WO2023141602A2 - Engineered retrons and methods of use - Google Patents

Engineered retrons and methods of use Download PDF

Info

Publication number
WO2023141602A2
WO2023141602A2 PCT/US2023/061038 US2023061038W WO2023141602A2 WO 2023141602 A2 WO2023141602 A2 WO 2023141602A2 US 2023061038 W US2023061038 W US 2023061038W WO 2023141602 A2 WO2023141602 A2 WO 2023141602A2
Authority
WO
WIPO (PCT)
Prior art keywords
retron
ncrna
sequence
engineered
nucleic acid
Prior art date
Application number
PCT/US2023/061038
Other languages
French (fr)
Other versions
WO2023141602A3 (en
Inventor
Alim Ladha
Vladimir PRESNYAK
Inna SHCHERBAKOVA
Brian Goodman
Mario Rodriguez MESTRE
Kyong-Rim KIEFFER-KWON
Socheata LY
Devin Scott QUINLAN
Muthusamy Jayaraman
Stephen Scully
Jacob LAYER
Original Assignee
Renagade Therapeutics Management 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 Renagade Therapeutics Management Inc. filed Critical Renagade Therapeutics Management Inc.
Publication of WO2023141602A2 publication Critical patent/WO2023141602A2/en
Priority to PCT/US2023/072872 priority Critical patent/WO2024044723A1/en
Publication of WO2023141602A3 publication Critical patent/WO2023141602A3/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/10Processes for the isolation, preparation or purification of DNA or RNA
    • C12N15/102Mutagenizing nucleic acids
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/87Introduction of foreign genetic material using processes not otherwise provided for, e.g. co-transformation
    • C12N15/90Stable introduction of foreign DNA into chromosome
    • C12N15/902Stable introduction of foreign DNA into chromosome using homologous recombination
    • C12N15/907Stable introduction of foreign DNA into chromosome using homologous recombination in mammalian cells
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N9/00Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
    • C12N9/10Transferases (2.)
    • C12N9/12Transferases (2.) transferring phosphorus containing groups, e.g. kinases (2.7)
    • C12N9/1241Nucleotidyltransferases (2.7.7)
    • C12N9/1276RNA-directed DNA polymerase (2.7.7.49), i.e. reverse transcriptase or telomerase
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N9/00Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
    • C12N9/14Hydrolases (3)
    • C12N9/16Hydrolases (3) acting on ester bonds (3.1)
    • C12N9/22Ribonucleases RNAses, DNAses
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12YENZYMES
    • C12Y207/00Transferases transferring phosphorus-containing groups (2.7)
    • C12Y207/07Nucleotidyltransferases (2.7.7)
    • C12Y207/07049RNA-directed DNA polymerase (2.7.7.49), i.e. telomerase or reverse-transcriptase
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/10Type of nucleic acid
    • C12N2310/20Type of nucleic acid involving clustered regularly interspaced short palindromic repeats [CRISPRs]
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/30Chemical structure
    • C12N2310/35Nature of the modification
    • C12N2310/351Conjugate
    • C12N2310/3519Fusion with another nucleic acid
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2740/00Reverse transcribing RNA viruses
    • C12N2740/00011Details
    • C12N2740/10011Retroviridae
    • C12N2740/10022New viral proteins or individual genes, new structural or functional aspects of known viral proteins or genes

Definitions

  • SEQUENCE LISTING [0003] This application contains a sequence listing filed in electronic form in eXtensible Markup Language (XML) format entitled RTX006_Sequence_Listing.xml, created on January 20, 2023 and having a size of 32,261,680 bytes. The content of the sequence listing is incorporated herein in its entirety.
  • TECHNICAL FIELD [0004] The present disclosure generally relates to systems, methods and compositions used for precise genome editing, including nucleic acid insertions, replacements, and deletions at targeted and precise genome sites, wherein said systems, methods, and compositions are based on novel and/or engineered retrons.
  • RNA-guided nucleases e.g., CRISPR nucleases
  • ZFN zinc-finger nucleases
  • TALENS transcription activator-like effector nucleases
  • HDR-dependent precise genome editing is the delivery of donor DNA template to the nuclease-induced DSB (e.g., see Ling et al., “Improving the efficiency of precise genome editing with site-specific Cas9-oligonucleotide conjugates,” Science Advances, 2020, Vol.6, No.15, pp.1-8).
  • Various methods aimed at boosting the efficiency of HDR-dependent editing have been reported, many of which involve the physical tethering of the DNA donor to a component of the precise editing system. Exemplary methods have been discussed in: K.
  • Retrons are defined by their unique ability to produce an unusual satellite DNA known as msDNA (multicopy single-stranded DNA).
  • DNA encoding retrons includes a reverse trancriptase (RT)-coding gene (ret) and a nucleic acid sequence encoding the non-coding RNA (ncRNA), which contains two contiguous and inverted non-coding sequences referred to as the msr and msd.
  • RT reverse trancriptase
  • ncRNA nucleic acid sequence encoding the non-coding RNA
  • the ret gene and the non-coding RNA are transcribed as a single RNA transcript, which becomes folded into a specific secondary structure following post-transcriptional processing.
  • the RT binds the RNA template downstream from the msd locus, initiating reverse transcription of the RNA towards its 5 ⁇ end, assisted by the 2’OH group present in a conserved branching guanosine residue that acts as a primer.
  • Reverse transcription halts before reaching the msr locus, and the resulting DNA, the msDNA, remains covalently attached to the RNA template via a 2’-5 ⁇ phosphodiester bond and base-pairing between the 3 ⁇ ends of the msDNA and the RNA template.
  • the external regions, at the 5 ⁇ and 3 ⁇ ends of the msd/msr transcript (a1 and a2, respectively) are complementary and can hybridize, leaving the structures located in the msr and msd regions in internal positions (see FIG.1A).
  • the msr locus which is not reverse transcribed, forms one to three short stem-loops of variable size, ranging from 3 to 10 base pairs, whereas the msd locus folds into a single/double long hairpin with a highly variable long stem of 10-50 bp in length that is also present in the final msDNA form.
  • retrons may be utilized as a means to provide donor DNA template for HDR-dependent genome editing (e.g., see Lopez et al., “Precise genome editing across kingdoms of life using retron-derived DNA,” Nature Chemical Biology, December 12, 2021, 18, pages199–206 (2022)), however, producing sufficient levels of donor DNA template intracellularly to sufficiently support efficient HDR-dependent editing remains a significant challenge.
  • Improved retron-based genome modification systems are highly desirous in the art.
  • the present disclosure provides recombinant retrons comprising one or more genetic modifications which improves the functionality and/or properties of a retron.
  • Such genetic modifications can include a mutation, insertion, deletion, inversion, replacement, substitution, or translocation of one or more contiguous or non-contiguous nucleobases in a nucleic acid molecule encoding a retron or a component of a retron, such as an ncRNA or a reverse transcriptase.
  • the retron that becomes modifed with the one or more genetic modifications is a naturally occurring retron or retron component (e.g., naturally occuring ncRNA of Table A or RT) ability to facilitate homology-dependent recombination (or HDR) in a cell, thereby resulting in a relative increase in the concentrations or amounts of msDNA comprising a DNA donor template.
  • a naturally occurring retron or retron component e.g., naturally occuring ncRNA of Table A or RT
  • HDR homology-dependent recombination
  • the recombinant retrons are based on and/or derived from a naturally-occurring retron, such as any retron-related sequence provided by Table X (the introduction of the one or more genetic modifications into a set of 7257 previously unknown retrons discovered through computational methods described herein (e.g., see Examples).
  • the recombinant retrons are based on introducing the one or more genetic modifications into previously available retron sequences (e.g., the “Mestre et al., Systematic Prediction of Genes Functionally Associated with Bacterial Retrons and Classification of The Encoded Tripartite Systems, Nucleic Acids Research, Volume 48, Issue 22, 16 December 2020, Pages 12632-12647” (incorporated herein by reference) to achieve recombinant retrons with the enhanced ability to produce increased concentrations or amounts of msDNA comprising a DNA donor template.
  • retron sequences e.g., the “Mestre et al., Systematic Prediction of Genes Functionally Associated with Bacterial Retrons and Classification of The Encoded Tripartite Systems, Nucleic Acids Research, Volume 48, Issue 22, 16 December 2020, Pages 12632-12647” (incorporated herein by reference) to achieve recombinant retrons with the enhanced ability to produce increased concentrations or amounts of msDNA
  • the present disclosure further provides nucleic acid molecules encoding the recombinant retrons and/or recombinant retron components (e.g., a recombinant ncRNA and/or a recombinant retron RT).
  • the present disclosure provides genome editing systems comprising recombinant retron components (e.g., recombinant ncRNA and/or recombinant RT), programmable nucleases (e.g., RNA-guided nucleases, such as CRISPR-Cas proteins, ZFPs, and TALENS), and guide RNAs (in the case where RNA-guide nucleases are used in said genome editing systems).
  • recombinant retron components e.g., recombinant ncRNA and/or recombinant RT
  • programmable nucleases e.g., RNA-guided nucleases, such as CRISPR-Cas proteins, ZFPs,
  • the disclosure provides nucleic acid molecules encoding the described genome editing systems and said components thereof, as well as polypeptides making up the components of said genome editing systems.
  • the disclosure provides vectors for transferring and/or expressing said genome editing systems, e.g., under in vitro, ex vivo, and in vivo conditions.
  • the disclosure provides cell-delivery compositions and methods, including compositions for passive and/or active transport to cells (e.g., plasmids), delivery by virus-based recombinant vectors (e.g., AAV and/or lentivirus vectors), delivery by non-virus-based systems (e.g., liposomes and LNPs), and delivery by virus-like particles.
  • the retron-based genome editing systems described herein may be delivered in the form of DNA (e.g., plasmids or DNA-based virus vectors), RNA (e.g., ncRNA and mRNA delivered by LNPs), a mixture of DNA and RNA, protein (e.g., virus-like particles), and ribonucleoprotein (RNP) complexes.
  • DNA e.g., plasmids or DNA-based virus vectors
  • RNA e.g., ncRNA and mRNA delivered by LNPs
  • protein e.g., virus-like particles
  • RNP ribonucleoprotein
  • each of the components of the retron-based genome editing system is delivered by an all-RNA system, e.g., the delivery of one or more RNA molecules (e.g., mRNA and/or ncRNA) by one or more LNPs, wherein the one or more RNA molecules form the ncRNA and guide RNA (as needed) and/or are translated into the polypeptide components (e.g., the RT and a programmable nuclease).
  • an all-RNA system e.g., the delivery of one or more RNA molecules (e.g., mRNA and/or ncRNA) by one or more LNPs, wherein the one or more RNA molecules form the ncRNA and guide RNA (as needed) and/or are translated into the polypeptide components (e.g., the RT and a programmable nuclease).
  • the disclosure provides methods for genome editing by introducing a retron-based genome editing system described herein into a cell (e.g., under in vitro, in vivo, or ex vivo conditions) comprising a target edit site, thereby resulting in an edit at the target edit.
  • a retron-based genome editing system described herein into a cell (e.g., under in vitro, in vivo, or ex vivo conditions) comprising a target edit site, thereby resulting in an edit at the target edit.
  • the disclosure provides formulations comprising any of the aforementioned components for delivery to cells and/or tissues, including in vitro, in vivo, and ex vivo delivery, recombinant cells and/or tissues modified by the recombinant retron-based genome modification systems and methods described herein, and methods of modifying cells by conducting genome editing and related DNA donor-dependent methods, such as recombineering, or cell recording, using the herein disclosed retron-based genome modification systems.
  • the disclosure also provides methods of making the recombinant retrons, retron-based genome modification systems, vectors, compositions and formulations described herein, as well as to pharmaceutical compositions and kits for modifying cells under in vitro, in vivo, and ex vivo conditions that comprise the herein disclosed genome editing and/or modification systems.
  • this disclosure or the inventions herein provide a gene editing system comprising one or more delivery vehicles, wherein: the delivery vehicle(s) comprise RNA cargo; the RNA cargo comprises (a) at least one mRNA molecule encoding (i) a nucleic acid programmable nuclease and (ii) a retron reverse transcriptase, (b) an engineered retron ncRNA, and (c) guide RNA for the programmable nuclease; and each delivery vehicle contains (a)(i) and/or (a)(ii) and/or (b) and/or (c); whereby one delivery vehicle or more than one delivery vehicle delivers (a)(i), (a)(ii), (b), and (c).
  • the delivery vehicle(s) comprise RNA cargo
  • the RNA cargo comprises (a) at least one mRNA molecule encoding (i) a nucleic acid programmable nuclease and (ii) a retron reverse transcriptase, (b) an engineered
  • (a)(i) and (a)(ii) comprise a single mRNA molecule encoding the nucleic acid programmable nuclease and the retron reverse transcriptase.
  • (a)(i) and (a)(ii) are encoded and expressed as a fusion protein.
  • the fusion protein comprises the C-terminal end of the nucleic acid programmable nuclease fused to the N-terminal end of the retron reverse transcriptase (nuclease:RT fusion); or the fusion protein comprises the N-terminal end of the nucleic acid programmable nuclease fused to the C-terminal end of the retron reverse transcriptase (RT:nuclease fusion).
  • (a)(i) and (a)(ii) comprise a first mRNA molecule encoding the nucleic acid programmable nuclease and a second mRNA molecule encoding the retron reverse transcriptase.
  • (c) is separate from (a)(i), (a)(ii) and (b) or is provided in trans.
  • the engineered retron ncRNA, and (c) the guide RNA are fused or are provided in cis.
  • the engineered retron ncRNA, and (c) the guide RNA are fused or are provided in cis and the guide RNA is fused to the 5’ end of the retron ncRNA.
  • the engineered retron ncRNA, and (c) the guide RNA are fused or are provided in cis and the guide RNA is fused to the 3’ end of the retron ncRNA.
  • the engineered retron ncRNA, and (c) the guide RNA are fused or are provided in cis and the engineered ncRNA comprises a first guide RNA fused to the 5’ end of the retron ncRNA, and a second guide RNA fused to the 3’ end of the retron ncRNA, and the first and second guide RNAs target different sequences.
  • guide RNA for the programmable nuclease can comprise one or more guides that target the same or different target sequences.
  • Such guide RNA(s) in an embodiment can be single guide RNA(s) or sgRNA(s); for instance, when the nucleic acid programmable nuclease comprises a Cas9.
  • the one or more delivery vehicles comprise a liposome or a lipid nanoparticle (LNP).
  • LNP lipid nanoparticle
  • the at least one mRNA molecule encoding (i) the nucleic acid programmable nuclease and (ii) the retron reverse transcriptase, and (b) the engineered retron ncRNA are in separate delivery vehicles.
  • the nucleic acid programmable nuclease and the retron reverse transcriptase are encoded on separate mRNA molecules and those separate mRNA molecules of (a)(i) and (a)(ii) are contained in the same delivery vehicle.
  • the nucleic acid programmable nuclease and the retron reverse transcriptase are encoded on separate mRNA molecules and those separate mRNA molecules of (a)(i) and (a)(ii) are contained in different delivery vehicles.
  • the engineered retron ncRNA includes a sequence of interest encoding a donor polynucleotide comprising an intended edit to be integrated at a target sequence in a cell, and wherein the donor polynucleotide is flanked by a 5' homology arm that hybridizes to a sequence 5’ to the target sequence and a 3' homology arm that hybridizes to a sequence 3' to the target sequence.
  • the donor polynucleotide can be heterologous to the cell.
  • the donor polynucleotide can be endogenous to the cell; for instance, the cell can contain a sequence that is typical for those in a population having a disease state and the donor polynucleotide can be a sequence that is typical for those in the population not having a non-disease state (e.g., the donor can be for a genetic correction or repair of a cell to modify the cell from having a mutation or modification that gives rise to a disease state to having a sequence typical of not having the disease state).
  • Such can be done in an animal cell, or a mammalian cell (e.g., a primate, a non-human primate, or a domesticated mammal such as a cat or dog or horse) or a human cell; for instance to correct, address, treat, mitigate a genetic condition in the animal, mammal, domesticated mammal, cat, dog, horse or human.
  • a mammalian cell e.g., a primate, a non-human primate, or a domesticated mammal such as a cat or dog or horse
  • a human cell for instance to correct, address, treat, mitigate a genetic condition in the animal, mammal, domesticated mammal, cat, dog, horse or human.
  • plant cells to introduce mutations that give rise to favorable phenotypic characteristics such as disease resistance or other favorable plant trait(s).
  • the nucleic acid programmable nuclease comprises a Cas9 nuclease, a TnpB nuclease, or a Cas12a nuclease.
  • the engineered retron ncRNA comprises: A) a pre-msr sequence having a first complementary region of the retron ncRNA; B) an msr sequence including an msr stem-loop structure; C) an msd sequence including an msd stem-loop structure and a sequence of interest, wherein said msd sequence templates a single strand DNA product (RT-DNA) in the presence of the retron reverse transcriptase; and D) a post- msd sequence having a second complementary region, wherein the first and second complementary regions form an a1/a2 duplex region of the retron ncRNA, wherein the msr stem- loop structure, the msd stem-loop structure, or the a1/a2 duplex comprise a modification which result in increased editing efficiency in the presence of a nucleic acid programmable nuclease that associates with the one or more guide RNAs, and wherein optionally one
  • the engineered retron ncRNA comprises A), B), C) and D
  • the sequence of interest can encode a donor polynucleotide comprising an intended edit to be integrated at a target sequence of a cell, wherein the donor polynucleotide is flanked by a 5' homology arm that hybridizes to a sequence 5’ to the target sequence and a 3' homology arm that hybridizes to a sequence 3' to the target sequence.
  • the engineered retron ncRNA comprises A), B), C) and D) (either with the sequence of interest encoding a donor polynucleotide or simply being a sequence of interest)
  • the ncRNA has a nucleotide sequence of Table B, or a nucleotide sequence having at least 75%, 80%, 85%, 90%, 95%, 99%, or 100% sequence identity with a sequence from Table B.
  • the donor polynucleotide can be heterologous to a cell. Alternatively, the donor polynucleotide can be endogenous to the cell.
  • the cell can contain a sequence that is typical for those in a population having a disease state and the donor polynucleotide can be a sequence that is typical for those in the population not having a non-disease state (e.g., the donor can be for a genetic correction or repair of a cell to modify the cell from having a mutation or modification that gives rise to a disease state to having a sequence typical of not having the disease state).
  • the gene editing system can comprise any combination(s) of the foregoing embodiments of the gene editing system.
  • this disclosure or the inventions herein provide a cell, such as an isolated cell comprising the gene editing system disclosed herein, such as in any of the foregoing paragraphs.
  • the cell e.g., isolated cell
  • the cell can be a eukaryotic cell.
  • the eukaryotic cell can be a plant cell or an animal cell or a mammalian cell, e.g., an isolated plant cell or an isolated animal cell or an isolated mammalian cell.
  • the mammalian cell e.g., an isolated mammalian cell
  • the cell can be a prokaryotic cell, e.g., a bacterial cell.
  • the donor polynucleotide can code for antibiotic susceptibility; and thus, the invention can involve a means for addressing antibiotic resistant bacteria by rendering such bacteria susceptible to antibiotics (and a subject to whom the gene editing system is administered can also then receive antibiotics to which the bacteria are rendered susceptible by the gene editing system).
  • this disclosure or the inventions herein provide a composition comprising: a) the gene editing system disclosed herein, such as in any of the foregoing paragraphs; and b)a pharmaceutically or veterinarily acceptable carrier.
  • the delivery vehicle in the composition can comprise a lipid nanoparticle comprising: a) one or more ionizable lipids; b) one or more structural lipids; c) one or more PEGylated lipids; and d)one or more phospholipids.
  • the one or more ionizable lipids comprises an ionizable lipid set forth in Table 2.
