (C-C motif) receptor 5 (CCR5) gene.
  • C-C motif chemokine receptor 5
  • the dCas9/gRNA complex comprises a guide RNA selected from: CACCGGGAGCCACGAAAACAGATCC (SEQ ID NO: 99);CACCGCGAAAACAGATCCAGGGACA (SEQ ID NO: 100); CACCGAGATCCAGGGACACGGTGCT (SEQ ID NO: 101); CACCGGACACGGTGCTAGGACAGTG (SEQ ID NO: 102); CACCGGAAAATGACCCAACAGCCTC (SEQ ID NO: 103); CACCGGCCTGGCCGGCCTGACCACT (SEQ ID NO: 104); CACCGCTGAGCACTGAAGGCCTGGC (SEQ ID NO: 105); CACCGTGGTTTCCACTGAGCACTGA (SEQ ID NO: 106); CACCGGATAGCCAGGAGTCCTTTCG (SEQ ID NO: 107); CACCGGCGCTTCCAGTGCTCAGACT (SEQ ID NO: 108); CACCGCAGTGCTCAGACTAGGGAAG (SEQ ID NO: 109
  • the guide RNAs are: AATCGAGAAGCGACTCGACA (SEQ ID NO: 425), and tgccctgcaggggagtgagc (SEQ ID NO: 426).
  • the guide RNAs are gaagcgactcgacatggagg (SEQ ID NO: 427) and cctgcaggggagtgagcagc (SEQ ID NO: 428).
  • guide RNAs (gRNAs) for targeting human genomic safe harbor sites using any of the gRNA-based targeting elements, e.g., without limitation dCas, in areas of open chromatin are as shown in TABLE 3A-3F.
  • guide RNAs for targeting human genomic safe harbor sites using any of the gRNA-based targeting elements, e.g., without limitation dCas, in areas of open chromatin are as shown in TABLE 3A: In embodiments, gRNAs for targeting human genomic safe harbor sites using any of the gRNA-based targeting elements, e.g., without limitation dCas, to the TTAA site in hROSA26 (e.g., hg38 chr3:9,396,133-9,396,305) are shown in TABLE 3B: In embodiments, gRNAs for targeting human genomic safe harbor sites using any of the gRNA-based targeting elements, e.g., without limitation dCas, to the AAVS1 (e.g., hg38 chr19:55,112,851-55,113,324) are shown in TABLE 3C: In embodiments, gRNAs for targeting human genomic safe harbor sites using any of the gRNA-
  • a Cas-based targeting element comprises Cas12 or a variant thereof, e.g., without limitation, Cas12a (e.g., dCas12a), or Cas12j (e.g., dCas12j), or Cas12k (e.g., dCas12k).
  • the targeting element comprises a Cas12 enzyme guide RNA complex.
  • the targeting element is selected from a zinc finger (ZF), catalytically inactive Zinc finger, transcription activator-like effector (TALE), meganuclease, and clustered regularly interspaced short palindromic repeat (CRISPR)- associated protein, any of which are, in embodiments, catalytically inactive.
  • ZF zinc finger
  • TALE transcription activator-like effector
  • CRISPR clustered regularly interspaced short palindromic repeat
  • the CRISPR-associated protein is selected from Cas9, CasX, CasY, Cas12a (Cpf1), and gRNA complexes thereof.
  • the CRISPR-associated protein is selected from Cas9, xCas9, Cas 6, Cas7, Cas8, Cas12a (Cpf1), Cas13a, Cas14, CasX, CasY, a Class 1 Cas protein, a Class 2 Cas protein, MAD7, MG1 nuclease, MG2 nuclease, MG3 nuclease, or catalytically inactive forms thereof, and gRNA complexes thereof.
  • the mobile element enzyme is capable of inserting a donor DNA at a TA dinucleotide site or a TTAA tetranucleotide site in a GSHS of a nucleic acid molecule.
  • the mobile element enzyme is suitable for causing insertion of the donor DNA in a GSHS when contacted with a biological cell.
  • the targeting element is suitable for directing the mobile element enzyme to the GSHS sequence.
  • the targeting element comprises transcription activator-like effector (TALE) DNA binding domain (DBD).
  • TALE DBD comprises one or more repeat sequences.
  • the TALE DBD comprises about 14, or about 15, or about, 16, or about 17, or about 18, or about 18.5 repeat sequences.
  • the TALE DBD repeat sequences comprise 33 or 34 amino acids.
  • the one or more of the TALE DBD repeat sequences comprise a repeat variable di-residue (RVD) at residue 12 or 13 of the 33 or 34 amino acids.
  • the targeting element e.g., TALE or Cas (e.g., Cas9 or Cas12, or variants thereof) DBDs cause the mammalian mobile element enzyme to bind specifically to human GSHS.
  • the TALEs or Cas DBDs sequester the mobile element enzyme to GSHS and promote transposition to nearby TA dinucleotide or a TTAA tetranucleotide sites which can be located in proximity to the repeat variable di-residues (RVD) TALE or gRNA nucleotide sequences.
  • the GSHS regions are located in open chromatin sites that are susceptible to mobile element enzyme activity. Accordingly, the mammalian mobile element enzyme does not only operate based on its ability to recognize TA or TTAA sites, but it also directs a donor DNA (having a transgene) to specific locations in proximity to a TALE or Cas DBD.
  • the chimeric mobile element enzyme in accordance with embodiments of the present disclosure has negligible risk of genotoxicity and exhibits superior features as compared to existing gene therapies.
  • a chimeric mobile element enzyme is mutated to be characterized by reduced or inhibited binding of off-target sequences and consequently reliant on a DBD fused thereto, such as a TALE or Cas DBD, for transposition.
  • a DBD fused thereto such as a TALE or Cas DBD
  • the described cells, compositions, and methods allow reducing vector and transgene insertions that increase a mutagenic risk.
  • the described cells and methods make use of a gene transfer system that reduces genotoxicity compared to viral- and nuclease-mediated gene therapies.
  • TALE or Cas DBDs are customizable, such as a TALE or Cas DBDs is selected for targeting a specific genomic location.
  • the genomic location is in proximity to a TA dinucleotide site or a TTAA (SEQ ID NO: 440) tetranucleotide site.
  • TTAA SEQ ID NO: 440
  • TALE repeat sequences e.g., modular arrays
  • gRNA e.g., gRNA which are linked together to recognize flanking DNA sequences.
  • TALE or gRNA can recognize certain base pair(s) or residue(s).
  • TALE nucleases TALENs
  • TALENs are a known tool for genome editing and introducing targeted double-stranded breaks. TALENs comprise endonucleases, such as FokI nuclease domain, fused to a customizable DBD. This DBD is composed of highly conserved repeats from TALEs, which are proteins secreted by Xanthomonas bacteria to alter transcription of genes in host plant cells.
  • the DBD includes a repeated highly conserved 33–34 amino acid sequence with divergent 12th and 13th amino acids. These two positions, referred to as the RVD, are highly variable and show a strong correlation with specific base pair or nucleotide recognition. This straightforward relationship between amino acid sequence and DNA recognition has allowed for the engineering of specific DBDs by selecting a combination of repeat segments containing the appropriate RVDs. Boch et al. Nature Biotechnology.2011; 29 (2): 135–6. Accordingly, TALENs can be readily designed using a “protein-DNA code” that relates modular DNA-binding TALE repeat domains to individual bases in a target-binding site. See Joung et al. Nat Rev Mol Cell Biol.2013;14(1):49-55.
  • the TALE DBD comprises one or more repeat sequences. In embodiments, the TALE DBD comprises about 15, or about, 16, or about 17, or about 18, or about 18.5 repeat sequences.
  • the TALE DBD repeat sequences comprise 33 or 34 amino acids.
  • the one or more of the TALE DBD repeat sequences comprise an RVD at residue 12 or 13 of the 33 or 34 amino acids.
  • the RVD can recognize certain base pair(s) or residue(s).
  • the RVD recognizes one base pair in the nucleic acid molecule.
  • the RVD recognizes a C residue in the nucleic acid molecule and is selected from HD, N(gap), HA, ND, and HI.
  • the RVD recognizes a G residue in the nucleic acid molecule and is selected from NN, NH, NK, HN, and NA.
  • the RVD recognizes an A residue in the nucleic acid molecule and is selected from NI and NS. In embodiments, the RVD recognizes a T residue in the nucleic acid molecule and is selected from NG, HG, H(gap), and IG.
  • the GSHS is in an open chromatin location in a chromosome. In embodiments, the GSHS is selected from adeno-associated virus site 1 (AAVS1), chemokine (C-C motif) receptor 5 (CCR5) gene, HIV-1 coreceptor; and human Rosa26 locus. In embodiments, the GSHS is located on human chromosome 2, 4, 6, 10, 11, 17, 22, or X.
  • the GSHS is selected from TALC1, TALC2, TALC3, TALC4, TALC5, TALC7, TALC8, AVS1, AVS2, AVS3, ROSA1, ROSA2, TALER1, TALER2, TALER3, TALER4, TALER5, SHCHR2-1, SHCHR2-2, SHCHR2-3, SHCHR2-4, SHCHR4-1, SHCHR4-2, SHCHR4-3, SHCHR6-1, SHCHR6-2, SHCHR6-3, SHCHR6-4, SHCHR10-1, SHCHR10-2, SHCHR10-3, SHCHR10-4, SHCHR10-5, SHCHR11-1, SHCHR11-2, SHCHR11-3, SHCHR17-1, SHCHR17-2, SHCHR17-3, and SHCHR17-4.
  • the GSHS comprises one or more of TGGCCGGCCTGACCACTGG (SEQ ID NO: 23), TGAAGGCCTGGCCGGCCTG (SEQ ID NO: 24), TGAGCACTGAAGGCCTGGC (SEQ ID NO: 25), TCCACTGAGCACTGAAGGC (SEQ ID NO: 26), TGGTTTCCACTGAGCACTG (SEQ ID NO: 27), TGGGGAAAATGACCCAACA (SEQ ID NO: 28), TAGGACAGTGGGGAAAATG (SEQ ID NO: 29), TCCAGGGACACGGTGCTAG (SEQ ID NO: 30), TCAGAGCCAGGAGTCCTGG (SEQ ID NO: 31), TCCTTCAGAGCCAGGAGTC (SEQ ID NO: 32), TCCTCCTTCAGAGCCAGGA (SEQ ID NO: 33), TCCAGCCCCTCCTCCTTCA (SEQ ID NO: 34), TCCGAGCTTGACCCTTGGA (SEQ ID NO: 35), TGGTTTCCGAGCTTGACCC (SEQ ID
  • the TALE DBD binds to one of TGGCCGGCCTGACCACTGG (SEQ ID NO: 23), TGAAGGCCTGGCCGGCCTG (SEQ ID NO: 24), TGAGCACTGAAGGCCTGGC (SEQ ID NO: 25), TCCACTGAGCACTGAAGGC (SEQ ID NO: 26), TGGTTTCCACTGAGCACTG (SEQ ID NO: 27), TGGGGAAAATGACCCAACA (SEQ ID NO: 28), TAGGACAGTGGGGAAAATG (SEQ ID NO: 29), TCCAGGGACACGGTGCTAG (SEQ ID NO: 30), TCAGAGCCAGGAGTCCTGG (SEQ ID NO: 31), TCCTTCAGAGCCAGGAGTC (SEQ ID NO: 32), TCCTCCTTCAGAGCCAGGA (SEQ ID NO: 33), TCCAGCCCCTCCTCCTTCA (SEQ ID NO: 34), TCCGAGCTTGACCCTTGGA (SEQ ID NO: 35), TGGTTTCCGAGCTTGACC
  • the TALE DBD comprises one or more of: NH NH HD HD NH NH HD HD NG NH NI HD HD NI HD NG NH NH, NH NI NI NH NH HD HD NG NH NH HD HD NH NH HD NG NH, NH NI NH HD NI HD NG NH NI NI NH NH HD HD NG NH NH HD, NH NI HD NG NH NI NH HD NI HD NG NH NI NI NH NH HD, NH NH NG NG NG HD HD NI HD NG NH NI NI HD NG NH, NH NH NH NH NH NI NI NI NI HD NG NH, NH NH NH NH NI NI NI NI NG NH NI HD NI HD NG NH, NH NH NH NH NI NI NI NI NG NH NI HD NI HD NG NH, NH NH
  • the TALE DBD comprises one or more of the sequences outlined herein or a variant sequence having at least about 90%, or at least about 93%, or at least about 95%, or at least about 97%, or at least about 98%, or at least about 99% identity thereto, or at least about 10 mutations, or at least about 9 mutations, or at least about 8 mutations, or at least about 7 mutations, or at least about 6 mutations, or at least about 5 mutations, or at least about 4 mutations, or at least about 3 mutations, or at least about 2 mutations, or at least about 1 mutation.
  • the GSHS and the TALE DBD sequences are selected from: TGGCCGGCCTGACCACTGG (SEQ ID NO: 23) and NH NH HD HD NH NH HD HD NG NH NI HD HD NI HD NG NH NH; TGAAGGCCTGGCCGGCCTG (SEQ ID NO: 24) and NH NI NI NH NH HD HD NG NH NH HD HD NH NH HD HD NG NH; TGAGCACTGAAGGCCTGGC (SEQ ID NO: 25) and NH NI NH HD NI HD NG NH NI NI NH NH HD HD NG NH NH HD; TCCACTGAGCACTGAAGGC (SEQ ID NO: 26) and HD HD NI HD NG NH NI NH HD NI HD NG NH NI NI NH NH HD; TGGTTTCCACTGAGCACTG (SEQ ID NO: 27) and NH NH NG NG NG HD HD NI HD NG NH NH
  • the GSHS is within about 25, or about 50, or about 100, or about 150, or about 200, or about 300, or about 500 nucleotides of the TA dinucleotide site or TTAA (SEQ ID NO: 440) tetranucleotide site.
  • Illustrative DNA binding codes for human genomic safe harbor in areas of open chromatin via TALEs, encompassed by various embodiments are provided in TABLE 4A-4F.
  • TALEs encompassed by various embodiments are provided in TABLE 4A-4F, e.g., having a sequence having at least about 90%, or at least about 93%, or at least about 95%, or at least about 97%, or at least about 98%, or at least about 99% identity to any of the sequences in TABLE 4A-4F.
  • TALEs for targeting human genomic safe harbor sites using any of the TALE-based targeting elements to the TTAA site in hROSA26 e.g., hg38 chr3:9,396,133-9,396,305
  • TABLE 4B TALEs for targeting human genomic safe harbor sites using any of the TALE-based targeting elements to the AAVS1 (e.g., hg38 chr19:55,112,851-55,113,324) are shown in TABLE 4C:
  • TALEs for targeting human genomic safe harbor sites using any of the TALE-based targeting elements to Chromosome 4 are shown in TABLE 4D:
  • TALEs for targeting human genomic safe harbor sites using any of the TALE-based targeting elements to Chromosome 22 are shown in TABLE 4E:
  • TALEs for targeting human genomic safe harbor sites using any of the TALE-based targeting elements to Chromosome X are shown in TABLE 4D:
  • TALEs for targeting human genomic safe harbor sites using any of the TALE-based targeting elements to Chromosome 22 are shown in embodiments.
  • TALEs for targeting human genomic safe harbor sites using any of the TALE-based targeting elements to Chromosome 22 are shown in TABLE 4E:
  • the mobile element enzyme is capable of inserting a donor DNA at a TTAA (SEQ ID NO: 440) tetranucleotide site.
  • TTAA SEQ ID NO: 440
  • Illustrative DNA binding codes for human genomic safe harbor in areas of open chromatin via ZNFs encompassed by various embodiments are provided in TABLE 5A-5E.
  • there is provided a variant of the ZNFs, encompassed by various embodiments are provided in TABLE 5A-5E, e.g., having a sequence having at least about 90%, or at least about 93%, or at least about 95%, or at least about 97%, or at least about 98%, or at least about 99% identity to any of the sequences in TABLE 5A-5E.
  • ZNFs for targeting human genomic safe harbor sites using any of the ZNF-based targeting elements to the TTAA site in hROSA26 are shown in TABLE 5A:
  • ZNFs for targeting human genomic safe harbor sites using any of the ZNF-based targeting elements to the AAVS1 are shown in TABLE 5B:
  • ZNFs for targeting human genomic safe harbor sites using any of the ZNF-based targeting elements to Chromosome 4 are shown in TABLE 5C:
  • ZNFs for targeting human genomic safe harbor sites using any of the ZNF-based targeting elements to Chromosome 22 are shown in TABLE 5D:
  • ZNFs for targeting human genomic safe harbor sites using any of the ZNF-based targeting elements to Chromosome X are shown in TABLE 5E:
  • the mobile element enzyme is capable of inserting a donor DNA at a TA dinucleotide site.
  • the mobile element enzyme is capable of inserting a donor DNA at a TTAA (SEQ ID NO: 440) tetranucleotide site.
  • the present disclosure relates to a system having nucleic acids encoding the enzyme and the donor DNA, respectively.
  • FIGs.1A-1D show examples of a system in accordance with embodiments of the present disclosure.
  • Linkers In embodiments, the targeting element comprises a nucleic acid binding component of the gene-editing system (targeting element).
  • the enzyme capable of targeted genomic integration e.g., without limitation, a chimeric mobile element enzyme
  • the targeting element e.g., nucleic acid binding component of the gene-editing system are fused or linked to one another.
  • the mobile element enzyme and the targeting element are fused or linked to one another.
  • the mobile element enzyme and the targeting element e.g., nucleic acid binding component of the gene-editing system are connected via a linker.
  • the linker is a flexible linker.
  • the flexible linker is substantially comprised of glycine and serine residues, optionally wherein the flexible linker comprises (Gly 4 Ser) n , where n is from about 1 to about 12.
  • the flexible linker is of about 20, or about 30, or about 40, or about 50, or about 60 amino acid residues.
  • the flexible linker is about 50, or about 100, or about 150, or about 200 amino acid residues in length. In embodiments, the flexible linker comprises at least about 150 nucleotides (nt), or at least about 200 nt, or at least about 250 nt, or at least about 300 nt, or at least about 350 nt, or at least about 400 nt, or at least about 450 nt, or at least about 500 nt, or at least about 500 nt, or at least about 600 nt. In embodiments, the flexible linker comprises from about 450 nt to about 500 nt.
  • the mobile element enzyme and the targeting element are encoded on a single polypeptide.
  • the donor DNA comprises a gene encoding a complete polypeptide.
  • the donor DNA comprises a gene which is defective or substantially absent in a disease state.
  • Inteins Inteins are mobile genetic elements that are protein domains, found in nature, with the capability to carry out the process of protein splicing. See Sarmiento & Camarero (2019) Current Protein & Peptide Science, 20(5), 408–424, which is incorporated by reference herein in its entirety.
  • Protein spicing is a post-translation biochemical modification which results in the cleavage and formation of peptide bonds between precursor polypeptide segments flanking the intein.
  • Inteins apply standard enzymatic strategies to excise themselves post-translationally from a precursor protein via protein splicing. Nanda et al., Microorganisms vol. 8,12 2004. 16 Dec. 2020, doi:10.3390/microorganisms8122004.
  • An intein can splice its flanking N- and C-terminal domains to become a mature protein and excise itself from a sequence. For example, split inteins have been used to control the delivery of heterologous genes into transgenic organisms.
  • intein-mediated incorporation of DNA binders such as, without limitation, dCas9, dCas12j, or TALEs, allows creation of a split-enzyme system such as, without limitation, split-MLT mobile element enzyme system, that permits reconstitution of the full-length enzyme, e.g., MLT mobile element enzyme, from two smaller fragments.
  • a nucleic acid encoding the enzyme capable of targeted genomic integration comprises an intein.
  • the nucleic acid encodes the enzyme in the form of first and second portions with the intein encoded between the first and second portions, such that the first and second portions are fused into a functional enzyme upon post-translational excision of the intein from the enzyme.
  • an intein is a suitable ligand-dependent intein, for example, an intein selected from those described in U.S. Patent No.9,200,045; Mootz et al., J. Am. Chem. Soc.2002; 124, 9044-9045; Mootz et al., J. Am. Chem. Soc. 2003; 125, 10561-10569; Buskirk et al., Proc. Natl.
  • the intein is NpuN (Intein-N) (SEQ ID NO: 423) and/or NpuC (Intein-C) (SEQ ID NO: 424), or a variant thereof, e.g., a sequence having at least about 90%, or at least about 93%, or at least about 95%, or at least about 97%, or at least about 98%, or at least about 99% identity thereto.
  • SEQ ID NO: 423 nucleotide sequence of NpuN (Intein-N) GGCGGATCTGGCGGTAGTGCTGAGTATTGTCTGAGTTACGAAACGGAAATACTCACGGTTGAGTATGGGCTTCTTCC AATTGGCAAAATCGTTGAAAAGCGCATAGAGTGTACGGTGTATTCCGTCGATAACAACGGTAATATCTACACCCAGC CGGTAGCTCAGTGGCACGACCGAGGCGAACAGGAAGTGTTCGAGTATTGCTTGGAAGATGGCTCCCTTATCCGCGCC ACTAAAGACCATAAGTTTATGACGGTTGACGGGCAGATGCTGCCTATAGACGAAATATTTGAGAGAGCTGGACTT GATGAGAGTCGATAATCTGCCAAAT SEQ ID NO: 424: nucleotide sequence of NpuC (Intein-C) GGCGGATCTGGCGGTAGTGGGGGTTCCGGATCCATAAAGATAGCTACTAGGAAATATCTTGGCAAACAAAACGTCTA TGACAAA
  • a nucleic acid encoding the transgene is DNA.
  • the enzyme e.g., without limitation, the mobile element enzyme
  • the nucleic acid is RNA, optionally a helper RNA.
  • the nucleic acid is RNA that has a 5’-m7G cap (cap0, or cap1, or cap2), optionally with pseudouridine substitution (e.g., without limitation n-methyl-pseudouridine), and optionally a poly-A tail of about 30, or about 50, or about 100, of about 150 nucleotides in length.
  • the poly-A tail is of about 30 nucleotides in length, optionally 34 nucleotides in length.
  • a nuclear localization signal is placed before the enzyme start codon at the N-terminus, optionally at the C-terminus.
  • the nucleic acid that is RNA has a 5’-m7G cap (cap 0, or cap 1, or cap 2).
  • the nucleic acid comprises a 5' cap structure, a 5'-UTR comprising a Kozak consensus sequence, a 5'-UTR comprising a sequence that increases RNA stability in vivo, a 3'-UTR comprising a sequence that increases RNA stability in vivo, and/or a 3' poly(A) tail.
  • the enzyme e.g., without limitation, a mobile element enzyme
  • the vector is a non-viral vector.
  • a nucleic acid encoding the enzyme in accordance with embodiments of the present disclosure is DNA.
  • a construct comprising a donor DNA is any suitable genetic construct, such as a nucleic acid construct, a plasmid, or a vector.
  • the construct is DNA, which is referred to herein as a donor DNA.
  • sequences of a nucleic acid encoding the donor DNA is codon optimized to provide improved mRNA stability and protein expression in mammalian systems.
  • the enzyme and the donor DNA are included in different vectors. In embodiments, the enzyme and the donor DNA are included in the same vector.
  • a nucleic acid encoding the enzyme capable of targeted genomic integration e.g., without limitation, a mobile element enzyme which is a chimeric mobile element enzyme
  • RNA e.g., helper RNA
  • a nucleic acid encoding a donor DNA is DNA.
  • a donor DNA often includes an open reading frame that encodes a transgene at the middle of donor DNA and terminal repeat sequences at the 5’ and 3’ end of the donor DNA. The translated mobile element enzyme binds to the 5’ and 3’ sequence of the donor DNA and carries out the transposition function.
  • donor DNA or transgene are used interchangeably with mobile elements, which are used to refer to polynucleotides capable of inserting copies of themselves into other polynucleotides.
  • the term donor DNA is well known to those skilled in the art and includes classes of donor DNAs that can be distinguished on the basis of sequence organization, for example inverted terminal sequences at each end, and/or directly repeated long terminal repeats (LTRs) at the ends.
  • LTRs long terminal repeats
  • the donor DNA as described herein may be described as a piggyBac like element, e.g., a donor DNA element that is characterized by its traceless excision, which recognizes TTAA (SEQ ID NO: 440) sequence and restores the sequence at the insert site back to the original TTAA (SEQ ID NO: 440) sequence after removal of the donor DNA.
  • the donor DNA is flanked by one or more end sequences or terminal ends.
  • the donor DNA is or comprises a gene encoding a complete polypeptide.
  • the donor DNA is or comprises a gene which is defective or substantially absent in a disease state.
  • the donor DNA includes a MLT mobile element enzyme (e.g., without limitation, a MLT mobile element enzyme having at least about 90% identity to the amino acid sequence of SEQ ID NO: 1, SEQ ID NO: 10, or SEQ ID NO: 11).
  • the mobile element enzyme can act on a left terminal end having a nucleotide sequence of SEQ ID NO: 431 or a sequence having at least about 90%, or at least about 93%, or at least about 95%, or at least about 97%, or at least about 98%, or at least about 99% identity thereto.
  • the donor DNA can act on a right terminal end having a nucleotide sequence of SEQ ID NO: 432 or a sequence having at least about 90%, or at least about 93%, or at least about 95%, or at least about 97%, or at least about 98%, or at least about 99% identity thereto.
  • the donor DNA acts on both MLT left donor DNA end and MLT right donor DNA end, having nucleotide sequences of SEQ ID NO: 431 and of SEQ ID NO: 432 respectively, or a sequence having at least about 90%, or at least about 93%, or at least about 95%, or at least about 97%, or at least about 98%, or at least about 99% identity thereto.
  • a MLT left donor DNA end is as follows TTAACACTTGGATTGCGGGAAACGAGTTAAGTCGGCTCGCGTGAATTGCGCGTACTCCGCGGGAGCCGTC TTAACTCGGTTCATATAGATTTGCGGTGGAGTGCGGGAAACGTGTAAACTCGGGCCGATTGTAACTGCGT ATTACCAAATATTTGTT (SEQ ID NO: 431)
  • a MLT right donor DNA end is as follows AATTATTTATGTACTGAATAGATAAAAAAATGTCTGTGATTGAATAAATTTTCATTTTACACAAGAAA CCGAAAATTTCATTTCAATCGAACCCATACTTCAAAAGATATAGGCATTTTAAACTAACTCTGATTTTGC GCGGGAAACCTAAATAATTGCCCGCGCCATCTTATATTTTGGCGGGAAATTCACCCGACACCGTGGTGTT AA (SEQ ID NO: 432).
  • a transgene is associated with various regulatory elements that are selected to ensure stable expression of a construct with the transgene.
  • a transgene is encoded by a non-viral vector (e.g., without limitation, a DNA plasmid) that can comprise one or more insulator sequences that prevent or mitigate activation or inactivation of nearby genes.
  • the insulators flank the donor DNA (transgene cassette) to reduce transcriptional silencing and position effects imparted by chromosomal sequences. As an additional effect, the insulators can eliminate functional interactions of the transgene enhancer and promoter sequences with neighboring chromosomal sequences.
  • the one or more insulator sequences comprise an HS4 insulator (1.2-kb 5’ -HS4 chicken ⁇ -globin (cHS4) insulator element) and an D4Z4 insulator (tandem macrosatellite repeats linked to Facio-Scapulo-Humeral Dystrophy (FSHD).
  • the sequences of the HS4 insulator and the D4Z4 insulator are as described in Rival-Gervier et al. Mol Ther.2013 Aug; 21(8):1536-50, which is incorporated herein by reference in its entirety.
  • the transgene is inserted into a GSHS location in a host genome.
  • GSHSs is defined as loci well-suited for gene transfer, as integrations within these sites are not associated with adverse effects such as proto-oncogene activation, tumor suppressor inactivation, or insertional mutagenesis.
  • GSHSs can defined by the following criteria: (1) distance of at least 50 kb from the 5’ end of any gene, (2) distance of at least 300 kb from any cancer-related gene, (3) distance of at least 300 kb from any microRNA (miRNA), (4) location outside a transcription unit, and (5) location outside ultra-conserved regions (UCRs) of the human genome. See Papapetrou et al. Nat Biotechnol 2011;29:73-8; Bejerano et al. Science 2004;304:1321-5.
  • CCR5 chemokine C-C motif receptor 5
  • a homozygous 32 bp deletion in the CCR5 gene confers resistance to HIV-1 virus infections in humans.
  • Disrupted CCR5 expression naturally occurring in about 1% of the Caucasian population, does not appear to result in any reduction in immunity.
  • a clinical trial has demonstrated safety and efficacy of disrupting CCR5 via targetable nucleases.
  • the donor DNA is under control of a tissue-specific promoter.
  • the tissue-specific promoter is, e.g., without limitation, a liver-specific promoter.
  • the liver-specific promoter is an LP1 promoter that, in embodiments, is a human LP1 promoter.
  • the LP1 promoter is described, e.g., in Nathwani et al. Blood vol. 2006;107(7):2653-61, and it is constructed, without limitation, as described in Nathawani et al.
  • the present nucleic acids include polymeric form of nucleotides of any length, either ribonucleotides or deoxyribonucleotides, or analogs or derivatives thereof.
  • transcriptionally- activated polynucleotides such as methylated or capped polynucleotides are provided.
  • the present compositions are mRNA or DNA.
  • the present non-viral vectors are linear or circular DNA molecules that comprise a polynucleotide encoding a polypeptide and is operably linked to control sequences, wherein the control sequences provide for expression of the polynucleotide encoding the polypeptide.
  • the non-viral vector comprises a promoter sequence, and transcriptional and translational stop signal sequences.
  • Such vectors may include, among others, chromosomal and episomal vectors, e.g., vectors bacterial plasmids, from donor DNAs, from yeast episomes, from insertion elements, from yeast chromosomal elements, and vectors from combinations thereof.
  • the present constructs may contain control regions that regulate as well as engender expression.
  • the construct comprising the enzyme and/or transgene is codon optimized.
  • Transgene codon optimization is used to optimize therapeutic potential of the transgene and its expression in the host organism. Codon optimization is performed to match the codon usage in the transgene with the abundance of transfer RNA (tRNA) for each codon in a host organism or cell. Codon optimization methods are known in the art and described in, for example, WO 2007/142954, which is incorporated by reference herein in its entirety. Optimization strategies can include, for example, the modification of translation initiation regions, alteration of mRNA structural elements, and the use of different codon biases.
  • the construct comprising the enzyme and/or transgene includes several other regulatory elements that are selected to ensure stable expression of the construct.
  • the non-viral vector is a DNA plasmid that can comprise one or more insulator sequences that prevent or mitigate activation or inactivation of nearby genes.
  • the one or more insulator sequences comprise an HS4 insulator (1.2-kb 5'-HS4 chicken ⁇ - globin (cHS4) insulator element) and an D4Z4 insulator (tandem macrosatellite repeats linked to Facio-Scapulo- Humeral Dystrophy (FSHD).
  • the sequences of the HS4 insulator and the D4Z4 insulator are as described in Rival-Gervier et al.
  • the gene of the construct comprising the enzyme and/or transgene is capable of transposition in the presence of a mobile element enzyme.
  • the non-viral vector in accordance with embodiments of the present disclosure comprises a nucleic acid construct encoding a mobile element enzyme.
  • the mobile element enzyme is an RNA mobile element enzyme plasmid.
  • the non-viral vector further comprises a nucleic acid construct encoding a DNA mobile element enzyme plasmid.
  • the mobile element enzyme is an in vitro-transcribed mRNA mobile element enzyme.
  • the mobile element enzyme is capable of excising and/or transposing the gene from the construct comprising the enzyme and/or transgene to site- or locus-specific genomic regions.
  • the enzyme and the donor DNA are included in the same vector.
  • the enzyme is disposed on the same (cis) or different vector (trans) than a donor DNA with a transgene. Accordingly, in embodiments, the enzyme and the donor DNA encompassing a transgene are in cis configuration such that they are included in the same vector. In embodiments, the enzyme and the donor DNA encompassing a transgene are in trans configuration such that they are included in different vectors.
  • the vector is any non-viral vector in accordance with the present disclosure.
  • a nucleic acid encoding the enzyme capable of targeted genomic integration in accordance with embodiments of the present disclosure is provided.
  • the nucleic acid is or comprises DNA or RNA.
  • the nucleic acid encoding the enzyme is DNA.
  • the nucleic acid encoding the enzyme capable of targeted genomic integration e.g., a chimeric mobile element enzyme
  • RNA such as, e.g., helper RNA.
  • the chimeric mobile element enzyme is incorporated into a vector.
  • the vector is a non-viral vector.
  • a nucleic acid encoding the transgene in accordance with embodiments of the present disclosure is provided.
  • the nucleic acid is or comprises DNA or RNA.
  • the nucleic acid encoding the transgene is DNA.
  • the nucleic acid encoding the e transgene is RNA such as, e.g., helper RNA.
  • the transgene is incorporated into a vector.
  • the vector is a non-viral vector.
  • the present enzyme can be in the form or an RNA or DNA and have one or two N-terminus nuclear localization signal (NLS) to shuttle the protein more efficiently into the nucleus.
  • NLS nuclear localization signal
  • the present enzyme further comprises one, two, three, four, five, or more NLSs.
  • NLS comprises the consensus sequence K(K/R)X(K/R) (SEQ ID NO: 348).
  • the NLS comprises the consensus sequence (K/R)(K/R)X 10-12 (K/R) 3/5 (SEQ ID NO: 349), where (K/R) 3/5 represents at least three of the five amino acids is either lysine or arginine.
  • the NLS comprises the c-myc NLS.
  • the c-myc NLS comprises the sequence PAAKRVKLD (SEQ ID NO: 350).
  • the NLS is the nucleoplasmin NLS.
  • the nucleoplasmin NLS comprises the sequence KRPAATKKAGQAKKKK (SEQ ID NO: 351).
  • the NLS comprises the SV40 Large T-antigen NLS.
  • the SV40 Large T-antigen NLS comprises the sequence PKKKRKV (SEQ ID NO: 352).
  • the NLS comprises three SV40 Large T-antigen NLSs (e.g., DPKKKRKVDPKKKRKVDPKKKRKV (SEQ ID NO: 353).
  • the NLS may comprise mutations/variations in the above sequences such that they contain 1 or more substitutions, additions or deletions (e.g., about 1, or about 2, or about 3, or about 4, or about 5, or about 10 substitutions, additions, or deletions).
  • a host cell comprising the nucleic acid in accordance with embodiments of the present disclosure is provided.
  • Lipids In embodiments, at least one of the first nucleic acid and the second nucleic acid is in the form of a lipid nanoparticle (LNP). In embodiments, a composition comprising the first and second nucleic acids is in the form of an LNP.
  • a nucleic acid encoding the enzyme and a nucleic acid encoding the transgene are contained within the same lipid nanoparticle (LNP).
  • the nucleic acid encoding the enzyme and the nucleic acid encoding the donor DNA are a mixture incorporated into or associated with the same LNP.
  • the nucleic acid encoding the enzyme and the nucleic acid encoding the donor DNA are in the form of a co-formulation incorporated into or associated with the same LNP.
  • the LNP is selected from 1,2-dioleoyl-3-trimethylammonium propane (DOTAP), a cationic cholesterol derivative mixed with dimethylaminoethane-carbamoyl (DC-Chol), phosphatidylcholine (PC), triolein (glyceryl trioleate), and 1,2-distearoyl-sn-glycero-3-phosphoethanolamine-N-[carboxy(polyethylene glycol)-2000] (DSPE-PEG), 1,2- dimyristoyl-rac-glycero-3-methoxypolyethyleneglycol – 2000 (DMG-PEG 2K), and 1,2 distearol -sn-glycerol- 3phosphocholine (DSPC) and/or comprising of one or more molecules selected from polyethylenimine (PEI) and poly(lactic-co-glycolic acid) (PLGA), and N-Acetylgalactosamine (GalNAc).
  • DOTAP 1,2-di
  • an LNP is as described, e.g., in Patel et al., J Control Release 2019; 303:91-100.
  • the LNP can comprise one or more of a structural lipid (e.g., DSPC), a PEG-conjugated lipid (CDM-PEG), a cationic lipid (MC3), cholesterol, and a targeting ligand (e.g., GalNAc).
  • a nanoparticle is a particle having a diameter of less than about 1000 nm.
  • nanoparticles of the present disclosure have a greatest dimension (e.g., diameter) of about 500 nm or less, or about 400 nm or less, or about 300 nm or less, or about 200 nm or less, or about 100 nm or less. In embodiments, nanoparticles of the present disclosure have a greatest dimension ranging between about 50 nm and about 150 nm, or between about 70 nm and about 130 nm, or between about 80 nm and about 120 nm, or between about 90 nm and about 110 nm. In embodiments, the nanoparticles of the present disclosure have a greatest dimension (e.g., a diameter) of about 100 nm.
  • a cell in accordance with the present disclosure is prepared via an in vivo genetic modification method.
  • a genetic modification in accordance with the present disclosure is performed via an ex vivo method.
  • a cell in accordance with the present disclosure is prepared by contacting the cell with an enzyme capable of targeted genomic integration (e.g., without limitation, a mammalian mobile element enzyme) in vivo.
  • the cell is contacted with the enzyme ex vivo.
  • the present method provides reduced insertional mutagenesis or oncogenesis as compared to a method with a non-chimeric mobile element enzyme.
  • the transgene of interest in accordance with embodiments of the present disclosure can encode various genes.
  • the enzyme (e.g., without limitations, a mobile element enzyme), and the donor DNA are included in the same pharmaceutical composition. In embodiments, the enzyme (e.g., without limitations, a mobile element enzyme) and the donor DNA are included in different pharmaceutical compositions. In embodiments, the enzyme and the donor DNA are co-transfected. In embodiments, the enzyme and the donor DNA are transfected separately. In embodiments, a method of producing a molecule with the transfected cell is provided, wherein the transfected cell is generated using a method of making a viral packaging or producer cell line in accordance with embodiments of the present disclosure.
  • the molecule is optionally a protein. In embodiments, the protein is an antibody.
  • the method of making a viral packaging or producer cell line of the present disclosure provides a stable cell with substantially reduced or ablated CAP expression. In embodiments, the method of making a viral packaging or producer cell line of the present disclosure provides a stable cell with REP expression or substantially enhanced REP expression. In embodiments, the method of making a viral packaging or producer cell line of the present disclosure provides a stable cell with substantially reduced or ablated CAP expression and REP expression or substantially enhanced REP expression. In embodiments, the method of making a viral packaging or producer cell line of the present disclosure provides a stable cell with REP expression or substantially enhanced VP1 expression. In embodiments, the method of making a viral packaging or producer cell line of the present disclosure provides a stable cell with substantially reduced or ablated CAP expression.
  • the viral packaging or producer cell line of the present disclosure is suitable for providing substantially reduced empty or cargo-free capsid.
  • a transfected cell for gene therapy is provided, wherein the transfected cell is generated using a method of making a viral packaging or producer cell line in accordance with embodiments of the present disclosure.
  • a method of delivering a cell therapy is provided, comprising administering to a patient in need thereof the transfected cell generated using a method of making a viral packaging or producer cell line in accordance with embodiments of the present disclosure.
  • a method of treating a disease or condition using a cell therapy comprising administering to a patient in need thereof the transfected cell generated using a method of making a viral packaging or producer cell line in accordance with embodiments of the present disclosure.
  • a method of treating a disease or condition using an antibody therapy comprising administering to a patient in need thereof the transfected cell generated using a method of making a viral packaging or producer cell line in accordance with embodiments of the present disclosure.
  • the disease or condition is or comprises cancer.
  • the cancer is or comprises an adrenal cancer, a biliary track cancer, a bladder cancer, a bone/bone marrow cancer, a brain cancer, a breast cancer, a cervical cancer, a colorectal cancer, a cancer of the esophagus, a gastric cancer, a head/neck cancer, a hepatobiliary cancer, a kidney cancer, a liver cancer, a lung cancer, an ovarian cancer, a pancreatic cancer, a pelvis cancer, a pleura cancer, a prostate cancer, a renal cancer, a skin cancer, a stomach cancer, a testis cancer, a thymus cancer, a thyroid cancer, a uterine cancer, a lymphoma, a melanoma, a multiple myeloma, or a leukemia.
  • an adrenal cancer a biliary track cancer, a bladder cancer, a bone/bone marrow cancer, a brain cancer, a breast cancer, a cervical cancer
  • the cancer is selected from one or more of the basal cell carcinoma, biliary tract cancer; bladder cancer; bone cancer; brain and central nervous system cancer; breast cancer; cancer of the peritoneum; cervical cancer; choriocarcinoma; colon and rectum cancer; connective tissue cancer; cancer of the digestive system; endometrial cancer; esophageal cancer; eye cancer; cancer of the head and neck; gastric cancer; glioblastoma; hepatic carcinoma; hepatoma; intra-epithelial neoplasm; kidney or renal cancer; larynx cancer; leukemia; liver cancer; lung cancer; melanoma; myeloma; neuroblastoma; oral cavity cancer; ovarian cancer; pancreatic cancer; prostate cancer; retinoblastoma; rhabdomyosarcoma; rectal cancer; cancer of the respiratory system; salivary gland carcinoma; sarcoma; skin cancer; squamous cell cancer; stomach cancer; testicular
  • the cancer is selected from one or more of basal cell carcinoma, biliary tract cancer; bladder cancer; bone cancer; brain and central nervous system cancer; breast cancer; cancer of the peritoneum; cervical cancer; choriocarcinoma; colon and rectum cancer; connective tissue cancer; cancer of the digestive system; endometrial cancer; esophageal cancer; eye cancer; cancer of the head and neck; gastric cancer (including gastrointestinal cancer); glioblastoma; hepatic carcinoma; hepatoma; intra-epithelial neoplasm; kidney or renal cancer; larynx cancer; leukemia; liver cancer; lung cancer (e.g., small-cell lung cancer, non-small cell lung cancer, adenocarcinoma of the lung, and squamous carcinoma of the lung); melanoma; myeloma; neuroblastoma; oral cavity cancer (lip, tongue, mouth, and pharynx); ovarian cancer; pancreatic cancer; prostate cancer;
  • the disease or condition is or comprises an infectious disease.
  • the infectious disease is a coronavirus infection, optionally selected from infection with SAR-CoV, MERS-CoV, and SARS-CoV-2, or variants thereof.
  • the infectious disease is or comprises a disease comprising a viral infection, a parasitic infection, or a bacterial infection.
  • the viral infection is caused by a virus of family Flaviviridae, a virus of family Picornaviridae, a virus of family Orthomyxoviridae, a virus of family Coronaviridae, a virus of family Retroviridae, a virus of family Paramyxoviridae, a virus of family Bunyaviridae, or a virus of family Reoviridae.
  • the virus of family Coronaviridae comprises a betacoronavirus or an alphacoronavirus, optionally wherein the betacoronavirus is selected from SARS-CoV-2, SARS-CoV, MERS-CoV, HCoV-HKU1, and HCoV-OC43, or the alphacoronavirus is selected from a HCoV-NL63 and HCoV-229E.
  • the infectious disease comprises a coronavirus infection 2019 (COVID-19).
  • the method requires a single administration.
  • the method requires a plurality of administrations.
  • the present disclosure provides an ex vivo gene therapy approach.
  • the method that is used to treat an inherited or acquired disease in a patient in need thereof comprises (a) contacting a cell obtained from a patient (autologous) or another individual (allogeneic) with a transfected cell in accordance with embodiments of the present disclosure; and (b) administering the cell to a patient in need thereof.
  • One of the advantages of ex vivo gene therapy is the ability to “sample” the transduced cells before patient administration. This facilitates efficacy and allows performing safety checks before introducing the cell(s) to the patient. For example, the transduction efficiency and/or the clonality of integration can be assessed before infusion of the product.
  • a composition comprising transfected cells in accordance with the present disclosure comprises a pharmaceutically acceptable carrier, excipient, or diluent.
  • compositions suitable for injectable use can include sterile aqueous solutions (where water soluble) or dispersions and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersion.
  • suitable carriers include physiological saline, bacteriostatic water, Cremophor ELTM (BASF, Parsippany, N.J.) or phosphate buffered saline (PBS).
  • the composition must be sterile, and the fluid should be easy to draw up by a syringe. It should be stable under the conditions of manufacture and storage and must be preserved against the contaminating action of microorganisms such as bacteria and fungi.
  • the carrier can be a solvent or dispersion medium containing, for example, water, ethanol, polyol (for example, glycerol, propylene glycol, and liquid polyethylene glycol, and the like), and suitable mixtures thereof.
  • the proper fluidity can be maintained, for example, by the use of a coating such as lecithin, by the maintenance of the required particle size in the case of dispersion and by the use of surfactants.
  • Prevention of the action of microorganisms can be achieved by various antibacterial and antifungal agents, for example, parabens, chlorobutanol, phenol, ascorbic acid, thimerosal, and the like.
  • isotonic agents for example, sugars, polyalcohols such as mannitol, sorbitol, and sodium chloride in the composition.
  • Prolonged absorption of the injectable compositions can be brought about by including in the composition an agent that delays absorption, for example, aluminum monostearate and gelatin.
  • Sterile injectable solutions can be prepared by incorporating the active compound in the required amount in an appropriate solvent with one or a combination of ingredients enumerated above, as required, followed by filtered sterilization.
  • dispersions are prepared by incorporating the active compound into a sterile vehicle, which contains a basic dispersion medium and the required other ingredients from those enumerated above.
  • sterile powders for the preparation of sterile injectable solutions, the preferred methods of preparation are vacuum drying and freeze-drying, which yield a powder of the active ingredient plus any additional desired ingredient from a previously sterile-filtered solution thereof.
  • Therapeutic compounds can be prepared with carriers that will protect the therapeutic compounds against rapid elimination from the body, such as a controlled release formulation, including implants and microencapsulated delivery systems.
  • Biodegradable, biocompatible polymers can be used, such as collagen, ethylene vinyl acetate, polyanhydrides (e.g., poly[1,3-bis(carboxyphenoxy)propane-co-sebacic-acid] (PCPP-SA) matrix, fatty acid dimer- sebacic acid (FAD-SA) copolymer, poly(lactide-co-glycolide)), polyglycolic acid, collagen, polyorthoesters, polyethyleneglycol-coated liposomes, and polylactic acid.
  • PCPP-SA poly[1,3-bis(carboxyphenoxy)propane-co-sebacic-acid]
  • FAD-SA fatty acid dimer- sebacic acid
  • polyglycolic acid collagen
  • polyorthoesters polyethyleneglycol-coated liposomes
  • polylactic acid polylactic acid
  • the stable integration comprises an introduction of a polynucleotide into a chromosome or mini-chromosome of the cell and, therefore, becomes a relatively permanent part of the cellular genome.
  • a transgenic organism that may comprise cells which have been transformed by the methods of the present disclosure.
  • the organism may be a mammal or an insect.
  • the organism When the organism is a mammal, the organism may include, but is not limited to, a mouse, a rat, a monkey, a dog, a rabbit, and the like. When the organism is an insect, the organism may include, but is not limited to, a fruit fly, a mosquito, a bollworm, and the like.
  • the cells produced in accordance with embodiments of the present disclosure, and/or components for generating cells is included in a container, kit, pack, or dispenser together with instructions for administration. Also provided herein are kits comprising: one or more genetic constructs encoding the present enzyme and donor DNA and instructions and/or reagents for the use of the same.
  • kits comprising: i) a transfected cell in accordance with embodiments of the present disclosure, ii) instructions for the use of the transfected cell.
  • a kit is provided for creating an AAV of packaging cell line or an AAV library of packaging cell lines, and instructions for creating a library. A subsequent user-defined transfection of the viral genome can be performed to generate a producer cell line.
  • the kit includes instructions for transfection of an AAV packaging cell line with a desired transgene.
  • kits includes an AAV library of packaging cell lines and instructions for creating a library
  • a kit is provided that comprises an enzyme (e.g., without limitation, a recombinant mammalian mobile element enzyme) or a nucleic acid in accordance with embodiments of the present disclosure, and instructions for introducing DNA and/or RNA into a cell using the enzyme.
  • an enzyme e.g., without limitation, a recombinant mammalian mobile element enzyme
  • nucleic acid in accordance with embodiments of the present disclosure
  • instructions for introducing DNA and/or RNA into a cell using the enzyme e.g., without limitation, a recombinant mammalian mobile element enzyme
  • an “effective amount,” when used in connection with medical uses is an amount that is effective for providing a measurable treatment, prevention, or reduction in the rate of pathogenesis of a disease of interest.
  • the term “in vivo” refers to an event that takes place in a subject’s body.
  • the term “ex vivo” refers to an event which involves treating or performing a procedure on a cell, tissue and/or organ which has been removed from a subject’s body.
  • the cell, tissue and/or organ may be returned to the subject’s body in a method of treatment or surgery.
  • the term “variant” encompasses but is not limited to nucleic acids or proteins which comprise a nucleic acid or amino acid sequence which differs from the nucleic acid or amino acid sequence of a reference by way of one or more substitutions, deletions and/or additions at certain positions.
  • the variant may comprise one or more conservative substitutions. Conservative substitutions may involve, e.g., the substitution of similarly charged or uncharged amino acids.
  • Carrier or “vehicle” as used herein refer to carrier materials suitable for drug administration.
  • Carriers and vehicles useful herein include any such materials known in the art, e.g., any liquid, gel, solvent, liquid diluent, solubilizer, surfactant, lipid, or the like, which is nontoxic, and which does not interact with other components of the composition in a deleterious manner.
  • pharmaceutically acceptable refers to those compounds, materials, compositions, and/or dosage forms that are, within the scope of sound medical judgment, suitable for use in contact with the tissues of human beings and animals without excessive toxicity, irritation, allergic response, or other problems or complications commensurate with a reasonable benefit/risk ratio.
  • pharmaceutically acceptable carrier or “pharmaceutically acceptable excipient” are intended to include any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents, and inert ingredients.
  • pharmaceutically acceptable carriers or pharmaceutically acceptable excipients for active pharmaceutical ingredients is well known in the art. Except insofar as any conventional pharmaceutically acceptable carrier or pharmaceutically acceptable excipient is incompatible with the active pharmaceutical ingredient, its use in the therapeutic compositions of the disclosure is contemplated. Additional active pharmaceutical ingredients, such as other drugs, can also be incorporated into the described compositions and methods. As referred to herein, all compositional percentages are by weight of the total composition, unless otherwise specified.
  • the word “include,” and its variants, is intended to be non-limiting, such that recitation of items in a list is not to the exclusion of other like items that may also be useful in the compositions and methods of this technology.
  • the terms “can” and “may” and their variants are intended to be non-limiting, such that recitation that an embodiment can or may comprise certain elements or features does not exclude other embodiments of the present technology that do not contain those elements or features.
  • compositions described herein needed for achieving a therapeutic effect may be determined empirically in accordance with conventional procedures for the particular purpose.
  • the therapeutic agents are given at a pharmacologically effective dose.
  • a “pharmacologically effective amount,” “pharmacologically effective dose,” “therapeutically effective amount,” or “effective amount” refers to an amount sufficient to produce the desired physiological effect or amount capable of achieving the desired result, particularly for treating the disorder or disease.
  • an effective amount as used herein would include an amount sufficient to, for example, delay the development of a symptom of the disorder or disease, alter the course of a symptom of the disorder or disease (e.g., slow the progression of a symptom of the disease), reduce or eliminate one or more symptoms or manifestations of the disorder or disease, and reverse a symptom of a disorder or disease.
  • Therapeutic benefit also includes halting or slowing the progression of the underlying disease or disorder, regardless of whether improvement is realized.
  • Effective amounts, toxicity, and therapeutic efficacy can be determined by standard pharmaceutical procedures in cell cultures or experimental animals, e.g., for determining the LD 50 (the dose lethal to about 50% of the population) and the ED 50 (the dose therapeutically effective in about 50% of the population).
  • the dosage can vary depending upon the dosage form employed and the route of administration utilized.
  • the dose ratio between toxic and therapeutic effects is the therapeutic index and can be expressed as the ratio LD 50 /ED 50 .
  • compositions and methods that exhibit large therapeutic indices are preferred.
  • a therapeutically effective dose can be estimated initially from in vitro assays, including, for example, cell culture assays.
  • a dose can be formulated in animal models to achieve a circulating plasma concentration range that includes the IC 50 as determined in cell culture, or in an appropriate animal model. Levels of the described compositions in plasma can be measured, for example, by high performance liquid chromatography. The effects of any particular dosage can be monitored by a suitable bioassay.
  • the dosage can be determined by a physician and adjusted, as necessary, to suit observed effects of the treatment.
  • “methods of treatment” are equally applicable to use of a composition for treating the diseases or disorders described herein and/or compositions for use and/or uses in the manufacture of a medicaments for treating the diseases or disorders described herein.
  • the present disclosure provides for any of the sequence provided herein, including the below, and a variant sequence having at least about 90%, or at least about 93%, or at least about 95%, or at least about 97%, or at least about 98%, or at least about 99% identity thereto, or at least about 10 mutations, or at least about 9 mutations, or at least about 8 mutations, or at least about 7 mutations, or at least about 6 mutations, or at least about 5 mutations, or at least about 4 mutations, or at least about 3 mutations, or at least about 2 mutations, or at least about 1 mutation.
  • SEQ ID NO: 1 MLT mobile element enzyme protein (amino acid sequence of a variant of the hyperactive mobile element enzyme with S at position 8 and C at position 13 (572 amino acids) MAQHSDYSDDEFCADKLSNYSCDSDLENASTSDEDSSDDEVMVRPRTLRRRRISSSSSDSESDIEGGREEWSHV DNPPVLEDFLGHQGLNTDAVINNIEDAVKLFIGDDFFEFLVEESNRYYNQNRNNFKLSKKSLKWKDITPQEMKK FLGLIVLMGQVRKDRRDDYWTTEPWTETPYFGKTMTRDRFRQIWKAWHFNNNADIVNESDRLCKVRPVLDYFVP KFINIYKPHQQLSLDEGIVPWRGRLFFRVYNAGKIVKYGILVRLLCESDTGYICNMEIYCGEGKRLLETIQTVV SPYTDSWYHIYMDNYYNSVANCEALMKNKFRICGTIRKNRGIPKDFQTISLKKGETKFIRKNDILL
  • FIG.2E illustrates a non-limiting example of a system comprising a nucleic acid (e.g., helper RNA) encoding an enzyme capable of performing targeted genomic integration and a nucleic acid encoding a mobile element enzyme (donor DNA).
  • a nucleic acid e.g., helper RNA
  • the helper RNA is translated into a bioengineered enzyme (e.g., integrase, recombinase, or mobile element enzyme) that recognizes specific ends and seamlessly inserts the donor DNA into the human genome in a site-specific manner without a footprint.
  • enzymes e.g., chimeric mobile element enzymes
  • FIGs.2A-D depict representations of chimeric mobile element enzyme designed using human GSHS TALE or Cas9/gRNA DBD.
  • FIG.2A. TALEs includes nuclear localization signals (NLS) and an activation domain (AD) to function as transcriptional activators.
  • a central tandem repeat domain confers specific DNA-binding and host specificity.
  • Translocation signal (TD) and four cryptic repeats required for initiation of DNA binding and for the recognition of 5’ -T 0 are located at the N-terminus (checkered rectangles).
  • Each 34 amino acid (aa) long repeat in the CRD binds to one nucleotide with specificity determined mainly by aa at position 13.
  • One sample repeat is shown below the protein scheme. Numbers 12/13 refer to aa positions within the repeat. See Jankele et al., Brief Funct Genomics 2014;13:409-19.
  • FIG.2C Repeat types are shown that have specificity for one or several nucleotides. Only bases of the DNA leading strand are shown.
  • FIG.2D Binding of the TALE to GSHS physically sequesters the mobile element enzyme to the same location and promotes transposition to the nearby TTAA sequences. All RVD are preceded by a thymine (T) to bind to the NTR shown in FIG.2A. All of these GSHS regions are in open chromatin and are susceptible to mobile element enzyme activity).
  • FIG.2C also illustrates (bottom) a chimeric mobile element enzyme construct comprising dCas9 linked to one or more guide RNAs.
  • An engineered chimeric mobile element enzyme may include: a guide RNA (gRNA) and an inactivated Cas protein.
  • the gRNA is a short synthetic RNA composed of a scaffold sequence necessary for Cas-binding and a user-defined ⁇ 20 nucleotide spacer that defines the genomic target to be modified.
  • the genomic target of the Cas protein is based upon the sequence present in the gRNA.
  • FIG.3B shows gRNA sequences that physically sequester the mobile element enzyme to GSHS and promotes transposition to the nearby TTAA sequences.
  • FIGs.3A-3E depict examples of construct templates (FIGs.3A-3D) and a system (FIG.3E) comprising a donor DNA and helper RNA.
  • FIG.3A depicts a plasmid construct template that transcribes mobile element enzyme RNA that is later processed with a 5’- m7G cap. Other mobile element enzymes can be substituted.
  • FIG.3B depicts a donor DNA construct template with the transgene VLDLR. Other tissue specific promoters and transgenes can be substituted.
  • FIG. 3C depicts a chimeric mobile element enzyme construct template with a TALE DNA binder. Other TALEs and mobile element enzymes can be substituted.
  • FIG.3D depicts a chimeric mobile element enzyme construct template with a dCas9/gRNA DNA binder. Other dCas9/gRNA and mobile element enzymes can be substituted.
  • FIG.3E depicts a non-limiting example of the system of FIG.1A.
  • the system comprises a nucleic acid (donor DNA), and a nucleic acid (helper RNA) encoding a mobile element enzyme (hyperactive ENGINEERED MOBILE ELEMENT ENZYME).
  • the donor DNA can be any gene of interest (GOI) including those replace, inactive, or provide suicide or helper functions.
  • the GOI can be driven by a predetermined promoter and flanked by insulators to prevent gene silencing.
  • the ITRs are specific for the mammal-derived mobile element enzyme (ENGINEERED MOBILE ELEMENT ENZYME).
  • FIG.3E, panel (B) depicts the helper RNA that is 5’- m7G capped (cap1) with flanking globin 5’- and 3’-UTRs, a 34 polyalanine tail region, and pseudouridine modification.
  • the mobile element enzyme e.g., a hyperactive mobile element enzyme in accordance with the present disclosure
  • FIG. 1A is a non-limiting representation of an AAV production strategy in accordance with embodiments of the present disclosure, using a donor vector to create a HEK293 producer cell line or another cell line including the E1a gene (e.g., engineered CHO-K1 or Vero cell line with E1a) that forms a replication deficient (rep-) AAV particle containing a transgene of interest.
  • E1a gene e.g., engineered CHO-K1 or Vero cell line with E1a
  • the single or dual donor comprising a transgene of interest is incorporated (e.g., by transfection such as electroporation) into a HEK293 cell line or another cell line including the E1a gene.
  • the HEK293 cells are expanded to create a producer cell line and culturing the producer cells creates replication deficient (rep-) AAV particles with the transgene of interest.
  • FIG.1B is a representation of an example of an inducible Rep/Cap and helper AAV donor plasmid construct (Kana r ) used with a helper RNA or DNA to create an AAV producer cell line, in accordance with embodiments of the present disclosure.
  • the donor DNA plasmid of FIG.1B includes helper E2A, E4 and VA genes flanked by insulators and mobile element enzyme recognition ends, to create an AAV producer cell line.
  • FIG.1C is a representation of an example of a nucleic acid (plasmid) encoding a transgene (gene of interest (GOI)) included between AAV inverted terminal repeats (ITRs), in accordance with embodiments of the present disclosure.
  • the plasmids shown in FIGs.1B and 1C can be combined or can be used separately for transfection into an E1+ cell line to produce AAV particle comprising the GOI.
  • FIG.4 depicts a non-limiting representation of a conventional rAAV production system.
  • AAV Rep/Cap genes the ITR-flanked gene of interest (GOI), as well as Ad-helper genes are provided as three separate plasmids, respectively, to a E1a/E1b containing HEK293 cell line or other engineered cell line that contains E1a (e.g., engineered CHO-K1 or Vero cells).
  • the present strategy is to include the AAV rep and cap genes, the AAV vector DNA sequences, and the essential Ad helper genes in a single donor plasmid flanked end sequences recognized by the mobile element enzyme in accordance with embodiments of the present disclosure.
  • the AAV rep and cap genes are included under the control of t-REx, leading to rep and cap gene amplification, the AAV vector DNA sequences, and the essential Ad helper genes in a single donor plasmid flanked by the end sequences recognized by the mobile element enzyme in accordance with embodiments of the present disclosure (FIG.1B).
  • the ability of the integrated AAV helper plasmid to direct the rescue, replication, and packaging of an AAV ITR-flanked transgene (e.g., the transgene of FIG.1C) will be assessed.
  • the integrated AAV helper plasmid can direct the rescue, replication, and packaging of an AAV ITR-flanked transgene.
  • This mobile element enzyme-mediated recombination system is expected to generate an integrated AAV helper plasmid that can facilitate the production of high-titer recombinant AAV particles in a simple two-plasmid transfection protocol.
  • the inducible nature of the rep and cap genes is employed by the use of reagents (e.g., rAAV-5 plasmid reagents) useful for the construction of rAAV-5 vectors bearing other reporter and/or therapeutic transgenes.
  • reagents e.g., rAAV-5 plasmid reagents
  • Example 4 Schematic Illustration for Constructs Used for Establishment of AAV producer stable cell lines, AAV2-H (Plasmid shown in FIG.5), AAV2-HB (Plasmid shown in FIG.6), and AAV-CDH (Plasmid shown in FIG.7) in HEK- 293R-22 tetR-expressing HEK-293 cells.
  • Plasmid with pSF-ITRP5TO1-AAV2H (FIG.5): pSF-ITRP5TO1-AAV2H is an engineered mobile element enzyme ITR containing plasmid that encodes 1) AAV2_REP78/68 ORFs under the control of P5TO1 promoter with AAV2_REP52/40 ORFs and CAPs (VP1/2/3) under their native P19 or P40 promoter, respectively, 2) Ad5 E2A_IRES_Ad5 E4 ORF6 under the control of the tetO-containing SV40 promoter, 3) VA RNAs under the control of HSV-1 ICP4 promoter, and 4) hygro-B resistant gene under the control of TK promoter.
  • ITR engineered mobile element enzyme ITR containing plasmid that encodes 1) AAV2_REP78/68 ORFs under the control of P5TO1 promoter with AAV2_REP52/40 ORFs and CAPs (VP1/2/3)
  • Plasmid with pSF-ITRP5TO1-AAV2HB (FIG. 6): pSFP5TO1-AAV2HB is an engineered mobile element enzyme ITR containing plasmid that encodes 1) AAV2_REP78/68 ORFs under the control of P5TO1 promoter and AAV2_REP52/40 ORFs under the native P19 promoter, 2) AAV2 CAPs under the tetO-containing P40 promoter with AAV9 P40 intron, 3) Ad5 E2A_IRES_Ad5 E4 ORF6 under the control of the tetO-containing SV40 promoter, 4) VA RNAs under the control of HSV-1 ICP4 promoter, and 5) hygro-B resistant gene under the control of TK promoter.
  • Plasmid pSFP5TO1-AAVCDH (FIG.7): pSFP5TO1-AAVCDH is an engineered mobile element enzyme ITR containing plasmid that encodes 1) AAV2_REP 78/68 ORFs under the control of P5TO1 promoter and AAV2_REP52/40 ORFs under the native P19 promoter, 2) Ad5 E2A_IRES_Ad5 E4 ORF6 under the control of the tetO-containing SV40 promoter, 3) VA RNAs under the control of HSV-1 ICP4 promoter, and 4) hygro-B resistant gene under the control of TK promoter.
  • Plasmid pSFA2-EP40CAP2WZ1_SB (FIG.8): This construct encodes AAV2 CAP under the control of the tetO-containing hCMV-derived enhancer element 3/AAV2 P40 promoter with a modified AAV2 P40 TATA element and modified AAV2 P40 intron_1 (a at position 276 substituted for C) and WPRE/modified ICP27 poly A, followed by ICP0 promoter-Zeocin resistant gene-ICP0 poly A cassette flanked with ITRs for Sleeping Beauty.
  • Plasmid pSFA2-EP40CAP2WZ2_SB (FIG.9): This construct encodes AAV2 CAP under the control of the tetO-containing hCMV enhancer element 3/AAV2 P40 promoter with a modified AAV2 P40 TATA element and modified AAV2 P40 intron_2 and WPRE/modified ICP27 poly A, followed by ICP0 promoter-Zeocin resistant gene-ICP0 poly A cassette flanked with SB ITRs. Establishment of AAV Producer/packaging cell lines a.
  • HEK-293R-22 cells were seeded at about 5 x 10e6 cells per 100 mm dish with normal HEK-293 cells growth medium.
  • Cells were co-transfected with pSF-ITRP5TO1-AAV2H, CMV-Engineered mobile element enzymeB VB200927-4525erk, and pSF-mCMVR at a DNA:Lipofectamine 2000 ratio of 1:2.
  • Transfection medium was removed at 3 and a half hour post transfection followed by addition of normal 293 cells growth medium.
  • Transfected cells were passed on day 3 post transfection and grew in 293 cells growth medium in the presence of hygro-B at a concentration of 100 ug/ml.
  • Hygro-B resistant single stable AAV-H colonies were then selected and analyzed by 1) ddPCR analyses with REP, E2A, ORF6, VA RNA and hygro-B resistant gene-specific primers, 2) western blot analysis with anti-AAV-2 REPs and CAPs specific antibodies, and 3) their ability to support AAV genome amplification following transduction with AAV2-EGFP viral particles.
  • HEK-293R-22 cells were seeded at about 5 x 10e6 cells per 100 mm dish with normal HEK- 293 cells growth medium.
  • Cells were co-transfected with VB220909-1308jwt_pSF-ITRP5TO1-AAV2HB, CMV- Engineered mobile element enzymeB VB200927-4525erk, and pSF-mCMVR at a DNA:Lipofectamine 2000 ratio of 1:2.
  • Transfection medium was removed at 3 and a half hour post transfection followed by addition of normal 293 cells growth medium.
  • Transfected cells were passed on day 2 post transfection and grew in 293 cells growth medium in the presence of hygro-B at a concentration of 100 ug/ml.
  • Hygro-B resistant single stable AAV-HB colonies were then selected and analyzed by 1) ddPCR analyses with REP, E2A, ORF6, VA RNA and hygro-B resistant gene-specific primers, 2) western blot analysis with anti-AAV-2 REPs and CAPs specific antibodies, and 3) their ability to support AAV genome amplification following transduction with AAV2-EGFP viral particles.
  • AAV serotype adaptable (Cap minus or Cap-) stable cell lines AAV-CDH (Plasmid shown in FIG.7) cells
  • HEK-293R-22 cells were seeded at about 5 x 10e6 cells per 100 mm dish with normal HEK-293 cells growth medium.
  • Cells were co-transfected with pSF-ITRP5TO1- AAVCDH, CMV-Engineered mobile element enzymeB VB200927-4525erk, and pSF-mCMVR at a DNA:Lipofectamine 2000 ratio of 1:2. Transfection medium was removed at 3 and a half hour post transfection followed by addition of normal 293 cells growth medium.
  • Transfected cells were passed on day 3 post transfection and grew in 293 cells growth medium in the presence of hygro-B at a concentration of 100 ug/ml or 50 ug/ml.
  • Hygro-B resistant single stable AAV-CDH Plasmid shown in FIG.7 colonies were then selected and analyzed by 1) ddPCR analyses with REP, E2A, ORF6, VA RNA and hygro-B resistant gene-specific primers, 2) western blot analysis with anti-AAV-2 REPs specific antibody, and 3) their ability to support AAV genome amplification following transduction with AAV2-EGFP viral particles. d.
  • AAV2-capsid minus stable lines Clone CDH-48, CDH-24 and CDH-129 cells HEK-293 were seeded at about 4 to 5.5 x 10e6 cells per 100 mm dish with normal HEK-293 cells growth medium.
  • Cells were co-transfected with of pSFA2-EP40CAP2WZ1-SB, pSFA2-EP40CAP2WZ2-SB, pSF-mCMVR, and sleeping beauty transposase SB100X Helper mRNA (VB210111- 1032qxg) at a DNA:Lipofectamine 2000 ratio of 1:2. Transfection medium was removed at 3 and a half hour post transfection followed by addition of normal 293 cells growth medium. Transfected cells were passed on day 2 post transfection with normal HEK-293 cells growth medium. Cells were then grown in HEK-293 cells growth medium in the presence of 100 ug/ml of zeocin.
  • Zeocin resistant single stable colonies were then selected and analyzed by 1) ddPCR analyses with REP, Cap, E2A, ORF6, VA RNA and zeocin resistant gene-specific primers, 2) western blot analysis with anti-AAV-2 REPs and CAPs specific antibodies, and 3) their ability to support AAV genome amplification following transduction with AAV2-EGFP viral particles.
  • ddPCR analyses for an engineered mobile element enzyme mediated Transposon Integration, AAV Genome Amplification/Rep Function, Cap Copy Insertion via Sleeping Beauty, and physical titer of rAAV particles.
  • AAV genome amplification is supported in either AAV2 producer cell line clone H-17 (FIG.10A – 10B, Cap+) or AAV serotype adaptable packaging cell line clone CDH-13 (FIG.11A – 11B, Cap-).
  • AAV genome amplification was used as a reference for expression of functional AAV Rep proteins in the AAV-H Clone H-17 or AAV-CDH (Plasmid shown in FIG.7) clone CDH-13 cells.
  • Commercially available rAAV2-EGFP particles from SignaGen at 75 VGC/cell was used to transduce 3 x 10e6 cells/dish of Clone H-17, CDH-13, or HEK-293 (AAV Rep- control) cells.
  • GFP images (FIG.10A – FIG.11B) were taken ⁇ 120 hours post-transduction using Zeiss AxioVert A1 microscope and Axiocam 705 mono camera, followed by cell harvesting, genomic DNA extraction, and ddPCR analyses for quantification of the rAAV2-EGFP genome copy number per nanogram (ng) of total genomic DNA.
  • ddPCR results at right The results clearly indicated that the rAAV-EGFP replicated in doxycycline dose responsive manner (ddPCR results at right), suggesting the inducible expression of functional AAV Rep proteins in the Clone H-17 and CDH-13 cells.
  • FIG.12A shows the results of an western blot analysis illustrating cells from selected AAV2-HB (Plasmid shown in FIG. 6) cell clones HB-36, HB-42, HB-118, and HB-120 were treated with10ng/ml doxycycline for 72 hours followed by extraction with RIPA buffer in the presence of proteinase inhibitor. The resulting cell lysate supernatants were subjected to electrophoresis and western blot analyses.
  • FIG.12B shows the results of a ddPCR analysis for AAV Genome Amplification / Rep function Assay.
  • the AAV-HB stable clone #36, 42, 118 and 120 cells were transduced with the rAAV2-EGFP particles from SignaGen at low MOI, and genomic DNA extracted/ddPCR performed for quantification of the rAAV2-GFP genome copy number per nanogram (ng) of total genomic DNA analyzed.
  • AAV2 full producer stable cell pools CDH-24/Cap-SB, CDH-48/Cap-SB, and CDH-129/Cap-SB with sleeping beauty transposase-mediated integration of AAV2 CAPs-expressing cassettes into AAV-CDH (Plasmid shown in FIG.7) serotype adaptable (Cap minus or Cap-) stable cell lines CDH-24, CDH48, and CDH- 129.
  • FIG.14A shows ddPCR analyses of SB-mediated AAV2 Cap transposon integration in each Zeocin-selected stable pools. Average Cap copy number integration in each pool is summarized at the bottle of the penel.
  • FIG.14B shows western blot analysis of SB-mediated AAV2 Cap transposon integration in each Zeocin-selected stable pools.
  • CDH-48/Cap-SB stable pool increased with doxycycline dosages and over time.
  • Cells from CDH-48/Cap-SB stable pools were incubated either in the presence of 0, 5, 10, 25, 50, or 100ng/ml doxycycline and harvest at 72 hours post induction; or incubated with 50ng/ml doxycycline and harvested at 24, 48, 72, or 96 hours post induction (FIG.16).
  • Cell extraction with RIPA buffer containing proteinase inhibitors, western blot, and ddPCR analyses of the CDH-48/Cap-SB (AAV2-capsid minus stable lines Clone CDH-48) samples were caried out as described previously.
  • the results show that expression of AAV2 Cap and Rep proteins increased over time in the presence of 50ng/ml doxycycline.
  • the Cap and Rep expression also increased in response to increasing amounts of doxycycline.
  • the cells from the CDH-48/Cap-SB stable pools were transduced with the rAAV2-EGFP particles from SignaGen at 500 VGC/cell in the presence or absence of 50ng/ml. 4 days post-transduction, culture media were removed, and cells harvested and washed twice with fresh growth media to remove the residue doxycycline.
  • rAAV-EGFP containing supernatants were collected following centrifugation at 2500rpm for 10 minutes AAV physical titer in each sample was measured by ddPCR as described above and data presented in FIG.17A.
  • the unconcentrated supernatants were used for transduction of CDH-48 Cap- cells at 1.5ml/well of the 6-well tissue culture plates in duplicates.
  • the transduction media were replaced next day with fresh growth media and cells were harvested after additional 2 days incubation followed by fgenomic DNA extraction.
  • ddPCR analyses of the genomic DNA samples were performed to evaluate copies of the rAAV-GFP copies introduced into CHD-48 Cap- cells through transduction and results presented in the FIG.17B.
  • the ddPCR results from both panels indicated that rAAV-EGFP particles were produced in response to doxycycline induction and the produced rAAV particles were competent for transduction. Characterization of Zeocin-resistant single clones selected from the CDH-48/Cap-SB stable pool.
  • Zeocin resistant single stable colonies were selected from the CDH-48/Cap-SB stable pool and analyzed by 1) ddPCR analysis for SB-mediated AAV2-Cap cassette integration (FIG.18A); 2) western blot analysis with antibodies specific for AAV2 CAP (FIG.18B), and AAV2 Rep (FIG.19A); and 3) ddPCR assays to evaluate their ability to support AAV genome amplification following transduction with low VGC/cell of AAV2-EGFP viral particles (FIG.19B).
  • FIG.18A indicated that SB-mediated AAV2-cap cassette integration ranged from 20-90 copies per ng of total genomic DNA.
  • Transposon copy number ddPCR The Transposon Copy Number ddPCR Assay quantifies the copy numbers of five elements in the Engineered mobile element enzyme transposon per genome of HEK293R-22 based cells. These elements are AAV2 Rep, AAV2 Cap, Ad5 E2A, Ad5 E4 Orf6, and VA RNA. Genomic DNA was extracted from samples with the Zymo Research Quick-DNA MiniPrep Kit (D3025) according to the manufacturer’s protocol.
  • Sample DNA concentrations were quantified by the Thermo Qubit 1X dsDNA Broad Range Assay Kit (Q33266) according to the manufacturer’s protocols and normalized to the same concentration in TE.
  • ddPCR reactions were assembled with Manufacturer’s Supermix for Probes (no dUTP), a FAM-labeled hydrolysis probe-based ddPCR assays for one of the transposon elements, a HEX-labeled hydrolysis probe-based ddPCR assay for RPP30, HindIII restriction enzyme, and water according to Manufacturer’s ddPCR protocol. Sample DNA was added to reaction wells at inputs of 4 ng and 20 ng per well to ensure that at least one reaction per sample was within the range of the assay.
  • AAV Genome Amplification / Rep function Assay quantifies the copy number of AAV-GFP genomes per nanogram (ng) of genomic DNA analyzed. Genomic DNA was extracted from cells with the Zymo Research Quick-DNA MiniPrep Kit (D3025) according to the manufacturer’s protocol. Sample DNA concentrations were quantified by the Thermo Qubit 1X dsDNA Broad Range Assay Kit (Q33266) according to the manufacturer’s protocols and normalized to the same concentration in TE.
  • ddPCR reactions were assembled with Manufacturer’s Supermix for Probes (no dUTP), a FAM-labeled hydrolysis probe-based ddPCR assay for EGFP, HindIII restriction enzyme, and water according to Manufacturer’s ddPCR protocol.
  • Sample DNA was added to reaction wells at inputs of 0.02 to 20 ng per well to ensure that at least one reaction per sample was within the range of the assay.
  • Droplets were generated by the automated QX200 AutoDG system and thermal cycled according to Manufacturer’s recommended parameters, with an annealing Tm of 60 ⁇ . Thermal cycled droplets were analyzed by Direct Quantitation on the QX200 Droplet Reader. Sample data was inspected for sufficient droplets (>10,000) and properly identified positive and negative droplets prior to analysis.
  • the QX200 droplet reader software automatically calculates the target (EGFP) copies per uL of reaction volume.
  • c Cap Copy Number ddPCR Assay: The Cap Copy Number ddPCR Assay quantifies the copy number of AAV2 Cap genes per genome of HEK293-based cells. Genomic DNA was extracted from samples with the Zymo Research Quick-DNA MiniPrep Kit (D3025) according to the manufacturer’s protocol.
  • Sample DNA concentrations were quantified by the Thermo Qubit 1X dsDNA Broad Range Assay Kit (Q33266) according to the manufacturer’s protocols and normalized to the same concentration in TE.
  • ddPCR reactions were assembled with Manufacturer’s Supermix for Probes (no dUTP), a FAM-labeled hydrolysis probe-based ddPCR assay for AAV2 Cap, a HEX-labeled hydrolysis probe-based ddPCR assay for RPP30, HindIII restriction enzyme, and water according to Manufacturer’s ddPCR protocol. Sample DNA was added to reaction wells at inputs of 4 ng and 20 ng per well to ensure that at least one reaction per sample was within the range of the assay.
  • AAV physical titer ddPCR quantifies the copy number of AAV-GFP genomes per mL of AAV sample analyzed.
  • a series of eight 10-fold serial dilutions (10E0-10E-7) of AAV samples were created to ensure that several dilutions would fall within the range of ddPCR quantitation.
  • Serial dilutions were created in nuclease-free water with 0.05% Pluronic F-68 (Gibco 24040-032).
  • AAV particles were lysed by incubation at 95 ⁇ for 10 minutes, followed by cooling to 4 ⁇ at a rate of 3 ⁇ per minute.
  • ddPCR reactions were assembled with Manufacturer ⁇ s Supermix for Probes (no dUTP), a FAM-labeled hydrolysis probe-based ddPCR assay for EGFP, HindIII restriction enzyme, and water according to Manufacturer ⁇ s ddPCR protocol. AAV sample dilutions were added to reaction wells at 5 uL per well. Droplets were generated by the automated QX200 AutoDG system and thermal cycled according to Manufacturer ⁇ s recommended parameters, with an annealing Tm of 60 ⁇ . Thermal cycled droplets were analyzed by Direct Quantitation on the QX200 Droplet Reader. Sample data was inspected for sufficient droplets (>10,000) and properly identified positive and negative droplets prior to analysis.
  • the QX200 droplet reader software automatically calculates the target (EGFP) copies per uL of reaction volume.
  • Western Blot analyses for detecting AAV Rep and Cap Expression a.
  • Cell lysate preparation Upon wash and scrape, ⁇ 1+E7 cells in 10 ml of same PBS containing 1mM PMSF and 100ug/ml TPCK or HaltTM Protease and Phosphatase Inhibitor Cocktail, EDTA-free (100X) (Cat# 78441) were pelleted down by centrifugation at 3000 rpm for 15 min at 4c and resuspend in 600 ul of RIPA buffer containing 1mM PMSF, 100ug/ml TPCK, and 50ug/ml leupeptin or Pierce Protease and Phosphatase Inhibitor Mini Tablets (cat# A32959) .
  • AAV Rep, AAV Cap, and host cell Cyclophilin B (a loading control) on PVDF membranes were probed with mouse anti-AAV2 Rep (clone 303.9, Progen Catlog# 61069 at 1:100x), mouse anti-AAV VP1/VP2/VP3 (clone B1, Progen 65158, supernatant at 1:25x), or mouse anti-Cyclophilin B (Abcam, ab236760 at 1:10,000x) followed by HRP conjugated secondary goat-anti-mouse IgG H&L antibody (AbCam, ab6789 at 2000x to 10,000x).

Landscapes

  • Micro-Organisms Or Cultivation Processes Thereof (AREA)

Abstract

A method of making a viral particle packaging and producer ceil line is provided. The cell line is based on an adeno-associated virus (AAV) vector, the cell line is caused to expresses, in association with the viral particle, a desired transgene.

Description

ADENO-ASSOCIATED VIRUS (AAV) PRODUCTION FIELD The present invention relates, in part, to a method of making a viral particle packaging and producer stable cell line using transposon(s) encoding genes required for AAV production and an enzyme capable of performing targeted genomic integration. CROSS-REFERENCE TO RELATED APPLICATIONS This application claims the benefit of priority to U.S. Provisional Patent Application No.63/432,220, filed November 7, 2022, which is incorporated by reference herein in its entirety. SEQUENCE LISTING The instant application contains a Sequence Listing that has been submitted electronically in XML file format and is hereby incorporated by reference in its entirety. The Sequence Listing for this application is labeled “SAL- 023PC_126933-5023_SequenceListing.XML”, which was created on November 6, 2023, and is 1,045,814 bytes in size. BACKGROUND Adeno-associated virus (AAV), first discovered in 1965 as a small, replication-defective viruses that infect humans and some other primate species, has since become a promising tool in gene therapy for treatment of various diseases and/or disorders. AAV is a very small (20–26 nm), icosahedral, and nonenveloped virus AAV particle containing a single-stranded DNA genome consisting of approximately 4.7 kb. The genome includes three open reading frames (ORFs) encoding for replication (non-structural) proteins (rep), capsid (structural) proteins (cap), and the assembly activating protein (AAP). These three genes give rise to at least nine gene products through the use of three promoters, alternative translation start sites, and differential splicing. See Naso et al., BioDrugs.2017;31(4):317-334. The coding sequences for the rep, cap, and AAP genes are flanked by inverted terminal repeats (ITRs). The 145-nt ITRs are partially paired, and they fold upon themselves to maximize base pairing and form a T-shaped hairpin structure. The rep gene encodes four Rep proteins (Rep78, Rep68, Rep52, and Rep40), which are required for viral genome replication and packaging. The cap gene expression results in viral capsid (Cap) proteins (VP; VP1/VP2/VP3), which form the outer capsid shell that protects the viral genome, as well as being actively involved in cell binding and internalization. Naso et al. (2017). The AAV genome contains two viral promoters known as p5 and p19, which regulate the transcription of the four Rep proteins with molecular masses of 78, 68, 52, and 40 kDa (Rep68 and Rep40 being the splice variants of Rep78 and Rep52, respectively). Expression of the cap gene is driven by the P40 promoter and regulated by alternative splicing and different translation initiation sites, resulting in three cap proteins (VP1, VP2, and VP3) that form an icosahedral capsid of 3.9 kDa. The molecular ratio of these proteins (VP1:VP2:VP3) is approximately 1:1:10. The AAV genome also encodes for the AAP in an alternative ORF of the cap gene that plays a major role for capsid assembly. See Penaud-Budloo et al., Mol Ther Methods Clin Dev 2018;8:166-80. AAV includes 12 different AAV serotypes. AAV belongs to the genus Dependoparvovirus (the Parvoviridae family) because it needs the presence of a helper virus for replication and assembly, e.g., adeno-, herpes-, human papilloma- or vaccinia viruses. After attachment of AAV to a cell surface receptor of the host cell, the virus gets internalized by endocytosis. Within the nucleus, the viral capsid sheds to release the single-stranded AAV genome which is then converted to double-stranded DNA. The free end of the ITR hairpin hereby acts as a primer for the DNA synthesis. Co- infection with one of the AAV helper viruses leads to the initiation of AAV gene expression, replication, and the production of AAV virions or viral particles. AAV can infect dividing or non-dividing cells. AAV derived vectors have several advantages for viral-based gene therapy. AAV is believed to be non-pathogenic for humans, which makes it a suitable vehicle for delivery of genetic material to human cells. AAV also has low cytotoxicity and elicits a very mild immune response. Therefore, AAV vectors are well suitable for in vivo gene delivery. Furthermore, different AAV serotypes can target various tissues. Due to their properties, AAV derived vectors are becoming a more preferable tool as compared to adenovirus- and retrovirus-derived vectors. Accordingly, there is an increasing demand to manufacture desired AAV serotypes in large quantities for pre-clinical and clinical trials, and various other purposes. SUMMARY In aspects, the present application provides methods of making viral particle packaging and producer cell lines. In embodiments, the methods comprise transfecting a cell with two or more nucleic acids. In embodiments, the nucleic acids encode one or more of: (a) an enzyme capable of performing targeted genomic integration, (b) an inducible viral replication (Rep) gene, (c) an inducible viral Capsid (cap) gene, (d) one or more adenoviral auxiliary genes (e.g., selected from one or more of E1A, E1B, E4, E2A, and VA of an AAV), (e) an insulator (e.g., selected from HS4, D4Z4), (f) one or more terminal ends recognized by the enzyme, and (g) a transgene flanked by AAV inverted terminal repeats (ITRs). In embodiments, the inducible viral replication (Rep) and Capsid (cap) genes are controlled by at least one of a tetO-containing P5 promoter, P19 promoter, SV40 promoter, P40 promoter, or a functional variant or fragment thereof. In embodiments, the resulting transfected cell expresses the transgene in association with the viral particle. In embodiments, the cell is transfected with two nucleic acids, and the first nucleic acid encodes (a) an enzyme capable of performing targeted genomic integration, and the second nucleic acid encodes an inducible viral Rep gene, (c) an inducible viral cap gene, (d) one or more adenoviral auxiliary genes, (e) an insulator, (f) one or more terminal ends recognized by the enzyme, and (g) a transgene flanked by AAV ITRs. In embodiments, the cell is transfected with three nucleic acids, and the first nucleic acid encodes (a) an enzyme capable of performing targeted genomic integration; the second nucleic acid encodes (b) an inducible viral Rep gene, (c) an inducible viral cap gene, (d) one or more adenoviral auxiliary genes, (e) an insulator, and (f) one or more terminal ends recognized by the enzyme; and the third nucleic acid encodes (g) a transgene flanked by AAV ITRs. In embodiments, the cell is transfected with four nucleic acids, and the first nucleic acid encodes (a) an enzyme capable of performing targeted genomic integration; the second nucleic acid encodes (b) an inducible viral Rep gene, (d) one or more adenoviral auxiliary genes, (e) an insulator, and (f) one or more terminal ends recognized by the enzyme; the third nucleic acid encodes (c) an inducible viral cap gene; and the fourth nucleic acid encodes (g) a transgene flanked by AAV ITRs. In aspects, the present application provides methods of transfecting an E1A, E1B+ cell with two or more nucleic acids. In embodiments, the nucleic acids encode one or more of: (a) an enzyme capable of performing targeted genomic integration, (b) an inducible viral replication (Rep) gene, (c) an inducible viral Capsid (cap) gene, (d) an insulator (e.g., selected from HS4, D4Z4), (e) one or more terminal ends recognized by the enzyme, and (f) a transgene flanked by AAV inverted terminal repeats (ITRs). In embodiments, the inducible viral replication (Rep) and Capsid (cap) genes are controlled by at least one of a tetO-containing P5 promoter, P19 promoter, SV40 promoter, P40 promoter, or a functional variant or fragment thereof. In embodiments, the resulting transfected cell expresses the transgene in association with the viral particle. In embodiments, the E1A, E1B+ cell is transfected with two nucleic acids, and the first nucleic acid encodes (a) an enzyme capable of performing targeted genomic integration, and the second nucleic acid encodes an inducible viral Rep gene, (c) an inducible viral cap gene, (d) an insulator, (e) one or more terminal ends recognized by the enzyme, and (f) a transgene flanked by AAV ITRs. In embodiments, the cell is transfected with three nucleic acids, and the first nucleic acid encodes (a) an enzyme capable of performing targeted genomic integration; the second nucleic acid encodes (b) an inducible viral Rep gene, (c) an inducible viral cap gene, (d) an insulator, and (e) one or more terminal ends recognized by the enzyme; and the third nucleic acid encodes (f) a transgene flanked by AAV ITRs. In embodiments, the cell is transfected with four nucleic acids, and the first nucleic acid encodes (a) an enzyme capable of performing targeted genomic integration; the second nucleic acid encodes (b) an inducible viral Rep gene, (d) an insulator, and (e) one or more terminal ends recognized by the enzyme; the third nucleic acid encodes (c) an inducible viral cap gene; and the fourth nucleic acid encodes (f) a transgene flanked by AAV ITRs. In aspects, methods for an adeno-associated virus (AAV) packaging and producer cell line are provided. In embodiments, the methods make use of a technique to include, among other elements, the AAV viral replication (Rep) and Capsid (Cap) genes under a control of an inducible promoter, in an E1A, E1B+ cell. In some embodiments, the inducible Rep and Cap genes (under a control of an inducible promoter), AAV vector DNA sequences, and essential helper genes are included in a single genetic construct such as, e.g., a donor plasmid flanked by end sequences recognized by an enzyme capable of performing targeted genomic integration (e.g., without limitation, a mobile element enzyme in accordance with embodiments of the present disclosure). A transgene of interest can be encoded by a separate nucleic acid, or it can be part of the genetic construct encoding the inducible Rep and Cap genes, AAV vector DNA sequences, and essential helper genes. In aspects, methods for making a packaging and producer cell line with a dual donor/helper system are provided. In the dual donor/helper system, inducible Rep and Cap genes (sometimes collectively referred to as “Rep/Cap genes”), and a transgene of interest are encoded by respective separate nucleic acids. In embodiments, the dual donor/helper system includes, without limitation, a nucleic acid encoding inducible Rep and Cap genes, a nucleic acid (e.g., a donor expression vector, such as a helper AAV donor plasmid) encoding a transgene of interest, and a nucleic acid encoding an enzyme capable of performing targeted genomic integration (e.g., without limitation, a mobile element enzyme). In aspects, the present application provides a method of making a viral particle packaging and producer cell line, the method comprising transfecting an E1A, E1B+ cell with a first nucleic acid encoding an enzyme capable of performing targeted genomic integration, a second nucleic acid encoding a donor DNA comprising inducible viral replication (Rep) and Capsid (cap) genes, adenoviral auxiliary genes selected from E1A and E1B, and insulators (optionally selected from HS4, D4Z4), and comprising terminal ends recognized by the enzyme, and a third nucleic acid encoding a donor DNA comprising a transgene flanked by AAV inverted terminal repeats (ITRs), to thereby result in a transfected cell that expresses the transgene in association with the viral particle. Accordingly, in aspects, a method of making a viral particle packaging and producer cell line is provided. In embodiments, the method comprises transfecting an E1A, E1B+ cell with: a first nucleic acid encoding an enzyme capable of performing targeted genomic integration; a second nucleic acid encoding a donor DNA comprising inducible viral replication (Rep) and Capsid (Cap) genes, adenoviral auxiliary genes selected from E4ORF6, E2A and VA RNA, and insulators (e.g., without limitation, HS4 and/or D4Z4), and comprising terminal ends recognized by the enzyme; wherein the inducible expression of AAV viral replication (Rep) and Capsid (cap) genes, as well as the adenoviral auxiliary genes E4ORF6 and E2A, are controlled by at least one of a tetO-containing P5 promoter, P19 promoter, SV40 promoter, P40 promoter, or a functional variant or fragment thereof, and a third nucleic acid encoding a donor DNA comprising a transgene flanked by AAV inverted terminal repeats (ITRs), to thereby result in a transfected cell that expresses the transgene in association with the viral particle. In aspects, methods for making a packaging and producer cell line with a single donor/helper system are provided. In the single donor/helper system, inducible Rep/Cap genes and a nucleic acid (e.g., a donor expression vector, such as a helper AAV donor plasmid) encoding a transgene of interest are encoded by a same construct. In embodiments, the single donor/helper system includes, without limitation, a nucleic acid (e.g., a donor expression vector) encoding inducible Rep and Cap genes and a transgene of interest, and a nucleic acid encoding an enzyme capable of performing targeted genomic integration (e.g., without limitation, a mobile element enzyme). Accordingly, in aspects, a method of making a viral particle packaging and producer cell line is provided. In embodiments, the method comprises transfecting an E1A, E1B+ cell with a first nucleic acid encoding an enzyme capable of performing targeted genomic integration; and a second nucleic acid encoding viral packaging genes, viral helper genes, insulators (e.g., without limitation, HS4 and D4Z4), and a transgene, and comprising terminal ends recognized by the enzyme, to thereby result in a transfected cell that expresses the transgene in association with a viral particle. In embodiments, the E1A, E1B+ cell is, without limitation, a HEK293, E1A, E1B+ engineered CHO-K1, or Sf9 cell line. In embodiments, the E1A, E1B+ cell is, without limitation, a Chinese hamster ovary (CHO), baby hamster kidney (BHK), human embryonic kidney (HEK293T) cells, Vero cell, or Spodoptera frugiperda 9 (Sf9) cell, In embodiments, the viral helper genes comprise adenoviral auxiliary genes. In embodiments, the adenoviral auxiliary genes are selected from one or more of E1A, E1B, E4ORF6, E2A, and VA of an AAV, optionally E1A and E1B. In embodiments, the viral packaging genes comprise rep and cap genes of viral replication (Rep) and Capsid (Cap) proteins of an AAV. In embodiments, the inducible viral replication (Rep) and Capsid (cap) genes are controlled by at least one of a tetO- containing P5 promoter, P19 promoter, SV40 promoter, P40 promoter, or a functional variant or fragment thereof. In embodiments, the tetO-containing AAV2 P40 promoter comprises a nucleotide sequence of SEQ ID NO: 816, or a functional variant or fragment thereof, or sequence having at least about 70%, or at least about 80%, or at least about 85%, or at least about 90%, or at least about 95%, or at least about 97% identity thereto, or at least about 98% identity thereto, or at least about 99% identity thereto or at least about 10 mutations, or at least about 9 mutations, or at least about 8 mutations, or at least about 7 mutations, or at least about 6 mutations, or at least about 5 mutations, or at least about 4 mutations, or at least about 3 mutations, or at least about 2 mutations, or at least about 1 mutation. In embodiments, the P40 intron is an AAV2 P40 intron. In embodiments, the AAV2 P40 intron comprises a C276A substitution, or a substitution at position corresponding thereto relative to SEQ ID NO: 818. In embodiments, the AAV2 P40 intron comprises one or more mutated translation start sites (ATGs), optionally wherein the translation start sites are mutated to one of CTG, ACG, and TTG. In embodiments, the AAV2 P40 intron comprises substitutions at one or more positions A13, A32, T42, A61, A71, A89, A203, A246, A258, and T282, or one or more positions corresponding thereto, relative to SEQ ID NO: 818. In embodiments, the AAV2 P40 intron comprises substitutions at one or more positions A13C, A32C, T42C, A61C, A71T, A89C, A203T, A246C, A258C, and T282C corresponding to SEQ ID NO: 818. In embodiments, the AAV2 P40 intron comprises the nucleotide sequence of SEQ ID NO: 818, or a functional variant or fragment thereof, or sequence having at least about 70%, or at least about 80%, or at least about 85%, or at least about 90%, or at least about 95%, or at least about 97% identity thereto, or at least about 98% identity thereto, or at least about 99% identity thereto, or at least about 10 mutations, or at least about 9 mutations, or at least about 8 mutations, or at least about 7 mutations, or at least about 6 mutations, or at least about 5 mutations, or at least about 4 mutations, or at least about 3 mutations, or at least about 2 mutations, or at least about 1 mutation. In embodiments, the AAV2 P40 intron comprises the nucleotide sequence of SEQ ID NO: 819, or a functional variant or fragment thereof, or sequence having at least about 70%, or at least about 80%, or at least about 85%, or at least about 90%, or at least about 95%, or at least about 97% identity thereto, or at least about 98% identity thereto, or at least about 99% identity thereto, or at least about 10 mutations, or at least about 9 mutations, or at least about 8 mutations, or at least about 7 mutations, or at least about 6 mutations, or at least about 5 mutations, or at least about 4 mutations, or at least about 3 mutations, or at least about 2 mutations, or at least about 1 mutation. In embodiments, the AAV2 P40 intron comprises the nucleotide sequence of SEQ ID NO: 817, or a functional variant or fragment thereof, or sequence having at least about 70%, or at least about 80%, or at least about 85%, or at least about 90%, or at least about 95%, or at least about 97% identity thereto, or at least about 98% identity thereto, or at least about 99% identity thereto, or at least about 10 mutations, or at least about 9 mutations, or at least about 8 mutations, or at least about 7 mutations, or at least about 6 mutations, or at least about 5 mutations, or at least about 4 mutations, or at least about 3 mutations, or at least about 2 mutations, or at least about 1 mutation. In embodiments, the modified TATA box element has the nucleotide sequence of TATATAA. In embodiments, the tetO- containing AAV2 P40 promoter or a functional variant or fragment thereof, comprising a modified TATA box element and a modified AAV2 P40 intron comprises the nucleotide sequence of SEQ ID NO: 823, or a functional variant or fragment thereof, or sequence having at least about 70%, or at least about 80%, or at least about 85%, or at least about 90%, or at least about 95%, or at least about 97% identity thereto, or at least about 98% identity thereto, or at least about 99% identity thereto, or at least about 10 mutations, or at least about 9 mutations, or at least about 8 mutations, or at least about 7 mutations, or at least about 6 mutations, or at least about 5 mutations, or at least about 4 mutations, or at least about 3 mutations, or at least about 2 mutations, or at least about 1 mutation. In embodiments, the tetO-containing P40 promoter is a tetO-containing AAV2 P40 promoter. In embodiments, the AAV2 P40 promoter comprises the nucleotide sequence of SEQ ID NO: 820, or a functional variant or fragment thereof, or sequence having at least about 70%, or at least about 80%, or at least about 85%, or at least about 90%, or at least about 95%, or at least about 97% identity thereto, or at least about 98% identity thereto, or at least about 99% identity thereto, or at least about 10 mutations, or at least about 9 mutations, or at least about 8 mutations, or at least about 7 mutations, or at least about 6 mutations, or at least about 5 mutations, or at least about 4 mutations, or at least about 3 mutations, or at least about 2 mutations, or at least about 1 mutation. In embodiments, the modified P40 intron is a AAV9 P40 intron. In embodiments, the modified AAV9 P40 intron comprises the nucleotide sequence of SEQ ID NO: 821, or a functional variant or fragment thereof, or sequence having at least about 70%, or at least about 80%, or at least about 85%, or at least about 90%, or at least about 95%, or at least about 97% identity thereto, or at least about 98% identity thereto, or at least about 99% identity thereto, or at least about 10 mutations, or at least about 9 mutations, or at least about 8 mutations, or at least about 7 mutations, or at least about 6 mutations, or at least about 5 mutations, or at least about 4 mutations, or at least about 3 mutations, or at least about 2 mutations, or at least about 1 mutation. In embodiments, the modified TATA box element has the nucleotide sequence of TATATAA. In embodiments, the tetO-containing AAV2 P40 promoter or a functional variant or fragment thereof, comprising a modified TATA box element and a modified AAV9 P40 intron comprises the nucleotide sequence of SEQ ID NO: 822, or a functional variant or fragment thereof, or sequence having at least about 70%, or at least about 80%, or at least about 85%, or at least about 90%, or at least about 95%, or at least about 97% identity thereto, or at least about 98% identity thereto, or at least about 99% identity thereto, or at least about 10 mutations, or at least about 9 mutations, or at least about 8 mutations, or at least about 7 mutations, or at least about 6 mutations, or at least about 5 mutations, or at least about 4 mutations, or at least about 3 mutations, or at least about 2 mutations, or at least about 1 mutation. In embodiments, the tetO-containing AAV2 P40 promoter or a functional variant or fragment thereof, comprising a modified TATA box element and a modified AAV9 P40 intron comprises a nucleotide sequence of SEQ ID NO: 824, or a functional variant or fragment thereof, or sequence having at least about 70%, or at least about 80%, or at least about 85%, or at least about 90%, or at least about 95%, or at least about 97% identity thereto, or at least about 98% identity thereto, or at least about 99% identity thereto, or at least about 10 mutations, or at least about 9 mutations, or at least about 8 mutations, or at least about 7 mutations, or at least about 6 mutations, or at least about 5 mutations, or at least about 4 mutations, or at least about 3 mutations, or at least about 2 mutations, or at least about 1 mutation. In embodiments, the P40 promoter further comprises one or more enhancer elements. In embodiments, the enhancer element comprises one or more cis-acting elements, optionally selected from an Sp1 binding site, GC rich sequence, GCGGAAC motif, TAATGARAT element, AP1 binding site, and CCAAT box element. In embodiments, the enhancer element comprises about 1 to about 5 Sp1 binding sites, optionally about 1, or about 2, or about 3, or about 4, or about 5 Sp1 binding sites. In embodiments, the enhancer element comprises about 1 or about 2 GC rich sequences. In embodiments, the enhancer element is derived from an hCMV Enhancer Element-3. In embodiments, the hCMV Enhancer Element-3 comprises the nucleotide sequence of SEQ ID NO: 830 or a functional variant or fragment thereof, or sequence having at least about 70%, or at least about 80%, or at least about 85%, or at least about 90%, or at least about 95%, or at least about 97% identity thereto, or at least about 98% identity thereto, or at least about 99% identity thereto, or at least about 10 mutations, or at least about 9 mutations, or at least about 8 mutations, or at least about 7 mutations, or at least about 6 mutations, or at least about 5 mutations, or at least about 4 mutations, or at least about 3 mutations, or at least about 2 mutations, or at least about 1 mutation. In embodiments, the expression of the Rep and Cap proteins is controlled by an inducible promoter, optionally an antibiotic-dependent promoter. In embodiments, the antibiotic-dependent promoter is tetracycline- or a variant thereof dependent promoter and a tetracycline-repressor-based (t-REx) system is used. In embodiments, the antibiotic- dependent promoter is tetracycline- or a variant thereof dependent promoter and a tetracycline-controlled transactivator (rtTA) system is used. In embodiments, the antibiotic-dependent promoter is a coumermycin/novobiocin promoter, or a variant thereof. In embodiments, the viral particle is an AAV, and wherein the AAV is selected from AAV1, AAV2, AAV3, AAV4, AAV5, AAV6, AAV7, AAV8, and AAV9. In embodiments, any one or more of the first, second, and third nucleic acids is encoded by a single nucleic acid. In embodiments, the second nucleic acid and/or the third nucleic acid are included in a single expression vector. In embodiments, the first and second nucleic acids are included in a single expression vector, and the third nucleic acid is included in an expression vector that is different from the expression vector including the first and second nucleic acids. In embodiments, the single expression vector comprises a plasmid. In embodiments, the plasmid comprises the nucleotide sequence of SEQ ID NO: 825 or a functional variant or fragment thereof, or sequence having at least about 70%, or at least about 80%, or at least about 85%, or at least about 90%, or at least about 95%, or at least about 97% identity thereto, or at least about 98% identity thereto, or at least about 99% identity thereto, or at least about 10 mutations, or at least about 9 mutations, or at least about 8 mutations, or at least about 7 mutations, or at least about 6 mutations, or at least about 5 mutations, or at least about 4 mutations, or at least about 3 mutations, or at least about 2 mutations, or at least about 1 mutation. In embodiments, the plasmid comprises the nucleotide sequence of SEQ ID NO: 826 or a functional variant or fragment thereof, or sequence having at least about 70%, or at least about 80%, or at least about 85%, or at least about 90%, or at least about 95%, or at least about 97% identity thereto, or at least about 98% identity thereto, or at least about 99% identity thereto, or at least about 10 mutations, or at least about 9 mutations, or at least about 8 mutations, or at least about 7 mutations, or at least about 6 mutations, or at least about 5 mutations, or at least about 4 mutations, or at least about 3 mutations, or at least about 2 mutations, or at least about 1 mutation. In embodiments, the plasmid comprises the nucleotide sequence of SEQ ID NO: 841 or a functional variant or fragment thereof, or sequence having at least about 70%, or at least about 80%, or at least about 85%, or at least about 90%, or at least about 95%, or at least about 97% identity thereto, or at least about 98% identity thereto, or at least about 99% identity thereto, or at least about 10 mutations, or at least about 9 mutations, or at least about 8 mutations, or at least about 7 mutations, or at least about 6 mutations, or at least about 5 mutations, or at least about 4 mutations, or at least about 3 mutations, or at least about 2 mutations, or at least about 1 mutation. In embodiments, the plasmid comprises the nucleotide sequence of SEQ ID NO: 842, or a functional variant or fragment thereof, or sequence having at least about 70%, or at least about 80%, or at least about 85%, or at least about 90%, or at least about 95%, or at least about 97% identity thereto, or at least about 98% identity thereto, or at least about 99% identity thereto, or at least about 10 mutations, or at least about 9 mutations, or at least about 8 mutations, or at least about 7 mutations, or at least about 6 mutations, or at least about 5 mutations, or at least about 4 mutations, or at least about 3 mutations, or at least about 2 mutations, or at least about 1 mutation. In embodiments, the plasmid comprises the nucleotide sequence of SEQ ID NO: 843, or a functional variant or fragment thereof, or sequence having at least about 70%, or at least about 80%, or at least about 85%, or at least about 90%, or at least about 95%, or at least about 97% identity thereto, or at least about 98% identity thereto, or at least about 99% identity thereto, or at least about 10 mutations, or at least about 9 mutations, or at least about 8 mutations, or at least about 7 mutations, or at least about 6 mutations, or at least about 5 mutations, or at least about 4 mutations, or at least about 3 mutations, or at least about 2 mutations, or at least about 1 mutation. In embodiments, one or more of the first, second, and third nucleic acids is RNA, optionally mRNA, optionally synthetic mRNA. In embodiments, one or more of the first, second, and third nucleic acids is DNA, optionally plasmid DNA. In embodiments, one or more of the first, second, and third nucleic acids is an expression vector, wherein the expression vector is optionally a plasmid. In embodiments, the transfected cell generates using the method of making a viral particle packaging and producer cell line in accordance with embodiments of the present disclosure comprises a donor DNA comprising the transgene flanked by AAV inverted terminal repeats (ITRs). In embodiments, the terminal ends or ITRs comprise the nucleotide sequence of SEQ ID NO: 831 and/or SEQ ID NO: 832, or a functional variant or fragment thereof, or sequence having at least about 70%, or at least about 80%, or at least about 85%, or at least about 90%, or at least about 95%, or at least about 97% identity thereto, or at least about 98% identity thereto, or at least about 99% identity thereto, or at least about 10 mutations, or at least about 9 mutations, or at least about 8 mutations, or at least about 7 mutations, or at least about 6 mutations, or at least about 5 mutations, or at least about 4 mutations, or at least about 3 mutations, or at least about 2 mutations, or at least about 1 mutation. In embodiments, the transgene has a size of about 200,000 base pairs or less. The methods in accordance with the present disclosure can be used to produce viral particles carrying a transgene of a large size. For example, in embodiments, the transgene has a size of at least 200,000 base pairs. In embodiments, the transgene has a size of about 200,000 base pairs. In embodiments, the method further comprises culturing the transfected cell in a medium that expands a population of the transfected cells to create a stably transfected packaging and producer cell line. In embodiments, the stably transfected producer cell line is capable of producing replication-deficient viral particles in association with the transgene. In embodiments, the transfection comprises electroporation, nucleofection, lipofection, or calcium phosphate transfection. In embodiments, the method is helper virus-free. In embodiments, an enzyme capable of performing targeted genomic integration causes the transgene to be inserted in a certain genomic locus and/or site (e.g., at a TA dinucleotide site or a TTAA tetranucleotide site in a genomic safe harbor site (GSHS)) of a nucleic acid molecule. In embodiments, the enzyme capable of performing targeted genomic integration is a recombinase. In embodiments, the enzyme has one or more mutations which confer hyperactivity. In embodiments, the recombinase is an integrase. In embodiments, the recombinase is an integrase or a mobile element enzyme. In embodiments, the integrase is a mobile element enzyme. In embodiments, the mobile element enzyme is an engineered mammalian mobile element enzyme. In embodiments, the mobile element enzyme is a mammal-derived RNA mobile element enzyme (e.g., a helper RNA mobile element enzyme). In embodiments, the mobile element enzyme is a mammal-derived DNA mobile element enzyme. In embodiments, the mobile element enzyme is a chimeric mobile element enzyme. In embodiments, the enzyme has gene cleavage activity (Exc+) and/or gene integration activity (Int+). In embodiments, enzyme has gene cleavage activity (Exc+) and/or a lack of gene integration activity (Int-). In embodiments, the mobile element enzyme comprises: (a) a gene-editing system, and (b) a mobile element enzyme that is capable of inserting the donor DNA comprising a transgene at a TA dinucleotide site or a TTAA tetranucleotide site in a genomic safe harbor site (GSHS). In embodiments, the transgene encodes a complete polypeptide. In embodiments, the transgene is defective or substantially absent in a disease state. In embodiments, the gene-editing system comprises a Cas9 enzyme guide RNA complex. In embodiments, the Cas9 enzyme guide RNA complex comprises a nuclease-deficient (or inactive, or dead) dCas9 guide RNA complex, also referred to as dCas9 guide RNA complex. In embodiments, the nuclease-deficient dCas9 guide RNA complex comprises a guide RNA selected from: GTTTAGCTCACCCGTGAGCC (SEQ ID NO: 91), CCCAATATTATTGTTCTCTG (SEQ ID NO: 92), GGGGTGGGATAGGGGATACG (SEQ ID NO: 93), GGATCCCCCTCTACATTTAA (SEQ ID NO: 94), GTGATCTTGTACAAATCATT (SEQ ID NO: 95), CTACACAGAATCTGTTAGAA (SEQ ID NO: 96), TAAGCTAGAGAATAGATCTC (SEQ ID NO: 97), and TCAATACACTTAATGATTTA (SEQ ID NO: 98), or a variant thereof. In embodiments, the enzyme is derived from Bombyx mori, Xenopus tropicalis, Trichoplusia ni, Myotis lucifugus, Rhinolophus ferrumequinum, Rousettus aegyptiacus, Phyllostomus discolor, Myotis myotis, Pteropus vampyrus, Pipistrellus kuhlii, Molossus molossus, Pan troglodytes, or Homo sapiens. In embodiments, the enzyme is an engineered version, including but not limited to hyperactive forms, of an enzyme (e.g., without limitation, a mobile element enzyme) derived from Bombyx mori, Xenopus tropicalis, Trichoplusia ni, Myotis lucifugus, Rhinolophus ferrumequinum, Rousettus aegyptiacus, Phyllostomus discolor, Myotis myotis, Pteropus vampyrus, Pipistrellus kuhlii, Molossus molossus, Pan troglodytes, or Homo sapiens. In embodiments, the mobile element enzyme is from one or more of the Sleeping beauty, Tn1, Tn2, Tn3, Tn5, Tn7, Tn9, Tn10, Tn552, Tn903, Tn1000/Gamma-delta, Tn/O, tnsA, tnsB, tnsC, tniQ, IS10, ISS, IS911, Minos, piggyBac, Tol2, Mos1, Himar1, Hermes, Tol2, Minos, Tel, P-element, MuA, Ty1, Chapaev, transib, Tc1/mariner, or Tc3 donor DNA system, or biologically active fragments variants thereof, inclusive of hyperactive variants. In embodiments, the enzyme is a mobile element enzyme such as a Myotis lucifugus mobile element enzyme (MLT mobile element enzyme), also referred to herein as an MLT mobile element enzyme. In embodiments, the MLT mobile element enzyme is a wild-type MLT mobile element enzyme. In embodiments, the MLT mobile element enzyme is a modified MLT mobile element enzyme, also referred to herein as a corrected MLT mobile element enzyme. In embodiments, the MLT mobile element enzyme (e.g., the wild-type MLT mobile element enzyme, the corrected MLT mobile element enzyme, or a variant thereof) has one or more mutations such as hyperactive mutations. In embodiments, the MLT mobile element enzyme is a modified MLT mobile element enzyme. In embodiments, the MLT mobile element enzyme has an amino acid sequence of SEQ ID NO: 1, or a variant having at least about 90%, or at least about 93%, or at least about 95%, or at least about 97%, or at least about 98%, or at least about 99% identity thereto, and one or more mutations selected from L573X, E574X, and S2X, wherein X is any amino acid or no amino acid, optionally X is A, G, or a deletion. In embodiments, the mutations are L573del E574del, and S2A (SEQ ID NO: 1). In embodiments, the MLT mobile element enzyme has an amino acid sequence of SEQ ID NO: 1 and S8P and C13R mutations (SEQ ID NO: 11). In embodiments, the MLT mobile element enzyme has an amino acid sequence having mutations at positions which correspond to at least one of S8P and C13R mutations relative to the amino acid of SEQ ID NO: 1 or a functional equivalent thereof. In embodiments, the MLT mobile element enzyme has an amino acid sequence having mutations at positions which correspond to S8P and C13R mutations relative to the amino acid of SEQ ID NO: 1 or a functional equivalent thereof. In embodiments, the MLT mobile element enzyme has an amino acid sequence having mutations at positions which correspond to S8P, C13R, and N125K mutations relative to the amino acid of SEQ ID NO: 1 or a functional equivalent thereof. In embodiments, the MLT mobile element enzyme has a nucleotide sequence of SEQ ID NO: 2 (which is codon- optimized) and an amino acid sequence SEQ ID NO: 1. In embodiments, the MLT mobile element enzyme has a nucleotide sequence of SEQ ID NO: 2, or a nucleotide sequence having at least about 90%, or at least about 93%, or at least about 95%, or at least about 97%, or at least about 98%, or at least about 99% identity thereto, or a codon- optimized form thereof. In embodiments, the MLT mobile element enzyme has an amino acid sequence SEQ ID NO: 1, or an amino acid sequence having at least about 90%, or at least about 93%, or at least about 95%, or at least about 97%, or at least about 98%, or at least about 99% identity thereto. In embodiments, the MLT mobile element enzyme includes a hyperactive mutation selected from TABLE 14A and TABLE 14B, e.g., about 1, or about 2, or about 3, or about 4, or about 5 hyperactive mutations selected from TABLE 14A or TABLE 14B, or combinations thereof. In embodiments, the MLT mobile element enzyme is a wild-type mobile element enzyme that has an amino acid sequence of SEQ ID NO: 10. In embodiments, the MLT mobile element enzyme is a modified mobile element enzyme having the amino acid sequence of SEQ ID NO: 11. In embodiments, at least one of the first, second, and third nucleic acids is in the form of a lipid nanoparticle (LNP). In embodiments, a nucleic acid encoding an enzyme capable of performing targeted genomic integration (e.g., without limitation, a mobile element enzyme) and a nucleic acid encoding a donor DNA are in the form of the same LNP, optionally in a co-formulation. In embodiments, a nucleic acid encoding an enzyme capable of performing targeted genomic integration (e.g., without limitation, a mobile element enzyme) and a nucleic acid encoding a donor DNA are in the same mixture with an LNP. In embodiments, a method of producing an AAV bearing a gene of interest is provided, to produce the AAV bearing the gene of interest. In embodiments, the method of making a viral packaging or producer cell line of the present disclosure provides a stable cell with substantially reduced or ablated CAP expression. In embodiments, the method of making a viral packaging or producer cell line of the present disclosure provides a stable cell with REP expression or substantially enhanced REP expression. In embodiments, the method of making a viral packaging or producer cell line of the present disclosure provides a stable cell with substantially reduced or ablated CAP expression and REP expression or substantially enhanced REP expression. In embodiments, the method of making a viral packaging or producer cell line of the present disclosure provides a stable cell with REP expression or substantially enhanced VP1 expression. In embodiments, the method of making a viral packaging or producer cell line of the present disclosure provides a stable cell with substantially reduced or ablated CAP expression. In embodiments, a cell for gene therapy is provided that is generated by a method in accordance with embodiments of the present disclosure. In some embodiments, a pharmaceutical composition comprising the cell is provided. In embodiments, a method for treating a disease or condition using gene therapy is provided, the method comprising administering to a subject in need thereof a transfected cell generated using a method in accordance with embodiments of the present disclosure. In embodiments, the disease or condition comprises cancer. In embodiments, the disease or condition comprises an infectious disease. In embodiments, the infectious disease is a coronavirus infection, optionally selected from infection with SAR-CoV, MERS-CoV, and SARS-CoV-2, or variants thereof. The details of the invention are set forth in the accompanying description below. Although methods and materials similar or equivalent to those described herein can be used in the practice or testing of the present invention, illustrative methods and materials are now described. Other features, objects, and advantages of the invention will be apparent from the description and from the claims. In the specification and the appended claims, the singular forms also include the plural unless the context clearly dictates otherwise. Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs. BRIEF DESCRIPTION OF DRAWINGS FIG.1A is a non-limiting representation of an AAV production strategy in accordance with embodiments of the present disclosure, using a donor vector to create a HEK293 producer cell line or another cell line expressing the AdV E1a and E1b genes (e.g., engineered CHO-K1 or Vero cell line with E1a, E1b) that forms a replication deficient AAV particle containing a transgene of interest. The single or dual donor comprising a transgene of interest is incorporated (e.g., by transfection such as electroporation) into a HEK293 cell line or another cell line expressing the E1a and E1b genes. The HEK293 cells are expanded to create a producer cell line and culturing the producer cells creates replication deficient (rep-) AAV particles with the transgene of interest. FIG.1B is a representation of an example of an inducible Rep/Cap and helper AAV donor plasmid construct (Kanar), encoding E2A, E4ORF6, and VA RNA helper genes flanked by insulators and mobile element enzyme recognition ends, and used with a helper RNA or DNA to create an AAV producer cell line, in accordance with embodiments of the present disclosure. FIG.1C is a representation of an example of a nucleic acid (plasmid) encoding a transgene (gene of interest (GOI)) included between AAV inverted terminal repeats (ITRs), in accordance with embodiments of the present disclosure. FIGs.2A-E depict non-limiting representations of chimeric, monomer or head-to-tail dimer mobile element enzymes that are designed to target human GSHS using TALE and Cas9/guide RNA DNA binders. FIG.2A. TALEs include nuclear localization signals (NLS) and an activation domain (AD) to function as transcriptional activators. The DNA binding domain has approximately 16.5 repeats of 33-34 amino acids with a residual variable di-residue (RVD) at position 12-13. FIG.2B. RVDs are shown that have specificity for one or several nucleotides. Only bases of the DNA leading strand are shown. FIG.2C. A chimeric mobile element enzyme construct comprising a TALE DNA-binding protein fused thereto by a linker that is greater than 23 amino acids in length (top) and a chimeric mobile element enzyme construct comprising dCas9 linked to one or more guide RNAs (bottom). FIG. 2D is a non-limiting representation of a system in accordance with embodiments of the present disclosure comprising a nucleic acid (e.g., helper RNA) encoding an enzyme capable of performing targeted genomic integration and a nucleic acid encoding a mobile element enzyme (donor DNA). The helper RNA is translated into a bioengineered enzyme (e.g., integrase, recombinase, or mobile element enzyme) that recognizes specific ends and seamlessly inserts the donor DNA into the human genome in a site-specific manner without a footprint. Chimeric mobile element enzymes form dimers or tetramers at open chromatin to insert donor DNA at TTAA recognition sites near DNA binding regions targeted by dCas9/gRNA or TALEs. Binding of the TALE or Cas9/gRNA to GSHS physically sequesters the mobile element enzyme as a monomer or dimer to the same location and promotes transposition to the nearby TTAA sequences. All RVDs are preceded by a thymine (T) to bind to the NTR shown in FIG.2A). FIG.2E is a non-limiting representation of a system in accordance with embodiments of the present disclosure comprising a nucleic acid (e.g., helper RNA) encoding an enzyme capable of performing targeted genomic integration and a nucleic acid encoding a mobile element enzyme (donor DNA). The helper RNA is translated into a bioengineered enzyme (e.g., integrase, recombinase, or mobile element enzyme) that recognizes specific ends and seamlessly inserts the donor DNA into the human genome in a site-specific manner without a footprint. FIGs.3A-3E depict construct templates. FIG.3A depicts a plasmid construct template that transcribes helper RNA that is later processed with a 5’- m7G cap and pseudouridine substitution. FIG.3B depicts a donor DNA construct template with the transgene VLDLR. FIG.3C depicts a chimeric mobile element enzyme construct template with a TALE DNA binder. Other TALEs and mobile element enzymes can be substituted. FIG.3D depicts a chimeric mobile element enzyme construct template with a dCas9/gRNA DNA binder. FIG.3E depicts a system comprising donor DNA (panel A) and helper RNA (panel B): panel (A) The donor DNA can be any gene of interest (GOI) including a gene that replaces, inactivates, or provides suicide or helper functions. The GOI can be driven by a predetermined promoter and flanked by insulators to prevent gene silencing. The internal terminal repeats (ITRs) can be specific for a mammal- derived mobile element enzyme. Panel (B) The helper RNA is 5’-m7G capped (cap 0, or cap1, or cap 2) with flanking globin 5’- and 3’-UTRs, an N-terminus nuclear localization signal (SV40 or nucleoplasmin), a 34 polyalanine tail region, and pseudouridine modification. The mobile element enzyme (e.g., hyperactive mobile element enzyme) is bioengineered to insert the donor DNA in a site- and/or locus specific manner in a human genome. FIG.4 depicts a non-limiting representation of a conventional rAAV production system. Production in adenovirus complementation systems is usually performed as plasmid transfection processes, where AAV Rep/Cap genes, the ITR-flanked gene of interest (GOI), as well as Ad-helper genes are provided as three separate plasmids, respectively, to a E1a/E1b containing HEK293 cell line or other engineered cell line that contains E1a/E1b (e.g., engineered CHO- K1 or Vero cells). FIG.5 depicts a non-limiting schematic of an engineered mobile element enzyme ITR containing plasmid that encodes 1) AAV2_REP78/68 ORFs under the control of P5TO1 promote with AAV2_REP52/40 ORFs and CAPs (VP1/2/3) under their native P19 or P40 promoter, respectively, 2) Ad5 E2A_IRES_Ad5 E4 ORF6 under the control of the tetO- containing SV40 promoter, 3) VA RNAs under the control of HSV-2 ICP4 promoter, and 4) hygro-B resistant gene under the control of TK promoter. FIG.6 depicts a non-limiting schematic of an engineered mobile element enzyme ITR containing plasmid that encodes 1) AAV2_REP78/68 ORFs under the control of P5TO1 promoter and AAV2_REP52/40 ORFs under the native P19 promoter, 2) AAV2 CAPs under the tetO-containing P40 promoter with AAV9 P40 intron, 3) Ad5 E2A_IRES_Ad5 E4 ORF6 under the control of the tetO-containing SV40 promoter, 4) VA RNAs under the control of HSV-2 ICP4 promoter, and 5) hygro-B resistant gene under the control of TK promoter. FIG.7 depicts a non-limiting schematic of an engineered mobile element enzyme ITR containing plasmid that encodes 1) AAV2_REP 78/68 ORFs under the control of P5TO1 promoter and AAV2_REP52/40 ORFs under the native P19 promoter, 2) Ad5 E2A_IRES_Ad5 E4 ORF6 under the control of the tetO-containing SV40 promoter, 3) VA RNAs under the control of HSV-2 ICP4 promoter, and 4) hygro-B resistant gene under the control of TK promoter. Note that the nucleotide sequence between the end of native AAV Rep ORFs and native poly A was deleted in the construct to create an AAV Cap VP1/2/3 deletion phenotype. FIG.8 depicts a non-limiting schematic of a construct encoding AAV2 CAP under the control of the tetO-containing hCMV-derived enhancer element 3/AAV2 P40 promoter with a modified AAV2 P40 TATA element and modified AAV2 P40 intron_1 (a at position 276 substituted for C) and WPRE/modified ICP27 poly A, followed by ICP0 promoter-Zeocin resistant gene-ICP0 poly A cassette flanked with ITRs for Sleeping Beauty (SB) transposase. FIG.9 depicts a non-limiting schematic of a construct encoding AAV2 CAP under the control of the tetO-containing hCMV enhancer element 3/AAV2 P40 promoter with a modified AAV2 P40 TATA element and MODIFIED AAV2 P40 intron_2 and WPRE/modified ICP27 poly A, followed by ICP0 promoter-Zeocin resistant gene-ICP0 poly A cassette flanked with SB ITRs. FIG.10A-10B depicts GFP images 120 hours post-transduction of AAVH clone 17+ (FIG.10A) and ddPCR analyses for quantification of the rAAV2-EGFP genome copy number per nanogram (ng) of total genomic DNA (FIG.10B). FIG.11A – 11B depicts GFP images 120 hours post-transduction of AAVH clone 13 + (FIG.10A) and ddPCR analyses for quantification of the rAAV2-EGFP genome copy number per nanogram (ng) of total genomic DNA (FIG.10B). FIG.12A – 12B depicts expression of functional AAV2 Rep proteins in AAV2-HB (Plasmid shown in FIG.6) clones as determined by western blot and ddPCR. FIG.12A depicts a western blot analysis illustrating cells from selected AAV2- H B cell clones HB-36, HB-42, HB-118, and HB-120 were treated with10ng/ml doxycycline for 72 hours followed by extraction with RIPA buffer in the presence of proteinase inhibitor. FIG.12B shows the results of a ddPCR analysis for AAV Genome Amplification / Rep function Assay. FIG.13A – 13B depicts expression of functional AAV2 Rep proteins in AAV-CDH (Plasmid shown in FIG.7) clones as determined by western blot (FIG.13A) and ddPCR (FIG.13B). FIG.14A – 14B depicts establishment of AAV2 full producer stable cell pools, CDH-24/Cap-SB, CDH-48/Cap-SB, and CDH-129/Cap-SB, with sleeping beauty transposase-mediated integration of AAV2 CAPs-expressing cassettes (Plasmid shown in FIG.7) into AAV-CDH Clone 24, 48, 129 serotype adaptable (Cap minus or Cap-) stable cell lines. FIG.14A shows ddPCR analyses of SB-mediated AAV2 Cap transposon integration in each Zeocin-selected stable pools. FIG.14B shows western blot analysis of SB-mediated AAV2 Cap transposon integration in each Zeocin-selected stable pools. FIG.15A – 15B depicts expression of functional AAV2 Rep proteins in CDH-24/Cap-SB, CDH-48/Cap-SB, and CDH- 129/Cap-SB pools as determined by western blot and ddPCR. FIG.15A shows ddPCR results of rAAV Rep proteins being expressed and FIG.15B shows expressed AAV2 Rep proteins are functionally active in supporting AAV genome amplification. FIG.16 depicts expression of AAV2 Cap and Rep in CDH-48/Cap-SB stable pool increased with doxycycline dosages and over time. Cells from CDH-48/Cap-SB stable pools were incubated either in the presence of 0, 5, 10, 25, 50, or 100ng/ml doxycycline and harvest at 72 hours post induction; or incubated with 50ng/ml doxycycline and harvested at 24, 48, 72, or 96 hours post induction (FIG.16). Cell extraction with RIPA buffer containing proteinase inhibitors, western blot, and ddPCR analyses of the CDH-48/Cap-SB (AAV2-capsid minus stable lines Clone CDH-48) samples were caried out as described previously. The results show that expression of AAV2 Cap and Rep proteins increased over time in the presence of 50ng/ml doxycycline. FIG.17A – 17B depicts production of competent rAAV2 particles from stably engineered CDH-48/Cap-SB cell pools in response to doxycycline. FIG.17A show a graph depicting rAAV2 physical titers from CDH-48/CAP-SB stable pool harvests. FIG.17B shows a graph depicting transduction of CDH-48 cells (AAV2-CAPSID MINUS STABLE LINES CLONE CDH-48) with unconcentrated rAAV supernatents. FIG.18A – 18B depicts characterization of Zeocin-resistant single clones selected from the CDH-48/Cap-SB stable pool. FIG.18A depicts ddPCR analysis for SB-mediated AAV2-Cap cassette integration. FIG.18B depicts western blot analysis with antibodies specific for AAV2 CAP. FIG.19A – 19B depicts characterization of Zeocin-resistant single clones selected from the CDH-48/Cap-SB stable pool. FIG.19A depicts western blot analysis with antibodies specific for AAV2 REP. FIG.19B shows ddPCR assays of AAV genome amplification following transduction with low VGC/cell of AAV2-EGFP viral particles. DETAILED DESCRIPTION The present invention is based, in part, on the discovery that a mobile element enzyme-mediated site-specific genomic integration can be used to produce a stable AAV-based cell line. The cell line is produced by insertion of inducible viral replication (rep) and Capsid (cap) genes and helper genes (E4ORF6, E2A, and VA) in specific genomic locations of a cell such as, e.g., a HEK293 (E1+) cell or a genetically engineered CHO-K1 or Vero cell line. An advantage of the approach in accordance with embodiments of the present disclosure is that it does not require DNA homology for recombination, and it is independent of the size of the donor or recipient DNA molecules. Also, the described method is performed in a brief enzyme-catalyzed reaction using an enzyme (e.g., without limitation, a mobile element enzyme) delivered as either DNA or RNA. In embodiments, this protocol allows for the production of stable expression human cell pools in about 4 weeks or in less than 4 weeks. In aspects, the present application provides methods of making viral particle packaging and producer cell lines. In embodiments, the methods comprise transfecting a cell with two or more nucleic acids. In embodiments, the nucleic acids encode one or more of: (a) an enzyme capable of performing targeted genomic integration, (b) an inducible viral replication (Rep) gene, (c) an inducible viral Capsid (cap) gene, (d) one or more adenoviral auxiliary genes (e.g., selected from one or more of E1A, E1B, E4, E2A, and VA of an AdV), (e) an insulator (e.g., selected from HS4, D4Z4), (f) one or more terminal ends recognized by the enzyme, and (g) a transgene flanked by AAV inverted terminal repeats (ITRs). In embodiments, the inducible viral replication (Rep) and Capsid (cap) genes are controlled by at least one of a tetO-containing P5 promoter, P19 promoter, SV40 promoter, P40 promoter, or a functional variant or fragment thereof. In embodiments, the resulting transfected cell expresses the transgene in association with the viral particle. In embodiments, the cell is transfected with two nucleic acids, and the first nucleic acid encodes (a) an enzyme capable of performing targeted genomic integration, and the second nucleic acid encodes an inducible viral Rep gene, (c) an inducible viral cap gene, (d) one or more adenoviral auxiliary genes, (e) an insulator, (f) one or more terminal ends recognized by the enzyme, and (g) a transgene flanked by AAV ITRs. In embodiments, the cell is transfected with three nucleic acids, and the first nucleic acid encodes (a) an enzyme capable of performing targeted genomic integration; the second nucleic acid encodes (b) an inducible viral Rep gene, (c) an inducible viral cap gene, (d) one or more adenoviral auxiliary genes, (e) an insulator, and (f) one or more terminal ends recognized by the enzyme; and the third nucleic acid encodes (g) a transgene flanked by AAV ITRs. In embodiments, the cell is transfected with four nucleic acids, and the first nucleic acid encodes (a) an enzyme capable of performing targeted genomic integration; the second nucleic acid encodes (b) an inducible viral Rep gene, (d) one or more adenoviral auxiliary genes, (e) an insulator, and (f) one or more terminal ends recognized by the enzyme; the third nucleic acid encodes (c) an inducible viral cap gene; and the fourth nucleic acid encodes (g) a transgene flanked by AAV ITRs. In aspects, the present application provides methods of transfecting an E1A, E1B+ cell with two or more nucleic acids. In embodiments, the nucleic acids encode one or more of: (a) an enzyme capable of performing targeted genomic integration, (b) an inducible viral replication (Rep) gene, (c) an inducible viral Capsid (cap) gene, (d) an insulator (e.g., selected from HS4, D4Z4), (e) one or more terminal ends recognized by the enzyme, and (f) a transgene flanked by AAV inverted terminal repeats (ITRs). In embodiments, the inducible viral replication (Rep) and Capsid (cap) genes are controlled by at least one of a tetO-containing P5 promoter, P19 promoter, SV40 promoter, P40 promoter, or a functional variant or fragment thereof. In embodiments, the resulting transfected cell expresses the transgene in association with the viral particle. In embodiments, the E1A, E1B+ cell is transfected with two nucleic acids, and the first nucleic acid encodes (a) an enzyme capable of performing targeted genomic integration, and the second nucleic acid encodes an inducible viral Rep gene, (c) an inducible viral cap gene, (d) an insulator, (e) one or more terminal ends recognized by the enzyme, and (f) a transgene flanked by AAV ITRs. In embodiments, the cell is transfected with three nucleic acids, and the first nucleic acid encodes (a) an enzyme capable of performing targeted genomic integration; the second nucleic acid encodes (b) an inducible viral Rep gene, (c) an inducible viral cap gene, (d) an insulator, and (e) one or more terminal ends recognized by the enzyme; and the third nucleic acid encodes (f) a transgene flanked by AAV ITRs. In embodiments, the cell is transfected with four nucleic acids, and the first nucleic acid encodes (a) an enzyme capable of performing targeted genomic integration; the second nucleic acid encodes (b) an inducible viral Rep gene, (d) an insulator, and (e) one or more terminal ends recognized by the enzyme; the third nucleic acid encodes (c) an inducible viral cap gene; and the fourth nucleic acid encodes (f) a transgene flanked by AAV ITRs. In aspects, the present application provides a method of making a viral particle packaging and producer cell line, the method comprising transfecting an E1A, E1B+ cell with a first nucleic acid encoding an enzyme capable of performing targeted genomic integration, a second nucleic acid encoding a donor DNA comprising inducible viral replication (Rep) and Capsid (cap) genes, adenoviral auxiliary genes selected from E1A and E1B, and insulators (optionally selected from HS4, D4Z4), and comprising terminal ends recognized by the enzyme, and a third nucleic acid encoding a donor DNA comprising a transgene flanked by AAV inverted terminal repeats (ITRs), to thereby result in a transfected cell that expresses the transgene in association with the viral particle. In aspects, a method of making a viral particle packaging and producer cell line is provided. In embodiments, the method comprises: transfecting an E1A, E1B+ cell with a first nucleic acid encoding an enzyme capable of performing targeted genomic integration; a second nucleic acid encoding a donor DNA comprising inducible viral replication (rep) and Capsid (cap) genes, adenoviral auxiliary genes selected from E2A, E4ORF6, and VA, and insulators (optionally selected from HS4, D4Z4), and comprising terminal ends recognized by the enzyme, wherein the inducible viral replication (Rep) and Capsid (cap) genes are controlled by at least one of a tetO-containing P5 promoter, P19 promoter, SV40 promoter, P40 promoter, or a functional variant or fragment thereof; and a third nucleic acid encoding a donor DNA comprising a transgene flanked by AAV inverted terminal repeats (ITRs), to thereby result in a transfected cell that expresses the transgene in association with the viral particle. In aspects, a method of making a viral particle packaging and producer cell line is provided. In embodiments, the method comprises: transfecting an E1A, E1B+ cell with a first nucleic acid encoding an enzyme capable of performing targeted genomic integration, and a second nucleic acid encoding viral packaging genes, viral helper genes, insulators (optionally selected from HS4 and D4Z4), and a transgene, and comprising terminal ends recognized by the enzyme to thereby result in a transfected cell that expresses the transgene in association with a viral particle. In embodiments, the inducible viral replication (Rep) and Capsid (cap) genes are controlled by at least one of a tetO- containing P5 promoter, P19 promoter, SV40 promoter, P40 promoter, or a functional variant or fragment thereof. In embodiments, a mammal-derived, helper RNA mobile element enzyme and donor DNA system is used to produce purified recombinant adeno-associated virus (rAAV) vector stocks. rAAV stocks, comprising an ITR-flanked therapeutic transgene or a gene of interest (GOI), are generated by a one plasmid or two-plasmid system, which creates a single producer cell line or separate packaging and producer cell lines. In mammalian cell-based production systems, the assembly of rAAV vectors typically requires (1) a recombinant vector genome comprising a gene of interest (GOI) and the regulation elements for the GOI expression in target cells (e.g., a promoter, poly A, introns, etc.) flanked by AAV ITRs, (2) the AAV rep and cap genes provided in trans, and (3) Helper functions from adeno-, herpes-, human papilloma- or vaccinia viruses, for replication and rescue of the recombinant genome. A conventional method for production of a vector is co-transfection of a vector plasmid containing the vector genome and a helper plasmid encoding the rep and cap genes into E1A-transformed human embryonic kidney cells (HEK293 cells) infected with Ad (see FIG.4). Aponte-Ubillus et al., Appl Microbiol Biotechnol 2018;102:1045-54. By this method, 100 to 1,000 vector particles per cell, or 108 to 109 particles per ml of crude stock lysate, can usually be prepared. Large-scale transfections are typically desired, and vector virions are required to be purified from liters of crude lysate. Although several modifications designed to increase DNA transfer efficiency and AAV helper gene expression have been developed, the existing protocols still require transfection, electroporation, and preparation of complicated conjugates. At the same time, it would be advantageous to develop a stable packaging cell line that can produce vector particles without a transfection step, thus allowing efficient large-scale stock preparation. Furthermore, AAV vector packaging strategies that rely on overexpression of the viral gene products face challenges such as issues associated with the cellular toxicity of Rep proteins, and the requirement that rep and cap expression levels be tightly regulated for cell viability hence maximal virion production. One strategy to increase viral gene expression during vector production is to use an inducible promoter (e.g., tetracycline (Tet), cumate, coumermycin/novobiocin, etc. see Zhao et al., Hum Gene Ther 2003;14:1619-29) to mimic the state that occurs during wild type AAV infection by amplifying intact fragments of the AAV genome containing the rep and cap genes. The most established method for production of rAAV vectors is the plasmid transfection of human embryonic HEK293 cells. Typically, HEK293 cells are simultaneously transfected by a vector plasmid (containing the GOI) and one or two helper plasmids. The helper plasmid(s) allow the expression of the four Rep proteins, the three AAV structural proteins VP1, VP2, and VP3, the AAP, and the adenoviral auxiliary functions E2A, E4ORF6, and VA RNA. The additional adenoviral E1A/E1B co-factors necessary for rAAV replication are expressed in HEK293 producer cells. See Qiao et al., J Virol 2002;76:1904-13; Yuan et al., Hum Gene Ther 2011;22:613-24; Lock et al., Hum Gene Ther 2010;21:1259- 71. Rep/cap genes and adenoviral helper sequences are either cloned on two separate plasmids or combined on one plasmid, hence evolving from a triple plasmid system to transfection with only two plasmids. Grimm et al. Hum Gene Ther 1998;9:2745-60. The triple plasmid protocol provides versatility with a cap gene that can be switched from one serotype to another. The plasmids are usually produced by conventional techniques in E. coli using bacterial origin and antibiotic-resistance gene or by minicircle (MC) technology. Schnodt et al., Mol Ther Nucleic Acids 2016;5:e355. Although transient transfection in adherent HEK293 cells has been used for large-scale manufacturing of rAAV vectors, it typically requires multiple production batches to fulfill the needs of clinical trials, resulting in lengthy and costly production campaigns. HEK293 cells have been adapted to suspension conditions to be economically viable in the long term. Grieger et al., Mol Ther 2016;24:287-97. Generation of AAV Vector Packaging Cell Lines. In embodiments, a method of making a viral particle packaging and producer cell line (e.g., the AAV cell line) is provided that comprises transfecting an E1A, E1B+ cell with (1) a first nucleic acid encoding an enzyme capable of performing targeted genomic integration, (2) a second nucleic acid encoding a donor DNA comprising inducible viral replication (rep) and Capsid (cap) genes, adenoviral auxiliary genes selected from E2A, E4ORF6, and VA, and insulators (optionally selected from HS4 and D4Z4), and comprising terminal ends recognized by the enzyme, wherein the inducible viral replication (Rep) and Capsid (cap) genes are controlled by at least one of a tetO-containing P5 promoter, P19 promoter, SV40 promoter, P40 promoter, or a functional variant or fragment thereof, and (3) a third nucleic acid encoding a donor DNA comprising a transgene flanked by AAV inverted terminal repeats (ITRs), to thereby result in a transfected cell that expresses the transgene in association with the viral particle. In embodiments, a method of making a viral particle packaging and producer cell line is provided that comprises transfecting an E1A, E1B+ cell with (1) a first nucleic acid encoding an enzyme capable of performing targeted genomic integration, and (2) a second nucleic acid encoding viral packaging genes, viral helper genes, insulators (optionally selected from HS4 and D4Z4), and a transgene, and comprising terminal ends recognized by the enzyme, to thereby result in a transfected cell that expresses the transgene in association with a viral particle. FIG.1A illustrates a non-limiting example of a method for generating AAV, using a single or dual donor DNA to create a producer cell line from a E1+ cell line (e.g., without limitation, an HEK293 cell line) that forms a replication deficient (rep-) AAV particle containing a transgene of interest. As shown in FIG.1A, a donor DNA construct encodes inducible Rep and Cap (“Capsid”) proteins (VP1, VP2, and VP3) and helper genes (E4, E2A, and VA) having end sequences recognized by an enzyme capable of performing targeted genomic integration (e.g., without limitation, a mobile element enzyme), as well as a transgene (or gene) of interest flanked by AAV ITRs. In embodiments, the donor DNA construct is a dual donor DNA comprising a nucleic acid encoding the inducible Rep and Cap proteins and helper genes that is separate from a nucleic acid encoding transgene. In embodiments, the donor DNA construct is a single donor DNA comprising a nucleic acid encoding the inducible Rep and Cap proteins, helper genes, and a transgene. FIG.1B shows an example of an inducible Rep/Cap and helper AAV donor plasmid construct (Kanar), encoding helper E2A, E4ORF6 and VA genes flanked by insulators and mobile element enzyme recognition ends, and used with a helper RNA or DNA (e.g., a mobile element enzyme) to create an AAV producer cell line. FIG.1C shows an example of a plasmid encoding a transgene included between AAV ITRs. The plasmids shown in FIGs.1B and 1C can be combined or can be used separately for transfection into an E1+ cell line to