  • this disclosure or the inventions herein provide uses of the gene editing system embodiments and/or the compositions disclosed herein, such as in any of the foregoing paragraphs; for instance, use in modifying a cell or genetically modifying a cell, e.g., a eukaryotic or a prokaryotic cell and/or an animal cell and/or a mammalian and/or a human cell and/or a bacterial cell and/or a plant cell, in vivo, in vitro or ex vivo (e.g., any cell discussed herein wherein the cell comprises an isolated cell).
  • a cell or genetically modifying a cell e.g., a eukaryotic or a prokaryotic cell and/or an animal cell and/or a mammalian and/or a human cell and/or a bacterial cell and/or a plant cell, in vivo, in vitro or ex vivo (e.g., any cell discussed herein wherein the cell comprises an isolated cell
  • this disclosure or the inventions herein provide uses of the gene editing system embodiments and/or the compositions disclosed herein, such as in any of the foregoing paragraphs; for instance, use in treating or addressing a genetic condition of a subject, [0032]
  • this disclosure or the inventions herein provide methods of genetically modifying a cell comprising: contacting a gene editing system as herein discussed, such as in any of the foregoing paragraphs, or a composition as herein discussed, such as in any of the foregoing paragraphs (which comprises a gene editing system as herein discussed, such as in any of the foregoing paragraphs), advantageously a gene editing system that includes a sequence of interest encoding a donor polynucleotide comprising an intended edit to be integrated at a target sequence in a cell, said method comprising contacting the composition or the gene editing system with the cell, thereby delivering the RNA cargo to the cell, wherein: the nucleic acid programmable nuclease forms a complex with the guide
  • the cell can be a eukaryotic or a prokaryotic cell or an animal cell or a mammalian cell or a human cell or a bacterial cell or a plant cell.
  • An engineered nucleic acid construct comprising: a) a first polynucleotide encoding a non-coding RNA (ncRNA), said first polynucleotide comprising: 1) an msr locus encoding the msr RNA portion of a multi-copy single- stranded DNA (msDNA); and 2) an msd locus encoding the msd RNA portion of the msDNA; and b) one or more heterologous nucleic acids inserted at or within a location selected from: the msd locus, upstream of the msr locus, upstream of the msd locus, and downstream of the msd locus, wherein the ncRNA comprises: (I) an ncRNA listed in Table B, or an ncRNA having at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 9
  • the engineered nucleic acid construct of paragraph 2, wherein the second polynucleotide comprises: III) a polynucleotide listed in Table A, or a polynucleotide having at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%%, at least 99.1%, at least 99.2%, at least 99.3%, at least 99.4%, at least 99.5%, at least 99.6%, at least 99.7%, at least 99.8% or at least 99.9% sequence identity to a polynucleotide listed in Table A; and/or IV) encodes a consensus amino acid sequence of Table C, or encodes an amino acid sequence having at least 50%, at least 55%, at least 60%, at least 65%, at least 70%
  • An engineered nucleic acid construct comprising: a) a first polynucleotide encoding a non-coding RNA (ncRNA), said first polynucleotide comprising: 1) an msr locus encoding the msr RNA portion of a multi-copy single- stranded DNA (msDNA); and 2) an msd locus encoding the msd RNA portion of the msDNA; b) one or more heterologous nucleic acids inserted at or within a location selected from: the msd locus, upstream of the msr locus, upstream of the msd locus, and downstream of the msd locus; and c) a second polynucleotide encoding a reverse transcriptase (RT), or a portion thereof, wherein the encoded RT or portion thereof is capable of synthesizing a DNA copy of at least a portion of the msd locus encoding the m
  • An engineered nucleic acid construct comprising: 1) an msr locus (that encodes the msr RNA portion of an msDNA); 2) an msd locus encoding the msd RNA portion of the msDNA; 3) a sequence encoding a retron reverse transcriptase (RT), wherein said msd RNA is capable of being reverse transcribed to form the msDNA by the retron reverse transcriptase (RT); and, 4) a heterologous nucleic acid inserted at or within the msd locus, upstream of the msr locus, upstream or downstream of the msd locus; wherein the engineered nucleic acid construct optionally has (a) a secondary structure of a wild-type ncRNA of any one of FIGs.2-27 or b) a variant of a), having: i) up to 1, 2, or 3 (e.g., up to 1) nucleotide changes per 10 red lettere
  • the engineered nucleic acid construct of any one of paragraphs 1 to 4a comprising one or more sequence modifications (e.g., an insertion, deletion, and/or substitution of one or more nucleotide(s)) in the msr locus and/or the msd locus that: a) modulates (e.g., enhances) reverse transcription, processivity, accuracy/fidelity, and/or production of the msDNA (e.g., in the mammalian cell); b) modulates (e.g., reduces) immunogenicity of ncRNA encoded by the engineered retron (e.g., the msr locus and/or the msd locus) in a host (e.g., a host comprising the mammalian cell); c) modulates (e.g., inhibits, either permanently or transiently) a function of the msDNA; and/or d) modulates (e.g., improves) efficiency of targeted genome editing /
  • the one or more heterologous nucleic acid sequences comprise: a) a heterologous nucleic acid (such as the coding sequence for an RNA aptamer or a ribozyme) inserted into the msr locus or the msd locus (such as in an S region (e.g., S1, S2, S3 and/or S4), or the tip of an L region (e.g., L1, L2, L3 and/or L4), or upstream or downstream of either the msr locus or the msd locus; or b) a first heterologous nucleic acid inserted into the msd locus, and a second heterologous nucleic acid inserted either upstream of the msr locus or downstream of the msd locus, wherein the second heterologous nucleic acid encodes a guide RNA.
  • a heterologous nucleic acid such as the coding sequence for an RNA aptamer or a ribozyme
  • said protein or peptide of interest comprises a therapeutic protein useful in treating a disease.
  • a sequence-specific nuclease such as a CRISPR/Cas effector enzyme, a ZFN, a TALEN, a meganuclease, TnpB, IscB, or a restriction endonuclease (RE)
  • a sequence-specific nuclease such as a CRISPR/Cas effector enzyme, a ZFN,
  • the sequence-specific nuclease is fused to the RT, optionally via a flexible linker (e.g., a flexible linker comprising Gly and Ser rich sequences such as G4S repeats or GS repeats) or by a generally disordered protein sequence (such as unstructured hydrophilic, biodegradable protein polymer, e.g., an XTEN peptide polymer).
  • a flexible linker e.g., a flexible linker comprising Gly and Ser rich sequences such as G4S repeats or GS repeats
  • a generally disordered protein sequence such as unstructured hydrophilic, biodegradable protein polymer, e.g., an XTEN peptide polymer.
  • nuclease is a CRISPR/Cas effector enzyme that forms a complex with a guide RNA (gRNA) recognizing a target sequence, wherein the gRNA is linked to the ncRNA and/or the msDNA, either directly or through a linker / spacer polynucleotide.
  • gRNA guide RNA
  • the DNA-repair modulating biomolecule is a regulatory protein that modulates (e.g., enhances) HDR, and the regulatory protein is fused to the RT or to the sequence-specific nuclease, optionally via a flexible linker (e.g., the flexible linker comprising Gly and Ser rich sequences such as G4S repeats or GS repeats) or by a generally disordered protein sequence (such as unstructured hydrophilic, biodegradable protein polymer, e.g., an XTEN peptide polymer). 17.
  • a flexible linker e.g., the flexible linker comprising Gly and Ser rich sequences such as G4S repeats or GS repeats
  • a generally disordered protein sequence such as unstructured hydrophilic, biodegradable protein polymer, e.g., an XTEN peptide polymer.
  • a vector system comprising one or more vectors comprising the engineered nucleic acid construct of any one of paragraphs 1-16, wherein the vector system is optionally all- RNA.
  • the vector system of paragraph 17 wherein the msr locus, the msd locus, and the polynucleotide encoding the RT are comprised within the same vector.
  • the same vector further comprises a promoter operably linked to the msr locus and/or the msd locus.
  • the promoter is further operably linked to the polynucleotide encoding the RT. 21.
  • a vector system comprising one or more vectors, comprising the engineered nucleic acid construct of paragraph 1 or 2, wherein the vector system further comprises a second polynucleotide encoding a reverse transcriptase (RT), or a portion thereof, wherein the encoded RT is capable of synthesizing a DNA copy of at least a portion of the msd locus encoding the msDNA, and wherein the msr locus, the msd locus, and the second polynucleotide encoding the RT are provided by at least two different vectors. 22.
  • RT reverse transcriptase
  • the second polynucleotide comprises: i) a polynucleotide listed in Table A, or a polynucleotide having at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%%, at least 99.1%, at least 99.2%, at least 99.3%, at least 99.4%, at least 99.5%, at least 99.6%, at least 99.7%, at least 99.8% or at least 99.9% sequence identity to a polynucleotide listed in Table A; and/or b) the second polynucleotide encodes: i) a polypeptide listed in Table A, or a polypeptide having at least 50%, at least 55%,
  • non-viral vector comprises a plasmid.
  • non-viral vector comprises a liposome, a lipid nanoparticle (LNP), a cationic polymer, a vesicle, or a gold nanoparticle.
  • LNP lipid nanoparticle
  • cationic polymer a cationic polymer
  • a vesicle a vesicle
  • gold nanoparticle a gold nanoparticle.
  • sequence-specific nuclease comprises an RNA-guided sequence-specific nuclease (e.g., a CRISPR/Cas effector enzyme, an engineered RNA-guided FokI-nuclease (e.g., dCas-FokI), an RNA-guided DNA endonuclease, TnpB, IscB, or a transposon-associated nuclease), or a non-RNA-guided sequence-specific nuclease (e.g., a meganuclease, a zinc finger nuclease (ZFN), a TALE nuclease (TALEN), or a restriction endonuclease (RE)).
  • RNA-guided sequence-specific nuclease e.g., a CRISPR/Cas effector enzyme, an engineered RNA-guided FokI-nuclease (e.g., dCas-F
  • the Cas effector enzyme is a Class 1, Type I, II, or III Cas; a Class 2, Type II Cas (e.g., Cas9); or a Class 2, Type V Cas (e.g., Cpfl). 32.
  • the RNA-guided sequence-specific nuclease comprises the CRISPR/Cas effector enzyme, the engineered RNA-guided FokI-nuclease (e.g., dCas-FokI), the RNA- guided DNA endonuclease, TnpB, IscB, IsrB, or the transposon-associated nuclease; or, 2) non-RNA-guided sequence-specific nuclease comprises the meganuclease, the zinc finger nuclease (ZFN), the TALE nuclease (TALEN), or the restriction endonuclease (RE).
  • ZFN zinc finger nuclease
  • TALEN TALE nuclease
  • RE restriction endonuclease
  • An engineered nucleic acid-enzyme construct comprising: a) a non-coding RNA (ncRNA) comprising: i) an msr locus encoding the msr RNA portion of a multi-copy single- stranded DNA (msDNA); and ii) an msd locus encoding the msd RNA portion of the msDNA; b) a heterologous nucleic acid inserted at or within a location selected from: the msd locus, upstream of the msr locus, upstream of the msd locus, and downstream of the msd locus; and c) a sequence encoding a reverse transcriptase (RT), or a domain thereof comprising: i) a polypeptide listed in Table A, or a polypeptide having at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%
  • An engineered nucleic acid-enzyme construct comprising: a) a non-coding RNA (ncRNA) comprising: i) an msr locus encoding the msr RNA portion of a multi-copy single- stranded DNA (msDNA); and ii) an msd locus encoding the msd RNA portion of the msDNA; wherein the ncRNA comprises: i) an ncRNA listed in Table B, or an ncRNA having at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%%, at least 99.1%, at least 99.2%, at least 99.3%, at least 99.4%, at least 99.5%, at least 99.6%
  • An engineered nucleic acid-enzyme construct comprising: a) a non-coding RNA (ncRNA) comprising: i) an msr locus encoding the msr RNA portion of a multi-copy single- stranded DNA (msDNA); and ii) an msd locus encoding the msd RNA portion of the msDNA; wherein, the ncRNA comprises: i) an ncRNA listed in Table B, or an ncRNA having at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%%, at least 99.1%, at least 99.2%, at least 99.3%, at least 99.4%, at least 99.5%, at least 99.
  • An isolated host cell comprising the engineered nucleic acid construct of any one of paragraphs 1-16, the vector system of any of paragraphs 17-33, the RNA molecule of paragraph 34, or the engineered nucleic acid-enzyme construct of any one of paragraphs 35-37.
  • 39. The isolated host cell of paragraph 38, wherein the host cell is a prokaryotic, archeon, or eukaryotic host cell.
  • 40. The isolated host cell of paragraph 38, wherein the eukaryotic host cell is a mammalian host cell. 41.
  • the isolated host cell of paragraph 39, wherein the eukaryotic host cell is a non-human host cell.
  • 42. The isolated host cell of paragraph 40 wherein the mammalian host cell is a human host cell. 43.
  • a pharmaceutical composition comprising: a) the engineered nucleic acid construct of any one of paragraphs 1-16, ncRNA encoded by the engineered nucleic acid construct of any one of paragraphs 1-16, the vector system of any one of paragraphs 17-33, the RNA molecule of paragraph 34, the engineered nucleic acid-enzyme construct of any one of paragraphs 35-37, and/or the isolated host cell of any one of paragraphs 38-43; and b) a pharmaceutically acceptable carrier.
  • a pharmaceutical composition comprising: a) a lipid nanoparticle (LNP); and b) the engineered nucleic acid construct of any one of paragraphs 1-16, ncRNA encoded by the engineered nucleic acid construct of any one of paragraphs 1-16, the vector system of any one of paragraphs 17-33, the RNA molecule of paragraph 34, and/or the engineered nucleic acid-enzyme construct of any one of paragraphs 35-37.
  • lipid nanoparticle comprises: a) one or more ionizable lipids; b) one or more structural lipids; c) one or more PEGylated lipids; and d) one or more phospholipids.
  • the one or more ionizable lipids is selected from the group consisting of those disclosed in Table 2.
  • the pharmaceutical composition of paragraph 47 or 48 wherein the one or more structural lipids are selected from the group consisting of cholesterol, fecosterol, beta sitosterol, sitosterol, ergosterol, campesterol, stigmasterol, brassicasterol, tomatidine, tomatine, ursolic acid, alpha-tocopherol, prednisolone, dexamethasone, prednisone, and hydrocortisone. 50.
  • the pharmaceutical composition of any one of paragraphs 47-49, wherein the one or more PEGylated lipids are selected from the group consisting of PEG-c-DOMG, PEG- DMG, PEG-DLPE, PEG-DMPE, PEG-DPPC, and PEG-DSPE.
  • any one of paragraphs 47-50, wherein the one or more phospholipids are selected from the group consisting of 1,2-distearoyl-sn-glycero-3- phosphocholine (DSPC), 1,2-dioleoyl-sn-glycero-3-phosphoethanolamine (DOPE), 1,2- dilinoleoyl-sn-glycero-3-phosphocholine (DLPC), 1,2-dimyristoyl-sn-glycero- phosphocholine (DMPC), 1.2-dioleoyl-sn-glycero-3-phosphocholine (DOPC), 1,2- dipalmitoyl-sn-glycero-3-phosphocholine (DPPC), 1,2-diundecanoyl-sn-glycero- phosphocholine (DUPC), 1-palmitoyl-2-oleoyl-sn-glycero-3-phosphocho line (POPC), 1,2-di-O-oc
  • lipid nanoparticle comprises about 48.5 mol % ionizable lipid, about 10 mol % phospholipid, about 40 mol % structural lipid, and about 1.5 mol% of PEG lipid.
  • the lipid nanoparticle comprises about 48.5 mol % ionizable lipid, about 10 mol % phospholipid, about 39 mol % structural lipid, and about 2.5 mol% of PEG lipid.
  • the LNP further comprises a targeting moiety operably connected to the LNP.
  • the LNP further comprises one or more additional components selected from the group consisting of DDAB, EPC, 14PA, 18BMP, DODAP, DOTAP, and C12-200.
  • the lipid nanoparticle comprises at least one cationic lipid selected from the group consisting of: a lipid in Table 2, a lipid having a structure of Formula (I), a lipid having a structure of Formula (II), a lipid having a structure of Formula (III), a lipid having a structure of Formula (IV), a lipid having a structure of Formula (V), a lipid having a structure of Formula (VI), and combinations thereof.
  • a lipid in Table 2 a lipid having a structure of Formula (I), a lipid having a structure of Formula (II), a lipid having a structure of Formula (III), a lipid having a structure of Formula (IV), a lipid having a structure of Formula (V), a lipid having a structure of Formula (VI), and combinations thereof.
  • a kit comprising the engineered nucleic acid construct of any one of paragraphs 1-16, ncRNA encoded by the engineered nucleic acid construct of any one of paragraphs 1-16, the vector system of any one of paragraphs 17-33, the RNA molecule of paragraph 34, the engineered nucleic acid-enzyme construct of any one of paragraphs 35-37, the host cell of any one of paragraphs 38-43, or the pharmaceutical composition of any one of paragraphs 44-56, and instructions for genetically modifying a cell with said engineered nucleic acid construct, ncRNA, vector system, host cell, or pharmaceutical composition. 58.
  • a method of modifying a target DNA sequence in a host (e.g., mammalian) cell comprising introducing into the mammalian cell the engineered nucleic acid construct of any one of paragraphs 1-16, ncRNA encoded by the engineered nucleic acid construct of any one of paragraphs 1-16, the vector system of any one of paragraphs 17- 33, the RNA molecule of paragraph 34, the engineered nucleic acid-enzyme construct of any one of paragraphs 35-37, or the pharmaceutical composition of any one of paragraphs 44-56, to allow production of the msDNA in the host (e.g., mammalian) cell, wherein the heterologous nucleic acid in the msDNA is integrated into the genome of the host (e.g., mammalian) cell at the target DNA sequence by homology-dependent recombination.
  • a method of treating a disease or condition in a subject in need thereof comprising administering a therapeutically effective amount of the engineered nucleic acid construct of any one of paragraphs 1-16, ncRNA encoded by the engineered nucleic acid construct of any one of paragraphs 1-16, the vector system of any one of paragraphs 17-33, the RNA molecule of paragraph 34, the engineered nucleic acid-enzyme construct of any one of paragraphs 35-37, the host cell of any one of paragraphs 38-43, or the pharmaceutical composition of any one of paragraphs 44-56 to the subject, thereby treating the disease or condition in the subject.
  • a method of treating a disease or condition in a subject in need thereof comprising administering a therapeutically effective amount of the host cell of any one of paragraphs 38-43 to the subject, thereby treating the disease or condition in the subject.
  • the host cell is autologous to the subject.
  • the host cell is allogeneic to the subject.
  • a gene editing system comprising one or more delivery vehicles, wherein: the delivery vehicle(s) comprise RNA cargo, said RNA cargo comprises (a) at least one mRNA molecule encoding (i) a nucleic acid programmable nuclease and (ii) a retron reverse transcriptase, (b) an engineered retron ncRNA, and (c) guide RNA for the programmable nuclease, each delivery vehicle contains (a)(i) and/or (a)(ii) and/or (b) and/or (c), whereby one delivery vehicle or more than one delivery vehicle delivers (a)(i), (a)(ii), (b), and (c). 2.
  • the delivery vehicle(s) comprise RNA cargo
  • said RNA cargo comprises (a) at least one mRNA molecule encoding (i) a nucleic acid programmable nuclease and (ii) a retron reverse transcriptase, (b) an engineered retron ncRNA, and (c
  • the fusion protein comprises the N- terminal end of the nucleic acid programmable nuclease fused to the C-terminal end of the retron reverse transcriptase (RT:nuclease fusion).
  • RT:nuclease fusion The gene editing system of paragraph 1, wherein (a)(i) and (a)(ii) comprise a first mRNA molecule encoding the nucleic acid programmable nuclease and a second mRNA molecule encoding the retron reverse transcriptase.