produce an AAV particle comprising the transgene (or gene of interest (GOI)). In embodiments, the method depicted in FIG.1A is performed using a system in accordance with embodiments of the present disclosure shown in FIG.2D. The system of FIG.2D is an integrative, non-viral donor DNA system for site- specific, stable genomic integration. The system allows producing any recombinant AAV serotype by the insertion of inducible AAV rep/cap genes and helper genes (E4, E2A, and VA) in specific genomic locations in a cell, such as, e.g., HEK293 (E1+) cells, a genetically engineered CHO-K1 cell line, or in other cells. The system of FIG.2D comprises comprising a nucleic acid (e.g., helper RNA [which is different from helper genes]) encoding an enzyme capable of performing targeted genomic integration (e.g., without limitation, a mobile element enzyme) and a nucleic acid encoding a transgene of interest (donor DNA). The helper RNA is translated into a bioengineered enzyme (e.g., integrase, recombinase, or mobile element enzyme) that recognizes specific ends and seamlessly inserts the transgene of interest into the human genome in a site-specific manner without a footprint. The present disclosure provides, in embodiments, methods for producing AAV packaging cell lines capable of regulated gene amplification. In embodiments, the methods of the present disclosure employ tetracycline repressor-controlled (tetR) tetO containing promoters (Yao F et al., Hum Gene Ther., 9:1939-1950, 1998) or reverse tetracycline-controlled transactivator (rtTA)-controlled gene expression and SV40 origin replication to regulate the expression of the AAV rep and cap genes, and the AdV5 E2A and E4ORF6. In embodiments, binding of doxycycline to t-REx or rtTA will initiate transcription from the tetO-containing promoters or SV40 T-antigen gene, leading to expression of the AAV rep and cap genes, and AdV5, E2A, and E4ORF6 genes (FIG. 1B). In embodiments, the construct shown in FIG.1B is permanently integrated into a E1a/E1b containing cell line (e.g., without limitations, an HEK293 cell line stably expressing tetR gene). Transfection in media with an inducer (e.g., without limitation, doxycycline) of inducible rep and cap genes results in high-level production of Rep and Cap proteins and AAV virion assembly. Because Rep protein expression is inducible, this strategy avoids the toxicity of Rep proteins. In embodiments, producer cells can be generated from these packaging lines by the addition of a vector construct comprising a transgene (or a GOI) flanked by AAV ITRs that can excise, replicate, and be packaged into virions once rep and cap are expressed. Alternatively, in embodiments, the transgene construct is included in the same plasmid as the donor DNA, in a “single plasmid” protocol. In embodiments, methods for an AAV packaging and producer cell line are provided. In embodiments, the AAV rep and cap genes are under control of an inducible promoter (e.g., t-Rex or rtTA), such that the rep and cap genes, the AAV vector DNA sequences, and the essential helper genes are encoded on a single donor plasmid flanked by mobile element enzyme recognition sequences (FIG.1B). In embodiments, a transgene is encoded separately from the donor plasmid (see, e.g., FIG.1C). In embodiments, the donor plasmid comprises a transgene such that a one-plasmid (e.g., without limitation, a minicircle) is created and used to generate a transgene-specific cell line (e.g., an E1A, E1B+ cell such as, without limitation, a HEK293 cell line) that is able to produce a clinical grade product for human gene therapy. In embodiments, methods for making a packaging and producer cell line with a dual donor/helper system are provided. In the dual donor/helper system, inducible Rep and Cap genes (sometimes collectively referred to as “Rep/Cap genes”), and a transgene of interest are encoded by respective separate nucleic acids. In embodiments, the dual donor/helper system includes, without limitation, a nucleic acid encoding inducible Rep and Cap genes, a nucleic acid (e.g., a donor expression vector, such as a helper AAV donor plasmid) encoding a transgene of interest, and a nucleic acid encoding an enzyme capable of performing targeted genomic integration (e.g., without limitation, a mobile element enzyme). Accordingly, in aspects, a method of making a viral particle packaging and producer cell line is provided. In embodiments, the method comprises transfecting an E1A, E1B+ cell with: a first nucleic acid encoding an enzyme capable of performing targeted genomic integration; a second nucleic acid encoding a donor DNA comprising inducible viral replication (Rep) and Capsid (Cap) genes, adenoviral auxiliary genes selected from E2A, E4ORF6, and VA, and insulators (e.g., without limitation, HS4 or D4Z4), and comprising terminal ends recognized by the enzyme; and a third nucleic acid encoding a donor DNA comprising a transgene flanked by AAV inverted terminal repeats (ITRs), to thereby result in a transfected cell that expresses the transgene in association with the viral particle. In aspects, methods for making a packaging and producer cell line with a single donor/helper system are provided. In the single donor/helper system, inducible Rep/Cap genes and a nucleic acid (e.g., a donor expression vector, such as a helper AAV donor plasmid) encoding a transgene of interest are encoded by a same construct. In embodiments, the single donor/helper system includes, without limitation, a nucleic acid (e.g., a donor expression vector) encoding inducible Rep and Cap genes and a transgene of interest, and a nucleic acid encoding an enzyme capable of performing targeted genomic integration (e.g., without limitation, a mobile element enzyme). Accordingly, in aspects, a method of making a viral particle packaging and producer cell line is provided. In embodiments, the method comprises transfecting an E1A, E1B+ cell with a first nucleic acid encoding an enzyme capable of performing targeted genomic integration; and a second nucleic acid encoding viral packaging genes, viral helper genes, insulators (e.g., without limitation, HS4, D4Z4), and a transgene, and comprising terminal ends recognized by the enzyme, to thereby result in a transfected cell that expresses the transgene in association with a viral particle. In embodiments, the E1A, E1B+ cell is, without limitation, HEK293, E1A, E1B+ engineered CHO-K1, or Sf9 cell line. In embodiments, the viral helper genes comprise adenoviral auxiliary genes. In embodiments, the adenoviral auxiliary genes are selected from one or more of E1A, E1B, E4, E2A, and VA of an AAV, optionally E1A and E1B. In embodiments, the viral packaging genes comprise rep and cap genes of viral replication (Rep) and Capsid (Cap) proteins of an AAV. In embodiments, the expression of the Rep and Cap proteins is controlled by an inducible promoter, optionally an antibiotic-dependent promoter. In embodiments, the antibiotic-dependent promoter is tetracycline- or a variant thereof dependent promoter and a tetracycline repressor-controlled (tetR) or tetracycline-controlled transactivator (rtTA) system is used. In embodiments, the antibiotic-dependent promoter is a coumermycin/novobiocin promoter, or a variant thereof. In embodiments, the inducible viral replication (Rep) and Capsid (cap) genes are controlled by at least one of a tetO- containing P5 promoter, P19 promoter, SV40 promoter, P40 promoter, or a functional variant or fragment thereof. In embodiments, the tetO-containing AAV2 P40 promoter comprises a nucleotide sequence of SEQ ID NO: 816, or a functional variant or fragment thereof, or sequence having at least about 70%, or at least about 80%, or at least about 85%, or at least about 90%, or at least about 95%, or at least about 97% identity thereto, or at least about 98% identity thereto, or at least about 99% identity thereto or at least about 10 mutations, or at least about 9 mutations, or at least about 8 mutations, or at least about 7 mutations, or at least about 6 mutations, or at least about 5 mutations, or at least about 4 mutations, or at least about 3 mutations, or at least about 2 mutations, or at least about 1 mutation. In embodiments, the P40 intron is an AAV2 P40 intron. In embodiments, the AAV2 P40 intron comprises a C276A substitution, or a substitution at position corresponding thereto relative to SEQ ID NO: 818. In embodiments, the AAV2 P40 intron comprises one or more mutated translation start sites (ATGs), optionally wherein the translation start sites are mutated to one of CTG, ACG, and TTG. In embodiments, the AAV2 P40 intron comprises substitutions at one or more positions A13, A32, T42, A61, A71, A89, A203, A246, A258, and T282, or one or more positions corresponding thereto, relative to SEQ ID NO: 818. In embodiments, the AAV2 P40 intron comprises substitutions at one or more positions A13C, A32C, T42C, A61C, A71T, A89C, A203T, A246C, A258C, and T282C corresponding to SEQ ID NO: 818. In embodiments, the AAV2 P40 intron comprises the nucleotide sequence of SEQ ID NO: 818, or a functional variant or fragment thereof, or sequence having at least about 70%, or at least about 80%, or at least about 85%, or at least about 90%, or at least about 95%, or at least about 97% identity thereto, or at least about 98% identity thereto, or at least about 99% identity thereto, or at least about 10 mutations, or at least about 9 mutations, or at least about 8 mutations, or at least about 7 mutations, or at least about 6 mutations, or at least about 5 mutations, or at least about 4 mutations, or at least about 3 mutations, or at least about 2 mutations, or at least about 1 mutation. In embodiments, the AAV2 P40 intron comprises the nucleotide sequence of SEQ ID NO: 819, or a functional variant or fragment thereof, or sequence having at least about 70%, or at least about 80%, or at least about 85%, or at least about 90%, or at least about 95%, or at least about 97% identity thereto, or at least about 98% identity thereto, or at least about 99% identity thereto, or at least about 10 mutations, or at least about 9 mutations, or at least about 8 mutations, or at least about 7 mutations, or at least about 6 mutations, or at least about 5 mutations, or at least about 4 mutations, or at least about 3 mutations, or at least about 2 mutations, or at least about 1 mutation. In embodiments, the AAV2 P40 intron comprises the nucleotide sequence of SEQ ID NO: 817, or a functional variant or fragment thereof, or sequence having at least about 70%, or at least about 80%, or at least about 85%, or at least about 90%, or at least about 95%, or at least about 97% identity thereto, or at least about 98% identity thereto, or at least about 99% identity thereto, or at least about 10 mutations, or at least about 9 mutations, or at least about 8 mutations, or at least about 7 mutations, or at least about 6 mutations, or at least about 5 mutations, or at least about 4 mutations, or at least about 3 mutations, or at least about 2 mutations, or at least about 1 mutation. In embodiments, the modified TATA box element has the nucleotide sequence of TATATAA. In embodiments, the tetO- containing AAV2 P40 promoter or a functional variant or fragment thereof, comprising a modified TATA box element and a modified AAV2 P40 intron comprises the nucleotide sequence of SEQ ID NO: 823, or a functional variant or fragment thereof, or sequence having at least about 70%, or at least about 80%, or at least about 85%, or at least about 90%, or at least about 95%, or at least about 97% identity thereto, or at least about 98% identity thereto, or at least about 99% identity thereto, or at least about 10 mutations, or at least about 9 mutations, or at least about 8 mutations, or at least about 7 mutations, or at least about 6 mutations, or at least about 5 mutations, or at least about 4 mutations, or at least about 3 mutations, or at least about 2 mutations, or at least about 1 mutation. In embodiments, the tetO-containing P40 promoter is a tetO-containing AAV2 P40 promoter. In embodiments, the AAV2 P40 promoter comprises the nucleotide sequence of SEQ ID NO: 820, or a functional variant or fragment thereof, or sequence having at least about 70%, or at least about 80%, or at least about 85%, or at least about 90%, or at least about 95%, or at least about 97% identity thereto, or at least about 98% identity thereto, or at least about 99% identity thereto, or at least about 10 mutations, or at least about 9 mutations, or at least about 8 mutations, or at least about 7 mutations, or at least about 6 mutations, or at least about 5 mutations, or at least about 4 mutations, or at least about 3 mutations, or at least about 2 mutations, or at least about 1 mutation. In embodiments, the modified P40 intron is a AAV9 P40 intron. In embodiments, the modified AAV9 P40 intron comprises the nucleotide sequence of SEQ ID NO: 821, or a functional variant or fragment thereof, or sequence having at least about 70%, or at least about 80%, or at least about 85%, or at least about 90%, or at least about 95%, or at least about 97% identity thereto, or at least about 98% identity thereto, or at least about 99% identity thereto, or at least about 10 mutations, or at least about 9 mutations, or at least about 8 mutations, or at least about 7 mutations, or at least about 6 mutations, or at least about 5 mutations, or at least about 4 mutations, or at least about 3 mutations, or at least about 2 mutations, or at least about 1 mutation. In embodiments, the modified TATA box element has the nucleotide sequence of TATATAA. In embodiments, the tetO-containing AAV2 P40 promoter or a functional variant or fragment thereof, comprising a modified TATA box element and a modified AAV9 P40 intron comprises the nucleotide sequence of SEQ ID NO: 822, or a functional variant or fragment thereof, or sequence having at least about 70%, or at least about 80%, or at least about 85%, or at least about 90%, or at least about 95%, or at least about 97% identity thereto, or at least about 98% identity thereto, or at least about 99% identity thereto, or at least about 10 mutations, or at least about 9 mutations, or at least about 8 mutations, or at least about 7 mutations, or at least about 6 mutations, or at least about 5 mutations, or at least about 4 mutations, or at least about 3 mutations, or at least about 2 mutations, or at least about 1 mutation. In embodiments, the tetO-containing AAV2 P40 promoter or a functional variant or fragment thereof, comprising a modified TATA box element and a modified AAV9 P40 intron comprises a nucleotide sequence of SEQ ID NO: 824, or a functional variant or fragment thereof, or sequence having at least about 70%, or at least about 80%, or at least about 85%, or at least about 90%, or at least about 95%, or at least about 97% identity thereto, or at least about 98% identity thereto, or at least about 99% identity thereto, or at least about 10 mutations, or at least about 9 mutations, or at least about 8 mutations, or at least about 7 mutations, or at least about 6 mutations, or at least about 5 mutations, or at least about 4 mutations, or at least about 3 mutations, or at least about 2 mutations, or at least about 1 mutation. In embodiments, the P40 promoter further comprises one or more enhancer elements. In embodiments, the enhancer element comprises one or more cis-acting elements, optionally selected from an Sp1 binding site, GC rich sequence, GCGGAAC motif, TAATGARAT element, AP1 binding site, and CCAAT box element. In embodiments, the enhancer element comprises about 1 to about 5 Sp1 binding sites, optionally about 1, or about 2, or about 3, or about 4, or about 5 Sp1 binding sites. In embodiments, the enhancer element comprises about 1 or about 2 GC rich sequences. In embodiments, the enhancer element is derived from an hCMV Enhancer Element-3. In embodiments, the hCMV Enhancer Element-3 comprises the nucleotide sequence of SEQ ID NO: 830 or a functional variant or fragment thereof, or sequence having at least about 70%, or at least about 80%, or at least about 85%, or at least about 90%, or at least about 95%, or at least about 97% identity thereto, or at least about 98% identity thereto, or at least about 99% identity thereto, or at least about 10 mutations, or at least about 9 mutations, or at least about 8 mutations, or at least about 7 mutations, or at least about 6 mutations, or at least about 5 mutations, or at least about 4 mutations, or at least about 3 mutations, or at least about 2 mutations, or at least about 1 mutation. In embodiments, the viral particle is an AAV of a suitable serotype. In embodiments, the AAV is selected from AAV1, AAV2, AAV5, AAV6, AAV7, AAV8, and AAV9. In some embodiments, the AAV serotype is a natural serotype or a synthetically engineered serotype. In embodiments, the AAV is AAV2. In embodiments, the AAV is AAV9. In embodiments, the viral particle is an AAV of any of the AAV serotypes. Today, 12 AAV serotypes are known, and more than 100 variants have been identified. Different serotype capsids can infect different tissues or culture cells in different ways, which depend on the primary receptor and co-receptors on the cell surface or the intracellular trafficking pathway itself. In embodiments, any one or more of the first, second, and third nucleic acids is encoded by a single nucleic acid. In embodiments, the second nucleic acid and the third nucleic acid are included in a single expression vector. In embodiments, the first and second nucleic acids are included in a single expression vector, and the third nucleic acid is included in an expression vector that is different from the expression vector including the first and second nucleic acids. In embodiments, the single expression vector comprises a plasmid. In embodiments, the plasmid comprises an AAV2 CAP under the control of the tetO-containing hCMV-derived enhancer element 3/AAV2 P40 promoter with a modified AAV2 P40 TATA element and modified AAV2 P40 intron_1 (a at position 276 substituted for C) and WPRE/modified ICP27 poly A followed by ICP0 promoter-Zeocin resistant gene-ICP0 poly A cassette flanked with SB ITRs. In embodiments, the plasmid comprises the nucleotide sequence of SEQ ID NO: 825 or a functional variant or fragment thereof, or sequence having at least about 70%, or at least about 80%, or at least about 85%, or at least about 90%, or at least about 95%, or at least about 97% identity thereto, or at least about 98% identity thereto, or at least about 99% identity thereto, or at least about 10 mutations, or at least about 9 mutations, or at least about 8 mutations, or at least about 7 mutations, or at least about 6 mutations, or at least about 5 mutations, or at least about 4 mutations, or at least about 3 mutations, or at least about 2 mutations, or at least about 1 mutation. In embodiments, the plasmid comprises an AAV2 CAP under the control of the tetO-containing hCMV enhancer element 3/AAV2 P40 promoter with a modified AAV2 P40 TATA element and MODIFIED AAV2 P40 intron_2 and WPRE/modified ICP27 poly A followed by ICP0 promoter-Zeocin resistant gene-ICP0 poly A cassette flanked with SB ITRs. In embodiments, the plasmid comprises the nucleotide sequence of SEQ ID NO: 826 or a functional variant or fragment thereof, or sequence having at least about 70%, or at least about 80%, or at least about 85%, or at least about 90%, or at least about 95%, or at least about 97% identity thereto, or at least about 98% identity thereto, or at least about 99% identity thereto, or at least about 10 mutations, or at least about 9 mutations, or at least about 8 mutations, or at least about 7 mutations, or at least about 6 mutations, or at least about 5 mutations, or at least about 4 mutations, or at least about 3 mutations, or at least about 2 mutations, or at least about 1 mutation. In embodiments, the plasmid comprises an engineered mobile element enzyme ITR containing plasmid that encodes 1) AAV2_REP 78/68 ORFs under the control of P5TO1 promoter, REP 52/40 under the control of P19 promoter, 2)Ad5 E2A_IRES_Ad5 E4 OR F6 under the control of the tetO-containing SV40 promoter, 3) VA RNAs under the control of HSV-2 ICP4 promoter, and 4) hygro-B resistant gene under the control of TK promoter. In embodiments, the plasmid comprises the nucleotide sequence of SEQ ID NO: 841 or a functional variant or fragment thereof, or sequence having at least about 70%, or at least about 80%, or at least about 85%, or at least about 90%, or at least about 95%, or at least about 97% identity thereto, or at least about 98% identity thereto, or at least about 99% identity thereto, or at least about 10 mutations, or at least about 9 mutations, or at least about 8 mutations, or at least about 7 mutations, or at least about 6 mutations, or at least about 5 mutations, or at least about 4 mutations, or at least about 3 mutations, or at least about 2 mutations, or at least about 1 mutation. In embodiments, the plasmid comprises an engineered mobile element enzyme ITR containing plasmid that encodes 1) AAV2_REP78/68 ORFs under the control of P5TO1 promoter, REP 52/40 under the control of P19 promoter, and CAPs under the native P40 promoter, 2) Ad5 E2A_IRES_Ad5 E4 ORF6 under the control of the tetO-containing SV40 promoter, 3) VA RNAs under the control of HSV-1 ICP4 promoter, and 4) hygro-B resistant gene under the control of TK promoter. In embodiments, the plasmid comprises the nucleotide sequence of SEQ ID NO: 842, or a functional variant or fragment thereof, or sequence having at least about 70%, or at least about 80%, or at least about 85%, or at least about 90%, or at least about 95%, or at least about 97% identity thereto, or at least about 98% identity thereto, or at least about 99% identity thereto, or at least about 10 mutations, or at least about 9 mutations, or at least about 8 mutations, or at least about 7 mutations, or at least about 6 mutations, or at least about 5 mutations, or at least about 4 mutations, or at least about 3 mutations, or at least about 2 mutations, or at least about 1 mutation. In embodiments, the plasmid comprises an engineered mobile element enzyme ITR containing plasmid that encodes 1) AAV2_REP78/68 ORFs under the control of P5TO1 promoter, REP 52/40 under the control of P19 promoter, 2) AAV2 CAPs under the tetO-containing P40 promoter with AAV9 P40 intron, 3) Ad5 E2A_IRES_Ad5 E4 ORF6 under the control of the tetO-containing SV40 promoter, 4) VA RNAs under the control of HSV-1 ICP4 promoter, and 5) hygro- B resistant gene under the control of TK promoter. In embodiments, the plasmid comprises the nucleotide sequence of SEQ ID NO: 843, or a functional variant or fragment thereof, or sequence having at least about 70%, or at least about 80%, or at least about 85%, or at least about 90%, or at least about 95%, or at least about 97% identity thereto, or at least about 98% identity thereto, or at least about 99% identity thereto, or at least about 10 mutations, or at least about 9 mutations, or at least about 8 mutations, or at least about 7 mutations, or at least about 6 mutations, or at least about 5 mutations, or at least about 4 mutations, or at least about 3 mutations, or at least about 2 mutations, or at least about 1 mutation. In embodiments, any one of the first, second, and third nucleic acids is RNA, optionally mRNA, optionally synthetic mRNA. In embodiments, any one of the first, second, and third nucleic acids is DNA, optionally plasmid DNA. In embodiments, any one of the first, second, and third nucleic acids is an expression vector, wherein the expression vector is optionally a plasmid. In embodiments, the transfected cell generates using the method of making a viral particle packaging and producer cell line in accordance with embodiments of the present disclosure comprises a donor DNA comprising the transgene flanked by AAV inverted terminal repeats (ITRs). In embodiments, the method further comprises culturing the transfected cell in a medium that expands a population of the transfected cells to create a stably transfected packaging and producer cell line. In embodiments, the stably transfected producer cell line is capable of producing replication-deficient viral particles in association with the transgene. In embodiments, the transfection comprises electroporation, nucleofection, lipofection, or calcium phosphate transfection. In embodiments, the method is helper virus-free. In embodiments, the cell is human embryonic kidney (HEK293), Chinese hamster ovary (CHO) E1A, E1B+ engineered CHO-K1, or Spodoptera frugiperda (Sf9) cell line, baby hamster kidney (BHK), vero cell. In embodiments, the viral helper genes comprise adenoviral auxiliary genes. In embodiments, the adenoviral auxiliary genes are selected from one or more of E1A, E1B, E4, E2A, and VA of an AAV, optionally E1A and E1B. In embodiments, the viral packaging genes comprise rep and cap genes of viral replication (Rep) and Capsid (Cap) proteins of an AAV. In embodiments, the expression of the Rep and Cap proteins is controlled by an inducible promoter. In embodiments, the inducible promoter is an antibiotic-dependent promoter, optionally a tetracycline-dependent promoter or a variant thereof, or a cumate or coumermycin/novobiocin promoter or a variant thereof. In embodiments, the viral particle is an AAV, and optionally wherein the AAV is selected from AAV1, AAV5, AAV2, AAV6, AAV7, AAV8, and AAV9. In embodiments, any one or more of the first, second, and third nucleic acids is encoded by a single nucleic acid. In embodiments, the second nucleic acid and the third nucleic acid are included in a single expression vector. In embodiments, the first and second nucleic acids are included in a single expression vector, and the third nucleic acid is included in an expression vector that is different from the expression vector including the first and second nucleic acids. In embodiments, the expression vector is or comprises a plasmid. In embodiments, the plasmid comprises an AAV2 CAP under the control of the tetO-containing hCMV-derived enhancer element 3/AAV2 P40 promoter with a modified AAV2 P40 TATA element and modified AAV2 P40 intron_1 (a at position 276 substituted for C) and WPRE/modified ICP27 poly A followed by ICP0 promoter-Zeocin resistant gene-ICP0 poly A cassette flanked with SB ITRs. In embodiments, the plasmid comprises the nucleotide sequence of SEQ ID NO: 825 or a functional variant or fragment thereof, or sequence having at least about 70%, or at least about 80%, or at least about 85%, or at least about 90%, or at least about 95%, or at least about 97% identity thereto, or at least about 98% identity thereto, or at least about 99% identity thereto, or at least about 10 mutations, or at least about 9 mutations, or at least about 8 mutations, or at least about 7 mutations, or at least about 6 mutations, or at least about 5 mutations, or at least about 4 mutations, or at least about 3 mutations, or at least about 2 mutations, or at least about 1 mutation. In embodiments, the plasmid comprises an AAV2 CAP under the control of the tetO-containing hCMV enhancer element 3/AAV2 P40 promoter with a modified AAV2 P40 TATA element and MODIFIED AAV2 P40 intron_2 and WPRE/modified ICP27 poly A followed by ICP0 promoter-Zeocin resistant gene-ICP0 poly A cassette flanked with SB ITRs. In embodiments, the plasmid comprises the nucleotide sequence of SEQ ID NO: 826 or a functional variant or fragment thereof, or sequence having at least about 70%, or at least about 80%, or at least about 85%, or at least about 90%, or at least about 95%, or at least about 97% identity thereto, or at least about 98% identity thereto, or at least about 99% identity thereto, or at least about 10 mutations, or at least about 9 mutations, or at least about 8 mutations, or at least about 7 mutations, or at least about 6 mutations, or at least about 5 mutations, or at least about 4 mutations, or at least about 3 mutations, or at least about 2 mutations, or at least about 1 mutation. In embodiments, the plasmid comprises an engineered mobile element enzyme ITR containing plasmid that encodes 1) AAV2_REP 78/68 ORFs under the control of P5TO1 promoter, REP 52/40 under the control of P19 promoter, 2)Ad5 E2A_IRES_Ad5 E4 OR F6 under the control of the tetO-containing SV40 promoter, 3) VA RNAs under the control of HSV-1 ICP4 promoter, and 4) hygro-B resistant gene under the control of TK promoter. In embodiments, the plasmid comprises the nucleotide sequence of SEQ ID NO: 841 or a functional variant or fragment thereof, or sequence having at least about 70%, or at least about 80%, or at least about 85%, or at least about 90%, or at least about 95%, or at least about 97% identity thereto, or at least about 98% identity thereto, or at least about 99% identity thereto, or at least about 10 mutations, or at least about 9 mutations, or at least about 8 mutations, or at least about 7 mutations, or at least about 6 mutations, or at least about 5 mutations, or at least about 4 mutations, or at least about 3 mutations, or at least about 2 mutations, or at least about 1 mutation. In embodiments, the plasmid comprises an engineered mobile element enzyme ITR containing plasmid that encodes 1) AAV2_REP78/68 ORFs under the control of P5TO1 promoter, REP 52/40 under the control of P19 promoter, and CAPs under the native P40 promoter, 2) Ad5 E2A_IRES_Ad5 E4 ORF6 under the control of the tetO-containing SV40 promoter, 3) VA RNAs under the control of HSV-1 ICP4 promoter, and 4) hygro-B resistant gene under the control of TK promoter. In embodiments, the plasmid comprises the nucleotide sequence of SEQ ID NO: 842, or a functional variant or fragment thereof, or sequence having at least about 70%, or at least about 80%, or at least about 85%, or at least about 90%, or at least about 95%, or at least about 97% identity thereto, or at least about 98% identity thereto, or at least about 99% identity thereto, or at least about 10 mutations, or at least about 9 mutations, or at least about 8 mutations, or at least about 7 mutations, or at least about 6 mutations, or at least about 5 mutations, or at least about 4 mutations, or at least about 3 mutations, or at least about 2 mutations, or at least about 1 mutation. In embodiments, the plasmid comprises an engineered mobile element enzyme ITR containing plasmid that encodes 1) AAV2_REP78/68 ORFs under the control of P5TO1 promoter and REP 52/40 under the control of P19 promoter, 2) AAV2 CAPs under the tetO-containing P40 promoter with AAV9 P40 intron, 3) Ad5 E2A_IRES_Ad5 E4 ORF6 under the control of the tetO-containing SV40 promoter, 4) VA RNAs under the control of HSV-1 ICP4 promoter, and 5) hygro- B resistant gene under the control of TK promoter. In embodiments, the plasmid comprises the nucleotide sequence of SEQ ID NO: 843, or a functional variant or fragment thereof, or sequence having at least about 70%, or at least about 80%, or at least about 85%, or at least about 90%, or at least about 95%, or at least about 97% identity thereto, or at least about 98% identity thereto, or at least about 99% identity thereto, or at least about 10 mutations, or at least about 9 mutations, or at least about 8 mutations, or at least about 7 mutations, or at least about 6 mutations, or at least about 5 mutations, or at least about 4 mutations, or at least about 3 mutations, or at least about 2 mutations, or at least about 1 mutation. In embodiments, any one of the first, second, and third nucleic acids is or comprises RNA, optionally mRNA, optionally synthetic mRNA or modified mRNA. In embodiments, any one of the first, second, and third nucleic acids is DNA, optionally plasmid DNA. In embodiments, any one of the first, second, and third nucleic acids is an expression vector, wherein the expression vector is optionally a plasmid. In embodiments, the transgene is flanked by AAV inverted terminal repeats (ITRs). In embodiments, the transgene is flanked by Sleeping beauty ITRs. SEQ ID NO: 831: Sleeping Beauty LE ITR Sequence (IR/DR(L) Lmut44) (231 bp including TATA): 5’- tatacagttgaagtcggaagtttacatacacttaagttggagtcattaaaactcgtttttcaactactccacaaatttcttgttaacaaacaatagttttggcaagtcagttag gacatctactttgtgcatgacacaagtcatttttccaacaattgtttacagacagattatttcacttataattcactgtatcacaattccagtgggtcagaagtttacatacact aa– 3’ SEQ ID NO: 832: Sleeping Beauty RE ITR Sequence (IR/DR (R) Rmut13, Δ130, 143, 150) (232 bp including TATA): 5’- ttgagtgtatgtaaacttctgacccactgggaatgtgatgaaagaaataaaagctgaaatgaatcattctctctactattattctgatatttcacattcttaaaataaagtggt gatcctaactgacctaagacagggaatttttactaggattaaatgtcaggaattgtgaaaaagtgagtttaaatgtatttggctaaggtgtatgtaaacttccgacttcaac tgtata – 3’ In embodiments, the ITRs comprise the nucleotide sequence of SEQ ID NO: 831 and/or SEQ ID NO: 832, or a functional variant or fragment thereof, or sequence having at least about 70%, or at least about 80%, or at least about 85%, or at least about 90%, or at least about 95%, or at least about 97% identity thereto, or at least about 98% identity thereto, or at least about 99% identity thereto, or at least about 10 mutations, or at least about 9 mutations, or at least about 8 mutations, or at least about 7 mutations, or at least about 6 mutations, or at least about 5 mutations, or at least about 4 mutations, or at least about 3 mutations, or at least about 2 mutations, or at least about 1 mutation. In embodiments, the transgene or transposon has a size of about 200,000 base pairs or less. In embodiments, the transgene or transposon has a size of at least 200,000 base pairs. In embodiments, the transgene or transposon has a size of about 200,000 base pairs. In embodiments, the transgene or transposon has a size of about 150,000 base pairs or less. In embodiments, the transgene or transposon has a size of at least 150,000 base pairs. In embodiments, the transgene or transposon has a size of about 150,000 base pairs. In embodiments, the transgene or transposon has a size of about 100,000 base pairs or less. In embodiments, the transgene or transposon has a size of at least 100,000 base pairs. In embodiments, the transgene or transposon has a size of about 100,000 base pairs. In embodiments, the transgene or transposon has a size of about 75,000 base pairs or less. In embodiments, the transgene or transposon has a size of at least 75,000 base pairs. In embodiments, the transgene or transposon has a size of about 75,000 base pairs. In embodiments, the transgene or transposon has a size of about 50,000 base pairs or less. In embodiments, the transgene or transposon has a size of at least 50,000 base pairs. In embodiments, the transgene or transposon has a size of about 50,000 base pairs. In embodiments, the method of the present disclosure further comprises culturing the transfected cell in a medium that expands a population of the transfected cells to create a stably transfected packaging and producer cell line. In embodiments, the stably transfected producer cell line is capable of producing replication-deficient viral particles in association with the transgene. In embodiments, the transfection comprises electroporation, nucleofection, lipofection, or calcium phosphate transfection. In embodiments, the method is helper virus-free. In embodiments, the enzyme capable of performing targeted genomic integration is a recombinase. In embodiments, the recombinase is an integrase or a mobile element enzyme. In embodiments, the enzyme is a mobile element enzyme. In embodiments, the enzyme is derived from Bombyx mori, Xenopus tropicalis, Trichoplusia ni, Myotis lucifugus, Rhinolophus ferrumequinum, Rousettus aegyptiacus, Phyllostomus discolor, Myotis myotis, Pteropus vampyrus, Pipistrellus kuhlii, Molossus molossus, Pan troglodytes, or Homo sapiens. In embodiments, the enzyme is an engineered version, including but not limited to hyperactive forms, of an enzyme derived from Bombyx mori, Xenopus tropicalis, Trichoplusia ni, Myotis lucifugus, Rhinolophus ferrumequinum, Rousettus aegyptiacus, Phyllostomus discolor, Myotis myotis, Pteropus vampyrus, Pipistrellus kuhlii, Molossus molossus, Pan troglodytes, or Homo sapiens. In embodiments, the mobile element enzyme is from one or more of the Sleeping beauty, Tn1, Tn2, Tn3, Tn5, Tn7, Tn9, Tn10, Tn552, Tn903, Tn1000/Gamma-delta, Tn/O, tnsA, tnsB, tnsC, tniQ, IS10, ISS, IS911, Minos, piggyBac, Tol2, Mos1, Himar1, Hermes, Tol2, Minos, Tel, P-element, MuA, Ty1, Chapaev, transib, Tc1/mariner, or Tc3 donor DNA system, or biologically active fragments variants thereof, inclusive of hyperactive variants. In embodiments, the mobile element enzyme has the amino acid sequence of SEQ ID NO: 1, or an amino acid sequence having at least about 80%, or an amino acid sequence having at least about 90%, or at least about 93%, or at least about 95%, or at least about 97%, or at least about 98%, or at least about 99% identity thereto. SEQ ID NO: 1: MLT mobile element enzyme protein (amino acid sequence of a variant of the hyperactive mobile element enzyme with S at position 8 and C at position 13 (572 amino acids) MAQHSDYSDDEFCADKLSNYSCDSDLENASTSDEDSSDDEVMVRPRTLRRRRISSSSSDSESDIEGGREEWSHV DNPPVLEDFLGHQGLNTDAVINNIEDAVKLFIGDDFFEFLVEESNRYYNQNRNNFKLSKKSLKWKDITPQEMKK FLGLIVLMGQVRKDRRDDYWTTEPWTETPYFGKTMTRDRFRQIWKAWHFNNNADIVNESDRLCKVRPVLDYFVP KFINIYKPHQQLSLDEGIVPWRGRLFFRVYNAGKIVKYGILVRLLCESDTGYICNMEIYCGEGKRLLETIQTVV SPYTDSWYHIYMDNYYNSVANCEALMKNKFRICGTIRKNRGIPKDFQTISLKKGETKFIRKNDILLQVWQSKKP VYLISSIHSAEMEESQNIDRTSKKKIVKPNALIDYNKHMKGVDRADQYLSYYSILRRTVKWTKRLAMYMINCAL FNSYAVYKSVRQRKMGFKMFLKQTAIHWLTDDIPEDMDIVPDLQPVPSTSGMRAKPPTSDPPCRLSMDMRKHTL QAIVGSGKKKNILRRCRVCSVHKLRSETRYMCKFCNIPLHKGACFEKYHTLKNY In embodiments, the mobile element enzyme comprises an amino acid other than serine at the position corresponding to position 2 of SEQ ID NO: 1. In embodiments, the amino acid is a non-polar aliphatic amino acid, optionally a non-polar aliphatic amino acid optionally selected from G, A, V, L, I and P, optionally A. In embodiments, the mobile element enzyme does not have additional residues at the C terminus relative to SEQ ID NO: 1. In embodiments, the enzyme has one or more mutations which confer hyperactivity. In embodiments, the enzyme has one or more amino acid substitutions selected from S8X1 and/or C13X2, or positions corresponding thereto relative to SEQ ID NO: 1. In embodiments, the enzyme has S8X1 and/or C13X2 substitutions, at positions corresponding thereto relative to SEQ ID NO: 1. In embodiments, the enzyme has S8X1 and C13X2 substitutions, at positions corresponding thereto relative to SEQ ID NO: 1. In embodiments, the enzyme has S8X1 substitution, at position corresponding thereto relative to SEQ ID NO: 1. In embodiments, the enzyme has C13X2 substitution, at positions corresponding thereto relative to SEQ ID NO: 1. In embodiments, X1 is selected from G, A, V, L, I, and P and X2 is selected from K, R, and H. In embodiments, X1 is P and X2 is R. In embodiments, the enzyme of the present disclosure comprises an amino acid sequence of SEQ ID NO: 11. In embodiments, the enzyme comprises at least one, at least five, at least seven, at least nine, or ten substitutions at positions corresponding to: 5, 8, 9, 10, 11, 14, 22, 36, 37, 54, 130, 239, 281, 282, 283, 284, 285, 294, 300, 310, 330, 331, 332, 333, 334, 335, 336, 337, 338, 339, 345, 375, 416, 427, 475, 481, 491, 520, and/or 561 of SEQ ID NO: 11. In embodiments, the enzyme comprises one, two, three, four, five, six, seven, eight, nine, or ten substitutions at positions corresponding to: 5, 8, 9, 10, 11, 14, 22, 36, 37, 54, 130, 239, 281, 282, 283, 284, 285, 294, 300, 310, 330, 331, 332, 333, 334, 335, 336, 337, 338, 339, 345, 375, 416, 427, 475, 481, 491, 520, and/or 561 of SEQ ID NO: 11. In embodiments, the enzyme comprises at least one, at least five, at least seven, at least nine, or ten substitutions selected from S5P, S8F, D9G, D10G, E11G, A14V, T22C, S36G, T37C, S54N, K130T, G239R, Y281A, C282A, G283A, E284A, G285A, T294A, T300A, N310A, G330A, T331A, I332A, R333A, K334A, N335A, R336A, G337A, I338A, P339A, I345V, T375G, D416A, R427H, D475G, M481V, P491Q, A520T, and A561T, wherein the positions are corresponding to positions of SEQ ID NO: 11. In embodiments, the enzyme comprises one, two, three, four, five, six, seven, eight, nine, or ten substitutions selected from S5P, S8F, D9G, D10G, E11G, A14V, T22C, S36G, T37C, S54N, K130T, G239R, Y281A, C282A, G283A, E284A, G285A, T294A, T300A, N310A, G330A, T331A, I332A, R333A, K334A, N335A, R336A, G337A, I338A, P339A, I345V, T375G, D416A, R427H, D475G, M481V, P491Q, A520T, and A561T, wherein the positions are corresponding to positions of SEQ ID NO: 11. In embodiments, the mobile element enzyme is an engineered mammalian mobile element enzyme. In embodiments, the mobile element enzyme is a mammal-derived, helper RNA mobile element enzyme. In embodiments, the mobile element enzyme is a mammal-derived, helper DNA mobile element enzyme. In embodiments, the enzyme is capable of inserting a donor DNA at a TA dinucleotide site. In embodiments, the enzyme is capable of inserting a donor DNA at a TTAA (SEQ ID NO: 440) tetranucleotide site. In embodiments, the mobile element enzyme has gene cleavage activity (Exc+) and/or gene integration activity (Int+), and the mobile element enzyme having at least about 90% identity to the amino acid sequence of SEQ ID NO: 1, SEQ ID NO: 3, SEQ ID NO: 4, SEQ ID NO: 6, SEQ ID NO: 7, SEQ ID NO: 8, SEQ ID NO: 9, SEQ ID NO: 10, SEQ ID NO: 11 or SEQ ID NO: 430, or a nucleotide sequence encoding the same. SEQ ID NO: 3: PGBD4 Amino Acid Sequence (585 Amino Acids). MSNPRKRSIP MRDSNTGLEQ LLAEDSFDES DFSEIDDSDN FSDSALEADK 50 IRPLSHLESD GKSSTSSDSG RSMKWSARAM IPRQRYDFTG TPGRKVDVSD 100 ITDPLQYFEL FFTEELVSKI TRETNAQAAL LASKPPGPKG FSRMDKWKDT 150 DNDELKVFFA VMLLQGIVQK PELEMFWSTR PLLDTPYLRQ IMTGERFLLL 200 FRCLHFVNNS SISAGQSKAQ ISLQKIKPVF DFLVNKFSTV YTPNRNIAVD 250 ESLMLFKGPL AMKQYLPTKR VRFGLKLYVL CESQSGYVWN ALVHTGPGMN 300 LKDSADGLKS SRIVLTLVND LLGQGYCVFL DNFNISPMLF RELHQNRTDA 350 VGTARLNRKQ IPNDLKKRIA KGTTVARFCG ELMALKWCDG KEVTMLSTFH 400 NDTVIEVNNR NGKKTKRPRV IVDYNENMGA VDSADQMLTS YPSERKRHKV 450 WYKKFFHHLL HITVLNSYIL FKKDNPEHTM SHINFRLALI ERMLEKHHKP 500 GQQHLRGRPC SDDVTPLRLS GRHFPKSIPA TSGKQNPTGR CKICCSQYDK 550 DGKKIRKETR YFCAECDVPL CVVPCFEIYH TKKNY 585 SEQ ID NO: 4: PGBD4 Hyperactive Mutant (S8P, G17R, K134K) Amino Acid Sequence (585 Amino Acids). MSNPRKRPIP MRDSNTRLEQ LLAEDSFDES DFSEIDDSDN FSDSALEADK 50 IRPLSHLESD GKSSTSSDSG RSMKWSARAM IPRQRYDFTG TPGRKVDVSD 100 ITDPLQYFEL FFTEELVSKI TRETNAQAAL LASKPPGPKG FSRMDKWKDT 150 DNDELKVFFA VMLLQGIVQK PELEMFWSTR PLLDTPYLRQ IMTGERFLLL 200 FRCLHFVNNS SISAGQSKAQ ISLQKIKPVF DFLVNKFSTV YTPNRNIAVD 250 ESLMLFKGPL AMKQYLPTKR VRFGLKLYVL CESQSGYVWN ALVHTGPGMN 300 LKDSADGLKS SRIVLTLVND LLGQGYCVFL DNFNISPMLF RELHQNRTDA 350 VGTARLNRKQ IPNDLKKRIA KGTTVARFCG ELMALKWCDG KEVTMLSTFH 400 NDTVIEVNNR NGKKTKRPRV IVDYNENMGA VDSADQMLTS YPSERKRHKV 450 WYKKFFHHLL HITVLNSYIL FKKDNPEHTM SHINFRLALI ERMLEKHHKP 500 GQQHLRGRPC SDDVTPLRLS GRHFPKSIPA TSGKQNPTGR CKICCSQYDK 550 DGKKIRKETR YFCAECDVPL CVVPCFEIYH TKKNY 585 SEQ ID NO: 6: PGBD1 Amino Acid Sequence (809 Amino Acids). MYEALPGPAP ENEDGLVKVK EEDPTWEQVC NSQEGSSHTQ EICRLRFRHF CYQEAHGPQE 60 ALAQLRELCH QWLRPEMHTK EQIMELLVLE QFLTILPKEL QPCVKTYPLE SGEEAVTVLE 120 NLETGSGDTG QQASVYIQGQ DMHPMVAEYQ GVSLECQSLQ LLPGITTLKC EPPQRPQGNP 180 QEVSGPVPHG SAHLQEKNPR DKAVVPVFNP VRSQTLVKTE EETAQAVAAE KWSHLSLTRR 240 NLCGNSAQET VMSLSPMTEE IVTKDRLFKA KQETSEEMEQ SGEASGKPNR ECAPQIPCST 300 PIATERTVAH LNTLKDRHPG DLWARMHISS LEYAAGDITR KGRKKDKARV SELLQGLSFS 360 GDSDVEKDNE PEIQPAQKKL KVSCFPEKSW TKRDIKPNFP SWSALDSGLL NLKSEKLNPV 420 ELFELFFDDE TFNLIVNETN NYASQKNVSL EVTVQEMRCV FGVLLLSGFM RHPRREMYWE 480 VSDTDQNLVR DAIRRDRFEL IFSNLHFADN GHLDQKDKFT KLRPLIKQMN KNFLLYAPLE 540 EYYCFDKSMC ECFDSDQFLN GKPIRIGYKI WCGTTTQGYL VWFEPYQEES TMKVDEDPDL 600 GLGGNLVMNF ADVLLERGQY PYHLCFDSFF TSVKLLSALK KKGVRATGTI RENRTEKCPL 660 MNVEHMKKMK RGYFDFRIEE NNEIILCRWY GDGIISLCSN AVGIEPVNEV SCCDADNEEI 720 PQISQPSIVK VYDECKEGVA KMDQIISKYR VRIRSKKWYS ILVSYMIDVA MNNAWQLHRA 780 CNPGASLDPL DFRRFVAHFY LEHNAHLSD 809 SEQ ID NO: 7: PGBD2 Amino Acid Sequence (592 Amino Acids). MASTSRDVIA GRGIHSKVKS AKLLEVLNAM EEEESNNNRE EIFIAPPDNA AGEFTDEDSG 60 DEDSQRGAHL PGSVLHASVL CEDSGTGEDN DDLELQPAKK RQKAVVKPQR IWTKRDIRPD 120 FGSWTASDPH IEDLKSQELS PVGLFELFFD EGTINFIVNE TNRYAWQKNV NLSLTAQELK 180 CVLGILILSG YISYPRRRMF WETSPDSHHH LVADAIRRDR FELIFSYLHF ADNNELDASD 240 RFAKVRPLII RMNCNFQKHA PLEEFYSFGE SMCEYFGHRG SKQLHRGKPV RLGYKIWCGT 300 TSRGYLVWFE PSQGTLFTKP DRSLDLGGSM VIKFVDALQE RGFLPYHIFF DKVFTSVKLM 360 SILRKKGVKA TGTVREYRTE RCPLKDPKEL KKMKRGSFDY KVDESEEIIV CRWHDSSVVN 420 ICSNAVGIEP VRLTSRHSGA AKTRTQVHQP SLVKLYQEKV GGVGRMDQNI AKYKVKIRGM 480 KWYSSFIGYV IDAALNNAWQ LHRICCQDAQ VDLLAFRRYI ACVYLESNAD TTSQGRRSRR 540 LETESRFDMI GHWIIHQDKR TRCALCHSQT NTRCEKCQKG VHAKCFREYH IR 592 SEQ ID NO: 8: PGBD3 Amino Acid Sequence (593 Amino Acids). MPRTLSLHEI TDLLETDDSI EASAIVIQPP ENATAPVSDE ESGDEEGGTI NNLPGSLLHT 60 AAYLIQDGSD AESDSDDPSY APKDDSPDEV PSTFTVQQPP PSRRRKMTKI LCKWKKADLT 120 VQPVAGRVTA PPNDFFTVMR TPTEILELFL DDEVIELIVK YSNLYACSKG VHLGLTSSEF 180 KCFLGIIFLS GYVSVPRRRM FWEQRTDVHN VLVSAAMRRD RFETIFSNLH VADNANLDPV 240 DKFSKLRPLI SKLNERCMKF VPNETYFSFD EFMVPYFGRH GCKQFIRGKP IRFGYKFWCG 300 ATCLGYICWF QPYQGKNPNT KHEEYGVGAS LVLQFSEALT EAHPGQYHFV FNNFFTSIAL 360 LDKLSSMGHQ ATGTVRKDHI DRVPLESDVA LKKKERGTFD YRIDGKGNIV CRWNDNSVVT 420 VASSGAGIHP LCLVSRYSQK LKKKIQVQQP NMIKVYNQFM GGVDRADENI DKYRASIRGK 480 KWYSSPLLFC FELVLQNAWQ LHKTYDEKPV DFLEFRRRVV CHYLETHGHP PEPGQKGRPQ 540 KRNIDSRYDG INHVIVKQGK QTRCAECHKN TTFRCEKCDV ALHVKCSVEY HTE 593 SEQ ID NO: 9: PGBD5 Amino Acid Sequence (524 Amino Acids). MAEGGGGARR RAPALLEAAR ARYESLHISD DVFGESGPDS GGNPFYSTSA ASRSSSAASS 60 DDEREPPGPP GAAPPPPRAP DAQEPEEDEA GAGWSAALRD RPPPRFEDTG GPTRKMPPSA 120 SAVDFFQLFV PDNVLKNMVV QTNMYAKKFQ ERFGSDGAWV EVTLTEMKAF LGYMISTSIS 180 HCESVLSIWS GGFYSNRSLA LVMSQARFEK ILKYFHVVAF RSSQTTHGLY KVQPFLDSLQ 240 NSFDSAFRPS QTQVLHEPLI DEDPVFIATC TERELRKRKK RKFSLWVRQC SSTGFIIQIY 300 VHLKEGGGPD GLDALKNKPQ LHSMVARSLC RNAAGKNYII FTGPSITSLT LFEEFEKQGI 360 YCCGLLRARK SDCTGLPLSM LTNPATPPAR GQYQIKMKGN MSLICWYNKG HFRFLTNAYS 420 PVQQGVIIKR KSGEIPCPLA VEAFAAHLSY ICRYDDKYSK YFISHKPNKT WQQVFWFAIS 480 IAINNAYILY KMSDAYHVKR YSRAQFGERL VRELLGLEDA SPTH 524 SEQ ID NO: 10: Myotis lucifugus (Wild-type) Amino Acid Sequence with Hyperactive Mutations (S8P, C13R, N125K) (572 Amino Acids). MSQHSDYPDD EFRADKLSNY SCDSDLENAS TSDEDSSDDE VMVRPRTLRR RRISSSSSDS 60 ESDIEGGREE WSHVDNPPVL EDFLGHQGLN TDAVINNIED AVKLFIGDDF FEFLVEESNR 120 YYNQKRNNFK LSKKSLKWKD ITPQEMKKFL GLIVLMGQVR KDRRDDYWTT EPWTETPYFG 180 KTMTRDRFRQ IWKAWHFNNN ADIVNESDRL CKVRPVLDYF VPKFINIYKP HQQLSLDEGI 240 VPWRGRLFFR VYNAGKIVKY GILVRLLCES DTGYICNMEI YCGEGKRLLE TIQTVVSPYT 300 DSWYHIYMDN YYNSVANCEA LMKNKFRICG TIRKNRGIPK DFQTISLKKG ETKFIRKNDI 360 LLQVWQSKKP VYLISSIHSA EMEESQNIDR TSKKKIVKPN ALIDYNKHMK GVDRADQYLS 420 YYSILRRTVK WTKRLAMYMI NCALFNSYAV YKSVRQRKMG FKMFLKQTAI HWLTDDIPED 480 MDIVPDLQPV PSTSGMRAKP PTSDPPCRLS MDMRKHTLQA IVGSGKKKNI LRRCRVCSVH 540 KLRSETRYMC KFCNIPLHKG ACFEKYHTLK NY 572 SEQ ID NO: 11: Myotis lucifugus Corrected Amino Acid Sequence with Hyperactive Mutations (S8P, C13R) (571 Amino Acids). MAQHSDYPDDEFRADKLSNYSCDSDLENASTSDEDSSDDEVMVRPRTLRRRRISSSSSDSESDIEGGREEWSHVDNPPVLEDFLGH QGLNTDAVINNIEDAVKLFIGDDFFEFLVEESNRYYNQNRNNFKLSKKSLKWKDITPQEMKKFLGLIVLMGQVRKDRRDDYWTTEP WTETPYFGKTMTRDRFRQIWKAWHFNNNADIVNESDRLCKVRPVLDYFVPKFINIYKPHQQLSLDEGIVPWRGRLFFRVYNAGKIV KYGILVRLLCESDTGYICNMEIYCGEGKRLLETIQTVVSPYTDSWYHIYMDNYYNSVANCEALMKNKFRICGTIRKNRGIPKDFQT ISLKKGETKFIRKNDILLQVWQSKKPVYLISSIHSAEMEESQNIDRTSKKKIVKPNALIDYNKHMKGVDRADQYLSYYSILRRTVK WTKRLAMYMINCALFNSYAVYKSVRQRKMGFKMFLKQTAIHWLTDDIPEDMDIVPDLQPVPSTSGMRAKPPTSDPPCRLSMDMRKH TLQAIVGSGKKKNILRRCRVCSVHKLRSETRYMCKFCNIPLHKGACFEKYHTLKNY SEQ ID NO: 430: Extended Pteropus vampyrus Amino Acid Sequence 584 Amino Acids. MSNPRKRSIP TCDVNFVLEQ LLAEDSFDES DFSEIDDSDD FSDSASEDYT VRPPSDSESD 60 GNSPTSADSG RALKWSTRVM IPRQRYDFTG TPGRKVDVSD TTDPLQYFEL FFTEELVSKI 120 TSEMNAQAAL LASKPPGPKG FSRMDKWKDT DNDELKVFFA VMLLQGIVQK PELEMFWSTR 180 PLLDIPYLRQ IMTGERFLLL LRCLHFVNNS SISAGQSKAQ ISLQKIKPVF DFLVNKFSTV 240 YTPNRNIAVD ESLMLFKGRL AMKQYIPTKC ARFGLKLYVL CESQSGYVWN ALVHTGPSMN 300 LKDSADGLKS SCIVLTLVND LLGQGYCVFL NNFYTSPMLF RELHQNRTDA VGTARLNRKQ 360 MPNDLKKRIA KGTTVARFCG ELMALKWCDK KEVTMLSTFH NDTVIEVDNR NGKKTKKPCV 420 IVDYNENMGA VDSADQMLTS YPTERKRHKF WYKKFFRHLL NITVLNSYIL FKKDNPEHTI 480 SHVNFRLTLI ERMLEKHHKP GQQRLRGRPC SDDVTPLRLS GRHFPKSIPP TSGKQNPTGR 540 CKVCCSHDKD GKKIRRETLY FCAECDVPLC VVPCFEIYHT KKNY In embodiments, the mobile element enzyme has at least about 95%, or at least about 96%, at least about 97%, at least about 98%, at least about 99% identity to the amino acid sequence of SEQ ID NO: 1, SEQ ID NO: 3, SEQ ID NO: 4, SEQ ID NO: 6, SEQ ID NO: 7, SEQ ID NO: 8, SEQ ID NO: 9, SEQ ID NO: 10, SEQ ID NO: 11 or SEQ ID NO: 430. In embodiments, the mobile element enzyme has one or more mutations which confer hyperactivity. In embodiments, the mobile element enzyme has an amino acid sequence of SEQ ID NO: 1, SEQ ID NO: 10, or SEQ ID NO: 11 or a functional equivalent thereof. In embodiments, the mobile element enzyme has the nucleotide sequence having at least about 90% identity to SEQ ID NO: 5 or a codon-optimized form thereof. SEQ ID NO: 5: PGBD4 Hyperactive Mutant (S8P, G17R, K134K) Nucleotide Sequence (1758 bp). ATGTCAAATC CTAGAAAACG TCCCATTCCT ATGCGTGATA GTAATACCCG TCTCGAACAG 60 TTGTTGGCTG AAGATTCATT TGATGAATCT GATTTTTCGG AAATAGATGA TTCTGATAAT 120 TTTTCGGATA GTGCTTTAGA AGCCGATAAG ATCAGGCCTC TGTCCCATTT AGAATCTGAT 180 GGAAAGAGCT CTACATCAAG TGACTCAGGG CGCTCCATGA AATGGTCAGC TCGTGCTATG 240 ATTCCACGTC AAAGGTATGA CTTTACCGGC ACACCTGGCA GAAAAGTCGA TGTCAGTGAT 300 ATCACTGACC CATTGCAGTA TTTTGAACTG TTCTTTACTG AGGAATTAGT TTCAAAAATT 360 ACTAGAGAAA CAAATGCCCA AGCTGCCTTG TTGGCTTCAA AGCCACCGGG TCCGAAAGGA 420 TTTTCGCGAA TGGATAAATG GAAAGACACT GACAATGACG AGCTCAAAGT CTTTTTTGCA 480 GTAATGTTAC TGCAAGGTAT TGTGCAGAAA CCTGAGCTGG AGATGTTTTG GTCAACAAGG 540 CCTCTTTTGG ATACACCTTA TCTCAGGCAA ATTATGACTG GTGAAAGATT TTTACTTTTG 600 TTTCGGTGCC TGCATTTTGT CAACAATTCT TCTATATCTG CTGGTCAATC AAAGGCCCAG 660 ATTTCATTGC AGAAGATCAA ACCTGTGTTC GACTTTCTTG TAAATAAATT TTCCACTGTA 720 TATACTCCAA ACAGAAACAT TGCAGTTGAT GAATCACTGA TGCTGTTCAA GGGGCCATTA 780 GCTATGAAGC AGTACCTCCC GACAAAACGA GTACGATTTG GTCTGAAGCT ATATGTACTT 840 TGTGAAAGTC AGTCTGGTTA TGTGTGGAAT GCGCTTGTTC ACACAGGGCC TGGCATGAAT 900 TTGAAAGATT CAGCGGATGG CCTGAAATCA TCACGCATTG TTCTTACCTT GGTCAATGAC 960 CTTCTTGGCC AAGGGTATTG TGTCTTCCTC GATAACTTTA ATATATCTCC CATGCTTTTC 1020 AGAGAATTAC ATCAAAATAG GACTGATGCA GTTGGGACAG CTCGTTTGAA CAGAAAACAG 1080 ATTCCAAATG ATCTGAAAAA AAGGATTGCA AAGGGGACGA CTGTAGCCAG ATTCTGTGGT 1140 GAACTTATGG CACTGAAATG GTGTGACGGC AAGGAGGTGA CAATGTTGTC AACATTCCAC 1200 AATGATACTG TGATTGAAGT AAACAATAGA AATGGAAAGA AAACTAAAAG GCCACGTGTC 1260 ATTGTGGATT ATAACGAGAA TATGGGAGCA GTGGACTCGG CTGATCAAAT GCTTACTTCT 1320 TATCCATCTG AGCGCAAAAG ACACAAGGTT TGGTATAAGA AATTCTTTCA CCATCTTCTA 1380 CACATTACAG TGCTGAACTC CTACATCCTG TTCAAGAAGG ATAATCCTGA GCACACGATG 1440 AGCCATATAA ACTTCAGACT GGCATTGATT GAAAGAATGC TGGAAAAGCA TCACAAGCCA 1500 GGGCAGCAAC ATCTTCGAGG TCGTCCTTGC TCCGATGATG TCACACCTCT TCGTCTGTCT 1560 GGAAGACATT TCCCCAAGAG CATACCAGCA ACGTCCGGGA AACAGAATCC AACTGGTCGC 1620 TGCAAAATTT GCTGCTCCCA ATACGACAAG GATGGCAAGA AGATCCGGAA AGAAACGCGC 1680 TATTTTTGTG CCGAATGTGA TGTTCCGCTT TGTGTTGTTC CGTGCTTTGA AATTTACCAC 1740 ACGAAAAAAA ATTATTAA 1758 In embodiments, the mobile element enzyme has an amino acid sequence having S8P and G17R mutations relative to the amino acid sequence of SEQ ID NO: 3 or SEQ ID NO: 4, or a functional equivalent thereof. In embodiments, the mobile element enzyme has an amino acid sequence having I83P and/or V118R mutation relative to the amino acid sequence of SEQ ID NO: 6 or a functional equivalent thereof. In embodiments, the mobile element enzyme has an amino acid sequence having S20P and/or A29R mutation relative to the amino acid sequence of SEQ ID NO: 7 or a functional equivalent thereof. In embodiments, the mobile element enzyme has an amino acid sequence having A12P and/or I28R mutation and/or R152K mutation relative to the amino acid sequence of SEQ ID NO: 9 or a functional equivalent thereof. In embodiments, the mobile element enzyme has an amino acid sequence having T4P and/or L13R mutation relative to the amino acid sequence of SEQ ID NO: 8 or a functional equivalent thereof. In embodiments, the donor DNA is included in a vector comprising left and right end sequences recognized by the mobile element enzyme. In embodiments, the end sequences are selected from MER, MER75A, MER75B, and MER85. In embodiments, the end sequences are selected from nucleotide sequences of SEQ ID NO: 12, SEQ ID NO: 13, SEQ ID NO: 14, SEQ ID NO: 15, SEQ ID NO: 16, SEQ ID NO: 17, SEQ ID NO: 18, SEQ ID NO: 19, SEQ ID NO: 20, SEQ ID NO: 21, SEQ ID NO: 441, and SEQ ID NO: 22, or a nucleotide sequence having at least about 90% identity thereto. SEQ ID NO: 12: Pteropus vampyrus Left End Sequence Sequence 381 bp. TTAACCCATT TCCTGTTTGC CCCGAGAATA CTCACCAGCG GCACTTGCAG CTGCAGCGTT 60 TACCCCGAGA TAACTCGTCG ATTACAGTCC TAACCTTACC CCCAAAGTTT GCCATGAAAT 120 ATCTCGCTTT TATTATTATT TTCGCATCGC TCTAGTATAT CGATAGTCTT TGGAAACAAA 180 TGACATCATT CTATTTACAG CATTCTGTTT TTAGTAGTGG TATTTCCATT TACAAAATAT 240 AGTAATTTTC TATCGCTGAA AATGTCAAAT CCTAGAAAAC GTAGCATTCC TACATGTGAT 300 GTTAACTTCG TTCTCGAACA GTTGTTAGCC GAAGATTCAT TTGATGAATC CGATTTTTCC 360 GAAATAGACG ATTCTGATGA T 381 SEQ ID NO: 13: PGBD4 Left End Nucleotide Sequence Sequence 373 bp. TTAACTCATT TCTCCTTAGC CCCGAGATTA CGCGCTGCTG TGCCTGCGAC TGCAGCGTTT 60 ACGCCGAGAT AACTCGTGGA TTACAGTGCC AACCTTACTC CCAAAGTTTG CCACGAAATA 120 TCTCGCTTCT GTTATTTTCG CATGGTTCTG GTATATTGAC TTTTGAAACA AAAGACATCA 180 TTCTGTTTAT AGCATTCTGT TTTTAGTAGT GGGATTTCCA TCTACAAAAT ATAGTAATTC 240 TCGATCGCTG AAATGTCAAA TCCTAGAAAA CGTAGCATTC CTATGCGTGA TAGTAATACC 300 GGTCTCGAAC AGTTGTTGGC TGAAGATTCA TTTGATGAAT CTGATTTTTC GGAAATAGAT 360 GATTCTGATA ATT 373 SEQ ID NO: 14: MER75 Left End Nucleotide Sequence Sequence 344 bp. TTAACCCTTT TCCCGTTTGC CCCGAGAATA CTCGCCGGCG GCGCTTGCGG CTGCAGCGTT 60 TACCCCGAGA TAACTTTGCC ACGAAATATC TCGCTTTTAT TATTATTTTC GCATCGCTCT 120 AGTATATCGA CTTTGGAAAC AAAAGACATC ATTCTATTTA TAGCATTCTG TTTTTAGTAG 180 TGGTATTTCC ATTTACAAAA TATAGTAATT CTCGATCGCT GAAAATGTCA AATCCTAGAA 240 AACGTAGCAT TCCTACGCGT GATGTTAACA TCGTTCTCGA ACAGTTGTTG GCCGAAGATT 300 CATTTGATGA ATCCGATTTT TCCGAAATAG ACGATTCTGA TGAT 344 SEQ ID NO: 15: MER75B Left End Nucleotide Sequence Sequence 91 bp. TTAACCCATT TCCCGTTTGC CCCGAGAATA CTCTTGTCTC TAATCCTAAT GTAACATCAT 60 ATACATTTCT GTTACATTAG GATTAGAGAC A 91 SEQ ID NO: 16: MER75A Left End Nucleotide Sequence Sequence 32 bp. TTAACCCATT TCCCGTTTGC CCCGAGAATA CT 32 SEQ ID NO: 17: Pteropus vampyrus Right End Sequence Sequence 171 bp. TAGGATTAGA GACAAGTTCT GTTTAGAAAT AACTCCAAGA ACAGTTTTTA TATTTTATTT 60 TCACATTGAA AACCAGTCAG ATTTGCTTCA GCCTCAAAGA GCATGTTTAT GTAAAATTAA 120 ATTAACGCTG GCAGCGAGCT GCACTTTTTT TCTAAACGGG AAATGGGTTA A 171 SEQ ID NO: 18: PGBD4 Right End Nucleotide Sequences Sequence 176 bp. CCTGGGATTA TAGGCATGAG CCACTGCGCC TAGCACCAAG AACAGTTTTT ATATTTTATT 60 TTCACATTGA AAATCAGTCA GATTTGCTTC AGCCTCAAAG AGGGTGTTTA TGTAAAACTA 120 AATGAGTGCA GGCAGCGAGC TACACTTTTT TTTTTCCTAA ATGGAAAATG GGTTAA 176 SEQ ID NO: 19: MER75 Right End Nucleotide Sequences Sequence 178 bp. TCAGACGATT CTGATGTTAG TTCTGTTTAG AAATAACTCC AAGAACAGTT TTTATATTTT 60 ATTTTCACAT TGAAAATCAG TCAGATTTGC TTCAGCCTCA AAGAGCGTGT TTATGTAAAA 120 TTAAATGAGC GCTGGCAGCG AGCTGCACTT TTTTTTTTCT AAACGGGAAA AGGGTTAA 178 SEQ ID NO: 20: MER75B Right End Nucleotide Sequences Sequence 160 bp. AGTTCTGTTT AGAAATAACT CCAAGAACAG TTTTTATATT TTATTTTCAC ATTGAAAATC 60 AGTCAGATTT GCTTCAGCCT CAAAGAGCGT GTTTATGTAA AATTAAATGA GCGCTGGCAG 120 CGAGCTGCAC TTTTTTTTTT CTAAACGGGA AAAGGGTTAA 160 SEQ ID NO: 21: MER75A Right End Nucleotide Sequences Sequence 46 bp. CGCTGGCAGC GAGCTGCACT TTTTTTCTAA ACGGGAAATG GGTTAA 46 In embodiments, one or more of the end sequences are optionally flanked by a TTAA (SEQ ID NO: 440) sequence. In embodiments, the end sequences include at least one repeat from a nucleotide sequence having at least about 90% identity to the nucleotide sequence of SEQ ID NO: 12, and wherein the at least one repeat from the nucleotide sequence having at least about 90% identity to the nucleotide sequence of SEQ ID NO: 12 is positioned at the 5’ end of the donor DNA. In embodiments, the end sequences include at least one repeat from a nucleotide sequence having at least about 90% identity to the nucleotide sequence of SEQ ID NO: 17, and wherein the at least one repeat from the nucleotide sequence having at least about 90% identity to the nucleotide sequence of SEQ ID NO: 17 is positioned at the 3’ end of the donor DNA. In embodiments, the end sequences are optionally flanked by a TTAA (SEQ ID NO: 440) sequence. In embodiments, the end sequences include at least one repeat from a nucleotide sequence having at least about 90% identity to the nucleotide sequence of SEQ ID NO: 13, and wherein the at least one repeat from the nucleotide sequence having at least about 90% identity to the nucleotide sequence of SEQ ID NO: 13 is positioned at the 5’ end of the donor DNA. In embodiments, the end sequences include at least one repeat from a nucleotide sequence having at least about 90% identity to the nucleotide sequence of SEQ ID NO: 18, and wherein the at least one repeat from the nucleotide sequence having at least about 90% identity to the nucleotide sequence of SEQ ID NO: 18 is positioned at the 3’ end of the donor DNA. In embodiments, the end sequences are optionally flanked by a TTAA (SEQ ID NO: 440) sequence. In embodiments, the end sequences include at least one repeat from a nucleotide sequence having at least about 90% identity to the nucleotide sequence of SEQ ID NO: 14, wherein the at least one repeat from the nucleotide sequence having at least about 90% identity to the nucleotide sequence of SEQ ID NO: 14 is positioned at the 5’ end of the donor DNA. In embodiments, the end sequences include at least one repeat from a nucleotide sequence having at least about 90% identity to the nucleotide sequence of SEQ ID NO: 19, wherein the at least one repeat from the nucleotide sequence having at least about 90% identity to the nucleotide sequence of SEQ ID NO: 19 is positioned at the 3’ end of the donor DNA. In embodiments, the end sequences are optionally flanked by a TTAA (SEQ ID NO: 440) sequence. In embodiments, the end sequences include at least one repeat from a nucleotide sequence having at least about 90% identity to the nucleotide sequence of SEQ ID NO: 15, wherein the at least one repeat from the nucleotide sequence having at least about 90% identity to the nucleotide sequence of SEQ ID NO: 15 is positioned at the 5’ end of the donor DNA. In embodiments, end sequences include at least one repeat from a nucleotide sequence having at least about 90% identity to the nucleotide sequence of SEQ ID NO: 20, wherein the at least one repeat from the nucleotide sequence having at least about 90% identity to the nucleotide sequence of SEQ ID NO: 20 is positioned at the 3’ end of the donor DNA. In embodiments, the end sequences are optionally flanked by a TTAA (SEQ ID NO: 440) sequence. In embodiments, the end sequences include at least one repeat from a nucleotide sequence having at least about 90% identity to the nucleotide sequence of SEQ ID NO: 16, wherein the at least one repeat from the nucleotide sequence having at least about 90% identity to the nucleotide sequence of SEQ ID NO: 16 is positioned at the 5’ end of the donor DNA. In embodiments, the end sequences include at least one repeat from a nucleotide sequence having at least about 90% identity to the nucleotide sequence of SEQ ID NO: 21 or SEQ ID NO: 441, wherein the at least one repeat from the nucleotide sequence having at least about 90% identity to the nucleotide sequence of SEQ ID NO: 21 or SEQ ID NO: 441 is positioned at the 3’ end of the donor DNA. In embodiments, the mobile element enzyme is an engineered form of a mobile element enzyme reconstructed from Homo sapiens or a predecessor thereof. In embodiments, the enzyme is in a monomeric or dimeric form. In embodiments, the enzyme is in a multimeric form. In embodiments, the method of the present disclosure provides an enzyme comprising (a) a targeting element, and (b) an enzyme that is capable of inserting the donor DNA comprising a transgene, optionally at a TA dinucleotide site or a TTAA (SEQ ID NO: 440) tetranucleotide site in a genomic safe harbor site (GSHS). In embodiments, the donor DNA comprises a transgene encoding a complete polypeptide. In embodiments, the donor DNA comprises a transgene which is defective or substantially absent in a disease state. In embodiments, the enzyme has one or more mutations which confer hyperactivity. In embodiments, the enzyme has gene cleavage (Exc) and/or gene integration activity (Int+). In embodiments, the enzyme has gene cleavage (Exc) and/or a lack of gene integration activity (Int-). In embodiments, the mobile element enzyme is a chimeric mobile element enzyme. In embodiments, the targeting element comprises one or more of a gRNA, optionally associated with a Cas enzyme, which is optionally catalytically inactive, transcription activator-like effector (TALE), catalytically inactive Zinc finger, catalytically inactive transcription factor, nickase, a transcriptional activator, a transcriptional repressor, a recombinase, a DNA methyltransferase, a histone methyltransferase, a paternally expressed gene 10 (PEG10), and a TnsD. In embodiments, the targeting element comprises a transcription activator-like effector (TALE) DNA binding domain (DBD). In embodiments, the TALE DBD comprises one or more repeat sequences. In embodiments, the TALE DBD comprises about 14, or about 15, or about, 16, or about 17, or about 18, or about 18.5 repeat sequences. In embodiments, the TALE DBD repeat sequences comprise 33 or 34 amino acids. In embodiments, the one or more of the TALE DBD repeat sequences comprise a repeat variable di-residue (RVD) at residue 12 or 13 of the 33 or 34 amino acids. In embodiments, the RVD recognizes one base pair in the nucleic acid molecule. In embodiments, the RVD recognizes a C residue in the nucleic acid molecule and is selected from HD, N(gap), HA, ND, and HI. In embodiments, the RVD recognizes a G residue in the nucleic acid molecule and is selected from NN, NH, NK, HN, and NA. In embodiments, the RVD recognizes an A residue in the nucleic acid molecule and is selected from NI and NS. In embodiments, the RVD recognizes a T residue in the nucleic acid molecule and is selected from NG, HG, H(gap), and IG. In embodiments, the GSHS is in an open chromatin location in a chromosome. In embodiments, the GSHS is selected from adeno-associated virus site 1 (AAVS1), chemokine (C-C motif) receptor 5 (CCR5) gene, HIV-1 coreceptor, and human Rosa26 locus. In embodiments, the GSHS is located on human chromosome 2, 4, 6, 10, 11, 17, 22, or X. In embodiments, the GSHS is selected from TALC1, TALC2, TALC3, TALC4, TALC5, TALC7, TALC8, AVS1, AVS2, AVS3, ROSA1, ROSA2, TALER1, TALER2, TALER3, TALER4, TALER5, SHCHR2-1, SHCHR2-2, SHCHR2-3, SHCHR2-4, SHCHR4-1, SHCHR4-2, SHCHR4-3, SHCHR6-1, SHCHR6-2, SHCHR6-3, SHCHR6-4, SHCHR10-1, SHCHR10-2, SHCHR10-3, SHCHR10-4, SHCHR10-5, SHCHR11-1, SHCHR11-2, SHCHR11-3, SHCHR17-1, SHCHR17-2, SHCHR17-3, and SHCHR17-4. In embodiments, the targeting element comprises a Cas9 enzyme guide RNA complex. In embodiments, the Cas9 enzyme guide RNA complex comprises a nuclease-deficient dCas9 guide RNA complex. In embodiments, the targeting element comprises a Cas12 enzyme guide RNA complex or wherein the targeting element comprises a nuclease- deficient dCas12 guide RNA complex, optionally dCas12j guide RNA complex or dCas12a guide RNA complex. In embodiments, the targeting element comprises: a gRNA of or comprising a sequence of TABLE 3A-3F, or a variant thereof; or a TALE DBD of or comprising a sequence of TABLE 4A-4F, or a variant thereof; or a ZNF of or comprising a sequence of TABLE 5A-5E, or a variant thereof. In embodiments, the targeting element is or comprises a nucleic acid binding component of the gene-editing system. In embodiments, the enzyme and the targeting element are connected. In embodiments, the enzyme and the targeting element are fused to one another or linked via a linker to one another. In embodiments, the linker is a flexible linker. In embodiments, the flexible linker is substantially comprised of glycine and serine residues, optionally wherein the flexible linker comprises (Gly4Ser)n, where n is from about 1 to about 12. In embodiments, the flexible linker is of about 20, or about 30, or about 40, or about 50, or about 60 amino acid residues. In embodiments, the donor DNA comprises a gene encoding a complete polypeptide. In embodiments, the donor DNA comprises a gene which is defective or substantially absent in a disease state. In embodiments, the donor DNA is flanked by one or more inverted terminal ends. In embodiments, at least one of the first nucleic acid and the second nucleic acid is in the form of a lipid nanoparticle (LNP). In embodiments, the first nucleic acid encoding the enzyme and the second nucleic acid encoding the donor DNA are in the form of the same LNP, optionally in a co-formulation. In embodiments, the LNP comprises one or more lipids selected from 1,2-dioleoyl-3-trimethylammonium propane (DOTAP), a cationic cholesterol derivative mixed with dimethylaminoethane-carbamoyl (DC-Chol), phosphatidylcholine (PC), triolein (glyceryl trioleate), and 1,2-distearoyl- sn-glycero-3-phosphoethanolamine-N-[carboxy(polyethylene glycol)-2000] (DSPE-PEG), 1,2-dimyristoyl-rac-glycero- 3-methoxypolyethyleneglycol – 2000 (DMG-PEG 2K), and 1,2 distearol -sn-glycerol-3phosphocholine (DSPC) and/or comprising of one or more molecules selected from polyethylenimine (PEI) and poly(lactic-co-glycolic acid) (PLGA), and N-Acetylgalactosamine (GalNAc). In embodiments, the enzyme is encoded by a recombinant or synthetic nucleic acid. In embodiments, the nucleic acid is mRNA or a helper RNA. In embodiments, the nucleic acid is RNA that has a 5’-m7G cap (cap0, cap1, or cap2) with pseudouridinesubstitution, and a poly-A tail of about 30, or about 50, or about 100, of about 150 nucleotides in length. In embodiments, the enzyme is incorporated into a vector or a vector-like particle. In embodiments, the vector is a non- viral vector. In embodiments, the enzyme and the donor DNA are included in the same vector. In embodiments, the enzyme and the donor DNA are included in different vectors. In embodiments, the enzyme and the donor DNA are included in a single pharmaceutical composition. In embodiments, the enzyme and the donor DNA are included in different pharmaceutical compositions. In embodiments, the enzyme and the donor DNA are co- administered. In embodiments, the enzyme and the donor DNA are administered separately. In embodiments, the present disclosure provides a method of producing an AAV bearing a gene of interest, comprising employing a method of the present disclosure to produce the AAV bearing the gene of interest. In embodiments, the present disclosure provides a cell for gene therapy, generated by a method of the present disclosure. In embodiments, the present disclosure provides a method of delivering a cell therapy, comprising administering to a patient in need thereof the transfected cell generated by a method of the present disclosure. In embodiments, the present disclosure provides a method of treating a disease or condition using a cell therapy, comprising administering to a patient in need thereof the transfected cell generated by a method of the present disclosure. In embodiments, the present disclosure provides a method of treating a disease or condition using a biologic, e.g., antibody, therapy, comprising administering to a patient in need thereof the transfected cell generated by a method of the present disclosure. Enzymes In embodiments, the enzyme capable of targeted genomic integration is any type of an enzyme that cause a transgene to be inserted from one location (e.g., without limitation, donor DNA) to a specific site and/or locus in a subject’s genome. In embodiments, the enzyme capable of targeted genomic integration is a recombinase. In embodiments, the recombinase is an integrase. In embodiments, the enzyme is a mobile element enzyme. In embodiments, the recombinase is an integrase or a mobile element enzyme. In embodiments, the mobile element enzyme is an engineered mammalian mobile element enzyme. In embodiments, the mobile element enzyme is a mammal-derived, helper RNA mobile element enzyme. Messenger RNA (mRNA) is an effective alternative to DNA as a source of a mobile element enzyme for targeting somatic cells and tissues, given that RNA is a safer alternative to DNA as a source of a mobile element enzyme for somatic gene therapy applications. See, e.g., Wilber et al., Mol. Ther.13, 625–630 (2006). Successful use of in vitro-transcribed mRNA as a transient source of mobile element enzyme and subsequent transposition in cultured human cells and in live mice was previously reported for Sleeping Beauty mobile element enzyme. See Wilber et al., Mol. Ther. 13, 625–630 (2006). It was demonstrated that in vitro-transcribed, UTR-stabilized mobile element enzyme-encoding mRNA can be used as a source of mobile element enzyme for Sleeping Beauty-mediated transposition in cultured somatic cells. Wilber et al., Mol. Ther.13, 625–630 (2006). Also, Hoerr et al. reported that a specific cytotoxic T cell response and circulating antigen-specific antibodies were detected after administration of in vitro-transcribed, UTR-stabilized, and protamine- condensed bacterial lacZ mRNA into the ear pinna of Balb/C mice. Hoerr et al., Eur. J. Immunol.2000; 30: 1-7; see also Wilber et al. (2006). In embodiments, the mobile element enzyme is a mammal-derived, DNA mobile element enzyme. In embodiments, the mobile element enzyme is a chimeric mobile element enzyme. In embodiments, the enzyme capable of targeted genomic integration is a mobile element enzyme, and the mobile element enzyme comprises (a) a targeting element that is or comprises a gene-editing system, and (b) a mobile element enzyme that is capable of inserting the donor DNA (e.g., mobile element) comprising a transgene at a TA dinucleotide site or a TTAA (SEQ ID NO: 440) tetranucleotide site in a GSHS, as described elsewhere herein. In embodiments, the enzyme is derived from Bombyx mori, Xenopus tropicalis, Trichoplusia ni, Myotis lucifugus, Rhinolophus ferrumequinum, Rousettus aegyptiacus, Phyllostomus discolor, Myotis myotis, Pteropus vampyrus, Pipistrellus kuhlii, Molossus molossus, Pan troglodytes, or Homo sapiens. In embodiments, the enzyme is an engineered version, including but not limited to hyperactive forms, of an enzyme derived from Bombyx mori, Xenopus tropicalis, Trichoplusia ni, Myotis lucifugus, Rhinolophus ferrumequinum, Rousettus aegyptiacus, Phyllostomus discolor, Myotis myotis, Pteropus vampyrus, Pipistrellus kuhlii, Molossus molossus, Pan troglodytes, or Homo sapiens. In embodiments, the enzyme is a mobile element enzyme derived from Bombyx mori, Xenopus tropicalis, Trichoplusia ni, Myotis lucifugus, Rhinolophus ferrumequinum, Rousettus aegyptiacus, Phyllostomus discolor, Myotis myotis, Pteropus vampyrus, Pipistrellus kuhlii, Molossus molossus, Pan troglodytes, or Homo sapiens. In embodiments, the enzyme is an engineered version, including but not limited to hyperactive forms, of a mobile element enzyme derived from Bombyx mori, Xenopus tropicalis, Trichoplusia ni, Myotis lucifugus, Rhinolophus ferrumequinum, Rousettus aegyptiacus, Phyllostomus discolor, Myotis myotis, Pteropus vampyrus, Pipistrellus kuhlii, Molossus molossus, Pan troglodytes, or Homo sapiens. In embodiments, the mobile element enzyme is from one or more of the Tn1, Tn2, Tn3, Tn5, Tn7, Tn9, Tn10, Tn552, Tn903, Tn1000/Gamma-delta, Tn/O, tnsA, tnsB, tnsC, tniQ, IS10, ISS, IS911, Minos, Sleeping beauty, piggyBac, Tol2, Mos1, Himar1, Hermes, Tol2, Minos, Tel, P-element, MuA, Ty1, Chapaev, transib, Tc1/mariner, or Tc3 donor DNA system, or biologically active fragments variants thereof, inclusive of hyperactive mutants (e.g., without limitation selected from TABLE 1, or equivalents thereof). In embodiments, the mobile element enzyme is from a MLT donor DNA system that is based on a cut-and-paste MLT element obtained from the little brown bat (Myotis lucifugus) or other bat mobile element enzymes, such as Rhinolophus ferrumequinum, Rousettus aegyptiacus, Phyllostomus discolor, Myotis myotis, Pipistrellus kuhlii, and Molossus molossus. See Mitra et al., Proc Natl Acad Sci U S A.2013 Jan 2;110(1):234-9; Jebb et al., Nature, volume 583, pages 578–584 (2020), which are incorporated by reference herein in their entireties. In embodiments, hyperactive forms of a bat mobile element enzyme are used. The MLT mobile element enzyme has been shown to be capable of transposition in bat, human, mammalian, and yeast cells. The hyperactive forms of the MLT mobile element enzyme enhance the transposition process. In addition, chimeric MLT mobile element enzymes are capable of site-specific excision without genomic integration. In embodiments, the mobile element enzyme is a Myotis lucifugus mobile element enzyme (MLT), which is either the wild type, monomer, dimer, tetramer (or another multimer), hyperactive, an Int-mutant, or of any other form. In embodiments, the MLT mobile element enzyme has an amino acid sequence of SEQ ID NO: 1, or a variant having at least about 80%, at least about 90%, or at least about 93%, or at least about 95%, or at least about 97%, or at least about 98%, or at least about 99% identity thereto, and one or more mutations selected from L573X, E574X, and S2X, wherein X is any amino acid or no amino acid, optionally X is A, G, or a deletion, optionally the mutations are L573del E574del, and S2A). In embodiments, the MLT mobile element enzyme has the nucleotide sequence of SEQ ID NO: 2 (which is a codon-optimized form of MLT), or a nucleotide sequence having at least about 90%, or at least about 93%, or at least about 95%, or at least about 97%, or at least about 98%, or at least about 99% identity thereto. SEQ ID NO: 1 is: MAQHSDYSDDEFCADKLSNYSCDSDLENASTSDEDSSDDEVMVRPRTLRRRRISSSSSDSESDIEGGREEWSHV DNPPVLEDFLGHQGLNTDAVINNIEDAVKLFIGDDFFEFLVEESNRYYNQNRNNFKLSKKSLKWKDITPQEMKK FLGLIVLMGQVRKDRRDDYWTTEPWTETPYFGKTMTRDRFRQIWKAWHFNNNADIVNESDRLCKVRPVLDYFVP KFINIYKPHQQLSLDEGIVPWRGRLFFRVYNAGKIVKYGILVRLLCESDTGYICNMEIYCGEGKRLLETIQTVV SPYTDSWYHIYMDNYYNSVANCEALMKNKFRICGTIRKNRGIPKDFQTISLKKGETKFIRKNDILLQVWQSKKP VYLISSIHSAEMEESQNIDRTSKKKIVKPNALIDYNKHMKGVDRADQYLSYYSILRRTVKWTKRLAMYMINCAL FNSYAVYKSVRQRKMGFKMFLKQTAIHWLTDDIPEDMDIVPDLQPVPSTSGMRAKPPTSDPPCRLSMDMRKHTL QAIVGSGKKKNILRRCRVCSVHKLRSETRYMCKFCNIPLHKGACFEKYHTLKNY In embodiments, the MLT mobile element enzyme has an amino acid sequence of SEQ ID NO: 1 or a variant having at least about 80%, at least about 90%, or at least about 93%, or at least about 95%, or at least about 97%, or at least about 98%, or at least about 99% identity thereto and comprises an amino acid other than serine at the position corresponding to position 2 of SEQ ID NO: 1. In embodiments, the amino acid is a non-polar aliphatic amino acid, optionally a non-polar aliphatic amino acid optionally selected from G, A, V, L, I and P, optionally A. In embodiments, the mobile element enzyme does not have additional residues at the C terminus relative to SEQ ID NO: 1. In embodiments, the MLT mobile element enzyme has an amino acid sequence of SEQ ID NO: 1 or a variant having at least about 80%, at least about 90%, or at least about 93%, or at least about 95%, or at least about 97%, or at least about 98%, or at least about 99% identity thereto and comprises an alanine at the position corresponding to position 2 of SEQ ID NO: 1. In embodiments, the MLT mobile element enzyme has an amino acid sequence of SEQ ID NO: 1 or a variant having at least about 80%, at least about 90%, or at least about 93%, or at least about 95%, or at least about 97%, or at least about 98%, or at least about 99% identity thereto and comprises an alanine at the position corresponding to position 2 of SEQ ID NO: 1 and no additional amino acids at the C terminal end. In embodiments, the MLT mobile element enzyme has a nucleotide sequence of SEQ ID NO: 2 (which is codon- optimized) and an amino acid sequence SEQ ID NO: 1, respectively. In embodiments, the MLT mobile element enzyme has a nucleotide sequence of SEQ ID NO: 2, or a nucleotide sequence having at least about 90%, or at least about 93%, or at least about 95%, or at least about 97%, or at least about 98%, or at least about 99% identity thereto, or a codon-optimized form thereof. In embodiments, the MLT mobile element enzyme has an amino acid sequence SEQ ID NO: 1, or an amino acid sequence having at least about 90%, or at least about 93%, or at least about 95%, or at least about 97%, or at least about 98%, or at least about 99% identity thereto. In embodiments, the mobile element enzyme can act on an MLT left terminal end, or a sequence having at least about 90%, or at least about 93%, or at least about 95%, or at least about 97%, or at least about 98%, or at least about 99% identity thereto, wherein the nucleotide sequence of the MLT left terminal end (5’ to 3’) is as follows: ttaacacttggattgcgggaaacgagttaagtcggctcgcgtgaattgcgcgtactccgcgggagccgtcttaactcggttcatatag atttgcggtggagtgcgggaaacgtgtaaactcgggccgattgtaactgcgtattaccaaatatttgtt (SEQ ID NO: 21) In embodiments, the mobile element enzyme can act on an MLT right terminal end, or a sequence having at least about 90%, or at least about 93%, or at least about 95%, or at least about 97%, or at least about 98%, or at least about 99% identity thereto, wherein the nucleotide sequence of the MLT right terminal end (5’ to 3’) is as follows: aattatttatgtactgaatagataaaaaaatgtctgtgattgaataaattttcattttttacacaagaaaccgaaaatttcatttcaatcgaa cccatacttcaaaagatataggcattttaaactaactctgattttgcgcgggaaacctaaataattgcccgcgccatcttatattttggcg ggaaattcacccgacaccgtAgtgttaa (SEQ ID NO: 22) In embodiments, the donor DNA is flanked by one or more terminal ends. In embodiments, the donor DNA is or comprises a gene encoding a compete polypeptide. In embodiments, the donor DNA is or comprises a gene which is defective or substantially absent in a disease state. In embodiments, the enzyme (e.g., without limitation, a mobile element enzyme, e.g., without limitation, MLT mobile element enzyme), inclusive of any described herein has one or more mutations which confer hyperactivity. In embodiments, the enzyme (e.g., without limitation, a mobile element enzyme, e.g., without limitation, MLT mobile element enzyme) has gene cleavage activity (Exc+) and/or gene integration activity (Int+). In embodiments, the enzyme (e.g., without limitation, a mobile element enzyme, e.g., without limitation, MLT mobile element enzyme) has gene cleavage activity (Exc+) and/or a lack of gene integration activity (Int-). In embodiments, the mobile element enzyme, e.g., without limitation, MLT mobile element enzyme includes a hyperactive mutation, e.g., about 1, or about 2, or about 3, or about 4, or about 5 hyperactive mutations or combinations thereof. In embodiments, the mobile element enzyme can include any number of any of the hyperactive mutations, or equivalents thereof, described herein. In embodiments, the MLT mobile element enzyme includes a hyperactive mutation, e.g., about 1, or about 2, or about 3, or about 4, or about 5 hyperactive mutations, or combinations thereof. In embodiments, the mobile element enzyme can include any number of any of the hyperactive mutations, or equivalents thereof, described herein. In embodiments, the enzyme comprises one or more mutations corresponding to TABLE 1, which, without being bound by theory, provides hyperactive mutations. Numbering relative to the amino acid sequence of protein of SEQ ID NO: 1, and nucleic acid sequence of SEQ ID NO: 2. TABLE 1:
Figure imgf000049_0001
In embodiments, the MLT mobile element enzyme has one or more amino acid substitutions selected from S8X1, C13X2 and/or N125X3, or positions corresponding thereto, relative to SEQ ID NO: 1, wherein X1 is selected from G, A, V, L, I and P, X2 is selected from K, R, and H, and X3 is selected from K, R, and H, or wherein: X1 is P, X2 is R, and/or X3 is K. In embodiments, the MLT mobile element enzyme has S8X1, C13X2 and N125X3 substitutions, at positions corresponding to SEQ ID NO: 1, wherein X1 is selected from G, A, V, L, I and P, X2 is selected from K, R, and H, and X3 is selected from K, R, and H, or wherein: X1 is P, X2 is R, and/or X3 is K. In embodiments, the MLT mobile element enzyme has S8X1 and C13X2 substitutions, at positions corresponding to SEQ ID NO: 1, wherein X1 is selected from G, A, V, L, I and P, X2 is selected from K, R, and H, and X3 is selected from K, R, and H, or wherein: X1 is P, X2 is R, and/or X3 is K. In embodiments, the MLT mobile element enzyme has S8X1 and N125X3 substitutions, at positions corresponding to SEQ ID NO: 1, wherein X1 is selected from G, A, V, L, I and P, X2 is selected from K, R, and H, and X3 is selected from K, R, and H, or wherein: X1 is P, X2 is R, and/or X3 is K. In embodiments, the MLT mobile element enzyme has C13X2 and N125X3 substitutions, at positions corresponding to SEQ ID NO: 1, wherein X1 is selected from G, A, V, L, I and P, X2 is selected from K, R, and H, and X3 is selected from K, R, and H, or wherein: X1 is P, X2 is R, and/or X3 is K. In embodiments, the MLT mobile element enzyme has an amino acid sequence of SEQ ID NO: 1, or a variant thereof, and S8P and C13R mutations (SEQ ID NO: 11). In embodiments, the MLT mobile element enzyme has an amino acid sequence having mutations at positions which correspond to at least one of S8P and C13R mutations relative to the amino acid of SEQ ID NO: 1 or a functional equivalent thereof. In embodiments, the MLT mobile element enzyme has an amino acid sequence having mutations at positions which correspond to S8P and C13R mutations relative to the amino acid of SEQ ID NO: 1 or a functional equivalent thereof. In embodiments, the MLT mobile element enzyme has an amino acid sequence of SEQ ID NO: 1, or a variant thereof, and S8P, C13R, and N125K mutations (SEQ ID NO: 10). In embodiments, a MLT mobile element enzyme comprising the amino acid sequence of SEQ ID NO: 1, or a variant thereof, and includes one or more hyperactive mutations selected from a substitution or deletion at one or more of positions S5, S8, D9, D10, E11, C13, A14, S36, S54, N125, K130, G239, T294, T300, I345, R427, D475, M481, P491, A520, and A561, or positions corresponding thereto. In embodiments, a MLT mobile element enzyme comprising the amino acid sequence of SEQ ID NO: 1, or a variant thereof, and includes one or more hyperactive mutations selected from S5P, S8P, S8P/C13R, D9G, D10G, E11G, C13R, A14V, S36G, S54N, N125K, K130T, G239S, T294A, T300A, I345V, R427H, D475G, M481V, P491Q, A520T, and A561T, or positions corresponding thereto. In embodiments, the MLT mobile element enzyme comprises one or more of hyperactive mutants selected from S8X1, C13X2 and/or N125X3 (e.g., all of S8X1, C13X2 and N125X3, S8X1 and C13X2, S8X1 and N125X3, and C13X2 and N125X3), where X1, X2, and X3 is each independently any amino acid, or X1 is a non-polar aliphatic amino acid, selected from G, A, V, L, I and P, X2 is a positively charged amino acid selected from K, R, and H, and/or X3 is a positively charged amino acid selected from K, R, and H. In embodiments, X1 is P, X2 is R, and/or X3 is K. In embodiments, the enzyme (e.g., without limitation, a mobile element enzyme, e.g., without limitation, MLT mobile element enzyme) has gene cleavage activity (Exc+) and/or gene integration activity (Int+). In embodiments, the enzyme (e.g., without limitation, a mobile element enzyme) has gene cleavage activity (Exc+) and/or a lack of gene integration activity (Int-). In embodiments, the MLT mobile element enzyme has gene cleavage activity (Exc+) and/or gene integration activity (Int+). In embodiments, the MLT mobile element enzyme has gene cleavage activity (Exc+) and/or a lack of gene integration activity (Int-). In embodiments, the mobile element enzyme, e.g., without limitation, MLT mobile element enzyme includes an integration reduced or deficient mutation, e.g., about 1, or about 2, or about 3, or about 4, or about 5 integration reduced or deficient mutations or combinations thereof. In embodiments, the mobile element enzyme can include any number of any of the integration reduced or deficient mutations, or equivalents thereof, described herein. In embodiments, the MLT mobile element enzyme includes a integration reduced or deficient mutations, e.g. about 1, or about 2, or about 3, or about 4, or about 5 integration reduced or deficient mutations, or combinations thereof. In embodiments, the mobile element enzyme can include any number of any of the integration reduced or deficient mutations, or equivalents thereof, described herein. In embodiments, the enzyme comprises one or more mutations corresponding to TABLE 2A, or positions corresponding thereto, which, without being bound by theory, provides integration reduced or deficient mutations. Numbering relative to the amino acid sequence of protein of SEQ ID NO: 1. TABLE 2A:
Figure imgf000051_0001
Figure imgf000052_0002
In embodiments, the enzyme comprises one or more mutations corresponding to TABLE 2B, or positions corresponding thereto, which, without being bound by theory, provides excision positive and integration deficient mutations. Numbering relative to the amino acid sequence of protein of SEQ ID NO: 1, and nucleic acid sequence of SEQ ID NO: 2. TABLE 2B:
Figure imgf000052_0001
Figure imgf000053_0001
In embodiments, a MLT mobile element enzyme comprising the amino acid sequence of SEQ ID NO: 1, or a variant thereof, and includes one or more mutations selected from S8P and/or C13R and one of R164N, W168V, M278A, K286A, R287A, R333A, K334A, N335A, K349A, K350A, K368A, K369A, and D416N, or positions corresponding thereto. In embodiments, a MLT mobile element enzyme comprising the amino acid sequence of SEQ ID NO: 1, or a variant thereof, and includes one or more mutations selected from S8P and/or C13R and one of R164N, W168V, M278A, K286A, R287A, R333A, K334A, N335A, K349A, K350A, K368A, K369A, and D416N and/or one or more of E284A, K286A, R287A, N310A, R333A, K334A, R336A, K349A, K350A, K368A, and K369A, or positions corresponding thereto. In embodiments, a MLT mobile element enzyme comprising the amino acid sequence of SEQ ID NO: 1, or a variant thereof, and includes one or more mutations selected from S8P and/or C13R and one of R164N, W168V, M278A, K286A, R287A, R333A, K334A, N335A, K349A, K350A, K368A, K369A, and D416N and/or one or more of E284A, K286A, R287A, N310A, R333A, K334A, R336A, K349A, K350A, K368A, and K369A and/or one R336A, or positions corresponding thereto. In embodiments, the mobile element enzyme is or is derived from any of Bombyx mori, Xenopus tropicalis, Trichoplusia ni, Rhinolophus ferrumequinum, Rousettus aegyptiacus, Phyllostomus discolor, Myotis myotis, Myotis lucifugus, Pipistrellus kuhlii, Pteropus vampyrus, and Molossus molossus. In embodiments, the mobile element enzyme is or is derived from any of Trichoplusia ni (SEQ ID NO: 433), Myotis myotis (SEQ ID NO: 435, SEQ ID NO: 436, SEQ ID NO: 438, or SEQ ID NO: 439), or Pteropus vampyrus (SEQ ID NO: 434). In embodiments, the mobile element enzymes have one or more hyperactive and/or integration deficient mutations selected from TABLE 1, TABLE 2A, and/or TABLE 2B, or equivalents thereof. One skilled in the art can correspond such mutants to mobile element enzymes from any of Trichoplusia ni (SEQ ID NO: 433), Myotis lucifugus (SEQ ID NO: 437), Myotis myotis (SEQ ID NO: 435, SEQ ID NO: 436, SEQ ID NO: 438, or SEQ ID NO: 439), or Pteropus vampyrus (SEQ ID NO: 434), e.g.: Trichnoplusia ni 1 MGSSLDDEHI LSALLQSDDE LVGEDSDSEI SDHVSEDDVQ SDTEEAFIDE VHEVQPTSSG 61 SEILDEQNVI EQPGSSLASN KILTLPQRTI RGKNKHCWST SKSTRRSRVS ALNIVRSQRG 121 PTRMCRNIYD PLLCFKLFFT DEIISEIVKW TNAEISLKRR ESMTGATFRD TNEDEIYAFF 181 GILVMTAVRK DNHMSTDDLF DRSLSMVYVS VMSRDRFDFL IRCLRMDDKS IRPTLRENDV 241 FTPVRKIWDL FIHQCIQNYT PGAHLTIDEQ LLGFRGRCPF RMYIPNKPSK YGIKILMMCD 301 SGTKYMINGM PYLGRGTQTN GVPLGEYYVK ELSKPVRGSC RNITCDNWFT SIPLAKNLLQ 361 EPYKLTIVGT VRSNKREIPE VLKNSRSRPV GTSMFCFDGP LTLVSYKPKP AKMVYLLSSC 421 DEDASINEST GKPQMVMYYN QTKGGVDTLD QMCSVMTCSR KTNRWPMALL YGMINIACIN 481 SFIIYSHNVS SKGEKVQSRK KFMRNLYMSL TSSFMRKRLE APTLKRYLRD NISNILPNEV 541 PGTSDDSTEE PVTKKRTYCT YCPSKIRRKA NASCKKCKKV ICREHNIDMC QSCF (SEQ ID NO: 433) Pteropus vampyrus 1 MSNPRKRSIP TCDVNFVLEQ LLAEDSFDES DFSEIDDSDD FSDSASEDYT VRPPSDSESD 61 GNSPTSADSG RALKWSTRVM IPRQRYDFTG TPGRKVDVSD TTDPLQYFEL FFTEELVSKI 121 TSEMNAQAAL LASKPPGPKG FSRMDKWKDT DNDELKVFFA VMLLQGIVQK PELEMFWSTR 181 PLLDIPYLRQ IMTGERFLLL LRCLHFVNNS SISAGQSKAQ ISLQKIKPVF DFLVNKFSTV 241 YTPNRNIAVD ESLMLFKGRL AMKQYIPTKM NLKDSADGLK (SEQ ID NO: 434) Myotis myotis (“2a”) 1 MDLRCQHTVL SIRESRGLLP NLKMKTSRMK KGDIIFSRKG DILLLAWKDK RVVRMISIHD 61 TSVSTTGKKN RKTGENIVKP ACIKEYNAHM KGVDRADQFL SCCSILRKMM KWTKKVVLYL 121 INCGLFNSFR VYNVLNPQAK MKYKQFLLSV ARDWIMDDNN EGSPEPETNL SSPSPGGARR 181 APRKDPPKRL SGDMKQHEPT CIPASGKKKF PTRACRVCAH GKRSESRYLC KFCLVPLHRG 241 KCFTQYHTLK KY (SEQ ID NO: 435) Myotis myotis (“1”) 1 MKAFLGVILN MGVLNHPNLQ SYWSMDFESH IPFFRSVFKR ERFLQIFWML HLKNDQKSSK 61 DLRTRTEKVN CFLSYLEMKF RERFCPGREI AVDEAVVGFK GKIHFITYNP KKPTKWGIRL 121 YVLSDSKCGY VHSFVPYYGG ITSETLVRPD LPFTSRIVLE LHERLKNSVP GSQGYHFFTD 181 RYYTSVTLAK ELFKEKTHLT GTIMPNRKDN PPVIKHQKLK KGEIVAFRDE NVMLLAWKDK 241 RIVTLSTWDS ETESVERRVG GGKEIVLKPK VVTNYTKFMG GVDIADYTST YCFMRKTLKW 301 WRTLFFWGLE VSVVNSYILY KECQKRKNEK PITHVKFIRK LVHDLVGEFR DGTLTSRGRL 361 LSTNLEQRLD GKLHIITPHP NKKHKDCVVC SNRKIKGGRR ETIYICETCE CKPGLHVGEC 421 FKKYHTMKNY RD (SEQ ID NO: 436) Myotis lucifugus (“2”) 1 MPSLRKRKET NETDTLPEVF NDNLSDIPSE IEDADDCFDD SGDDSTDSTD SEIIRPVRKR 61 KVAVLSSDSD TDEATDNCWS EIDTPPRLQM FEGHAGVTTF PSQCDSVPSV TNLFFGDELF 121 EMLCKELSNY HDQTAMKRKT PSRTLKWSPV TQKDIKKFLG LIILMGQTRK DSLKDYWSTD 181 PLICTPIFPQ TMSRHRFEQI WTFWHFNDNA KMDSRSGRLF KIQPVLDYFL HKFRTIYKPK 241 QQLSLDEGMI PWRGRFKFRT YNPAKITKYG LLVRMVCESD TGYICSMEIY TAEGRKLQET 301 VLSVLGPYLG IWHHIYQDNY YNATSTAELL LQNKTRVCGT IRESRGLPPN LEMKTSRMKK 361 GDIIFSRKGD ILLLAWKDKR VVRMISTIHD TSVSTTGKKN RKTGENIVKP TCIKEYNAHM 421 KGVDRADQFL SCCSILRKTM KWTKKVVLYL INCGLFNSFR VYNVLNPQAK MKYKQFLLSV 481 ARDWITDDNN EGSPEPETNL SSPSPGGARR APRKDPPKRL SGDMKQHEPT CIPASGKKKF 541 PTRACRVCAA HGKRSESRYL CKFCLVPLHR GKCFTQYHTL KKYMDLRCQH TVLSTVGRGY 601 SVLARFKPRT NERTGSSHCH VQVPAGGQGP PSTIIANGCG CKLEPMVRTR SPTCLVIEFG 661 CM (SEQ ID NO: 437) Myotis myotis (“2”) 1 MPSLRKRKET NETDTLPEVF NDNLSDIPSE IEDADDCFDD SGDDSTDSTE SEIIRPVRKR 61 KVAVLSSDSN TDEATDNCWS EIDTPPRLQM FEGHAGVTTF PSQCDSVPSV TNLFFGDELF 121 EMLCKELSNY HDQTAMKRKT PSRTLKWSPV TQKDIKKFLG LIILMGQTRK DSWKDYWSTD 181 PLICTPIFPQ TMSRHRFEQI WTFWHFNDNA KMDSCSGRLF KIQPVLDYFL HKFRTIYKPK 241 QQLSLDEGMI PWRGRLKFTY NPAITKYGLL VRMVCESDTG YICNMEIYTA ERKKLQETVL 301 SVLGPYLGIW HHIYQDNYYN ATSTAELLLQ NKTRVCGTIR ESRGLPPNLK MKTSRMKKGD 361 IIFSRKGDIL LLAWKDKRVV RMISTIHDTS VSTTGKKNRK TGENIVKPTC IKEYNAHMKG 421 VDRADQFLSC CSILRKTTKW TKKVVLYLIN CGLFNSFRVY NILNPQAKMK YKQFLLSVAR 481 DWITDDNNEG SPEPETNLSS PSSGGARRAP RKDQPKRLSG DMKQHEPTCI PASGKKKFPT 541 ACRVCAAHGK RSESRYLRKF CFVPLRGKCF MYHTLKKYSE LFSLIVVSKI QNVIIYKTTK 601 VYMRYVMRSH CPLSFLVFAP SVKDRSRVFS FFTRHLLWTL DVNTLSCPHR MKRSHWWKPC 661 RSIYEKLYNC TNP (SEQ ID NO: 438) Myotis myotis (“2b”) 1 MDLRCQHTVL SIRESRGLPP NLKMKTSRMK KGDIIFSRKG DILLLAWKDK RVVRMISTIH 61 DTSVSTTGKK NRKTGENIVK PACIKEYNAH MKGVDRADQF LSCCSILRKT MKWTKKVVLY 121 LINCGLFNSF RVYNVLNPQA KMKYKQFLLS VARDWITDDN NEGSPEPETN LSSPSPGGAR 181 RAPRKDPPKR LSGDMKQHEP TCIPASGKKK FPTRACRVCA AHGKRSESRY LCKFCLVPLH 241 RGKCFTQYHT LKKY (SEQ ID NO: 439) In embodiments, the mobile element enzyme is derived from Bombyx mori, Xenopus tropicalis, or Trichoplusia ni. In embodiments, the mobile element enzyme is an engineered version of a mobile element enzyme, including but not limited to monomers, dimers, tetramers, hyperactive, or Int-forms, derived from Bombyx mori, Xenopus tropicalis, or Trichoplusia ni. In embodiments, the mobile element enzyme is derived from Bombyx mori, Xenopus tropicalis, Trichoplusia ni, or Myotis lucifugus. In embodiments, the mobile element enzyme is an engineered version, including but not limited to a mobile element enzyme that is a monomer, dimer, tetramer (or another multimer), hyperactive, or has a reduced interaction with non-TTAA (SEQ ID NO: 440) recognitions sites (Int-), derived from Bombyx mori, Xenopus tropicalis, Trichoplusia ni or Myotis lucifugus. In embodiments, the mobile element enzymes have one or more hyperactive and/or integration deficient mutations selected from TABLE 1, TABLE 2A, and TABLE 2B, or equivalents thereof. In embodiments, one skilled in the art can correspond such mutants to mobile element enzymes from any of Bombyx mori, Xenopus tropicalis, Trichoplusia ni, Rhinolophus ferrumequinum, Rousettus aegyptiacus, Phyllostomus discolor, Myotis myotis, Myotis lucifugus, Pipistrellus kuhlii, Pteropus vampyrus, Pan troglodytes, and Molossus molossus. In embodiments, the mobile element enzyme has a nucleotide sequence having at least about 90%, or at least about 93%, or at least about 95%, or at least about 97%, or at least about 98%, or at least about 99% identity to a nucleotide sequence of any of Rhinolophus ferrumequinum, Rousettus aegyptiacus, Phyllostomus discolor, Myotis myotis, Myotis lucifugus, Pteropus vampyrus, Pipistrellus kuhliim, Pan troglodytes, and Molossus molossus. In embodiments, the mobile element enzyme has an amino acid sequence having at least about 90%, or at least about 93%, or at least about 95%, or at least about 97%, or at least about 98%, or at least about 99% identity to an amino acid sequence of any of Rhinolophus ferrumequinum, Rousettus aegyptiacus, Phyllostomus discolor, Myotis myotis, Myotis lucifugus, Pteropus vampyrus, Pipistrellus kuhlii, and Molossus molossus. See Jebb, et al. (2020). In embodiments, the enzyme (e.g., without limitation, a mobile element enzyme) is derived from Bombyx mori, Xenopus tropicalis, Trichoplusia ni, Myotis lucifugus, Rhinolophus ferrumequinum, Rousettus aegyptiacus, Phyllostomus discolor, Myotis myotis, Pteropus vampyrus, Pipistrellus kuhlii, Molossus molossus, Pan troglodytes, or Homo sapiens. In embodiments, the enzyme (e.g., without limitation, a mobile element enzyme) is an engineered version, including but not limited to hyperactive forms, of a mobile element enzyme derived from Bombyx mori, Xenopus tropicalis, Trichoplusia ni, Myotis lucifugus, Rhinolophus ferrumequinum, Rousettus aegyptiacus, Phyllostomus discolor, Myotis myotis, Pteropus vampyrus, Pipistrellus kuhlii, Molossus molossus, Pan troglodytes, or Homo sapiens. The enzyme is either the wild type, monomer, dimer, tetramer, hyperactive, or an Int-mutant. In embodiments, the mobile element enzymes have one or more hyperactive and/or integration deficient mutations selected from TABLE 1, TABLE 2A, and/or TABLE 2B, or equivalents thereof. In embodiments, the mobile element enzyme has a nucleotide sequence having at least about 90%, or at least about 93%, or at least about 95%, or at least about 97%, or at least about 98%, or at least about 99% identity to a nucleotide sequence of any of Rhinolophus ferrumequinum, Rousettus aegyptiacus, Phyllostomus discolor, Myotis myotis, Pteropus vampyrus, Pipistrellus kuhlii, Molossus molossus, and Pan troglodytes. In embodiments, the mobile element enzyme has an amino acid sequence having at least about 90%, or at least about 93%, or at least about 95%, or at least about 97%, or at least about 98%, or at least about 99% identity to an amino acid sequence of any of Rhinolophus ferrumequinum, Rousettus aegyptiacus, Phyllostomus discolor, Myotis myotis, Pteropus vampyrus, Pipistrellus kuhlii, Molossus molossus, Pan troglodytes, and Homo sapiens. In embodiments, the mobile element enzyme is an engineered version, including but not limited to a mobile element enzyme that is a monomer, dimer, tetramer, hyperactive, or has a reduced interaction with non-TTAA (SEQ ID NO: 440) recognitions sites (Int-), derived from any of Bombyx mori, Xenopus tropicalis, Trichoplusia ni, Rhinolophus ferrumequinum, Rousettus aegyptiacus, Phyllostomus discolor, Myotis myotis, Myotis lucifugus, Pipistrellus kuhlii, Pteropus vampyrus, and Molossus molossus Bombyx mori, Xenopus tropicalis, Trichoplusia ni, Pan troglodytes, Myotis lucifugus, and Homo sapiens. The mobile element enzyme is either the wild type, monomer, dimer, tetramer or another multimer, hyperactive, or a an Int-mutant. In embodiments, the mobile element enzyme is from a Tc1/mariner donor DNA system. See, e.g., Plasterk et al. Trends in Genetics.1999; 15(8):326–32. In embodiments, the mobile element enzyme is from a Sleeping Beauty donor DNA system (see, e.g., Cell. 1997;91:501–510), e.g., a hyperactive form of Sleeping Beauty (hypSB), e.g., SB100X (see Gene Therapy volume 18, pages 849–856(2011), or a piggyBac (PB) donor DNA system (see, e.g., Trends Biotechnol.2015 Sep;33(9):525-33, which is incorporated herein by reference in its entirety), e.g., a hyperactive form of PB mobile element enzyme (hypPB), e.g., with seven amino acid substitutions (e.g., I30V, S103P, G165S, M282V, S509G, N570S, N538K on mPB, or functional equivalents in non-mPB, see Mol Ther Nucleic Acids.2012 Oct; 1(10): e50, which is incorporated herein by reference in its entirety); see also Yusa et al., PNAS January 25, 2011108 (4) 1531-1536; Voigt et al., Nature Communications volume 7, Article number: 11126 (2016). The piggyBac mobile element enzymes belong to the IS4 mobile element enzyme family. De Palmenaer et al., BMC Evolutionary Biology.2008;8:18. doi: 10.1186/1471-2148-8-18. The piggyBac family includes a large diversity of donor DNAs, and any of these donor DNAs can be used in embodiments of the present disclosure. See, e.g., Bouallègue et al., Genome Biol Evol.2017;9(2):323-339. The founding member of the piggyBac (super)family, insect piggyBac, was originally identified in the cabbage looper moth (Trichoplusiani ni) and studied both in vivo and in vitro. Insect piggyBac is known to transpose by a canonical cut-and-paste mechanism promoted by an element-encoded mobile element enzyme with a catalytic site resembling the RNase H fold shared by many recombinases. The insect piggyBac donor DNA system has been shown to be highly active in a wide range of animals, including Drosophila and mice, where it has been developed as a powerful tool for gene tagging and genome engineering. Other donor DNAs affiliated to the piggyBac superfamily are common in arthropods and vertebrates including Xenopus and Bombyx. Mammalian piggyBac donor DNAs and mobile element enzymes, including hyperactive mammalian piggyBac variants, which can be used in embodiments of the present disclosure, are described, e.g., in International Application WO2010085699, which is incorporated herein by reference in its entirety. In embodiments, the mobile element enzyme is from a LEAP-IN 1 type or LEAP-IN donor DNA system (Biotechnol J. 2018 Oct;13(10):e1700748. doi: 10.1002/biot.201700748. Epub 2018 Jun 11). The LEAPIN mobile element enzyme system includes a mobile element enzyme (e.g., without limitation, a mobile element enzyme mRNA) and a vector containing one or more genes of interest (donor DNAs), selection markers, regulatory elements, insulators, etc., flanked by the donor DNA cognate inverted terminal ends and the transposition recognition motif (TTAT). Upon co-transfection of vector DNA and mobile element enzyme mRNA, the transiently expressed enzyme catalyzes high-efficiency and precise integration of a single copy of the donor DNA cassette (all sequences between the terminal ends) at one or more sites across the genome of the host cell. Hottentot et al. In Genotyping: Methods and Protocols. White SJ, Cantsilieris S, eds: 185–196. (New York, NY: Springer): 2017. pp.185–196. The LEAPIN mobile element enzyme generates stable transgene integrants with various advantageous characteristics, including single copy integrations at multiple genomic loci, primarily in open chromatin segments; no payload limit, so multiple independent transcriptional units may be expressed from a single construct; the integrated transgenes maintain their structural and functional integrity; and maintenance of transgene integrity ensures the desired chain ratio in every recombinant cell. In embodiments, the mobile element enzyme is an engineered form of a mobile element enzyme reconstructed from Homo sapiens or a predecessor thereof. Donor DNAs in Humans have 5 inactive elements, designated PiggyBac domain (PGBD)1, PGBD2, PGBD3, PGBD4, and PGBD5. PGBD1, PGBD2, and PGBD3 have multiple coding exons, but in each case the mobile element enzyme- related sequence is encoded by a single uninterrupted 3' terminal exon. Thus, PGBD1 and PGBD2 may resemble the PGBD3 donor DNA in which the mobile element enzyme ORF is flanked upstream by a 3' splice site and downstream by a polyadenylation site. See Newman et al., PLoS Genet 2008;4:e1000031. PLoS Genet 4(3): e1000031. https://doi.org/10.1371/journal.pgen.1000031; Gray et al., PLoS Genet 8(9): e1002972. https://doi.org/10.1371/journal.pgen.1002972. The PGBD5 inactive mobile element enzyme sequence belongs to the RNase H clan of Pfam structures, while PGBD3 has sustained only a single D to N mutation in the essential catalytic triad DDD(D) and retains the ability to bind the upstream piggyBac terminal inverted repeat. Bailey et al., DNA Repair (Amst) 2012;11:488-501. The PGBD5 mobile element enzyme does not retain the catalytic DDD (D) motif found in active elements, and the mobile element enzyme is not only inactive but fails to associate with either DNA or chromatin in vivo. Pavelitz et al., Mob DNA 2013;4:23. However, in vitro studies showed that it is transpositionally active in HEK293 cells. See Henssen et al., Elife 2015;4. PGBD1 and PGBD2 are thought to be present in the common ancestor of mammals, while PGBD3 and PGBD4 are restricted to primates. See Sarkar et al., Mol Genet Genomics 2003;270:173-80. The Pteropus vampyrus mobile element enzyme is closely related to PGBD4 and shares DDD catalytic domain and the C-terminal region that are involved in excision mechanisms. See Mitra et al., EMBO J 2008;27:1097-109. A mammalian mobile element enzyme, which has gene cleavage and/or gene integration activity, can be constructed based on alignment of the amino acid sequence of Pteropus vampyrus mobile element enzyme to PGBD1, PGBD2, PGBD3, PGBD4, and PGBD5 sequences. Also, in embodiments. the mammalian mobile element enzyme has mutations that confers hyperactivity to a recombinant mammalian mobile element enzyme. Accordingly, in embodiments, the mobile element enzyme has gene cleavage activity (Exc+) and/or gene integration activity (Int+). In embodiments, the mobile element enzyme has gene cleavage activity (Exc+) and/or lacks gene integration activity (Int-). In some aspects, an enzyme capable of targeted genomic integration is a recombinant mammalian mobile element enzyme that was derived by, in part, aligning several inactive mobile element enzyme sequences from a human genome to Pteropus vampyrus mobile element enzyme sequence. In embodiments, the Pteropus vampyrus mobile element enzyme has an amino acid sequence having at least 90% identity (e.g., a sequence having at least about 90%, or at least about 93%, or at least about 95%, or at least about 97%, or at least about 98%, or at least about 99% identity) to SEQ ID NO: 430 (or a functional equivalent thereof. In embodiments, the Pteropus vampyrus mobile element enzyme has an amino acid sequence of SEQ ID NO: 430, or a functional equivalent thereof. In embodiments, the Pteropus vampyrus mobile element enzyme has a nucleotide sequence having at least 90% identity (e.g., a sequence having at least about 90%, or at least about 93%, or at least about 95%, or at least about 97%, or at least about 98%, or at least about 99% identity) to SEQ ID NO: 429 or a codon-optimized variant thereof. In embodiments, the mobile element enzyme is a mammalian mobile element enzyme, such as a mobile element enzyme from a bat, e.g., without limitation, Pteropus vampyrus. In embodiments, the mobile element enzyme is an engineered form that is based on a mobile element enzyme reconstructed from Homo sapiens or a predecessor thereof. In embodiments, the mobile element enzyme includes but is not limited to an engineered version that is a monomer, dimer, tetramer (or another multimer), hyperactive, or has a reduced interaction with non-TTAA (SEQ ID NO: 440) recognitions sites (Int-), of an engineered version of a mobile element enzyme reconstructed from Homo sapiens or a predecessor thereof. In embodiments, the mobile element enzyme is an engineered form that is based on a mobile element enzyme reconstructed from mammalian species. In embodiments, the mobile element enzyme includes but is not limited to an engineered that is a monomer, dimer, tetramer (or another multimer), hyperactive, or has a reduced interaction with non-TTAA (SEQ ID NO: 440) recognitions sites (Int-), of a mobile element enzyme reconstructed from mammalian species. In embodiments, the donor DNA is included in a vector comprising left and right end sequences recognized by the mobile element enzyme. In embodiments, the end sequences are selected from MER, MER75A, MER75B, and MER85. In embodiments, the end sequences are selected from nucleotide sequences of SEQ ID NO: 12, SEQ ID NO: 13 , SEQ ID NO: 14, SEQ ID NO: 15, SEQ ID NO: 16, SEQ ID NO: 17, SEQ ID NO: 18, SEQ ID NO: 19, SEQ ID NO: 20, SEQ ID NO: 21, SEQ ID NO: 441, and SEQ ID NO: 22, or a nucleotide sequence having at least about 90% identity (e.g., a sequence having at least about 90%, or at least about 93%, or at least about 95%, or at least about 97%, or at least about 98%, or at least about 99% identity) thereto. In embodiments, one or more of the end sequences are optionally flanked by a TTAA (SEQ ID NO: 440) sequence. In embodiments, the end sequences include at least one repeat from a nucleotide sequence having at least about 90% (e.g., a sequence having at least about 90%, or at least about 93%, or at least about 95%, or at least about 97%, or at least about 98%, or at least about 99% identity) identity to the nucleotide sequence of SEQ ID NO: 12, and wherein the at least one repeat from the nucleotide sequence having at least about 90% identity (e.g., a sequence having at least about 90%, or at least about 93%, or at least about 95%, or at least about 97%, or at least about 98%, or at least about 99% identity) to the nucleotide sequence of SEQ ID NO: 12 is positioned at the 5’ end of the donor DNA. The end sequences can further include at least one repeat from a nucleotide sequence having at least about 90% identity (e.g., a sequence having at least about 90%, or at least about 93%, or at least about 95%, or at least about 97%, or at least about 98%, or at least about 99% identity to the nucleotide sequence of SEQ ID NO: 17, and wherein the at least one repeat from the nucleotide sequence having at least about 90% identity (e.g., a sequence having at least about 90%, or at least about 93%, or at least about 95%, or at least about 97%, or at least about 98%, or at least about 99% identity) to the nucleotide sequence of SEQ ID NO: 17 is positioned at the 3’ end of the donor DNA. The end sequences, which can be from, e.g., Pteropus vampyrus, are optionally flanked by a TTAA (SEQ ID NO: 440) sequence. In embodiments, the end sequences include at least one repeat from a nucleotide sequence having at least about 90% identity (e.g., a sequence having at least about 90%, or at least about 93%, or at least about 95%, or at least about 97%, or at least about 98%, or at least about 99% identity) to the nucleotide sequence of SEQ ID NO: 13, and wherein the at least one repeat from the nucleotide sequence having at least about 90% identity (e.g., a sequence having at least about 90%, or at least about 93%, or at least about 95%, or at least about 97%, or at least about 98%, or at least about 99% identity) to the nucleotide sequence of SEQ ID NO: 13 is positioned at the 5’ end of the donor DNA. The end sequences can further include at least one repeat from a nucleotide sequence having at least about 90% identity (e.g., a sequence having at least about 90%, or at least about 93%, or at least about 95%, or at least about 97%, or at least about 98%, or at least about 99% identity) to the nucleotide sequence of SEQ ID NO: 18, and wherein the at least one repeat from the nucleotide sequence having at least about 90% identity (e.g., a sequence having at least about 90%, or at least about 93%, or at least about 95%, or at least about 97%, or at least about 98%, or at least about 99% identity) to the nucleotide sequence of SEQ ID NO: 18 is positioned at the 3’ end of the donor DNA. The end sequences, which can be, e.g., PGBD4, are optionally flanked by a TTAA (SEQ ID NO: 440) sequence. In embodiments, the end sequences include at least one repeat from a nucleotide sequence having at least about 90% identity (e.g., a sequence having at least about 90%, or at least about 93%, or at least about 95%, or at least about 97%, or at least about 98%, or at least about 99% identity) to the nucleotide sequence of SEQ ID NO: 14, wherein the at least one repeat from the nucleotide sequence having at least about 90% identity (e.g., a sequence having at least about 90%, or at least about 93%, or at least about 95%, or at least about 97%, or at least about 98%, or at least about 99% identity) to the nucleotide sequence of SEQ ID NO: 14 is positioned at the 5’ end of the donor DNA. The end sequences include at least one repeat from a nucleotide sequence having at least about 90% identity (e.g., a sequence having at least about 90%, or at least about 93%, or at least about 95%, or at least about 97%, or at least about 98%, or at least about 99% identity) to the nucleotide sequence of SEQ ID NO: 18, wherein the at least one repeat from the nucleotide sequence having at least about 90% identity (e.g., a sequence having at least about 90%, or at least about 93%, or at least about 95%, or at least about 97%, or at least about 98%, or at least about 99% identity) to the nucleotide sequence of SEQ ID NO: 19 is positioned at the 3’ end of the donor DNA. The end sequences, which can be, e.g., MER75, are optionally flanked by a TTAA (SEQ ID NO: 440) sequence. In embodiments, the end sequences include at least one repeat from a nucleotide sequence having at least about 90% identity (e.g., a sequence having at least about 90%, or at least about 93%, or at least about 95%, or at least about 97%, or at least about 98%, or at least about 99% identity) to the nucleotide sequence of SEQ ID NO: 15, wherein the at least one repeat from the nucleotide sequence having at least about 90% identity (e.g., a sequence having at least about 90%, or at least about 93%, or at least about 95%, or at least about 97%, or at least about 98%, or at least about 99% identity) to the nucleotide sequence of SEQ ID NO: 15 is positioned at the 5’ end of the donor DNA. The end sequences include at least one repeat from a nucleotide sequence having at least about 90% identity (e.g., a sequence having at least about 90%, or at least about 93%, or at least about 95%, or at least about 97%, or at least about 98%, or at least about 99% identity) to the nucleotide sequence of SEQ ID NO: 20, wherein the at least one repeat from the nucleotide sequence having at least about 90% identity(e.g. a sequence having at least about 90%, or at least about 93%, or at least about 95%, or at least about 97%, or at least about 98%, or at least about 99% identity) to the nucleotide sequence of SEQ ID NO: 20 is positioned at the 3’ end of the donor DNA. The end sequences, which can be, e.g., MER75B, are optionally flanked by a TTAA (SEQ ID NO: 440) sequence. In embodiments, the end sequences include at least one repeat from a nucleotide sequence having at least about 90% (e.g., a sequence having at least about 90%, or at least about 93%, or at least about 95%, or at least about 97%, or at least about 98%, or at least about 99% identity) identity to the nucleotide sequence of SEQ ID NO: 16, wherein the at least one repeat from the nucleotide sequence having at least about 90% identity (e.g., a sequence having at least about 90%, or at least about 93%, or at least about 95%, or at least about 97%, or at least about 98%, or at least about 99% identity) to the nucleotide sequence of SEQ ID NO: 16 is positioned at the 5’ end of the donor DNA. The end sequences include at least one repeat from a nucleotide sequence having at least about 90% identity (e.g., a sequence having at least about 90%, or at least about 93%, or at least about 95%, or at least about 97%, or at least about 98%, or at least about 99% identity) to the nucleotide sequence of SEQ ID NO: 21 or SEQ ID NO: 441, wherein the at least one repeat from the nucleotide sequence having at least about 90% identity (e.g., a sequence having at least about 90%, or at least about 93%, or at least about 95%, or at least about 97%, or at least about 98%, or at least about 99% identity) to the nucleotide sequence of SEQ ID NO: 21 or SEQ ID NO: 441 is positioned at the 3’ end of the donor DNA. The end sequences, which can be, e.g., MER75A, are optionally flanked by a TTAA (SEQ ID NO: 440) sequence. In embodiments, a donor DNA is or comprises a vector comprising a donor DNA comprising one or more end sequences recognized by an enzyme such as, for example a mobile element enzyme. In embodiments, the end sequences are selected from Pteropus vampyrus, MER75, MER75A, and MER75B. MERs contain end sequences with similarity to piggyBac-like mobile elements and exhibit duplications of their presumed TTAA (SEQ ID NO: 440) target sites. In embodiments, the end sequences are selected from nucleotide sequences of SEQ ID NO: 12, SEQ ID NO: 13, SEQ ID NO: 14, SEQ ID NO: 15, SEQ ID NO: 16, SEQ ID NO: 17, SEQ ID NO: 18, SEQ ID NO: 19, SEQ ID NO: 20, SEQ ID NO: 21, SEQ ID NO: 441, and SEQ ID NO: 22, or a nucleotide sequence having at least about 90% identity (e.g. a sequence having at least about 90%, or at least about 93%, or at least about 95%, or at least about 97%, or at least about 98%, or at least about 99% identity) thereto. In embodiments, the mobile element enzyme has an amino acid sequence of SEQ ID NO: 3, SEQ ID NO: 4, SEQ ID NO: 6, SEQ ID NO: 7, SEQ ID NO: 8, or SEQ ID NO: 9, or a variant sequence having at least about 90%, or at least about 93%, or at least about 95%, or at least about 97%, or at least about 98%, or at least about 99% identity thereto, or at least about 10 mutations, or at least about 9 mutations, or at least about 8 mutations, or at least about 7 mutations, or at least about 6 mutations, or at least about 5 mutations, or at least about 4 mutations, or at least about 3 mutations, or at least about 2 mutations, or at least about 1 mutation. In embodiments, the mobile element enzyme has an amino acid sequence having S8P, G17R, and/or K134K mutation relative to the amino acid sequence of SEQ ID NO: 4 or a functional equivalent thereof. In embodiments, the mobile element enzyme has an amino acid sequence having S8P, G17R, and/or K134K mutation relative to the amino acid sequence of SEQ ID NO: 5 or a functional equivalent thereof. In embodiments, the mobile element enzyme has an amino acid sequence having I83P and/or V118R mutation relative to the amino acid sequence of SEQ ID NO: 6 or a functional equivalent thereof. In embodiments, the mobile element enzyme has an amino acid sequence having S20P and/or A29R mutation relative to the amino acid sequence of SEQ ID NO: 7 or a functional equivalent thereof. In embodiments, the mobile element enzyme has an amino acid sequence having T4P and/or L13R mutation relative to the amino acid sequence of SEQ ID NO: 8 or a functional equivalent thereof. In embodiments, the mobile element enzyme has an amino acid sequence having A12P and/or I28R mutation and/or R152K mutation relative to the amino acid sequence of SEQ ID NO: 9 or a functional equivalent thereof. In embodiments, the enzyme capable of targeted genomic integration (e.g., without limitations, a mobile element enzyme) is in a monomeric or dimeric form. In embodiments, the enzyme capable of targeted genomic integration (e.g., without limitations, a mobile element enzyme) is in a multimeric form. In embodiments, the enzyme (e.g., without limitation, a mobile element enzyme) is an engineered version, including but not limited to a mobile element enzyme that is a monomer, dimer, tetramer, hyperactive, or has a reduced interaction with non-TTAA (SEQ ID NO: 440) recognitions sites (Int-), and is derived from any of Bombyx mori, Xenopus tropicalis, Trichoplusia ni, Myotis lucifugus, Rhinolophus ferrumequinum, Rousettus aegyptiacus, Phyllostomus discolor, Myotis myotis, Pteropus vampyrus, Pipistrellus kuhlii, Pan troglodytes, Molossus molossus, or Homo sapiens. In embodiments, the mobile element enzyme is either the wild type, monomer, dimer, tetramer or another multimer, hyperactive, or an Int-mutant. Targeting Chimeric Constructs In aspects, the present disclosure provides for targeted chimeras, e.g., in embodiments, the enzyme, without limitation, a mobile element enzyme, comprises a targeting element. In embodiments, the targeting element is or comprises a gene-editing system, e.g., that is catalytically inactive (or “dead”). in embodiments, the enzyme, without limitation, a mobile element enzyme, associated with the targeting element, is capable of inserting the donor DNA (e.g., mobile element) comprising a transgene, optionally at a TA dinucleotide site or a TTAA (SEQ ID NO: 440) tetranucleotide site in a GSHS. In embodiments, the enzyme, without limitation, a mobile element enzyme, associated with the targeting element has one or more mutations which confer hyperactivity. In embodiments, the enzyme, without limitation, a mobile element enzyme, associated with the targeting element has gene cleavage activity (Exc+) and/or gene integration activity (Int+). In embodiments, the enzyme, without limitation, a mobile element enzyme, associated with the targeting element has gene cleavage activity (Exc+) and/or a lack of gene integration activity (Int-). In embodiments, the targeting element comprises one or more proteins or nucleic acids that are capable of binding to a nucleic acid. In embodiments, the targeting element comprises one or more of a of a gRNA, optionally associated with a Cas enzyme, which is optionally catalytically inactive, transcription activator-like effector (TALE), catalytically inactive Zinc finger, catalytically inactive transcription factor, nickase, a transcriptional activator, a transcriptional repressor, a recombinase, a DNA methyltransferase, a histone methyltransferase, paternally expressed gene 10 (PEG10), and TnsD. In embodiments, the targeting element comprises a transcription activator-like effector (TALE) DNA binding domain (DBD). In embodiments, the TALE DBD comprises one or more repeat sequences. In embodiments, the TALE DBD comprises about 14, or about 15, or about, 16, or about 17, or about 18, or about 18.5 repeat sequences. In embodiments, the TALE DBD repeat sequences comprise 33 or 34 amino acids. In embodiments, the TALE DBD repeat sequences comprise a repeat variable di-residue (RVD) at residue 12 or 13 of the 33 or 34 amino acids. In embodiments, the RVD recognizes one base pair in the nucleic acid molecule. In embodiments, the RVD recognizes a C residue in the nucleic acid molecule and is selected from HD, N(gap), HA, ND, and HI. In embodiments, the RVD recognizes a G residue in the nucleic acid molecule and is selected from NN, NH, NK, HN, and NA. In embodiments, the RVD recognizes an A residue in the nucleic acid molecule and is selected from NI and NS. In embodiments, the RVD recognizes a T residue in the nucleic acid molecule and is selected from NG, HG, H(gap), and IG. In embodiments, the GSHS is in an open chromatin location in a chromosome. In embodiments, the GSHS is selected from adeno-associated virus site 1 (AAVS1), chemokine (C-C motif) receptor 5 (CCR5) gene, HIV-1 coreceptor, and human Rosa26 locus. In embodiments, the GSHS is located on human chromosome 2, 4, 6, 10, 11, 17, 22, or X. In embodiments, the GSHS is selected from TALC1, TALC2, TALC3, TALC4, TALC5, TALC7, TALC8, AVS1, AVS2, AVS3, ROSA1, ROSA2, TALER1, TALER2, TALER3, TALER4, TALER5, SHCHR2-1, SHCHR2-2, SHCHR2-3, SHCHR2-4, SHCHR4-1, SHCHR4-2, SHCHR4-3, SHCHR6-1, SHCHR6-2, SHCHR6-3, SHCHR6-4, SHCHR10-1, SHCHR10-2, SHCHR10-3, SHCHR10-4, SHCHR10-5, SHCHR11-1, SHCHR11-2, SHCHR11-3, SHCHR17-1, SHCHR17-2, SHCHR17-3, and SHCHR17-4. In embodiments, the targeting element comprises a Cas9 enzyme guide RNA complex. In embodiments, the Cas9 enzyme guide RNA complex comprises a nuclease-deficient dCas9 guide RNA complex. In embodiments, the targeting element comprises a Cas12 enzyme guide RNA complex. In embodiments, the targeting element comprises a nuclease-deficient dCas12 guide RNA complex, optionally dCas12j guide RNA complex or dCas12a guide RNA complex. In embodiments, the targeting element comprises a Cas12k enzyme guide RNA complex. In embodiments, the targeting element comprises a nuclease-deficient dCas12 guide RNA complex, optionally dCas12k guide RNA complex. In embodiments, a targeting chimeric system or construct, having a DBD fused to a mobile element enzyme, directs binding of an enzyme capable of targeted genomic integration (e.g., without limitation, a mobile element enzyme) to a specific sequence (e.g., transcription activator-like effector proteins (TALE) repeat variable di-residues (RVD) or gRNA) near an enzyme recognition site. The enzyme is thus prevented from binding to random recognition sites. In embodiments, the targeting chimeric construct binds to human GSHS. In embodiments, dCas9 (i.e., deficient for nuclease activity) is programmed with gRNAs directed to bind at a desired sequence of DNA in GSHS. In embodiments, TALEs described herein can physically sequester the enzyme such as, e.g., a mobile element enzyme, to GSHS and promote transposition to nearby TTAA (SEQ ID NO: 440) sequences in close proximity to the RVD TALE nucleotide sequences. GSHS in open chromatin sites are specifically targeted based on the predilection for mobile element enzymes to insert into open chromatin. In embodiments, an enzyme capable of targeted genomic integration (e.g., without limitation, a recombinase, integrase, or a mobile element enzyme such as, without limitation, a mammalian mobile element enzyme) is linked to or fused with a TALE DNA binding domain (DBD) or a Cas-based gene-editing system, such as, e.g., Cas9 or a variant thereof. In embodiments, the targeting element targets the enzyme to a locus of interest. In embodiments, the targeting element comprises CRISPR (Clustered Regularly Interspaced Short Palindromic Repeat) associated protein 9 (Cas9), or a variant thereof. A CRISPR/Cas9 tool only requires Cas9 nuclease for DNA cleavage and a single-guide RNA (sgRNA) for target specificity. See Jinek et al. (2012) Science 337, 816–821; Chylinski et al. (2014) Nucleic Acids Res 42, 6091– 6105. The inactivated form of Cas9, which is a nuclease-deficient (or inactive, or “catalytically dead” Cas9, is typically denoted as “dCas9,” has no substantial nuclease activity. Qi, L. S. et al. (2013). Cell 152, 1173–1183. CRISPR/dCas9 binds precisely to specific genomic sequences through targeting of guide RNA (gRNA) sequences. See Dominguez et al., Nat Rev Mol Cell Biol.2016;17:5–15; Wang et al., Annu Rev Biochem.2016;85:227–64. dCas9 is utilized to edit gene expression when applied to the transcription binding site of a desired site and/or locus in a genome. When the dCas9 protein is coupled to guide RNA (gRNA) to create dCas9 guide RNA complex, dCas9 prevents the proliferation of repeating codons and DNA sequences that might be harmful to an organism's genome. Essentially, when multiple repeat codons are produced, it elicits a response, or recruits an abundance of dCas9 to combat the overproduction of those codons and results in the shut-down of transcription. Thus, dCas9 works synergistically with gRNA and directly affects the DNA polymerase II from continuing transcription. In embodiments, the targeting element comprises a nuclease-deficient Cas enzyme guide RNA complex. In embodiments, the targeting element comprises a nuclease-deficient (or inactive, or “catalytically dead” Cas, e.g., Cas9, typically denoted as “dCas” or “dCas9” ) guide RNA complex. In embodiments, the dCas9/gRNA complex comprises a guide RNA selected from: GTTTAGCTCACCCGTGAGCC (SEQ ID NO: 91), CCCAATATTATTGTTCTCTG (SEQ ID NO: 92), GGGGTGGGATAGGGGATACG (SEQ ID NO: 93), GGATCCCCCTCTACATTTAA (SEQ ID NO: 94), GTGATCTTGTACAAATCATT (SEQ ID NO: 95), CTACACAGAATCTGTTAGAA (SEQ ID NO: 96), TAAGCTAGAGAATAGATCTC (SEQ ID NO: 97), and TCAATACACTTAATGATTTA (SEQ ID NO: 98), wherein the guide RNA directs the enzyme to a chemokine (C-C motif) receptor 5 (CCR5) gene. In embodiments, the dCas9/gRNA complex comprises a guide RNA selected from: CACCGGGAGCCACGAAAACAGATCC (SEQ ID NO: 99);CACCGCGAAAACAGATCCAGGGACA (SEQ ID NO: 100); CACCGAGATCCAGGGACACGGTGCT (SEQ ID NO: 101); CACCGGACACGGTGCTAGGACAGTG (SEQ ID NO: 102); CACCGGAAAATGACCCAACAGCCTC (SEQ ID NO: 103); CACCGGCCTGGCCGGCCTGACCACT (SEQ ID NO: 104); CACCGCTGAGCACTGAAGGCCTGGC (SEQ ID NO: 105); CACCGTGGTTTCCACTGAGCACTGA (SEQ ID NO: 106); CACCGGATAGCCAGGAGTCCTTTCG (SEQ ID NO: 107); CACCGGCGCTTCCAGTGCTCAGACT (SEQ ID NO: 108); CACCGCAGTGCTCAGACTAGGGAAG (SEQ ID NO: 109); CACCGGCCCCTCCTCCTTCAGAGCC (SEQ ID NO: 110); CACCGTCCTTCAGAGCCAGGAGTCC (SEQ ID NO: 111); CACCGTGGTTTCCGAGCTTGACCCT (SEQ ID NO: 112); CACCGCTGCAGAGTATCTGCTGGGG (SEQ ID NO: 113); CACCGCGTTCCTGCAGAGTATCTGC (SEQ ID NO: 114); TCCCCTCCCAGAAAGACCTG (SEQ ID NO: 131); TGGGCTCCAAGCAATCCTGG (SEQ ID NO: 132); GTGGCTCAGGAGGTACCTGG (SEQ ID NO: 133); GAGCCACGAAAACAGATCCA (SEQ ID NO: 134); AAGTGAACGGGGAAGGGAGG (SEQ ID NO: 135); GACAAAAGCCGAAGTCCAGG (SEQ ID NO: 136); GTGGTTGATAAACCCACGTG (SEQ ID NO: 137); TGGGAACAGCCACAGCAGGG (SEQ ID NO: 138); GCAGGGGAACGGGGATGCAG (SEQ ID NO: 139); GAGATGGTGGACGAGGAAGG (SEQ ID NO: 140); GAGATGGCTCCAGGAAATGG (SEQ ID NO: 141); TAAGGAATCTGCCTAACAGG (SEQ ID NO: 142); TCAGGAGACTAGGAAGGAGG (SEQ ID NO: 143); TATAAGGTGGTCCCAGCTCG (SEQ ID NO: 144); CTGGAAGATGCCATGACAGG (SEQ ID NO: 145); GCACAGACTAGAGAGGTAAG (SEQ ID NO: 146); ACAGACTAGAGAGGTAAGGG (SEQ ID NO: 147); GAGAGGTGACCCGAATCCAC (SEQ ID NO: 148); GCACAGGCCCCAGAAGGAGA (SEQ ID NO: 149); CCGGAGAGGACCCAGACACG (SEQ ID NO: 150); GAGAGGACCCAGACACGGGG (SEQ ID NO: 151); GCAACACAGCAGAGAGCAAG (SEQ ID NO: 152); GAAGAGGGAGTGGAGGAAGA (SEQ ID NO: 153); AAGACGGAACCTGAAGGAGG (SEQ ID NO: 154); AGAAAGCGGCACAGGCCCAG (SEQ ID NO: 155); GGGAAACAGTGGGCCAGAGG (SEQ ID NO: 156); GTCCGGACTCAGGAGAGAGA (SEQ ID NO: 157); GGCACAGCAAGGGCACTCGG (SEQ ID NO: 158); GAAGAGGGGAAGTCGAGGGA (SEQ ID NO: 159); GGGAATGGTAAGGAGGCCTG (SEQ ID NO: 160); GCAGAGTGGTCAGCACAGAG (SEQ ID NO: 161); GCACAGAGTGGCTAAGCCCA (SEQ ID NO: 162); GACGGGGTGTCAGCATAGGG (SEQ ID NO: 163); GCCCAGGGCCAGGAACGACG (SEQ ID NO: 164); GGTGGAGTCCAGCACGGCGC (SEQ ID NO: 165); ACAGGCCGCCAGGAACTCGG (SEQ ID NO: 166); ACTAGGAAGTGTGTAGCACC (SEQ ID NO: 167); ATGAATAGCAGACTGCCCCG (SEQ ID NO: 168); ACACCCCTAAAAGCACAGTG (SEQ ID NO: 169); CAAGGAGTTCCAGCAGGTGG (SEQ ID NO: 170); AAGGAGTTCCAGCAGGTGGG (SEQ ID NO: 171); TGGAAAGAGGAGGGAAGAGG (SEQ ID NO: 172); TCGAATTCCTAACTGCCCCG (SEQ ID NO: 173); GACCTGCCCAGCACACCCTG (SEQ ID NO: 174); GGAGCAGCTGCGGCAGTGGG (SEQ ID NO: 175); GGGAGGGAGAGCTTGGCAGG (SEQ ID NO: 176); GTTACGTGGCCAAGAAGCAG (SEQ ID NO: 177); GCTGAACAGAGAAGAGCTGG (SEQ ID NO: 178); TCTGAGGGTGGAGGGACTGG (SEQ ID NO: 179); GGAGAGGTGAGGGACTTGGG (SEQ ID NO: 180); GTGAACCAGGCAGACAACGA (SEQ ID NO: 181); CAGGTACCTCCTGAGCCACG (SEQ ID NO: 182); GGGGGAGTAGGGGCATGCAG (SEQ ID NO: 183); GCAAATGGCCAGCAAGGGTG (SEQ ID NO: 184); CAAATGGCCAGCAAGGGTGG (SEQ ID NO: 309); GCAGAACCTGAGGATATGGA (SEQ ID NO: 310); AATACACAGAATGAAAATAG (SEQ ID NO: 311); CTGGTGACTAGAATAGGCAG (SEQ ID NO: 312); TGGTGACTAGAATAGGCAGT (SEQ ID NO: 313); TAAAAGAATGTGAAAAGATG (SEQ ID NO: 314); TCAGGAGTTCAAGACCACCC (SEQ ID NO: 315); TGTAGTCCCAGTTATGCAGG (SEQ ID NO: 316); GGGTTCACACCACAAATGCA (SEQ ID NO: 317); GGCAAATGGCCAGCAAGGGT (SEQ ID NO: 318); AGAAACCAATCCCAAAGCAA (SEQ ID NO: 319); GCCAAGGACACCAAAACCCA (SEQ ID NO: 320); AGTGGTGATAAGGCAACAGT (SEQ ID NO: 321); CCTGAGACAGAAGTATTAAG (SEQ ID NO: 322); AAGGTCACACAATGAATAGG (SEQ ID NO: 323); CACCATACTAGGGAAGAAGA (SEQ ID NO: 324); CAATACCCTGCCCTTAGTGG (SEQ ID NO: 327); AATACCCTGCCCTTAGTGGG (SEQ ID NO: 325); TTAGTGGGGGGTGGAGTGGG (SEQ ID NO: 326); GTGGGGGGTGGAGTGGGGGG (SEQ ID NO: 328); GGGGGGTGGAGTGGGGGGTG (SEQ ID NO: 329); GGGGTGGAGTGGGGGGTGGG (SEQ ID NO: 330); GGGTGGAGTGGGGGGTGGGG (SEQ ID NO: 331); GGGGGTGGGGAAAGACATCG (SEQ ID NO: 332); GCAGCTGTGAATTCTGATAG (SEQ ID NO: 333); GAGATCAGAGAAACCAGATG (SEQ ID NO: 334); TCTATACTGATTGCAGCCAG (SEQ ID NO: 335); CACCGAATCGAGAAGCGACTCGACA (SEQ ID NO: 185); CACCGGTCCCTGGGCGTTGCCCTGC (SEQ ID NO: 186); CACCGCCCTGGGCGTTGCCCTGCAG (SEQ ID NO: 187); CACCGCCGTGGGAAGATAAACTAAT (SEQ ID NO: 188); CACCGTCCCCTGCAGGGCAACGCCC (SEQ ID NO: 189); CACCGGTCGAGTCGCTTCTCGATTA (SEQ ID NO: 190); CACCGCTGCTGCCTCCCGTCTTGTA (SEQ ID NO: 191); CACCGGAGTGCCGCAATACCTTTAT (SEQ ID NO: 192); CACCGACACTTTGGTGGTGCAGCAA (SEQ ID NO: 193); CACCGTCTCAAATGGTATAAAACTC (SEQ ID NO: 194); CACCGAATCCCGCCCATAATCGAGA (SEQ ID NO: 195); CACCGTCCCGCCCATAATCGAGAAG (SEQ ID NO: 196); CACCGCCCATAATCGAGAAGCGACT (SEQ ID NO: 197); CACCGGAGAAGCGACTCGACATGGA (SEQ ID NO: 198); CACCGGAAGCGACTCGACATGGAGG (SEQ ID NO: 199); CACCGGCGACTCGACATGGAGGCGA (SEQ ID NO: 200); AAACTGTCGAGTCGCTTCTCGATTC (SEQ ID NO: 201); AAACGCAGGGCAACGCCCAGGGACC (SEQ ID NO: 202); AAACCTGCAGGGCAACGCCCAGGGC (SEQ ID NO: 203); AAACATTAGTTTATCTTCCCACGGC (SEQ ID NO: 204); AAACGGGCGTTGCCCTGCAGGGGAC (SEQ ID NO: 205); AAACTAATCGAGAAGCGACTCGACC (SEQ ID NO: 206); AAACTACAAGACGGGAGGCAGCAGC (SEQ ID NO: 207); AAACATAAAGGTATTGCGGCACTCC (SEQ ID NO: 208); AAACTTGCTGCACCACCAAAGTGTC (SEQ ID NO: 209); AAACGAGTTTTATACCATTTGAGAC (SEQ ID NO: 210); AAACTCTCGATTATGGGCGGGATTC (SEQ ID NO: 211); AAACCTTCTCGATTATGGGCGGGAC (SEQ ID NO: 212); AAACAGTCGCTTCTCGATTATGGGC (SEQ ID NO: 213); AAACTCCATGTCGAGTCGCTTCTCC (SEQ ID NO: 214); AAACCCTCCATGTCGAGTCGCTTCC (SEQ ID NO: 215); AAACTCGCCTCCATGTCGAGTCGCC (SEQ ID NO: 216); CACCGACAGGGTTAATGTGAAGTCC (SEQ ID NO: 217); CACCGTCCCCCTCTACATTTAAAGT (SEQ ID NO: 218); CACCGCATTTAAAGTTGGTTTAAGT (SEQ ID NO: 219); CACCGTTAGAAAATATAAAGAATAA (SEQ ID NO: 220); CACCGTAAATGCTTACTGGTTTGAA (SEQ ID NO: 221); CACCGTCCTGGGTCCAGAAAAAGAT (SEQ ID NO: 222); CACCGTTGGGTGGTGAGCATCTGTG (SEQ ID NO: 223); CACCGCGGGGAGAGTGGAGAAAAAG (SEQ ID NO: 224); CACCGGTTAAAACTCTTTAGACAAC (SEQ ID NO: 225); CACCGGAAAATCCCCACTAAGATCC (SEQ ID NO: 226); AAACGGACTTCACATTAACCCTGTC (SEQ ID NO: 227); AAACACTTTAAATGTAGAGGGGGAC (SEQ ID NO: 228); AAACACTTAAACCAACTTTAAATGC (SEQ ID NO: 229); AAACTTATTCTTTATATTTTCTAAC (SEQ ID NO: 230); AAACTTCAAACCAGTAAGCATTTAC (SEQ ID NO: 231); AAACATCTTTTTCTGGACCCAGGAC (SEQ ID NO: 232); AAACCACAGATGCTCACCACCCAAC (SEQ ID NO: 233); AAACCTTTTTCTCCACTCTCCCCGC (SEQ ID NO: 234); AAACGTTGTCTAAAGAGTTTTAACC (SEQ ID NO: 235); AAACGGATCTTAGTGGGGATTTTCC (SEQ ID NO: 236); AGTAGCAGTAATGAAGCTGG (SEQ ID NO: 237); ATACCCAGACGAGAAAGCTG (SEQ ID NO: 238); TACCCAGACGAGAAAGCTGA (SEQ ID NO: 239); GGTGGTGAGCATCTGTGTGG (SEQ ID NO: 240); AAATGAGAAGAAGAGGCACA (SEQ ID NO: 241); CTTGTGGCCTGGGAGAGCTG (SEQ ID NO: 242); GCTGTAGAAGGAGACAGAGC (SEQ ID NO: 243); GAGCTGGTTGGGAAGACATG (SEQ ID NO: 244); CTGGTTGGGAAGACATGGGG (SEQ ID NO: 245); CGTGAGGATGGGAAGGAGGG (SEQ ID NO: 246); ATGCAGAGTCAGCAGAACTG (SEQ ID NO: 247); AAGACATCAAGCACAGAAGG (SEQ ID NO: 248); TCAAGCACAGAAGGAGGAGG (SEQ ID NO: 249); AACCGTCAATAGGCAAAGGG (SEQ ID NO: 250); CCGTATTTCAGACTGAATGG (SEQ ID NO: 251); GAGAGGACAGGTGCTACAGG (SEQ ID NO: 252); AACCAAGGAAGGGCAGGAGG (SEQ ID NO: 253); GACCTCTGGGTGGAGACAGA (SEQ ID NO: 254); CAGATGACCATGACAAGCAG (SEQ ID NO: 255); AACACCAGTGAGTAGAGCGG (SEQ ID NO: 256); AGGACCTTGAAGCACAGAGA (SEQ ID NO: 257); TACAGAGGCAGACTAACCCA (SEQ ID NO: 258); ACAGAGGCAGACTAACCCAG (SEQ ID NO: 259); TAAATGACGTGCTAGACCTG (SEQ ID NO: 260); AGTAACCACTCAGGACAGGG (SEQ ID NO: 261); ACCACAAAACAGAAACACCA (SEQ ID NO: 262); GTTTGAAGACAAGCCTGAGG (SEQ ID NO: 263); GCTGAACCCCAAAAGACAGG (SEQ ID NO: 264); GCAGCTGAGACACACACCAG (SEQ ID NO: 265); AGGACACCCCAAAGAAGCTG (SEQ ID NO: 266); GGACACCCCAAAGAAGCTGA (SEQ ID NO: 267); CCAGTGCAATGGACAGAAGA (SEQ ID NO: 268); AGAAGAGGGAGCCTGCAAGT (SEQ ID NO: 269); GTGTTTGGGCCCTAGAGCGA (SEQ ID NO: 270); CATGTGCCTGGTGCAATGCA (SEQ ID NO: 271); TACAAAGAGGAAGATAAGTG (SEQ ID NO: 272); GTCACAGAATACACCACTAG (SEQ ID NO: 273); GGGTTACCCTGGACATGGAA (SEQ ID NO: 274); CATGGAAGGGTATTCACTCG (SEQ ID NO: 275); AGAGTGGCCTAGACAGGCTG (SEQ ID NO: 276); CATGCTGGACAGCTCGGCAG (SEQ ID NO: 277); AGTGAAAGAAGAGAAAATTC (SEQ ID NO: 278); TGGTAAGTCTAAGAAACCTA (SEQ ID NO: 279); CCCACAGCCTAACCACCCTA (SEQ ID NO: 280); AATATTTCAAAGCCCTAGGG (SEQ ID NO: 281); GCACTCGGAACAGGGTCTGG (SEQ ID NO: 282); AGATAGGAGCTCCAACAGTG (SEQ ID NO: 283); AAGTTAGAGCAGCCAGGAAA (SEQ ID NO: 284); TAGAGCAGCCAGGAAAGGGA (SEQ ID NO: 285); TGAATACCCTTCCATGTCCA (SEQ ID NO: 286); CCTGCATTGCACCAGGCACA (SEQ ID NO: 287); TCTAGGGCCCAAACACACCT (SEQ ID NO: 288); TCCCTCCATCTATCAAAAGG (SEQ ID NO: 289); AGCCCTGAGACAGAAGCAGG (SEQ ID NO: 290); GCCCTGAGACAGAAGCAGGT (SEQ ID NO: 291); AGGAGATGCAGTGATACGCA (SEQ ID NO: 292); ACAATACCAAGGGTATCCGG (SEQ ID NO: 293); TGATAAAGAAAACAAAGTGA (SEQ ID NO: 294); AAAGAAAACAAAGTGAGGGA (SEQ ID NO: 295); GTGGCAAGTGGAGAAATTGA (SEQ ID NO: 296); CAAGTGGAGAAATTGAGGGA (SEQ ID NO: 297); GTGGTGATGATTGCAGCTGG (SEQ ID NO: 298); CTATGTGCCTGACACACAGG (SEQ ID NO: 299); GGGTTGGACCAGGAAAGAGG (SEQ ID NO: 300); GATGCCTGGAAAAGGAAAGA (SEQ ID NO: 301); TAGTATGCACCTGCAAGAGG (SEQ ID NO: 302); TATGCACCTGCAAGAGGCGG (SEQ ID NO: 303); AGGGGAAGAAGAGAAGCAGA (SEQ ID NO: 304); GCTGAATCAAGAGACAAGCG (SEQ ID NO: 305); AAGCAAATAAATCTCCTGGG (SEQ ID NO: 306); AGATGAGTGCTAGAGACTGG (SEQ ID NO: 307); and CTGATGGTTGAGCACAGCAG (SEQ ID NO: 308). In embodiments, the guide RNAs are: AATCGAGAAGCGACTCGACA (SEQ ID NO: 425), and tgccctgcaggggagtgagc (SEQ ID NO: 426). In embodiments, the guide RNAs are gaagcgactcgacatggagg (SEQ ID NO: 427) and cctgcaggggagtgagcagc (SEQ ID NO: 428). In embodiments, guide RNAs (gRNAs) for targeting human genomic safe harbor sites using any of the gRNA-based targeting elements, e.g., without limitation dCas, in areas of open chromatin are as shown in TABLE 3A-3F. In embodiments, guide RNAs (gRNAs) for targeting human genomic safe harbor sites using any of the gRNA-based targeting elements, e.g., without limitation dCas, in areas of open chromatin are as shown in TABLE 3A:
Figure imgf000069_0001
Figure imgf000070_0001
Figure imgf000071_0001
Figure imgf000072_0001
Figure imgf000073_0001
Figure imgf000074_0001
Figure imgf000075_0001
In embodiments, gRNAs for targeting human genomic safe harbor sites using any of the gRNA-based targeting elements, e.g., without limitation dCas, to the TTAA site in hROSA26 (e.g., hg38 chr3:9,396,133-9,396,305) are shown in TABLE 3B:
Figure imgf000076_0001
Figure imgf000077_0001
In embodiments, gRNAs for targeting human genomic safe harbor sites using any of the gRNA-based targeting elements, e.g., without limitation dCas, to the AAVS1 (e.g., hg38 chr19:55,112,851-55,113,324) are shown in TABLE 3C:
Figure imgf000077_0002
Figure imgf000078_0001