  • (c) is separate from (a)(i), (a)(ii) and (b) or is provided in trans. 8.
  • the gene editing system of paragraph 1 wherein the one or more delivery vehicles comprise a liposome or a lipid nanoparticle (LNP). 13. The gene editing system of paragraph 1, wherein (a) the at least one mRNA molecule encoding (i) the nucleic acid programmable nuclease and (ii) the retron reverse transcriptase, and (b) the engineered retron ncRNA are in the same delivery vehicle. 14. The gene editing system of paragraph 1, wherein the (a) the at least one mRNA molecule encoding (i) the nucleic acid programmable nuclease and (ii) the retron reverse transcriptase, and (b) the engineered retron ncRNA are in separate delivery vehicles. 15.
  • nucleic acid programmable nuclease and the retron reverse transcriptase are encoded on separate mRNA molecules and those separate mRNA molecules of (a)(i) and (a)(ii) are contained in the same delivery vehicle.
  • nucleic acid programmable nuclease and the retron reverse transcriptase are encoded on separate mRNA molecules and those separate mRNA molecules of (a)(i) and (a)(ii) are contained in different delivery vehicles. 17.
  • the engineered retron ncRNA includes a sequence of interest encoding a donor polynucleotide comprising an intended edit to be integrated at a target sequence in a cell, and wherein the donor polynucleotide is flanked by a 5' homology arm that hybridizes to a sequence 5’ to the target sequence and a 3' homology arm that hybridizes to a sequence 3' to the target sequence.
  • the nucleic acid programmable nuclease comprises a Cas9 nuclease, a TnpB nuclease, or a Cas12a nuclease. 19.
  • nucleic acid programmable nuclease comprises a Cas9 nuclease.
  • retron reverse transcriptase has an amino acid sequence of Table A, or an amino acid sequence having at least 90%, 95%, 99%, or 100% sequence identity with any amino acid sequence of Table A. 21.
  • the engineered retron ncRNA comprises: A) a pre-msr sequence having a first complementary region of the retron ncRNA; B) an msr sequence including an msr stem-loop structure; C) an msd sequence including an msd stem-loop structure and a sequence of interest, wherein said msd sequence templates a single strand DNA product (RT-DNA) in the presence of the retron reverse transcriptase; and D) a post-msd sequence having a second complementary region, wherein the first and second complementary regions form an a1/a2 duplex region of the retron ncRNA, wherein the msr stem-loop structure, the msd stem-loop structure, or the a1/a2 duplex comprise a modification which result in increased editing efficiency in the presence of a nucleic acid programmable nuclease that associates with the one or more guide RNAs, and wherein optionally one or more of the guide
  • sequence of interest encodes a donor polynucleotide comprising an intended edit to be integrated at a target sequence of a cell, wherein the donor polynucleotide is flanked by a 5' homology arm that hybridizes to a sequence 5’ to the target sequence and a 3' homology arm that hybridizes to a sequence 3' to the target sequence.
  • the ncRNA has a nucleotide sequence of any sequences of Table B, or a nucleotide sequence having at least 90%, 95%, 99%, or 100% sequence identity with any sequence of Table B.
  • An isolated cell comprising the gene editing system of paragraph 1. 25.
  • a composition comprising: a) the gene editing system of paragraph 1; and b) a pharmaceutically or veterinarily acceptable carrier.
  • the delivery vehicle is a lipid nanoparticle comprising: a) one or more ionizable lipids; b) one or more structural lipids; c) one or more PEGylated lipids; and d) one or more phospholipids. 29.
  • the composition of paragraph 28, wherein the one or more ionizable lipids comprises an ionizable lipid set forth in Table 2.
  • a method of genetically modifying a cell comprising: contacting the gene editing system of paragraph 17 with the cell, thereby delivering the RNA cargo to the cell, wherein: the nucleic acid programmable nuclease forms a complex with the guide RNA, wherein said guide RNA directs the complex to the target sequence, the nucleic acid programmable nuclease creates a double-stranded break in in the target sequence, the retron reverse transcriptase and engineered retron ncRNA create RT DNA that comprises the donor polynucleotide, and the donor polynucleotide becomes integrated at the target sequence.
  • An engineered retron comprising: a) an msr locus (that encodes the msr RNA portion of an msDNA); b) an msd locus encoding the msd RNA portion of the msDNA; c) a retron reverse transcriptase (RT), wherein said msd RNA is capable of being reverse transcribed to form the msDNA by the retron reverse transcriptase (RT); d) a heterologous nucleic acid inserted at or within the msd locus, upstream of the msr locus, upstream or downstream of the msd locus; and, e) a sequence modification (e.g., insertion, deletion, and/or substitution of one or more nucleotide(s)) in the msr locus and/or the msd locus that: i) modulates (e.g., enhances) reverse transcription, processivity, acuracy/fidelity, and/or production of
  • sequence modification comprises, in the encoded retron ncRNA, one or more of: (i) a modified (e.g., mutated, reduced, or eliminated) bulge in a1, a2 or both a1 and a2; (ii) an extension or shortening of a1, a2 or both a1 and a2; (iii) an extension or shortening of a spacer sequence between hairpin loops (e.g., S1, S2, S3, S4, S5 and/or S6); (iv) a modified (e.g., mutated or eliminated) bulge in hairpin loops (e.g., L1, L2, L3, L4, L5 and/or L6 (e.g., by removing unpaired bases in the bulge, or by replacing unpaired bases with an equivalent number of base pairs)); (v) a modified (e.g., extended or shortened) length of hairpin loops (e.g.
  • a heterologous nucleic acid such as the coding sequence for an RNA aptamer or a ribozyme
  • a heterologous nucleic acid such as the coding sequence for an RNA aptamer or a ribozyme
  • the msr locus or the msd locus such as in an S region (e.g., S1, S2, S3, S4, S5 and/or S6), or the tip of an L region (e.g., L1, L2, L3, L4, L5 and/or L6)
  • gRNA CRISPR/Cas guide RNA
  • RT retron reverse transcriptase
  • heterologous nucleic acid encodes a protein or peptide of interest
  • said heterologous nucleic acid comprises or encodes a donor / template sequence (e.g., a donor that corrects / repairs / removes a mutation at the target genome site, such as a mutated exon in a disease gene; a functional DNA element (such as a promoter, an enhancer, a protein binding sequence, a methylation site, a homology region for assiting gene editing, etc.); or a coding sequence for a functional RNA element (ncRNAs, etc)).
  • a donor / template sequence e.g., a donor that corrects / repairs / removes a mutation at the target genome site, such as a mutated exon in a disease gene
  • a functional DNA element such as a promoter, an enhancer, a protein binding sequence, a methylation site, a homology region for assiting gene editing, etc.
  • ncRNAs a
  • said protein or peptide of interest comprises a therapeutic protein (such as a wt protein defective in a disease cell, or a therapeutic antibody or antigen-binding fragment thereof) useful in treating a disease.
  • a therapeutic protein such as a wt protein defective in a disease cell, or a therapeutic antibody or antigen-binding fragment thereof
  • said donor / template sequence corrects/repairs/removes a mutation at the target genome site (such as a mutated exon in a disease gene).
  • a sequence-specific nuclease such as a CRISPR/Cas effector enzyme, a ZFN, a TALEN, a meganuclease, TnpB, IscB, or a restriction endonuclease (RE)
  • a sequence-specific nuclease such as a CRISPR/Cas effector enzyme, a ZFN, a TALEN, a meganuclease, TnpB, IscB, or a restriction endonuclease (RE)
  • sequence-specific nuclease is fused to the RT, optionally via a flexible linker (e.g., a flexible linker comprising Gly and Ser rich sequences such as G4S repeats or GS repeats) or by a generally disordered protein sequence (such as unstructured hydrophilic, biodegradable protein polymer, e.g., an XTEN peptide polymer).
  • a flexible linker e.g., a flexible linker comprising Gly and Ser rich sequences such as G4S repeats or GS repeats
  • a generally disordered protein sequence such as unstructured hydrophilic, biodegradable protein polymer, e.g., an XTEN peptide polymer.
  • nuclease is a CRISPR/Cas effector enzyme that forms a complex with a guide RNA (gRNA) recognizing a target sequence, wherein the gRNA is linked to the ncRNA and/or the msDNA, either directly or through a linker / spacer polynucleotide.
  • gRNA guide RNA
  • the DNA-repair modulating biomolecule is a regulatory protein that modulates (e.g., enhances) HDR, and the regulatory protein is fused to the RT or to the sequence-specific nuclease, optionally via a flexible linker (e.g., the flexible linker comprising Gly and Ser rich sequences such as G4S repeats or GS repeats) or by a generally disordered protein sequence (such as unstructured hydrophilic, biodegradable protein polymer, e.g., an XTEN peptide polymer). 14.
  • a vector system comprising a vector comprising the engineered retron of any one of paragraphs 1-13. 15.
  • the vector system of paragraph 14 wherein the msr locus, the msd locus, and the RT gene are provided by the same vector.
  • the vector system of paragraph 14 or 15 wherein the same vector comprises a promoter operably linked to the msr locus and/or the msd locus. 17.
  • the msr locus, the msd locus, and the RT gene are provided by at least two different vectors. 19.
  • sequence-specific nuclease comprises an RNA-guided sequence-specific nuclease (e.g., a CRISPR/Cas effector enzyme, an engineered RNA-guided FokI-nuclease (e.g., dCas-FokI), an RNA-guided DNA endonuclease, TnpB, IscB, or a transposon-associated nuclease), or a non-RNA-guided sequence-specific nuclease (e.g., a meganuclease, a zinc finger nuclease (ZFN), a TALE nuclease (TALEN), or a restriction endonuclease (RE)).
  • RNA-guided sequence-specific nuclease e.g., a CRISPR/Cas effector enzyme, an engineered RNA-guided FokI-nuclease (e.g., dCas-F
  • the Cas effector enzyme is a Class 1, Type I, II, or III Cas; a Class 2, Type II Cas (e.g., Cas9); or a Class 2, Type V Cas (e.g., Cpfl). 25.
  • the RNA-guided sequence-specific nuclease comprises the CRISPR/Cas effector enzyme, the engineered RNA-guided FokI-nuclease (e.g., dCas-FokI), the RNA- guided DNA endonuclease, TnpB, IscB, IsrB, or the transposon-associated nuclease; or, 2) non-RNA-guided sequence-specific nuclease comprises the meganuclease, the zinc finger nuclease (ZFN), the TALE nuclease (TALEN), or the restriction endonuclease (RE). 26.
  • the engineered RNA-guided FokI-nuclease e.g., dCas-FokI
  • the RNA- guided DNA endonuclease TnpB, IscB, IsrB, or the transposon-associated nuclease
  • 27. An isolated host cell comprising the engineered retron of any one of paragraphs 1-13, or the vector system of any of paragraphs 14-26.
  • 28. The host cell of paragraph 27, wherein the host cell is a prokaryotic, archeon, or eukaryotic host cell.
  • 29. The host cell of paragraph 28, wherein the eukaryotic host cell is a mammalian host cell.
  • 31. The host cell of paragraph 29, wherein the mammalian host cell is a human host cell. 32.
  • 33. A pharmaceutical composition comprising the engineered retron of any one of paragraphs 1-13.
  • 34. A delivery vehicle comprising the engineered retron of any one of paragraphs 1-13 or ncRNA encoded by the engineered retron of any one of paragraphs 1-13, the vector of any one of paragraphs 14-26, the host cell of any one of paragraphs 27-32, or the pharmaceutical composition of paragraph 33.
  • the lipid nanoparticle comprises at least one cationic lipid selected from the group consists of: a lipid in Table (I), a lipid having a structure of Formula (I), a lipid having a structure of Formula (II), a lipid having a structure of Formula (III), a lipid having a structure of Formula (IV), a lipid having a structure of Formula (V), a lipid having a structure of Formula (VI), and combinations thereof. 37.
  • a kit comprising the engineered retron of any one of paragraphs 1-13 or ncRNA encoded by the engineered retron of any one of paragraphs 1-13, the vector or vector system of any one of claims 14-26, the host cell of any one of claims 27-32, the pharmaceutical composition of claim 33, or the delivery vehicle of any one of claims 34-36, and instructions for genetically modifying a cell with said engineered retron, ncRNA, vector / vector system, host cell, pharmaceutical composition, or delivery vehicle. 38.
  • a method of modifying a target DNA sequence in a host (e.g., mammalian) cell comprising introducing into the mammalian cell the engineered retron of any one of claims 1-13 or ncRNA encoded by the engineered retron of any one of claims 1-13, the vector or vector system of any one of claims 14-26, the pharmaceutical composition of claim 33, or the delivery vehicle of any one of claims 34-36, to allow the production of the msDNA in the host (e.g., mammalian) cell, wherein the encoded heterologous nucleic acid in the msDNA is integrated into the genome of the host (e.g., mammalian) cell at the target DNA sequence.
  • the engineered retron is engineered from a type IA/IIA1 retron, the ncRNA of which has a secondary structure of comprising a consensus secondary structure illustrated in FIG.2 and comprising a1-S1-L1-L2-S2-L3-S3-a2-S4-L4, wherein: a1/a2 is a stem 8 bp in length; L1 is a stem of 5 bps with a 10-nt tip; L2 is a stem of 7 bps with a 5-nt tip, and a 1/1 bulge 3 nt from the tip; L3 is a stem of 23 bps with a 22-nt tip, and a 2/2 bulge 21 bps from the tip; L4 is a stem of 11 bps with a 5-nt tip; S1 is a single-stranded spacer region between the a1/a2 stem and L1, with no spacer between L1 and L2; S2 is a single-
  • a1/a2 is a stem 6 bps in length with a 2/2 bulge 3 bps from the tip, wherein a1 has a 2-nt overhang and a2 has a 6-nt overhang
  • L1 is a stem of 14 bps with a 3-nt tip, a 1/0 bulge 4 bps from the tip, and a 0/6 bulge 10 bps from the tip
  • L2 is a stem of 23 bps with a 5-nt tip, a 1/1 bulge 4 bps from the tip, and a 0/1 bulge 18 bps from the tip
  • S1 is a single-stranded spacer region between a1/a2 and L1
  • S2 is a single-stranded spacer region between L1 and L2
  • S3 is a single-stranded spacer region between L2 and a1/a2; and conserved nucleotides are as shown in FIG.3;
  • the a1/a2 stem is 16 bp in length, with a 17-base 5 ⁇ overhang and a 16-base 3 ⁇ overhang;
  • L1 is a stem of 6 bps with a 4-nt tip;
  • L2 is a stem of 4 bps with a 4-nt tip, with a 2/2 bulge 2 nts from the tip;
  • L3 is a stem of 3 bps with a 5-nt tip;
  • L4 is a stem of 9 bps with a 5-nt tip, and a 1/1 bulge 4 nts from the tip;
  • S1, S2, S3, and S4 are single-stranded spacer regions between the a1/a2 stem and L1, L2 and L3, L3 and L4, and L4 and the a1/a2 stem, respectively, with no spacer between L1 and L2; wherein the last 5 nts of S1 and the 5
  • a1/a2 is a stem 13 bps in length
  • L1 is a stem of 9 bps with a 3-nt tip
  • L2 is a stem of 10 bps with a 5-nt tip
  • S1 is a single-stranded spacer region between a1/a2 and L1
  • S2 is a single-stranded spacer region between L1 and L2
  • S3 is a single-stranded spacer region between L2 and a1/a2
  • conserved nucleotides are as shown in FIG.5; or e) the engineered retron is engineered from a type IIA1 retron, the ncRNA of which has a secondary structure comprising a consensus secondary structure illustrated in FIG.
  • a1/a2 is a stem 10 bps in length with a 1-nt overhang on a2
  • L1 is a stem of 10 bps with a 3-nt tip
  • L2 is a stem of 7 bps with a 5-nt tip
  • L3 is a stem of 27 bps with a 8-nt tip and a 0/2 bulge 26 bps from the tip
  • S1 is a single-stranded spacer region between a1/a2 and L1
  • S2 is a single-stranded spacer region between L1 and L2
  • S3 is a single-stranded spacer region between L2 and L3
  • S4 is a single-stranded spacer region between L3 and a1/a2
  • conserved nucleotides are as shown in FIG.6; or f) the engineered retron is engineered from a type IIA2 retron, the n
  • a1/a2 is a stem 7 bp in length with no overhangs
  • L1 is a stem of 8 bps with a 3-nt tip
  • L2 is a stem of 30 bps with a 8-nt tip, a 1/1 bulge 2 bps from the tip, and a 1/1 bulge 27 bps from the tip
  • L3 is a stem of 8 bps with a 5-nt tip, and a 0/1 bulge 3 nt from the tip
  • S1 is a single-stranded spacer region between the a1/a2 stem and L1
  • S2 is a single-stranded spacer region between L1 and L2
  • S3 is a single-stranded spacer region between L3 and the a1/a2 stem
  • conserved nucleotides are as shown in FIG.7; or g) the engineered retron is engineered from a type IIA3 retron
  • a1/a2 is a stem 6 bps in length
  • L1 is a stem of 8 bps with a 9-nt tip
  • L2 is a stem of 8 bps with a 3-nt tip
  • S1 is a single-stranded spacer region between a1/a2 and L1
  • S2 is a single-stranded spacer region between L2 and L3
  • S3 is a single-stranded spacer region between L3 and a1/a2
  • conserved nucleotides are as shown in FIG.8 h)
  • the engineered retron is engineered from a type IIA4 retron, the ncRNA of which has a secondary structure comprising a consensus secondary structure illustrated in FIG.