In embodiments, gRNAs for targeting human genomic safe harbor sites using any of the gRNA-based targeting elements, e.g., without limitation dCas, to Chromosome 4 (e.g., hg38 chr4:30,793,534-30,875,476 or hg38 chr4:30,793,533-30,793,537 (9677); chr4:30,875,472-30,875,476 (8948)) are shown in TABLE 3D:
Figure imgf000078_0002
Figure imgf000079_0001
In embodiments, gRNAs for targeting human genomic safe harbor sites using any of the gRNA-based targeting elements, e.g., without limitation dCas, to Chromosome 22 (e.g., hg38 chr22:35,370,000-35,380,000 or hg38 chr22:35,373,912-35,373,916 (861); chr22:35,377,843-35,377,847 (1153)) are shown in TABLE 3E:
Figure imgf000079_0002
Figure imgf000080_0001
Figure imgf000081_0001
In embodiments, gRNAs for targeting human genomic safe harbor sites using any of the gRNA-based targeting elements, e.g., without limitation dCas, to Chromosome X (e.g., hg38 chrX:134,419,661-134,541,172 or hg38 chrX:134,476,304-134,476,307 (85); chrX:134,476,337-134,476,340 (51)) are shown in TABLE 3F:
Figure imgf000081_0002
Figure imgf000082_0001
In embodiments, the gRNA comprises one or more of the sequences outlined herein or a variant sequence having at least about 10 mutations, or at least about 9 mutations, or at least about 8 mutations, or at least about 7 mutations, or at least about 6 mutations, or at least about 5 mutations, or at least about 4 mutations, or at least about 3 mutations, or at least about 2 mutations, or at least about 1 mutation. In embodiments, a Cas-based targeting element comprises Cas12 or a variant thereof, e.g., without limitation, Cas12a (e.g., dCas12a), or Cas12j (e.g., dCas12j), or Cas12k (e.g., dCas12k). In embodiments, the targeting element comprises a Cas12 enzyme guide RNA complex. In embodiments, comprises a nuclease-deficient dCas12 guide RNA complex, optionally dCas12j guide RNA complex or dCas12a guide RNA complex. In embodiments, the targeting element is selected from a zinc finger (ZF), catalytically inactive Zinc finger, transcription activator-like effector (TALE), meganuclease, and clustered regularly interspaced short palindromic repeat (CRISPR)- associated protein, any of which are, in embodiments, catalytically inactive. In embodiments, the CRISPR-associated protein is selected from Cas9, CasX, CasY, Cas12a (Cpf1), and gRNA complexes thereof. In embodiments, the CRISPR-associated protein is selected from Cas9, xCas9, Cas 6, Cas7, Cas8, Cas12a (Cpf1), Cas13a, Cas14, CasX, CasY, a Class 1 Cas protein, a Class 2 Cas protein, MAD7, MG1 nuclease, MG2 nuclease, MG3 nuclease, or catalytically inactive forms thereof, and gRNA complexes thereof. In embodiments, the mobile element enzyme is capable of inserting a donor DNA at a TA dinucleotide site or a TTAA tetranucleotide site in a GSHS of a nucleic acid molecule. The mobile element enzyme is suitable for causing insertion of the donor DNA in a GSHS when contacted with a biological cell. In embodiments, the targeting element is suitable for directing the mobile element enzyme to the GSHS sequence. In embodiments, the targeting element comprises transcription activator-like effector (TALE) DNA binding domain (DBD). The TALE DBD comprises one or more repeat sequences. For example, in embodiments, the TALE DBD comprises about 14, or about 15, or about, 16, or about 17, or about 18, or about 18.5 repeat sequences. In embodiments, the TALE DBD repeat sequences comprise 33 or 34 amino acids. In embodiments, the one or more of the TALE DBD repeat sequences comprise a repeat variable di-residue (RVD) at residue 12 or 13 of the 33 or 34 amino acids. In embodiments, the targeting element (e.g., TALE or Cas (e.g., Cas9 or Cas12, or variants thereof) DBDs cause the mammalian mobile element enzyme to bind specifically to human GSHS. In embodiments, the TALEs or Cas DBDs sequester the mobile element enzyme to GSHS and promote transposition to nearby TA dinucleotide or a TTAA tetranucleotide sites which can be located in proximity to the repeat variable di-residues (RVD) TALE or gRNA nucleotide sequences. The GSHS regions are located in open chromatin sites that are susceptible to mobile element enzyme activity. Accordingly, the mammalian mobile element enzyme does not only operate based on its ability to recognize TA or TTAA sites, but it also directs a donor DNA (having a transgene) to specific locations in proximity to a TALE or Cas DBD. The chimeric mobile element enzyme in accordance with embodiments of the present disclosure has negligible risk of genotoxicity and exhibits superior features as compared to existing gene therapies. In embodiments, a chimeric mobile element enzyme is mutated to be characterized by reduced or inhibited binding of off-target sequences and consequently reliant on a DBD fused thereto, such as a TALE or Cas DBD, for transposition. The described cells, compositions, and methods allow reducing vector and transgene insertions that increase a mutagenic risk. The described cells and methods make use of a gene transfer system that reduces genotoxicity compared to viral- and nuclease-mediated gene therapies. The dual system is designed to avoid the persistence of an active mobile element enzyme and efficiently transfect human cell lines without significant cytotoxicity. In embodiments, TALE or Cas DBDs are customizable, such as a TALE or Cas DBDs is selected for targeting a specific genomic location. In embodiments, the genomic location is in proximity to a TA dinucleotide site or a TTAA (SEQ ID NO: 440) tetranucleotide site. Embodiments of the present disclosure make use of the ability of TALE or Cas or dCas9/gRNA DBDs to target specific sites in a host genome. The DNA targeting ability of a TALE or Cas DBD or dCas9/gRNA DBD is provided by TALE repeat sequences (e.g., modular arrays) or gRNA which are linked together to recognize flanking DNA sequences. Each TALE or gRNA can recognize certain base pair(s) or residue(s). TALE nucleases (TALENs) are a known tool for genome editing and introducing targeted double-stranded breaks. TALENs comprise endonucleases, such as FokI nuclease domain, fused to a customizable DBD. This DBD is composed of highly conserved repeats from TALEs, which are proteins secreted by Xanthomonas bacteria to alter transcription of genes in host plant cells. The DBD includes a repeated highly conserved 33–34 amino acid sequence with divergent 12th and 13th amino acids. These two positions, referred to as the RVD, are highly variable and show a strong correlation with specific base pair or nucleotide recognition. This straightforward relationship between amino acid sequence and DNA recognition has allowed for the engineering of specific DBDs by selecting a combination of repeat segments containing the appropriate RVDs. Boch et al. Nature Biotechnology.2011; 29 (2): 135–6. Accordingly, TALENs can be readily designed using a “protein-DNA code” that relates modular DNA-binding TALE repeat domains to individual bases in a target-binding site. See Joung et al. Nat Rev Mol Cell Biol.2013;14(1):49-55. doi:10.1038/nrm3486. The following table, for example, shows such code:
Figure imgf000084_0001
It has been demonstrated that TALENs can be used to target essentially any DNA sequence of interest in human cell. Miller et al. Nat Biotechnol.2011;29:143–148. Guidelines for selection of potential target sites and for use of particular TALE repeat domains (harboring NH residues at the hypervariable positions) for recognition of G bases have been proposed. See Streubel et al. Nat Biotechnol.2012;30:593–595. Accordingly, in embodiments, the TALE DBD comprises one or more repeat sequences. In embodiments, the TALE DBD comprises about 15, or about, 16, or about 17, or about 18, or about 18.5 repeat sequences. In embodiments, the TALE DBD repeat sequences comprise 33 or 34 amino acids. In embodiments, the one or more of the TALE DBD repeat sequences comprise an RVD at residue 12 or 13 of the 33 or 34 amino acids. The RVD can recognize certain base pair(s) or residue(s). In embodiments, the RVD recognizes one base pair in the nucleic acid molecule. In embodiments, the RVD recognizes a C residue in the nucleic acid molecule and is selected from HD, N(gap), HA, ND, and HI. In embodiments, the RVD recognizes a G residue in the nucleic acid molecule and is selected from NN, NH, NK, HN, and NA. In embodiments, the RVD recognizes an A residue in the nucleic acid molecule and is selected from NI and NS. In embodiments, the RVD recognizes a T residue in the nucleic acid molecule and is selected from NG, HG, H(gap), and IG. In embodiments, the GSHS is in an open chromatin location in a chromosome. In embodiments, the GSHS is selected from adeno-associated virus site 1 (AAVS1), chemokine (C-C motif) receptor 5 (CCR5) gene, HIV-1 coreceptor; and human Rosa26 locus. In embodiments, the GSHS is located on human chromosome 2, 4, 6, 10, 11, 17, 22, or X. In embodiments, the GSHS is selected from TALC1, TALC2, TALC3, TALC4, TALC5, TALC7, TALC8, AVS1, AVS2, AVS3, ROSA1, ROSA2, TALER1, TALER2, TALER3, TALER4, TALER5, SHCHR2-1, SHCHR2-2, SHCHR2-3, SHCHR2-4, SHCHR4-1, SHCHR4-2, SHCHR4-3, SHCHR6-1, SHCHR6-2, SHCHR6-3, SHCHR6-4, SHCHR10-1, SHCHR10-2, SHCHR10-3, SHCHR10-4, SHCHR10-5, SHCHR11-1, SHCHR11-2, SHCHR11-3, SHCHR17-1, SHCHR17-2, SHCHR17-3, and SHCHR17-4. In embodiments, the GSHS comprises one or more of TGGCCGGCCTGACCACTGG (SEQ ID NO: 23), TGAAGGCCTGGCCGGCCTG (SEQ ID NO: 24), TGAGCACTGAAGGCCTGGC (SEQ ID NO: 25), TCCACTGAGCACTGAAGGC (SEQ ID NO: 26), TGGTTTCCACTGAGCACTG (SEQ ID NO: 27), TGGGGAAAATGACCCAACA (SEQ ID NO: 28), TAGGACAGTGGGGAAAATG (SEQ ID NO: 29), TCCAGGGACACGGTGCTAG (SEQ ID NO: 30), TCAGAGCCAGGAGTCCTGG (SEQ ID NO: 31), TCCTTCAGAGCCAGGAGTC (SEQ ID NO: 32), TCCTCCTTCAGAGCCAGGA (SEQ ID NO: 33), TCCAGCCCCTCCTCCTTCA (SEQ ID NO: 34), TCCGAGCTTGACCCTTGGA (SEQ ID NO: 35), TGGTTTCCGAGCTTGACCC (SEQ ID NO: 36), TGGGGTGGTTTCCGAGCTT (SEQ ID NO: 37), TCTGCTGGGGTGGTTTCCG (SEQ ID NO: 38), TGCAGAGTATCTGCTGGGG (SEQ ID NO: 39), CCAATCCCCTCAGT (SEQ ID NO: 40), CAGTGCTCAGTGGAA (SEQ ID NO: 41), GAAACATCCGGCGACTCA (SEQ ID NO: 42), TCGCCCCTCAAATCTTACA (SEQ ID NO: 43), TCAAATCTTACAGCTGCTC (SEQ ID NO: 44), TCTTACAGCTGCTCACTCC (SEQ ID NO: 45), TACAGCTGCTCACTCCCCT (SEQ ID NO: 46), TGCTCACTCCCCTGCAGGG (SEQ ID NO: 47), TCCCCTGCAGGGCAACGCC (SEQ ID NO: 48), TGCAGGGCAACGCCCAGGG (SEQ ID NO: 49), TCTCGATTATGGGCGGGAT (SEQ ID NO: 50), TCGCTTCTCGATTATGGGC (SEQ ID NO: 51), TGTCGAGTCGCTTCTCGAT (SEQ ID NO: 52), TCCATGTCGAGTCGCTTCT (SEQ ID NO: 53), TCGCCTCCATGTCGAGTCG (SEQ ID NO: 54), TCGTCATCGCCTCCATGTC (SEQ ID NO: 55), TGATCTCGTCATCGCCTCC (SEQ ID NO: 56), GCTTCAGCTTCCTA (SEQ ID NO: 57), CTGTGATCATGCCA (SEQ ID NO: 58), ACAGTGGTACACACCT (SEQ ID NO: 59), CCACCCCCCACTAAG (SEQ ID NO: 60), CATTGGCCGGGCAC (SEQ ID NO: 61), GCTTGAACCCAGGAGA (SEQ ID NO: 62), ACACCCGATCCACTGGG (SEQ ID NO: 63), GCTGCATCAACCCC (SEQ ID NO: 64), GCCACAAACAGAAATA (SEQ ID NO: 65), GGTGGCTCATGCCTG (SEQ ID NO: 66), GATTTGCACAGCTCAT (SEQ ID NO: 67), AAGCTCTGAGGAGCA (SEQ ID NO: 68), CCCTAGCTGTCCC (SEQ ID NO: 69), GCCTAGCATGCTAG (SEQ ID NO: 70), ATGGGCTTCACGGAT (SEQ ID NO: 71), GAAACTATGCCTGC (SEQ ID NO: 72), GCACCATTGCTCCC (SEQ ID NO: 73), GACATGCAACTCAG (SEQ ID NO: 74), ACACCACTAGGGGT (SEQ ID NO: 75), GTCTGCTAGACAGG (SEQ ID NO: 76), GGCCTAGACAGGCTG (SEQ ID NO: 77), GAGGCATTCTTATCG (SEQ ID NO: 78), GCCTGGAAACGTTCC (SEQ ID NO: 79), GTGCTCTGACAATA (SEQ ID NO: 80), GTTTTGCAGCCTCC (SEQ ID NO: 81), ACAGCTGTGGAACGT (SEQ ID NO: 82), GGCTCTCTTCCTCCT (SEQ ID NO: 83), CTATCCCAAAACTCT (SEQ ID NO: 84), GAAAAACTATGTAT (SEQ ID NO: 85), AGGCAGGCTGGTTGA (SEQ ID NO: 86), CAATACAACCACGC (SEQ ID NO: 87), ATGACGGACTCAACT (SEQ ID NO: 88), CACAACATTTGTAA (SEQ ID NO: 89), and ATTTCCAGTGCACA (SEQ ID NO: 90). In embodiments, the TALE DBD binds to one of TGGCCGGCCTGACCACTGG (SEQ ID NO: 23), TGAAGGCCTGGCCGGCCTG (SEQ ID NO: 24), TGAGCACTGAAGGCCTGGC (SEQ ID NO: 25), TCCACTGAGCACTGAAGGC (SEQ ID NO: 26), TGGTTTCCACTGAGCACTG (SEQ ID NO: 27), TGGGGAAAATGACCCAACA (SEQ ID NO: 28), TAGGACAGTGGGGAAAATG (SEQ ID NO: 29), TCCAGGGACACGGTGCTAG (SEQ ID NO: 30), TCAGAGCCAGGAGTCCTGG (SEQ ID NO: 31), TCCTTCAGAGCCAGGAGTC (SEQ ID NO: 32), TCCTCCTTCAGAGCCAGGA (SEQ ID NO: 33), TCCAGCCCCTCCTCCTTCA (SEQ ID NO: 34), TCCGAGCTTGACCCTTGGA (SEQ ID NO: 35), TGGTTTCCGAGCTTGACCC (SEQ ID NO: 36), TGGGGTGGTTTCCGAGCTT (SEQ ID NO: 37), TCTGCTGGGGTGGTTTCCG (SEQ ID NO: 38), TGCAGAGTATCTGCTGGGG (SEQ ID NO: 39), CCAATCCCCTCAGT (SEQ ID NO: 40), CAGTGCTCAGTGGAA (SEQ ID NO: 41), GAAACATCCGGCGACTCA (SEQ ID NO: 42), TCGCCCCTCAAATCTTACA (SEQ ID NO: 43), TCAAATCTTACAGCTGCTC (SEQ ID NO: 44), TCTTACAGCTGCTCACTCC (SEQ ID NO: 45), TACAGCTGCTCACTCCCCT (SEQ ID NO: 46), TGCTCACTCCCCTGCAGGG (SEQ ID NO: 47), TCCCCTGCAGGGCAACGCC (SEQ ID NO: 48), TGCAGGGCAACGCCCAGGG (SEQ ID NO: 49), TCTCGATTATGGGCGGGAT (SEQ ID NO: 50), TCGCTTCTCGATTATGGGC (SEQ ID NO: 51), TGTCGAGTCGCTTCTCGAT (SEQ ID NO: 52), TCCATGTCGAGTCGCTTCT (SEQ ID NO: 53), TCGCCTCCATGTCGAGTCG (SEQ ID NO: 54), TCGTCATCGCCTCCATGTC (SEQ ID NO: 55), TGATCTCGTCATCGCCTCC (SEQ ID NO: 56), GCTTCAGCTTCCTA (SEQ ID NO: 57), CTGTGATCATGCCA (SEQ ID NO: 58), ACAGTGGTACACACCT (SEQ ID NO: 59), CCACCCCCCACTAAG (SEQ ID NO: 60), CATTGGCCGGGCAC (SEQ ID NO: 61), GCTTGAACCCAGGAGA (SEQ ID NO: 62), ACACCCGATCCACTGGG (SEQ ID NO: 63), GCTGCATCAACCCC (SEQ ID NO: 64), GCCACAAACAGAAATA (SEQ ID NO: 65), GGTGGCTCATGCCTG (SEQ ID NO: 66), GATTTGCACAGCTCAT (SEQ ID NO: 67), AAGCTCTGAGGAGCA (SEQ ID NO: 68), CCCTAGCTGTCCC (SEQ ID NO: 69), GCCTAGCATGCTAG (SEQ ID NO: 70), ATGGGCTTCACGGAT (SEQ ID NO: 71), GAAACTATGCCTGC (SEQ ID NO: 72), GCACCATTGCTCCC (SEQ ID NO: 73), GACATGCAACTCAG (SEQ ID NO: 74), ACACCACTAGGGGT (SEQ ID NO: 75), GTCTGCTAGACAGG (SEQ ID NO: 76), GGCCTAGACAGGCTG (SEQ ID NO: 77), GAGGCATTCTTATCG (SEQ ID NO: 78), GCCTGGAAACGTTCC (SEQ ID NO: 79), GTGCTCTGACAATA (SEQ ID NO: 80), GTTTTGCAGCCTCC (SEQ ID NO: 81), ACAGCTGTGGAACGT (SEQ ID NO: 82), GGCTCTCTTCCTCCT (SEQ ID NO: 83), CTATCCCAAAACTCT (SEQ ID NO: 84), GAAAAACTATGTAT (SEQ ID NO: 85), AGGCAGGCTGGTTGA (SEQ ID NO: 86), CAATACAACCACGC (SEQ ID NO: 87), ATGACGGACTCAACT (SEQ ID NO: 88), CACAACATTTGTAA (SEQ ID NO: 89), and ATTTCCAGTGCACA (SEQ ID NO: 90). In embodiments, the TALE DBD comprises one or more of: NH NH HD HD NH NH HD HD NG NH NI HD HD NI HD NG NH NH, NH NI NI NH NH HD HD NG NH NH HD HD NH NH HD HD NG NH, NH NI NH HD NI HD NG NH NI NI NH NH HD HD NG NH NH HD, HD HD NI HD NG NH NI NH HD NI HD NG NH NI NI NH NH HD, NH NH NG NG NG HD HD NI HD NG NH NI NH HD NI HD NG NH, NH NH NH NH NI NI NI NI NG NH NI HD HD HD NI NI HD NI, NI NH NH NI HD NI NH NG NH NH NH NH NI NI NI NI NG NH, HD HD NI NH NH NH NI HD NI HD NH NH NG NH HD NG NI NH, HD NI NH NI NH HD HD NI NH NH NI NH NG HD HD NG NH NH, HD HD NG NG HD NI NH NI NH HD HD NI NH NH NI NH NG HD, HD HD NG HD HD NG NG HD NI NH NI NH HD HD NI NH NH NI, HD HD NI NH HD HD HD HD NG HD HD NG HD HD NG NG HD NI, HD HD NH NI NH HD NG NG NH NI HD HD HD NG NG NH NH NI, NH NH NG NG NG HD HD NH NI NH HD NG NG NH NI HD HD HD, NH NH NH NH NG NH NH NG NG NG HD HD NH NI NH HD NG NG, HD NG NH HD NG NH NH NH NH NG NH NH NG NG NG HD HD NH, NH HD NI NH NI NH NG NI NG HD NG NH HD NG NH NH NH NH, HD HD NI NI NG HD HD HD HD NG HD NI NH NG, HD NI NH NG NH HD NG HD NI NH NG NH NH NI NI, NH NI NI NI HD NI NG HD HD NH NH HD NH NI HD NG HD NI, HD NH HD HD HD HD NG HD NI NI NI NG HD NG NG NI HD NI, HD NI NI NI NG HD NG NG NI HD NI NH HD NG NH HD NG HD, HD NG NG NI HD NI NH HD NG NH HD NG HD NI HD NG HD HD, NI HD NI NH HD NG NH HD NG HD NI HD NG HD HD HD HD NG, NH HD NG HD NI HD NG HD HD HD HD NG NH HD NI NH NH NH, HD HD HD HD NG NH HD NI NH NH NH HD NI NI HD NH HD HD, NH HD NI NH NH NH HD NI NI HD NH HD HD HD NI NH NH NH, HD NG HD NH NI NG NG NI NG NH NH NH HD NH NH NH NI NG, HD NH HD NG NG HD NG HD NH NI NG NG NI NG NH NH NH HD, NH NG HD NH NI NH NG HD NH HD NG NG HD NG HD NH NI NG, HD HD NI NG NH NG HD NH NI NH NG HD NH HD NG NG HD NG, HD NH HD HD NG HD HD NI NG NH NG HD NH NI NH NG HD NH, HD NH NG HD NI NG HD NH HD HD NG HD HD NI NG NH NG HD, NH NI NG HD NG HD NH NG HD NI NG HD NH HD HD NG HD HD, NH HD NG NG HD NI NH HD NG NG HD HD NG NI, HD NG NK NG NH NI NG HD NI NG NH HD HD NI, NI HD NI NN NG NN NN NG NI HD NI HD NI HD HD NG, HD HD NI HD HD HD HD HD HD NI HD NG NI NI NN, HD NI NG NG NN NN HD HD NN NN NN HD NI HD, NN HD NG NG NN NI NI HD HD HD NI NN NN NI NN NI, NI HD NI HD HD HD NN NI NG HD HD NI HD NG NN NN NN, NN HD NG NN HD NI NG HD NI NI HD HD HD HD, NN NN HD NI HD NN NI NI NI HD NI HD HD HD NG HD HD, NN NN NG NN NN HD NG HD NI NG NN HD HD NG NN, NN NI NG NG NG NN HD NI HD NI NN HD NG HD NI NG, NI NI NH HD NG HD NG NH NI NH NH NI NH HD, HD HD HD NG NI NK HD NG NH NG HD HD HD HD, NH HD HD NG NI NH HD NI NG NH HD NG NI NH, NI NG NH NH NH HD NG NG HD NI HD NH NH NI NG, NH NI NI NI HD NG NI NG NH HD HD NG NH HD, NH HD NI HD HD NI NG NG NH HD NG HD HD HD, NH NI HD NI NG NH HD NI NI HD NG HD NI NH, NI HD NI HD HD NI HD NG NI NH NH NH NH NG, NH NG HD NG NH HD NG NI NH NI HD NI NH NH, NH NH HD HD NG NI NH NI HD NI NH NH HD NG NH, NH NI NH NH HD NI NG NG HD NG NG NI NG HD NH, NN HD HD NG NN NN NI NI NI HD NN NG NG HD HD, NN NG NN HD NG HD NG NN NI HD NI NI NG NI, NN NG NG NG NG NN HD NI NN HD HD NG HD HD, NI HD NI NN HD NG NN NG NN NN NI NI HD NN NG, HD NI NI NN NI HD HD NN NI NN HD NI HD NG NN HD NG NN, HD NG NI NG HD HD HD NI NI NI NI HD NG HD NG, NH NI NI NI NI NI HD NG NI NG NH NG NI NG, NI NH NH HD NI NH NH HD NG NH NH NG NG NH NI, HD NI NI NG NI HD NI NI HD HD NI HD NN HD, NI NG NN NI HD NN NN NI HD NG HD NI NI HD NG, HD NI HD NI NI HD NI NG NG NG NN NG NI NI, and NI NG NG NG HD HD NI NN NG NN HD NI HD NI. In embodiments, the TALE DBD comprises one or more of the sequences outlined herein or a variant sequence having at least about 90%, or at least about 93%, or at least about 95%, or at least about 97%, or at least about 98%, or at least about 99% identity thereto, or at least about 10 mutations, or at least about 9 mutations, or at least about 8 mutations, or at least about 7 mutations, or at least about 6 mutations, or at least about 5 mutations, or at least about 4 mutations, or at least about 3 mutations, or at least about 2 mutations, or at least about 1 mutation. In embodiments, the GSHS and the TALE DBD sequences are selected from: TGGCCGGCCTGACCACTGG (SEQ ID NO: 23) and NH NH HD HD NH NH HD HD NG NH NI HD HD NI HD NG NH NH; TGAAGGCCTGGCCGGCCTG (SEQ ID NO: 24) and NH NI NI NH NH HD HD NG NH NH HD HD NH NH HD HD NG NH; TGAGCACTGAAGGCCTGGC (SEQ ID NO: 25) and NH NI NH HD NI HD NG NH NI NI NH NH HD HD NG NH NH HD; TCCACTGAGCACTGAAGGC (SEQ ID NO: 26) and HD HD NI HD NG NH NI NH HD NI HD NG NH NI NI NH NH HD; TGGTTTCCACTGAGCACTG (SEQ ID NO: 27) and NH NH NG NG NG HD HD NI HD NG NH NI NH HD NI HD NG NH; TGGGGAAAATGACCCAACA (SEQ ID NO: 28) and NH NH NH NH NI NI NI NI NG NH NI HD HD HD NI NI HD NI; TAGGACAGTGGGGAAAATG (SEQ ID NO: 29) and NI NH NH NI HD NI NH NG NH NH NH NH NI NI NI NI NG NH; TCCAGGGACACGGTGCTAG (SEQ ID NO: 30) and HD HD NI NH NH NH NI HD NI HD NH NH NG NH HD NG NI NH; TCAGAGCCAGGAGTCCTGG (SEQ ID NO: 31) and HD NI NH NI NH HD HD NI NH NH NI NH NG HD HD NG NH NH; TCCTTCAGAGCCAGGAGTC (SEQ ID NO: 32) and HD HD NG NG HD NI NH NI NH HD HD NI NH NH NI NH NG HD; TCCTCCTTCAGAGCCAGGA (SEQ ID NO: 33) and HD HD NG HD HD NG NG HD NI NH NI NH HD HD NI NH NH NI; TCCAGCCCCTCCTCCTTCA (SEQ ID NO: 34) and HD HD NI NH HD HD HD HD NG HD HD NG HD HD NG NG HD NI; TCCGAGCTTGACCCTTGGA (SEQ ID NO: 35) and HD HD NH NI NH HD NG NG NH NI HD HD HD NG NG NH NH NI; TGGTTTCCGAGCTTGACCC (SEQ ID NO: 36) and NH NH NG NG NG HD HD NH NI NH HD NG NG NH NI HD HD HD; TGGGGTGGTTTCCGAGCTT (SEQ ID NO: 37) and NH NH NH NH NG NH NH NG NG NG HD HD NH NI NH HD NG NG; TCTGCTGGGGTGGTTTCCG (SEQ ID NO: 38) and HD NG NH HD NG NH NH NH NH NG NH NH NG NG NG HD HD NH; TGCAGAGTATCTGCTGGGG (SEQ ID NO: 39) and NH HD NI NH NI NH NG NI NG HD NG NH HD NG NH NH NH NH; CCAATCCCCTCAGT (SEQ ID NO: 40) and HD HD NI NI NG HD HD HD HD NG HD NI NH NG; CAGTGCTCAGTGGAA (SEQ ID NO: 41) and HD NI NH NG NH HD NG HD NI NH NG NH NH NI NI; GAAACATCCGGCGACTCA (SEQ ID NO: 42) and NH NI NI NI HD NI NG HD HD NH NH HD NH NI HD NG HD NI; TCGCCCCTCAAATCTTACA (SEQ ID NO: 43) and HD NH HD HD HD HD NG HD NI NI NI NG HD NG NG NI HD NI; TCAAATCTTACAGCTGCTC (SEQ ID NO: 44) and HD NI NI NI NG HD NG NG NI HD NI NH HD NG NH HD NG HD; TCTTACAGCTGCTCACTCC (SEQ ID NO: 45) and HD NG NG NI HD NI NH HD NG NH HD NG HD NI HD NG HD HD; TACAGCTGCTCACTCCCCT (SEQ ID NO: 46) and NI HD NI NH HD NG NH HD NG HD NI HD NG HD HD HD HD NG; TGCTCACTCCCCTGCAGGG (SEQ ID NO: 47) and NH HD NG HD NI HD NG HD HD HD HD NG NH HD NI NH NH NH; TCCCCTGCAGGGCAACGCC (SEQ ID NO: 48) and HD HD HD HD NG NH HD NI NH NH NH HD NI NI HD NH HD HD; TGCAGGGCAACGCCCAGGG (SEQ ID NO: 49) and NH HD NI NH NH NH HD NI NI HD NH HD HD HD NI NH NH NH; TCTCGATTATGGGCGGGAT (SEQ ID NO: 50) and HD NG HD NH NI NG NG NI NG NH NH NH HD NH NH NH NI NG; TCGCTTCTCGATTATGGGC (SEQ ID NO: 51) and HD NH HD NG NG HD NG HD NH NI NG NG NI NG NH NH NH HD; TGTCGAGTCGCTTCTCGAT (SEQ ID NO: 52) and NH NG HD NH NI NH NG HD NH HD NG NG HD NG HD NH NI NG; TCCATGTCGAGTCGCTTCT (SEQ ID NO: 53) and HD HD NI NG NH NG HD NH NI NH NG HD NH HD NG NG HD NG; TCGCCTCCATGTCGAGTCG (SEQ ID NO: 54) and HD NH HD HD NG HD HD NI NG NH NG HD NH NI NH NG HD NH; TCGTCATCGCCTCCATGTC (SEQ ID NO: 55) and HD NH NG HD NI NG HD NH HD HD NG HD HD NI NG NH NG HD; TGATCTCGTCATCGCCTCC (SEQ ID NO: 56) and NH NI NG HD NG HD NH NG HD NI NG HD NH HD HD NG HD HD; GCTTCAGCTTCCTA (SEQ ID NO: 57) and NH HD NG NG HD NI NH HD NG NG HD HD NG NI; CTGTGATCATGCCA (SEQ ID NO: 58) and HD NG NK NG NH NI NG HD NI NG NH HD HD NI; ACAGTGGTACACACCT (SEQ ID NO: 59) and NI HD NI NN NG NN NN NG NI HD NI HD NI HD HD NG; CCACCCCCCACTAAG (SEQ ID NO: 60) and HD HD NI HD HD HD HD HD HD NI HD NG NI NI NN; CATTGGCCGGGCAC (SEQ ID NO: 61) and HD NI NG NG NN NN HD HD NN NN NN HD NI HD; GCTTGAACCCAGGAGA (SEQ ID NO: 62) and NN HD NG NG NN NI NI HD HD HD NI NN NN NI NN NI; ACACCCGATCCACTGGG (SEQ ID NO: 63) and NI HD NI HD HD HD NN NI NG HD HD NI HD NG NN NN NN; GCTGCATCAACCCC (SEQ ID NO: 64) and NN HD NG NN HD NI NG HD NI NI HD HD HD HD; GCCACAAACAGAAATA (SEQ ID NO: 65) and NN NN HD NI HD NN NI NI NI HD NI HD HD HD NG HD HD; GGTGGCTCATGCCTG (SEQ ID NO: 66) and NN NN NG NN NN HD NG HD NI NG NN HD HD NG NN; GATTTGCACAGCTCAT (SEQ ID NO: 67) and NN NI NG NG NG NN HD NI HD NI NN HD NG HD NI NG; AAGCTCTGAGGAGCA (SEQ ID NO: 68) and NI NI NH HD NG HD NG NH NI NH NH NI NH HD; CCCTAGCTGTCCC (SEQ ID NO: 69) and HD HD HD NG NI NK HD NG NH NG HD HD HD HD; GCCTAGCATGCTAG (SEQ ID NO: 70) and NH HD HD NG NI NH HD NI NG NH HD NG NI NH; ATGGGCTTCACGGAT (SEQ ID NO: 71) and NI NG NH NH NH HD NG NG HD NI HD NH NH NI NG; GAAACTATGCCTGC (SEQ ID NO: 72) and NH NI NI NI HD NG NI NG NH HD HD NG NH HD; GCACCATTGCTCCC (SEQ ID NO: 73) and NH HD NI HD HD NI NG NG NH HD NG HD HD HD; GACATGCAACTCAG (SEQ ID NO: 74) and NH NI HD NI NG NH HD NI NI HD NG HD NI NH; ACACCACTAGGGGT (SEQ ID NO: 75) and NI HD NI HD HD NI HD NG NI NH NH NH NH NG; GTCTGCTAGACAGG (SEQ ID NO: 76) and NH NG HD NG NH HD NG NI NH NI HD NI NH NH; GGCCTAGACAGGCTG (SEQ ID NO: 77) and NH NH HD HD NG NI NH NI HD NI NH NH HD NG NH; GAGGCATTCTTATCG (SEQ ID NO: 78) and NH NI NH NH HD NI NG NG HD NG NG NI NG HD NH; GCCTGGAAACGTTCC (SEQ ID NO: 79) and NN HD HD NG NN NN NI NI NI HD NN NG NG HD HD; GTGCTCTGACAATA (SEQ ID NO: 80) and NN NG NN HD NG HD NG NN NI HD NI NI NG NI; GTTTTGCAGCCTCC (SEQ ID NO: 81) and NN NG NG NG NG NN HD NI NN HD HD NG HD HD; ACAGCTGTGGAACGT (SEQ ID NO: 82) and NI HD NI NN HD NG NN NG NN NN NI NI HD NN NG; GGCTCTCTTCCTCCT (SEQ ID NO: 83) and HD NI NI NN NI HD HD NN NI NN HD NI HD NG NN HD NG NN; CTATCCCAAAACTCT (SEQ ID NO: 84) and HD NG NI NG HD HD HD NI NI NI NI HD NG HD NG; GAAAAACTATGTAT (SEQ ID NO: 85) and NH NI NI NI NI NI HD NG NI NG NH NG NI NG; AGGCAGGCTGGTTGA (SEQ ID NO: 86) and NI NH NH HD NI NH NH HD NG NH NH NG NG NH NI; CAATACAACCACGC (SEQ ID NO: 87) and HD NI NI NG NI HD NI NI HD HD NI HD NN HD; ATGACGGACTCAACT (SEQ ID NO: 88) and NI NG NN NI HD NN NN NI HD NG HD NI NI HD NG; and CACAACATTTGTAA (SEQ ID NO: 89) and HD NI HD NI NI HD NI NG NG NG NN NG NI NI. In embodiments, the GSHS is within about 25, or about 50, or about 100, or about 150, or about 200, or about 300, or about 500 nucleotides of the TA dinucleotide site or TTAA (SEQ ID NO: 440) tetranucleotide site. Illustrative DNA binding codes for human genomic safe harbor in areas of open chromatin via TALEs, encompassed by various embodiments are provided in TABLE 4A-4F. In embodiments, there is provided a variant of the TALEs, encompassed by various embodiments are provided in TABLE 4A-4F, e.g., having a sequence having at least about 90%, or at least about 93%, or at least about 95%, or at least about 97%, or at least about 98%, or at least about 99% identity to any of the sequences in TABLE 4A-4F. Illustrative DNA binding codes for human genomic safe harbor in areas of open chromatin via TALEs, encompassed by various embodiments are provided in TABLE 4A:
Figure imgf000093_0001
Figure imgf000094_0001
Figure imgf000095_0001
Figure imgf000096_0001
Figure imgf000097_0001
In embodiments, TALEs for targeting human genomic safe harbor sites using any of the TALE-based targeting elements to the TTAA site in hROSA26 (e.g., hg38 chr3:9,396,133-9,396,305) are shown in TABLE 4B:
Figure imgf000097_0002
In embodiments, TALEs for targeting human genomic safe harbor sites using any of the TALE-based targeting elements to the AAVS1 (e.g., hg38 chr19:55,112,851-55,113,324) are shown in TABLE 4C:
Figure imgf000097_0003
Figure imgf000098_0001
In embodiments, TALEs for targeting human genomic safe harbor sites using any of the TALE-based targeting elements to Chromosome 4 (e.g., hg38 chr4:30,793,534-30,875,476 or hg38 chr4:30,793,533-30,793,537 (9677); chr4:30,875,472-30,875,476 (8948)) are shown in TABLE 4D:
Figure imgf000098_0002
Figure imgf000099_0001
In embodiments, TALEs for targeting human genomic safe harbor sites using any of the TALE-based targeting elements to Chromosome 22 (e.g., hg38 chr22:35,370,000-35,380,000 or hg38 chr22:35,373,912-35,373,916 (861); chr22:35,377,843-35,377,847 (1153)) are shown in TABLE 4E:
Figure imgf000099_0002
Figure imgf000100_0001
In embodiments, TALEs for targeting human genomic safe harbor sites using any of the TALE-based targeting elements to Chromosome X (e.g., hg38 chrX:134,419,661-134,541,172 or hg38 chrX:134,476,304-134,476,307 (85); chrX:134,476,337-134,476,340 (51)) are shown in TABLE 4F:
Figure imgf000100_0002
Figure imgf000101_0002
In embodiments, the mobile element enzyme is capable of inserting a donor DNA at a TA dinucleotide site. In embodiments, the mobile element enzyme is capable of inserting a donor DNA at a TTAA (SEQ ID NO: 440) tetranucleotide site. Illustrative DNA binding codes for human genomic safe harbor in areas of open chromatin via ZNFs, encompassed by various embodiments are provided in TABLE 5A-5E. In embodiments, there is provided a variant of the ZNFs, encompassed by various embodiments are provided in TABLE 5A-5E, e.g., having a sequence having at least about 90%, or at least about 93%, or at least about 95%, or at least about 97%, or at least about 98%, or at least about 99% identity to any of the sequences in TABLE 5A-5E. In embodiments, ZNFs for targeting human genomic safe harbor sites using any of the ZNF-based targeting elements to the TTAA site in hROSA26 (e.g., hg38 chr3:9,396,133-9,396,305) are shown in TABLE 5A:
Figure imgf000101_0001
Figure imgf000102_0001
In embodiments, ZNFs for targeting human genomic safe harbor sites using any of the ZNF-based targeting elements to the AAVS1 (e.g., hg38 chr19:55,112,851-55,113,324) are shown in TABLE 5B:
Figure imgf000102_0002
Figure imgf000103_0001
In embodiments, ZNFs for targeting human genomic safe harbor sites using any of the ZNF-based targeting elements to Chromosome 4 (e.g., hg38 chr4:30,793,534-30,875,476 or hg38 chr4:30,793,533-30,793,537 (9677); chr4:30,875,472-30,875,476 (8948)) are shown in TABLE 5C:
Figure imgf000103_0002
Figure imgf000104_0001
In embodiments, ZNFs for targeting human genomic safe harbor sites using any of the ZNF-based targeting elements to Chromosome 22 (e.g., hg38 chr22:35,370,000-35,380,000 or hg38 chr22:35,373,912-35,373,916 (861); chr22:35,377,843-35,377,847 (1153)) are shown in TABLE 5D:
Figure imgf000104_0002
Figure imgf000105_0001
Figure imgf000106_0002
In embodiments, ZNFs for targeting human genomic safe harbor sites using any of the ZNF-based targeting elements to Chromosome X (e.g., hg38 chrX:134,419,661-134,541,172 or hg38 chrX:134,476,304-134,476,307 (85); chrX:134,476,337-134,476,340 (51)) are shown in TABLE 5E:
Figure imgf000106_0001
In embodiments, the mobile element enzyme is capable of inserting a donor DNA at a TA dinucleotide site. In embodiments, the mobile element enzyme is capable of inserting a donor DNA at a TTAA (SEQ ID NO: 440) tetranucleotide site. In embodiments, the present disclosure relates to a system having nucleic acids encoding the enzyme and the donor DNA, respectively. FIGs.1A-1D show examples of a system in accordance with embodiments of the present disclosure. Linkers In embodiments, the targeting element comprises a nucleic acid binding component of the gene-editing system (targeting element). In embodiments, the enzyme capable of targeted genomic integration (e.g., without limitation, a chimeric mobile element enzyme) and the targeting element, e.g., nucleic acid binding component of the gene-editing system are fused or linked to one another. For example, in embodiments, the mobile element enzyme and the targeting element, e.g., nucleic acid binding component of the gene-editing system are fused or linked to one another. In embodiments, the mobile element enzyme and the targeting element, e.g., nucleic acid binding component of the gene- editing system are connected via a linker. In embodiments, the linker is a flexible linker. In embodiments, the flexible linker is substantially comprised of glycine and serine residues, optionally wherein the flexible linker comprises (Gly4Ser)n, where n is from about 1 to about 12. In embodiments, the flexible linker is of about 20, or about 30, or about 40, or about 50, or about 60 amino acid residues. In embodiments, the flexible linker is about 50, or about 100, or about 150, or about 200 amino acid residues in length. In embodiments, the flexible linker comprises at least about 150 nucleotides (nt), or at least about 200 nt, or at least about 250 nt, or at least about 300 nt, or at least about 350 nt, or at least about 400 nt, or at least about 450 nt, or at least about 500 nt, or at least about 500 nt, or at least about 600 nt. In embodiments, the flexible linker comprises from about 450 nt to about 500 nt. In embodiments, the mobile element enzyme and the targeting element, e.g., nucleic acid binding component of the gene-editing system are encoded on a single polypeptide. In embodiments, the donor DNA comprises a gene encoding a complete polypeptide. In embodiments, the donor DNA comprises a gene which is defective or substantially absent in a disease state. Inteins Inteins (INTervening protEINS) are mobile genetic elements that are protein domains, found in nature, with the capability to carry out the process of protein splicing. See Sarmiento & Camarero (2019) Current Protein & Peptide Science, 20(5), 408–424, which is incorporated by reference herein in its entirety. Protein spicing is a post-translation biochemical modification which results in the cleavage and formation of peptide bonds between precursor polypeptide segments flanking the intein. Inteins apply standard enzymatic strategies to excise themselves post-translationally from a precursor protein via protein splicing. Nanda et al., Microorganisms vol. 8,12 2004. 16 Dec. 2020, doi:10.3390/microorganisms8122004. An intein can splice its flanking N- and C-terminal domains to become a mature protein and excise itself from a sequence. For example, split inteins have been used to control the delivery of heterologous genes into transgenic organisms. See Wood & Camarero (2014) J Biol Chem.289(21):14512-14519. This approach relies on splitting the target protein into two segments, which are then post-translationally reconstituted in vivo by protein trans-splicing (PTS). See Aboye & Camarero (2012) J. Biol. Chem.287, 27026–27032. More recently, an intein-mediated split-Cas9 system has been developed to incorporate Cas9 into cells and reconstitute nuclease activity efficiently. Truong et al., Nucleic Acids Res.2015, 43 (13), 6450–6458. The protein splicing excises the internal region of the precursor protein, which is then followed by the ligation of the N-extein and C-extein fragments, resulting in two polypeptides – the excised intein and the new polypeptide produced by joining the C- and N-exteins. Sarmiento & Camarero (2019). In embodiments, intein-mediated incorporation of DNA binders such as, without limitation, dCas9, dCas12j, or TALEs, allows creation of a split-enzyme system such as, without limitation, split-MLT mobile element enzyme system, that permits reconstitution of the full-length enzyme, e.g., MLT mobile element enzyme, from two smaller fragments. This allows avoiding the need to express DNA binders at the N- or C-terminus of an enzyme, e.g., MLT mobile element enzyme. In this approach, the two portions of an enzyme, e.g., MLT mobile element enzyme, are fused to the intein and, after co-expression, the intein allows producing a full-length enzyme, e.g., MLT mobile element enzyme, by post- translation modification. Thus, in embodiments, a nucleic acid encoding the enzyme capable of targeted genomic integration comprises an intein. In embodiments, the nucleic acid encodes the enzyme in the form of first and second portions with the intein encoded between the first and second portions, such that the first and second portions are fused into a functional enzyme upon post-translational excision of the intein from the enzyme. In embodiments, an intein is a suitable ligand-dependent intein, for example, an intein selected from those described in U.S. Patent No.9,200,045; Mootz et al., J. Am. Chem. Soc.2002; 124, 9044-9045; Mootz et al., J. Am. Chem. Soc. 2003; 125, 10561-10569; Buskirk et al., Proc. Natl. Acad. Sci. USA.2004; 101, 10505-10510; Skretas & Wood. Protein Sci.2005; 14, 523-532; Schwartz, et al., Nat. Chem. Biol.2007; 3, 50-54; Peck et al., Chem. Biol.2011; 18 (5), 619- 630; the entire contents of each of which are hereby incorporated by reference herein. In embodiments the intein is NpuN (Intein-N) (SEQ ID NO: 423) and/or NpuC (Intein-C) (SEQ ID NO: 424), or a variant thereof, e.g., a sequence having at least about 90%, or at least about 93%, or at least about 95%, or at least about 97%, or at least about 98%, or at least about 99% identity thereto. SEQ ID NO: 423: nucleotide sequence of NpuN (Intein-N) GGCGGATCTGGCGGTAGTGCTGAGTATTGTCTGAGTTACGAAACGGAAATACTCACGGTTGAGTATGGGCTTCTTCC AATTGGCAAAATCGTTGAAAAGCGCATAGAGTGTACGGTGTATTCCGTCGATAACAACGGTAATATCTACACCCAGC CGGTAGCTCAGTGGCACGACCGAGGCGAACAGGAAGTGTTCGAGTATTGCTTGGAAGATGGCTCCCTTATCCGCGCC ACTAAAGACCATAAGTTTATGACGGTTGACGGGCAGATGCTGCCTATAGACGAAATATTTGAGAGAGAGCTGGACTT GATGAGAGTCGATAATCTGCCAAAT SEQ ID NO: 424: nucleotide sequence of NpuC (Intein-C) GGCGGATCTGGCGGTAGTGGGGGTTCCGGATCCATAAAGATAGCTACTAGGAAATATCTTGGCAAACAAAACGTCTA TGACATAGGAGTTGAGCGAGATCACAATTTTGCTTTGAAGAATGGGTTCATCGCGTCTAATTGCTTCAACGCTAGCG GCGGGTCAGGAGGCTCTGGTGGAAGC Nucleic Acids of the Disclosure In embodiments, a nucleic acid encoding the enzyme is RNA. In embodiments, a nucleic acid encoding the transgene is DNA. In embodiments, the enzyme (e.g., without limitation, the mobile element enzyme) is encoded by a recombinant or synthetic nucleic acid. In embodiments, the nucleic acid is RNA, optionally a helper RNA. In embodiments, the nucleic acid is RNA that has a 5’-m7G cap (cap0, or cap1, or cap2), optionally with pseudouridine substitution (e.g., without limitation n-methyl-pseudouridine), and optionally a poly-A tail of about 30, or about 50, or about 100, of about 150 nucleotides in length. In embodiments, the poly-A tail is of about 30 nucleotides in length, optionally 34 nucleotides in length. In embodiments, a nuclear localization signal is placed before the enzyme start codon at the N-terminus, optionally at the C-terminus. In embodiments, the nucleic acid that is RNA has a 5’-m7G cap (cap 0, or cap 1, or cap 2). In embodiments, the nucleic acid comprises a 5' cap structure, a 5'-UTR comprising a Kozak consensus sequence, a 5'-UTR comprising a sequence that increases RNA stability in vivo, a 3'-UTR comprising a sequence that increases RNA stability in vivo, and/or a 3' poly(A) tail. In embodiments, the enzyme (e.g., without limitation, a mobile element enzyme) is incorporated into a vector or a vector-like particle. In embodiments, the vector is a non-viral vector. In embodiments, a nucleic acid encoding the enzyme in accordance with embodiments of the present disclosure, is DNA. In various embodiments, a construct comprising a donor DNA is any suitable genetic construct, such as a nucleic acid construct, a plasmid, or a vector. In various embodiments, the construct is DNA, which is referred to herein as a donor DNA. In embodiments, sequences of a nucleic acid encoding the donor DNA is codon optimized to provide improved mRNA stability and protein expression in mammalian systems. In embodiments, the enzyme and the donor DNA are included in different vectors. In embodiments, the enzyme and the donor DNA are included in the same vector. In various embodiments, a nucleic acid encoding the enzyme capable of targeted genomic integration (e.g., without limitation, a mobile element enzyme which is a chimeric mobile element enzyme) is RNA (e.g., helper RNA), and a nucleic acid encoding a donor DNA is DNA. As would be appreciated in the art, a donor DNA often includes an open reading frame that encodes a transgene at the middle of donor DNA and terminal repeat sequences at the 5’ and 3’ end of the donor DNA. The translated mobile element enzyme binds to the 5’ and 3’ sequence of the donor DNA and carries out the transposition function. In embodiments, donor DNA or transgene are used interchangeably with mobile elements, which are used to refer to polynucleotides capable of inserting copies of themselves into other polynucleotides. The term donor DNA is well known to those skilled in the art and includes classes of donor DNAs that can be distinguished on the basis of sequence organization, for example inverted terminal sequences at each end, and/or directly repeated long terminal repeats (LTRs) at the ends. In embodiments, the donor DNA as described herein may be described as a piggyBac like element, e.g., a donor DNA element that is characterized by its traceless excision, which recognizes TTAA (SEQ ID NO: 440) sequence and restores the sequence at the insert site back to the original TTAA (SEQ ID NO: 440) sequence after removal of the donor DNA. In embodiments, the donor DNA is flanked by one or more end sequences or terminal ends. In embodiments, the donor DNA is or comprises a gene encoding a complete polypeptide. In embodiments, the donor DNA is or comprises a gene which is defective or substantially absent in a disease state. In embodiments, the donor DNA includes a MLT mobile element enzyme (e.g., without limitation, a MLT mobile element enzyme having at least about 90% identity to the amino acid sequence of SEQ ID NO: 1, SEQ ID NO: 10, or SEQ ID NO: 11). For example, the mobile element enzyme can act on a left terminal end having a nucleotide sequence of SEQ ID NO: 431 or a sequence having at least about 90%, or at least about 93%, or at least about 95%, or at least about 97%, or at least about 98%, or at least about 99% identity thereto. In embodiments, the donor DNA can act on a right terminal end having a nucleotide sequence of SEQ ID NO: 432 or a sequence having at least about 90%, or at least about 93%, or at least about 95%, or at least about 97%, or at least about 98%, or at least about 99% identity thereto. In embodiments, the donor DNA acts on both MLT left donor DNA end and MLT right donor DNA end, having nucleotide sequences of SEQ ID NO: 431 and of SEQ ID NO: 432 respectively, or a sequence having at least about 90%, or at least about 93%, or at least about 95%, or at least about 97%, or at least about 98%, or at least about 99% identity thereto. In embodiments, a MLT left donor DNA end (5’ to 3’) is as follows TTAACACTTGGATTGCGGGAAACGAGTTAAGTCGGCTCGCGTGAATTGCGCGTACTCCGCGGGAGCCGTC TTAACTCGGTTCATATAGATTTGCGGTGGAGTGCGGGAAACGTGTAAACTCGGGCCGATTGTAACTGCGT ATTACCAAATATTTGTT (SEQ ID NO: 431) In embodiments, a MLT right donor DNA end (5’ to 3’) is as follows AATTATTTATGTACTGAATAGATAAAAAAATGTCTGTGATTGAATAAATTTTCATTTTTTACACAAGAAA CCGAAAATTTCATTTCAATCGAACCCATACTTCAAAAGATATAGGCATTTTAAACTAACTCTGATTTTGC GCGGGAAACCTAAATAATTGCCCGCGCCATCTTATATTTTGGCGGGAAATTCACCCGACACCGTGGTGTT AA (SEQ ID NO: 432). In embodiments, a transgene is associated with various regulatory elements that are selected to ensure stable expression of a construct with the transgene. Thus, in embodiments, a transgene is encoded by a non-viral vector (e.g., without limitation, a DNA plasmid) that can comprise one or more insulator sequences that prevent or mitigate activation or inactivation of nearby genes. The insulators flank the donor DNA (transgene cassette) to reduce transcriptional silencing and position effects imparted by chromosomal sequences. As an additional effect, the insulators can eliminate functional interactions of the transgene enhancer and promoter sequences with neighboring chromosomal sequences. In embodiments, the one or more insulator sequences comprise an HS4 insulator (1.2-kb 5’ -HS4 chicken β-globin (cHS4) insulator element) and an D4Z4 insulator (tandem macrosatellite repeats linked to Facio-Scapulo-Humeral Dystrophy (FSHD). In embodiments, the sequences of the HS4 insulator and the D4Z4 insulator are as described in Rival-Gervier et al. Mol Ther.2013 Aug; 21(8):1536-50, which is incorporated herein by reference in its entirety. In embodiments, the transgene is inserted into a GSHS location in a host genome. GSHSs is defined as loci well-suited for gene transfer, as integrations within these sites are not associated with adverse effects such as proto-oncogene activation, tumor suppressor inactivation, or insertional mutagenesis. GSHSs can defined by the following criteria: (1) distance of at least 50 kb from the 5’ end of any gene, (2) distance of at least 300 kb from any cancer-related gene, (3) distance of at least 300 kb from any microRNA (miRNA), (4) location outside a transcription unit, and (5) location outside ultra-conserved regions (UCRs) of the human genome. See Papapetrou et al. Nat Biotechnol 2011;29:73-8; Bejerano et al. Science 2004;304:1321-5. Furthermore, the use of GSHS locations can allow stable transgene expression across multiple cell types. One such site, chemokine C-C motif receptor 5 (CCR5) has been identified and used for integrative gene transfer. CCR5 is a member of the beta chemokine receptor family and is required for the entry of R5 tropic viral strains involved in primary infections. A homozygous 32 bp deletion in the CCR5 gene confers resistance to HIV-1 virus infections in humans. Disrupted CCR5 expression, naturally occurring in about 1% of the Caucasian population, does not appear to result in any reduction in immunity. Lobritz at al., Viruses 2010;2:1069-105. A clinical trial has demonstrated safety and efficacy of disrupting CCR5 via targetable nucleases. Tebas at al., HIV. N Engl J Med 2014;370:901-10. In embodiments, the donor DNA is under control of a tissue-specific promoter. The tissue-specific promoter is, e.g., without limitation, a liver-specific promoter. In embodiments, the liver-specific promoter is an LP1 promoter that, in embodiments, is a human LP1 promoter. The LP1 promoter is described, e.g., in Nathwani et al. Blood vol. 2006;107(7):2653-61, and it is constructed, without limitation, as described in Nathawani et al. It should be appreciated however that a variety of promoters can be used, including other tissue-specific promoters, inducible promoters, constitutive promoters, etc. In embodiments, the present nucleic acids include polymeric form of nucleotides of any length, either ribonucleotides or deoxyribonucleotides, or analogs or derivatives thereof. In embodiments, there is provided double- and single- stranded DNA, as well as double- and single-stranded RNA, and RNA-DNA hybrids. In embodiments, transcriptionally- activated polynucleotides such as methylated or capped polynucleotides are provided. In embodiments, the present compositions are mRNA or DNA. In embodiments, the present non-viral vectors are linear or circular DNA molecules that comprise a polynucleotide encoding a polypeptide and is operably linked to control sequences, wherein the control sequences provide for expression of the polynucleotide encoding the polypeptide. In embodiments, the non-viral vector comprises a promoter sequence, and transcriptional and translational stop signal sequences. Such vectors may include, among others, chromosomal and episomal vectors, e.g., vectors bacterial plasmids, from donor DNAs, from yeast episomes, from insertion elements, from yeast chromosomal elements, and vectors from combinations thereof. The present constructs may contain control regions that regulate as well as engender expression. In embodiments, the construct comprising the enzyme and/or transgene is codon optimized. Transgene codon optimization is used to optimize therapeutic potential of the transgene and its expression in the host organism. Codon optimization is performed to match the codon usage in the transgene with the abundance of transfer RNA (tRNA) for each codon in a host organism or cell. Codon optimization methods are known in the art and described in, for example, WO 2007/142954, which is incorporated by reference herein in its entirety. Optimization strategies can include, for example, the modification of translation initiation regions, alteration of mRNA structural elements, and the use of different codon biases. In embodiments, the construct comprising the enzyme and/or transgene includes several other regulatory elements that are selected to ensure stable expression of the construct. Thus, in embodiments, the non-viral vector is a DNA plasmid that can comprise one or more insulator sequences that prevent or mitigate activation or inactivation of nearby genes. In embodiments, the one or more insulator sequences comprise an HS4 insulator (1.2-kb 5'-HS4 chicken ȕ- globin (cHS4) insulator element) and an D4Z4 insulator (tandem macrosatellite repeats linked to Facio-Scapulo- Humeral Dystrophy (FSHD). In embodiments, the sequences of the HS4 insulator and the D4Z4 insulator are as described in Rival-Gervier et al. Mol Ther.2013 Aug; 21(8):1536-50, which is incorporated herein by reference in its entirety. In embodiments, the gene of the construct comprising the enzyme and/or transgene is capable of transposition in the presence of a mobile element enzyme. In embodiments, the non-viral vector in accordance with embodiments of the present disclosure comprises a nucleic acid construct encoding a mobile element enzyme. The mobile element enzyme is an RNA mobile element enzyme plasmid. In embodiments, the non-viral vector further comprises a nucleic acid construct encoding a DNA mobile element enzyme plasmid. In embodiments, the mobile element enzyme is an in vitro-transcribed mRNA mobile element enzyme. The mobile element enzyme is capable of excising and/or transposing the gene from the construct comprising the enzyme and/or transgene to site- or locus-specific genomic regions. In embodiments, the enzyme and the donor DNA are included in the same vector. In embodiments, the enzyme is disposed on the same (cis) or different vector (trans) than a donor DNA with a transgene. Accordingly, in embodiments, the enzyme and the donor DNA encompassing a transgene are in cis configuration such that they are included in the same vector. In embodiments, the enzyme and the donor DNA encompassing a transgene are in trans configuration such that they are included in different vectors. The vector is any non-viral vector in accordance with the present disclosure. In some aspects, a nucleic acid encoding the enzyme capable of targeted genomic integration (e.g., a mobile element enzyme or a chimeric mobile element enzyme) in accordance with embodiments of the present disclosure is provided. The nucleic acid is or comprises DNA or RNA. In embodiments, the nucleic acid encoding the enzyme is DNA. In embodiments, the nucleic acid encoding the enzyme capable of targeted genomic integration (e.g., a chimeric mobile element enzyme) is RNA such as, e.g., helper RNA. In embodiments, the chimeric mobile element enzyme is incorporated into a vector. In embodiments, the vector is a non-viral vector. In embodiments, a nucleic acid encoding the transgene in accordance with embodiments of the present disclosure is provided. The nucleic acid is or comprises DNA or RNA. In embodiments, the nucleic acid encoding the transgene is DNA. In embodiments, the nucleic acid encoding the e transgene is RNA such as, e.g., helper RNA. In embodiments, the transgene is incorporated into a vector. In embodiments, the vector is a non-viral vector. In embodiments, the present enzyme can be in the form or an RNA or DNA and have one or two N-terminus nuclear localization signal (NLS) to shuttle the protein more efficiently into the nucleus. For example, in embodiments, the present enzyme further comprises one, two, three, four, five, or more NLSs. Examples of NLS are provided in Kosugi et al. (J. Biol. Chem. (2009) 284:478-485; incorporated by reference herein). In a particular embodiment, the NLS comprises the consensus sequence K(K/R)X(K/R) (SEQ ID NO: 348). In an embodiment, the NLS comprises the consensus sequence (K/R)(K/R)X10-12(K/R)3/5 (SEQ ID NO: 349), where (K/R)3/5 represents at least three of the five amino acids is either lysine or arginine. In an embodiment, the NLS comprises the c-myc NLS. In a particular embodiment, the c-myc NLS comprises the sequence PAAKRVKLD (SEQ ID NO: 350). In a particular embodiment, the NLS is the nucleoplasmin NLS. In embodiments, the nucleoplasmin NLS comprises the sequence KRPAATKKAGQAKKKK (SEQ ID NO: 351). In embodiments, the NLS comprises the SV40 Large T-antigen NLS. In embodiments, the SV40 Large T-antigen NLS comprises the sequence PKKKRKV (SEQ ID NO: 352). In a particular embodiment, the NLS comprises three SV40 Large T-antigen NLSs (e.g., DPKKKRKVDPKKKRKVDPKKKRKV (SEQ ID NO: 353). In embodiments, the NLS may comprise mutations/variations in the above sequences such that they contain 1 or more substitutions, additions or deletions (e.g., about 1, or about 2, or about 3, or about 4, or about 5, or about 10 substitutions, additions, or deletions). In some aspects, a host cell comprising the nucleic acid in accordance with embodiments of the present disclosure is provided. Lipids In embodiments, at least one of the first nucleic acid and the second nucleic acid is in the form of a lipid nanoparticle (LNP). In embodiments, a composition comprising the first and second nucleic acids is in the form of an LNP. In embodiments, a nucleic acid encoding the enzyme and a nucleic acid encoding the transgene are contained within the same lipid nanoparticle (LNP). In embodiments, the nucleic acid encoding the enzyme and the nucleic acid encoding the donor DNA are a mixture incorporated into or associated with the same LNP. In embodiments, the nucleic acid encoding the enzyme and the nucleic acid encoding the donor DNA are in the form of a co-formulation incorporated into or associated with the same LNP. In embodiments, the LNP is selected from 1,2-dioleoyl-3-trimethylammonium propane (DOTAP), a cationic cholesterol derivative mixed with dimethylaminoethane-carbamoyl (DC-Chol), phosphatidylcholine (PC), triolein (glyceryl trioleate), and 1,2-distearoyl-sn-glycero-3-phosphoethanolamine-N-[carboxy(polyethylene glycol)-2000] (DSPE-PEG), 1,2- dimyristoyl-rac-glycero-3-methoxypolyethyleneglycol – 2000 (DMG-PEG 2K), and 1,2 distearol -sn-glycerol- 3phosphocholine (DSPC) and/or comprising of one or more molecules selected from polyethylenimine (PEI) and poly(lactic-co-glycolic acid) (PLGA), and N-Acetylgalactosamine (GalNAc). In embodiments, an LNP is as described, e.g., in Patel et al., J Control Release 2019; 303:91-100. The LNP can comprise one or more of a structural lipid (e.g., DSPC), a PEG-conjugated lipid (CDM-PEG), a cationic lipid (MC3), cholesterol, and a targeting ligand (e.g., GalNAc). In embodiments, a nanoparticle is a particle having a diameter of less than about 1000 nm. In embodiments, nanoparticles of the present disclosure have a greatest dimension (e.g., diameter) of about 500 nm or less, or about 400 nm or less, or about 300 nm or less, or about 200 nm or less, or about 100 nm or less. In embodiments, nanoparticles of the present disclosure have a greatest dimension ranging between about 50 nm and about 150 nm, or between about 70 nm and about 130 nm, or between about 80 nm and about 120 nm, or between about 90 nm and about 110 nm. In embodiments, the nanoparticles of the present disclosure have a greatest dimension (e.g., a diameter) of about 100 nm. In some aspects, a cell in accordance with the present disclosure is prepared via an in vivo genetic modification method. In embodiments, a genetic modification in accordance with the present disclosure is performed via an ex vivo method. In some aspects, a cell in accordance with the present disclosure is prepared by contacting the cell with an enzyme capable of targeted genomic integration (e.g., without limitation, a mammalian mobile element enzyme) in vivo. In embodiments, the cell is contacted with the enzyme ex vivo. In embodiments, the present method provides reduced insertional mutagenesis or oncogenesis as compared to a method with a non-chimeric mobile element enzyme. Therapeutic Applications In embodiments, the transgene of interest in accordance with embodiments of the present disclosure can encode various genes. In embodiments, the enzyme (e.g., without limitations, a mobile element enzyme), and the donor DNA are included in the same pharmaceutical composition. In embodiments, the enzyme (e.g., without limitations, a mobile element enzyme) and the donor DNA are included in different pharmaceutical compositions. In embodiments, the enzyme and the donor DNA are co-transfected. In embodiments, the enzyme and the donor DNA are transfected separately. In embodiments, a method of producing a molecule with the transfected cell is provided, wherein the transfected cell is generated using a method of making a viral packaging or producer cell line in accordance with embodiments of the present disclosure. The molecule is optionally a protein. In embodiments, the protein is an antibody. In embodiments, the method of making a viral packaging or producer cell line of the present disclosure provides a stable cell with substantially reduced or ablated CAP expression. In embodiments, the method of making a viral packaging or producer cell line of the present disclosure provides a stable cell with REP expression or substantially enhanced REP expression. In embodiments, the method of making a viral packaging or producer cell line of the present disclosure provides a stable cell with substantially reduced or ablated CAP expression and REP expression or substantially enhanced REP expression. In embodiments, the method of making a viral packaging or producer cell line of the present disclosure provides a stable cell with REP expression or substantially enhanced VP1 expression. In embodiments, the method of making a viral packaging or producer cell line of the present disclosure provides a stable cell with substantially reduced or ablated CAP expression. In embodiments, the viral packaging or producer cell line of the present disclosure is suitable for providing substantially reduced empty or cargo-free capsid. In embodiments, a transfected cell for gene therapy is provided, wherein the transfected cell is generated using a method of making a viral packaging or producer cell line in accordance with embodiments of the present disclosure. In embodiments, a method of delivering a cell therapy is provided, comprising administering to a patient in need thereof the transfected cell generated using a method of making a viral packaging or producer cell line in accordance with embodiments of the present disclosure. In embodiments, a method of treating a disease or condition using a cell therapy, comprising administering to a patient in need thereof the transfected cell generated using a method of making a viral packaging or producer cell line in accordance with embodiments of the present disclosure. In embodiments, a method of treating a disease or condition using an antibody therapy, comprising administering to a patient in need thereof the transfected cell generated using a method of making a viral packaging or producer cell line in accordance with embodiments of the present disclosure. In embodiments, the disease or condition is or comprises cancer. In embodiments, the cancer is or comprises an adrenal cancer, a biliary track cancer, a bladder cancer, a bone/bone marrow cancer, a brain cancer, a breast cancer, a cervical cancer, a colorectal cancer, a cancer of the esophagus, a gastric cancer, a head/neck cancer, a hepatobiliary cancer, a kidney cancer, a liver cancer, a lung cancer, an ovarian cancer, a pancreatic cancer, a pelvis cancer, a pleura cancer, a prostate cancer, a renal cancer, a skin cancer, a stomach cancer, a testis cancer, a thymus cancer, a thyroid cancer, a uterine cancer, a lymphoma, a melanoma, a multiple myeloma, or a leukemia. In embodiments, the cancer is selected from one or more of the basal cell carcinoma, biliary tract cancer; bladder cancer; bone cancer; brain and central nervous system cancer; breast cancer; cancer of the peritoneum; cervical cancer; choriocarcinoma; colon and rectum cancer; connective tissue cancer; cancer of the digestive system; endometrial cancer; esophageal cancer; eye cancer; cancer of the head and neck; gastric cancer; glioblastoma; hepatic carcinoma; hepatoma; intra-epithelial neoplasm; kidney or renal cancer; larynx cancer; leukemia; liver cancer; lung cancer; melanoma; myeloma; neuroblastoma; oral cavity cancer; ovarian cancer; pancreatic cancer; prostate cancer; retinoblastoma; rhabdomyosarcoma; rectal cancer; cancer of the respiratory system; salivary gland carcinoma; sarcoma; skin cancer; squamous cell cancer; stomach cancer; testicular cancer; thyroid cancer; uterine or endometrial cancer; cancer of the urinary system; vulval cancer; Hodgkin's lymphoma; non-Hodgkin's lymphoma; B-cell lymphoma; small lymphocytic (SL) NHL; intermediate grade/follicular NHL; intermediate grade diffuse NHL; high grade immunoblastic NHL; high grade lymphoblastic NHL; high grade small non-cleaved cell NHL; bulky disease NHL; mantle cell lymphoma; AIDS-related lymphoma; Waldenstrom's Macroglobulinemia; chronic lymphocytic leukemia (CLL); acute lymphoblastic leukemia (ALL); and Hairy cell leukemia. In embodiments, the cancer is selected from one or more of basal cell carcinoma, biliary tract cancer; bladder cancer; bone cancer; brain and central nervous system cancer; breast cancer; cancer of the peritoneum; cervical cancer; choriocarcinoma; colon and rectum cancer; connective tissue cancer; cancer of the digestive system; endometrial cancer; esophageal cancer; eye cancer; cancer of the head and neck; gastric cancer (including gastrointestinal cancer); glioblastoma; hepatic carcinoma; hepatoma; intra-epithelial neoplasm; kidney or renal cancer; larynx cancer; leukemia; liver cancer; lung cancer (e.g., small-cell lung cancer, non-small cell lung cancer, adenocarcinoma of the lung, and squamous carcinoma of the lung); melanoma; myeloma; neuroblastoma; oral cavity cancer (lip, tongue, mouth, and pharynx); ovarian cancer; pancreatic cancer; prostate cancer; retinoblastoma; rhabdomyosarcoma; rectal cancer; cancer of the respiratory system; salivary gland carcinoma; sarcoma; skin cancer; squamous cell cancer; stomach cancer; testicular cancer; thyroid cancer; uterine or endometrial cancer; cancer of the urinary system; vulvar cancer; lymphoma including Hodgkin's and non-Hodgkin's lymphoma, as well as B-cell lymphoma (including low grade/follicular non-Hodgkin's lymphoma (NHL); small lymphocytic (SL) NHL; intermediate grade/follicular NHL; intermediate grade diffuse NHL; high grade immunoblastic NHL; high grade lymphoblastic NHL; high grade small non-cleaved cell NHL; bulky disease NHL; mantle cell lymphoma; AIDS-related lymphoma; and Waldenstrom's Macroglobulinemia; chronic lymphocytic leukemia (CLL); acute lymphoblastic leukemia (ALL); Hairy cell leukemia; chronic myeloblastic leukemia; as well as other carcinomas and sarcomas; and post-transplant lymphoproliferative disorder (PTLD), as well as abnormal vascular proliferation associated with phakomatoses, edema (e.g. that associated with brain tumors), and Meigs syndrome. In embodiments, the disease or condition is or comprises an infectious disease. In embodiments, the infectious disease is a coronavirus infection, optionally selected from infection with SAR-CoV, MERS-CoV, and SARS-CoV-2, or variants thereof. In embodiments, the infectious disease is or comprises a disease comprising a viral infection, a parasitic infection, or a bacterial infection. In embodiments, the viral infection is caused by a virus of family Flaviviridae, a virus of family Picornaviridae, a virus of family Orthomyxoviridae, a virus of family Coronaviridae, a virus of family Retroviridae, a virus of family Paramyxoviridae, a virus of family Bunyaviridae, or a virus of family Reoviridae. In embodiments, the virus of family Coronaviridae comprises a betacoronavirus or an alphacoronavirus, optionally wherein the betacoronavirus is selected from SARS-CoV-2, SARS-CoV, MERS-CoV, HCoV-HKU1, and HCoV-OC43, or the alphacoronavirus is selected from a HCoV-NL63 and HCoV-229E. In embodiments, the infectious disease comprises a coronavirus infection 2019 (COVID-19). In embodiments, the method requires a single administration. In embodiments, the method requires a plurality of administrations. Isolated Cell In some aspects of the present disclosure, an isolated cell is provided that comprises the transfected cell in accordance with embodiments of the present disclosure. In some aspects, the present disclosure provides an ex vivo gene therapy approach. Accordingly, in embodiments, the method that is used to treat an inherited or acquired disease in a patient in need thereof comprises (a) contacting a cell obtained from a patient (autologous) or another individual (allogeneic) with a transfected cell in accordance with embodiments of the present disclosure; and (b) administering the cell to a patient in need thereof. One of the advantages of ex vivo gene therapy is the ability to “sample” the transduced cells before patient administration. This facilitates efficacy and allows performing safety checks before introducing the cell(s) to the patient. For example, the transduction efficiency and/or the clonality of integration can be assessed before infusion of the product. The present disclosure provides transfected cells and methods that can be effectively used for ex vivo gene modification. In embodiments, a composition comprising transfected cells in accordance with the present disclosure comprises a pharmaceutically acceptable carrier, excipient, or diluent. Methods of formulating suitable pharmaceutical compositions are known in the art, see, e.g., Remington: The Science and Practice of Pharmacy, 21st ed., 2005; and the books in the series Drugs and the Pharmaceutical Sciences: a Series of Textbooks and Monographs (Dekker, N.Y.). For example, pharmaceutical compositions suitable for injectable use can include sterile aqueous solutions (where water soluble) or dispersions and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersion. For intravenous administration, suitable carriers include physiological saline, bacteriostatic water, Cremophor EL™ (BASF, Parsippany, N.J.) or phosphate buffered saline (PBS). In all cases, the composition must be sterile, and the fluid should be easy to draw up by a syringe. It should be stable under the conditions of manufacture and storage and must be preserved against the contaminating action of microorganisms such as bacteria and fungi. The carrier can be a solvent or dispersion medium containing, for example, water, ethanol, polyol (for example, glycerol, propylene glycol, and liquid polyethylene glycol, and the like), and suitable mixtures thereof. The proper fluidity can be maintained, for example, by the use of a coating such as lecithin, by the maintenance of the required particle size in the case of dispersion and by the use of surfactants. Prevention of the action of microorganisms can be achieved by various antibacterial and antifungal agents, for example, parabens, chlorobutanol, phenol, ascorbic acid, thimerosal, and the like. In many cases, it will be preferable to include isotonic agents, for example, sugars, polyalcohols such as mannitol, sorbitol, and sodium chloride in the composition. Prolonged absorption of the injectable compositions can be brought about by including in the composition an agent that delays absorption, for example, aluminum monostearate and gelatin. Sterile injectable solutions can be prepared by incorporating the active compound in the required amount in an appropriate solvent with one or a combination of ingredients enumerated above, as required, followed by filtered sterilization. Generally, dispersions are prepared by incorporating the active compound into a sterile vehicle, which contains a basic dispersion medium and the required other ingredients from those enumerated above. In the case of sterile powders for the preparation of sterile injectable solutions, the preferred methods of preparation are vacuum drying and freeze-drying, which yield a powder of the active ingredient plus any additional desired ingredient from a previously sterile-filtered solution thereof. Therapeutic compounds can be prepared with carriers that will protect the therapeutic compounds against rapid elimination from the body, such as a controlled release formulation, including implants and microencapsulated delivery systems. Biodegradable, biocompatible polymers can be used, such as collagen, ethylene vinyl acetate, polyanhydrides (e.g., poly[1,3-bis(carboxyphenoxy)propane-co-sebacic-acid] (PCPP-SA) matrix, fatty acid dimer- sebacic acid (FAD-SA) copolymer, poly(lactide-co-glycolide)), polyglycolic acid, collagen, polyorthoesters, polyethyleneglycol-coated liposomes, and polylactic acid. Such formulations can be prepared using standard techniques, or obtained commercially, e.g., from Alza Corporation and Nova Pharmaceuticals, Inc. Liposomal suspensions can also be used as pharmaceutically acceptable carriers. These can be prepared according to methods known to those skilled in the art, for example, as described in U.S. Pat. No.4,522,811. Semisolid, gelling, soft-gel, or other formulations (including controlled release) can be used, e.g., when administration to a surgical site is desired. Methods of making such formulations are known in the art and can include the use of biodegradable, biocompatible polymers. See, e.g., Sawyer et al., Yale J Biol Med.2006; 79(3-4): 141-152. In embodiments, there is provided a method of transforming a cell using the construct comprising the enzyme and/or transgene described herein in the presence of a mobile element enzyme to produce a stably transfected cell which results from the stable integration of a gene of interest into the cell. In embodiments, the stable integration comprises an introduction of a polynucleotide into a chromosome or mini-chromosome of the cell and, therefore, becomes a relatively permanent part of the cellular genome. In embodiments, there is provided a transgenic organism that may comprise cells which have been transformed by the methods of the present disclosure. In embodiments, the organism may be a mammal or an insect. When the organism is a mammal, the organism may include, but is not limited to, a mouse, a rat, a monkey, a dog, a rabbit, and the like. When the organism is an insect, the organism may include, but is not limited to, a fruit fly, a mosquito, a bollworm, and the like. In embodiments, the cells produced in accordance with embodiments of the present disclosure, and/or components for generating cells, is included in a container, kit, pack, or dispenser together with instructions for administration. Also provided herein are kits comprising: one or more genetic constructs encoding the present enzyme and donor DNA and instructions and/or reagents for the use of the same. Also provided herein are kits comprising: i) a transfected cell in accordance with embodiments of the present disclosure, ii) instructions for the use of the transfected cell. Furthermore, in embodiments, a kit is provided for creating an AAV of packaging cell line or an AAV library of packaging cell lines, and instructions for creating a library. A subsequent user-defined transfection of the viral genome can be performed to generate a producer cell line. In embodiments, the kit includes instructions for transfection of an AAV packaging cell line with a desired transgene. In embodiments, a kit includes an AAV library of packaging cell lines and instructions for creating a library In some aspects, a kit is provided that comprises an enzyme (e.g., without limitation, a recombinant mammalian mobile element enzyme) or a nucleic acid in accordance with embodiments of the present disclosure, and instructions for introducing DNA and/or RNA into a cell using the enzyme. Definitions The following definitions are used in connection with the disclosure disclosed herein. Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood to one of skill in the art to which this invention belongs. As used herein, “a,” “an,” or “the” can mean one or more than one. Further, the term “about” when used in connection with a referenced numeric indication means the referenced numeric indication plus or minus up to 10% of that referenced numeric indication. For example, the language “about 50” covers the range of 45 to 55. An “effective amount,” when used in connection with medical uses is an amount that is effective for providing a measurable treatment, prevention, or reduction in the rate of pathogenesis of a disease of interest. The term “in vivo” refers to an event that takes place in a subject’s body. The term “ex vivo” refers to an event which involves treating or performing a procedure on a cell, tissue and/or organ which has been removed from a subject’s body. Aptly, the cell, tissue and/or organ may be returned to the subject’s body in a method of treatment or surgery. As used herein, the term “variant” encompasses but is not limited to nucleic acids or proteins which comprise a nucleic acid or amino acid sequence which differs from the nucleic acid or amino acid sequence of a reference by way of one or more substitutions, deletions and/or additions at certain positions. The variant may comprise one or more conservative substitutions. Conservative substitutions may involve, e.g., the substitution of similarly charged or uncharged amino acids. “Carrier” or “vehicle” as used herein refer to carrier materials suitable for drug administration. Carriers and vehicles useful herein include any such materials known in the art, e.g., any liquid, gel, solvent, liquid diluent, solubilizer, surfactant, lipid, or the like, which is nontoxic, and which does not interact with other components of the composition in a deleterious manner. The phrase “pharmaceutically acceptable” refers to those compounds, materials, compositions, and/or dosage forms that are, within the scope of sound medical judgment, suitable for use in contact with the tissues of human beings and animals without excessive toxicity, irritation, allergic response, or other problems or complications commensurate with a reasonable benefit/risk ratio. The terms “pharmaceutically acceptable carrier” or “pharmaceutically acceptable excipient” are intended to include any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents, and inert ingredients. The use of such pharmaceutically acceptable carriers or pharmaceutically acceptable excipients for active pharmaceutical ingredients is well known in the art. Except insofar as any conventional pharmaceutically acceptable carrier or pharmaceutically acceptable excipient is incompatible with the active pharmaceutical ingredient, its use in the therapeutic compositions of the disclosure is contemplated. Additional active pharmaceutical ingredients, such as other drugs, can also be incorporated into the described compositions and methods. As referred to herein, all compositional percentages are by weight of the total composition, unless otherwise specified. As used herein, the word “include,” and its variants, is intended to be non-limiting, such that recitation of items in a list is not to the exclusion of other like items that may also be useful in the compositions and methods of this technology. Similarly, the terms “can” and “may” and their variants are intended to be non-limiting, such that recitation that an embodiment can or may comprise certain elements or features does not exclude other embodiments of the present technology that do not contain those elements or features. Although the open-ended term “comprising,” as a synonym of terms such as including, containing, or having, is used herein to describe and claim the invention, the present invention, or embodiments thereof, may alternatively be described using alternative terms such as “consisting of” or “consisting essentially of.” As used herein, the words “preferred” and “preferably” refer to embodiments of the technology that afford certain benefits, under certain circumstances. However, other embodiments may also be preferred, under the same or other circumstances. Furthermore, the recitation of one or more preferred embodiments does not imply that other embodiments are not useful and is not intended to exclude other embodiments from the scope of the technology. The amount of compositions described herein needed for achieving a therapeutic effect may be determined empirically in accordance with conventional procedures for the particular purpose. Generally, for administering therapeutic agents for therapeutic purposes, the therapeutic agents are given at a pharmacologically effective dose. A “pharmacologically effective amount,” “pharmacologically effective dose,” “therapeutically effective amount,” or “effective amount” refers to an amount sufficient to produce the desired physiological effect or amount capable of achieving the desired result, particularly for treating the disorder or disease. An effective amount as used herein would include an amount sufficient to, for example, delay the development of a symptom of the disorder or disease, alter the course of a symptom of the disorder or disease (e.g., slow the progression of a symptom of the disease), reduce or eliminate one or more symptoms or manifestations of the disorder or disease, and reverse a symptom of a disorder or disease. Therapeutic benefit also includes halting or slowing the progression of the underlying disease or disorder, regardless of whether improvement is realized. Effective amounts, toxicity, and therapeutic efficacy can be determined by standard pharmaceutical procedures in cell cultures or experimental animals, e.g., for determining the LD50 (the dose lethal to about 50% of the population) and the ED50 (the dose therapeutically effective in about 50% of the population). The dosage can vary depending upon the dosage form employed and the route of administration utilized. The dose ratio between toxic and therapeutic effects is the therapeutic index and can be expressed as the ratio LD50/ED50. In embodiments, compositions and methods that exhibit large therapeutic indices are preferred. A therapeutically effective dose can be estimated initially from in vitro assays, including, for example, cell culture assays. Also, a dose can be formulated in animal models to achieve a circulating plasma concentration range that includes the IC50 as determined in cell culture, or in an appropriate animal model. Levels of the described compositions in plasma can be measured, for example, by high performance liquid chromatography. The effects of any particular dosage can be monitored by a suitable bioassay. The dosage can be determined by a physician and adjusted, as necessary, to suit observed effects of the treatment. As used herein, “methods of treatment” are equally applicable to use of a composition for treating the diseases or disorders described herein and/or compositions for use and/or uses in the manufacture of a medicaments for treating the diseases or disorders described herein. SELECTED SEQUENCES In embodiments, the present disclosure provides for any of the sequence provided herein, including the below, and a variant sequence having at least about 90%, or at least about 93%, or at least about 95%, or at least about 97%, or at least about 98%, or at least about 99% identity thereto, or at least about 10 mutations, or at least about 9 mutations, or at least about 8 mutations, or at least about 7 mutations, or at least about 6 mutations, or at least about 5 mutations, or at least about 4 mutations, or at least about 3 mutations, or at least about 2 mutations, or at least about 1 mutation. SEQ ID NO: 1: MLT mobile element enzyme protein (amino acid sequence of a variant of the hyperactive mobile element enzyme with S at position 8 and C at position 13 (572 amino acids) MAQHSDYSDDEFCADKLSNYSCDSDLENASTSDEDSSDDEVMVRPRTLRRRRISSSSSDSESDIEGGREEWSHV DNPPVLEDFLGHQGLNTDAVINNIEDAVKLFIGDDFFEFLVEESNRYYNQNRNNFKLSKKSLKWKDITPQEMKK FLGLIVLMGQVRKDRRDDYWTTEPWTETPYFGKTMTRDRFRQIWKAWHFNNNADIVNESDRLCKVRPVLDYFVP KFINIYKPHQQLSLDEGIVPWRGRLFFRVYNAGKIVKYGILVRLLCESDTGYICNMEIYCGEGKRLLETIQTVV SPYTDSWYHIYMDNYYNSVANCEALMKNKFRICGTIRKNRGIPKDFQTISLKKGETKFIRKNDILLQVWQSKKP VYLISSIHSAEMEESQNIDRTSKKKIVKPNALIDYNKHMKGVDRADQYLSYYSILRRTVKWTKRLAMYMINCAL FNSYAVYKSVRQRKMGFKMFLKQTAIHWLTDDIPEDMDIVPDLQPVPSTSGMRAKPPTSDPPCRLSMDMRKHTL QAIVGSGKKKNILRRCRVCSVHKLRSETRYMCKFCNIPLHKGACFEKYHTLKNY SEQ ID NO: 2: Codon-optimized MLT mobile element enzyme DNA. Nucleotide sequence of SEQ ID NO: 1 (1719 bp) ATGGCCCAGCACAGCGACTACAGCGACGACGAGTTCTGTGCCGATAAGCTGAGTAACTACAGCTGCGACAGCGA CCTGGAAAACGCCAGCACATCCGACGAGGACAGCTCTGACGACGAGGTGATGGTGCGGCCCAGAACCCTGAGAC GGAGAAGAATCAGCAGCTCTAGCAGCGACTCTGAATCCGACATCGAGGGCGGCCGGGAAGAGTGGAGCCACGTG GACAACCCTCCTGTTCTGGAAGATTTTCTGGGCCATCAGGGCCTGAACACCGACGCCGTGATCAACAACATCGA GGATGCCGTGAAGCTGTTCATAGGAGATGATTTCTTTGAGTTCCTGGTCGAGGAATCCAACCGCTATTACAACC AGAATAGAAACAACTTCAAGCTGAGCAAGAAAAGCCTGAAGTGGAAGGACATCACCCCTCAGGAGATGAAAAAG TTCCTGGGACTGATCGTTCTGATGGGACAGGTGCGGAAGGACAGAAGGGATGATTACTGGACAACCGAACCTTG GACCGAGACCCCTTACTTTGGCAAGACCATGACCAGAGACAGATTCAGACAGATCTGGAAAGCCTGGCACTTCA ACAACAATGCTGATATCGTGAACGAGTCTGATAGACTGTGTAAAGTGCGGCCAGTGTTGGATTACTTCGTGCCT AAGTTCATCAACATCTATAAGCCTCACCAGCAGCTGAGCCTGGATGAAGGCATCGTGCCCTGGCGGGGCAGACT GTTCTTCAGAGTGTACAATGCTGGCAAGATCGTCAAATACGGCATCCTGGTGCGCCTTCTGTGCGAGAGCGATA CAGGCTACATCTGTAATATGGAAATCTACTGCGGCGAGGGCAAAAGACTGCTGGAAACCATCCAGACCGTCGTT TCCCCTTATACCGACAGCTGGTACCACATCTACATGGACAACTACTACAATTCTGTGGCCAACTGCGAGGCCCT GATGAAGAACAAGTTTAGAATCTGCGGCACAATCAGAAAAAACAGAGGCATCCCTAAGGACTTCCAGACCATCT CTCTGAAGAAGGGCGAAACCAAGTTCATCAGAAAGAACGACATCCTGCTCCAAGTGTGGCAGTCCAAGAAACCC GTGTACCTGATCAGCAGCATCCATAGCGCCGAGATGGAAGAAAGCCAGAACATCGACAGAACAAGCAAGAAGAA GATCGTGAAGCCCAATGCTCTGATCGACTACAACAAGCACATGAAAGGCGTGGACCGGGCCGACCAGTACCTGT CTTATTACTCTATCCTGAGAAGAACAGTGAAATGGACCAAGAGACTGGCCATGTACATGATCAATTGCGCCCTG TTCAACAGCTACGCCGTGTACAAGTCCGTGCGACAAAGAAAAATGGGATTCAAGATGTTCCTGAAGCAGACAGC CATCCACTGGCTGACAGACGACATTCCTGAGGACATGGACATTGTGCCAGATCTGCAACCTGTGCCCAGCACCT CTGGTATGAGAGCTAAGCCTCCCACCAGCGATCCTCCATGTAGACTGAGCATGGACATGCGGAAGCACACCCTG CAGGCCATCGTCGGCAGCGGCAAGAAGAAGAACATCCTTAGACGGTGCAGGGTGTGCAGCGTGCACAAGCTGCG GAGCGAGACTCGGTACATGTGCAAGTTTTGCAACATTCCCCTGCACAAGGGAGCCTGCTTCGAGAAGTACCACA CCCTGAAGAATTACTAG SEQ ID NO: 3: PGBD4 Amino Acid Sequence (585 Amino Acids). MSNPRKRSIP MRDSNTGLEQ LLAEDSFDES DFSEIDDSDN FSDSALEADK 50 IRPLSHLESD GKSSTSSDSG RSMKWSARAM IPRQRYDFTG TPGRKVDVSD 100 ITDPLQYFEL FFTEELVSKI TRETNAQAAL LASKPPGPKG FSRMDKWKDT 150 DNDELKVFFA VMLLQGIVQK PELEMFWSTR PLLDTPYLRQ IMTGERFLLL 200 FRCLHFVNNS SISAGQSKAQ ISLQKIKPVF DFLVNKFSTV YTPNRNIAVD 250 ESLMLFKGPL AMKQYLPTKR VRFGLKLYVL CESQSGYVWN ALVHTGPGMN 300 LKDSADGLKS SRIVLTLVND LLGQGYCVFL DNFNISPMLF RELHQNRTDA 350 VGTARLNRKQ IPNDLKKRIA KGTTVARFCG ELMALKWCDG KEVTMLSTFH 400 NDTVIEVNNR NGKKTKRPRV IVDYNENMGA VDSADQMLTS YPSERKRHKV 450 WYKKFFHHLL HITVLNSYIL FKKDNPEHTM SHINFRLALI ERMLEKHHKP 500 GQQHLRGRPC SDDVTPLRLS GRHFPKSIPA TSGKQNPTGR CKICCSQYDK 550 DGKKIRKETR YFCAECDVPL CVVPCFEIYH TKKNY 585 SEQ ID NO: 4: PGBD4 Hyperactive Mutant (S8P, G17R, K134K) Amino Acid Sequence (585 Amino Acids). MSNPRKRPIP MRDSNTRLEQ LLAEDSFDES DFSEIDDSDN FSDSALEADK 50 IRPLSHLESD GKSSTSSDSG RSMKWSARAM IPRQRYDFTG TPGRKVDVSD 100 ITDPLQYFEL FFTEELVSKI TRETNAQAAL LASKPPGPKG FSRMDKWKDT 150 DNDELKVFFA VMLLQGIVQK PELEMFWSTR PLLDTPYLRQ IMTGERFLLL 200 FRCLHFVNNS SISAGQSKAQ ISLQKIKPVF DFLVNKFSTV YTPNRNIAVD 250 ESLMLFKGPL AMKQYLPTKR VRFGLKLYVL CESQSGYVWN ALVHTGPGMN 300 LKDSADGLKS SRIVLTLVND LLGQGYCVFL DNFNISPMLF RELHQNRTDA 350 VGTARLNRKQ IPNDLKKRIA KGTTVARFCG ELMALKWCDG KEVTMLSTFH 400 NDTVIEVNNR NGKKTKRPRV IVDYNENMGA VDSADQMLTS YPSERKRHKV 450 WYKKFFHHLL HITVLNSYIL FKKDNPEHTM SHINFRLALI ERMLEKHHKP 500 GQQHLRGRPC SDDVTPLRLS GRHFPKSIPA TSGKQNPTGR CKICCSQYDK 550 DGKKIRKETR YFCAECDVPL CVVPCFEIYH TKKNY 585 SEQ ID NO: 5: PGBD4 Hyperactive Mutant (S8P, G17R, K134K) Nucleotide Sequence (1758 bp). ATGTCAAATC CTAGAAAACG TCCCATTCCT ATGCGTGATA GTAATACCCG TCTCGAACAG 60 TTGTTGGCTG AAGATTCATT TGATGAATCT GATTTTTCGG AAATAGATGA TTCTGATAAT 120 TTTTCGGATA GTGCTTTAGA AGCCGATAAG ATCAGGCCTC TGTCCCATTT AGAATCTGAT 180 GGAAAGAGCT CTACATCAAG TGACTCAGGG CGCTCCATGA AATGGTCAGC TCGTGCTATG 240 ATTCCACGTC AAAGGTATGA CTTTACCGGC ACACCTGGCA GAAAAGTCGA TGTCAGTGAT 300 ATCACTGACC CATTGCAGTA TTTTGAACTG TTCTTTACTG AGGAATTAGT TTCAAAAATT 360 ACTAGAGAAA CAAATGCCCA AGCTGCCTTG TTGGCTTCAA AGCCACCGGG TCCGAAAGGA 420 TTTTCGCGAA TGGATAAATG GAAAGACACT GACAATGACG AGCTCAAAGT CTTTTTTGCA 480 GTAATGTTAC TGCAAGGTAT TGTGCAGAAA CCTGAGCTGG AGATGTTTTG GTCAACAAGG 540 CCTCTTTTGG ATACACCTTA TCTCAGGCAA ATTATGACTG GTGAAAGATT TTTACTTTTG 600 TTTCGGTGCC TGCATTTTGT CAACAATTCT TCTATATCTG CTGGTCAATC AAAGGCCCAG 660 ATTTCATTGC AGAAGATCAA ACCTGTGTTC GACTTTCTTG TAAATAAATT TTCCACTGTA 720 TATACTCCAA ACAGAAACAT TGCAGTTGAT GAATCACTGA TGCTGTTCAA GGGGCCATTA 780 GCTATGAAGC AGTACCTCCC GACAAAACGA GTACGATTTG GTCTGAAGCT ATATGTACTT 840 TGTGAAAGTC AGTCTGGTTA TGTGTGGAAT GCGCTTGTTC ACACAGGGCC TGGCATGAAT 900 TTGAAAGATT CAGCGGATGG CCTGAAATCA TCACGCATTG TTCTTACCTT GGTCAATGAC 960 CTTCTTGGCC AAGGGTATTG TGTCTTCCTC GATAACTTTA ATATATCTCC CATGCTTTTC 1020 AGAGAATTAC ATCAAAATAG GACTGATGCA GTTGGGACAG CTCGTTTGAA CAGAAAACAG 1080 ATTCCAAATG ATCTGAAAAA AAGGATTGCA AAGGGGACGA CTGTAGCCAG ATTCTGTGGT 1140 GAACTTATGG CACTGAAATG GTGTGACGGC AAGGAGGTGA CAATGTTGTC AACATTCCAC 1200 AATGATACTG TGATTGAAGT AAACAATAGA AATGGAAAGA AAACTAAAAG GCCACGTGTC 1260 ATTGTGGATT ATAACGAGAA TATGGGAGCA GTGGACTCGG CTGATCAAAT GCTTACTTCT 1320 TATCCATCTG AGCGCAAAAG ACACAAGGTT TGGTATAAGA AATTCTTTCA CCATCTTCTA 1380 CACATTACAG TGCTGAACTC CTACATCCTG TTCAAGAAGG ATAATCCTGA GCACACGATG 1440 AGCCATATAA ACTTCAGACT GGCATTGATT GAAAGAATGC TGGAAAAGCA TCACAAGCCA 1500 GGGCAGCAAC ATCTTCGAGG TCGTCCTTGC TCCGATGATG TCACACCTCT TCGTCTGTCT 1560 GGAAGACATT TCCCCAAGAG CATACCAGCA ACGTCCGGGA AACAGAATCC AACTGGTCGC 1620 TGCAAAATTT GCTGCTCCCA ATACGACAAG GATGGCAAGA AGATCCGGAA AGAAACGCGC 1680 TATTTTTGTG CCGAATGTGA TGTTCCGCTT TGTGTTGTTC CGTGCTTTGA AATTTACCAC 1740 ACGAAAAAAA ATTATTAA 1758 SEQ ID NO: 6: PGBD1 Amino Acid Sequence (809 Amino Acids). MYEALPGPAP ENEDGLVKVK EEDPTWEQVC NSQEGSSHTQ EICRLRFRHF CYQEAHGPQE 60 ALAQLRELCH QWLRPEMHTK EQIMELLVLE QFLTILPKEL QPCVKTYPLE SGEEAVTVLE 120 NLETGSGDTG QQASVYIQGQ DMHPMVAEYQ GVSLECQSLQ LLPGITTLKC EPPQRPQGNP 180 QEVSGPVPHG SAHLQEKNPR DKAVVPVFNP VRSQTLVKTE EETAQAVAAE KWSHLSLTRR 240 NLCGNSAQET VMSLSPMTEE IVTKDRLFKA KQETSEEMEQ SGEASGKPNR ECAPQIPCST 300 PIATERTVAH LNTLKDRHPG DLWARMHISS LEYAAGDITR KGRKKDKARV SELLQGLSFS 360 GDSDVEKDNE PEIQPAQKKL KVSCFPEKSW TKRDIKPNFP SWSALDSGLL NLKSEKLNPV 420 ELFELFFDDE TFNLIVNETN NYASQKNVSL EVTVQEMRCV FGVLLLSGFM RHPRREMYWE 480 VSDTDQNLVR DAIRRDRFEL IFSNLHFADN GHLDQKDKFT KLRPLIKQMN KNFLLYAPLE 540 EYYCFDKSMC ECFDSDQFLN GKPIRIGYKI WCGTTTQGYL VWFEPYQEES TMKVDEDPDL 600 GLGGNLVMNF ADVLLERGQY PYHLCFDSFF TSVKLLSALK KKGVRATGTI RENRTEKCPL 660 MNVEHMKKMK RGYFDFRIEE NNEIILCRWY GDGIISLCSN AVGIEPVNEV SCCDADNEEI 720 PQISQPSIVK VYDECKEGVA KMDQIISKYR VRIRSKKWYS ILVSYMIDVA MNNAWQLHRA 780 CNPGASLDPL DFRRFVAHFY LEHNAHLSD 809 SEQ ID NO: 7: PGBD2 Amino Acid Sequence (592 Amino Acids). MASTSRDVIA GRGIHSKVKS AKLLEVLNAM EEEESNNNRE EIFIAPPDNA AGEFTDEDSG 60 DEDSQRGAHL PGSVLHASVL CEDSGTGEDN DDLELQPAKK RQKAVVKPQR IWTKRDIRPD 120 FGSWTASDPH IEDLKSQELS PVGLFELFFD EGTINFIVNE TNRYAWQKNV NLSLTAQELK 180 CVLGILILSG YISYPRRRMF WETSPDSHHH LVADAIRRDR FELIFSYLHF ADNNELDASD 240 RFAKVRPLII RMNCNFQKHA PLEEFYSFGE SMCEYFGHRG SKQLHRGKPV RLGYKIWCGT 300 TSRGYLVWFE PSQGTLFTKP DRSLDLGGSM VIKFVDALQE RGFLPYHIFF DKVFTSVKLM 360 SILRKKGVKA TGTVREYRTE RCPLKDPKEL KKMKRGSFDY KVDESEEIIV CRWHDSSVVN 420 ICSNAVGIEP VRLTSRHSGA AKTRTQVHQP SLVKLYQEKV GGVGRMDQNI AKYKVKIRGM 480 KWYSSFIGYV IDAALNNAWQ LHRICCQDAQ VDLLAFRRYI ACVYLESNAD TTSQGRRSRR 540 LETESRFDMI GHWIIHQDKR TRCALCHSQT NTRCEKCQKG VHAKCFREYH IR 592 SEQ ID NO: 8: PGBD3 Amino Acid Sequence (593 Amino Acids). MPRTLSLHEI TDLLETDDSI EASAIVIQPP ENATAPVSDE ESGDEEGGTI NNLPGSLLHT 60 AAYLIQDGSD AESDSDDPSY APKDDSPDEV PSTFTVQQPP PSRRRKMTKI LCKWKKADLT 120 VQPVAGRVTA PPNDFFTVMR TPTEILELFL DDEVIELIVK YSNLYACSKG VHLGLTSSEF 180 KCFLGIIFLS GYVSVPRRRM FWEQRTDVHN VLVSAAMRRD RFETIFSNLH VADNANLDPV 240 DKFSKLRPLI SKLNERCMKF VPNETYFSFD EFMVPYFGRH GCKQFIRGKP IRFGYKFWCG 300 ATCLGYICWF QPYQGKNPNT KHEEYGVGAS LVLQFSEALT EAHPGQYHFV FNNFFTSIAL 360 LDKLSSMGHQ ATGTVRKDHI DRVPLESDVA LKKKERGTFD YRIDGKGNIV CRWNDNSVVT 420 VASSGAGIHP LCLVSRYSQK LKKKIQVQQP NMIKVYNQFM GGVDRADENI DKYRASIRGK 480 KWYSSPLLFC FELVLQNAWQ LHKTYDEKPV DFLEFRRRVV CHYLETHGHP PEPGQKGRPQ 540 KRNIDSRYDG INHVIVKQGK QTRCAECHKN TTFRCEKCDV ALHVKCSVEY HTE 593 SEQ ID NO: 9: PGBD5 Amino Acid Sequence (524 Amino Acids). MAEGGGGARR RAPALLEAAR ARYESLHISD DVFGESGPDS GGNPFYSTSA ASRSSSAASS 60 DDEREPPGPP GAAPPPPRAP DAQEPEEDEA GAGWSAALRD RPPPRFEDTG GPTRKMPPSA 120 SAVDFFQLFV PDNVLKNMVV QTNMYAKKFQ ERFGSDGAWV EVTLTEMKAF LGYMISTSIS 180 HCESVLSIWS GGFYSNRSLA LVMSQARFEK ILKYFHVVAF RSSQTTHGLY KVQPFLDSLQ 240 NSFDSAFRPS QTQVLHEPLI DEDPVFIATC TERELRKRKK RKFSLWVRQC SSTGFIIQIY 300 VHLKEGGGPD GLDALKNKPQ LHSMVARSLC RNAAGKNYII FTGPSITSLT LFEEFEKQGI 360 YCCGLLRARK SDCTGLPLSM LTNPATPPAR GQYQIKMKGN MSLICWYNKG HFRFLTNAYS 420 PVQQGVIIKR KSGEIPCPLA VEAFAAHLSY ICRYDDKYSK YFISHKPNKT WQQVFWFAIS 480 IAINNAYILY KMSDAYHVKR YSRAQFGERL VRELLGLEDA SPTH 524 SEQ ID NO: 10: Myotis lucifugus (Wild-type) Amino Acid Sequence with Hyperactive Mutations (S8P, C13R, N125K) 572 Amino Acids. MSQHSDYPDD EFRADKLSNY SCDSDLENAS TSDEDSSDDE VMVRPRTLRR RRISSSSSDS 60 ESDIEGGREE WSHVDNPPVL EDFLGHQGLN TDAVINNIED AVKLFIGDDF FEFLVEESNR 120 YYNQKRNNFK LSKKSLKWKD ITPQEMKKFL GLIVLMGQVR KDRRDDYWTT EPWTETPYFG 180 KTMTRDRFRQ IWKAWHFNNN ADIVNESDRL CKVRPVLDYF VPKFINIYKP HQQLSLDEGI 240 VPWRGRLFFR VYNAGKIVKY GILVRLLCES DTGYICNMEI YCGEGKRLLE TIQTVVSPYT 300 DSWYHIYMDN YYNSVANCEA LMKNKFRICG TIRKNRGIPK DFQTISLKKG ETKFIRKNDI 360 LLQVWQSKKP VYLISSIHSA EMEESQNIDR TSKKKIVKPN ALIDYNKHMK GVDRADQYLS 420 YYSILRRTVK WTKRLAMYMI NCALFNSYAV YKSVRQRKMG FKMFLKQTAI HWLTDDIPED 480 MDIVPDLQPV PSTSGMRAKP PTSDPPCRLS MDMRKHTLQA IVGSGKKKNI LRRCRVCSVH 540 KLRSETRYMC KFCNIPLHKG ACFEKYHTLK NY 572 SEQ ID NO: 11: Myotis lucifugus Corrected Amino Acid Sequence with Hyperactive Mutations (S8P, C13R) 572 Amino Acids. MAQHSDYPDDEFRADKLSNYSCDSDLENASTSDEDSSDDEVMVRPRTLRRRRISSSSSDSESDIEGGREEWSHVDNPPVLEDFLGH QGLNTDAVINNIEDAVKLFIGDDFFEFLVEESNRYYNQNRNNFKLSKKSLKWKDITPQEMKKFLGLIVLMGQVRKDRRDDYWTTEP WTETPYFGKTMTRDRFRQIWKAWHFNNNADIVNESDRLCKVRPVLDYFVPKFINIYKPHQQLSLDEGIVPWRGRLFFRVYNAGKIV KYGILVRLLCESDTGYICNMEIYCGEGKRLLETIQTVVSPYTDSWYHIYMDNYYNSVANCEALMKNKFRICGTIRKNRGIPKDFQT ISLKKGETKFIRKNDILLQVWQSKKPVYLISSIHSAEMEESQNIDRTSKKKIVKPNALIDYNKHMKGVDRADQYLSYYSILRRTVK WTKRLAMYMINCALFNSYAVYKSVRQRKMGFKMFLKQTAIHWLTDDIPEDMDIVPDLQPVPSTSGMRAKPPTSDPPCRLSMDMRKH TLQAIVGSGKKKNILRRCRVCSVHKLRSETRYMCKFCNIPLHKGACFEKYHTLKNY SEQ ID NO: 12: Pteropus vampyrus Left End Sequence Sequence 381 bp. TTAACCCATT TCCTGTTTGC CCCGAGAATA CTCACCAGCG GCACTTGCAG CTGCAGCGTT 60 TACCCCGAGA TAACTCGTCG ATTACAGTCC TAACCTTACC CCCAAAGTTT GCCATGAAAT 120 ATCTCGCTTT TATTATTATT TTCGCATCGC TCTAGTATAT CGATAGTCTT TGGAAACAAA 180 TGACATCATT CTATTTACAG CATTCTGTTT TTAGTAGTGG TATTTCCATT TACAAAATAT 240 AGTAATTTTC TATCGCTGAA AATGTCAAAT CCTAGAAAAC GTAGCATTCC TACATGTGAT 300 GTTAACTTCG TTCTCGAACA GTTGTTAGCC GAAGATTCAT TTGATGAATC CGATTTTTCC 360 GAAATAGACG ATTCTGATGA T 381 SEQ ID NO: 13: PGBD4 Left End Nucleotide Sequence Sequence 373 bp. TTAACTCATT TCTCCTTAGC CCCGAGATTA CGCGCTGCTG TGCCTGCGAC TGCAGCGTTT 60 ACGCCGAGAT AACTCGTGGA TTACAGTGCC AACCTTACTC CCAAAGTTTG CCACGAAATA 120 TCTCGCTTCT GTTATTTTCG CATGGTTCTG GTATATTGAC TTTTGAAACA AAAGACATCA 180 TTCTGTTTAT AGCATTCTGT TTTTAGTAGT GGGATTTCCA TCTACAAAAT ATAGTAATTC 240 TCGATCGCTG AAATGTCAAA TCCTAGAAAA CGTAGCATTC CTATGCGTGA TAGTAATACC 300 GGTCTCGAAC AGTTGTTGGC TGAAGATTCA TTTGATGAAT CTGATTTTTC GGAAATAGAT 360 GATTCTGATA ATT 373 SEQ ID NO: 14: MER75 Left End Nucleotide Sequence Sequence 344 bp. TTAACCCTTT TCCCGTTTGC CCCGAGAATA CTCGCCGGCG GCGCTTGCGG CTGCAGCGTT 60 TACCCCGAGA TAACTTTGCC ACGAAATATC TCGCTTTTAT TATTATTTTC GCATCGCTCT 120 AGTATATCGA CTTTGGAAAC AAAAGACATC ATTCTATTTA TAGCATTCTG TTTTTAGTAG 180 TGGTATTTCC ATTTACAAAA TATAGTAATT CTCGATCGCT GAAAATGTCA AATCCTAGAA 240 AACGTAGCAT TCCTACGCGT GATGTTAACA TCGTTCTCGA ACAGTTGTTG GCCGAAGATT 300 CATTTGATGA ATCCGATTTT TCCGAAATAG ACGATTCTGA TGAT 344 SEQ ID NO: 15: MER75B Left End Nucleotide Sequence Sequence 91 bp. TTAACCCATT TCCCGTTTGC CCCGAGAATA CTCTTGTCTC TAATCCTAAT GTAACATCAT 60 ATACATTTCT GTTACATTAG GATTAGAGAC A 91 SEQ ID NO: 16: MER75A Left End Nucleotide Sequence Sequence 32 bp. TTAACCCATT TCCCGTTTGC CCCGAGAATA CT 32 SEQ ID NO: 17: Pteropus vampyrus Right End Sequence Sequence 171 bp. TAGGATTAGA GACAAGTTCT GTTTAGAAAT AACTCCAAGA ACAGTTTTTA TATTTTATTT 60 TCACATTGAA AACCAGTCAG ATTTGCTTCA GCCTCAAAGA GCATGTTTAT GTAAAATTAA 120 ATTAACGCTG GCAGCGAGCT GCACTTTTTT TCTAAACGGG AAATGGGTTA A 171 SEQ ID NO: 18: PGBD4 Right End Nucleotide Sequences Sequence 176 bp. CCTGGGATTA TAGGCATGAG CCACTGCGCC TAGCACCAAG AACAGTTTTT ATATTTTATT 60 TTCACATTGA AAATCAGTCA GATTTGCTTC AGCCTCAAAG AGGGTGTTTA TGTAAAACTA 120 AATGAGTGCA GGCAGCGAGC TACACTTTTT TTTTTCCTAA ATGGAAAATG GGTTAA 176 SEQ ID NO: 19: MER75 Right End Nucleotide Sequences Sequence 178 bp. TCAGACGATT CTGATGTTAG TTCTGTTTAG AAATAACTCC AAGAACAGTT TTTATATTTT 60 ATTTTCACAT TGAAAATCAG TCAGATTTGC TTCAGCCTCA AAGAGCGTGT TTATGTAAAA 120 TTAAATGAGC GCTGGCAGCG AGCTGCACTT TTTTTTTTCT AAACGGGAAA AGGGTTAA 178 SEQ ID NO: 20: MER75B Right End Nucleotide Sequences Sequence 160 bp. AGTTCTGTTT AGAAATAACT CCAAGAACAG TTTTTATATT TTATTTTCAC ATTGAAAATC 60 AGTCAGATTT GCTTCAGCCT CAAAGAGCGT GTTTATGTAA AATTAAATGA GCGCTGGCAG 120 CGAGCTGCAC TTTTTTTTTT CTAAACGGGA AAAGGGTTAA 160 SEQ ID NO: 441: MER75A Right End Nucleotide Sequences Sequence 46 bp. CGCTGGCAGC GAGCTGCACT TTTTTTCTAA ACGGGAAATG GGTTAA 46 SEQ ID NO: 429: Extended Pteropus vampyrus Nucleotide Sequence* 2210 BP CCCATTTCCT GTTTGCCCCG AGAATACTCA CCAGCGGCAC TTGCAGCTGC AGCGTTTACC 60 CCGAGATAAC TCGYCGATTA CAGTCCTAAC CTTACCCCCA AAGTTTGCCA TGAAATATCT 120 CGCTTTTATT ATTATTTTCG CATCGCTCTA GTATATCGAT AGTCTTTGGA AACAAATGAC 180 ATCATTNTAT TTACAGCATT CTGTTTTTAN TAGTGGTATT TCCATTTACA AAATATAGTA 240 ATTTTCTATC GCTGAAAATG TCAAATCCTA GAAAACGTAG CATTCCTACA TGTGATGTTA 300 ACTTCGTTCT CGAACAGTTG TTAGCCGAAG ATTCATTTGA TGAATCCGAT TTTTCCGAAA 360 TAGACGATTC TGATGATTTT TCGGATAGTG CTTCGGAAGA CTATACGGTC AGGCCTCCGT 420 CCGATTCGGA ATCTGATGGA AATAGCCCTA CATCAGCTGA CTCGGGTCGC GCTCTGAAAT 480 GGTCAACTCG TGTTATGATT CCACGTCAAA GGTATGACTT TACCGGCACA CCTGGCAGAA 540 AAGTTGATGT CAGTGATACC ACTGACCCAC TGCAGTATTT TGAACTGTTC TTTACTGAGG 600 AATTAGTTTC AAAAATTACC AGTGAAATGA ATGCCCAAGC TGCCTTGTTG GCTTCAAAGC 660 CACCTGGTCC GAAAGGATTT TCGCGAATGG ATAAATGGAA AGACACTGAC AATGATGAAC 720 TGAAAGTCTT TTTTGCAGTA ATGTTACTGC AAGGTATTGT GCAGAAACCT GAGCTGGAGA 780 TGTTTTGGTC GACAAGGCCT CTTTTGGATA TACCTTATCT CAGGCAAATT ATGACTGGTG 840 AAAGATTTTT ACTTTTGCTT CGGTGCCTGC ATTTTGTCAA CAATTCTTCC ATATCCGCTG 900 GTCAATCAAA GGCCCAGATT TCATTGCAGA AGATCAAACC TGTGTTCGAC TTTCTTGTAA 960 ATAAGTTTTC AACTGTATAT ACTCCAAACA GAAACATTGC AGTCGATGAA TCACTGATGC 1020 TGTTCAAGGG GCGGTTAGCT ATGAAGCAGT ACATCCCGAC GAAATGtGCA CGATTTGGTC 1080 TCAAGCTNTA TGTACTTTGT GAAAGTCAAT CTGGTTACGT GTGGAATGCG CTTGTTCACA 1140 CAGGGCCCAG TATGAATTTG AAAGATTCAG CTGATGGTCT GAAATCGTCA TGCATTGTTC 1200 TTACCTTGGT CAATGACCTT CTTGGCCAAG GATATTGTGT CTTCCTCAAT AACTTTTATA 1260 CATCTCCCAT GCTTTTCAGA GAATTACATC AAAACAGGAC TGATGCAGTT GGGACAGCTC 1320 GTTTGAACAG AAAACAGATG CCAAATGATC TGAAAAAAAG GATTGCAAAG GGGACGACTG 1380 TAGCCAGATT CTGTGGTGAA CTTATGGCAC TGAAATGGTG TGACAAGAAG GAGGTGACAA 1440 TGTTGTCAAC ATTCCACAAT GATACTGTGA TTGAAGTAGA CAACAGAAAT GGAAAGAAAA 1500 CTAAGAAGCC ATGTGTCATT GTGGATTATA ACGAGAATAT GGGAGCAGTG GACTCGGCTG 1560 ATCAGATGCT CACTTCTTAT CCAACTGAGC GCAAAAGGCA CAAGTTTTGG TATAAGAAAT 1620 TCTTTCGCCA CCTTCTAAAC ATTACAGTGC TGAACTCCTA CATCCTGTTC AAGAAGGACA 1680 ATCCTGAGCA CACGATCAGC CATGTAAACT TCAGACTGAC GTTGATTGAA AGAATGCTGG 1740 AAAAGCATCA CAAGCCAGGG CAGCAACGTC TTCGAGGTCG TCCGTGCTCT GATGATGTCA 1800 CACCTCTTCG CCTGTCTGGA AGACATTTCC CCAAGAGCAT ACCACCAACA TCAGGGAAAC 1860 AGAATCCAAC TGGTCGCTGC AAAGTTTGCT GCTCGCACGA CAAGGATGGC AAGAAGATCC 1920 GGAGAGAAAC GTtATATTTT TGTGCGGAAT GTGATGTTCC GCTTTGTGTT GTTCCGTGCT 1980 TTGAAATTTA CCACACGAAA AAAAATTATT AAATACTGAT CATCATATAC ATTTCTGTTA 2040 CATTAGGATT AGAGACAAGT TCTGTTTAGA AATAACTCCA AGAACAGTTT TTATATTTTA 2100 TTTTCACATT GAAAACCAGT CAGATTTGCT TCAGCCTCAA AGAGCATGTT TATGTAAAAT 2160 TAAATTAACG CTGGCAGCGA GCTGCACTTN TTTTCTAAAC GGGAAATGGG 2210 SEQ ID NO: 430: Extended Pteropus vampyrus Amino Acid Sequence 584 Amino Acids. MSNPRKRSIP TCDVNFVLEQ LLAEDSFDES DFSEIDDSDD FSDSASEDYT VRPPSDSESD 60 GNSPTSADSG RALKWSTRVM IPRQRYDFTG TPGRKVDVSD TTDPLQYFEL FFTEELVSKI 120 TSEMNAQAAL LASKPPGPKG FSRMDKWKDT DNDELKVFFA VMLLQGIVQK PELEMFWSTR 180 PLLDIPYLRQ IMTGERFLLL LRCLHFVNNS SISAGQSKAQ ISLQKIKPVF DFLVNKFSTV 240 YTPNRNIAVD ESLMLFKGRL AMKQYIPTKC ARFGLKLYVL CESQSGYVWN ALVHTGPSMN 300 LKDSADGLKS SCIVLTLVND LLGQGYCVFL NNFYTSPMLF RELHQNRTDA VGTARLNRKQ 360 MPNDLKKRIA KGTTVARFCG ELMALKWCDK KEVTMLSTFH NDTVIEVDNR NGKKTKKPCV 420 IVDYNENMGA VDSADQMLTS YPTERKRHKF WYKKFFRHLL NITVLNSYIL FKKDNPEHTI 480 SHVNFRLTLI ERMLEKHHKP GQQRLRGRPC SDDVTPLRLS GRHFPKSIPP TSGKQNPTGR 540 CKVCCSHDKD GKKIRRETLY FCAECDVPLC VVPCFEIYHT KKNY SEQ ID NO: 431: A MLT left end (5’ to 3’) is as follows: TTAACACTTGGATTGCGGGAAACGAGTTAAGTCGGCTCGCGTGAATTGCGCGTACTCCGCGGGAGCCGTC TTAACTCGGTTCATATAGATTTGCGGTGGAGTGCGGGAAACGTGTAAACTCGGGCCGATTGTAACTGCGT ATTACCAAATATTTGTT SEQ ID NO: 432: A MLT right end (5’ to 3’) is as follows: AATTATTTATGTACTGAATAGATAAAAAAATGTCTGTGATTGAATAAATTTTCATTTTTTACAC AAGAAACCGAAAATTTCATTTCAATCGAACCCATACTTCAAAAGATATAGGCATTTTAAACTAA CTCTGATTTTGCGCGGGAAACCTAAATAATTGCCCGCGCCATCTTATATTTTGGCGGGAAATTC ACCCGACACCGTGGTGTTAA SEQ ID NO: 433: Trichnoplusia ni 1 MGSSLDDEHI LSALLQSDDE LVGEDSDSEI SDHVSEDDVQ SDTEEAFIDE VHEVQPTSSG 61 SEILDEQNVI EQPGSSLASN KILTLPQRTI RGKNKHCWST SKSTRRSRVS ALNIVRSQRG 121 PTRMCRNIYD PLLCFKLFFT DEIISEIVKW TNAEISLKRR ESMTGATFRD TNEDEIYAFF 181 GILVMTAVRK DNHMSTDDLF DRSLSMVYVS VMSRDRFDFL IRCLRMDDKS IRPTLRENDV 241 FTPVRKIWDL FIHQCIQNYT PGAHLTIDEQ LLGFRGRCPF RMYIPNKPSK YGIKILMMCD 301 SGTKYMINGM PYLGRGTQTN GVPLGEYYVK ELSKPVRGSC RNITCDNWFT SIPLAKNLLQ 361 EPYKLTIVGT VRSNKREIPE VLKNSRSRPV GTSMFCFDGP LTLVSYKPKP AKMVYLLSSC 421 DEDASINEST GKPQMVMYYN QTKGGVDTLD QMCSVMTCSR KTNRWPMALL YGMINIACIN 481 SFIIYSHNVS SKGEKVQSRK KFMRNLYMSL TSSFMRKRLE APTLKRYLRD NISNILPNEV 541 PGTSDDSTEE PVTKKRTYCT YCPSKIRRKA NASCKKCKKV ICREHNIDMC QSCF SEQ ID NO: 434: Pteropus vampyrus 1 MSNPRKRSIP TCDVNFVLEQ LLAEDSFDES DFSEIDDSDD FSDSASEDYT VRPPSDSESD 61 GNSPTSADSG RALKWSTRVM IPRQRYDFTG TPGRKVDVSD TTDPLQYFEL FFTEELVSKI 121 TSEMNAQAAL LASKPPGPKG FSRMDKWKDT DNDELKVFFA VMLLQGIVQK PELEMFWSTR 181 PLLDIPYLRQ IMTGERFLLL LRCLHFVNNS SISAGQSKAQ ISLQKIKPVF DFLVNKFSTV 241 YTPNRNIAVD ESLMLFKGRL AMKQYIPTKM NLKDSADGLK SEQ ID NO: 435: Myotis myotis (“2a”) 1 MDLRCQHTVL SIRESRGLLP NLKMKTSRMK KGDIIFSRKG DILLLAWKDK RVVRMISIHD 61 TSVSTTGKKN RKTGENIVKP ACIKEYNAHM KGVDRADQFL SCCSILRKMM KWTKKVVLYL 121 INCGLFNSFR VYNVLNPQAK MKYKQFLLSV ARDWIMDDNN EGSPEPETNL SSPSPGGARR 181 APRKDPPKRL SGDMKQHEPT CIPASGKKKF PTRACRVCAH GKRSESRYLC KFCLVPLHRG 241 KCFTQYHTLK KY SEQ ID NO: 436: Myotis myotis (“1”) 1 MKAFLGVILN MGVLNHPNLQ SYWSMDFESH IPFFRSVFKR ERFLQIFWML HLKNDQKSSK 61 DLRTRTEKVN CFLSYLEMKF RERFCPGREI AVDEAVVGFK GKIHFITYNP KKPTKWGIRL 121 YVLSDSKCGY VHSFVPYYGG ITSETLVRPD LPFTSRIVLE LHERLKNSVP GSQGYHFFTD 181 RYYTSVTLAK ELFKEKTHLT GTIMPNRKDN PPVIKHQKLK KGEIVAFRDE NVMLLAWKDK 241 RIVTLSTWDS ETESVERRVG GGKEIVLKPK VVTNYTKFMG GVDIADYTST YCFMRKTLKW 301 WRTLFFWGLE VSVVNSYILY KECQKRKNEK PITHVKFIRK LVHDLVGEFR DGTLTSRGRL 361 LSTNLEQRLD GKLHIITPHP NKKHKDCVVC SNRKIKGGRR ETIYICETCE CKPGLHVGEC 421 FKKYHTMKNY RD SEQ ID NO: 437: Myotis lucifugus (“2”) 1 MPSLRKRKET NETDTLPEVF NDNLSDIPSE IEDADDCFDD SGDDSTDSTD SEIIRPVRKR 61 KVAVLSSDSD TDEATDNCWS EIDTPPRLQM FEGHAGVTTF PSQCDSVPSV TNLFFGDELF 121 EMLCKELSNY HDQTAMKRKT PSRTLKWSPV TQKDIKKFLG LIILMGQTRK DSLKDYWSTD 181 PLICTPIFPQ TMSRHRFEQI WTFWHFNDNA KMDSRSGRLF KIQPVLDYFL HKFRTIYKPK 241 QQLSLDEGMI PWRGRFKFRT YNPAKITKYG LLVRMVCESD TGYICSMEIY TAEGRKLQET 301 VLSVLGPYLG IWHHIYQDNY YNATSTAELL LQNKTRVCGT IRESRGLPPN LEMKTSRMKK 361 GDIIFSRKGD ILLLAWKDKR VVRMISTIHD TSVSTTGKKN RKTGENIVKP TCIKEYNAHM 421 KGVDRADQFL SCCSILRKTM KWTKKVVLYL INCGLFNSFR VYNVLNPQAK MKYKQFLLSV 481 ARDWITDDNN EGSPEPETNL SSPSPGGARR APRKDPPKRL SGDMKQHEPT CIPASGKKKF 541 PTRACRVCAA HGKRSESRYL CKFCLVPLHR GKCFTQYHTL KKYMDLRCQH TVLSTVGRGY 601 SVLARFKPRT NERTGSSHCH VQVPAGGQGP PSTIIANGCG CKLEPMVRTR SPTCLVIEFG 661 CM SEQ ID NO: 438: Myotis myotis (“2”) 1 MPSLRKRKET NETDTLPEVF NDNLSDIPSE IEDADDCFDD SGDDSTDSTE SEIIRPVRKR 61 KVAVLSSDSN TDEATDNCWS EIDTPPRLQM FEGHAGVTTF PSQCDSVPSV TNLFFGDELF 121 EMLCKELSNY HDQTAMKRKT PSRTLKWSPV TQKDIKKFLG LIILMGQTRK DSWKDYWSTD 181 PLICTPIFPQ TMSRHRFEQI WTFWHFNDNA KMDSCSGRLF KIQPVLDYFL HKFRTIYKPK 241 QQLSLDEGMI PWRGRLKFTY NPAITKYGLL VRMVCESDTG YICNMEIYTA ERKKLQETVL 301 SVLGPYLGIW HHIYQDNYYN ATSTAELLLQ NKTRVCGTIR ESRGLPPNLK MKTSRMKKGD 361 IIFSRKGDIL LLAWKDKRVV RMISTIHDTS VSTTGKKNRK TGENIVKPTC IKEYNAHMKG 421 VDRADQFLSC CSILRKTTKW TKKVVLYLIN CGLFNSFRVY NILNPQAKMK YKQFLLSVAR 481 DWITDDNNEG SPEPETNLSS PSSGGARRAP RKDQPKRLSG DMKQHEPTCI PASGKKKFPT 541 ACRVCAAHGK RSESRYLRKF CFVPLRGKCF MYHTLKKYSE LFSLIVVSKI QNVIIYKTTK 601 VYMRYVMRSH CPLSFLVFAP SVKDRSRVFS FFTRHLLWTL DVNTLSCPHR MKRSHWWKPC 661 RSIYEKLYNC TNP SEQ ID NO: 439: Myotis myotis (“2b”) 1 MDLRCQHTVL SIRESRGLPP NLKMKTSRMK KGDIIFSRKG DILLLAWKDK RVVRMISTIH 61 DTSVSTTGKK NRKTGENIVK PACIKEYNAH MKGVDRADQF LSCCSILRKT MKWTKKVVLY 121 LINCGLFNSF RVYNVLNPQA KMKYKQFLLS VARDWITDDN NEGSPEPETN LSSPSPGGAR 181 RAPRKDPPKR LSGDMKQHEP TCIPASGKKK FPTRACRVCA AHGKRSESRY LCKFCLVPLH 241 RGKCFTQYHT LKKY TTAA (SEQ ID NO: 440) SEQ ID NO: 816: Tet-operator-containing AAV2 P40 promoter with a modified AAV2 P40 TATA element gaaggtcaccaagcaggaagtcaaagactttttccggtgggcaaaggatcacgtggttgaggtggagcatgaattctacgtcaaaaaggg tggagccaagaaaagacccgcccccagtgacgcaggtatataagtgagcccCTCCCTATCAGTGATAGAGATCTCCCTATCAGTGATA GAGACCaaCcgggtgcgcgagtcagtt gcgcagccat cgCcgtcagagcggaagct tcgatcaact tcgcagacag SEQ ID NO: 817: Modified AAV2 P40 intron_2 GggtGaGTaCc aaCtgttctc TaCgctgtttc cctgcaAaca Cgcgaaga Ttgaatcaga attcaaatCt ctgcttcact cacggacCga gactgttt gtgcttt cccgtgtcag aatctcaacccgtttctgtc gtcaagg cgtatcagaa actgtgctac attcatcata tcTtgggaaaggtgccagac gcttgcactg cctgcgatct ggtcaCtgtg gatttggCtaactgcatctttccacCatTT aCCa tttT tcaggt SEQ ID NO: 818: Modified AAV2 P40 intron_1 (a at position 276 substituted for C), in which aataaa is changed to Cataaa: gtaccaaaac aaatgttctc gtcacgtgggcatgaatctg atgctgtttc cctgcagaca atgcgagaga atgaatcaga attcaaatat ctgcttcact cacggacaga aagactgttt agagtgcttt cccgtgtcag aatctcaacccgtttctgtc gtcaaaaagg cgtatcagaa actgtgctac attcatcata tcatgggaaaggtgccagac gcttgcactg cctgcgatct ggtcaatgtg gatttggatg actgcatctt tgaacCataa atga tttaaa tcaggt atggctgc cgatggttatcttccag SEQ ID NO: 819: Modified AAV2 P40 intron_2, which contains a-C change and the potential translation start site ATGs have been changed to either Ctg, aCg, or Ttg: gtaccaaaac aaCtgttctc gtcacgtgggcCtgaatctg aCgctgtttc cctgcagaca Ctgcgagaga Ttgaatcaga attcaaatCt ctgcttcact cacggacaga aagactgttt agagtgcttt cccgtgtcag aatctcaacccgtttctgtc gtcaaaaagg cgtatcagaa actgtgctac attcatcata tcTtgggaaaggtgccagac gcttgcactg cctgcgatct ggtcaCtgtg gatttggCtg actgcatctt tgaacCataa aCga tttaaa tcaggt atggctgc cgatggttatcttccag SEQ ID NO: 820: tetO-containing P40 promoter with a modified TATA element (AAV2) GAAGGTCACCAAGCAGGAAGTCAAAGACTTTTTCCGGTGGGCAAAGGATCACGTGGTTGAGGTGGAGCATGAA TTCTACGTCAAAAAGGGTGGAGCCAAGAAAAGACCCGCCCCCAGTGACGCAGGTATATAAGTGAGCCCCTCTC CCTATCAGTGATAGAGATCTCCCTATCAGTGATGAGCCCAAACGGGTGCGCGAGTCAGTTGCGCAGCCATCGA CGTCAGACGCGGAAGCTTCGATCAACT ACGCAGACAG SEQ ID NO: 821: AAV9 P40 intron gtac caaaacaaat gttctcgtca cgcgggcatg cttcagatgc tgcttccctg caaaacgtgc gagagaatga atcagaattt caacatttgc ttcacacacg gggtcagaga ctgctcagag tgtttccccg gcgtgtcaga atctcaaccg gtcgtcagaa agaggacgta tcggaaactc tgtgcgattc atcatctgct ggggcgggct cccgagattg cttgctcggc ctgcgatctg gtcaacgtgg acctggatga ctgtgtttct gagcaataaa tgacttaaac caggtatggc tgccgatggt tatcttccag attggctcga ggacaacct SEQ ID NO: 822: tetO-containing P40 promoter with a modified TATA element (upper case) and AAV9 P40 intron (lower case): GAAGGTCACCAAGCAGGAAGTCAAAGACTTTTTCCGGTGGGCAAAGGATCACGTGGTTGAGGTGGAGCATGAA TTCTACGTCAAAAAGGG TGGAGCCAAGAAAAGACCCGCCCCCAGTGACGCAGGTATATAAGTGAGCCCCTCTCCCTATCAGTGATAGAGAT CTCCCTATCAGTGATGAGCCCAAACGGGTGCGCGAGTCAGTTGCGCAGCCATCGACGTCAGACGCGGAAGCTT CGATCAACT ACGCAGACAG gtac caaaacaaat gttctcgtca cgcgggcatg cttcagatgc tgcttccctg caaaacgtgc gagagaatga atcagaattt caacatttgc ttcacacacg gggtcagaga ctgctcagag tgtttccccg gcgtgtcaga atctcaaccg gtcgtcagaa agaggacgta tcggaaactc tgtgcgattc atcatctgct ggggcgggct cccgagattg cttgctcggc ctgcgatctg gtcaacgtgg acctggatga ctgtgtttct gagcaataaa tgacttaaac caggtatggc tgccgatggt tatcttccag attggctcga ggacaacct SEQ ID NO: 823: Tet-operator-containing AAV2 P40 promoter with a modified AAV2 P40 TATA element and modified AAV2 P40 intron_2 gaaggtcaccaagcaggaagtcaaagactttttccggtgggcaaaggatcacgtggttgaggtggagcatgaattctacgtcaaaaaggg tggagccaagaaaagacccgcccccagtgacgcaggtatataagtgagcccCTCCCTATCAGTGATAGAGATCTCCCTATCAGTGATA GAGACCaaCcgggtgcgcgagtcagtt gcgcagccat cgCcgtcagagcggaagct tcgatcaact tcgcagacag GggtGaGTaCc aaCtgttctc TaCgctgtttc cctgcaAaca Cgcgaaga Ttgaatcaga attcaaatCt ctgcttcact cacggacCga gactgttt gtgcttt cccgtgtcag aatctcaacccgtttctgtc gtcaagg cgtatcagaa actgtgctac attcatcata tcTtgggaaaggtgccagac gcttgcactg cctgcgatct ggtcaCtgtg gatttggCtaactgcatctttccacCatTT aCCa tttT tcaggt SEQ ID NO: 824: AAV2 CAP under the control of the tetO-containing AAV2 P40 promoter with a modified AAV2 P40 TATA element and AAV9 P40 intron and AAV9 poly A 5’- gaaggtcaccaagcaggaagtcaaagactttttccggtgggcaaaggatcacgtggttgaggtggagcatgaattctacgtcaaaaagggtggagccaagaaaa gacccgcccccagtgacgcaggtatataagtgagcccCTCCCTATCAGTGATAGAGATCTCCCTATCAGTGATAGAGACCaaacgg gtgcgcgagtcagttgcgcagccatcgacgtcagacgcggaagcttcgatcaactacgcagacaggtaccaaaacaaatgttctcgtcacgcgggcatgcttcag atgctgcttccctgcaaaacgtgcgagagaatgaatcagaatttcaacatttgcttcacacacggggtcagagactgctcagagtgtttccccggcgtgtcagaatctc aaccggtcgtcagaaagaggacgtatcggaaactctgtgcgattcatcatctgctggggcgggctcccgagattgcttgctcggcctgcgatctggtcaacgtggacc tggatgactgtgtttctgagcaataaatgacttaaaccaggtatggctgccgatggttatcttccagattggctcgaggacactctctctgaaggaataagacagtggtg gaagctcaaacctggcccaccaccaccaaagcccgcagagcggcataaggacgacagcaggggtcttgtgcttcctgggtacaagtacctcggacccttcaacg gactcgacaagggagagccggtcaacgaggcagacgccgcggccctcgagcacgacaaagcctacgaccggcagctcgacagcggagacaacccgtacct caagtacaaccacgccgacgcggagtttcaggagcgccttaaagaagatacgtcttttgggggcaacctcggacgagcagtcttccaggcgaaaaagagggttct tgaacctctgggcctggttgaggaacctgttaagacggctccgggaaaaaagaggccggtagagcactctcctgtggagccagactcctcctcgggaaccggaaa ggcgggccagcagcctgcaagaaaaagattgaattttggtcagactggagacgcagactcagtacctgacccccagcctctcggacagccaccagcagccccct ctggtctgggaactaatacgatggctacaggcagtggcgcaccaatggcagacaataacgagggcgccgacggagtgggtaattcctcgggaaattggcattgcg attccacatggatgggcgacagagtcatcaccaccagcacccgaacctgggccctgcccacctacaacaaccacctctacaaacaaatttccagccaatcagga gcctcgaacgacaatcactactttggctacagcaccccttgggggtattttgacttcaacagattccactgccacttttcaccacgtgactggcaaagactcatcaacaa caactggggattccgacccaagagactcaacttcaagctctttaacattcaagtcaaagaggtcacgcagaatgacggtacgacgacgattgccaataaccttacc agcacggttcaggtgtttactgactcggagtaccagctcccgtacgtcctcggctcggcgcatcaaggatgcctcccgccgttcccagcagacgtcttcatggtgccac agtatggatacctcaccctgaacaacgggagtcaggcagtaggacgctcttcattttactgcctggagtactttccttctcagatgctgcgtaccggaaacaactttacct tcagctacacttttgaggacgttcctttccacagcagctacgctcacagccagagtctggaccgtctcatgaatcctctcatcgaccagtacctgtattacttgagcagaa caaacactccaagtggaaccaccacgcagtcaaggcttcagttttctcaggccggagcgagtgacattcgggaccagtctaggaactggcttcctggaccctgttac cgccagcagcgagtatcaaagacatctgcggataacaacaacagtgaatactcgtggactggagctaccaagtaccacctcaatggcagagactctctggtgaat ccgggcccggccatggcaagccacaaggacgatgaagaaaagttttttcctcagagcggggttctcatctttgggaagcaaggctcagagaaaacaaatgtggac attgaaaaggtcatgattacagacgaagaggaaatcaggacaaccaatcccgtggctacggagcagtatggttctgtatctaccaacctccagagaggcaacaga caagcagctaccgcagatgtcaacacacaaggcgttcttccaggcatggtctggcaggacagagatgtgtaccttcaggggcccatctgggcaaagattccacac acggacggacattttcacccctctcccctcatgggtggattcggacttaaacaccctcctccacagattctcatcaagaacaccccggtacctgcgaatccttcgacca ccttcagtgcggcaaagtttgcttccttcatcacacagtactccacgggacaggtcagcgtggagatcgagtgggagctgcagaaggaaaacagcaaacgctgga atcccgaaattcagtacacttccaactacaacaagtctgttaatgtggactttactgtggacactaatggcgtgtattcagagcctcgccccattggcaccagatacctg actcgtaatctgtaattgcctgttaatcaataaaccggttaattcgtttcagttgaactttggtctctgcggtatttctttcttat – 3’ SEQ ID NO: 825: SFA2-EP40CAP2WZ1_SB: AAV2 CAP under the control of the tetO-containing hCMV-derived enhancer element 3/AAV2 P40 promoter with a modified AAV2 P40 TATA element and modified AAV2 P40 intron_1 (a at position 276 substituted for C) and WPRE/modified ICP27 poly A followed by ICP0 promoter-Zeocin resistant gene-ICP0 poly A cassette flanked with SB ITRs. 5’- GCGGCCGCTatacagttgaagtcggaagtttacatacacttaagttggagtcattaaaactcgtttttcaactactccacaaatttcttgttaacaaacaatagttttg gcaagtcagttaggacatctactttgtgcatgacacaagtcatttttccaacaattgtttacagacagattatttcacttataattcactgtatcacaattccagtgggtcaga agtttacatacactaaGATATCTCAACGGGACTTTCCAAAATcccgccGGTTACATAACTTACGGTAAATGGCCCGCCTGGC TGACCGCCCAACGACCCCCGCCCATTGACGTCAATAATGACGTATGTTCCCATAGTAACGCCAATAGGGACTTT CCATTGACGTCAATGGGTGGACTATTTACGGTAAACTGCCACGGTAAATGGCCCGCCTGGCCACTGGGCGTGG ATAGCGGTTTGACTCACGGGGATTTCCAAAATGTCGTAACAACTCCGCCCCATTGACGTCAATGGGCGGTTTGT TTTGGCACCAAAATGGCGGGgaaggtcaccaagcaggaagtcaaagactttttccggtgggcaaaggatcacgtggttgaggtggagcatgaattct acgtcaaaaagggtggagccaagaaaagacccgcccccagtgacgcaggtatataagtgagcccCTCCCTATCAGTGATAGAGATCTCCCTA TCAGTGATAGAGACCaaacgggtgcgcgagtcagtt gcgcagccat cgacgtcagacgcggaagct tcgatcaact acgcagacag gtaccaaaac aaatgttctc gtcacgtgggcatgaatctg atgctgtttc cctgcagaca atgcgagaga atgaatcaga attcaaatat ctgcttcact cacggacaga aagactgttt agagtgcttt cccgtgtcag aatctcaacccgtttctgtc gtcaaaaagg cgtatcagaa actgtgctac attcatcata tcatgggaaaggtgccagac gcttgcactg cctgcgatct ggtcaatgtg gatttggatg actgcatctttgaacCataa atga tttaaa tcaggt atggctgc cgatggttatcttccagatt ggctcgagga cactctctct gaaggaataa gacagtggtg gaagctcaaacctggcccac caccaccaaa gcccgcagag cggcataagg acgacagcag gggtcttgtgcttcctgggt acaagtacct cggacccttc aacggactcg acaagggaga gccggtcaacgaggcagacg ccgcggccct cgagcacgac aaagcctacg accggcagct cgacagcggagacaacccgt acctcaagta caaccacgcc gacgcggagt ttcaggagcg ccttaaagaagatacgtctt ttgggggcaa cctcggacga gcagtcttcc aggcgaaaaa gagggttcttgaacctctgg gcctggttga ggaacctgtt aagacggctc cgggaaaaaa gaggccggtagagcactctc ctgtggagcc agactcctcc tcgggaaccg gaaaggcggg ccagcagcctgcaagaaaaa gattgaattt tggtcagact ggagacgcag actcagtacc tgacccccagcctctcggac agccaccagc agccccctct ggtctgggaa ctaatacgat ggctacaggcagtggcgcac caatggcaga caataacgag ggcgccgacg gagtgggtaa ttcctcgggaaattggcatt gcgattccac atggatgggc gacagagtca tcaccaccag cacccgaacctgggccctgc ccacctacaa caaccacctc tacaaacaaa tttccagcca atcaggagcctcgaacgaca atcactactt tggctacagc accccttggg ggtattttga cttcaacagattccactgcc acttttcacc acgtgactgg caaagactca tcaacaacaa ctggggattc cgacccaaga gactcaactt caagctcttt aacattcaag tcaaagaggt cacgcagaatgacggtacga cgacgattgc caataacctt accagcacgg ttcaggtgtt tactgactcggagtaccagc tcccgtacgt cctcggctcg gcgcatcaag gatgcctccc gccgttcccagcagacgtct tcatggtgcc acagtatgga tacctcaccc tgaacaacgg gagtcaggca gtaggacgct cttcatttta ctgcctggag tactttcctt ctcagatgct gcgtaccgga aacaacttta ccttcagcta cacttttgag gacgttcctt tccacagcag ctacgctcacagccagagtc tggaccgtct catgaatcct ctcatcgacc agtacctgta ttacttgagc agaacaaaca ctccaagtgg aaccaccacg cagtcaaggc ttcagttttc tcaggccgga gcgagtgaca ttcgggacca gtctaggaac tggcttcctg gaccctgtta ccgccagcagcgagtatcaa agacatctgc ggataacaac aacagtgaat actcgtggac tggagctaccaagtaccacc tcaatggcag agactctctg gtgaatccgg gcccggccat ggcaagccacaaggacgatg aagaaaagtt ttttcctcag agcggggttc tcatctttgg gaagcaaggctcagagaaaa caaatgtgga cattgaaaag gtcatgatta cagacgaaga ggaaatcaggacaaccaatc ccgtggctac ggagcagtat ggttctgtat ctaccaacct ccagagaggcaacagacaag cagctaccgc agatgtcaac acacaaggcg ttcttccagg catggtctggcaggacagag atgtgtacct tcaggggccc atctgggcaa agattccaca cacggacggacattttcacc cctctcccct catgggtgga ttcggactta aacaccctcc tccacagattctcatcaaga acaccccggt acctgcgaat ccttcgacca ccttcagtgc ggcaaagtttgcttccttca tcacacagta ctccacggga caggtcagcg tggagatcga gtgggagctgcagaaggaaa acagcaaacg ctggaatccc gaaattcagt acacttccaa ctacaacaag tctgttaatg tggactttac tgtggacact aatggcgtgt attcagagcc tcgccccattggcaccagat acctgactcgtaatctgtaattgcttgttaatcGGATCCaatcaacctctggattacaaaatttgtgaaagattgactggtattcttaactatgttgctccttttacgctatgt ggatacgctgctttaatgcctttgtatcatgctattgcttcccgtatggctttcattttctcctccttgtataaatcctggttgctgtctctttatgaggagttgtggcccgttgtcagg caacgtggcgtggtgtgcactgtgtttgctgacgcaacccccactggttggggcattgccaccacctgtcagctcctttccgggactttcgctttccccctccctattgcca cggcggaactcatcgccgcctgccttgcccgctgctggacaggggctcggctgttgggcactgacaattccgtggtgttgtcggggaagctgacgtcctttccatggct gctcgcctgtgttgccacctggattctgcgcgggacgtccttctgctacgtcccttcggccctcaatccagcggaccttccttcccgcggcctgctgccggctctgcggcc tcttccgcgtcttcgccttcgccctcagacgagtcggatctccctttgggccgcctccccgcctggaattcgagctcggtacgatcagcctcgaaAATATTTTTAT TGTAACTAATGTTTTAGGTACAATAAAAGATCTTTCATTAGATCAGGTGTTGGTTTTTTGTGTGGGTGACCGGTaa CGGCCCCTCGCGCATTGGCCCGCGGGTCGCTCAATGAACCCGCATTGGTCCCCTGGGGTTCCGGGTATGGTA ATGAGTTTCTTCGGGAAGGCGGGAAGCCCCGGGGCACCGACGCAGGCCAAGCCCCTGTTGCGTCGGCGGGAG GGGCATGCTAATGGGGTTCTTTGGGGGACACCGGGTTGGGCCCCCAAATCGGGGGCCGGGCCGTGCATGCTA ATGATATTCTTTGGGGGCGCCGGGTTGGTCCCCGGGGACGGGGCCGCCCCGCGGTGGGCCTGCCTCCCCTG GGACGCGCGGCCATTGGGGGAATCGTCACTGCCGCCCCTTTGGGGAGGGGAAAGGCGTGGGGTATATAAGCA GAGCTCGTCGCATTTGCACCTCGGCACTCGGAGCGAGACGCAGCAGCCAGGCAGACTCGccaccatggccaagttgac cagtgccgttccggtgctcaccgcgcgcgacgtcgccggagcggtcgagttctggaccgaccggctcgggttctcccgggacttcgtggaggacgacttcgccggt gtggtccgggacgacgtgaccctgttcatcagcgcggtccaggaccaggtggtgccggacaacaccctggcctgggtgtgggtgcgcggcctggacgagctgtac gccgagtggtcggaggtcgtgtccacgaacttccgggacgcctccgggccggccatgaccgagatcggcgagcagccgtgggggcgggagttcgccctgcgcg acccggccggcaactgcgtgcacttcgtggccgaggagcaggactgataacacaagctttcttttgtgttggacctaataaaaaaaactcaggggatttttgctgtctgt tgggaaataaaggtttacttttgtatcttttgggtgtctgtgttggatgACGCGTttgagtgtatgtaaacttctgacccactgggaatgtgatgaaagaaataaaagctg aaatgaatcattctctctactattattctgatatttcacattcttaaaataaagtggtgatcctaactgacctaagacagggaatttttactaggattaaatgtcaggaattgtg aaaaagtgagtttaaatgtatttggctaaggtgtatgtaaacttccgacttcaactgtata GCTAGC– 3’ SEQ ID NO: 826: SFA2-EP40CAP2WZ2_SB: AAV2 CAP under the control of the tetO-containing hCMV enhancer element 3/AAV2 P40 promoter with a modified AAV2 P40 TATA element and MODIFIED AAV2 P40 intron_2 and WPRE/modified ICP27 poly A followed by ICP0 promoter-Zeocin resistant gene-ICP0 poly A cassette flanked with SB ITRs. 5’- GCGGCCGCTatacagttgaagtcggaagtttacatacacttaagttggagtcattaaaactcgtttttcaactactccacaaatttcttgttaacaaacaatagttttg gcaagtcagttaggacatctactttgtgcatgacacaagtcatttttccaacaattgtttacagacagattatttcacttataattcactgtatcacaattccagtgggtcaga agtttacatacactaaGATATCTCAACGGGACTTTCCAAAATcccgccGGTTACATAACTTACGGTAAATGGCCCGCCTGGC TGACCGCCCAACGACCCCCGCCCATTGACGTCAATAATGACGTATGTTCCCATAGTAACGCCAATAGGGACTTT CCATTGACGTCAATGGGTGGACTATTTACGGTAAACTGCCACGGTAAATGGCCCGCCTGGCCACTGGGCGTGG ATAGCGGTTTGACTCACGGGGATTTCCAAAATGTCGTAACAACTCCGCCCCATTGACGTCAATGGGCGGTTTGT TTTGGCACCAAAATGGCGGGgaaggtcaccaagcaggaagtcaaagactttttccggtgggcaaaggatcacgtggttgaggtggagcatgaattct acgtcaaaaagggtggagccaagaaaagacccgcccccagtgacgcaggtatataagtgagcccCTCCCTATCAGTGATAGAGATCTCCCTA TCAGTGATAGAGACCaaacgggtgcgcgagtcagtt gcgcagccat cgacgtcagacgcggaagct tcgatcaact acgcagacag gtaccaaaac aaCtgttctc gtcacgtgggcCtgaatctg aCgctgtttc cctgcagaca Ctgcgagaga Ttgaatcaga attcaaatCt ctgcttcact cacggacaga aagactgttt agagtgcttt cccgtgtcag aatctcaacccgtttctgtc gtcaaaaagg cgtatcagaa actgtgctac attcatcata tcTtgggaaaggtgccagac gcttgcactg cctgcgatct ggtcaCtgtg gatttggCtg actgcatctttgaacCataa aCga tttaaa tcaggt atggctgc cgatggttatcttccagatt ggctcgagga cactctctct gaaggaataa gacagtggtg gaagctcaaacctggcccac caccaccaaa gcccgcagag cggcataagg acgacagcag gggtcttgtgcttcctgggt acaagtacct cggacccttc aacggactcg acaagggaga gccggtcaacgaggcagacg ccgcggccct cgagcacgac aaagcctacg accggcagct cgacagcggagacaacccgt acctcaagta caaccacgcc gacgcggagt ttcaggagcg ccttaaagaagatacgtctt ttgggggcaa cctcggacga gcagtcttcc aggcgaaaaa gagggttcttgaacctctgg gcctggttga ggaacctgtt aagacggctc cgggaaaaaa gaggccggtagagcactctc ctgtggagcc agactcctcc tcgggaaccg gaaaggcggg ccagcagcctgcaagaaaaa gattgaattt tggtcagact ggagacgcag actcagtacc tgacccccagcctctcggac agccaccagc agccccctct ggtctgggaa ctaatacgat ggctacaggcagtggcgcac caatggcaga caataacgag ggcgccgacg gagtgggtaa ttcctcgggaaattggcatt gcgattccac atggatgggc gacagagtca tcaccaccag cacccgaacctgggccctgc ccacctacaa caaccacctc tacaaacaaa tttccagcca atcaggagcctcgaacgaca atcactactt tggctacagc accccttggg ggtattttga cttcaacagattccactgcc acttttcacc acgtgactgg caaagactca tcaacaacaa ctggggattc cgacccaaga gactcaactt caagctcttt aacattcaag tcaaagaggt cacgcagaatgacggtacga cgacgattgc caataacctt accagcacgg ttcaggtgtt tactgactcggagtaccagc tcccgtacgt cctcggctcg gcgcatcaag gatgcctccc gccgttcccagcagacgtct tcatggtgcc acagtatgga tacctcaccc tgaacaacgg gagtcaggca gtaggacgct cttcatttta ctgcctggag tactttcctt ctcagatgct gcgtaccgga aacaacttta ccttcagcta cacttttgag gacgttcctt tccacagcag ctacgctcacagccagagtc tggaccgtct catgaatcct ctcatcgacc agtacctgta ttacttgagc agaacaaaca ctccaagtgg aaccaccacg cagtcaaggc ttcagttttc tcaggccgga gcgagtgaca ttcgggacca gtctaggaac tggcttcctg gaccctgtta ccgccagcagcgagtatcaa agacatctgc ggataacaac aacagtgaat actcgtggac tggagctaccaagtaccacc tcaatggcag agactctctg gtgaatccgg gcccggccat ggcaagccacaaggacgatg aagaaaagtt ttttcctcag agcggggttc tcatctttgg gaagcaaggctcagagaaaa caaatgtgga cattgaaaag gtcatgatta cagacgaaga ggaaatcaggacaaccaatc ccgtggctac ggagcagtat ggttctgtat ctaccaacct ccagagaggcaacagacaag cagctaccgc agatgtcaac acacaaggcg ttcttccagg catggtctggcaggacagag atgtgtacct tcaggggccc atctgggcaa agattccaca cacggacggacattttcacc cctctcccct catgggtgga ttcggactta aacaccctcc tccacagattctcatcaaga acaccccggt acctgcgaat ccttcgacca ccttcagtgc ggcaaagtttgcttccttca tcacacagta ctccacggga caggtcagcg tggagatcga gtgggagctgcagaaggaaa acagcaaacg ctggaatccc gaaattcagt acacttccaa ctacaacaag tctgttaatg tggactttac tgtggacact aatggcgtgt attcagagcc tcgccccattggcaccagat acctgactcgtaatctgtaattgcttgttaatcGATCCAatcaacctctggattacaaaatttgtgaaagattgactggtattcttaactatgttgctccttttacgctatgtg gatacgctgctttaatgcctttgtatcatgctattgcttcccgtatggctttcattttctcctccttgtataaatcctggttgctgtctctttatgaggagttgtggcccgttgtcaggc aacgtggcgtggtgtgcactgtgtttgctgacgcaacccccactggttggggcattgccaccacctgtcagctcctttccgggactttcgctttccccctccctattgccac ggcggaactcatcgccgcctgccttgcccgctgctggacaggggctcggctgttgggcactgacaattccgtggtgttgtcggggaagctgacgtcctttccatggctg ctcgcctgtgttgccacctggattctgcgcgggacgtccttctgctacgtcccttcggccctcaatccagcggaccttccttcccgcggcctgctgccggctctgcggcctc ttccgcgtcttcgccttcgccctcagacgagtcggatctccctttgggccgcctccccgcctggaattcgagctcggtacgatcagcctcgaaAATATTTTTATT GTAACTAATGTTTTAGGTACAATAAAAGATCTTTCATTAGATCAGGTGTTGGTTTTTTGTGTGGGTGACCGGTaaC GGCCCCTCGCGCATTGGCCCGCGGGTCGCTCAATGAACCCGCATTGGTCCCCTGGGGTTCCGGGTATGGTAAT GAGTTTCTTCGGGAAGGCGGGAAGCCCCGGGGCACCGACGCAGGCCAAGCCCCTGTTGCGTCGGCGGGAGG GGCATGCTAATGGGGTTCTTTGGGGGACACCGGGTTGGGCCCCCAAATCGGGGGCCGGGCCGTGCATGCTAA TGATATTCTTTGGGGGCGCCGGGTTGGTCCCCGGGGACGGGGCCGCCCCGCGGTGGGCCTGCCTCCCCTGG GACGCGCGGCCATTGGGGGAATCGTCACTGCCGCCCCTTTGGGGAGGGGAAAGGCGTGGGGTATATAAGCAG AGCTCGTCGCATTTGCACCTCGGCACTCGGAGCGAGACGCAGCAGCCAGGCAGACTCGccaccatggccaagttgacca gtgccgttccggtgctcaccgcgcgcgacgtcgccggagcggtcgagttctggaccgaccggctcgggttctcccgggacttcgtggaggacgacttcgccggtgtg gtccgggacgacgtgaccctgttcatcagcgcggtccaggaccaggtggtgccggacaacaccctggcctgggtgtgggtgcgcggcctggacgagctgtacgcc gagtggtcggaggtcgtgtccacgaacttccgggacgcctccgggccggccatgaccgagatcggcgagcagccgtgggggcgggagttcgccctgcgcgacc cggccggcaactgcgtgcacttcgtggccgaggagcaggactgataacacaagctttcttttgtgttggacctaataaaaaaaactcaggggatttttgctgtctgttgg gaaataaaggtttacttttgtatcttttgggtgtctgtgttggatgACGCGTttgagtgtatgtaaacttctgacccactgggaatgtgatgaaagaaataaaagctgaa atgaatcattctctctactattattctgatatttcacattcttaaaataaagtggtgatcctaactgacctaagacagggaatttttactaggattaaatgtcaggaattgtgaa aaagtgagtttaaatgtatttggctaaggtgtatgtaaacttccgacttcaactgtata GCTAGC– 3’ SEQ ID NO: 841: Plasmid VB220803-2044esq_pSFP5TO1-AAVCDH: pSFP5TO1-AAVCDH is an engineered mobile element enzyme ITR containing plasmid that encodes 1) AAV2_REP 78/68 ORFs under the control of P5TO1 promoter, 2)Ad5 E2A_IRES_Ad5 E4ORF6 under the control of the tetO-containing SV40 promoter, 3) VA RNAs under the control of HSV-2 ICP4 promoter, and 4) hygro-B resistant gene under the control of TK promoter. 5’- cgtacgggcgcgccgcggccgccaactttgtatagaaaagttgttaacacttggattgcgggaaacgagttaagtcggctcgcgtgaattgcgcgtactccgcggga gccgtcttaactcggttcatatagatttgcggtggagtgcgggaaacgtgtaaactcgggccgattgtaactgcgtattaccaaatatttgttagatcctctagcgataagc ttgatatcgaattcgaggtcgactcaattgttaattaatgggcgtggatagcggtttgactcacggggatttccaagtctccaccccattgacgtcaatgggagtttgttttg gcaccaaaatcaacgggactttccaaaatgtcgtaacaactccgccccattgacgcaaatgggcggggtcctgtattagaggtcacgtgagtgttttgcgacattttgc gacaccatgtggtcacgctgggtatataagcaaccggtctccctatcagtgatagagatctccctatcagtgatagagatcggcccgagtgagcacgcagggtctcc gcggccgcgcgcagccaccatgccggggttttacgagattgtgattaaggtccccagcgaccttgacgagcatctgcccggcatttctgacagctttgtgaactgggt ggccgagaaggaatgggagttgccgccagattctgacatggatctgaatctgattgagcaggcacccctgaccgtggccgagaagctgcagcgcgactttctgacg gaatggcgccgtgtgagtaaggccccggaggcccttttctttgtgcaatttgagaagggagagagctacttccacatgcacgtgctcgtggaaaccaccggggtgaa atccatggttttgggacgtttcctgagtcagattcgcgaaaaactgattcagagaatttaccgcgggatcgagccgactttgccaaactggttcgcggtcacaaagacc agaaatggcgccggaggcgggaacaaggtggtggatgagtgctacatccccaattacttgctccccaaaacccagcctgagctccagtgggcgtggactaatatg gaacagtatttaagcgcctgtttgaatctcacggagcgtaaacggttggtggcgcagcatctgacgcacgtgtcgcagacgcaggagcagaacaaagagaatcag aatcccaattctgatgcgccggtgatcagatcaaaaacttcagccaggtacatggagctggtcgggtggctcgtggacaaggggattacctcggagaagcagtgga tccaggaggaccaggcctcatacatctccttcaatgcggcctccaactcgcggtcccaaatcaaggctgccttggacaatgcgggaaagattatgagcctgactaaa accgcccccgactacctggtgggccagcagcccgtggaggacatttccagcaatcggatttataaaattttggaactaaacgggtacgatccccaatatgcggcttcc gtctttctgggatgggccacgaaaaagttcggcaagaggaacaccatctggctgtttgggcctgcaactaccgggaagaccaacatcgcggaggccatagcccac actgtgcccttctacgggtgcgtaaactggaccaatgagaactttcccttcaacgactgtgtcgacaagatggtgatctggtgggaggaggggaagatgaccgccaa ggtcgtggagtcggccaaagccattctcggaggaagcaaggtgcgcgtggaccagaaatgcaagtcctcggcccagatagacccgactcccgtgatcgtcacctc caacaccaacatgtgcgccgtgattgacgggaactcaacgaccttcgaacaccagcagccgttgcaagaccggatgttcaaatttgaactcacccgccgtctggat catgactttgggaaggtcaccaagcaggaagtcaaagactttttccggtgggcaaaggatcacgtggttgaggtggagcatgaattctacgtcaaaaagggtggag ccaagaaaagacccgcccccagtgacgcagatataagtgagcccaaacgggtgcgcgagtcagttgcgcagccatcgacgtcagacgcggaagcttcgatca actacgcagacaggtaccaaaacaaatgttctcgtcacgtgggcatgaatctgatgctgtttccctgcagacaatgcgagagaatgaatcagaattcaaatatctgctt cactcacggacagaaagactgtttagagtgctttcccgtgtcagaatctcaacccgtttctgtcgtcaaaaaggcgtatcagaaactgtgctacattcatcatatcatggg aaaggtgccagacgcttgcactgcctgcgatctggtcaatgtggatttggatgactgcatctttgaacaataaatgatttaaatcaggtatggctgccgatggttatcttcc agattggctcgaggacactctctctgaaggaataagataatctgtaattgcttgttaatcaataaaccgtttaattcgtttcagttgaactttggtctctgcgtatttctttcttatc tagtttccatggctacgtacccgggcaagtttgtacaaaaaagcaggctctgtggaatgtgtgtcagttagggtgtggaaagtccccaggctccccagcaggcagaa gtatgcaaagcatgcatctcaattagtcagcaaccaggtgtggaaagtccccaggctccccagcaggcagaagtatgcaaagcatgcatctcaattagtcagcaac catagtcccgcccctaactccgcccatcccgcccctaactccgcccagttccgcccattctccgccccatggctgacaggcgccgggtatataagcagagctctccct atcagtgatagagatctccctatcagtgatagagatcgagctcgcgtgtgcgaggccgaggccgcctcgccaccatggccagccgcgaggaggagcagcgcga gaccacccccgagcgcggccgcggcgccgcccgccgcccccccaccatggaggacgtgagcagccccagccccagccccccccccccccgcgcccccccc aagaagcgcatgcgccgccgcatcgagagcgaggacgaggaggacagcagccaggacgccctggtgccccgcacccccagcccccgccccagcaccagc gccgccgacctggccatcgcccccaagaagaagaagaagcgccccagccccaagcccgagcgcccccccagccccgaggtgatcgtggacagcgaggag gagcgcgaggacgtggccctgcagatggtgggcttcagcaacccccccgtgctgatcaagcacggcaagggcggcaagcgcaccgtgcgccgcctgaacgag gacgaccccgtggcccgcggcatgcgcacccaggaggaggaggaggagcccagcgaggccgagagcgagatcaccgtgatgaaccccctgagcgtgccca tcgtgagcgcctgggagaagggcatggaggccgcccgcgccctgatggacaagtaccacgtggacaacgacctgaaggccaacttcaagctgctgcccgacca ggtggaggccctggccgccgtgtgcaagacctggctgaacgaggagcaccgcggcctgcagctgaccttcaccagcaacaagaccttcgtgaccatgatgggcc gcttcctgcaggcctacctgcagagcttcgccgaggtgacctacaagcaccacgagcccaccggctgcgccctgtggctgcaccgctgcgccgagatcgagggc gagctgaagtgcctgcacggcagcatcatgatcaacaaggagcacgtgatcgagatggacgtgaccagcgagaacggccagcgcgccctgaaggagcagag cagcaaggccaagatcgtgaagaaccgctggggccgcaacgtggtgcagatcagcaacaccgacgcccgctgctgcgtgcacgacgccgcctgccccgccaa ccagttcagcggcaagagctgcggcatgttcttcagcgagggcgccaaggcccaggtggccttcaagcagatcaaggccttcatgcaggccctgtaccccaacgc ccagaccggccacggccacctgctgatgcccctgcgctgcgagtgcaacagcaagcccggccacgcccccttcctgggccgccagctgcccaagctgaccccct tcgccctgagcaacgccgaggacctggacgccgacctgatcagcgacaagagcgtgctggccagcgtgcaccaccccgccctgatcgtgttccagtgctgcaac cccgtgtaccgcaacagccgcgcccagggcggcggccccaactgcgacttcaagatcagcgcccccgacctgctgaacgccctggtgatggtgcgcagcctgtg gagcgagaacttcaccgagctgccccgcatggtggtgcccgagttcaagtggagcaccaagcaccagtaccgcaacgtgagcctgcccgtggcccacagcgac gcccgccagaaccccttcgacttctgaagcaggtttccccaatgacacaaaacgtgcaacttgaaactccgcctggtctttccaggtctagaggggtaacactttgtac tgcgtttggctccacgctcgatccactggcgagtgttagtaacagcactgttgcttcgtagcggagcatgacggccgtgggaactcctccttggtaacaaggacccac ggggccaaaagccacgcccacacgggcccgtcatgtgtgcaaccccagcacggcgactttactgcgaaacccactttaaagtgacattgaaactggtacccaca cactggtgacaggctaaggatgcccttcaggtaccccgaggtaacacgcgacactcgggatctgagaaggggactggggcttctataaaagcgctcggtttaaaa agcttctatgcctgaataggtgaccggaggtcggcacctttcctttgcaattactgaccctatgaatacagccaccatgaccaccagcggcgtgcccttcggcatgacc ctgcgccccacccgcagccgcctgagccgccgcaccccctacagccgcgaccgcctgccccccttcgagaccgagacccgcgccaccatcctggaggaccac cccctgctgcccgagtgcaacaccctgaccatgcacaacgtgagctacgtgcgcggcctgccctgcagcgtgggcttcaccctgatccaggagtgggtggtgccct gggacatggtgctgacccgcgaggagctggtgatcctgcgcaagtgcatgcacgtgtgcctgtgctgcgccaacatcgacatcatgaccagcatgatgatccacgg ctacgagagctgggccctgcactgccactgcagcagccccggcagcctgcagtgcatcgccggcggccaggtgctggccagctggttccgcatggtggtggacg gcgccatgttcaaccagcgcttcatctggtaccgcgaggtggtgaactacaacatgcccaaggaggtgatgttcatgagcagcgtgttcatgcgcggccgccacctg atctacctgcgcctgtggtacgacggccacgtgggcagcgtggtgcccgccatgagcttcggctacagcgccctgcactgcggcatcctgaacaacatcgtggtgct gtgctgcagctactgcgccgacctgagcgagatccgcgtgcgctgctgcgcccgccgcacccgccgcctgatgctgcgcgccgtgcgcatcatcgccgaggagac caccgccatgctgtacagctgccgcaccgagcgccgccgccagcagttcatccgcgccctgctgcagcaccaccgccccatcctgatgcacgactacgacagca cccccatgtaacagaacttgtttattgcagcttataatggttacaaataaagcaatagcatcacaaatttcacaaataaagcatttttttcactgcattctagttgtggtttgtc caaactcatcaatgtatcttatcatgtctgaagcttggcacccagctttcttgtacaaagtgggggccggcgggggccaacgggagcgcggggccggcatctcattac cacgaacccggaagggcaggggagcgagcccgcccgcgacgagggtctcattagcatcgcgggcggaagcggaagccgcccgcgccgggcgctaatgaga tgccgcgcgggcggagcggcggcggcgcgaccaacgggccgccgccacggacgcggacgcgcgggcgtcggggcggggccgcgcataatgcggttccacc tgggggcggaaccccggcgagccggggcgcggcggcgtcgatcgctcctcctccgcgtcctcctcctttccccccgccccgcgcgccccgaggactatatgagcc aggcgacggggcgatcgggcactcttccgtggtctggtggataaattcgcaagggtatcatggcggacgaccggggttcgagccccgtatccggccgtccgccgtg atccatgcggttaccgcccgcgtgtcgaacccaggtgtgcgacgtcagacaacgggggagtgctccttttggcttccttccaggcgcggcggctgctgcgctagcttttt tggccactggccgcgcgcagcgtaagcggttaggctggaaagcgaaagcattaagtggctcgctccctgtagccggagggttattttccaagggttgagtcgcggg acccccggttcgagtctcggaccggccggactgcggcgaacgggggtttgcctccccgtcatgcaagaccccgcttgcaaattcctccggaaacagggacgagcc cctttttcaaatatttttattgcaactccctgttttaggtacaataaaaacaaaacatttcaaacaaatcgcccctcgtgttgtccttctttgctcatggccggcggacgcgtac gcgtcaaaccccgcccagcgtcttgtcattggcgaattcgaacacgcagatgcagtcggggcggcgcggtccgaggtccacttcgctatataaggtgacgcgtgtg gcctcgaacaccgagcgaccctgcagcgacccgcttaacagcggccaccatgaaaaagcctgaactcaccgcgacgtctgtcgagaagtttctgatcgaaaagtt cgacagcgtctccgacctgatgcagctctcggagggcgaagaatctcgtgctttcagcttcgatgtaggagggcgtggatatgtcctgcgggtaaatagctgcgccga tggtttctacaaagatcgttatgtttatcggcactttgcatcggccgcgctcccgattccggaagtgcttgacattggggaattcagcgagagcctgacctattgcatctcc cgccgtgcacagggtgtcacgttgcaagacctgcctgaaaccgaactgcccgctgttctgcagccggtcgcggaggccatggatgcgatcgctgcggccgatctta gccagacgagcgggttcggcccattcggaccgcaaggaatcggtcaatacactacatggcgtgatttcatatgcgcgattgctgatccccatgtgtatcactggcaaa ctgtgatggacgacaccgtcagtgcgtccgtcgcgcaggctctcgatgagctgatgctttgggccgaggactgccccgaagtccggcacctcgtgcacgcggatttc ggctccaacaatgtcctgacggacaatggccgcataacagcggtcattgactggagcgaggcgatgttcggggattcccaatacgaggtcgccaacatcttcttctg gaggccgtggttggcttgtatggagcagcagacgcgctacttcgagcggaggcatccggagcttgcaggatcgccgcggctccgggcgtatatgctccgcattggtc ttgaccaactctatcagagcttggttgacggcaatttcgatgatgcagcttgggcgcagggtcgatgcgacgcaatcgtccgatccggagccgggactgtcgggcgta cacaaatcgcccgcagaagcgcggccgtctggaccgatggctgtgtagaagtactcgccgatagtggaaaccgacgccccagcactcgtccggatcgggagatg ggggaggctaactgaaacaataaaagatctttattttcattagatctgtgtgttggttttttgtgtgcatatgaattatttatgtactgaatagataaaaaaatgtctgtgattga ataaattttcattttttacacaagaaaccgaaaatttcatttcaatcgaacccatacttcaaaagatataggcattttaaactaactctgattttgcgcgggaaacctaaata attgcccgcgccatcttatattttggcgggaaattcacccgacaccgtagtgttaacaactttattatacatagttgttaattaaggcgctcttccgcttcctcgctcactgact cgctgcgctcggtcgttcggctgcggcgagcggtatcagctcactcaaaggcggtaatacggttatccacagaatcaggggataacgcaggaaagaacatgtgag caaaaggccagcaaaaggccaggaaccgtaaaaaggccgcgttgctggcgtttttccataggctccgcccccctgacgagcatcacaaaaatcgacgctcaagt cagaggtggcgaaacccgacaggactataaagataccaggcgtttccccctggaagctccctcgtgcgctctcctgttccgaccctgccgcttaccggatacctgtcc gcctttctctcttcgggaagcgtggcgctttctcatagctcacgctgtaggtatctcagttcggtgtaggtcgttcgctccaagctgggctgtgtgcacgaaccccccgttca gcccgaccgctgcgccttatccggtaactatcgtcttgagtccaacccggtaagacacgacttatcgccactggcagcagccactggtaacaggattagcagagcg aggtatgtaggcggtgctacagagttcttgaagtggtggcctaactacggctacactagaagaacagtatttggtatctgcgctctgctgaagccagttaccttcggaaa aagagttggtagctcttgatccggcaaacaaaccaccgctggtagcggtggtttttttgtttgcaagcagcagattacgcgcagaaaaaaaggatctcaagaagatcc tttgatcttttctacggggtctgacgctcagtggaacgaaaactcacgttaagggattttggtcatgagattatcaaaaaggatcttcacctagatccttttaaattaaaaat gaagttttaaatcaatctaaagtatatatgagtaaacttggtctgacagttagaaaaactcatcgagcatcaaatgaaactgcaatttattcatatcaggattatcaatacc atatttttgaaaaagccgtttctgtaatgaaggagaaaactcaccgaggcagttccataggatggcaagatcctggtatcggtctgcgattccgactcgtccaacatca atacaacctattaatttcccctcgtcaaaaataaggttatcaagtgagaaatcaccatgagtgacgactgaatccggtgagaatggcaaaagtttatgcatttctttccag acttgttcaacaggccagccattacgctcgtcatcaaaatcactcgcatcaaccaaaccgttattcattcgtgattgcgcctgagcgagacgaaatacgcgatcgctgtt aaaaggacaattacaaacaggaatcgaatgcaaccggcgcaggaacactgccagcgcatcaacaatattttcacctgaatcaggatattcttctaatacctggaat gctgtttttccggggatcgcagtggtgagtaaccatgcatcatcaggagtacggataaaatgcttgatggtcggaagaggcataaattccgtcagccagtttagtctga ccatctcatctgtaacatcattggcaacgctacctttgccatgtttcagaaacaactctggcgcatcgggcttcccatacaagcgatagattgtcgcacctgattgcccga cattatcgcgagcccatttatacccatataaatcagcatccatgttggaatttaatcgcggcctcgacgtttcccgttgaatatggctcatactcttcctttttcaatattattga agcatttatcagggttattgtctcatgagcggatacatatttgaatgtatttagaaaaataaacaaataggggttccgcgcacatttccccgaaaagtgccacctgacgtc taagaaaccattattatcatgacattaacctataaaaataggcgtatcacgaggccctttcgtc – 3’ SEQ ID NO: 842: Plasmid with pSF-ITRP5TO1-AAV2H: pSF-ITRP5TO1-AAV2H is an engineered mobile element enzyme ITR containing plasmid that encodes 1) AAV2_REP78/68 ORFs under the control of P5TO1 promoter and CAPs under the native P40 promoter, 2) Ad5 E2A_IRES_Ad5 E4 ORF6 under the control of the tetO-containing SV40 promoter, 3) VA RNAs under the control of HSV-1 ICP4 promoter, and 4) hygro-B resistant gene under the control of TK promoter. 5’- cgtacgggcgcgccgcggccgccaactttgtatagaaaagttgttaacacttggattgcgggaaacgagttaagtcggctcgcgtgaattgcgcgtactccgcggga gccgtcttaactcggttcatatagatttgcggtggagtgcgggaaacgtgtaaactcgggccgattgtaactgcgtattaccaaatatttgttagatcctctagcgataagc ttgatatcgaattcgaggtcgactttaattaaccccattgacgtcaatgggcggcgtggatagcggtttgactcacggggatttccaagtctccaccccattgacgtcaat gggagtttgttttggcaccaaaatcaacgggactttccaaaatgtcgtaacaactccgccccattgacgcaaatgggcggggtcctgtattagaggtcacgtgagtgttt tgcgacattttgcgacaccatgtggtcacgctgggtatataagcaaccggtctccctatcagtgatagagatctccctatcagtgatagagatcggcccgagtgagcac gcagggtctccgcggccgcgcgcagccaccatgccggggttttacgagattgtgattaaggtccccagcgaccttgacgagcatctgcccggcatttctgacagcttt gtgaactgggtggccgagaaggaatgggagttgccgccagattctgacatggatctgaatctgattgagcaggcacccctgaccgtggccgagaagctgcagcgc gactttctgacggaatggcgccgtgtgagtaaggccccggaggcccttttctttgtgcaatttgagaagggagagagctacttccacatgcacgtgctcgtggaaacc accggggtgaaatccatggttttgggacgtttcctgagtcagattcgcgaaaaactgattcagagaatttaccgcgggatcgagccgactttgccaaactggttcgcgg tcacaaagaccagaaatggcgccggaggcgggaacaaggtggtggatgagtgctacatccccaattacttgctccccaaaacccagcctgagctccagtgggcg tggactaatatggaacagtatttaagcgcctgtttgaatctcacggagcgtaaacggttggtggcgcagcatctgacgcacgtgtcgcagacgcaggagcagaaca aagagaatcagaatcccaattctgatgcgccggtgatcagatcaaaaacttcagccaggtacatggagctggtcgggtggctcgtggacaaggggattacctcgga gaagcagtggatccaggaggaccaggcctcatacatctccttcaatgcggcctccaactcgcggtcccaaatcaaggctgccttggacaatgcgggaaagattatg agcctgactaaaaccgcccccgactacctggtgggccagcagcccgtggaggacatttccagcaatcggatttataaaattttggaactaaacgggtacgatcccc aatatgcggcttccgtctttctgggatgggccacgaaaaagttcggcaagaggaacaccatctggctgtttgggcctgcaactaccgggaagaccaacatcgcgga ggccatagcccacactgtgcccttctacgggtgcgtaaactggaccaatgagaactttcccttcaacgactgtgtcgacaagatggtgatctggtgggaggagggga agatgaccgccaaggtcgtggagtcggccaaagccattctcggaggaagcaaggtgcgcgtggaccagaaatgcaagtcctcggcccagatagacccgactcc cgtgatcgtcacctccaacaccaacatgtgcgccgtgattgacgggaactcaacgaccttcgaacaccagcagccgttgcaagaccggatgttcaaatttgaactca cccgccgtctggatcatgactttgggaaggtcaccaagcaggaagtcaaagactttttccggtgggcaaaggatcacgtggttgaggtggagcatgaattctacgtca aaaagggtggagccaagaaaagacccgcccccagtgacgcagatataagtgagcccaaacgggtgcgcgagtcagttgcgcagccatcgacgtcagacgcg gaagcttcgatcaactacgcagacaggtaccaaaacaaatgttctcgtcacgtgggcatgaatctgatgctgtttccctgcagacaatgcgagagaatgaatcagaa ttcaaatatctgcttcactcacggacagaaagactgtttagagtgctttcccgtgtcagaatctcaacccgtttctgtcgtcaaaaaggcgtatcagaaactgtgctacatt catcatatcatgggaaaggtgccagacgcttgcactgcctgcgatctggtcaatgtggatttggatgactgcatctttgaacaataaatgatttaaatcaggtatggctgc cgatggttatcttccagattggctcgaggacactctctctgaaggaataagacagtggtggaagctcaaacctggcccaccaccaccaaagcccgcagagcggca taaggacgacagcaggggtcttgtgcttcctgggtacaagtacctcggacccttcaacggactcgacaagggagagccggtcaacgaggcagacgccgcggcc ctcgagcacgacaaagcctacgaccggcagctcgacagcggagacaacccgtacctcaagtacaaccacgccgacgcggagtttcaggagcgccttaaagaa gatacgtcttttgggggcaacctcggacgagcagtcttccaggcgaaaaagagggttcttgaacctctgggcctggttgaggaacctgttaagacggctccgggaaa aaagaggccggtagagcactctcctgtggagccagactcctcctcgggaaccggaaaggcgggccagcagcctgcaagaaaaagattgaattttggtcagactg gagacgcagactcagtacctgacccccagcctctcggacagccaccagcagccccctctggtctgggaactaatacgatggctacaggcagtggcgcaccaatg gcagacaataacgagggcgccgacggagtgggtaattcctcgggaaattggcattgcgattccacatggatgggcgacagagtcatcaccaccagcacccgaac ctgggccctgcccacctacaacaaccacctctacaaacaaatttccagccaatcaggagcctcgaacgacaatcactactttggctacagcaccccttgggggtattt tgacttcaacagattccactgccacttttcaccacgtgactggcaaagactcatcaacaacaactggggattccgacccaagagactcaacttcaagctctttaacatt caagtcaaagaggtcacgcagaatgacggtacgacgacgattgccaataaccttaccagcacggttcaggtgtttactgactcggagtaccagctcccgtacgtcct cggctcggcgcatcaaggatgcctcccgccgttcccagcagacgtcttcatggtgccacagtatggatacctcaccctgaacaacgggagtcaggcagtaggacg ctcttcattttactgcctggagtactttccttctcagatgctgcgtaccggaaacaactttaccttcagctacacttttgaggacgttcctttccacagcagctacgctcacag ccagagtctggaccgtctcatgaatcctctcatcgaccagtacctgtattacttgagcagaacaaacactccaagtggaaccaccacgcagtcaaggcttcagttttct caggccggagcgagtgacattcgggaccagtctaggaactggcttcctggaccctgttaccgccagcagcgagtatcaaagacatctgcggataacaacaacagt gaatactcgtggactggagctaccaagtaccacctcaatggcagagactctctggtgaatccgggcccggccatggcaagccacaaggacgatgaagaaaagttt tttcctcagagcggggttctcatctttgggaagcaaggctcagagaaaacaaatgtggacattgaaaaggtcatgattacagacgaagaggaaatcaggacaacc aatcccgtggctacggagcagtatggttctgtatctaccaacctccagagaggcaacagacaagcagctaccgcagatgtcaacacacaaggcgttcttccaggc atggtctggcaggacagagatgtgtaccttcaggggcccatctgggcaaagattccacacacggacggacattttcacccctctcccctcatgggtggattcggactt aaacaccctcctccacagattctcatcaagaacaccccggtacctgcgaatccttcgaccaccttcagtgcggcaaagtttgcttccttcatcacacagtactccacgg gacaggtcagcgtggagatcgagtgggagctgcagaaggaaaacagcaaacgctggaatcccgaaattcagtacacttccaactacaacaagtctgttaatgtgg actttactgtggacactaatggcgtgtattcagagcctcgccccattggcaccagatacctgactcgtaatctgtaattgcttgttaatcaataaaccgtttaattcgtttcagt tgaactttggtctctgcgtatttctttcttatctagtttccatggctacgtacaagtttgtacaaaaaagcaggctctgtggaatgtgtgtcagttagggtgtggaaagtcccca ggctccccagcaggcagaagtatgcaaagcatgcatctcaattagtcagcaaccaggtgtggaaagtccccaggctccccagcaggcagaagtatgcaaagcat gcatctcaattagtcagcaaccatagtcccgcccctaactccgcccatcccgcccctaactccgcccagttccgcccattctccgccccatggctgacaggcgccgg gtatataagcagagctctccctatcagtgatagagatctccctatcagtgatagagatcgagctcgcgtgtgcgaggccgaggccgcctcgccaccatggccagccg cgaggaggagcagcgcgagaccacccccgagcgcggccgcggcgccgcccgccgcccccccaccatggaggacgtgagcagccccagccccagcccccc ccccccccgcgccccccccaagaagcgcatgcgccgccgcatcgagagcgaggacgaggaggacagcagccaggacgccctggtgccccgcacccccagc ccccgccccagcaccagcgccgccgacctggccatcgcccccaagaagaagaagaagcgccccagccccaagcccgagcgcccccccagccccgaggtg atcgtggacagcgaggaggagcgcgaggacgtggccctgcagatggtgggcttcagcaacccccccgtgctgatcaagcacggcaagggcggcaagcgcac cgtgcgccgcctgaacgaggacgaccccgtggcccgcggcatgcgcacccaggaggaggaggaggagcccagcgaggccgagagcgagatcaccgtgatg aaccccctgagcgtgcccatcgtgagcgcctgggagaagggcatggaggccgcccgcgccctgatggacaagtaccacgtggacaacgacctgaaggccaac ttcaagctgctgcccgaccaggtggaggccctggccgccgtgtgcaagacctggctgaacgaggagcaccgcggcctgcagctgaccttcaccagcaacaaga ccttcgtgaccatgatgggccgcttcctgcaggcctacctgcagagcttcgccgaggtgacctacaagcaccacgagcccaccggctgcgccctgtggctgcaccg ctgcgccgagatcgagggcgagctgaagtgcctgcacggcagcatcatgatcaacaaggagcacgtgatcgagatggacgtgaccagcgagaacggccagcg cgccctgaaggagcagagcagcaaggccaagatcgtgaagaaccgctggggccgcaacgtggtgcagatcagcaacaccgacgcccgctgctgcgtgcacg acgccgcctgccccgccaaccagttcagcggcaagagctgcggcatgttcttcagcgagggcgccaaggcccaggtggccttcaagcagatcaaggccttcatgc aggccctgtaccccaacgcccagaccggccacggccacctgctgatgcccctgcgctgcgagtgcaacagcaagcccggccacgcccccttcctgggccgcca gctgcccaagctgacccccttcgccctgagcaacgccgaggacctggacgccgacctgatcagcgacaagagcgtgctggccagcgtgcaccaccccgccctg atcgtgttccagtgctgcaaccccgtgtaccgcaacagccgcgcccagggcggcggccccaactgcgacttcaagatcagcgcccccgacctgctgaacgccctg gtgatggtgcgcagcctgtggagcgagaacttcaccgagctgccccgcatggtggtgcccgagttcaagtggagcaccaagcaccagtaccgcaacgtgagcct gcccgtggcccacagcgacgcccgccagaaccccttcgacttctgaagcaggtttccccaatgacacaaaacgtgcaacttgaaactccgcctggtctttccaggtc tagaggggtaacactttgtactgcgtttggctccacgctcgatccactggcgagtgttagtaacagcactgttgcttcgtagcggagcatgacggccgtgggaactcctc cttggtaacaaggacccacggggccaaaagccacgcccacacgggcccgtcatgtgtgcaaccccagcacggcgactttactgcgaaacccactttaaagtgac attgaaactggtacccacacactggtgacaggctaaggatgcccttcaggtaccccgaggtaacacgcgacactcgggatctgagaaggggactggggcttctata aaagcgctcggtttaaaaagcttctatgcctgaataggtgaccggaggtcggcacctttcctttgcaattactgaccctatgaatacagccaccatgaccaccagcgg cgtgcccttcggcatgaccctgcgccccacccgcagccgcctgagccgccgcaccccctacagccgcgaccgcctgccccccttcgagaccgagacccgcgcc accatcctggaggaccaccccctgctgcccgagtgcaacaccctgaccatgcacaacgtgagctacgtgcgcggcctgccctgcagcgtgggcttcaccctgatcc aggagtgggtggtgccctgggacatggtgctgacccgcgaggagctggtgatcctgcgcaagtgcatgcacgtgtgcctgtgctgcgccaacatcgacatcatgac cagcatgatgatccacggctacgagagctgggccctgcactgccactgcagcagccccggcagcctgcagtgcatcgccggcggccaggtgctggccagctggtt ccgcatggtggtggacggcgccatgttcaaccagcgcttcatctggtaccgcgaggtggtgaactacaacatgcccaaggaggtgatgttcatgagcagcgtgttcat gcgcggccgccacctgatctacctgcgcctgtggtacgacggccacgtgggcagcgtggtgcccgccatgagcttcggctacagcgccctgcactgcggcatcctg aacaacatcgtggtgctgtgctgcagctactgcgccgacctgagcgagatccgcgtgcgctgctgcgcccgccgcacccgccgcctgatgctgcgcgccgtgcgc atcatcgccgaggagaccaccgccatgctgtacagctgccgcaccgagcgccgccgccagcagttcatccgcgccctgctgcagcaccaccgccccatcctgat gcacgactacgacagcacccccatgtaacagaacttgtttattgcagcttataatggttacaaataaagcaatagcatcacaaatttcacaaataaagcatttttttcact gcattctagttgtggtttgtccaaactcatcaatgtatcttatcatgtctgaagcttggcacccagctttcttgtacaaagtgggggccggcgggggccaacgggagcgc ggggccggcatctcattaccacgaacccggaagggcaggggagcgagcccgcccgcgacgagggtctcattagcatcgcgggcggaagcggaagccgcccg cgccgggcgctaatgagatgccgcgcgggcggagcggcggcggcgcgaccaacgggccgccgccacggacgcggacgcgcgggcgtcggggcggggcc gcgcataatgcggttccacctgggggcggaaccccggcgagccggggcgcggcggcgtcgatcgctcctcctccgcgtcctcctcctttccccccgccccgcgcgc cccgaggactatatgagccaggcgacggggcgatcgggcactcttccgtggtctggtggataaattcgcaagggtatcatggcggacgaccggggttcgagcccc gtatccggccgtccgccgtgatccatgcggttaccgcccgcgtgtcgaacccaggtgtgcgacgtcagacaacgggggagtgctccttttggcttccttccaggcgcg gcggctgctgcgctagcttttttggccactggccgcgcgcagcgtaagcggttaggctggaaagcgaaagcattaagtggctcgctccctgtagccggagggttatttt ccaagggttgagtcgcgggacccccggttcgagtctcggaccggccggactgcggcgaacgggggtttgcctccccgtcatgcaagaccccgcttgcaaattcctc cggaaacagggacgagcccctttttcaaatatttttattgcaactccctgttttaggtacaataaaaacaaaacatttcaaacaaatcgcccctcgtgttgtccttctttgctc atggccggcggacgcgtcaaaccccgcccagcgtcttgtcattggcgaattcgaacacgcagatgcagtcggggcggcgcggtccgaggtccacttcgctatataa ggtgacgcgtgtggcctcgaacaccgagcgaccctgcagcgacccgcttaacagcggccaccatgaaaaagcctgaactcaccgcgacgtctgtcgagaagttt ctgatcgaaaagttcgacagcgtctccgacctgatgcagctctcggagggcgaagaatctcgtgctttcagcttcgatgtaggagggcgtggatatgtcctgcgggta aatagctgcgccgatggtttctacaaagatcgttatgtttatcggcactttgcatcggccgcgctcccgattccggaagtgcttgacattggggaattcagcgagagcctg acctattgcatctcccgccgtgcacagggtgtcacgttgcaagacctgcctgaaaccgaactgcccgctgttctgcagccggtcgcggaggccatggatgcgatcgc tgcggccgatcttagccagacgagcgggttcggcccattcggaccgcaaggaatcggtcaatacactacatggcgtgatttcatatgcgcgattgctgatccccatgt gtatcactggcaaactgtgatggacgacaccgtcagtgcgtccgtcgcgcaggctctcgatgagctgatgctttgggccgaggactgccccgaagtccggcacctcg tgcacgcggatttcggctccaacaatgtcctgacggacaatggccgcataacagcggtcattgactggagcgaggcgatgttcggggattcccaatacgaggtcgc caacatcttcttctggaggccgtggttggcttgtatggagcagcagacgcgctacttcgagcggaggcatccggagcttgcaggatcgccgcggctccgggcgtatat gctccgcattggtcttgaccaactctatcagagcttggttgacggcaatttcgatgatgcagcttgggcgcagggtcgatgcgacgcaatcgtccgatccggagccgg gactgtcgggcgtacacaaatcgcccgcagaagcgcggccgtctggaccgatggctgtgtagaagtactcgccgatagtggaaaccgacgccccagcactcgtc cggatcgggagatgggggaggctaactgaaacaataaaagatctttattttcattagatctgtgtgttggttttttgtgtgcatatgaattatttatgtactgaatagataaaa aaatgtctgtgattgaataaattttcattttttacacaagaaaccgaaaatttcatttcaatcgaacccatacttcaaaagatataggcattttaaactaactctgattttgcgc gggaaacctaaataattgcccgcgccatcttatattttggcgggaaattcacccgacaccgtagtgttaacaactttattatacatagttgttaattaaggcgctcttccgct tcctcgctcactgactcgctgcgctcggtcgttcggctgcggcgagcggtatcagctcactcaaaggcggtaatacggttatccacagaatcaggggataacgcagg aaagaacatgtgagcaaaaggccagcaaaaggccaggaaccgtaaaaaggccgcgttgctggcgtttttccataggctccgcccccctgacgagcatcacaaa aatcgacgctcaagtcagaggtggcgaaacccgacaggactataaagataccaggcgtttccccctggaagctccctcgtgcgctctcctgttccgaccctgccgctt accggatacctgtccgcctttctctcttcgggaagcgtggcgctttctcatagctcacgctgtaggtatctcagttcggtgtaggtcgttcgctccaagctgggctgtgtgca cgaaccccccgttcagcccgaccgctgcgccttatccggtaactatcgtcttgagtccaacccggtaagacacgacttatcgccactggcagcagccactggtaaca ggattagcagagcgaggtatgtaggcggtgctacagagttcttgaagtggtggcctaactacggctacactagaagaacagtatttggtatctgcgctctgctgaagc cagttaccttcggaaaaagagttggtagctcttgatccggcaaacaaaccaccgctggtagcggtggtttttttgtttgcaagcagcagattacgcgcagaaaaaaag gatctcaagaagatcctttgatcttttctacggggtctgacgctcagtggaacgaaaactcacgttaagggattttggtcatgagattatcaaaaaggatcttcacctaga tccttttaaattaaaaatgaagttttaaatcaatctaaagtatatatgagtaaacttggtctgacagttagaaaaactcatcgagcatcaaatgaaactgcaatttattcata tcaggattatcaataccatatttttgaaaaagccgtttctgtaatgaaggagaaaactcaccgaggcagttccataggatggcaagatcctggtatcggtctgcgattcc gactcgtccaacatcaatacaacctattaatttcccctcgtcaaaaataaggttatcaagtgagaaatcaccatgagtgacgactgaatccggtgagaatggcaaaa gtttatgcatttctttccagacttgttcaacaggccagccattacgctcgtcatcaaaatcactcgcatcaaccaaaccgttattcattcgtgattgcgcctgagcgagacg aaatacgcgatcgctgttaaaaggacaattacaaacaggaatcgaatgcaaccggcgcaggaacactgccagcgcatcaacaatattttcacctgaatcaggata ttcttctaatacctggaatgctgtttttccggggatcgcagtggtgagtaaccatgcatcatcaggagtacggataaaatgcttgatggtcggaagaggcataaattccgt cagccagtttagtctgaccatctcatctgtaacatcattggcaacgctacctttgccatgtttcagaaacaactctggcgcatcgggcttcccatacaagcgatagattgtc gcacctgattgcccgacattatcgcgagcccatttatacccatataaatcagcatccatgttggaatttaatcgcggcctcgacgtttcccgttgaatatggctcatactctt cctttttcaatattattgaagcatttatcagggttattgtctcatgagcggatacatatttgaatgtatttagaaaaataaacaaataggggttccgcgcacatttccccgaaa agtgccacctgacgtctaagaaaccattattatcatgacattaacctataaaaataggcgtatcacgaggccctttcgtc – 3’ SEQ ID NO: 843: Plasmid with pSF-ITRP5TO1-AAV2HB: pSFP5TO1-AAV2HB is an engineered mobile element enzyme ITR containing plasmid that encodes 1) AAV2_REP78/68 ORFs under the control of P5TO1 promoter, 2) AAV2 CAPs under the tetO-containing P40 promoter with AAV9 P40 intron, 3) Ad5 E2A_IRES_Ad5 E4 ORF6 under the control of the tetO-containing SV40 promoter, 4) VA RNAs under the control of HSV-1 ICP4 promoter, and 5) hygro- B resistant gene under the control of TK promoter. 