  • a1/a2 is a stem 3 bp in length with no overhangs and a 7-nt tip
  • L1 is a stem of 7 bps with a 3-nt tip
  • L2 is a stem of 6 bps with a 4-nt tip
  • L3 is a stem of 40 bps with a 5-nt tip, and a 2/2 bulge 3 bps from the tip, a 5/4 bulge 10 bps from the tip, and a 12/15 bulge 30 bps from the tip
  • L4 is a stem of 4 bps with a 9-nt tip
  • S1 is a single-stranded spacer region between L1 and L2/L3
  • S2 is a single-stranded spacer region between L2/L3 and L4
  • S3 is a single-stranded spacer region between L4 and the 3 ⁇ end of the ncRNA; and conserved nucleotides are
  • a1/a2 is a stem 15 bps in length with a 1-nt overhang on a1, a 13-nt overhang on a2, and a 7/5 bulge 13-nt from the tip;
  • L1 is a stem of 10 bps with a 3-nt tip;
  • L2 is a stem of 35 bps with a 3-nt tip;
  • L3 is a stem of 6 bps with a 5-nt tip;
  • S1 is a single-stranded spacer region between a1/a2 and L1;
  • S2 is a single-stranded spacer region between L1 and L2;
  • S3 is a single-stranded spacer region between L2 and L3;
  • S4 is a single-stranded spacer region between L3 and a1/a2; and conserved nucleotides are as shown in FIG.10; or j) the engineered retron is engineered from
  • a1/a2 is a stem 2 bps in length with a 1-nt overhang on a2
  • L1 is a stem of 8 bps with a 4-nt tip
  • L2 is a stem of 9 bps with a 3-nt tip and a 1/1 bulge 3 bps from the tip
  • L3 is a stem of 20 bps with a 3-nt tip and a 1/2 bulge 3 bps from the tip
  • S1 is a single-stranded spacer region between a1/a2 and L1
  • S2 is a single-stranded spacer region between L2 and L3
  • S3 is a single-stranded spacer region between L3 and a1/a2
  • conserved nucleotides are as shown in FIG.11; or k) the engineered retron is engineered from a type IIIA2 retron, the ncRNA of which has a secondary structure comprising a
  • a1/a2 is a stem 15 bp in length
  • L1 is a stem of 6 bps with a 4-nt tip
  • L2 is a stem of 13 bps with a 5-nt tip
  • L3 is a stem of 4 bps with a 8-nt tip
  • L4 is a stem of 20 bps with a 4-nt tip and a 2/2 bulge 6 bp from the tip
  • S1 is a single-stranded spacer region between a1/a2 and L1
  • S2 is a single-stranded spacer region between L1 and L2
  • S3 is a single-stranded spacer region between L2 and L3
  • S4 is a single-stranded spacer region between L3 and L4
  • S5 is a single-stranded spacer region between L4 and a1/a2; and conserved nucleotides
  • a1/a2 is a stem 24 bps in length and having a 1/0 bulge 15 bps from the tip, and a 1/1 bulge 19 bps from the tip;
  • L1 is a stem of 7 bps with a 4-nt tip;
  • L2 is a stem of 9 bps with a 8-nt tip;
  • L3 is a stem of 8 bps with a 4-nt tip;
  • L4 is a stem of 4 bps with a 9-nt tip, and a 2/2 bulge 3 bps from the tip;
  • L5 is a stem of 19 bps with a 18-nt tip;
  • L6 is a stem of 5 bps with a 3-nt tip;
  • S1 is a single-stranded spacer region between a1/a2 and L1;
  • S2 is a single-stranded spacer region between L1 and L2
  • a1/a2 is a stem 5 bps in length with a 1/2 bulge 2 bps from the tip
  • L1 is a stem of 8 bps with a 6-nt tip
  • L2 is a stem of 8 bps with a 5-nt tip
  • L3 is a stem of 13 bps with a 14-nt tip and a 1/0 bulge 2 bps from the tip
  • S1 is a single-stranded spacer region between a1/a2 and L1
  • S2 is a single-stranded spacer region between L1 and L2
  • S3 is a single-stranded spacer region between L2 and L3
  • S4 is a single-stranded spacer region between L3 and a1/a2
  • conserved nucleotides are as shown in SEQ ID NO: 19 or FIG.20 or FIG.14; or n) the engineered retron is engineered from
  • the a1/a2 stem is 11 bp in length with no overhang;
  • L1 is a stem of 9 bps with a 3-nt tip;
  • L2 is a stem of 14 bps with a 5-nt tip;
  • L3 is a stem of 9 bps with a 7-nt tip;
  • L4 is a stem of 15 bps with a 7-nt tip;
  • S1, S2, and S3 are single-stranded spacer regions between the a1/a2 stem and L1, L2 and L3, and L4 and the a1/a2 stem, respectively, with no spacer between L1 and L2, and no spacer between L2 and L3; wherein the 5 th -2 nd last nts of S2 and the 3 rd -6 th nts of S3 forms a 4-bp stem, and, conserved nucleotides are as shown in FIG.15; or o
  • a1/a2 is a stem 9 bp in length with no overhang
  • L1 is a stem of 5 bps with a 6-nt tip
  • L2 is a stem of 9 bps with a 4-nt tip
  • L3 is a stem of 26 bps with a 5-nt tip, a 0/1 bulge 7 bps from the tip, and a 0/1 bulge 9 bps from the tip
  • S1 is a single-stranded spacer region between the a1/a2 stem and L1, with no spacer region between L1 and L2
  • S2 is a single-stranded spacer region between L2 and L3
  • S3 is a single-stranded spacer region between L3 and the a1/a2 stem
  • conserved nucleotides are as shown in FIG.17; or q) the engineered retron is engineered from a type IX retron, the ncRNA
  • a1/a2 is a stem 12 bp in length, wherein a1 has a 14-nt overhang and a2 has a 2-nt overhang;
  • L1 is a stem of 11 bps with a 3-nt tip and a 1/3 bulge 7 bp from the tip;
  • L2 is a stem of 25 bps with a 7-nt tip;
  • S1 is a single-stranded spacer region between L1 and L2;
  • S2 is a single-stranded spacer region between L2 and a1/a2; and conserved nucleotides are as shown in FIG.18; or r)
  • the engineered retron is engineered from a type V retron, the ncRNA of which has a secondary structure comprising a consensus secondary structure illustrated in FIG.19 and comprising a1-S1-L1-S2-L2-S3-a2, wherein: a1/a2 is a stem 13
  • a1/a2 is a stem 4 bp in length with a 1 bp 5 ⁇ overhang
  • L1 is a stem of 7 bps with a 4-nt tip
  • L2 is a stem of 8 bps with a 4-nt tip
  • L3 is a stem of 16 bps with a 6-nt tip, a 3/4 bulge 3 bps from the tip, a 2/3 bulge 5 bps from the tip, and a 3/1 bulge 8 bps from the tip
  • S1 is a single-stranded spacer region between the a1/a2 stem and L1
  • S2 is a single-stranded spacer region between L1 and L2
  • S3 is a single-stranded spacer region between L2 and L3, with no spacer region between L3 and L4
  • S4 is a single-stranded spacer region between L4 and the a1/a2 stem
  • a1/a2 is a stem 13 bps in length with a 1-nt overhang on a2
  • L1 is a stem of 7 bps with a 3-nt tip, and a 2/2 bulge 1 bp from the tip
  • L2 is a stem of 8 bps with a 19-nt tip
  • S1 is a single-stranded spacer region between a1/a2 and L1
  • S2 is a single-stranded spacer region between L1 and L2
  • S3 is a single-stranded spacer region between L2 and a1/a2
  • conserved nucleotides are as shown in FIG.23
  • the engineered retron is engineered from a type XIII retron, the ncRNA of which has a secondary structure comprising a consensus secondary structure illustrated in FIG.
  • a1/a2 is a stem 7 bp in length with no overhangs
  • L1 is a stem of 8 bps with a 3-nt tip
  • L2 is a stem of 30 bps with a 8-nt tip, a 1/1 bulge 2 bps from the tip, and a 1/1 bulge 27 bps from the tip
  • L3 is a stem of 8 bps with a 5-nt tip, and a 0/1 bulge 3 nt from the tip
  • S1 is a single-stranded spacer region between the a1/a2 stem and L1
  • S2 is a single-stranded spacer region between L1 and L2
  • S3 is a single-stranded spacer region between L3 and the a1/a2 stem
  • conserved nucleotides are as shown in FIG.24; or x) the engineered retron is engineered from a type XIV
  • the a1/a2 stem is 15 bp in length with no overhang, and a 4/2 bulge 7 bps from the 5 ⁇ end of a1;
  • L1 is a stem of 8 bps with a 5-nt tip;
  • L2 is a stem of 7 bps with a 5-nt tip;
  • L3 is a stem of 13 bps with a 2-nt tip, a 5/9 bulge 5 bps from the tip, and a 5/5 bulge 8 bps from the tip;
  • S1, S2, and S3 are single-stranded spacer regions between the a1/a2 stem and L1, L2 and L3, and L3 and the a1/a2 stem, respectively, with no spacer between L1 and L2; and wherein the 5 th -3 rd last nts of S1 and the 2nd-5th 4 nts of S2 forms a 3-bp stem; and conserved nucleo
  • sequence modification comprises, in the ncRNA, one or more of: (i) a mutated, reduced, or eliminated bulge in a1, a2 or both a1 and a2; (ii) an extension or shortening of a1, a2 or both a1 and a2; (iii) an extension or shortening of a spacer sequence between hairpin loops S1, S2, S3, S4, S5 and/or S6; (iv) a mutated or eliminated bulge in hairpin loops L1, L2, L3, L4, L5 and/or L6 from removal of unpaired bases in the bulge, or by replacing unpaired bases with an equivalent number of base pairs; or (v) extended or shortened length of hairpin loops L1, L2, L3, L4, L5 and/or L6; (vi) an alternative L1 and/or L2 having complement, reverse, or reverse complement sequences; (vii) an increased number of unpaired bases at the tip of hair
  • the engineered retron of any one of paragraphs 1-4 comprising: (i) a heterologous to the retron of the rRT and to the retron of the msRNA nucleic acid inserted into the msr locus or the msd locus in S1, S2, S3, S4, S5 and/or S6, or the tip of an L1, L2, L3, L4, L5 and/or L6, or upstream or downstream of either the msr locus or the msd locus; or, (ii) a first heterologous to the retron of the rRT and to the retron of the msRNA nucleic acid inserted into the msd locus, and a second heterologous to the retron of the rRT and to the retron of the msRNA nucleic acid inserted either upstream of the msr locus or downstream of the msd locus, wherein the second heterologous nucleic acid encodes a CRISPR-Cas guide RNA (
  • the engineered retron of paragraph 8 wherein the donor sequence that corrects or repairs or removes a mutation at a target genome site, or repairs or removes a mutation in an exon in a disease gene; or provides a functional DNA element comprising a promoter, an enhancer, a protein binding sequence, a methylation site, or a homology region for gene editing.
  • said protein or peptide of interest comprises a therapeutic protein.
  • the therapeutic protein comprises a wild type protein that is expressed in a defective manner in a disease cell, or a therapeutic antibody or an antigen-binding fragment thereof. 12.
  • a system comprising: a) a retron, or the rRT and the ncRNA of the retron, or RNA encoding the rRT and the ncRNA of the retron, wherein the retron comprises: i) an engineered retron of any one of paragraphs 1-11, or ii) a retron set forth in Table X or a retron having a sequence having at least 85%, or 90%, or 95% or 99% sequence identity with the sequence of a retron set forth in Table X, or iii) a retron comprising a rRT sequence set forth in Table A, or a sequence having at least 85%, or 90%, or 95% or 99% sequence identity with a rRT sequence set forth in Table A, or iv) a retron encoding an ncRNA comprising a sequence set forth in Table B, or a sequence having at least 85%, or 90%, or 95% or 99% sequence identity with a ncRNA sequence set forth in Table b, or
  • b) comprises a programmable nuclease or a CRISPR-Cas enzyme and guide RNA for the programmable nuclease or the CRISPR-Cas enzyme, or a Class 1, Type I, II, or III Cas, or a Class 2, Type II Cas and guide RNA therefor, or a Cas9 and guide RNA therefor, or a Class 2, Type V Cas and guide RNA therefor, or a Cpfl and guide RNA therefor, or a Zinc Finger Nuclease, or a transcription activator-like effector nuclease (TALEN), or a meganuclease, or TnpB, or IscB, or a restriction endonuclease (RE), or an engineered RNA-guided FokI-nuclease, or a dCas- FokI, or a transposon-associated nuclease; or b) comprises nucleic acid sequence
  • b) includes a regulatory protein that enhances homology directed repair (HDR). 22.
  • HDR homology directed repair
  • the regulatory protein is fused to the rRT or to the sequence-specific nuclease, optionally via a flexible linker or by a generally disordered protein sequence.
  • 23. The system of any one of paragraphs 12-22, further comprising one or more delivery vehicles. 24.
  • the one or more delivery vehicles comprise a liposome or a lipid nanoparticle (LNP); or, wherein the LNP comprises at least one cationic lipid comprising a lipid in Table (I), a lipid having a structure of Formula (I), a lipid having a structure of Formula (II), a lipid having a structure of Formula (III), a lipid having a structure of Formula (IV), a lipid having a structure of Formula (V), a lipid having a structure of Formula (VI), or combinations thereof; or wherein the LNP comprises a) one or more ionizable lipids, b) one or more structural lipids, c) one or more PEGylated lipids, and d) one or more phospholipids; or wherein or wherein the LNP comprises a) one or more ionizable lipids, b) one or more structural lipids, c) one or more PEGylated lipids, and d) one or more phospho
  • RNA cargo comprises (a) at least one mRNA molecule encoding (i) the nucleic acid programmable nuclease or CRISPR-Cas enzyme and (ii) the rRT, (b) and the ncRNA, and (c) guide RNA for the programmable nuclease
  • each delivery vehicle contains (a)(i) and/or (a)(ii) and/or (b) and/or (c), whereby one delivery vehicle or more than one delivery vehicle delivers (a)(i), (a)(ii), (b), and (c). 26.
  • (a)(i) and (a)(ii) comprise a single mRNA molecule encoding the nucleic acid programmable nuclease or CRISPR-Cas enzyme and the retron reverse transcriptase.
  • (a)(i) and (a)(ii) are encoded and expressed as a fusion protein.
  • the fusion protein comprises the C-terminal end of the nucleic acid programmable nuclease or CRISPR-Cas enzyme fused to the N-terminal end of the retron reverse transcriptase (nuclease:RT fusion).
  • the fusion protein comprises the N-terminal end of the nucleic acid programmable nuclease or CRISPR-Cas enzyme fused to the C-terminal end of the retron reverse transcriptase (RT:nuclease fusion).
  • RT:nuclease fusion the retron reverse transcriptase fusion.
  • (a)(i) and (a)(ii) comprise a first mRNA molecule encoding the nucleic acid programmable nuclease or CRISPR-Cas enzyme and a second mRNA molecule encoding the retron reverse transcriptase.
  • (c) is separate from (a)(i), (a)(ii) and (b) or is provided in trans. 32.
  • nucleic acid programmable nuclease or CRISPR-Cas enzyme and the retron reverse transcriptase are encoded on separate mRNA molecules and those separate mRNA molecules of (a)(i) and (a)(ii) are contained in the same delivery vehicle.
  • nucleic acid programmable nuclease or CRISPR-Cas enzyme and the retron reverse transcriptase are encoded on separate mRNA molecules and those separate mRNA molecules of (a)(i) and (a)(ii) are contained in different delivery vehicles.
  • the ncRNA includes a sequence of interest encoding a donor polynucleotide comprising an intended edit to be integrated at a target sequence in a cell, and wherein the donor polynucleotide is flanked by a 5' homology arm that hybridizes to a sequence 5’ to the target sequence and a 3' homology arm that hybridizes to a sequence 3' to the target sequence.
  • the nucleic acid programmable nuclease or CRISPR-Cas enzyme comprises a Cas9 nuclease, a TnpB nuclease, or a Cas12a nuclease. 42.
  • the nucleic acid programmable nuclease comprises a Cas9 nuclease.
  • the ncRNA comprises: A) a pre-msr sequence having a first complementary region of the ncRNA; B) an msr sequence including an msr stem-loop structure; C) an msd sequence including an msd stem-loop structure and a sequence of interest, wherein said msd sequence templates a single strand DNA product (RT-DNA) in the presence of the retron reverse transcriptase; and D) a post-msd sequence having a second complementary region, wherein the first and second complementary regions form an a1/a2 duplex region of the retron ncRNA, wherein the msr stem-loop structure, the msd stem-loop structure, or the a1/a2 duplex comprise a modification which result in increased editing efficiency in the presence of a nucleic acid programmable nuclease.
  • sequence of interest encodes a donor polynucleotide comprising an intended edit to be integrated at a target sequence of a cell, wherein the donor polynucleotide is flanked by a 5' homology arm that hybridizes to a sequence 5’ to the target sequence and a 3' homology arm that hybridizes to a sequence 3' to the target sequence.
  • system is a gene editing system.
  • a vector system comprising vector(s) comprising the engineered retron of any one of paragraphs 1-11, or comprising the system of any one of paragraphs 12-24 or the system of paragraph 45 as dependent on paragraphs 12-24. 47.
  • the vector system of paragraph 46 wherein the msr locus, the msd locus, and the RT gene are provided by the same vector.
  • the vector system of paragraph 46 or 47, wherein the same vector comprises a promoter operably linked to the msr locus and/or the msd locus.
  • 50. The vector system of paragraph 46, wherein the msr locus, the msd locus, and the ret locus are provided by at least two different vectors. 51.
  • the vector system of paragraph 46 or 50 wherein said ret locus is provided in trans with respect to the msr gene and/or the msd gene.
  • 52. The vector system of any one of paragraphs 46-51, wherein the one or more vectors comprise a viral vector and/or a nonviral vector.
  • 53. The vector system of paragraph 52, wherein the nonviral vector comprises a DNA plasmid or mRNA.
  • the viral vector comprises an adenovirus or a lentivirus or an adeno-associated virus. 55.
  • 56. An isolated cell comprising an engineered retron or system of any of the preceding paragraphs. 57. The cell of paragraph 56, wherein the cell is a prokaryotic, archaeon, or eukaryotic cell. 58. The cell of paragraph 57, wherein the eukaryotic cell is a mammalian cell. 59. The cell of paragraph 58, wherein the mammalian cell is a non-human cell. 60. The cell of paragraph 58, wherein the mammalian cell is a human cell. 61.
  • a composition comprising an engineered retron or system or cell of any of the preceding paragraphs, and a pharmaceutical or veterinarily acceptable carrier.
  • a kit comprising an engineered retron or system or cell or composition of any of the preceding paragraphs, and instructions for genetically modifying a cell with the engineered retron or the system or the composition, or for use of the cell. 64.
  • An engineered retron or system or cell or composition or kit of any of the preceding paragraphs for use in modifying a cell or for genetically modifying a cell, or for treatment of a subject or for effecting modification of a target DNA sequence in a host or subject; or use of an engineered retron or system or cell or composition or kit of any of the preceding paragraphs for modifying or genetically modifying a cell, or in treatment of a subject, or for effecting modification of a target DNA sequence in a host or subject.
  • a method of modifying a cell comprising: contacting the engineered retron or system or cell or composition of any of the preceding paragraphs with the cell. 66.
  • a method of genetically modifying a cell comprising: contacting the cell with a system as described in any one of paragraphs 12-45, wherein: a complex is formed and guided to a target sequence, the complex provides a break in the target sequence, the rRT and ncRNA create msDNA that comprises a donor polynucleotide, and the donor polynucleotide becomes integrated at the target sequence.
  • the invention does not intend to encompass within the scope of the invention any product, process, or making of the product or method of using the product, which does not meet the written description and enablement requirements of the USPTO (35 U.S.C. ⁇ 112, first paragraph) or the EPO (Article 83 of the EPC), such that Applicants reserve the right and hereby disclose a disclaimer of any previously described product, process of making the product, or method of using the product. It may be advantageous in the practice of the invention to be in compliance with Art.53(c) EPC and Rule 28(b) and (c) EPC.
  • FIG.1A is a schematic that depicts naturally occurring retrons from genomic DNA stage through production of the msDNA chimeric microsatellite molecule.
  • Retrons are encoded in the bacterial genome and comprise a non-coding RNA (ncRNA) portion and a portion encoding a specialized reverse transcriptase (RT).
  • ncRNA and the RT initially are transcribed from the retron DNA as a single polycistronic message.
  • the initial transcript is processed resulting in the removal or separation of the transcript encoding the retron RT.
  • the remaining transcript is the ncRNA, which undergoes folding to form a secondary structure having several characteristic stem-loops and a duplex formed between the 5 ⁇ and 3 ⁇ regions of the ncRNA (i.e., the a1/a2 duplex).
  • the folded ncRNA is recognized by the accompanying RT which is separately translated and provided in trans.
  • the translated RT typically recognizes certain secondary structures in the ncRNA, and binds the RNA template downstream from the msd region.
  • the RT initiates reverse transcription of the RNA towards its 5 ⁇ end, starting from the 2’-end of a conserved guanosine (G) residue found immediately after a double-stranded RNA structure (the a1/a2 region) within the ncRNA.
  • G conserved guanosine
  • a portion (i.e., the msd region) of the ncRNA serves as a template for reverse transcription, and reverse transcription terminates before reaching the msr locus.
  • cellular RNase H degrades the segment of the ncRNA that serves as template, but not other parts of the ncRNA.
  • FIG.1B is a schematic depicting an embodiment of a recombinant retron contemplated by this disclosure.
  • a nucleic acid molecule comprises a nucleotide sequence encoding a retron ncRNA region (the msr/msd region) is depicted at the top left.