5’- cgtacgggcgcgccgcggccgccaactttgtatagaaaagttgttaacacttggattgcgggaaacgagttaagtcggctcgcgtgaattgcgcgtactccgcggga gccgtcttaactcggttcatatagatttgcggtggagtgcgggaaacgtgtaaactcgggccgattgtaactgcgtattaccaaatatttgttagatcctctagcgataagc ttgatatcgaattcgaggtcgactcaattgttaattaatgggcgtggatagcggtttgactcacggggatttccaagtctccaccccattgacgtcaatgggagtttgttttg gcaccaaaatcaacgggactttccaaaatgtcgtaacaactccgccccattgacgcaaatgggcggggtcctgtattagaggtcacgtgagtgttttgcgacattttgc gacaccatgtggtcacgctgggtatataagcaaccggtctccctatcagtgatagagatctccctatcagtgatagagatcggcccgagtgagcacgcagggtctcc gcggccgcgcgcagccaccatgccggggttttacgagattgtgattaaggtccccagcgaccttgacgagcatctgcccggcatttctgacagctttgtgaactgggt ggccgagaaggaatgggagttgccgccagattctgacatggatctgaatctgattgagcaggcacccctgaccgtggccgagaagctgcagcgcgactttctgacg gaatggcgccgtgtgagtaaggccccggaggcccttttctttgtgcaatttgagaagggagagagctacttccacatgcacgtgctcgtggaaaccaccggggtgaa atccatggttttgggacgtttcctgagtcagattcgcgaaaaactgattcagagaatttaccgcgggatcgagccgactttgccaaactggttcgcggtcacaaagacc agaaatggcgccggaggcgggaacaaggtggtggatgagtgctacatccccaattacttgctccccaaaacccagcctgagctccagtgggcgtggactaatatg gaacagtatttaagcgcctgtttgaatctcacggagcgtaaacggttggtggcgcagcatctgacgcacgtgtcgcagacgcaggagcagaacaaagagaatcag aatcccaattctgatgcgccggtgatcagatcaaaaacttcagccaggtacatggagctggtcgggtggctcgtggacaaggggattacctcggagaagcagtgga tccaggaggaccaggcctcatacatctccttcaatgcggcctccaactcgcggtcccaaatcaaggctgccttggacaatgcgggaaagattatgagcctgactaaa accgcccccgactacctggtgggccagcagcccgtggaggacatttccagcaatcggatttataaaattttggaactaaacgggtacgatccccaatatgcggcttcc gtctttctgggatgggccacgaaaaagttcggcaagaggaacaccatctggctgtttgggcctgcaactaccgggaagaccaacatcgcggaggccatagcccac actgtgcccttctacgggtgcgtaaactggaccaatgagaactttcccttcaacgactgtgtcgacaagatggtgatctggtgggaggaggggaagatgaccgccaa ggtcgtggagtcggccaaagccattctcggaggaagcaaggtgcgcgtggaccagaaatgcaagtcctcggcccagatagacccgactcccgtgatcgtcacctc caacaccaacatgtgcgccgtgattgacgggaactcaacgaccttcgaacaccagcagccgttgcaagaccggatgttcaaatttgaactcacccgccgtctggat catgactttgggaaggtcaccaagcaggaagtcaaagactttttccggtgggcaaaggatcacgtggttgaggtggagcatgaattctacgtcaaaaagggtggag ccaagaaaagacccgcccccagtgacgcagatataagtgagcccaaacgggtgcgcgagtcagttgcgcagccatcgacgtcagacgcggaagcttcgatca actacgcagacaggtaccaaaacaaatgttctcgtcacgtgggcatgaatctgatgctgtttccctgcagacaatgcgagagaatgaatcagaattcaaatatctgctt cactcacggacagaaagactgtttagagtgctttcccgtgtcagaatctcaacccgtttctgtcgtcaaaaaggcgtatcagaaactgtgctacattcatcatatcatggg aaaggtgccagacgcttgcactgcctgcgatctggtcaatgtggatttggatgactgcatctttgaacaataaatgatttaaatcaggtatggctgccgatggttatcttcc agattggctcgaggacactctctctgaaggaataagataatctgtaattgcttgttaatcaataaaccgtttaattcgtttcagttgaactttggtctctgcgtatttctttcttatc tagtttccatggctacgtacccgggcaagtttgtacaaaaaagcaggctctgtggaatgtgtgtcagttagggtgtggaaagtccccaggctccccagcaggcagaa gtatgcaaagcatgcatctcaattagtcagcaaccaggtgtggaaagtccccaggctccccagcaggcagaagtatgcaaagcatgcatctcaattagtcagcaac catagtcccgcccctaactccgcccatcccgcccctaactccgcccagttccgcccattctccgccccatggctgacaggcgccgggtatataagcagagctctccct atcagtgatagagatctccctatcagtgatagagatcgagctcgcgtgtgcgaggccgaggccgcctcgccaccatggccagccgcgaggaggagcagcgcga gaccacccccgagcgcggccgcggcgccgcccgccgcccccccaccatggaggacgtgagcagccccagccccagccccccccccccccgcgcccccccc aagaagcgcatgcgccgccgcatcgagagcgaggacgaggaggacagcagccaggacgccctggtgccccgcacccccagcccccgccccagcaccagc gccgccgacctggccatcgcccccaagaagaagaagaagcgccccagccccaagcccgagcgcccccccagccccgaggtgatcgtggacagcgaggag gagcgcgaggacgtggccctgcagatggtgggcttcagcaacccccccgtgctgatcaagcacggcaagggcggcaagcgcaccgtgcgccgcctgaacgag gacgaccccgtggcccgcggcatgcgcacccaggaggaggaggaggagcccagcgaggccgagagcgagatcaccgtgatgaaccccctgagcgtgccca tcgtgagcgcctgggagaagggcatggaggccgcccgcgccctgatggacaagtaccacgtggacaacgacctgaaggccaacttcaagctgctgcccgacca ggtggaggccctggccgccgtgtgcaagacctggctgaacgaggagcaccgcggcctgcagctgaccttcaccagcaacaagaccttcgtgaccatgatgggcc gcttcctgcaggcctacctgcagagcttcgccgaggtgacctacaagcaccacgagcccaccggctgcgccctgtggctgcaccgctgcgccgagatcgagggc gagctgaagtgcctgcacggcagcatcatgatcaacaaggagcacgtgatcgagatggacgtgaccagcgagaacggccagcgcgccctgaaggagcagag cagcaaggccaagatcgtgaagaaccgctggggccgcaacgtggtgcagatcagcaacaccgacgcccgctgctgcgtgcacgacgccgcctgccccgccaa ccagttcagcggcaagagctgcggcatgttcttcagcgagggcgccaaggcccaggtggccttcaagcagatcaaggccttcatgcaggccctgtaccccaacgc ccagaccggccacggccacctgctgatgcccctgcgctgcgagtgcaacagcaagcccggccacgcccccttcctgggccgccagctgcccaagctgaccccct tcgccctgagcaacgccgaggacctggacgccgacctgatcagcgacaagagcgtgctggccagcgtgcaccaccccgccctgatcgtgttccagtgctgcaac cccgtgtaccgcaacagccgcgcccagggcggcggccccaactgcgacttcaagatcagcgcccccgacctgctgaacgccctggtgatggtgcgcagcctgtg gagcgagaacttcaccgagctgccccgcatggtggtgcccgagttcaagtggagcaccaagcaccagtaccgcaacgtgagcctgcccgtggcccacagcgac gcccgccagaaccccttcgacttctgaagcaggtttccccaatgacacaaaacgtgcaacttgaaactccgcctggtctttccaggtctagaggggtaacactttgtac tgcgtttggctccacgctcgatccactggcgagtgttagtaacagcactgttgcttcgtagcggagcatgacggccgtgggaactcctccttggtaacaaggacccac ggggccaaaagccacgcccacacgggcccgtcatgtgtgcaaccccagcacggcgactttactgcgaaacccactttaaagtgacattgaaactggtacccaca cactggtgacaggctaaggatgcccttcaggtaccccgaggtaacacgcgacactcgggatctgagaaggggactggggcttctataaaagcgctcggtttaaaa agcttctatgcctgaataggtgaccggaggtcggcacctttcctttgcaattactgaccctatgaatacagccaccatgaccaccagcggcgtgcccttcggcatgacc ctgcgccccacccgcagccgcctgagccgccgcaccccctacagccgcgaccgcctgccccccttcgagaccgagacccgcgccaccatcctggaggaccac cccctgctgcccgagtgcaacaccctgaccatgcacaacgtgagctacgtgcgcggcctgccctgcagcgtgggcttcaccctgatccaggagtgggtggtgccct gggacatggtgctgacccgcgaggagctggtgatcctgcgcaagtgcatgcacgtgtgcctgtgctgcgccaacatcgacatcatgaccagcatgatgatccacgg ctacgagagctgggccctgcactgccactgcagcagccccggcagcctgcagtgcatcgccggcggccaggtgctggccagctggttccgcatggtggtggacg gcgccatgttcaaccagcgcttcatctggtaccgcgaggtggtgaactacaacatgcccaaggaggtgatgttcatgagcagcgtgttcatgcgcggccgccacctg atctacctgcgcctgtggtacgacggccacgtgggcagcgtggtgcccgccatgagcttcggctacagcgccctgcactgcggcatcctgaacaacatcgtggtgct gtgctgcagctactgcgccgacctgagcgagatccgcgtgcgctgctgcgcccgccgcacccgccgcctgatgctgcgcgccgtgcgcatcatcgccgaggagac caccgccatgctgtacagctgccgcaccgagcgccgccgccagcagttcatccgcgccctgctgcagcaccaccgccccatcctgatgcacgactacgacagca cccccatgtaacagaacttgtttattgcagcttataatggttacaaataaagcaatagcatcacaaatttcacaaataaagcatttttttcactgcattctagttgtggtttgtc caaactcatcaatgtatcttatcatgtctgaagcttggcacccagctttcttgtacaaagtggataagaaagaaataccgcagagaccaaagttcaactgaaacgaatt aaccggtttattgattaacaggcaattacagattacgagtcaggtatctggtgccaatggggcgaggctctgaatacacgccattagtgtccacagtaaagtccacatt aacagacttgttgtagttggaagtgtactgaatttcgggattccagcgtttgctgttttccttctgcagctcccactcgatctccacgctgacctgtcccgtggagtactgtgtg atgaaggaagcaaactttgccgcactgaaggtggtcgaaggattcgcaggtaccggggtgttcttgatgagaatctgtggaggagggtgtttaagtccgaatccacc catgaggggagaggggtgaaaatgtccgtccgtgtgtggaatctttgcccagatgggcccctgaaggtacacatctctgtcctgccagaccatgcctggaagaacgc cttgtgtgttgacatctgcggtagctgcttgtctgttgcctctctggaggttggtagatacagaaccatactgctccgtagccacgggattggttgtcctgatttcctcttcgtct gtaatcatgaccttttcaatgtccacatttgttttctctgagccttgcttcccaaagatgagaaccccgctctgaggaaaaaacttttcttcatcgtccttgtggcttgccatgg ccgggcccggattcaccagagagtctctgccattgaggtggtacttggtagctccagtccacgagtattcactgttgttgttatccgcagatgtctttgatactcgctgctgg cggtaacagggtccaggaagccagttcctagactggtcccgaatgtcactcgctccggcctgagaaaactgaagccttgactgcgtggtggttccacttggagtgtttg ttctgctcaagtaatacaggtactggtcgatgagaggattcatgagacggtccagactctggctgtgagcgtagctgctgtggaaaggaacgtcctcaaaagtgtagc tgaaggtaaagttgtttccggtacgcagcatctgagaaggaaagtactccaggcagtaaaatgaagagcgtcctactgcctgactcccgttgttcagggtgaggtatc catactgtggcaccatgaagacgtctgctgggaacggcgggaggcatccttgatgcgccgagccgaggacgtacgggagctggtactccgagtcagtaaacacct gaaccgtgctggtaaggttattggcaatcgtcgtcgtaccgtcattctgcgtgacctctttgacttgaatgttaaagagcttgaagttgagtctcttgggtcggaatccccag ttgttgttgatgagtctttgccagtcacgtggtgaaaagtggcagtggaatctgttgaagtcaaaatacccccaaggggtgctgtagccaaagtagtgattgtcgttcgag gctcctgattggctggaaatttgtttgtagaggtggttgttgtaggtgggcagggcccaggttcgggtgctggtggtgatgactctgtcgcccatccatgtggaatcgcaat gccaatttcccgaggaattacccactccgtcggcgccctcgttattgtctgccattggtgcgccactgcctgtagccatcgtattagttcccagaccagagggggctgct ggtggctgtccgagaggctgggggtcaggtactgagtctgcgtctccagtctgaccaaaattcaatctttttcttgcaggctgctggcccgcctttccggttcccgaggag gagtctggctccacaggagagtgctctaccggcctcttttttcccggagccgtcttaacaggttcctcaaccaggcccagaggttcaagaaccctctttttcgcctggaa gactgctcgtccgaggttgcccccaaaagacgtatcttctttaaggcgctcctgaaactccgcgtcggcgtggttgtacttgaggtacgggttgtctccgctgtcgagctg ccggtcgtaggctttgtcgtgctcgagggccgcggcgtctgcctcgttgaccggctctcccttgtcgagtccgttgaagggtccgaggtacttgtacccaggaagcaca agacccctgctgtcgtccttatgccgctctgcgggctttggtggtggtgggccaggtttgagcttccaccactgtcttattccttcagagagagtgtcctcgagccaatctgg aagataaccatcggcagccatacctggtttaagtcatttattgctcagaaacacagtcatccaggtccacgttgaccagatcgcaggccgagcaagcaatctcggga gcccgccccagcagatgatgaatcgcacagagtttccgatacgtcctctttctgacgaccggttgagattctgacacgccggggaaacactctgagcagtctctgacc ccgtgtgtgaagcaaatgttgaaattctgattcattctctcgcacgttttgcagggaagcagcatctgaagcatgcccgcgtgacgagaacatttgttttggtacctgtctg cgtagttgatcgaagcttccgcgtctgacgtcgatggctgcgcaactgactcgcgcacccgtttGGTCTCTATCACTGATAGGGAGATCTCTATC ACTGATAGGGAGgggctcacttatatacctgcgtcactgggggcgggtcttttcttggctccaccctttttgacgtagaattcatgctccacctcaaccacgtgatc ctttgcccaccggaaaaagtctttgacttcctgcttggtgaccttcgggccggcgggggccaacgggagcgcggggccggcatctcattaccacgaacccggaagg gcaggggagcgagcccgcccgcgacgagggtctcattagcatcgcgggcggaagcggaagccgcccgcgccgggcgctaatgagatgccgcgcgggcgga gcggcggcggcgcgaccaacgggccgccgccacggacgcggacgcgcgggcgtcggggcggggccgcgcataatgcggttccacctgggggcggaacccc ggcgagccggggcgcggcggcgtcgatcgctcctcctccgcgtcctcctcctttccccccgccccgcgcgccccgaggactatatgagccaggcgacggggcgat cgggcactcttccgtggtctggtggataaattcgcaagggtatcatggcggacgaccggggttcgagccccgtatccggccgtccgccgtgatccatgcggttaccgc ccgcgtgtcgaacccaggtgtgcgacgtcagacaacgggggagtgctccttttggcttccttccaggcgcggcggctgctgcgctagcttttttggccactggccgcgc gcagcgtaagcggttaggctggaaagcgaaagcattaagtggctcgctccctgtagccggagggttattttccaagggttgagtcgcgggacccccggttcgagtct cggaccggccggactgcggcgaacgggggtttgcctccccgtcatgcaagaccccgcttgcaaattcctccggaaacagggacgagcccctttttcaaatatttttatt gcaactccctgttttaggtacaataaaaacaaaacatttcaaacaaatcgcccctcgtgttgtccttctttgctcatggccggcggacgcgtacgcgtcaaaccccgcc cagcgtcttgtcattggcgaattcgaacacgcagatgcagtcggggcggcgcggtccgaggtccacttcgctatataaggtgacgcgtgtggcctcgaacaccgag cgaccctgcagcgacccgcttaacagcggccaccatgaaaaagcctgaactcaccgcgacgtctgtcgagaagtttctgatcgaaaagttcgacagcgtctccga cctgatgcagctctcggagggcgaagaatctcgtgctttcagcttcgatgtaggagggcgtggatatgtcctgcgggtaaatagctgcgccgatggtttctacaaagat cgttatgtttatcggcactttgcatcggccgcgctcccgattccggaagtgcttgacattggggaattcagcgagagcctgacctattgcatctcccgccgtgcacagggt gtcacgttgcaagacctgcctgaaaccgaactgcccgctgttctgcagccggtcgcggaggccatggatgcgatcgctgcggccgatcttagccagacgagcgggt tcggcccattcggaccgcaaggaatcggtcaatacactacatggcgtgatttcatatgcgcgattgctgatccccatgtgtatcactggcaaactgtgatggacgacac cgtcagtgcgtccgtcgcgcaggctctcgatgagctgatgctttgggccgaggactgccccgaagtccggcacctcgtgcacgcggatttcggctccaacaatgtcct gacggacaatggccgcataacagcggtcattgactggagcgaggcgatgttcggggattcccaatacgaggtcgccaacatcttcttctggaggccgtggttggctt gtatggagcagcagacgcgctacttcgagcggaggcatccggagcttgcaggatcgccgcggctccgggcgtatatgctccgcattggtcttgaccaactctatcag agcttggttgacggcaatttcgatgatgcagcttgggcgcagggtcgatgcgacgcaatcgtccgatccggagccgggactgtcgggcgtacacaaatcgcccgca gaagcgcggccgtctggaccgatggctgtgtagaagtactcgccgatagtggaaaccgacgccccagcactcgtccggatcgggagatgggggaggctaactga aacaataaaagatctttattttcattagatctgtgtgttggttttttgtgtgcatatgaattatttatgtactgaatagataaaaaaatgtctgtgattgaataaattttcattttttaca caagaaaccgaaaatttcatttcaatcgaacccatacttcaaaagatataggcattttaaactaactctgattttgcgcgggaaacctaaataattgcccgcgccatctt atattttggcgggaaattcacccgacaccgtagtgttaacaactttattatacatagttgttaattaaggcgctcttccgcttcctcgctcactgactcgctgcgctcggtcgtt cggctgcggcgagcggtatcagctcactcaaaggcggtaatacggttatccacagaatcaggggataacgcaggaaagaacatgtgagcaaaaggccagcaa aaggccaggaaccgtaaaaaggccgcgttgctggcgtttttccataggctccgcccccctgacgagcatcacaaaaatcgacgctcaagtcagaggtggcgaaa cccgacaggactataaagataccaggcgtttccccctggaagctccctcgtgcgctctcctgttccgaccctgccgcttaccggatacctgtccgcctttctctcttcggg aagcgtggcgctttctcatagctcacgctgtaggtatctcagttcggtgtaggtcgttcgctccaagctgggctgtgtgcacgaaccccccgttcagcccgaccgctgcg ccttatccggtaactatcgtcttgagtccaacccggtaagacacgacttatcgccactggcagcagccactggtaacaggattagcagagcgaggtatgtaggcggt gctacagagttcttgaagtggtggcctaactacggctacactagaagaacagtatttggtatctgcgctctgctgaagccagttaccttcggaaaaagagttggtagctc ttgatccggcaaacaaaccaccgctggtagcggtggtttttttgtttgcaagcagcagattacgcgcagaaaaaaaggatctcaagaagatcctttgatcttttctacgg ggtctgacgctcagtggaacgaaaactcacgttaagggattttggtcatgagattatcaaaaaggatcttcacctagatccttttaaattaaaaatgaagttttaaatcaat ctaaagtatatatgagtaaacttggtctgacagttagaaaaactcatcgagcatcaaatgaaactgcaatttattcatatcaggattatcaataccatatttttgaaaaagc cgtttctgtaatgaaggagaaaactcaccgaggcagttccataggatggcaagatcctggtatcggtctgcgattccgactcgtccaacatcaatacaacctattaattt cccctcgtcaaaaataaggttatcaagtgagaaatcaccatgagtgacgactgaatccggtgagaatggcaaaagtttatgcatttctttccagacttgttcaacaggc cagccattacgctcgtcatcaaaatcactcgcatcaaccaaaccgttattcattcgtgattgcgcctgagcgagacgaaatacgcgatcgctgttaaaaggacaatta caaacaggaatcgaatgcaaccggcgcaggaacactgccagcgcatcaacaatattttcacctgaatcaggatattcttctaatacctggaatgctgtttttccgggga tcgcagtggtgagtaaccatgcatcatcaggagtacggataaaatgcttgatggtcggaagaggcataaattccgtcagccagtttagtctgaccatctcatctgtaac atcattggcaacgctacctttgccatgtttcagaaacaactctggcgcatcgggcttcccatacaagcgatagattgtcgcacctgattgcccgacattatcgcgagccc atttatacccatataaatcagcatccatgttggaatttaatcgcggcctcgacgtttcccgttgaatatggctcatactcttcctttttcaatattattgaagcatttatcagggtt attgtctcatgagcggatacatatttgaatgtatttagaaaaataaacaaataggggttccgcgcacatttccccgaaaagtgccacctgacgtctaagaaaccattatt atcatgacattaacctataaaaataggcgtatcacgaggccctttcgtc – 3’ SEQ ID NO: 830: Synthetic hCMV-derived Enhancer Element-3 (contains 2 copies of 21 bp repeat, 3 copies of 18 bp repeats, 3 copies of 19 bp repeats and 2 additional SP1 sites): TCAACGGGACTTTCCAAAATcccgccGGTTACATAACTTACGGTAAATGGCCCGCCTGGCTGACCGCCCAACGACC CCCGCCCATTGACGTCAATAATGACGTATGTTCCCATAGTAACGCCAATAGGGACTTTCCATTGACGTCAATGGG TGGACTATTTACGGTAAACTGCCACGGTAAATGGCCCGCCTGGCCACTGGGCGTGGATAGCGGTTTGACTCAC GGGGATTTCCAAAATGTCGTAACAACTCCGCCCCATTGACGTCAATGGGCGGTTTGTTTTGGCACCAAAATGGC GGG SEQ ID NO: 831: Sleeping Beauty LE ITR Sequence (IR/DR(L) Lmut44) (231 bp including TATA): 5’- tatacagttgaagtcggaagtttacatacacttaagttggagtcattaaaactcgtttttcaactactccacaaatttcttgttaacaaacaatagttttggcaagtcagttag gacatctactttgtgcatgacacaagtcatttttccaacaattgtttacagacagattatttcacttataattcactgtatcacaattccagtgggtcagaagtttacatacact aa– 3’ SEQ ID NO: 832: Sleeping Beauty RE ITR Sequence (IR/DR (R) Rmut13, Δ130, 143, 150) (232 bp including TATA): 5’- ttgagtgtatgtaaacttctgacccactgggaatgtgatgaaagaaataaaagctgaaatgaatcattctctctactattattctgatatttcacattcttaaaataaagtggt gatcctaactgacctaagacagggaatttttactaggattaaatgtcaggaattgtgaaaaagtgagtttaaatgtatttggctaaggtgtatgtaaacttccgacttcaac tgtata – 3’ SEQ ID NO: 833: P5TO1 promoter sequence: Ttaattaatgggcgtggatagcggtttgactcacggggatttccaagtctccaccccattgacgtcaatgggagtttgttttggcaccaaaatcaacgggactttccaaa atgtcgtaacaactccgccccattgacgcaaatgggcggggtcctgtattagaggtcacgtgagtgttttgcgacattttgcgacaccatgtggtcacgctgggtatata agcaaccggtctccctatcagtgatagagatctccctatcagtgatagagatcggcccgagtgagcacgcagggtctccgcggccgc SEQ ID NO: 834: SV40TO promoter sequence: Cccgggcaagtttgtacaaaaaagcaggctctgtggaatgtgtgtcagttagggtgtggaaagtccccaggctccccagcaggcagaagtatgcaaagcatgcat ctcaattagtcagcaaccaggtgtggaaagtccccaggctccccagcaggcagaagtatgcaaagcatgcatctcaattagtcagcaaccatagtcccgcccctaa ctccgcccatcccgcccctaactccgcccagttccgcccattctccgccccatggctgacaggcgccgggtatataagcagagctctccctatcagtgatagagatct ccctatcagtgatagagatcgagctcgcgtgtgcgaggccgaggccgcctc SEQ ID NO: 835: HSV-2 ICP4 promoter sequence followed by VA RNAs and ICP27 poly A ’ gggccggcgggggccaacgggagcgcggggccggcatctcattaccacgaacccggaagggcaggggagcgagcccgcccgcgacgagggtctcattagc atcgcgggcggaagcggaagccgcccgcgccgggcgctaatgagatgccgcgcgggcggagcggcggcggcgcgaccaacgggccgccgccacggacg cggacgcgcgggcgtcggggcggggccgcgcataatgcggttccacctgggggcggaaccccggcgagccggggcgcggcggcgtcgatcgctcctcctccg cgtcctcctcctttccccccgccccgcgcgccccgaggactatatgagccaggcgacggggcgatcgggcactcttccgtggtctggtggataaattcgcaagggtat catggcggacgaccggggttcgagccccgtatccggccgtccgccgtgatccatgcggttaccgcccgcgtgtcgaacccaggtgtgcgacgtcagacaacggg ggagtgctccttttggcttccttccaggcgcggcggctgctgcgctagcttttttggccactggccgcgcgcagcgtaagcggttaggctggaaagcgaaagcattaag tggctcgctccctgtagccggagggttattttccaagggttgagtcgcgggacccccggttcgagtctcggaccggccggactgcggcgaacgggggtttgcctcccc gtcatgcaagaccccgcttgcaaattcctccggaaacagggacgagcccctttttcaaatatttttattgcaactccctgttttaggtacaataaaaacaaaacatttcaa acaaatcgcccctcgtgttgtccttctttgctcatggccggcggacgcgtacgcgt – 3’ SEQ ID NO: 836: TK Promoter – hygro_resistant gene_synthetic poly A: 5’- caaaccccgcccagcgtcttgtcattggcgaattcgaacacgcagatgcagtcggggcggcgcggtccgaggtccacttcgctatataaggtgacgcgtgtggcctc gaacaccgagcgaccctgcagcgacccgcttaacagcggccaccatgaaaaagcctgaactcaccgcgacgtctgtcgagaagtttctgatcgaaaagttcgac agcgtctccgacctgatgcagctctcggagggcgaagaatctcgtgctttcagcttcgatgtaggagggcgtggatatgtcctgcgggtaaatagctgcgccgatggtt tctacaaagatcgttatgtttatcggcactttgcatcggccgcgctcccgattccggaagtgcttgacattggggaattcagcgagagcctgacctattgcatctcccgcc gtgcacagggtgtcacgttgcaagacctgcctgaaaccgaactgcccgctgttctgcagccggtcgcggaggccatggatgcgatcgctgcggccgatcttagcca gacgagcgggttcggcccattcggaccgcaaggaatcggtcaatacactacatggcgtgatttcatatgcgcgattgctgatccccatgtgtatcactggcaaactgtg atggacgacaccgtcagtgcgtccgtcgcgcaggctctcgatgagctgatgctttgggccgaggactgccccgaagtccggcacctcgtgcacgcggatttcggctc caacaatgtcctgacggacaatggccgcataacagcggtcattgactggagcgaggcgatgttcggggattcccaatacgaggtcgccaacatcttcttctggaggc cgtggttggcttgtatggagcagcagacgcgctacttcgagcggaggcatccggagcttgcaggatcgccgcggctccgggcgtatatgctccgcattggtcttgacc aactctatcagagcttggttgacggcaatttcgatgatgcagcttgggcgcagggtcgatgcgacgcaatcgtccgatccggagccgggactgtcgggcgtacacaa atcgcccgcagaagcgcggccgtctggaccgatggctgtgtagaagtactcgccgatagtggaaaccgacgccccagcactcgtccggatcgggagatggggg aggctaactgaaacaataaaagatctttattttcattagatctgtgtgttggttttttgtgtg – 3’ SEQ ID NO: 837: Zeocin Resistant Gene under the control of a modified HSV-1 ICP0 promoter with modified ICP0 poly A 5’- CGGCCCCTCGCGCATTGGCCCGCGGGTCGCTCAATGAACCCGCATTGGTCCCCTGGGGTTCCGGGTATGGTA ATGAGTTTCTTCGGGAAGGCGGGAAGCCCCGGGGCACCGACGCAGGCCAAGCCCCTGTTGCGTCGGCGGGAG GGGCATGCTAATGGGGTTCTTTGGGGGACACCGGGTTGGGCCCCCAAATCGGGGGCCGGGCCGTGCATGCTA ATGATATTCTTTGGGGGCGCCGGGTTGGTCCCCGGGGACGGGGCCGCCCCGCGGTGGGCCTGCCTCCCCTG GGACGCGCGGCCATTGGGGGAATCGTCACTGCCGCCCCTTTGGGGAGGGGAAAGGCGTGGGGTATATAAGCA GAGCTCGTCGCATTTGCACCTCGGCACTCGGAGCGAGACGCAGCAGCCAGGCAGACTC Gccaccatggccaagttgaccagtgccgttccggtgctcaccgcgcgcgacgtcgccggagcggtcgagttctggaccgaccggctcgggttctcccgggacttcg tggaggacgacttcgccggtgtggtccgggacgacgtgaccctgttcatcagcgcggtccaggaccaggtggtgccggacaacaccctggcctgggtgtgggtgc gcggcctggacgagctgtacgccgagtggtcggaggtcgtgtccacgaacttccgggacgcctccgggccggccatgaccgagatcggcgagcagccgtgggg gcgggagttcgccctgcgcgacccggccggcaactgcgtgcacttcgtggccgaggagcaggactga taacacaa gctttcttt tgtgttggac ctaataaaaaaaactcaggggatttttgctgtctgttgggaaataaaggt ttacttttgt atcttttggg tgtctgtgtt ggatg – 3’ SEQ ID NO: 838: WPRE REGULATAORY ELEMENTS PLUS BGH POLY A: Aatcaacctctggattacaaaatttgtgaaagattgactggtattcttaactatgttgctccttttacgctatgtggatacgctgctttaatgcctttgtatcatgctattgcttccc gtatggctttcattttctcctccttgtataaatcctggttgctgtctctttatgaggagttgtggcccgttgtcaggcaacgtggcgtggtgtgcactgtgtttgctgacgcaaccc ccactggttggggcattgccaccacctgtcagctcctttccgggactttcgctttccccctccctattgccacggcggaactcatcgccgcctgccttgcccgctgctgga caggggctcggctgttgggcactgacaattccgtggtgttgtcggggaagctgacgtcctttccatggctgctcgcctgtgttgccacctggattctgcgcgggacgtcct tctgctacgtcccttcggccctcaatccagcggaccttccttcccgcggcctgctgccggctctgcggcctcttccgcgtcttcgccttcgccctcagacgagtcggatctc cctttgggccgcctccccgcctggaattcgagctcggtacgatcagcctcgaCtgtgccttctagttgccagccatctgttgtttgcccctcccccgtgccttccttgaccct ggaaggtgccactcccactgtcctttcctaataaaatgaggaaattgcatcgcattgtctgagtaggtgtcattctattctggggggtggggtggggcaggacagcaag ggggaggattgggaagacaatagcaggcatgc SEQ ID NO: 839: AAV2 REPs coding sequence plus its native poly A tail: gcgcagccaccatgccggggttttacgagattgtgattaaggtccccagcgaccttgacgagcatctgcccggcatttctgacagctttgtgaactgggtggccgaga aggaatgggagttgccgccagattctgacatggatctgaatctgattgagcaggcacccctgaccgtggccgagaagctgcagcgcgactttctgacggaatggcg ccgtgtgagtaaggccccggaggcccttttctttgtgcaatttgagaagggagagagctacttccacatgcacgtgctcgtggaaaccaccggggtgaaatccatggt tttgggacgtttcctgagtcagattcgcgaaaaactgattcagagaatttaccgcgggatcgagccgactttgccaaactggttcgcggtcacaaagaccagaaatgg cgccggaggcgggaacaaggtggtggatgagtgctacatccccaattacttgctccccaaaacccagcctgagctccagtgggcgtggactaatatggaacagtat ttaagcgcctgtttgaatctcacggagcgtaaacggttggtggcgcagcatctgacgcacgtgtcgcagacgcaggagcagaacaaagagaatcagaatcccaat tctgatgcgccggtgatcagatcaaaaacttcagccaggtacatggagctggtcgggtggctcgtggacaaggggattacctcggagaagcagtggatccaggag gaccaggcctcatacatctccttcaatgcggcctccaactcgcggtcccaaatcaaggctgccttggacaatgcgggaaagattatgagcctgactaaaaccgccc ccgactacctggtgggccagcagcccgtggaggacatttccagcaatcggatttataaaattttggaactaaacgggtacgatccccaatatgcggcttccgtctttctg ggatgggccacgaaaaagttcggcaagaggaacaccatctggctgtttgggcctgcaactaccgggaagaccaacatcgcggaggccatagcccacactgtgc ccttctacgggtgcgtaaactggaccaatgagaactttcccttcaacgactgtgtcgacaagatggtgatctggtgggaggaggggaagatgaccgccaaggtcgtg gagtcggccaaagccattctcggaggaagcaaggtgcgcgtggaccagaaatgcaagtcctcggcccagatagacccgactcccgtgatcgtcacctccaacac caacatgtgcgccgtgattgacgggaactcaacgaccttcgaacaccagcagccgttgcaagaccggatgttcaaatttgaactcacccgccgtctggatcatgactt tgggaaggtcaccaagcaggaagtcaaagactttttccggtgggcaaaggatcacgtggttgaggtggagcatgaattctacgtcaaaaagggtggagccaagaa aagacccgcccccagtgacgcagatataagtgagcccaaacgggtgcgcgagtcagttgcgcagccatcgacgtcagacgcggaagcttcgatcaactacgca gacaggtaccaaaacaaatgttctcgtcacgtgggcatgaatctgatgctgtttccctgcagacaatgcgagagaatgaatcagaattcaaatatctgcttcactcacg gacagaaagactgtttagagtgctttcccgtgtcagaatctcaacccgtttctgtcgtcaaaaaggcgtatcagaaactgtgctacattcatcatatcatgggaaaggtg ccagacgcttgcactgcctgcgatctggtcaatgtggatttggatgactgcatctttgaacaataaatgatttaaatcaggtatggctgccgatggttatcttccagattggc tcgaggacactctctctgaaggaataagataatctgtaattgcttgttaatcaataaaccgtttaattcgtttcagttgaactttggtctctgcgtatttctttcttatctagtttccat ggctacgta SEQ ID NO: 840: Ad5 E2A coding sequence followed by IRES and Ad5 ORF 6 coding sequence and SV40 poly A 5’- gccaccatggccagccgcgaggaggagcagcgcgagaccacccccgagcgcggccgcggcgccgcccgccgcccccccaccatggaggacgtgagcagc cccagccccagccccccccccccccgcgccccccccaagaagcgcatgcgccgccgcatcgagagcgaggacgaggaggacagcagccaggacgccctg gtgccccgcacccccagcccccgccccagcaccagcgccgccgacctggccatcgcccccaagaagaagaagaagcgccccagccccaagcccgagcgcc cccccagccccgaggtgatcgtggacagcgaggaggagcgcgaggacgtggccctgcagatggtgggcttcagcaacccccccgtgctgatcaagcacggca agggcggcaagcgcaccgtgcgccgcctgaacgaggacgaccccgtggcccgcggcatgcgcacccaggaggaggaggaggagcccagcgaggccgag agcgagatcaccgtgatgaaccccctgagcgtgcccatcgtgagcgcctgggagaagggcatggaggccgcccgcgccctgatggacaagtaccacgtggaca acgacctgaaggccaacttcaagctgctgcccgaccaggtggaggccctggccgccgtgtgcaagacctggctgaacgaggagcaccgcggcctgcagctgac cttcaccagcaacaagaccttcgtgaccatgatgggccgcttcctgcaggcctacctgcagagcttcgccgaggtgacctacaagcaccacgagcccaccggctg cgccctgtggctgcaccgctgcgccgagatcgagggcgagctgaagtgcctgcacggcagcatcatgatcaacaaggagcacgtgatcgagatggacgtgacc agcgagaacggccagcgcgccctgaaggagcagagcagcaaggccaagatcgtgaagaaccgctggggccgcaacgtggtgcagatcagcaacaccgac gcccgctgctgcgtgcacgacgccgcctgccccgccaaccagttcagcggcaagagctgcggcatgttcttcagcgagggcgccaaggcccaggtggccttcaa gcagatcaaggccttcatgcaggccctgtaccccaacgcccagaccggccacggccacctgctgatgcccctgcgctgcgagtgcaacagcaagcccggccac gcccccttcctgggccgccagctgcccaagctgacccccttcgccctgagcaacgccgaggacctggacgccgacctgatcagcgacaagagcgtgctggccag cgtgcaccaccccgccctgatcgtgttccagtgctgcaaccccgtgtaccgcaacagccgcgcccagggcggcggccccaactgcgacttcaagatcagcgccc ccgacctgctgaacgccctggtgatggtgcgcagcctgtggagcgagaacttcaccgagctgccccgcatggtggtgcccgagttcaagtggagcaccaagcacc agtaccgcaacgtgagcctgcccgtggcccacagcgacgcccgccagaaccccttcgacttctgaagcaggtttccccaatgacacaaaacgtgcaacttgaaac tccgcctggtctttccaggtctagaggggtaacactttgtactgcgtttggctccacgctcgatccactggcgagtgttagtaacagcactgttgcttcgtagcggagcatg acggccgtgggaactcctccttggtaacaaggacccacggggccaaaagccacgcccacacgggcccgtcatgtgtgcaaccccagcacggcgactttactgcg aaacccactttaaagtgacattgaaactggtacccacacactggtgacaggctaaggatgcccttcaggtaccccgaggtaacacgcgacactcgggatctgaga aggggactggggcttctataaaagcgctcggtttaaaaagcttctatgcctgaataggtgaccggaggtcggcacctttcctttgcaattactgaccctatgaatacagc caccatgaccaccagcggcgtgcccttcggcatgaccctgcgccccacccgcagccgcctgagccgccgcaccccctacagccgcgaccgcctgccccccttcg agaccgagacccgcgccaccatcctggaggaccaccccctgctgcccgagtgcaacaccctgaccatgcacaacgtgagctacgtgcgcggcctgccctgcag cgtgggcttcaccctgatccaggagtgggtggtgccctgggacatggtgctgacccgcgaggagctggtgatcctgcgcaagtgcatgcacgtgtgcctgtgctgcg ccaacatcgacatcatgaccagcatgatgatccacggctacgagagctgggccctgcactgccactgcagcagccccggcagcctgcagtgcatcgccggcggc caggtgctggccagctggttccgcatggtggtggacggcgccatgttcaaccagcgcttcatctggtaccgcgaggtggtgaactacaacatgcccaaggaggtgat gttcatgagcagcgtgttcatgcgcggccgccacctgatctacctgcgcctgtggtacgacggccacgtgggcagcgtggtgcccgccatgagcttcggctacagcg ccctgcactgcggcatcctgaacaacatcgtggtgctgtgctgcagctactgcgccgacctgagcgagatccgcgtgcgctgctgcgcccgccgcacccgccgcct gatgctgcgcgccgtgcgcatcatcgccgaggagaccaccgccatgctgtacagctgccgcaccgagcgccgccgccagcagttcatccgcgccctgctgcagc accaccgccccatcctgatgcacgactacgacagcacccccatgtaacagaacttgtttattgcagcttataatggttacaaataaagcaatagcatcacaaatttcac aaataaagcatttttttcactgcattctagttgtggtttgtccaaactcatcaatgtatcttatcatgtctgaagcttggcacccagctttcttgtacaaagtgg – 3’ This invention is further illustrated by the following non-limiting examples. EXAMPLES Hereinafter, the present disclosure will be described in further detail with reference to examples. These examples are illustrative purposes only and are not to be construed to limit the scope of the present invention. In addition, various modifications and variations can be made without departing from the technical scope of the present invention. Example 1 – Design of Chimeric Mobile Element Enzymes with Transcription Activator-Like Effector (TALE) DNA Binding Domains (DBDs) or dCas9/gRNA that Target Human Genomic Safe Harbor Sites (GSHS) FIG.2E illustrates a non-limiting example of a system comprising a nucleic acid (e.g., helper RNA) encoding an enzyme capable of performing targeted genomic integration and a nucleic acid encoding a mobile element enzyme (donor DNA). The helper RNA is translated into a bioengineered enzyme (e.g., integrase, recombinase, or mobile element enzyme) that recognizes specific ends and seamlessly inserts the donor DNA into the human genome in a site-specific manner without a footprint. In this example, enzymes, e.g., chimeric mobile element enzymes, are designed using human GSHS TALE or Cas9/gRNA DBD. FIGs.2A-D depict representations of chimeric mobile element enzyme designed using human GSHS TALE or Cas9/gRNA DBD. FIG.2A. TALEs includes nuclear localization signals (NLS) and an activation domain (AD) to function as transcriptional activators. A central tandem repeat domain confers specific DNA-binding and host specificity. Translocation signal (TD) and four cryptic repeats required for initiation of DNA binding and for the recognition of 5’ -T0 are located at the N-terminus (checkered rectangles). Each 34 amino acid (aa) long repeat in the CRD binds to one nucleotide with specificity determined mainly by aa at position 13. One sample repeat is shown below the protein scheme. Numbers 12/13 refer to aa positions within the repeat. See Jankele et al., Brief Funct Genomics 2014;13:409-19. FIG.2B. Repeat types are shown that have specificity for one or several nucleotides. Only bases of the DNA leading strand are shown. FIG.2C. A chimeric mobile element enzyme having a TALE DNA-binding protein fused thereto by a linker that is greater than 23 amino acids in length (top). See Hew et al., Synth Biol (Oxf) 2019;4:ysz018. FIG.2D. Binding of the TALE to GSHS physically sequesters the mobile element enzyme to the same location and promotes transposition to the nearby TTAA sequences. All RVD are preceded by a thymine (T) to bind to the NTR shown in FIG.2A. All of these GSHS regions are in open chromatin and are susceptible to mobile element enzyme activity). FIG.2C also illustrates (bottom) a chimeric mobile element enzyme construct comprising dCas9 linked to one or more guide RNAs. An engineered chimeric mobile element enzyme may include: a guide RNA (gRNA) and an inactivated Cas protein. The gRNA is a short synthetic RNA composed of a scaffold sequence necessary for Cas-binding and a user-defined ~20 nucleotide spacer that defines the genomic target to be modified. Thus, the genomic target of the Cas protein is based upon the sequence present in the gRNA. FIG.3B shows gRNA sequences that physically sequester the mobile element enzyme to GSHS and promotes transposition to the nearby TTAA sequences. FIGs.3A-3E depict examples of construct templates (FIGs.3A-3D) and a system (FIG.3E) comprising a donor DNA and helper RNA. FIG.3A depicts a plasmid construct template that transcribes mobile element enzyme RNA that is later processed with a 5’- m7G cap. Other mobile element enzymes can be substituted. FIG.3B depicts a donor DNA construct template with the transgene VLDLR. Other tissue specific promoters and transgenes can be substituted. FIG. 3C depicts a chimeric mobile element enzyme construct template with a TALE DNA binder. Other TALEs and mobile element enzymes can be substituted. FIG.3D depicts a chimeric mobile element enzyme construct template with a dCas9/gRNA DNA binder. Other dCas9/gRNA and mobile element enzymes can be substituted. FIG.3E depicts a non-limiting example of the system of FIG.1A. In FIG.3E, the system comprises a nucleic acid (donor DNA), and a nucleic acid (helper RNA) encoding a mobile element enzyme (hyperactive ENGINEERED MOBILE ELEMENT ENZYME). As shown in FIG.3E, panel (A), the donor DNA can be any gene of interest (GOI) including those replace, inactive, or provide suicide or helper functions. The GOI can be driven by a predetermined promoter and flanked by insulators to prevent gene silencing. The ITRs are specific for the mammal-derived mobile element enzyme (ENGINEERED MOBILE ELEMENT ENZYME). FIG.3E, panel (B) depicts the helper RNA that is 5’- m7G capped (cap1) with flanking globin 5’- and 3’-UTRs, a 34 polyalanine tail region, and pseudouridine modification. The mobile element enzyme (e.g., a hyperactive mobile element enzyme in accordance with the present disclosure) is bioengineered to insert the donor DNA in a site- or locus specific manner. Example 2 – Creation of an AAV Packaging and Producer Cell Line A goal of this study was to assess the ability to produce high-titer recombinant AAV particles in a two-plasmid transfection protocol. FIG. 1A is a non-limiting representation of an AAV production strategy in accordance with embodiments of the present disclosure, using a donor vector to create a HEK293 producer cell line or another cell line including the E1a gene (e.g., engineered CHO-K1 or Vero cell line with E1a) that forms a replication deficient (rep-) AAV particle containing a transgene of interest. The single or dual donor comprising a transgene of interest is incorporated (e.g., by transfection such as electroporation) into a HEK293 cell line or another cell line including the E1a gene. The HEK293 cells are expanded to create a producer cell line and culturing the producer cells creates replication deficient (rep-) AAV particles with the transgene of interest. FIG.1B is a representation of an example of an inducible Rep/Cap and helper AAV donor plasmid construct (Kanar) used with a helper RNA or DNA to create an AAV producer cell line, in accordance with embodiments of the present disclosure. The donor DNA plasmid of FIG.1B includes helper E2A, E4 and VA genes flanked by insulators and mobile element enzyme recognition ends, to create an AAV producer cell line. FIG.1C is a representation of an example of a nucleic acid (plasmid) encoding a transgene (gene of interest (GOI)) included between AAV inverted terminal repeats (ITRs), in accordance with embodiments of the present disclosure. The plasmids shown in FIGs.1B and 1C can be combined or can be used separately for transfection into an E1+ cell line to produce AAV particle comprising the GOI. FIG.4 depicts a non-limiting representation of a conventional rAAV production system. Production in adenovirus complementation systems is usually performed as plasmid transfection processes, where AAV Rep/Cap genes, the ITR-flanked gene of interest (GOI), as well as Ad-helper genes are provided as three separate plasmids, respectively, to a E1a/E1b containing HEK293 cell line or other engineered cell line that contains E1a (e.g., engineered CHO-K1 or Vero cells). The present strategy is to include the AAV rep and cap genes, the AAV vector DNA sequences, and the essential Ad helper genes in a single donor plasmid flanked end sequences recognized by the mobile element enzyme in accordance with embodiments of the present disclosure. The AAV rep and cap genes are included under the control of t-REx, leading to rep and cap gene amplification, the AAV vector DNA sequences, and the essential Ad helper genes in a single donor plasmid flanked by the end sequences recognized by the mobile element enzyme in accordance with embodiments of the present disclosure (FIG.1B). The ability of the integrated AAV helper plasmid to direct the rescue, replication, and packaging of an AAV ITR-flanked transgene (e.g., the transgene of FIG.1C) will be assessed. The integrated AAV helper plasmid can direct the rescue, replication, and packaging of an AAV ITR-flanked transgene. This mobile element enzyme-mediated recombination system is expected to generate an integrated AAV helper plasmid that can facilitate the production of high-titer recombinant AAV particles in a simple two-plasmid transfection protocol. In addition, the inducible nature of the rep and cap genes is employed by the use of reagents (e.g., rAAV-5 plasmid reagents) useful for the construction of rAAV-5 vectors bearing other reporter and/or therapeutic transgenes. Example 3 – Identification of Excision Positive and Integration Negative Mutants TABLES 14A-14B depict hyperactive MLT mutants (TABLE 14A) and Integration-deficient (Int-) mutants (TABLE 14B) from MLT mobile element enzyme DNA. TABLE 14A. Hyperactive MLT mutants from mobile element enzyme protein of SEQ ID NO: 1 and mobile element enzyme DNA of SEQ ID NO: 2
Figure imgf000148_0001
Figure imgf000149_0001
TABLE 14B. Integration deficient MLT mutants from mobile element enzyme protein of SEQ ID NO: 1 and mobile element enzyme DNA of SEQ ID NO: 2
Figure imgf000149_0002
Example 4: Schematic Illustration for Constructs Used for Establishment of AAV producer stable cell lines, AAV2-H (Plasmid shown in FIG.5), AAV2-HB (Plasmid shown in FIG.6), and AAV-CDH (Plasmid shown in FIG.7) in HEK- 293R-22 tetR-expressing HEK-293 cells. Constructs pSF-ITRP5TO1-AAV2H, pSFP5TO1-AAV2HB, or pSFP5TO1-AAVCDH were used to co-transfect HEK- 293R-22 cells with pCMV-Engineered mobile element enzymeB VB200927-4525erk, and pSF-mCMVR to generate AAV stable producer/packaging cell pools, AAV2-H (Plasmid shown in FIG.5), AAV2-HB (Plasmid shown in FIG.6), and AAV-CDH (Plasmid shown in FIG.7), for subsequent single cell cloning, respectively. a. Plasmid with pSF-ITRP5TO1-AAV2H (FIG.5): pSF-ITRP5TO1-AAV2H is an engineered mobile element enzyme ITR containing plasmid that encodes 1) AAV2_REP78/68 ORFs under the control of P5TO1 promoter with AAV2_REP52/40 ORFs and CAPs (VP1/2/3) under their native P19 or P40 promoter, respectively, 2) Ad5 E2A_IRES_Ad5 E4 ORF6 under the control of the tetO-containing SV40 promoter, 3) VA RNAs under the control of HSV-1 ICP4 promoter, and 4) hygro-B resistant gene under the control of TK promoter. b. Plasmid with pSF-ITRP5TO1-AAV2HB (FIG. 6): pSFP5TO1-AAV2HB is an engineered mobile element enzyme ITR containing plasmid that encodes 1) AAV2_REP78/68 ORFs under the control of P5TO1 promoter and AAV2_REP52/40 ORFs under the native P19 promoter, 2) AAV2 CAPs under the tetO-containing P40 promoter with AAV9 P40 intron, 3) Ad5 E2A_IRES_Ad5 E4 ORF6 under the control of the tetO-containing SV40 promoter, 4) VA RNAs under the control of HSV-1 ICP4 promoter, and 5) hygro-B resistant gene under the control of TK promoter. c. Plasmid pSFP5TO1-AAVCDH (FIG.7): pSFP5TO1-AAVCDH is an engineered mobile element enzyme ITR containing plasmid that encodes 1) AAV2_REP 78/68 ORFs under the control of P5TO1 promoter and AAV2_REP52/40 ORFs under the native P19 promoter, 2) Ad5 E2A_IRES_Ad5 E4 ORF6 under the control of the tetO-containing SV40 promoter, 3) VA RNAs under the control of HSV-1 ICP4 promoter, and 4) hygro-B resistant gene under the control of TK promoter. Note that the nucleotide sequence between the end of native AAV Rep ORFs and native poly A was deleted in pSFP5TO1-AAVCDH to create an AAV Cap VP1/2/3 deletion phenotype. d. Plasmid pSFA2-EP40CAP2WZ1_SB (FIG.8): This construct encodes AAV2 CAP under the control of the tetO-containing hCMV-derived enhancer element 3/AAV2 P40 promoter with a modified AAV2 P40 TATA element and modified AAV2 P40 intron_1 (a at position 276 substituted for C) and WPRE/modified ICP27 poly A, followed by ICP0 promoter-Zeocin resistant gene-ICP0 poly A cassette flanked with ITRs for Sleeping Beauty. e. Plasmid pSFA2-EP40CAP2WZ2_SB (FIG.9): This construct encodes AAV2 CAP under the control of the tetO-containing hCMV enhancer element 3/AAV2 P40 promoter with a modified AAV2 P40 TATA element and modified AAV2 P40 intron_2 and WPRE/modified ICP27 poly A, followed by ICP0 promoter-Zeocin resistant gene-ICP0 poly A cassette flanked with SB ITRs. Establishment of AAV Producer/packaging cell lines a. Establishment of AAV2 full producer stable cell lines, AAVH cells, in HEK-293R-22 tetR-expressing HEK-293 cells: HEK-293R-22 cells were seeded at about 5 x 10e6 cells per 100 mm dish with normal HEK-293 cells growth medium. Cells were co-transfected with pSF-ITRP5TO1-AAV2H, CMV-Engineered mobile element enzymeB VB200927-4525erk, and pSF-mCMVR at a DNA:Lipofectamine 2000 ratio of 1:2. Transfection medium was removed at 3 and a half hour post transfection followed by addition of normal 293 cells growth medium. Transfected cells were passed on day 3 post transfection and grew in 293 cells growth medium in the presence of hygro-B at a concentration of 100 ug/ml. Hygro-B resistant single stable AAV-H colonies were then selected and analyzed by 1) ddPCR analyses with REP, E2A, ORF6, VA RNA and hygro-B resistant gene-specific primers, 2) western blot analysis with anti-AAV-2 REPs and CAPs specific antibodies, and 3) their ability to support AAV genome amplification following transduction with AAV2-EGFP viral particles. b. Establishment of second generation AAV2 full producer stable cell lines, AAVHB cells, in HEK-293R-22 tetR- expressing HEK-293 cells: HEK-293R-22 cells were seeded at about 5 x 10e6 cells per 100 mm dish with normal HEK- 293 cells growth medium. Cells were co-transfected with VB220909-1308jwt_pSF-ITRP5TO1-AAV2HB, CMV- Engineered mobile element enzymeB VB200927-4525erk, and pSF-mCMVR at a DNA:Lipofectamine 2000 ratio of 1:2. Transfection medium was removed at 3 and a half hour post transfection followed by addition of normal 293 cells growth medium. Transfected cells were passed on day 2 post transfection and grew in 293 cells growth medium in the presence of hygro-B at a concentration of 100 ug/ml. Hygro-B resistant single stable AAV-HB colonies were then selected and analyzed by 1) ddPCR analyses with REP, E2A, ORF6, VA RNA and hygro-B resistant gene-specific primers, 2) western blot analysis with anti-AAV-2 REPs and CAPs specific antibodies, and 3) their ability to support AAV genome amplification following transduction with AAV2-EGFP viral particles. c. Establishment of AAV serotype adaptable (Cap minus or Cap-) stable cell lines, AAV-CDH (Plasmid shown in FIG.7) cells, in HEK-293R-22 tetR-expressing HEK-293 cells: HEK-293R-22 cells were seeded at about 5 x 10e6 cells per 100 mm dish with normal HEK-293 cells growth medium. Cells were co-transfected with pSF-ITRP5TO1- AAVCDH, CMV-Engineered mobile element enzymeB VB200927-4525erk, and pSF-mCMVR at a DNA:Lipofectamine 2000 ratio of 1:2. Transfection medium was removed at 3 and a half hour post transfection followed by addition of normal 293 cells growth medium. Transfected cells were passed on day 3 post transfection and grew in 293 cells growth medium in the presence of hygro-B at a concentration of 100 ug/ml or 50 ug/ml. Hygro-B resistant single stable AAV-CDH (Plasmid shown in FIG.7) colonies were then selected and analyzed by 1) ddPCR analyses with REP, E2A, ORF6, VA RNA and hygro-B resistant gene-specific primers, 2) western blot analysis with anti-AAV-2 REPs specific antibody, and 3) their ability to support AAV genome amplification following transduction with AAV2-EGFP viral particles. d. Establishment of AAV2 REPs and CAPs expressing stable cell lines with sleeping beauty transposon- mediated integration of AAV2 CAPs-expressing cassettes into AAV2 capsid-minus stable cell lines: AAV2-capsid minus stable lines Clone CDH-48, CDH-24 and CDH-129 cells HEK-293 were seeded at about 4 to 5.5 x 10e6 cells per 100 mm dish with normal HEK-293 cells growth medium. Cells were co-transfected with of pSFA2-EP40CAP2WZ1-SB, pSFA2-EP40CAP2WZ2-SB, pSF-mCMVR, and sleeping beauty transposase SB100X Helper mRNA (VB210111- 1032qxg) at a DNA:Lipofectamine 2000 ratio of 1:2. Transfection medium was removed at 3 and a half hour post transfection followed by addition of normal 293 cells growth medium. Transfected cells were passed on day 2 post transfection with normal HEK-293 cells growth medium. Cells were then grown in HEK-293 cells growth medium in the presence of 100 ug/ml of zeocin. Zeocin resistant single stable colonies were then selected and analyzed by 1) ddPCR analyses with REP, Cap, E2A, ORF6, VA RNA and zeocin resistant gene-specific primers, 2) western blot analysis with anti-AAV-2 REPs and CAPs specific antibodies, and 3) their ability to support AAV genome amplification following transduction with AAV2-EGFP viral particles. ddPCR analyses for an engineered mobile element enzyme mediated Transposon Integration, AAV Genome Amplification/Rep Function, Cap Copy Insertion via Sleeping Beauty, and physical titer of rAAV particles. AAV genome amplification is supported in either AAV2 producer cell line clone H-17 (FIG.10A – 10B, Cap+) or AAV serotype adaptable packaging cell line clone CDH-13 (FIG.11A – 11B, Cap-). AAV genome amplification was used as a reference for expression of functional AAV Rep proteins in the AAV-H Clone H-17 or AAV-CDH (Plasmid shown in FIG.7) clone CDH-13 cells. Commercially available rAAV2-EGFP particles from SignaGen at 75 VGC/cell was used to transduce 3 x 10e6 cells/dish of Clone H-17, CDH-13, or HEK-293 (AAV Rep- control) cells. At 4 hours post- transduction, cells were switched to growth media with or without Doxycycline in the indicated concentration. GFP images (FIG.10A – FIG.11B) were taken ~120 hours post-transduction using Zeiss AxioVert A1 microscope and Axiocam 705 mono camera, followed by cell harvesting, genomic DNA extraction, and ddPCR analyses for quantification of the rAAV2-EGFP genome copy number per nanogram (ng) of total genomic DNA. The results clearly indicated that the rAAV-EGFP replicated in doxycycline dose responsive manner (ddPCR results at right), suggesting the inducible expression of functional AAV Rep proteins in the Clone H-17 and CDH-13 cells. Negative/low intensity EGFP image from transduced HEK-293 Rep negative cells also support this conclusion. Expression of functional AAV2 Rep proteins in AAV2-HB (Plasmid shown in FIG.6) clones as determined by western blot and ddPCR. FIG.12A shows the results of an western blot analysis illustrating cells from selected AAV2-HB (Plasmid shown in FIG. 6) cell clones HB-36, HB-42, HB-118, and HB-120 were treated with10ng/ml doxycycline for 72 hours followed by extraction with RIPA buffer in the presence of proteinase inhibitor. The resulting cell lysate supernatants were subjected to electrophoresis and western blot analyses. Lysate supernatants from HEK- 293 cells with or without transfection of pSFP5TO1-AAV2HB plasmid were analyzed as positive and negative controls. Cyclophilin B western blot was performed as a loading control. FIG.12B shows the results of a ddPCR analysis for AAV Genome Amplification / Rep function Assay. The AAV-HB stable clone #36, 42, 118 and 120 cells were transduced with the rAAV2-EGFP particles from SignaGen at low MOI, and genomic DNA extracted/ddPCR performed for quantification of the rAAV2-GFP genome copy number per nanogram (ng) of total genomic DNA analyzed. Western blot results clearly demonstrated that the rAAV Rep proteins are expressed and ddPCR results indicate the expressed AAV2 Rep proteins are functionally active in supporting AAV genome amplification, both in doxycycline responsive manner. Expression of functional AAV2 Rep proteins in AAV-CDH (Plasmid shown in FIG.7) clones as determined by western blot (FIG.13A) and ddPCR (FIG.13B). Cell extraction with RIPA buffer containing proteinase inhibitors, western blot, and ddPCR analyses of selected AAV- CDH (Plasmid shown in FIG.7) Clones of CDH-6, CDH-24, CDH-48, CDH-129, and CDH-175 were caried out as described in legend above. Like the AAVHB clones, western blot results clearly demonstrated that the rAAV Rep proteins are expressed while ddPCR results indicate the expressed AAV2 Rep proteins are functionally active in supporting AAV genome amplification, both in response to doxycycline. Note that CDH-48 and CDH-129 represent the top clones selected based on their Rep expression level, genome copy number/ng gDNA and the fold of induction. Establishment of AAV2 full producer stable cell pools, CDH-24/Cap-SB, CDH-48/Cap-SB, and CDH-129/Cap-SB with sleeping beauty transposase-mediated integration of AAV2 CAPs-expressing cassettes into AAV-CDH (Plasmid shown in FIG.7) serotype adaptable (Cap minus or Cap-) stable cell lines CDH-24, CDH48, and CDH- 129. Procedures for Establishment of AAV2 full producer stable cell pools, CDH-24/Cap-SB, CDH-48/Cap-SB, and CDH- 129/Cap-SB, with sleeping beauty (SB) transposase-mediated integration of AAV2 CAPs-expressing cassettes into AAV-CDH clone 24, 48, and 129 have been described above. FIG.14A shows ddPCR analyses of SB-mediated AAV2 Cap transposon integration in each Zeocin-selected stable pools. Average Cap copy number integration in each pool is summarized at the bottle of the penel. FIG.14B shows western blot analysis of SB-mediated AAV2 Cap transposon integration in each Zeocin-selected stable pools. Cells from the CDH-24/Cap-SB, CDH-48/Cap-SB, and CDH- 129/Cap-SB pools were treated with 20ng/ml doxycycline for 72 hours followed by extraction with RIPA buffer in the presence of proteinase inhibitor. Western blot analysis was performed similarly as describe above. The results show that stable integration of 25-40 copies of AAV-cap transposon enables detection of Cap expression in response to doxycycline. The lysate supernatants from HEK-293 cells with or without transient transfection of pSFA2- EP40CAP2WZ2-SB plasmid were analyzed as positive and negative controls. Cyclophilin B western blot was performed as a loading control. Expression of functional AAV2 Rep proteins in CDH-24/Cap-SB, CDH-48/Cap-SB, and CDH-129/Cap-SB pools as determined by western blot and ddPCR. Cell extraction with RIPA buffer containing proteinase inhibitors, western blot, and ddPCR analyses of the CDH-24/Cap- SB, CDH-48/Cap-SB, and CDH-129/Cap-SB cell pools were caried out as described previously. The western blot results in FIG.15A clearly demonstrated that the rAAV Rep proteins are expressed while ddPCR results in FIG.15B indicate the expressed AAV2 Rep proteins are functionally active in supporting AAV genome amplification, both in response to doxycycline. Expression of AAV2 Cap and Rep in CDH-48/Cap-SB stable pool increased with doxycycline dosages and over time. Cells from CDH-48/Cap-SB stable pools were incubated either in the presence of 0, 5, 10, 25, 50, or 100ng/ml doxycycline and harvest at 72 hours post induction; or incubated with 50ng/ml doxycycline and harvested at 24, 48, 72, or 96 hours post induction (FIG.16). Cell extraction with RIPA buffer containing proteinase inhibitors, western blot, and ddPCR analyses of the CDH-48/Cap-SB (AAV2-capsid minus stable lines Clone CDH-48) samples were caried out as described previously. The results show that expression of AAV2 Cap and Rep proteins increased over time in the presence of 50ng/ml doxycycline. The Cap and Rep expression also increased in response to increasing amounts of doxycycline. Production of competent rAAV2 particles from stably engineered CDH-48/Cap-SB cell pools in response to doxycycline. The cells from the CDH-48/Cap-SB stable pools were transduced with the rAAV2-EGFP particles from SignaGen at 500 VGC/cell in the presence or absence of 50ng/ml. 4 days post-transduction, culture media were removed, and cells harvested and washed twice with fresh growth media to remove the residue doxycycline. Upon resuspending in 7ml of fresh growth media without doxycycline, cells were subjected to 3 cycles of flash-freeze and thaw. rAAV-EGFP containing supernatants were collected following centrifugation at 2500rpm for 10 minutes AAV physical titer in each sample was measured by ddPCR as described above and data presented in FIG.17A. To test the presence of the competent rAAV2-EGFP particles in dox- and dox+ samples, the unconcentrated supernatants were used for transduction of CDH-48 Cap- cells at 1.5ml/well of the 6-well tissue culture plates in duplicates. The transduction media were replaced next day with fresh growth media and cells were harvested after additional 2 days incubation followed by fgenomic DNA extraction. ddPCR analyses of the genomic DNA samples were performed to evaluate copies of the rAAV-GFP copies introduced into CHD-48 Cap- cells through transduction and results presented in the FIG.17B. The ddPCR results from both panels indicated that rAAV-EGFP particles were produced in response to doxycycline induction and the produced rAAV particles were competent for transduction. Characterization of Zeocin-resistant single clones selected from the CDH-48/Cap-SB stable pool. Zeocin resistant single stable colonies were selected from the CDH-48/Cap-SB stable pool and analyzed by 1) ddPCR analysis for SB-mediated AAV2-Cap cassette integration (FIG.18A); 2) western blot analysis with antibodies specific for AAV2 CAP (FIG.18B), and AAV2 Rep (FIG.19A); and 3) ddPCR assays to evaluate their ability to support AAV genome amplification following transduction with low VGC/cell of AAV2-EGFP viral particles (FIG.19B). Specifically, data in FIG.18A indicated that SB-mediated AAV2-cap cassette integration ranged from 20-90 copies per ng of total genomic DNA. AAV2 Cap and Rep expression profile (both in expression level and ratio among the different Cap or Rep proteins) varies greatly among different clones as expected. Finally, all selected clones are capable to support AAV genome amplification to different degree and fold of induction varies as well. Materials and Methods a. Transposon copy number ddPCR: The Transposon Copy Number ddPCR Assay quantifies the copy numbers of five elements in the Engineered mobile element enzyme transposon per genome of HEK293R-22 based cells. These elements are AAV2 Rep, AAV2 Cap, Ad5 E2A, Ad5 E4 Orf6, and VA RNA. Genomic DNA was extracted from samples with the Zymo Research Quick-DNA MiniPrep Kit (D3025) according to the manufacturer’s protocol. Sample DNA concentrations were quantified by the Thermo Qubit 1X dsDNA Broad Range Assay Kit (Q33266) according to the manufacturer’s protocols and normalized to the same concentration in TE. ddPCR reactions were assembled with Manufacturer’s Supermix for Probes (no dUTP), a FAM-labeled hydrolysis probe-based ddPCR assays for one of the transposon elements, a HEX-labeled hydrolysis probe-based ddPCR assay for RPP30, HindIII restriction enzyme, and water according to Manufacturer’s ddPCR protocol. Sample DNA was added to reaction wells at inputs of 4 ng and 20 ng per well to ensure that at least one reaction per sample was within the range of the assay. Droplets were generated by the automated QX200 AutoDG system and thermal cycled according to Manufacturer̓s recommended parameters, with an annealing Tm of 60Υ. Thermal cycled droplets were analyzed by the CNV algorithm on the QX200 Droplet Reader, with the reference RPP30 assay set to 3 copies per genome (RPP30 is located on Chr10, which is triploid in the HEK293 cell line). Sample data was inspected for sufficient droplets (>10,000) and properly identified positive and negative droplets prior to analysis. The QX200 droplet reader software automatically calculates the copies per uL of reaction volume for the target (transposon element) and reference (RPP30) and reports the target copies per genome using the reference RPP30 = 3 copies per genome. Calculated copy numbers for all transposon elements were used to determine the integrity of the inserted transposon, as well as the copy number. Intact transposons had an integer copy number (e.g.1, 2, etc.) across all elements. b. AAV Genome Amplification / Rep function Assay: The Genomic Amplification Assay quantifies the copy number of AAV-GFP genomes per nanogram (ng) of genomic DNA analyzed. Genomic DNA was extracted from cells with the Zymo Research Quick-DNA MiniPrep Kit (D3025) according to the manufacturer’s protocol. Sample DNA concentrations were quantified by the Thermo Qubit 1X dsDNA Broad Range Assay Kit (Q33266) according to the manufacturer’s protocols and normalized to the same concentration in TE. ddPCR reactions were assembled with Manufacturer’s Supermix for Probes (no dUTP), a FAM-labeled hydrolysis probe-based ddPCR assay for EGFP, HindIII restriction enzyme, and water according to Manufacturer’s ddPCR protocol. Sample DNA was added to reaction wells at inputs of 0.02 to 20 ng per well to ensure that at least one reaction per sample was within the range of the assay. Droplets were generated by the automated QX200 AutoDG system and thermal cycled according to Manufacturer’s recommended parameters, with an annealing Tm of 60Υ. Thermal cycled droplets were analyzed by Direct Quantitation on the QX200 Droplet Reader. Sample data was inspected for sufficient droplets (>10,000) and properly identified positive and negative droplets prior to analysis. The QX200 droplet reader software automatically calculates the target (EGFP) copies per uL of reaction volume. The copy number of AAV-GFP genomes/ng DNA (y) was calculated by y = (a*b)/c, where a = EGFP copies per uL, b = 20uL total reaction volume, and c = ng DNA per reaction volume. c. Cap Copy Number ddPCR Assay: The Cap Copy Number ddPCR Assay quantifies the copy number of AAV2 Cap genes per genome of HEK293-based cells. Genomic DNA was extracted from samples with the Zymo Research Quick-DNA MiniPrep Kit (D3025) according to the manufacturer’s protocol. Sample DNA concentrations were quantified by the Thermo Qubit 1X dsDNA Broad Range Assay Kit (Q33266) according to the manufacturer’s protocols and normalized to the same concentration in TE. ddPCR reactions were assembled with Manufacturer’s Supermix for Probes (no dUTP), a FAM-labeled hydrolysis probe-based ddPCR assay for AAV2 Cap, a HEX-labeled hydrolysis probe-based ddPCR assay for RPP30, HindIII restriction enzyme, and water according to Manufacturer’s ddPCR protocol. Sample DNA was added to reaction wells at inputs of 4 ng and 20 ng per well to ensure that at least one reaction per sample was within the range of the assay. Droplets were generated by the automated QX200 AutoDG system and thermal cycled according to Manufacturer̓s recommended parameters, with an annealing Tm of 60Υ. Thermal cycled droplets were analyzed by the CNV algorithm on the QX200 Droplet Reader, with the reference RPP30 assay set to 3 copies per genome (RPP30 is located on Chr10, which is triploid in the HEK293 cell line). Sample data was inspected for sufficient droplets (>10,000) and properly identified positive and negative droplets prior to analysis. The QX200 droplet reader software automatically calculates the copies per uL of reaction volume for the target (AAV2 Cap) and reference (RPP30) and reports the target (AAV2 Cap) copies per genome using the reference RPP30 = 3 copies per genome. d. AAV physical titer ddPCR: The AAV Physical Titer ddPCR Assay quantifies the copy number of AAV-GFP genomes per mL of AAV sample analyzed. A series of eight 10-fold serial dilutions (10E0-10E-7) of AAV samples were created to ensure that several dilutions would fall within the range of ddPCR quantitation. Serial dilutions were created in nuclease-free water with 0.05% Pluronic F-68 (Gibco 24040-032). AAV particles were lysed by incubation at 95Υ for 10 minutes, followed by cooling to 4Υ at a rate of 3Υ per minute. ddPCR reactions were assembled with Manufacturer̓s Supermix for Probes (no dUTP), a FAM-labeled hydrolysis probe-based ddPCR assay for EGFP, HindIII restriction enzyme, and water according to Manufacturer̓s ddPCR protocol. AAV sample dilutions were added to reaction wells at 5 uL per well. Droplets were generated by the automated QX200 AutoDG system and thermal cycled according to Manufacturer̓s recommended parameters, with an annealing Tm of 60Υ. Thermal cycled droplets were analyzed by Direct Quantitation on the QX200 Droplet Reader. Sample data was inspected for sufficient droplets (>10,000) and properly identified positive and negative droplets prior to analysis. The QX200 droplet reader software automatically calculates the target (EGFP) copies per uL of reaction volume. The titer of AAV genomes per uL of sample (y) was calculated by y = (20*A*B*1000)/C, where 20 = total uL of reaction volume, A = AAV-GFP genome copies/uL of reaction volume, B = dilution factor of AAV sample, 1000 = uL per mL (conversion factor), and C = uL of AAV sample dilution in reaction volume. Western Blot analyses for detecting AAV Rep and Cap Expression: a. Cell lysate preparation: Upon wash and scrape, ~1+E7 cells in 10 ml of same PBS containing 1mM PMSF and 100ug/ml TPCK or Halt™ Protease and Phosphatase Inhibitor Cocktail, EDTA-free (100X) (Cat# 78441) were pelleted down by centrifugation at 3000 rpm for 15 min at 4c and resuspend in 600 ul of RIPA buffer containing 1mM PMSF, 100ug/ml TPCK, and 50ug/ml leupeptin or Pierce Protease and Phosphatase Inhibitor Mini Tablets (cat# A32959) . Samples were Incubated on ice for 40 min – vortex each tube for 10 seconds every 15 minutes followed by centrifugation at 13,000 rpm for 2 min at 4c. The supernatants were quick-frozen down in 100 ul to 150 ul aliquots with dry ice and stored at -80 C. b. Protein concentration, SDS-PAGE and western blot analyses: Protein concentration from each cell lysate supernatants were determined in triplicates using Manufacturer DC Protein Quantitation Assay (Cat no: 5000112) according to the manufacturer’s instruction. Upon heat treatment in 1x laemmi buffer containing 10% BME and 0.5M UREA, equal amounts of protein samples (up to 100ug /well) were loaded onto Manufacturer Criterion TGX gradient (8-16% or 4-20%) gels, separated by electrophoresis at 85V for ~2.5hr followed by protein transfer to PVDF membrane using Manufacturer Trans-Blot Turbo system. Blocking and antibody incubation steps were performed using EveryBlot Blocking Buffer (cat# 1201002). AAV Rep, AAV Cap, and host cell Cyclophilin B (a loading control) on PVDF membranes were probed with mouse anti-AAV2 Rep (clone 303.9, Progen Catlog# 61069 at 1:100x), mouse anti-AAV VP1/VP2/VP3 (clone B1, Progen 65158, supernatant at 1:25x), or mouse anti-Cyclophilin B (Abcam, ab236760 at 1:10,000x) followed by HRP conjugated secondary goat-anti-mouse IgG H&L antibody (AbCam, ab6789 at 2000x to 10,000x). Upon adding developing solution (1:1 peroxide SuperSignal™ West Pico PLUS Chemiluminescent Substrate for REP/CAP, or 1:1 peroxide: enhancer: ECL for Cyclophilin B), the western blot images were visualized and captured using iBright Imaging System. GFP images a. GFP expression images were captured using Zeiss AxioVert A1 microscope, Axiocam 705 mono camera, 10x objective, 150ms GFP exposure. EQUIVALENTS
While the invention has been described in connection with specific embodiments thereof, it will be understood that it is capable of further modifications and this application is intended to cover any variations, uses, or adaptations of the invention following, in general, the principles of the invention and including such departures from the present disclosure as come within known or customary practice within the art to which the invention pertains and as may be applied to the essential features herein set forth and as follows in the scope of the appended claims.
Those skilled in the art will recognize, or be able to ascertain, using no more than routine experimentation, numerous equivalents to the specific embodiments described specifically herein. Such equivalents are intended to be encompassed in the scope of the following claims.
INCORPORATION BY REFERENCE
All patents and publications referenced herein are hereby incorporated by reference in their entireties.
The publications discussed herein are provided solely for their disclosure prior to the filing date of the present application. Nothing herein is to be construed as an admission that the present invention is not entitled to antedate such publication by virtue of prior invention.
As used herein, all headings are simply for organization and are not intended to limit the disclosure in any manner. The content of any individual section may be equally applicable to all sections.