  • the msr region has been modified by introducing one or more nucleotide modifications (e.g., a nucleotide substitution, deletion, or insertion). For example, it may be desirable to introduce one or more nucleotide substitutions in the msr to enhance functionality (e.g., binding of the corresponding ncRNA to RT, improved stability, improved folding, etc.).
  • the modified msr is referred to as msr’.
  • the msd has been modified by introducing a heterologous nucleotide sequence encoding an HDR donor template.
  • the retron DNA has been modifed to introduce a nucleotide sequence encoding a guide RNA at the 3 ⁇ end of the retron DNA sequence.
  • a DNA vector e.g., a plasmid
  • the DNA is shown to be transcribed as a polycistronic message that includes the msr’/msd’ region (forming the ncRNA) which is fused at its 3 ⁇ end to the guide RNA (in other embodiments, the guide RNA could be fused to the 5 ⁇ end of the retron ncRNA.
  • This intermediate is shown to form a complex with a reverse transcriptase provided in trans (e.g., by way of a separate expression vector or delivered mRNA).
  • the top right schematic shows the formation of a complex between the recombinant ncRNA and the RT and the beginning of reverse transcription from the covalently-linked conserved guanosine (G) (i.e., the “priming G” or “priming guanosine”) using the msd RNA as a template sequence.
  • G covalently-linked conserved guanosine
  • a recombinant msDNA is formed which comprises three modifications, as shown: (a) a guide RNA linked to the 3 ⁇ end of the msDNA, (b) a nucleotide change in the msr’, and (c) the reverse-transcribed single-strand DNA comprises a region that is an HDR donor template.
  • FIG.1C is a schematic depicting a recombinant retron-based genome editing system described herein.
  • the components of such a system may include (a) a guide RNA provided in cis (e.g., fused to the recombinant retron msDNA) and/or in trans (e.g., separately expressed in a cell), (b) a recombinant ncRNA (including at least a sequence encoding an HDR donor template and optionally a guide RNA fused ot the ncRNA), (c) a reverse transcriptase, and (d) a programmable nuclease.
  • a guide RNA provided in cis (e.g., fused to the recombinant retron msDNA) and/or in trans (e.g., separately expressed in a cell)
  • a recombinant ncRNA including at least a sequence encoding an HDR donor template and optionally a guide RNA fused ot the ncRNA
  • a reverse transcriptase e.g., a reverse transcriptas
  • FIG.1D provides a simplified schematic of a the natural lifecycle of a retron.
  • Retrons typically comprise a reverse transcriptase (RT) and two non-coding contiguous inverted sequences (msr and msd) transcribed as a single RNA that is folded into a specific secondary structure.
  • RT reverse transcriptase
  • msr and msd non-coding contiguous inverted sequences
  • the conserved NAXXH motif and VTG triplet in retron RTs are indicated.
  • the RT binds downstream from the msd locus in the RNA, initiating reverse transcription of the RNA template towards its 5 ⁇ end, assisted by the 2 ⁇ OH group present in a conserved branching G residue acting as a primer.
  • FIG.1E provides a detailed representation of the natural biological pathway of a retron in a cell, concluding with the generation of the msDNA satellite molecule. This figure parallels FIG.1D but more completely depicts the stages of msDNA production.
  • (1) depicts the retron locus which includes an ncRNA locus having an msr locus and a msd locus (both of which are non-coding) and a reverse transcriptase (RT) locus.
  • the ncDNA locus and the RT locus are transcribed as a single RNA transcript, which is depicted in (2).
  • the colors representing each component in (1) are carried through each of stages (2) through (6).
  • Stages (3) and (4) depict the folding of the ncRNA portion into a series of stem loops, wherein the 5 ⁇ end and the 3 ⁇ ends of the ncRNA form a duplex.
  • the position of the conserved branching guanosine residue having a 2’OH group is show.
  • the branching guanosine serves as a future priming site for the reverse transcriptase.
  • Stage (4) further shows that the region of the transcript encoding the reverse transcriptase is removed, separately being translated to produce the reverse transcriptas enzyme.
  • the reverse transcriptase associates with the folded ncRNA and begins polymerization of a single strand of DNA (i.e., the reverse transcription product) from the primer site (i.e., the conserved branching guanosine residue having the 2’OH end) and using the msd RNA sequence as a template. Reverse transcription terminates at the msr region.
  • the msd RNA template is exonucleotically removed, thereby resulting in a chimeric molecule comprising the msr RNA region which is covalently joined to the ssDNA transcription product through covalent linkage to the conserved guanosine primer residue.
  • FIG.1F is a schematic depicting that the herein disclosed recombinant retron-based genome modification systems may be implemented as (a) cell recorder systems, (b) genome editing systems, and (c) recombineering systems. These uses are not intended to be limiting.
  • FIG.1G is a schematic depicting various configurations contemplated for the recombinant retron disclosed herein.
  • FIG.1H is a schematic that emphasizes that any suitable configuration for presenting the components of the recombinant retron-based genome modification systems disclosed herein to a cell are contemplated, including where the RT and/or the programmable nuclease are provided in trans relative to the retron ncRNA.
  • the RT and the programmable nuclease may be provided as a fusion protein.
  • FIG.1I depicts that the RT and programmable nuclease may be provided as fusion proteins (top, middle) or provided separate from one another.
  • FIG.1J depicts that nuclear localization signals may be engineered into the polypeptides of the disclosure (e.g., a RNA-guide nuclease) to facilitate translocation into the nuclease of cell where editing occurs.
  • FIG.1K is a schematic (not to scale) representation depicting an embodiment of genome editing, in which double-stranded break (DSB) created by a suitable nuclease (such as a CRISPR/Cas effector enzyme, a ZFN, TALEN, meganuclease, TnpB, IscB, or restriction enzymes (Res)) promotes the insertion of a donor or template sequence (shown here as a “marker” flanked by homologous sequences matching those flanking the DSB, provided on a “donor vector”).
  • DSB double-stranded break
  • a suitable nuclease such as a CRISPR/Cas effector enzyme, a ZFN, TALEN, meganuclease, TnpB, IscB, or restriction enzymes (Res) promotes the insertion of a donor or template sequence (shown here as a “marker” flanked by homologous sequences matching those flanking the
  • FIG.2 is a schematic representation of a consensus secondary structure of a retron ncRNA msr/msd of Type IA/IIA1 retron produced by computational structural alignment of ncRNA sequences from Table B as described in Example 3.
  • the colored dots represents the probability a base is at that location (e.g., red circle represents the presence of a base in 97% of the cases, black represents the presence of a base in 90-97% of the cases, grey represents the presence of a base in 75-90% of the cases, and white represents the presence of a base in 50-75% of the cases), as opposed to a gap (no base), whereas the colored letters represent bases that are conserved to different degrees (e.g., with red representing 97%+ conserved, black being 90%+ conserved, and grey at least 75% conserved).
  • Each highlighted base-pair represents a significantly covarying basepair.
  • FIG.3 is a schematic representation of a consensus secondary structure of a retron ncRNA msr/msd of Type IB1 retrons produced by computational structural alignment of ncRNA sequences from Table B as described in Example 3.
  • the colored dots represents the probability a base is at that location (e.g., red circle represents the presence of a base in 97% of the cases, black represents the presence of a base in 90-97% of the cases, grey represents the presence of a base in 75-90% of the cases, and white represents the presence of a base in 50-75% of the cases), as opposed to a gap (no base), whereas the colored letters represent bases that are conserved to different degrees (e.g., with red representing 97%+ conserved, black being 90%+ conserved, and grey at least 75% conserved).
  • FIG.4 is a schematic representation of a consensus secondary structure of a retron ncRNA msr/msd of Type IB2 retrons produced by computational structural alignment of ncRNA sequences from Table B as described in Example 3.
  • the colored dots represents the probability a base is at that location (e.g., red circle represents the presence of a base in 97% of the cases, black represents the presence of a base in 90-97% of the cases, grey represents the presence of a base in 75-90% of the cases, and white represents the presence of a base in 50-75% of the cases), as opposed to a gap (no base), whereas the colored letters represent bases that are conserved to different degrees (e.g., with red representing 97%+ conserved, black being 90%+ conserved, and grey at least 75% conserved).
  • Each highlighted base-pair represents a significantly covarying basepair.
  • FIG.5 is a schematic representation of a consensus secondary structure of a retron ncRNA msr/msd of Type IC retrons produced by computational structural alignment of ncRNA sequences from Table B as described in Example 3.
  • the colored dots represents the probability a base is at that location (e.g., red circle represents the presence of a base in 97% of the cases, black represents the presence of a base in 90-97% of the cases, grey represents the presence of a base in 75-90% of the cases, and white represents the presence of a base in 50-75% of the cases), as opposed to a gap (no base), whereas the colored letters represent bases that are conserved to different degrees (e.g., with red representing 97%+ conserved, black being 90%+ conserved, and grey at least 75% conserved).
  • FIG.6 is a schematic representation of a consensus secondary structure of a retron ncRNA msr/msd of Type IIA other retrons produced by computational structural alignment of ncRNA sequences from Table B as described in Example 3.
  • the colored dots represents the probability a base is at that location (e.g., red circle represents the presence of a base in 97% of the cases, black represents the presence of a base in 90-97% of the cases, grey represents the presence of a base in 75-90% of the cases, and white represents the presence of a base in 50-75% of the cases), as opposed to a gap (no base), whereas the colored letters represent bases that are conserved to different degrees (e.g., with red representing 97%+ conserved, black being 90%+ conserved, and grey at least 75% conserved).
  • Each highlighted base-pair represents a significantly covarying basepair.
  • FIG.7 is a schematic representation of a consensus secondary structure of a retron ncRNA msr/msd of Type IIA2 retrons produced by computational structural alignment of ncRNA sequences from Table B as described in Example 3.
  • the colored dots represents the probability a base is at that location (e.g., red circle represents the presence of a base in 97% of the cases, black represents the presence of a base in 90-97% of the cases, grey represents the presence of a base in 75-90% of the cases, and white represents the presence of a base in 50-75% of the cases), as opposed to a gap (no base), whereas the colored letters represent bases that are conserved to different degrees (e.g., with red representing 97%+ conserved, black being 90%+ conserved, and grey at least 75% conserved).
  • FIG.8 is a schematic representation of a consensus secondary structure of a retron ncRNA msr/msd of Type IIA3 retrons produced by computational structural alignment of ncRNA sequences from Table B as described in Example 3.
  • the colored dots represents the probability a base is at that location (e.g., red circle represents the presence of a base in 97% of the cases, black represents the presence of a base in 90-97% of the cases, grey represents the presence of a base in 75-90% of the cases, and white represents the presence of a base in 50-75% of the cases), as opposed to a gap (no base), whereas the colored letters represent bases that are conserved to different degrees (e.g., with red representing 97%+ conserved, black being 90%+ conserved, and grey at least 75% conserved).
  • Each highlighted base-pair represents a significantly covarying basepair.
  • FIG.9 is a schematic representation of a consensus secondary structure of a retron ncRNA msr/msd of Type IIA4 retrons produced by computational structural alignment of ncRNA sequences from Table B as described in Example 3.
  • the colored dots represents the probability a base is at that location (e.g., red circle represents the presence of a base in 97% of the cases, black represents the presence of a base in 90-97% of the cases, grey represents the presence of a base in 75-90% of the cases, and white represents the presence of a base in 50-75% of the cases), as opposed to a gap (no base), whereas the colored letters represent bases that are conserved to different degrees (e.g., with red representing 97%+ conserved, black being 90%+ conserved, and grey at least 75% conserved).
  • FIG.10 is a schematic representation of a consensus secondary structure of a retron ncRNA msr/msd of Type IIA5 retrons produced by computational structural alignment of ncRNA sequences from Table B as described in Example 3.
  • the colored dots represents the probability a base is at that location (e.g., red circle represents the presence of a base in 97% of the cases, black represents the presence of a base in 90-97% of the cases, grey represents the presence of a base in 75-90% of the cases, and white represents the presence of a base in 50-75% of the cases), as opposed to a gap (no base), whereas the colored letters represent bases that are conserved to different degrees (e.g., with red representing 97%+ conserved, black being 90%+ conserved, and grey at least 75% conserved).
  • Each highlighted base-pair represents a significantly covarying basepair.
  • FIG.11 is a schematic representation of a consensus secondary structure of a retron ncRNA msr/msd of Type IIIA1 retrons produced by computational structural alignment of ncRNA sequences from Table B as described in Example 3.
  • the colored dots represents the probability a base is at that location (e.g., red circle represents the presence of a base in 97% of the cases, black represents the presence of a base in 90-97% of the cases, grey represents the presence of a base in 75-90% of the cases, and white represents the presence of a base in 50-75% of the cases), as opposed to a gap (no base), whereas the colored letters represent bases that are conserved to different degrees (e.g., with red representing 97%+ conserved, black being 90%+ conserved, and grey at least 75% conserved).
  • FIG.12 is a schematic representation of a consensus secondary structure of a retron ncRNA msr/msd of Type IIIA2 retrons produced by computational structural alignment of ncRNA sequences from Table B as described in Example 3.
  • the colored dots represents the probability a base is at that location (e.g., red circle represents the presence of a base in 97% of the cases, black represents the presence of a base in 90-97% of the cases, grey represents the presence of a base in 75-90% of the cases, and white represents the presence of a base in 50-75% of the cases), as opposed to a gap (no base), whereas the colored letters represent bases that are conserved to different degrees (e.g., with red representing 97%+ conserved, black being 90%+ conserved, and grey at least 75% conserved).
  • Each highlighted base-pair represents a significantly covarying basepair.
  • FIG.13 is a schematic representation of a consensus secondary structure of a retron ncRNA msr/msd of IIIA3 retrons produced by computational structural alignment of ncRNA sequences from Table B as described in Example 3.
  • the colored dots represents the probability a base is at that location (e.g., red circle represents the presence of a base in 97% of the cases, black represents the presence of a base in 90-97% of the cases, grey represents the presence of a base in 75-90% of the cases, and white represents the presence of a base in 50-75% of the cases), as opposed to a gap (no base), whereas the colored letters represent bases that are conserved to different degrees (e.g., with red representing 97%+ conserved, black being 90%+ conserved, and grey at least 75% conserved).
  • FIG.14 is a schematic representation of a consensus secondary structure of a retron ncRNA msr/msd of Type IIIA4 retrons produced by computational structural alignment of ncRNA sequences from Table B as described in Example 3.
  • the colored dots represents the probability a base is at that location (e.g., red circle represents the presence of a base in 97% of the cases, black represents the presence of a base in 90-97% of the cases, grey represents the presence of a base in 75-90% of the cases, and white represents the presence of a base in 50-75% of the cases), as opposed to a gap (no base), whereas the colored letters represent bases that are conserved to different degrees (e.g., with red representing 97%+ conserved, black being 90%+ conserved, and grey at least 75% conserved).
  • Each highlighted base-pair represents a significantly covarying basepair.
  • FIG.15 is a schematic representation of a consensus secondary structure of a retron ncRNA msr/msd of Type IIIA5 retrons produced by computational structural alignment of ncRNA sequences from Table B as described in Example 3.
  • the colored dots represents the probability a base is at that location (e.g., red circle represents the presence of a base in 97% of the cases, black represents the presence of a base in 90-97% of the cases, grey represents the presence of a base in 75-90% of the cases, and white represents the presence of a base in 50-75% of the cases), as opposed to a gap (no base), whereas the colored letters represent bases that are conserved to different degrees (e.g., with red representing 97%+ conserved, black being 90%+ conserved, and grey at least 75% conserved).
  • FIG.16 is a schematic representation of a consensus secondary structure of a retron ncRNA msr/msd of Type IIIunk retrons produced by computational structural alignment of ncRNA sequences from Table B as described in Example 3.
  • the colored dots represents the probability a base is at that location (e.g., red circle represents the presence of a base in 97% of the cases, black represents the presence of a base in 90-97% of the cases, grey represents the presence of a base in 75-90% of the cases, and white represents the presence of a base in 50-75% of the cases), as opposed to a gap (no base), whereas the colored letters represent bases that are conserved to different degrees (e.g., with red representing 97%+ conserved, black being 90%+ conserved, and grey at least 75% conserved).
  • Each highlighted base-pair represents a significantly covarying basepair.
  • FIG.17 is a schematic representation of a consensus secondary structure of a retron ncRNA msr/msd of Type IV retrons produced by computational structural alignment of ncRNA sequences from Table B as described in Example 3.
  • the colored dots represents the probability a base is at that location (e.g., red circle represents the presence of a base in 97% of the cases, black represents the presence of a base in 90-97% of the cases, grey represents the presence of a base in 75-90% of the cases, and white represents the presence of a base in 50-75% of the cases), as opposed to a gap (no base), whereas the colored letters represent bases that are conserved to different degrees (e.g., with red representing 97%+ conserved, black being 90%+ conserved, and grey at least 75% conserved).
  • FIG.18 is a schematic representation of a consensus secondary structure of a retron ncRNA msr/msd of Type IX retrons produced by computational structural alignment of ncRNA sequences from Table B as described in Example 3.
  • the colored dots represents the probability a base is at that location (e.g., red circle represents the presence of a base in 97% of the cases, black represents the presence of a base in 90-97% of the cases, grey represents the presence of a base in 75-90% of the cases, and white represents the presence of a base in 50-75% of the cases), as opposed to a gap (no base), whereas the colored letters represent bases that are conserved to different degrees (e.g., with red representing 97%+ conserved, black being 90%+ conserved, and grey at least 75% conserved).
  • Each highlighted base-pair represents a significantly covarying basepair.
  • FIG.19 is a schematic representation of a consensus secondary structure of a retron ncRNA msr/msd of Type V retrons produced by computational structural alignment of ncRNA sequences from Table B as described in Example 3.
  • the colored dots represents the probability a base is at that location (e.g., red circle represents the presence of a base in 97% of the cases, black represents the presence of a base in 90-97% of the cases, grey represents the presence of a base in 75-90% of the cases, and white represents the presence of a base in 50-75% of the cases), as opposed to a gap (no base), whereas the colored letters represent bases that are conserved to different degrees (e.g., with red representing 97%+ conserved, black being 90%+ conserved, and grey at least 75% conserved).
  • FIG.20 is a schematic representation of a consensus secondary structure of a retron ncRNA msr/msd of Type VI retrons produced by computational structural alignment of ncRNA sequences from Table B as described in Example 3.
  • the colored dots represents the probability a base is at that location (e.g., red circle represents the presence of a base in 97% of the cases, black represents the presence of a base in 90-97% of the cases, grey represents the presence of a base in 75-90% of the cases, and white represents the presence of a base in 50-75% of the cases), as opposed to a gap (no base), whereas the colored letters represent bases that are conserved to different degrees (e.g., with red representing 97%+ conserved, black being 90%+ conserved, and grey at least 75% conserved).
  • Each highlighted base-pair represents a significantly covarying basepair.
  • FIG.21 is a schematic representation of a consensus secondary structure of a retron ncRNA msr/msd of Type XI Group 1 retrons produced by computational structural alignment of ncRNA sequences from Table B as described in Example 3.
  • the colored dots represents the probability a base is at that location (e.g., red circle represents the presence of a base in 97% of the cases, black represents the presence of a base in 90-97% of the cases, grey represents the presence of a base in 75-90% of the cases, and white represents the presence of a base in 50-75% of the cases), as opposed to a gap (no base), whereas the colored letters represent bases that are conserved to different degrees (e.g., with red representing 97%+ conserved, black being 90%+ conserved, and grey at least 75% conserved).
  • Each highlighted base-pair represents a significantly covarying basepair.
  • FIG.22 is a schematic representation of a consensus secondary structure of a retron ncRNA msr/msd of Type XI Group 2 retrons produced by computational structural alignment of ncRNA sequences from Table B as described in Example 3.
  • the colored dots represents the probability a base is at that location (e.g., red circle represents the presence of a base in 97% of the cases, black represents the presence of a base in 90-97% of the cases, grey represents the presence of a base in 75-90% of the cases, and white represents the presence of a base in 50-75% of the cases), as opposed to a gap (no base), whereas the colored letters represent bases that are conserved to different degrees (e.g., with red representing 97%+ conserved, black being 90%+ conserved, and grey at least 75% conserved).
  • Each highlighted base-pair represents a significantly covarying basepair.
  • FIG.23 is a schematic representation of a consensus secondary structure of a retron ncRNA msr/msd of Type XII retrons produced by computational structural alignment of ncRNA sequences from Table B as described in Example 3.
  • the colored dots represents the probability a base is at that location (e.g., red circle represents the presence of a base in 97% of the cases, black represents the presence of a base in 90-97% of the cases, grey represents the presence of a base in 75-90% of the cases, and white represents the presence of a base in 50-75% of the cases), as opposed to a gap (no base), whereas the colored letters represent bases that are conserved to different degrees (e.g., with red representing 97%+ conserved, black being 90%+ conserved, and grey at least 75% conserved).
  • Each highlighted base-pair represents a significantly covarying basepair.
  • FIG.24 is a schematic representation of a consensus secondary structure of a retron ncRNA msr/msd of Type XIII retrons produced by computational structural alignment of ncRNA sequences from Table B as described in Example 3.
  • the colored dots represents the probability a base is at that location (e.g., red circle represents the presence of a base in 97% of the cases, black represents the presence of a base in 90-97% of the cases, grey represents the presence of a base in 75-90% of the cases, and white represents the presence of a base in 50-75% of the cases), as opposed to a gap (no base), whereas the colored letters represent bases that are conserved to different degrees (e.g., with red representing 97%+ conserved, black being 90%+ conserved, and grey at least 75% conserved).
  • FIG.25 is a schematic representation of a consensus secondary structure of a retron ncRNA msr/msd of Type XIV retrons produced by computational structural alignment of ncRNA sequences from Table B as described in Example 3.
  • the colored dots represents the probability a base is at that location (e.g., red circle represents the presence of a base in 97% of the cases, black represents the presence of a base in 90-97% of the cases, grey represents the presence of a base in 75-90% of the cases, and white represents the presence of a base in 50-75% of the cases), as opposed to a gap (no base), whereas the colored letters represent bases that are conserved to different degrees (e.g., with red representing 97%+ conserved, black being 90%+ conserved, and grey at least 75% conserved).
  • Each highlighted base-pair represents a significantly covarying basepair.
  • FIG.26 is a schematic representation of a consensus secondary structure of a retron ncRNA msr/msd of Ec107 retrons produced by computational structural alignment of ncRNA sequences from Table B as described in Example 3.
  • the colored dots represents the probability a base is at that location (e.g., red circle represents the presence of a base in 97% of the cases, black represents the presence of a base in 90-97% of the cases, grey represents the presence of a base in 75-90% of the cases, and white represents the presence of a base in 50-75% of the cases), as opposed to a gap (no base), whereas the colored letters represent bases that are conserved to different degrees (e.g., with red representing 97%+ conserved, black being 90%+ conserved, and grey at least 75% conserved).
  • FIG.27 is a schematic representation of a consensus secondary structure of a retron ncRNA msr/msd of Outgroup A retrons produced by computational structural alignment of ncRNA sequences from Table B as described in Example 3.
  • the colored dots represents the probability a base is at that location (e.g., red circle represents the presence of a base in 97% of the cases, black represents the presence of a base in 90-97% of the cases, grey represents the presence of a base in 75-90% of the cases, and white represents the presence of a base in 50-75% of the cases), as opposed to a gap (no base), whereas the colored letters represent bases that are conserved to different degrees (e.g., with red representing 97%+ conserved, black being 90%+ conserved, and grey at least 75% conserved). Each highlighted base-pair represents a significantly covarying basepair.
  • FIG.28 is a phylogenetic tree of RT sequences constructed in accordance with Example 3.
  • FIG.29 is a structural representation of the retron loci associated with each retron type in FIG.28.
  • FIG.30 shows the position of certain retrons (EcoI, Eco3, Eco5, AcoI, RTX003_2042, RTX003_6083v1, and RTX003_6943) within the phylogenetic retron tree of FIG.28.
  • FIG.31A is a plasmid map of an exemplary retron (EcoI) tested in the Examples herein.
  • FIG.31B is a linear representation in 5’ to 3’ direction of the plasmid map of FIG.31A.
  • FIG.31C is a plasmid map of an exemplary retron (RTX3_6083v1) tested in the Examples herein.
  • FIG.31D is a linear representation in 5’ to 3’ direction of the plasmid map of FIG.31C.
  • FIG.32 is a representation of a plasmid-based assay used to measure retron precise edits and indels as performed in the Examples.
  • Step 1 a plasmid (e.g., that of FIG. 31A or FIG.31C) is transfected into human cells (e.g., HEK293t cells) which are engineered to express Cas9. Editing is allowed to occur for 72 hours at 37°C.
  • Step 2 the genomic DNA is extracted from the cells and used to prepare a next-generation sequencing (NGS) library for sequencing.
  • the library is sequenced over the target site (e.g., EMX1) of editing to generate sequence reads.
  • Step 3 the sequencing reads are analyzed to obtain a frequency of sequence reads containing the desired edit (percentage of precision editing) and a frequence of indels at the desired edit site (percentage of indels).
  • FIG.33 is an equivalent representation of FIG.32.
  • FIG.34 is a representation of a methodology for transfecting HEK293T cells. Cells were seeded in 24 well plate one day prior to transfection.
  • FIG.35 is an example of reference sequence and desired editing outcome for Eco3 retron at he EMX1 genomic site. Analysis of editing outcomes is performed using CRISPresso2 pipeline.
  • the editing template inserts a 10 bp insertion into the EMX1 gene (TTACGTCTGC) along with a 6 bp substitution to mutate the PAM sequence (GAAGGG>AAAGTT).
  • FIG.36 shows the results of plasmid-based assay (e.g., according to FIG.33) demonstrating up to about 0.3% precise edits and as low as 40% indels with Eco1 retron in Cas9 expressing HEK293T cells.
  • the plasmid that encodes Eco1 RT and Eco1 ncRNA-sgRNA fusion targeting EMX1 was transfected via lipofection using two different amounts of Lipofectamine.
  • FIG.37 shows the results of plasmid-based assay (e.g., according to FIG.33) demonstrating up to about 0.1% precise edits and as low as 3% indels with AcoI.
  • Aco1 retron has not been experimentally validated to produce msDNA.
  • FIG.38 shows the results of plasmid-based assay (e.g., according to FIG.33) demonstrating up to about 0.3% precise edits and as low as 5% indels with RTX003_2042.
  • This retron can achieve a comparable precise editing to Eco1 but with significantly lower indels (10- fold).
  • RTX003_2042 is a novel retron and precise editing activity observed in this experiment strongly support that RTX003_2042 retron could generate msDNA inside human cells.
  • FIG.39A shows the results of plasmid-based assay demonstrating up to about 0.05 ⁇ 0.08% precise edits and as low as 2.5 ⁇ 4% indels with RTX003_6083v1 and 6943. Both are novel retrons and precise editing activity observed in this experiment strongly support that RTX003_6083v1 and 6943 retron could generate msDNA inside human cells.
  • FIG.39B shows follow up experiments using the same assay of FIG.39A indicated that RTX3_6083v1 and RTX3_6943 generated 3-4 fold more precise edits than Eco1 while indels generated from these two retrons were 2-3 fold lower.
  • FIG.39C shows follow up experiments using the same assay of FIG.39A indicated that RTX3_6083v1 and RTX3_6943 generated 3-4 fold more precise edits than Eco1 while indels generated from these two retrons were 2-3 fold lower.
  • RTX3_2042 showed precise editing at similar frequencies to Eco1 but had more variability than other samples.
  • FIG.39D shows the results of plasmid-based assay demonstrating up to about 0.7% precise edits and as low as ⁇ 4% indels with RTX003_0637, RTX003_1262, and RTX003_6342 compared to empty vector and EcoI.
  • FIG.39E shows the results of plasmid-based assay demonstrating precise editing and indel generation for an array of retrons, including EcoI, Eco3, RTX3_2042_RT_inactivated, RTX3_2042, RTX3_6083v1, RTX3_6943, RTX3_6943, RTX3_1262, RTX3_6342S, and RTX3_6342L.
  • FIG.40 is a representation of an two-RNA editing assay used in the Examples to measure the relative editing efficiency of exemplary retrons using electroporation-based delivery of two RNA components into HEK293T cells.
  • FIG.41 shows the results of two-RNA system (RT mRNA + ncRNA-sgRNA fusion) delivered to Cas9 expressing HEK293T cells by electroporation. Eco1, Eco3 and Eco5 retrons were tested. Results showed precise edits (left graph) up to 0.4% for Eco3 and as low as 10% indels (right graph) for Eco3.
  • FIG.42 shows the results of titration of two-component Eco3 RNA system (RT mRNA + ncRNA-sgRNA fusion) delivered to Cas9 expressing 293T cells by electroporation.
  • the RT mRNA and the ncRNA were mixed at ratios of 1:2, 1:3, 1:4, 1:5, 1:8, 1:10, respectively, and delivered at two different amounts of RT mRNA (0.2 or 0.5 ⁇ g).
  • FIG.43 is a representation of a three-RNA retron editing system which involves delivery by electroporation of three RNA components (RT mRNA, retron ncRNA-sgRNA fusion, and Cas9 mRNA) into HEK293T cells.
  • FIG.44 shows the results of Cas9 mRNA titration of three-component Eco3 RNA system (RT mRNA + ncRNA-sgRNA fusion + Cas9 mRNA) delivered to 293T cells by electroporation.
  • FIG.45 depicts a process of lipofection using three RNA system in HEK293T cells. Cells were seeded in 96 well plate one day prior to transfection.
  • FIG.46 shows the results of three-component Aco1 RNA system (RT mRNA + ncRNA +Cas9 mRNA) delivered to HEK293T cells by lipofection.
  • FIG.47 shows the results of minimal Cas9 nuclease activity when sgRNA is fused to ncRNA of Eco3 retron.
  • FIG.48 is a representation of the all-RNA editing assay used in the Examples to measure the relative editing efficiency of the sample retrons in an all-RNA format, modified with a step of in trans guide RNA spike-in. Electroporation using three RNA system + sgRNA trans spike-in in HEK293T cells.
  • FIG.49 shows the results of guide RNA spike-in in all RNA system (RT mRNA + ncRNA-sgRNA fusion + Cas9 mRNA + sgRNA) delivered to HEK293T cells by electroporation.
  • the guide RNA spike-in in all RNA system increased precise editing up to ⁇ 50 fold.
  • the increasing amount of guide RNA gradually increased precise editing and 1:8 of RT mRNA:ncRNA-sgRNA fusion performed slightly better than 1:6, reaching 13% of precise editing.
  • frequency of indels is shown for respective conditions.
  • FIG.50 represents a lipofection process using three RNA system + gRNA trans spike-in in HEK293T cells.
  • Cells were seeded in 96 well plate one day prior to transfection. Appropriate amount of RT mRNA, ncRNA-sgRNA fusion, Cas9 mRNA, sgRNA and Lipofectamine reagent were mixed and transferred to cells. After 72 hours incubation, genomic DNA was extracted and targeting region was amplified into sequencing libraries. Sequencing data was analyzed by CRISPResso2 and precise edit and indels were calculated.
  • FIG.51 shows the results of guide RNA spike-in (i.e., delivering a separate molecule bolus of guide RNA which in all RNA system (RT mRNA + ncRNA-sgRNA fusion + Cas9 mRNA + sgRNA) delivered to HEK293T cells by lipofection.
  • RT mRNA + ncRNA-sgRNA fusion + Cas9 mRNA + sgRNA RNA spike-in
  • the amount of guide RNA spike- in is titrated at 2, 5 and 10 ng.
  • the guide RNA spike-in in all RNA system increased precise editing up to 3.5 fold, 12% of efficiency. The increasing amount of guide RNA at this range did not further increase precise editing.
  • FIG.52 shows the results of ncRNA-sgRNA fusion separation in all RNA system (RT mRNA + Cas9 mRNA + either ncRNA-sgRNA fusion OR separate ncRNA + sgRNA) delivered to HEK293T cells by lipofection.
  • RT mRNA + Cas9 mRNA + either ncRNA-sgRNA fusion OR separate ncRNA + sgRNA delivered to HEK293T cells by lipofection.
  • the amount of guide RNA spike-in is titrated at 0, 2, 5, 10, 50 and 100 ng.
  • precise editing peaked at 2.23% compared to 1.78% with ncRNA-sgRNA fusion.
  • FIG.53 is a schematic of improved templates used for in vitro transcription to produce ncRNA (left or A) and ncRNA modifications (right or B).
  • A relates to the optimization of RNA production by in vitro transcription.
  • Previously made in vitro transcription experiments to produce RNA used a double-stranded DNA template containing a 3’ overhang (on same strand as T7 promoter sequence).
  • a new template with a blunt end was designed and tested and as shown in FIG.54 results in increased precise editing efficiency.
  • FIG.54 shows the results of ncRNA-sgRNA fusion separation in 4 component all-RNA system (RT mRNA + Cas9 mRNA + ncRNA + sgRNA) delivered to HEK293T cells by Lipofectamine MessengerMAX. All RNA was transfected at a fixed amount as shown on the graphs. Using RNA generated from linearized plasmid template containing a 3’ overhang produced 1.35% precise edits.
  • FIG.55 provides schematics for end protection of RNA from cellular nuclease activity by capping and tailing.
  • A a 7-methylguanosine cap0 was added to 5’ triphosphate of RNA.
  • B a poly-A tail was added to the 3’ end by enzymatic addition. Tail length is estimated over 50 nucleotides.
  • FIG.56 shows the result of end protection of ncRNA-sgRNA fusion by cap and tail in 4 component all-RNA system (RT mRNA + Cas9 mRNA + ncRNA + sgRNA) delivered to HEK293T cells by Lipofectamine MessengerMAX. All RNA was transfected at a fixed amount RT mRNA 100 ng, ncRNA-sgRNA 400ng, Cas9 mRNA 100 ng, and sgRNA 5 ng.
  • RT mRNA + Cas9 mRNA + ncRNA + sgRNA 4 component all-RNA system
  • ncRNA-gRNA fusion was either capped (+cap –tail) or poly-A tailed (-cap +tail) or both capped and poly-A tailed (+cap +tail).
  • RNA without end protection (-cap –tail) produced ⁇ 4.5 % precise edits and the editing was dependent on retron since the absence of RT abrogated precise editing.
  • RNA with either or both protection by cap and tail produced lower precise editing (left graph) but lowered indels (right graph) than without cap and tail.
  • FIG.57 shows the result that shortening msd stems can modulate precising editing in a retron specific manner.
  • Modified retrons include RTX3_4536 (long (L) and short (S) versions), RTX3_6279 (long (L) and short (S) versions), RTX3_6342 (long (L) and short (S) versions), RTX3_6438 (long (L) and short (S) versions), RTX3_6549 (long (L) and short (S) versions), and RTX3_6605 (long (L) and short (S) versions).
  • FIG.58 shows the result that shortening msd stems can modulate precising editing in a retron specific manner.
  • Modified retrons include RTX3_5752 (long (L) and short (S) versions), RTX3_6221 (long (L) and short (S) versions), and RTX3_6034 (long (L) and short (S) versions).
  • FIG.59A Demonstrates that four retrons from various clades are capable of insertions of up to 100 bps to EMX1 locus.
  • Figure shows the results of testing different template lengths for four different retrons in all RNA system delivered to HEK293T cells by lipofection. At given amount of RT mRNA and Cas9 mRNA, ncRNA-sgRNA fusion with different template lengths ranging from 10 to 100 bp were added.
  • FIG.59B Demonstrates that four retrons from various clades are capable of insertions of up to 100 bps to EMX1 locus.
  • Figure shows indel results corresponding with the constructs of FIG.59A, graphing frequency of indels for respective conditions.
  • FIG.60A Demonstrates that additional sgRNA targeting the same genomic locus increases the indel and insert frequencies.
  • FIG.60B Demonstrates that additional sgRNA targeting the same genomic locus increases the indel and insert frequencies. This figure reports the frequency of indels associated with the retrons of FIG.60A.
  • FIG.61A is a schematic showing unedited and edited allele by GFP gene insertion at EMX1 locus using retron editing system.
  • FIG.61B Demonstrates three retrons from various clades are capable of GFP integration at EMX1 locus.
  • the figure provides qPCR data depicting GFP insertion at 5’ or 3’ junctions at EMX1 locus by three retrons.
  • ncRNA-sgRNA engineered to contain GFP gene insert was transfected to HEK293T cells, together with RT mRNA of each retron, Cas9 mRNA, with/without additional sgRNA. Genomic DNA was analyzed by indicated primer pairs to detect GFP gene insertion at 5’ or 3’ junctions on the edited allele.
  • ⁇ Ct was obtained by subtracting +RT sample’s Ct value from –RT sample’s Ct value for relative quantification of GFP insertion at EMX1 locus.
  • the bigger value of ⁇ Ct indicates the higher frequency of insert in the sample.
  • the figure provides qPCR data depicting GFP insertion at 5’ or 3’ junctions at EMX1 locus by two retrons.
  • ncRNA-sgRNA engineered to contain GFP gene insert was transfected to HEK293T cells, together with RT mRNA of each retron, Cas9 mRNA, with/without additional sgRNA.
  • Genomic DNA was analyzed by indicated primer pairs in the table to detect GFP gene insertion at 5’ or 3’ junctions on the edited allele.
  • ⁇ Ct was obtained by subtracting junction Ct value from EMX1 Ct value for relative quantification of GFP insertion at EMX1 locus. With all two retrons, amplicons of both 5’ and 3’ junctions are detectable significantly above background level.
  • FIG.61D shows PCR products for Aco1 were run on Agilent Tapestation High Sensitivity D1000 screentape. In the presence of RT (+RT sample, left on the gel), the expected size of amplicons were detected at both 5’ and 3’ junctions of edited alleles as indicated by arrows. Edited alleles were not detected in the absence of RT (-RT sample, right on the gel). Triangles indicate the expected amplicon for unedited alleles, which is present in both -RT and +RT samples.
  • FIG.62A Demonstrates that order of sgRNA and ncRNA within fused RNA wherein sgRNA is positioned upstream of ncRNA is more active in precise editing and indels.
  • frequency of indels is shown for respective conditions.
  • FIG.62B Demonstrates that where sgRNA is positioned downstream of ncRNA, additional sgRNA compensates for lower editing activity of configuration. Same as previous figure but with the addition of sgRNA, In the presence of additional of sgRNA, the differences between ncRNA-sgRNA and sgRNA-ncRNA depends on each retron. For Eco3 and R2042, sgRNA-ncRNA performed better than ncRNA-sgRNA. For Aco1, ncRNA-sgRNA performed slightly better than sgRNA-ncRNA. On the bottom row of graphs, frequency of indels is shown for respective conditions. [00152] FIG.63 Demonstrates that RT and Cas9 are required for precise editing.
  • ncRNA/ncRNA-sgRNA/sgRNA-ncRNA were added. All insertion templates tested were 25 bp. The best ncRNA format depends on each retron. For Eco3, the highest precise editing was observed with separate ncRNA + additional sgRNA (9.88%). For Aco1, the highest precise editing was observed with ncRNA-sgRNA + additional sgRNA (2.96%). For R2042, the highest precise editing was observed with separate ncRNA + additional sgRNA (7.4%). For R6943, the highest precise editing was observed with ncRNA-sgRNA fusion + additional sgRNA (0.39%).