Claims

CLAIMS What is claimed is: 1. A method of making a viral particle packaging and producer cell line, the method comprising: transfecting a cell with two or more nucleic acids, wherein the nucleic acids encode one or more of: (a) an enzyme capable of performing targeted genomic integration, (b) an inducible viral replication (Rep) gene, (c) an inducible viral Capsid (cap) gene, (d) one or more adenoviral auxiliary genes, optionally selected from one or more of E1A, E1B, E4, E2A, and VA of an AAV, optionally E1A and E1B, (e) an insulator, optionally selected from HS4, D4Z4, (f) one or more terminal ends recognized by the enzyme, and (g) a transgene flanked by AAV inverted terminal repeats (ITRs), wherein the inducible viral replication (Rep) and Capsid (cap) genes are controlled by at least one of a tetO- containing P5 promoter, P19 promoter, SV40 promoter, P40 promoter, or a functional variant or fragment thereof, to thereby result in a transfected cell that expresses the transgene in association with the viral particle. 2. A method of making a viral particle packaging and producer cell line, the method comprising: transfecting an E1A, E1B+ cell with two or more nucleic acids, wherein the nucleic acids encode one or more of: (a) an enzyme capable of performing targeted genomic integration, (b) an inducible viral replication (Rep) gene, (c) an inducible viral Capsid (cap) gene, (d) an insulator, optionally selected from HS4, D4Z4, (e) one or more terminal ends recognized by the enzyme, and (f) a transgene flanked by AAV inverted terminal repeats (ITRs), wherein the inducible viral replication (Rep) and Capsid (cap) genes are controlled by at least one of a tetO- containing P5 promoter, P19 promoter, SV40 promoter, P40 promoter, or a functional variant or fragment thereof, to thereby result in a transfected cell that expresses the transgene in association with the viral particle. 3. The method of claim 1, wherein the cell is transfected with: (1) two nucleic acids, and wherein the first nucleic acid encodes (a), and the second nucleic acid encodes (b), (c), (d), (e), (f), and (g); (2) three nucleic acids, and wherein the first nucleic acid encodes (a), the second nucleic acid encodes (b), (c), (d), (e), and (f), and the third nucleic acid encodes (g); or (3) four nucleic acids, and wherein the first nucleic acid encodes (a), the second nucleic acid encodes (b), (d), (e), and (f), the third nucleic acid encodes (c), and the fourth nucleic acid encodes (g). 4. The method of claim 2, wherein the cell is transfected with: (1) two nucleic acids, and wherein the first nucleic acid encodes (a), and the second nucleic acid encodes (b), (c), (d), (e), and (f); (2) three nucleic acids, and wherein the first nucleic acid encodes (a), the second nucleic acid encodes (b), (c), (d), and (e), and the third nucleic acid encodes (f); or (3) four nucleic acids, and wherein the first nucleic acid encodes (a), the second nucleic acid encodes (b), (d), and (e), the third nucleic acid encodes (c), and the fourth nucleic acid encodes (f). 5. The method of claim 1 or claim 2, wherein the cell is human embryonic kidney (HEK293), Chinese hamster ovary (CHO) E1A, E1B+ engineered CHO-K1, or Spodoptera frugiperda (Sf9) cell line, baby hamster kidney (BHK), vero cell. 6. The method of claim 1, wherein the inducible viral replication (Rep) and Capsid (cap) genes are controlled by a tetO-containing P40 promoter. 7. The method of claim 6, wherein the tetO-containing P40 promoter, or a functional variant or fragment thereof, further comprises a modified TATA box element and a modified P40 intron. 8. The method of claim 7, wherein the tetO-containing P40 promoter is a tetO-containing AAV2 P40 promoter. 9. The method of claim 8, wherein the tetO-containing AAV2 P40 promoter comprises a nucleotide sequence of SEQ ID NO: 816, or a functional variant or fragment thereof, or sequence having at least about 70%, or at least about 80%, or at least about 85%, or at least about 90%, or at least about 95%, or at least about 97% identity thereto, or at least about 98% identity thereto, or at least about 99% identity thereto or at least about 10 mutations, or at least about 9 mutations, or at least about 8 mutations, or at least about 7 mutations, or at least about 6 mutations, or at least about 5 mutations, or at least about 4 mutations, or at least about 3 mutations, or at least about 2 mutations, or at least about 1 mutation. 10. The method of any one of claims 6-9, wherein the P40 intron is an AAV2 P40 intron. 11. The method of claim 10, wherein the AAV2 P40 intron comprises a C276A substitution, or a substitution at position corresponding thereto relative to SEQ ID NO: 818. 12. The method of claim 10 or 11, wherein the AAV2 P40 intron comprises one or more mutated translation start sites (ATGs), optionally wherein the translation start sites are mutated to one of CTG, ACG, and TTG. 13. The method of any one of claims 10-12, wherein the AAV2 P40 intron comprises substitutions at one or more positions A13, A32, T42, A61, A71, A89, A203, A246, A258, and T282, or one or more positions corresponding thereto, relative to SEQ ID NO: 818. 14. The method of any one of claims 10-13, wherein the AAV2 P40 intron comprises substitutions at one or more positions A13C, A32C, T42C, A61C, A71T, A89C, A203T, A246C, A258C, and T282C corresponding to SEQ ID NO: 818. 15. The method of any one of claims 10-14, wherein the AAV2 P40 intron comprises the nucleotide sequence of SEQ ID NO: 818, or a functional variant or fragment thereof, or sequence having at least about 70%, or at least about 80%, or at least about 85%, or at least about 90%, or at least about 95%, or at least about 97% identity thereto, or at least about 98% identity thereto, or at least about 99% identity thereto, or at least about 10 mutations, or at least about 9 mutations, or at least about 8 mutations, or at least about 7 mutations, or at least about 6 mutations, or at least about 5 mutations, or at least about 4 mutations, or at least about 3 mutations, or at least about 2 mutations, or at least about 1 mutation. 16. The method of any one of claims 10-15, wherein the AAV2 P40 intron comprises the nucleotide sequence of SEQ ID NO: 819, or a functional variant or fragment thereof, or sequence having at least about 70%, or at least about 80%, or at least about 85%, or at least about 90%, or at least about 95%, or at least about 97% identity thereto, or at least about 98% identity thereto, or at least about 99% identity thereto, or at least about 10 mutations, or at least about 9 mutations, or at least about 8 mutations, or at least about 7 mutations, or at least about 6 mutations, or at least about 5 mutations, or at least about 4 mutations, or at least about 3 mutations, or at least about 2 mutations, or at least about 1 mutation. 17. The method of any one of claims 10-15, wherein the AAV2 P40 intron comprises the nucleotide sequence of SEQ ID NO: 817, or a functional variant or fragment thereof, or sequence having at least about 70%, or at least about 80%, or at least about 85%, or at least about 90%, or at least about 95%, or at least about 97% identity thereto, or at least about 98% identity thereto, or at least about 99% identity thereto, or at least about 10 mutations, or at least about 9 mutations, or at least about 8 mutations, or at least about 7 mutations, or at least about 6 mutations, or at least about 5 mutations, or at least about 4 mutations, or at least about 3 mutations, or at least about 2 mutations, or at least about 1 mutation. 18. The method of any one of claims 6-17, wherein the modified TATA box element has the nucleotide sequence of TATATAA. 19. The method of any one of claims 6-18, wherein the tetO-containing AAV2 P40 promoter or a functional variant or fragment thereof, comprising a modified TATA box element and a modified AAV2 P40 intron comprises the nucleotide sequence of SEQ ID NO: 823, or a functional variant or fragment thereof, or sequence having at least about 70%, or at least about 80%, or at least about 85%, or at least about 90%, or at least about 95%, or at least about 97% identity thereto, or at least about 98% identity thereto, or at least about 99% identity thereto, or at least about 10 mutations, or at least about 9 mutations, or at least about 8 mutations, or at least about 7 mutations, or at least about 6 mutations, or at least about 5 mutations, or at least about 4 mutations, or at least about 3 mutations, or at least about 2 mutations, or at least about 1 mutation. 20. The method of claim 6, wherein the tetO-containing P40 promoter is a tetO-containing AAV2 P40 promoter. 21. The method of claim 20, wherein the AAV2 P40 promoter comprises the nucleotide sequence of SEQ ID NO: 820, or a functional variant or fragment thereof, or sequence having at least about 70%, or at least about 80%, or at least about 85%, or at least about 90%, or at least about 95%, or at least about 97% identity thereto, or at least about 98% identity thereto, or at least about 99% identity thereto, or at least about 10 mutations, or at least about 9 mutations, or at least about 8 mutations, or at least about 7 mutations, or at least about 6 mutations, or at least about 5 mutations, or at least about 4 mutations, or at least about 3 mutations, or at least about 2 mutations, or at least about 1 mutation. 22. The method of claim20 or 21, wherein the modified P40 intron is a AAV9 P40 intron. 23. The method of claim 22, wherein the modified AAV9 P40 intron comprises the nucleotide sequence of SEQ ID NO: 821, or a functional variant or fragment thereof, or sequence having at least about 70%, or at least about 80%, or at least about 85%, or at least about 90%, or at least about 95%, or at least about 97% identity thereto, or at least about 98% identity thereto, or at least about 99% identity thereto, or at least about 10 mutations, or at least about 9 mutations, or at least about 8 mutations, or at least about 7 mutations, or at least about 6 mutations, or at least about 5 mutations, or at least about 4 mutations, or at least about 3 mutations, or at least about 2 mutations, or at least about 1 mutation. 24. The method of any one of claims 20-23, wherein the modified TATA box element has the nucleotide sequence of TATATAA.
25. The method of any one of claims 20-24, wherein tetO-containing AAV2 P40 promoter or a functional variant or fragment thereof, comprising a modified TATA box element and a modified AAV9 P40 intron comprises the nucleotide sequence of SEQ ID NO: 822, or a functional variant or fragment thereof, or sequence having at least about 70%, or at least about 80%, or at least about 85%, or at least about 90%, or at least about 95%, or at least about 97% identity thereto, or at least about 98% identity thereto, or at least about 99% identity thereto, or at least about 10 mutations, or at least about 9 mutations, or at least about 8 mutations, or at least about 7 mutations, or at least about 6 mutations, or at least about 5 mutations, or at least about 4 mutations, or at least about 3 mutations, or at least about 2 mutations, or at least about 1 mutation. 26. The method of any one of claim 6-9 or 22-24, wherein the tetO-containing AAV2 P40 promoter or a functional variant or fragment thereof, comprising a modified TATA box element and a modified AAV9 P40 intron comprises a nucleotide sequence of SEQ ID NO: 824, or a functional variant or fragment thereof, or sequence having at least about 70%, or at least about 80%, or at least about 85%, or at least about 90%, or at least about 95%, or at least about 97% identity thereto, or at least about 98% identity thereto, or at least about 99% identity thereto, or at least about 10 mutations, or at least about 9 mutations, or at least about 8 mutations, or at least about 7 mutations, or at least about 6 mutations, or at least about 5 mutations, or at least about 4 mutations, or at least about 3 mutations, or at least about 2 mutations, or at least about 1 mutation. 27. The method of any one of claims 6-26, wherein the P40 promoter further comprises one or more enhancer elements. 28. The method of claim 27, wherein the enhancer element comprises one or more cis-acting elements, optionally selected from an Sp1 binding site, GC rich sequence, GCGGAAC motif, TAATGARAT element, AP1 binding site, and CCAAT box element. 29. The method of claim 27 or 28, wherein the enhancer element comprises about 1 to about 5 Sp1 binding sites, optionally about 1, or about 2, or about 3, or about 4, or about 5 Sp1 binding sites. 30. The method of any one of claims 27-29, wherein the enhancer element comprises about 1 or about 2 GC rich sequences. 31. The method of any one of claims 27-30, wherein the enhancer element is derived from an hCMV Enhancer Element-3. 32. The method of claim 31, wherein the hCMV Enhancer Element-3 comprises the nucleotide sequence of SEQ ID NO: 830 or a functional variant or fragment thereof, or sequence having at least about 70%, or at least about 80%, or at least about 85%, or at least about 90%, or at least about 95%, or at least about 97% identity thereto, or at least about 98% identity thereto, or at least about 99% identity thereto, or at least about 10 mutations, or at least about 9 mutations, or at least about 8 mutations, or at least about 7 mutations, or at least about 6 mutations, or at least about 5 mutations, or at least about 4 mutations, or at least about 3 mutations, or at least about 2 mutations, or at least about 1 mutation. 33. The method of claim 1, wherein the inducible promoter is an antibiotic-dependent promoter, optionally a tetracycline-dependent promoter or a variant thereof, or a cumate or coumermycin/novobiocin promoter or a variant thereof. 34. The method of any one of claims 1-33, wherein the viral particle is an AAV, and optionally wherein the AAV is selected from AAV1, AAV2, AAV5, AAV6, AAV7, AAV8, and AAV9. 35. The method of any one of claims 1-34, wherein any one or more of the first, second, and third nucleic acids is encoded by a single nucleic acid. 36. The method of any one of claims 1-35, wherein the second nucleic acid and/or the third nucleic acid are included in a single expression vector. 37. The method of any one of claims 1-36, wherein the first and second nucleic acids are included in a single expression vector, and the third nucleic acid is included in an expression vector that is different from the expression vector including the first and second nucleic acids. 38. The method of claim 36 or claim 37, wherein the expression vector is or comprises a plasmid. 39. The method of claim 38, wherein the plasmid comprises the nucleotide sequence of SEQ ID NO: 825 or a functional variant or fragment thereof, or sequence having at least about 70%, or at least about 80%, or at least about 85%, or at least about 90%, or at least about 95%, or at least about 97% identity thereto, or at least about 98% identity thereto, or at least about 99% identity thereto, or at least about 10 mutations, or at least about 9 mutations, or at least about 8 mutations, or at least about 7 mutations, or at least about 6 mutations, or at least about 5 mutations, or at least about 4 mutations, or at least about 3 mutations, or at least about 2 mutations, or at least about 1 mutation. 40. The method of claim 38, wherein the plasmid is substantially of the form as show in FIG. 8, or a functional equivalent thereof. 41. The method of claim 38, wherein the plasmid comprises a AAV CAP under the control of an adeno-associated viral (AAV) system encoding AAV2 CAP comprising: a tetO-containing P40 promoter with a modified AAV2 P40 TATA element and modified AAV2 P40 intron, wherein the AAV2 P40 intron comprises the nucleotide sequence of SEQ ID NO: 818 or a functional variant or fragment thereof, an hCMV-derived enhancer element, a Zeocin resistance gene, a WPRE regulatory element, and a cassette flanked with Sleeping beauty ITRs
42. The method of claim 38, wherein the plasmid comprises the nucleotide sequence of SEQ ID NO: 826 or a functional variant or fragment thereof, or sequence having at least about 70%, or at least about 80%, or at least about 85%, or at least about 90%, or at least about 95%, or at least about 97% identity thereto, or at least about 98% identity thereto, or at least about 99% identity thereto, or at least about 10 mutations, or at least about 9 mutations, or at least about 8 mutations, or at least about 7 mutations, or at least about 6 mutations, or at least about 5 mutations, or at least about 4 mutations, or at least about 3 mutations, or at least about 2 mutations, or at least about 1 mutation. 43. The method of claim 38, wherein the plasmid is substantially of the form as show in FIG. 9, or a functional equivalent thereof. 44. The method of claim 38, wherein the plasmid comprises a AAV CAP under the control of an adeno-associated viral (AAV) system encoding AAV2 CAP comprising: a tetO-containing P40 promoter with a modified AAV2 P40 TATA element and modified AAV2 P40 intron, wherein the AAV2 P40 intron comprises the nucleodie sequence of SEQ ID NO: 819 or a functional variant or fragment thereof, an hCMV-derived enhancer element, a Zeocin resistance gene, a WPRE regulatory element, and a cassette flanked with Sleeping beauty ITRs. 45. The method of claim 38, wherein the plasmid comprises the nucleotide sequence of SEQ ID NO: 841 or a functional variant or fragment thereof, or sequence having at least about 70%, or at least about 80%, or at least about 85%, or at least about 90%, or at least about 95%, or at least about 97% identity thereto, or at least about 98% identity thereto, or at least about 99% identity thereto, or at least about 10 mutations, or at least about 9 mutations, or at least about 8 mutations, or at least about 7 mutations, or at least about 6 mutations, or at least about 5 mutations, or at least about 4 mutations, or at least about 3 mutations, or at least about 2 mutations, or at least about 1 mutation. 46. The method of claim 38, wherein the plasmid is substantially of the form as show in FIG. 7, or a functional equivalent thereof. 47. The method of claim 38, wherein the plasmid comprises a tetO-containing P5 promoter, a tetO-containing SV40 early promoter, a modified TATA box element, an enhancer element derived from a HSV-1 ICP4 promoter, a hygromycin B resistance gene and a cassette flanked with inverted terminal repeats (ITRs) derived from an engineered mobile element enzyme. 48. The method of claim 38, wherein the plasmid comprises the nucleotide sequence of SEQ ID NO: 842, or a functional variant or fragment thereof, or sequence having at least about 70%, or at least about 80%, or at least about 85%, or at least about 90%, or at least about 95%, or at least about 97% identity thereto, or at least about 98% identity thereto, or at least about 99% identity thereto, or at least about 10 mutations, or at least about 9 mutations, or at least about 8 mutations, or at least about 7 mutations, or at least about 6 mutations, or at least about 5 mutations, or at least about 4 mutations, or at least about 3 mutations, or at least about 2 mutations, or at least about 1 mutation.
49. The method of claim 38, wherein the plasmid is substantially of the form as show in FIG. 5, or a functional equivalent thereof. 50. The method of claim 38, wherein the plasmid comprises comprises a tetO-containing P5 promoter, a P40 promoter, a tetO-containing SV40 containing promoter, a modified TATA box element, a HSV-2 ICP4 promoter, a hygromycin B resistance gene, and a cassette flanked with inverted terminal repeats (ITRs) derived from an engineered mobile element enzyme. 51. The method of claim 38, the plasmid comprises the nucleotide sequence of SEQ ID NO: 843, or a functional variant or fragment thereof, or sequence having at least about 70%, or at least about 80%, or at least about 85%, or at least about 90%, or at least about 95%, or at least about 97% identity thereto, or at least about 98% identity thereto, or at least about 99% identity thereto, or at least about 10 mutations, or at least about 9 mutations, or at least about 8 mutations, or at least about 7 mutations, or at least about 6 mutations, or at least about 5 mutations, or at least about 4 mutations, or at least about 3 mutations, or at least about 2 mutations, or at least about 1 mutation. 52. The method of claim 38, wherein the plasmid is substantially of the form as show in FIG. 6, or a functional equivalent thereof. 53. The method of claim 38, wherein the plasmid comprises a tetO-containing P5 promoter, a tetO-containing P40 promoter with AAV9 P40 intron, a tetO-containing SV40 containing promoter, a modified TATA box element, a HSV-1 ICP4 promoter, a hygromycin B resistance gene, and a cassette flanked with inverted terminal repeats (ITRs) derived from an engineered mobile element enzyme. 54. The method of any one of claims 1-53, wherein one or more of the first, second, and third nucleic acids is or comprises RNA, optionally mRNA, optionally synthetic mRNA or modified mRNA. 55. The method of any one of claims 1-54, wherein one or more of the first, second, and third nucleic acids is DNA, optionally plasmid DNA. 56. The method of any one of claims 1-55, wherein one or more of the first, second, and third nucleic acids is an expression vector, wherein the expression vector is optionally a plasmid. The method of any one of claims 1-56, wherein the transgene is flanked by AAV inverted terminal repeats (ITRs). 58. The method of claim 1 or 57, wherein the terminal ends or ITRs comprise the nucleotide sequence of SEQ ID NO: 831 and/or SEQ ID NO: 832, or a functional variant or fragment thereof, or sequence having at least about 70%, or at least about 80%, or at least about 85%, or at least about 90%, or at least about 95%, or at least about 97% identity thereto, or at least about 98% identity thereto, or at least about 99% identity thereto, or at least about 10 mutations, or at least about 9 mutations, or at least about 8 mutations, or at least about 7 mutations, or at least about 6 mutations, or at least about 5 mutations, or at least about 4 mutations, or at least about 3 mutations, or at least about 2 mutations, or at least about 1 mutation. 59. The method of any one of claims 1-58, wherein the transgene has a size of about 200,000 bases or less. 60. The method of any one of claims 1-59, further comprising culturing the transfected cell in a medium that expands a population of the transfected cells to create a stably transfected packaging and producer cell line. 61. The method of claim 60, wherein the stably transfected producer cell line is capable of producing replication- deficient viral particles in association with the transgene. 62. The method of any one of claims 1-61, wherein the transfection comprises electroporation, nucleofection, lipofection, or calcium phosphate transfection. 63. The method of any one of claims 1-62, wherein the method is helper virus-free. 64. The method of any one of claims 1-63, wherein the enzyme capable of performing targeted genomic integration is a recombinase. 65. The method of claim 64, wherein the recombinase is an integrase or a mobile element enzyme. 66. The method of claim 64 or 65, wherein the enzyme capable of performing targeted genomic integration is a mobile element enzyme. 67. The method of any one of claims 1-66, wherein the enzyme is derived from Bombyx mori, Xenopus tropicalis, Trichoplusia ni, Myotis lucifugus, Rhinolophus ferrumequinum, Rousettus aegyptiacus, Phyllostomus discolor, Myotis myotis, Pteropus vampyrus, Pipistrellus kuhlii, Molossus molossus, Pan troglodytes, or Homo sapiens. 68. The method of any one of claims 1-67, wherein the enzyme is an engineered version, including but not limited to hyperactive forms, of an enzyme derived from Bombyx mori, Xenopus tropicalis, Trichoplusia ni, Myotis lucifugus, Rhinolophus ferrumequinum, Rousettus aegyptiacus, Phyllostomus discolor, Myotis myotis, Pteropus vampyrus, Pipistrellus kuhlii, Molossus molossus, Pan troglodytes, or Homo sapiens. 69. The method of any one of claims 1-68, wherein the mobile element enzyme is from one or more of the Sleeping beauty, Tn1, Tn2, Tn3, Tn5, Tn7, Tn9, Tn10, Tn552, Tn903, Tn1000/Gamma-delta, Tn/O, tnsA, tnsB, tnsC, tniQ, IS10, ISS, IS911, Minos, piggyBac, Tol2, Mos1, Himar1, Hermes, Tol2, Minos, Tel, P-element, MuA, Ty1, Chapaev, transib, Tc1/mariner, or Tc3 donor DNA system, or biologically active fragments variants thereof, inclusive of hyperactive variants. 70. The method of any one of claims 1-69, wherein the mobile element enzyme has the amino acid sequence of SEQ ID NO: 1, or an amino acid sequence having at least about 80%, or an amino acid sequence having at least about 90%, or at least about 93%, or at least about 95%, or at least about 97%, or at least about 98%, or at least about 99% identity thereto. 71. The method of any one of claims 1-70, wherein the mobile element enzyme comprises an amino acid other than serine at the position corresponding to position 2 of SEQ ID NO: 1. 72. The method of claim 71, wherein the amino acid is a non-polar aliphatic amino acid, optionally a non-polar aliphatic amino acid optionally selected from G, A, V, L, I and P, optionally A. 73. The method of any one of claims 70-72, wherein the mobile element enzyme does not have additional residues at the C terminus relative to SEQ ID NO: 1. 74. The method of any one of claims 1-73, wherein the enzyme has one or more mutations which confer hyperactivity. 75. The method of any one of claims 70-74, wherein the enzyme has one or more amino acid substitutions selected from S8X1 and/or C13X2, or positions corresponding thereto relative to SEQ ID NO: 1. 76. The method of claim 75, wherein the enzyme has S8X1 and/or C13X2 substitutions, at positions corresponding thereto relative to SEQ ID NO: 1. 77. The method of claim 76, wherein the enzyme has S8X1 and C13X2 substitutions, at positions corresponding thereto relative to SEQ ID NO: 1. 78. The method of claim 77, wherein the enzyme has S8X1 substitution, at position corresponding thereto relative to SEQ ID NO: 1. 79. The method of claim 78, wherein the enzyme has C13X2 substitution, at positions corresponding thereto relative to SEQ ID NO: 1. 80. The method of any one of claims 75-79, wherein X1 is selected from G, A, V, L, I, and P and X2 is selected from K, R, and H. 81. The method of claim 80, wherein: X1 is P and X2 is R. 82. The method of any one of claims 66-81, wherein the enzyme comprises an amino acid sequence of SEQ ID NO: 11. 83. The method of any one of claims 66-82, wherein the enzyme comprises at least one, at least five, at least seven, at least nine, or ten substitutions at positions corresponding to: 5, 8, 9, 10, 11, 14, 22, 36, 37, 54, 130, 239, 281, 282, 283, 284, 285, 294, 300, 310, 330, 331, 332, 333, 334, 335, 336, 337, 338, 339, 345, 375, 416, 427, 475, 481, 491, 520, and/or 561 of SEQ ID NO: 11.
84. The method of any one of claims 66-83, wherein the enzyme comprises one, two, three, four, five, six, seven, eight, nine, or ten substitutions at positions corresponding to: 5, 8, 9, 10, 11, 14, 22, 36, 37, 54, 130, 239, 281, 282, 283, 284, 285, 294, 300, 310, 330, 331, 332, 333, 334, 335, 336, 337, 338, 339, 345, 375, 416, 427, 475, 481, 491, 520, and/or 561 of SEQ ID NO: 11. 85. The method of any one of claims 66-84, wherein the enzyme comprises at least one, at least five, at least seven, at least nine, or ten substitutions selected from S5P, S8F, D9G, D10G, E11G, A14V, T22C, S36G, T37C, S54N, K130T, G239R, Y281A, C282A, G283A, E284A, G285A, T294A, T300A, N310A, G330A, T331A, I332A, R333A, K334A, N335A, R336A, G337A, I338A, P339A, I345V, T375G, D416A, R427H, D475G, M481V, P491Q, A520T, and A561T, wherein the positions are corresponding to positions of SEQ ID NO: 11. 86. The method of any one of claims 66-85, wherein the enzyme comprises one, two, three, four, five, six, seven, eight, nine, or ten substitutions selected from S5P, S8F, D9G, D10G, E11G, A14V, T22C, S36G, T37C, S54N, K130T, G239R, Y281A, C282A, G283A, E284A, G285A, T294A, T300A, N310A, G330A, T331A, I332A, R333A, K334A, N335A, R336A, G337A, I338A, P339A, I345V, T375G, D416A, R427H, D475G, M481V, P491Q, A520T, and A561T, wherein the positions are corresponding to positions of SEQ ID NO: 11. 87. The method of any one of claims 66-86, wherein the mobile element enzyme is an engineered mammalian mobile element enzyme. 88. The method of any one of claims 66-87, wherein the mobile element enzyme is a mammal-derived, helper RNA mobile element enzyme. 89. The method of any one of claims 66-88, wherein the mobile element enzyme is a mammal-derived, helper DNA mobile element enzyme. 90. The method of any one of claims 64-89, wherein the enzyme is capable of inserting a donor DNA at a TA dinucleotide site. 91. The method of any one of claims 64-90, wherein the enzyme is capable of inserting a donor DNA at a TTAA (SEQ ID NO: 440) tetranucleotide site. 92. The method of any one of claims 64-91, wherein the mobile element enzyme has gene cleavage activity (Exc+) and/or gene integration activity (Int+), and the mobile element enzyme having at least about 90% identity to the amino acid sequence of SEQ ID NO: 1, SEQ ID NO: 3, SEQ ID NO: 4, SEQ ID NO: 6, SEQ ID NO: 7, SEQ ID NO: 8, SEQ ID NO: 9, SEQ ID NO: 10, SEQ ID NO: 11 or SEQ ID NO: 430, or a nucleotide sequence encoding the same. 93. The method of claim 92, wherein the mobile element enzyme has at least about 95%, or at least about 96%, at least about 97%, at least about 98%, at least about 99% identity to the amino acid sequence of SEQ ID NO: 1, SEQ ID NO: 3, SEQ ID NO: 4, SEQ ID NO: 6, SEQ ID NO: 7, SEQ ID NO: 8, SEQ ID NO: 9, SEQ ID NO: 10, SEQ ID NO: 11 or SEQ ID NO: 430. 94. The method of claim 92 or 93, wherein the mobile element enzyme has one or more mutations which confer hyperactivity. 95. The method of any one of claims 92-94, wherein the mobile element enzyme has an amino acid sequence of SEQ ID NO: 1, SEQ ID NO: 10, or SEQ ID NO: 11 or a functional equivalent thereof. 96. The method of one of claims 92-94, wherein the mobile element enzyme has the nucleotide sequence having at least about 90% identity to SEQ ID NO: 5 or a codon-optimized form thereof. 97. The method of claim 93, wherein the mobile element enzyme has an amino acid sequence having S8P and G17R mutations relative to the amino acid sequence of SEQ ID NO: 3 or SEQ ID NO: 4, or a functional equivalent thereof. 98. The method of claim 93, wherein the mobile element enzyme has an amino acid sequence having I83P and/or V118R mutation relative to the amino acid sequence of SEQ ID NO: 6 or a functional equivalent thereof. 99. The method of claim 93, wherein the mobile element enzyme has an amino acid sequence having S20P and/or A29R mutation relative to the amino acid sequence of SEQ ID NO: 7 or a functional equivalent thereof. 100. The method of claim 93, wherein the mobile element enzyme has an amino acid sequence having A12P and/or I28R mutation and/or R152K mutation relative to the amino acid sequence of SEQ ID NO: 9 or a functional equivalent thereof. 101. The method of claim 93, wherein the mobile element enzyme has an amino acid sequence having T4P and/or L13R mutation relative to the amino acid sequence of SEQ ID NO: 8 or a functional equivalent thereof. 102. The method of any one of claims 66-101, wherein the donor DNA is included in a vector comprising left and right end sequences recognized by the mobile element enzyme. 103. The method of claim 102, wherein the end sequences are selected from MER, MER75A, MER75B, and MER85. 104. The method of claim 103, wherein the end sequences are selected from nucleotide sequences of SEQ ID NO: 12, SEQ ID NO: 13, SEQ ID NO: 14, SEQ ID NO: 15, SEQ ID NO: 16, SEQ ID NO: 17, SEQ ID NO: 18, SEQ ID NO: 19, SEQ ID NO: 20, SEQ ID NO: 21, SEQ ID NO: 441, and SEQ ID NO: 22, or a nucleotide sequence having at least about 90% identity thereto. 105. The method of any one of claims 102-104, wherein one or more of the end sequences are optionally flanked by a TTAA (SEQ ID NO: 440) sequence.
106. The method of claim 104, wherein the end sequences include at least one repeat from a nucleotide sequence having at least about 90% identity to the nucleotide sequence of SEQ ID NO: 12, and wherein the at least one repeat from the nucleotide sequence having at least about 90% identity to the nucleotide sequence of SEQ ID NO: 12 is positioned at the 5’ end of the donor DNA. 107. The method of claim 104, wherein the end sequences include at least one repeat from a nucleotide sequence having at least about 90% identity to the nucleotide sequence of SEQ ID NO: 17, and wherein the at least one repeat from the nucleotide sequence having at least about 90% identity to the nucleotide sequence of SEQ ID NO: 17 is positioned at the 3’ end of the donor DNA. 108. The method of claim 106 or 107, wherein the end sequences are optionally flanked by a TTAA (SEQ ID NO: 440) sequence. 109. The method of claim 104, wherein the end sequences include at least one repeat from a nucleotide sequence having at least about 90% identity to the nucleotide sequence of SEQ ID NO: 13, and wherein the at least one repeat from the nucleotide sequence having at least about 90% identity to the nucleotide sequence of SEQ ID NO: 13 is positioned at the 5’ end of the donor DNA. 110. The method of claim 104, wherein the end sequences include at least one repeat from a nucleotide sequence having at least about 90% identity to the nucleotide sequence of SEQ ID NO: 18, and wherein the at least one repeat from the nucleotide sequence having at least about 90% identity to the nucleotide sequence of SEQ ID NO: 18 is positioned at the 3’ end of the donor DNA. 111. The method of claim 109 or 110, wherein the end sequences are optionally flanked by a TTAA (SEQ ID NO: 440) sequence. 112. The method of claim 104, wherein the end sequences include at least one repeat from a nucleotide sequence having at least about 90% identity to the nucleotide sequence of SEQ ID NO: 14, wherein the at least one repeat from the nucleotide sequence having at least about 90% identity to the nucleotide sequence of SEQ ID NO: 14 is positioned at the 5’ end of the donor DNA. 113. The method of claim 104, wherein the end sequences include at least one repeat from a nucleotide sequence having at least about 90% identity to the nucleotide sequence of SEQ ID NO: 19, wherein the at least one repeat from the nucleotide sequence having at least about 90% identity to the nucleotide sequence of SEQ ID NO: 19 is positioned at the 3’ end of the donor DNA. 114. The method of claim 112 or 113, wherein the end sequences are optionally flanked by a TTAA (SEQ ID NO: 440) sequence.
115. The method of claim 104, wherein the end sequences include at least one repeat from a nucleotide sequence having at least about 90% identity to the nucleotide sequence of SEQ ID NO: 15, wherein the at least one repeat from the nucleotide sequence having at least about 90% identity to the nucleotide sequence of SEQ ID NO: 15 is positioned at the 5’ end of the donor DNA. 116. The method of claim 104, wherein end sequences include at least one repeat from a nucleotide sequence having at least about 90% identity to the nucleotide sequence of SEQ ID NO: 20, wherein the at least one repeat from the nucleotide sequence having at least about 90% identity to the nucleotide sequence of SEQ ID NO: 20 is positioned at the 3’ end of the donor DNA. 117. The method of claim 115 or 116, wherein the end sequences are optionally flanked by a TTAA (SEQ ID NO: 440) sequence. 118. The method of claim 104, wherein the end sequences include at least one repeat from a nucleotide sequence having at least about 90% identity to the nucleotide sequence of SEQ ID NO: 16, wherein the at least one repeat from the nucleotide sequence having at least about 90% identity to the nucleotide sequence of SEQ ID NO: 16 is positioned at the 5’ end of the donor DNA. 119. The method of claim 104, wherein the end sequences include at least one repeat from a nucleotide sequence having at least about 90% identity to the nucleotide sequence of SEQ ID NO: 21 or SEQ ID NO: 441, wherein the at least one repeat from the nucleotide sequence having at least about 90% identity to the nucleotide sequence of SEQ ID NO: 21 or SEQ ID NO: 441 is positioned at the 3’ end of the donor DNA. 120. The method of any one of claims 104-119, wherein the mobile element enzyme is an engineered form of a mobile element enzyme reconstructed from Homo sapiens or a predecessor thereof. 121. The method of any one of claims 1-120, wherein the enzyme is in a monomeric or dimeric form. 122. The method of any one of claims 1-121, wherein the enzyme is in a multimeric form. 123. The method of any one of claims 1-122, wherein the enzyme comprises: (a) a targeting element, and (b) an enzyme that is capable of inserting a donor DNA (e.g. a mobile element) comprising a transgene, optionally at a TA dinucleotide site or a TTAA (SEQ ID NO: 440) tetranucleotide site in a genomic safe harbor site (GSHS). 124. The method of any one of claims 1-123, wherein the donor DNA comprises a transgene encoding a complete polypeptide.
125. The method of any one of claims 1-124, wherein the donor DNA comprises a transgene which is defective or substantially absent in a disease state. 126. The method of any one of claims 1-125, wherein the enzyme has one or more mutations which confer hyperactivity. 127. The method of any one of claims 1-126, wherein the enzyme has gene cleavage activity (Exc+) and/or gene integration activity (Int+). 128. The method of any one of claims 1-127, wherein the enzyme has gene cleavage activity (Exc+) and/or a lack of gene integration activity (Int-). 129. The method of any one of claims 1-128, wherein the mobile element enzyme is a chimeric mobile element enzyme. 130. The method of any one of claims 123-129, wherein the targeting element comprises one or more of a gRNA, optionally associated with a Cas enzyme, which is optionally catalytically inactive, transcription activator-like effector (TALE), catalytically inactive Zinc finger, catalytically inactive transcription factor, nickase, a transcriptional activator, a transcriptional repressor, a recombinase, a DNA methyltransferase, a histone methyltransferase, a paternally expressed gene 10 (PEG10), and a TnsD. 131. The method of any one of claims 123-130, wherein the targeting element comprises a transcription activator- like effector (TALE) DNA binding domain (DBD). 132. The method of claim 131, wherein the TALE DBD comprises one or more repeat sequences. 133. The method of claim 132, wherein the TALE DBD comprises about 14, or about 15, or about, 16, or about 17, or about 18, or about 18.5 repeat sequences. 134. The method of claim 132 or claim 133, wherein the TALE DBD repeat sequences comprise 33 or 34 amino acids. 135. The method of claim 131, wherein the one or more of the TALE DBD repeat sequences comprise a repeat variable di-residue (RVD) at residue 12 or 13 of the 33 or 34 amino acids. 136. The method of claim 135, wherein the RVD recognizes one base pair in the nucleic acid molecule. 137. The method of claim 135, wherein the RVD recognizes a C residue in the nucleic acid molecule and is selected from HD, N(gap), HA, ND, and HI. 138. The method of claim 135, wherein the RVD recognizes a G residue in the nucleic acid molecule and is selected from NN, NH, NK, HN, and NA.
139. The method of claim 135, wherein the RVD recognizes an A residue in the nucleic acid molecule and is selected from NI and NS. 140. The method of claim 135, wherein the RVD recognizes a T residue in the nucleic acid molecule and is selected from NG, HG, H(gap), and IG. 141. The method of any one of claims 123-140, wherein the GSHS is in an open chromatin location in a chromosome. 142. The method of any one of claims 123-140, wherein the GSHS is selected from adeno-associated virus site 1 (AAVS1), chemokine (C-C motif) receptor 5 (CCR5) gene, HIV-1 coreceptor, and human Rosa26 locus. 143. The method of any one of claims 123-140, wherein the GSHS is located on human chromosome 2, 4, 6, 10, 11, 17, 22, or X. 144. The method of any one of claims 123-140, wherein the GSHS is selected from TALC1, TALC2, TALC3, TALC4, TALC5, TALC7, TALC8, AVS1, AVS2, AVS3, ROSA1, ROSA2, TALER1, TALER2, TALER3, TALER4, TALER5, SHCHR2-1, SHCHR2-2, SHCHR2-3, SHCHR2-4, SHCHR4-1, SHCHR4-2, SHCHR4-3, SHCHR6-1, SHCHR6-2, SHCHR6-3, SHCHR6-4, SHCHR10-1, SHCHR10-2, SHCHR10-3, SHCHR10-4, SHCHR10-5, SHCHR11-1, SHCHR11-2, SHCHR11-3, SHCHR17-1, SHCHR17-2, SHCHR17-3, and SHCHR17-4. 145. The method of any one of claims 123-130, wherein the targeting element comprises a Cas9 enzyme guide RNA complex. 146. The method of claim 145, wherein the Cas9 enzyme guide RNA complex comprises a nuclease-deficient dCas9 guide RNA complex. 147. The method of any one of claims 123-130, wherein the targeting element comprises a Cas12 enzyme guide RNA complex or wherein the targeting element comprises a nuclease-deficient dCas12 guide RNA complex, optionally dCas12j guide RNA complex or dCas12a guide RNA complex. 148. The method of any one of claims 123-147, wherein the targeting element comprises: a gRNA of or comprising a sequence of TABLE 3A-3F, or a variant thereof; or a TALE DBD of or comprising a sequence of TABLE 4A-4F, or a variant thereof; or a ZNF of or comprising a sequence of TABLE 5A-5E, or a variant thereof. 149. The method of any one of claims 123-130, wherein the targeting element is or comprises a nucleic acid binding component of the gene-editing system. 150. The method of any one of claims 123-149, wherein the enzyme and the targeting element are connected.
151. The method of any one of claims 123-149, wherein the enzyme and the targeting element are fused to one another or linked via a linker to one another. 152. The method of claim 151, wherein the linker is a flexible linker. 153. The method of claim 152, wherein the flexible linker is substantially comprised of glycine and serine residues, optionally wherein the flexible linker comprises (Gly4Ser)n, where n is from about 1 to about 12. 154. The method of claim 152, wherein the flexible linker is of about 20, or about 30, or about 40, or about 50, or about 60 amino acid residues. 155. The method of any one of claims 1-154, wherein the nucleic acid comprises a gene encoding a complete polypeptide. 156. The method of any one of claims 1-155, wherein the nucleic acid comprises a gene which is defective or substantially absent in a disease state. 157. The method of any one of claims 1-156, wherein the transgene is flanked by one or more inverted terminal ends. 158. The method of any one of claims 1-157, wherein at least one of the first nucleic acid and the second nucleic acid is in the form of a lipid nanoparticle (LNP). 159. The method of any one of claims 1-158, wherein the first nucleic acid encoding the enzyme and the second nucleic acid are in the form of the same LNP, optionally in a co-formulation. 160. The method of claim 158 or claim 159, wherein the LNP comprises one or more lipids selected from 1,2- dioleoyl-3-trimethylammonium propane (DOTAP), a cationic cholesterol derivative mixed with dimethylaminoethane- carbamoyl (DC-Chol), phosphatidylcholine (PC), triolein (glyceryl trioleate), and 1,2-distearoyl-sn-glycero-3- phosphoethanolamine-N-[carboxy(polyethylene glycol)-2000] (DSPE-PEG), 1,2-dimyristoyl-rac-glycero-3- methoxypolyethyleneglycol – 2000 (DMG-PEG 2K), and 1,2 distearol -sn-glycerol-3phosphocholine (DSPC) and/or comprising of one or more molecules selected from polyethylenimine (PEI) and poly(lactic-co-glycolic acid) (PLGA), and N-Acetylgalactosamine (GalNAc). 161. The method of any one of claims 1-160, wherein the enzyme is encoded by a recombinant or synthetic nucleic acid. 162. The method of claim 161, wherein the nucleic acid is mRNA or a helper RNA. 163. The method of claim 162, wherein the nucleic acid is RNA that has a 5’-m7G cap (cap0, cap1, or cap2) with pseudouridinesubstitution, and a poly-A tail of about 30, or about 50, or about 100, of about 150 nucleotides in length.
164. The method of claim 162, wherein the enzyme is incorporated into a vector or a vector-like particle. 165. The method of claim 164, wherein the vector is a non-viral vector. 166. The method of any of claims 1-165, wherein the enzyme and the one or more elements (a)-(g) are included in the same vector. 167. The method of any of claims 1-166, wherein the enzyme and the one or more elements (a)-(g) are included in different vectors. 168. The method of any one of claims 1-167, wherein the enzyme and the one or more elements (a)-(g) are included in a single pharmaceutical compositions. 169. The method of any one of claims 1-168, wherein the enzyme and the one or more elements (a)-(g) are included in different pharmaceutical compositions. 170. The method of any one of claims 1-169, wherein the enzyme and the one or more elements (a)-(g) are co- administered. 171. The method of any one of claims 1-170, wherein the enzyme and the one or more elements (a)-(g) are administered separately. 172. The method of any one of claims 1-171, wherein the viral particle packaging and producer cell line provides a stable cell with substantially reduced or ablated CAP expression. 173. The method of any one of claims 1-171, wherein the viral particle packaging and producer cell line provides a stable cell with REP expression or substantially enhanced REP expression. 174. The method of any one of claims 1-171, wherein the viral particle packaging and producer cell line provides a stable cell with substantially reduced or ablated CAP expression and REP expression or substantially enhanced REP expression. 175. The method of any one of claims 1-171, wherein the viral particle packaging and producer cell line provides a stable cell with REP expression or substantially enhanced VP1 expression. 176. The method of any one of claims 1-171, wherein the viral particle packaging and producer cell line provides a stable cell with substantially reduced or ablated CAP expression. 177. The method of any one of claims 1-171, wherein the viral particle packaging and producer cell line is suitable for providing substantially reduced empty or cargo-free capsid. 178. A method of producing an AAV bearing a gene of interest, comprising employing a method of any one of claims 1-177, to produce the AAV bearing the gene of interest.
179. A cell for gene therapy, generated by a method of any one of claims 1-178.
180. A method of delivering a cell therapy, comprising administering to a patient in need thereof the transfected cell generated by a method of any one of claims 1-179.
181. A method of treating a disease or condition using a cell therapy, comprising administering to a patient in need thereof the transfected cell generated by a method of any one of claims 1-180.
182. A method of treating a disease or condition using a biologic, e.g., antibody, therapy, comprising administering to a patient in need thereof the transfected cell generated by a method of any one of claims 1-181.
PCT/US2023/078957 2022-11-07 2023-11-07 Adeno-associated virus (aav) production WO2024102739A2 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US202263423220P 2022-11-07 2022-11-07
US63/423,220 2022-11-07

Publications (1)

Publication Number Publication Date
WO2024102739A2 true WO2024102739A2 (en) 2024-05-16

Family

ID=91033437

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2023/078957 WO2024102739A2 (en) 2022-11-07 2023-11-07 Adeno-associated virus (aav) production

Country Status (1)

Country Link
WO (1) WO2024102739A2 (en)

Similar Documents

Publication Publication Date Title
JP2024003220A (en) Gene editing using modified closed-ended DNA (CEDNA)
US20210062161A1 (en) Methods for Adeno-Associated Viral Vector Production
KR102526711B1 (en) Adeno-associated virus vector variants for high efficiency genome editing and methods thereof
CN111868242A (en) Closed-ended DNA vectors obtainable from cell-free synthesis and method for obtaining a ceDNA vector
KR20180091863A (en) Extensible method for producing recombinant adeno-associated virus (AAV) vectors in serum-free suspension cell culture systems suitable for clinical use
EP3759217A1 (en) Closed-ended dna (cedna) vectors for insertion of transgenes at genomic safe harbors (gsh) in humans and murine genomes
JPH09509578A (en) Integrable recombinant adenoviruses, their production and their therapeutic use
MX2012011374A (en) Pharmacologically induced transgene ablation system.
EP3310369B1 (en) Self-limiting viral vectors encoding nucleases
CN112639110A (en) Vectors for gene delivery to persist in cells
CN113316640A (en) Modified closed-end DNA (CEDNA) comprising a symmetrical modified inverted terminal repeat
CN113454232A (en) Modulation of REP protein activity in closed end dna (cedna) production
US6806080B2 (en) Hybrid vectors for gene therapy
CN113874508A (en) Non-viral DNA vectors and their use for expressing phenylalanine hydroxylase (PAH) therapeutics
US20210108197A1 (en) Shrna expression cassette, polynucleotide sequence carrying same, and application thereof
CN113874513A (en) Non-viral DNA vectors and their use for expressing FVIII therapeutics
US20030190746A1 (en) Gene expression control system and its use in recombinant virus packaging cell lines
WO2024102739A2 (en) Adeno-associated virus (aav) production
CN117858891A (en) Modified AAV capsids for gene therapy and methods thereof
Hacobian et al. Pushing the right buttons: Improving efficacy of therapeutic DNA vectors
US20220380812A1 (en) Crispr/cas9 system as an agent for inhibition of polyoma jc infection
WO2023240162A1 (en) Aav vectors for gene editing
WO2023081816A2 (en) Transposable mobile elements with enhanced genomic site selection
CA3236684A1 (en) Mobile elements and chimeric constructs thereof
WO2023220386A1 (en) Adeno-associated viral vectors for targeting brain microvasculature