  • FIG.65 Demonstrates that retrons can achieve precise editing at AAVS1 gene locus. Results of testing four different retrons to insert 25 bp template into AAVS1 locus. Retrons were delivered as all RNA to HEK293T cells by lipofection. For all four retrons, the best precise editing was observed in the presence of additional sgRNA and ranged from 0.73% to 2.05% depending on the retron used. On the right, frequency of indels is shown for respective conditions. [00155] FIG.66 Demonstrates that retrons can achieve precise editing at AAVS1 gene locus. Results of negative controls for AAVS1 experiments in previous figure.
  • FIG.67 Demonstrates that template for either target or non-target strand can be used for precise editing. Results of testing templates on different strands relative to Cas9 sgRNA target strand for two different retrons in all RNA system delivered to HEK293T cells by lipofection. At given amount of RT mRNA, Cas9 mRNA and additional sgRNA, ncRNA- sgRNA was added. All insertion templates tested were 25 bp.
  • Cas9 nickase activity in 4 component all-RNA system (RT mRNA + Cas9 mRNA + ncRNA/ncRNA-sgRNA + sgRNA) delivered to HEK293T cells by Lipofectamine MessengerMAX. All RNA was transfected at a fixed amount RT mRNA 100 ng, ncRNA/ncRNA-sgRNA 400ng, Cas9 variant mRNA 100 ng, and sgRNA 5 ng. Either separated ncRNA or ncRNA-sgRNA fusion was used. Precise editing by Cas9 WT (Wild Type) was ⁇ 9% when either ncRNA or ncRNA-sgRNA was used with additional sgRNA.
  • FIG.69 Demonstrates that retron mediated precise editing by Cas9 nickase is dependent on each component of all RNA system.
  • FIG.70 demonstrates that dual sgRNAs increases precise editing, but not indels. The figure shows the results of testing dual sgRNAs for R2042 in all RNA system delivered to HEK293T cells by lipofection.
  • a ncRNA includes a plurality of ncRNAs and reference to “the reverse transcriptase” includes reference to one or more RTs and equivalents thereof known to those skilled in the art, and so forth.
  • the claims may be drafted to exclude any optional element. As such, this statement is intended to serve as antecedent basis for use of such exclusive terminology as “solely,” “only” and the like in connection with the recitation of claim elements, or use of a “negative” limitation. For example, claims may be drafted to exclude certain RT sequences. [00164] It is appreciated that certain features of the invention, which are, for clarity, described in the context of separate embodiments, may also be provided in combination in a single embodiment.
  • biologically active refers to a characteristic of an agent (e.g., DNA, RNA, or protein) that has activity in a biological system (including in vitro and in vivo biological system), and particularly in a living organism, such as in a mammal, including human and non-human mammals. For instance, an agent when administered to an organism has a biological effect on that organism, is considered to be biologically active.
  • a biological system including in vitro and in vivo biological system
  • a living organism such as in a mammal, including human and non-human mammals.
  • an agent when administered to an organism has a biological effect on that organism, is considered to be biologically active.
  • bulge refers to a small region of unpaired base(s) that interrupts a “stem” of base-paired nucleotides.
  • the bulge may comprise one or two single- stranded or unbase-paired nucleotides joined at both ends by base-paired nucleotides of the stem.
  • the bulge can be symmetrical (viz., the two unbase-paired single-stranded regions have the same number of nucleotides), or asymmetrical (viz., the unbase-paired single stranded region(s) have different or unequal numbers of nucleotides), or there is only one unbase-paired nucleotide on one strand.
  • a bulge can be described as A/B (such as a “2/2 bulge,” or a “1/0 bulge”) wherein A represents the number of unpaired nucleotides on the upstream strand of the stem, and B represents the number of unpaired nucleotides on the downstream strand of the stem.
  • An upstream strand of a bulge is more 5 ⁇ to a downstream strand of the bulge in the primary nucleotide sequence.
  • nucleic acid e.g., RNA, DNA
  • RNA nucleic acid
  • anneal or “hybridize”
  • nucleic acid specifically binds to a complementary nucleic acid
  • Standard Watson-Crick base-pairing includes: adenine (A) pairing with thymidine (T), adenine (A) pairing with uracil (U), and guanine (G) pairing with cytosine (C) [DNA, RNA].
  • adenine (A) pairing with thymidine (T) adenine (A) pairing with uracil (U)
  • guanine (G) can also base pair with uracil (U).
  • G/U base-pairing is at least partially responsible for the degeneracy (i.e., redundancy) of the genetic code in the context of tRNA anti- codon base-pairing with codons in mRNA.
  • a guanine (G) is considered complementary to both a uracil (U) and to an adenine (A).
  • U uracil
  • A adenine
  • G/U base-pair can be made at a given nucleotide position of a dsRNA duplex of a guide RNA molecule, the position is not considered to be non-complementary, but is instead considered to be complementary.
  • sequence of a polynucleotide need not be 100% complementary to that of its target nucleic acid to be specifically hybridizable or hybridizable. Moreover, a polynucleotide may hybridize over one or more segments such that intervening or adjacent segments are not involved in the hybridization event (e.g., a bulge, a loop structure or hairpin structure, etc.).
  • a polynucleotide can comprise 60% or more, 65% or more, 70% or more, 75% or more, 80% or more, 85% or more, 90% or more, 95% or more, 98% or more, 99% or more, 99.5% or more, or 100% sequence complementarity to a target region within the target nucleic acid sequence to which it will hybridize.
  • an antisense nucleic acid in which 18 of 20 nucleotides of the antisense compound are complementary to a target region, and would therefore specifically hybridize, would represent 90 percent complementarity.
  • the remaining noncomplementary nucleotides may be clustered or interspersed with complementary nucleotides and need not be contiguous to each other or to complementary nucleotides.
  • Percent complementarity between particular stretches of nucleic acid sequences within nucleic acids can be determined using any convenient method.
  • Example methods include BLAST programs (basic local alignment search tools) and PowerBLAST programs (Altschul et al., J. Mol. Biol., 1990, 215, 403-410; Zhang and Madden, Genome Res., 1997, 7, 649-656), the Gap program (Wisconsin Sequence Analysis Package, Version 8 for Unix, Genetics Computer Group, University Research Park, Madison Wis.), e.g., using default settings, which uses the algorithm of Smith and Waterman (Adv. Appl. Math., 1981, 2, 482-489), and the like.
  • DNA-guided nuclease is a type of “programmable nuclease,” and a specific type of “nucleic acid-guided nuclease.”
  • An example of a DNA-guided nuclease is reported in Varshney et al., DNA-guided genome editing using structure-guided endonucleases, Genome Biology, 2016, 17(1), 187, which may be used in the context of the present disclosure and is incorporated herein by reference.
  • DNA- guided nuclease or “DNA-guided endonuclease” refers to a nuclease that associates covalently or non-covalently with a guide RNA thereby forming a complex between the guide RNA and the DNA-guided nuclease.
  • the guide RNA comprises a spacer sequence which comprises a nucleotide sequence having complementarity with a strand of a target DNA sequence.
  • the DNA-guided nuclease is indirectly guided or programmed to localize to a specific site in a DNA molecule through its association with the guide RNA, which directly binds or anneals to a strand of the target DNA through its complementarity region via Watson-Crick base-pairing.
  • DNA regulatory sequences can be used interchangeably herein to refer to transcriptional and translational control sequences, such as promoters, enhancers, polyadenylation signals, terminators, protein degradation signals, and the like, that provide for and/or regulate transcription of a non-coding sequence (e.g., guide RNA) or a coding sequence and/or regulate translation of a mRNA into an encoded polypeptide.
  • a non-coding sequence e.g., guide RNA
  • a coding sequence e.g., coding sequence and/or regulate translation of a mRNA into an encoded polypeptide.
  • Donor nucleic acid or “donor polynucleotide” or “donor DNA” or “HDR donor DNA” it is meant a single-stranded DNA to be inserted at a site cleaved by a programmable nuclease (e.g., a CRISPR/Cas effector protein; a TALEN; a ZFN; a meganuclease) (e.g., after dsDNA cleavage, after nicking a target DNA, after dual nicking a target DNA, and the like).
  • a programmable nuclease e.g., a CRISPR/Cas effector protein; a TALEN; a ZFN; a meganuclease
  • the donor polynucleotide can contain sufficient homology to a genomic sequence at the target site, e.g.70%, 80%, 85%, 90%, 95%, or 100% homology with the nucleotide sequences flanking the target site, e.g., within about 200 bases or less of the target site, e.g., within about 190 bases or less of the target site, e.g., within about 180 bases or less of the target site, e.g., within about 170 bases or less of the target site, e.g., within about 160 bases or less of the target site, e.g., within about 150 bases or less of the target site, e.g., within about 140 bases or less of the target site, e.g., within about 130 bases or less of the target site, e.g., within about 120 bases or less of the target site, e.g., within about 110 bases or less of the target site, e.g., within about 100 bases or less of the target site, e.
  • RNA sequence that “encodes” a particular RNA is a DNA nucleotide sequence that is transcribed into RNA.
  • a DNA polynucleotide may encode an RNA (mRNA) that is translated into protein (and therefore the DNA and the mRNA both encode the protein), or a DNA polynucleotide may encode an RNA that is not translated into protein (e.g. tRNA, rRNA, microRNA (miRNA), a “non-coding” RNA (ncRNA), a guide RNA, etc.).
  • the retron DNA may encode the ncRNA loci (which includes the msr and msd regions) as well as a retron RT.
  • Engineered retron As used herein, the term “engineered retron” or equivalently, “recombinant retron,” refers to a retron that does not occur in nature.
  • engineered retrons can include wildtype or naturally-occurring retrons that are modified to contain at least one modification, including a single nucleotide substitution, insertion, or deletion, or a substitution, insertion, or deletion of more than one nucleotide, i.e., up to 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, 69, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92
  • nucleotide of a starting point retron e.g., a wildtype retron
  • the nucleotides may be contiguous or non-contiguous.
  • an engineered retron as a whole is not naturally-occurring, it may include components such as nucleotide sequences that do occur in nature.
  • an engineered retron can have nucleotide sequences from different organisms (e.g., from different bacteria species), or from completely synthetic / artificial / recombinant nucleic acid sequences.
  • an engineered retron can have a bacterial nucleotide sequence, a human nucleotide sequence, a viral nucleotide sequence, and/or a synthetic / artificial / recombinant nucleotide sequence, and/or combinations of such sequences.
  • modifications of the recombinant retrons disclosed herein include the insertion of a heterologous nucleic acid sequence in a retron, for example, inserted into the ncRNA locus, such as in the msr or the msd loci.
  • Linking guide RNA molecules to the 5 ⁇ and/or 3 ⁇ ends also represent another modification envisioned by the recombinant retrons disclosed herein.
  • the guide RNA molecules may also be categorized or referred to more generally as types of heterologous nucleic acid sequences used to modify starting point retrons.
  • Exosomes [0001] As used herein, the term “exosomes” refer to small membrane bound vesicles with an endocytic origin.
  • exosomes are generally released into an extracellular environment from host/progenitor cells post fusion of multivesicular bodies the cellular plasma membrane.
  • exosomes can include components of the progenitor membrane in addition to designed components (e.g. engineered retron).
  • Exosome membranes are generally lamellar, composed of a bilayer of lipids, with an aqueous inter-nanoparticle space.
  • expression vector or “expression construct” refers to a vector that includes one or more expression control sequences, and an “expression control sequence” is a DNA sequence that controls and regulates the transcription and/or translation of another DNA sequence.
  • Suitable expression vectors include, without limitation, plasmids and viral vectors derived from, for example, bacteriophage, baculoviruses, tobacco mosaic virus, herpes viruses, cytomegalovirus, retroviruses, vaccinia viruses, adenoviruses, and adeno- associated viruses.
  • Numerous vectors and expression systems are commercially available, such as from Novagen (Madison, WI), Clontech (Palo Alto, CA), Stratagene (La Jolla, CA), and Invitrogen/Life Technologies (Carlsbad, CA).
  • the present invention comprehends recombinant vectors that may include viral vectors, bacterial vectors, protozoan vectors, DNA vectors, or recombinants thereof.
  • heterologous nucleic acid sequence refers to a genotypically distinct entity from that of the rest of the entity to which it is compared or into which it is introduced or incorporated.
  • a polynucleotide introduced by genetic engineering techniques into a different cell type is a heterologous polynucleotide (e.g., DNA or RNA) and, if expressed, can encode a heterologous polypeptide.
  • a cellular sequence e.g., a gene or portion thereof
  • the heterologous sequence inserted into the wild- type retron regions does not naturally insert into such regions (e.g., the engineered retron with the inserted heterologous sequence is not naturally existing).
  • the heterologous sequence can be from the same species of bacteria in which the wild-type retron is normally found, so long as the heterologous sequence is not naturally inserted in the wild-type retron at the location in which the heterologous sequence is inserted.
  • the heterologous sequence is a mammalian sequence (e.g., a human sequence), or a reverse complement thereof.
  • lipid nanoparticle refers to a type of lipid particle delivery system formed of small solid or semi-solid particles possessing an exterior lipid layer with a hydrophilic exterior surface that is exposed to the non-LNP environment, an interior space which may aqueous (vesicle like) or non-aqueous (micelle like), and at least one hydrophobic inter-membrane space.
  • LNP membranes may be lamellar or non-lamellar and may be comprised of 1, 2, 3, 4, 5 or more layers.
  • LNPs may comprise a nucleic acid (e.g. engineered retron) into their interior space, into the inter membrane space, onto their exterior surface, or any combination thereof.
  • an LNP of the present disclosure comprises an ionizable lipid, a structural lipid, a PEGylated lipid (aka PEG lipid), and a phospholipid.
  • an LNP comprises an ionizable lipid, a structural lipid, a PEGylated lipid (aka PEG lipid), and a zwitterionic amino acid lipid.
  • Linker refers to a molecule linking or joining two other molecules or moieties.
  • the linker can be an amino acid sequence in the case of a linker joining two fusion proteins.
  • an RNA-guided nuclease e.g., Cas12a
  • the linker can also be a nucleotide sequence in the case of joining two nucleotide sequences together.
  • a ncRNA at its 5 ⁇ and/or 3 ⁇ ends may be linked by a nucleotide sequence linker to one or more guide RNAs.
  • the linker is an organic molecule, group, polymer, or chemical moiety.
  • the linker is 5-100 amino acids in length, for example, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 30- 35, 35-40, 40- 45, 45-50, 50-60, 60-70, 70-80, 80-90, 90-100, 100-150, or 150-200 amino acids in length. Longer or shorter linkers are also contemplated.
  • Liposomes [00179] As used herein, the term “liposomes” refer to a type of lipid particle delivery system comprising small vesicles that contain at least one lipid membrane surrounding an aqueous inner-nanoparticle space that is generally not derived from a progenitor/host cell.
  • Liposomes are a versatile carrier platform in that they are capable of transporting hydrophobic or hydrophilic molecules, including small molecules, proteins, and nucleic acids into cells. They were the earliest developed generation of nanoscale medicine delivery platform. Numerous liposomal drug formulations have been approved for human medicines, e.g., Doxil, a lipid nanoparticle formulation of the antitumor agent doxorubicin. Further discuss of liposomes can be found, for example, in Tenchov et al., “Lipid Nanoparticles – From Liposomes to mRNA Vaccine Delivery, a Landscape of Diversity and Advancement,” ACS Nano, 2021, 15, pp. 16982-17015 (the contents of which are incorporated by reference).
  • loop in a polynucleotide refers to a single stranded stretch of one or more nucleotides, such as 2, 3, 4, 5, 6, 7, 8, 9, or 10 nucleotides, wherein the most 5 ⁇ nucleotide and the most 3 ⁇ nucleotide of the loop are each linked to a base- paired nucleotide in a stem.
  • Micelles As used herein, the term “micelles” refer to small particles which do not have an aqueous intra-particle space.
  • Nanoparticle [00181] As used herein, the term “nanoparticle” refers to any nanoscale particle typically ranging in size from about 1 nm to 1000 nm.
  • nuclear localization sequence [00182]
  • the term“nuclear localization sequence” or“NLS” refers to an amino acid sequence that promotes import of a protein (e.g., a RNA-guided nuclease) into the cell nucleus, for example, by nuclear transport.
  • Nuclear localization sequences are known in the art. For example, NLS sequences are described in Plank et al., international PCT application, PCT/EP2000/011690, filed November 23, 2000, published as WO/2001/038547 on May 31, 2001, the contents of which are incorporated herein by reference for its disclosure of exemplary nuclear localization sequences.
  • nucleic acid or “nucleic acid molecule” or “nucleic acid sequence” or “polynucleotide” generally refer to deoxyribonucleic or ribonucleic oligonucleotides in either single- or double-stranded form. The term may also encompass oligonucleotides containing known analogues of natural nucleotides.
  • RNA Ribonucleic acid
  • DNA DNA
  • cDNA including RT DNA
  • DNA or RNA
  • nucleotides Adenine (A), Thymine (T), Guanine (G) and Cytosine (C) also may (or may not) encompass nucleotide modifications, e.g., methylated and/or hydroxylated nucleotides, e.g., Cytosine (C) encompasses 5-methylcytosine and 5- hydroxymethylcytosine.
  • nucleic acid-guided nuclease or “nucleic acid-guided endonuclease” refers to a nuclease that associates covalently or non-covalently with a guide nucleic acid (e.g., a guide RNA or a guide DNA) thereby forming a complex between the guide nucleic acid and the nucleic acid-guided nuclease.
  • the guide nucleic acid comprises a spacer sequence which comprises a nucleotide sequence having complementarity with a strand of a target DNA sequence.
  • the nucleic acid-guided nuclease is indirectly guided or programmed to localize to a specific site in a DNA molecule through its association with the guide nucleic acid, which directly binds or anneals to a strand of the target DNA through its complementarity region via Watson-Crick base-pairing.
  • the nucleic acid- guided nuclease will include a DNA-binding activity (e.g., as in the case for CRISPR Cas9).
  • nucleic acid-guided nuclease is programmed by associating with a guide RNA molecule and in such cases the nuclease may be called “RNA-guided nuclease.” When programmed by a guide DNA, the nuclease may be called a “DNA-guided nuclease.”
  • Nucleic acid-guided, RNA-guided, or DNA-guided nucleases may also be referred to as “programmable nucleases,” which also include other classes of programmable nucleases which associate with specific DNA sequences through amino acid / nucleotide sequence recognition (e.g., zinc fingers nucleases (ZFN) and transcription activator like effector nucleases (TALEN)) rather than through guide RNAs.
  • ZFN zinc fingers nucleases
  • TALEN transcription activator like effector nucleases
  • any nuclease contemplated herein may also be engineered to remove, inactivate, or otherwise eliminate one or more nuclease activities (e.g., by introducing a nuclease-inactiving mutation in the active site(s) of a nuclease).
  • a nuclease that has been modified to remove, inactivate, or otherwise eliminate all nuclease activity may be referred to as a “dead” nuclease.
  • a dead nuclease is not able to cut either strand of a double-stranded DNA molecule.
  • a nuclease that has been modified to remove, inactivate, or otherwise eliminate at least one nuclease activity but which still retains at least one nuclease activity may be referred to as a “nickase” nuclease.
  • a nickase nuclease cuts one strand of a double-stranded DNA molecule, but not both strands.
  • a CRISPR Cas9 naturally comprises two distinct nuclease activity domains, namely, the HNH domain and the RuvC domain. The HNH domain cuts the strand of DNA bound to the guide RNA and the RuvC domain cuts the protospacer strand.
  • RNA-guided nuclease may be similarly converted to nickases and/or dead nucleases by inactivating one or more of the existing nuclease domains.
  • operably linked refers to the correct location and orientation in relation to a polynucleotide (e.g., a coding sequence) to control the initiation of transcription by RNA polymerase and expression of the coding sequence, such as one for the msr gene, msd gene, and/or the ret gene.
  • a polynucleotide e.g., a coding sequence
  • Other transcriptional control regulatory elements e.g., enhancer sequences, transcription factor binding sites
  • programmable nuclease is meant to refer to a polypeptide that has the property of selective localization to a specific desired nucleotide sequence target in a nucleic acid molecule (e.g., to a specific gene target) due to one or more targeting functions.
  • targeting functions can include one or more DNA-binding domains, such as zinc finger domains characteristic of many different types of DNA binding proteins or TALE domains characteristic of TALEN proteins.
  • Such targeting function may also include the ability to associate and/or form a complex with a guide RNA, which then localizes to a specific site on the DNA which bears a sequence that is complementary to a portion of the guide RNA (i.e., the spacer of the guide RNA).
  • the programmable nuclease may be a single protein which comprises both a domain that binds directly (e.g., a ZF protein) or indirectly (e.g., an RNA-guided protein) to a target DNA site, as well as a nuclease domain.
  • the programmable nuclease may be a composite of two or more separate proteins or domains (from different proteins) which together provide the necessary functions of selective DNA binding and nuclease activity.
  • the programmable nuclease may comprise a (a) nuclease-inactive RNA-guided nuclease (which still is capable of binding a guide RNA, localizing to a target DNA, and binding to the target DNA, but not capable of cutting or nicking the strands) fused to a (b) nuclease protein or domain, such as a FokI nuclease.
  • promoter refers to a nucleic acid molecule with a sequence recognized by the cellular transcription machinery and which is able to initiate transcription of a downstream gene.
  • a promoter can be constitutively active, meaning that the promoter is always active in a given cellular context, or conditionally active, meaning that the promoter is only active in the presence of a specific condition.
  • a conditional promoter may only be active in the presence of a specific protein that connects a protein associated with a regulatory element in the promoter to the basic transcriptional machinery, or only in the absence of an inhibitory molecule.
  • a “recombinant nucleic acid” or “recombinant nucleotide” refers to a molecule that is constructed by joining nucleic acid molecules, which optionally may self-replicate in a live cell.
  • Retron refers to a specific type of naturally-occuring and distinct DNA sequence found in the genome of many bacteria which typically encodes three distinct components, namely, (a) a non-coding RNA (“ncRNA”) (comprising continguous inverted sequences (msr and msd), (b) a reverse transcriptase (RT)-coding gene (ret), and (c) in many cases, a retron-associated gene of unknown function. Retrons are particularly defined by their unique ability to produce a satellite DNA known as msDNA (multicopy single-stranded DNA).
  • msDNA multicopy single-stranded DNA
  • ncRNA comprising the msr and msd elements
  • ret gene are transcribed as a single polycitronic RNA transcript which processed into the ncRNA transcript and a transcript encoding the ret gene.
  • the ncRNA then becomes folded into a specific secondary structure.
  • the RT then binds the folded ncRNA and reverse transcribes the msd region to form a single strand of cDNA (the msDNA) that remains covalently attached to the RNA template via a 2’-5 ⁇ phophodiester bond and base-pairing between the 3 ⁇ ends of the msDNA and the RNA template.
  • Retron component refers to a distinct element or feature of a retron, namely (a) a non-coding RNA (“ncRNA”) (comprising continguous inverted sequences (msr and msd), (b) a reverse transcriptase (RT)-coding gene (ret), and (c) in many cases, a retron-associated gene of unknown function.
  • ncRNA non-coding RNA
  • RT reverse transcriptase-coding gene
  • RNA-guided nuclease is a type of “programmable nuclease,” and a specific type of “nucleic acid-guided nuclease.”
  • RNA-guided nuclease or “RNA-guided endonuclease” refers to a nuclease that associates covalently or non- covalently with a guide RNA thereby forming a complex between the guide RNA and the RNA- guided nuclease.
  • the guide RNA comprises a spacer sequence which comprises a nucleotide sequence having complementarity with a strand of a target DNA sequence.
  • sequence identity refers to the overall relatedness between polymeric molecules, e.g., between polynucleotide molecules (e.g., DNA molecules and/or RNA molecules) and/or between polypeptide molecules.
  • Calculation of the percent identity of two polynucleotide sequences can be performed by aligning the two sequences for optimal comparison purposes (e.g., gaps can be introduced in one or both of a first and a second nucleic acid sequences for optimal alignment and non-identical sequences can be disregarded for comparison purposes).
  • the length of a sequence aligned for comparison purposes is at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, at least 95%, or 100% of the length of the reference sequence.
  • the nucleotides at corresponding nucleotide positions are then compared.
  • the percent identity between the two sequences is a function of the number of identical positions shared by the sequences, taking into account the number of gaps, and the length of each gap, which needs to be introduced for optimal alignment of the two sequences.
  • the comparison of sequences and determination of percent identity between two sequences can be accomplished using a mathematical algorithm.
  • the percent identity between two nucleotide sequences can be determined using methods such as those described in Computational Molecular Biology, Lesk, A. M., ed., Oxford University Press, New York, 1988; Biocomputing: Informatics and Genome Projects, Smith, D.
  • the percent identity between two nucleotide sequences can be determined using the algorithm of Meyers and Miller (CABIOS, 1989, 4:11- 17), which has been incorporated into the ALIGN program (version 2.0) using a PAM120 weight residue table, a gap length penalty of 12 and a gap penalty of 4.
  • the percent identity between two nucleotide sequences can, alternatively, be determined using the GAP program in the GCG software package using an NWSgapdna. CMP matrix. Methods commonly employed to determine percent identity between sequences include, but are not limited to those disclosed in Carillo, H. and Lipman, D., SIAM J Applied Math., 48:1073 (1988); incorporated herein by reference.
  • the term“subject” refers to an individual organism, for example, an individual mammal. In some embodiments, the subject is a human. In some embodiments, the subject is a non-human mammal. In some embodiments, the subject is a non-human primate.
  • the subject is a rodent. In some embodiments, the subject is a sheep, a goat, a cattle, a cat, or a dog. In some embodiments, the subject is a vertebrate, an amphibian, a reptile, a fish, an insect, a fly, or a nematode. In some embodiments, the subject is a research animal. In some embodiments, the subject is genetically engineered, e.g., a genetically engineered non- human subject. The subject may be of either sex and at any stage of development. The terms “individual,” “subject,” “host,” and “patient,” used interchangeably herein.
  • the term “stem” refers to two or more base pairs, such as 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20 or more base pairs, formed by inverted repeat sequences connected at a “tip,” where the more 5 ⁇ or “upstream” strand of the stem bends to allows the more 3 ⁇ or “downstream” strand to base-pair with the upstream strand.
  • the number of base pairs in a stem is the “length” of the stem.
  • the tip of the stem is typically at least 3 nucleotides, but can be 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15 or more nucleotides.
  • An otherwise continuous stem may be interrupted by one or more bulges as defined herein.
  • the number of unpaired nucleotides in the bulge(s) are not included in the length of the stem.
  • the position of a bulge closest to the tip can be described by the number of base pairs between the bulge and the tip (e.g., the bulge is 4 bps from the tip).
  • the position of the other bulges (if any) further away from the tip can be described by the number of base pairs in the stem between the bulge in question and the tip, excluding any unpaired bases of other bulges in between.
  • Synthetic or artificial nucleic acid refers nucleic acids that are non-naturally occurring sequences. Such sequences do not originate from, or are not known to be present in any living organism (e.g., based on sequence search in existing sequence databases). Recombinant nucleic acids and synthetic nucleic acids also include those molecules that result from the replication of either of the foregoing.
  • Engineered nucleic acid constructs of the present disclosure such as the engineer ed retron described herein, may be encoded by a single molecule (e.g., encoded by or present on the same plasmid or other suitable vector) or by multiple different molecules (e.g., multiple independently-replicating vectors).
  • Target site is a polynucleotide (e.g., DNA such as genomic DNA) that includes a site or specific locus (“target site” or “target sequence”) targeted by a recombinant retron genome modification system disclosed herein.
  • target site is a polynucleotide (e.g., DNA such as genomic DNA) that includes a site or specific locus (“target site” or “target sequence”) targeted by a recombinant retron genome modification system disclosed herein.
  • target sequence is the sequence to which the guide sequence of a guide nucleic acid (e.g., guide RNA) will hybridize.
  • the target site (or target sequence) 5 ⁇ -GTCAATGGACC-3 ⁇ within a target nucleic acid is targeted by (or is bound by, or hybridizes with, or is complementary to) the sequence 5 ⁇ -GGTCCATTGAC-3 ⁇ .
  • Suitable hybridization conditions include physiological conditions normally present in a cell.
  • the strand of the target nucleic acid that is complementary to and hybridizes with the guide RNA is referred to as the “complementary strand” or “target strand”; while the strand of the target nucleic acid that is complementary to the “target strand” (and is therefore not complementary to the guide RNA) is referred to as the “non-target strand” or “non- complementary strand.”
  • Treatment refers to a clinical intervention aimed to reverse, alleviate, delay the onset of, or inhibit the progress of a disease or disorder, or one or more symptoms thereof, as described herein.
  • treatment may be administered after one or more symptoms have developed and/or after a disease has been diagnosed.
  • treatment may be administered in the absence of symptoms, e.g., to prevent or delay onset of a symptom or inhibit onset or progression of a disease.
  • treatment may be administered to a susceptible individual prior to the onset of symptoms (e.g., in light of a history of symptoms and/or in light of genetic or other susceptibility factors). Treatment may also be continued after symptoms have resolved, for example, to prevent or delay their recurrence.
  • upstream and downstream are terms of relativity that define the linear position of at least two elements located in a nucleic acid molecule (whether single or double-stranded) that is orientated in a 5 ⁇ -to-3 ⁇ direction.
  • a first element is said to be upstream of a second element in a nucleic acid molecule where the first element is positioned somewhere that is 5 ⁇ to the second element.
  • a first element is downstream of a second element in a nucleic acid molecule where the first element is positioned somewhere that is 3 ⁇ to the second element.
  • variant retron RT is retron RT comprising one or more changes in amino acid residues as compared to a wild type retron RT amino acid sequence.
  • variant retron RT encompasses homologous proteins having at least 75%, or at least 80%, or at least 85%, or at least 90%, or at least 95%, or at least 99% percent identity with a reference sequence and having the same or substantially the same functional activity or activities as the reference sequence.
  • vector permits or facilitates the transfer of a polynucleotide from one environment to another. It is a replicon such as a plasmid, phage, or cosmid into which another DNA segment may be inserted so as to bring about the replication of the inserted segment (e.g., the subject engineered retron). Generally, a vector is capable of replication when associated with the proper control elements.
  • vector may include cloning and expression vectors, as well as viral vectors and integrating vectors.
  • Wild type is a term of the art understood by skilled persons and means the typical form of an organism, strain, gene, protein, or characteristic as it occurs in nature as distinguished from mutant or variant forms.
  • DETAILED DESCRIPTION [00202] The present disclosure provides systems, methods and compositions used for precise genome editing, including installing nucleic acid insertions, replacements, and deletions at targeted and precise genome sites, wherein said systems, methods, and compositions are based on novel and/or modified retrons or components thereof, such as modified versions of the retron RTs of Table X, modified versions of the ncRNAs of Table A, and modified versions of the RTs of Table B.
  • the present disclosure provides recombinant retrons comprising one or more genetic modifications which improves the functionality and/or properties of a retron.
  • Such genetic modifications can include a mutation, insertion, deletion, inversion, replacement, substitution, or translocation of one or more contiguous or non-contiguous nucleobases in a nucleic acid molecule encoding a retron or a component of a retron, such as an ncRNA or a reverse transcriptase.
  • the retron that becomes modifed with the one or more genetic modifications is a naturally occurring retron or retron component (e.g., naturally occuring ncRNA of Table A or RT) ability to facilitate homology-dependent recombination (or HDR) in a cell, thereby resulting in a relative increase in the concentrations or amounts of msDNA comprising a DNA donor template.
  • a naturally occurring retron or retron component e.g., naturally occuring ncRNA of Table A or RT
  • HDR homology-dependent recombination
  • the recombinant retrons are based on and/or derived from a naturally-occurring retron, such as any retron-related sequence provided by Table X (the introduction of the one or more genetic modifications into a set of 7257 previously unknown retrons discovered through computational methods described herein (e.g., see Examples).
  • the recombinant retrons are based on introducing the one or more genetic modifications into previously available retron sequences (e.g., the “Mestre et al., Systematic Prediction of Genes Functionally Associated with Bacterial Retrons and Classification of The Encoded Tripartite Systems, Nucleic Acids Research, Volume 48, Issue 22, 16 December 2020, Pages 12632-12647” (incorporated herein by reference) to achieve recombinant retrons with the enhanced ability to produce increased concentrations or amounts of msDNA comprising a DNA donor template.
  • retron sequences e.g., the “Mestre et al., Systematic Prediction of Genes Functionally Associated with Bacterial Retrons and Classification of The Encoded Tripartite Systems, Nucleic Acids Research, Volume 48, Issue 22, 16 December 2020, Pages 12632-12647” (incorporated herein by reference) to achieve recombinant retrons with the enhanced ability to produce increased concentrations or amounts of msDNA
  • the present disclosure further provides nucleic acid molecules encoding the recombinant retrons and/or recombinant retron components (e.g., a recombinant ncRNA and/or a recombinant retron RT).
  • the present disclosure provides genome editing systems comprising recombinant retron components (e.g., recombinant ncRNA and/or recombinant RT), programmable nucleases (e.g., RNA-guided nucleases, such as CRISPR-Cas proteins, ZFPs, and TALENS), and guide RNAs (in the case where RNA-guide nucleases are used in said genome editing systems).
  • recombinant retron components e.g., recombinant ncRNA and/or recombinant RT
  • programmable nucleases e.g., RNA-guided nucleases, such as CRISPR-Cas proteins, ZFPs,
  • the disclosure provides nucleic acid molecules encoding the described genome editing systems and said components thereof, as well as polypeptides making up the components of said genome editing systems.
  • the disclosure provides vectors for transferring and/or expressing said genome editing systems, e.g., under in vitro, ex vivo, and in vivo conditions.
  • the disclosure provides cell-delivery compositions and methods, including compositions for passive and/or active transport to cells (e.g., plasmids), delivery by virus-based recombinant vectors (e.g., AAV and/or lentivirus vectors), delivery by non-virus-based systems (e.g., liposomes and LNPs), and delivery by virus-like particles.
  • the retron-based genome editing systems described herein may be delivered in the form of DNA (e.g., plasmids or DNA-based virus vectors), RNA (e.g., ncRNA and mRNA delivered by LNPs), a mixture of DNA and RNA, protein (e.g., virus-like particles), and ribonucleoprotein (RNP) complexes.
  • DNA e.g., plasmids or DNA-based virus vectors
  • RNA e.g., ncRNA and mRNA delivered by LNPs
  • protein e.g., virus-like particles
  • RNP ribonucleoprotein
  • each of the components of the retron-based genome editing system is delivered by an all-RNA system, e.g., the delivery of one or more RNA molecules (e.g., mRNA and/or ncRNA) by one or more LNPs, wherein the one or more RNA molecules form the ncRNA and guide RNA (as needed) and/or are translated into the polypeptide components (e.g., the RT and a programmable nuclease).
  • an all-RNA system e.g., the delivery of one or more RNA molecules (e.g., mRNA and/or ncRNA) by one or more LNPs, wherein the one or more RNA molecules form the ncRNA and guide RNA (as needed) and/or are translated into the polypeptide components (e.g., the RT and a programmable nuclease).
  • the disclosure provides methods for genome editing by introducing a retron-based genome editing system described herein into a cell (e.g., under in vitro, in vivo, or ex vivo conditions) comprising a target edit site, thereby resulting in an edit at the target edit.
  • a retron-based genome editing system described herein into a cell (e.g., under in vitro, in vivo, or ex vivo conditions) comprising a target edit site, thereby resulting in an edit at the target edit.
  • the disclosure provides formulations comprising any of the aforementioned components for delivery to cells and/or tissues, including in vitro, in vivo, and ex vivo delivery, recombinant cells and/or tissues modified by the recombinant retron-based genome modification systems and methods described herein, and methods of modifying cells by conducting genome editing and related DNA donor-dependent methods, such as recombineering, or cell recording, using the herein disclosed retron-based genome modification systems.
  • the disclosure also provides methods of making the recombinant retrons, retron-based genome modification systems, vectors, compositions and formulations described herein, as well as to pharmaceutical compositions and kits for modifying cells under in vitro, in vivo, and ex vivo conditions that comprise the herein disclosed genome editing and/or modification systems.
  • Described herein are engineered retrons comprising one or more heterologous nucleic acids.
  • the one or more heterologous nucleic acids may be inserted, for example, at or within a location selected from: the msd locus, upstream of the msr locus, upstream of the msd locus, and downstream of the msd locus.
  • the engineered retrons have structural improvements over their naturally existing counterparts or wild-type retrons at least with respect to the encoded ncRNA and/or the reverse transcriptase (RT), such that the engineered retron or the encoded ncRNA thereof, when delivered to a host cell, such as a mammalian host cell, exhibits various functional improvements over its naturally existing/wild- type retron elements.
  • RT reverse transcriptase
  • Exemplary (non-limiting functional improvements) may include any one or more of the features described herein.
  • the engineered retron may comprise a sequence modification (e.g., insertion, deletion, and/or substitution of one or more nucleotide(s)) in the msr locus and/or the msd locus that: i) modulates (e.g., enhances) reverse transcription, processivity, accuracy/fidelity, and/or production of the msDNA (e.g., in the mammalian cell); ii) modulates (e.g., reduces) immunogenicity of the ncRNA encoded by the engineered retron (e.g., encoded by the msr locus and/or the msd locus) in a host (e.g., a host comprising the mammalian cell); iii) comprises a nucleotide sequence that modulates (e.g., inhibits or antagonizes) a function of the msDNA; and/or iv) modulates (e.g., improves
  • a sequence modification
  • the engineered retron is an engineered nucleic acid construct comprising: a) a first polynucleotide encoding a non-coding RNA (ncRNA), said first polynucleotide comprising: i) an msr locus encoding the msr RNA portion of a multi-copy single-stranded DNA (msDNA); and ii) an msd locus encoding the msd RNA portion of the msDNA; and [00208] b) one or more heterologous nucleic acids inserted at or within a location selected from: the msd locus, upstream of the msr locus, upstream of the msd locus, and downstream of the msd locus.
  • ncRNA non-coding RNA
  • the engineered nucleic acid construct may further comprise a second polynucleotide encoding a reverse transcriptase (RT), or a portion thereof, wherein the encoded RT is capable of synthesizing a DNA copy of at least a portion of the msd locus encoding the msDNA.
  • RT reverse transcriptase
  • the engineered retron of the invention encodes a reverse transcriptase (RT) or a functional domain thereof, comprising: i) a polypeptide listed in Table A, or a polypeptide having at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%%, at least 99.1%, at least 99.2%, at least 99.3%, at least 99.4%, at least 99.5%, at least 99.6%, at least 99.7%, at least 99.8% or at least 99.9% sequence identity to a polypeptide listed in Table A; and/or ii) a polypeptide listed in any one of Table C.
  • RT reverse transcriptase
  • the RT does not comprise a polypeptide listed in Table X.
  • the engineered retron of the invention encodes a reverse transcriptase (RT) or a functional domain thereof, comprising: i) a polynucleotide listed in Table A, or a polynucleotide having at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%%, at least 99.1%, at least 99.2%, at least 99.3%, at least 99.4%, at least 99.5%, at least 99.6%, at least 99.7%, at least 99.8% or at least 99.9% sequence identity to a polynucleotide listed in Table A; and/or ii) a reverse transcriptase (RT) or a
  • the polynucleotide encoding the RT does not comprise a polynucleotide of Table X.
  • the engineered retron of the invention encodes an ncRNA comprising: (I) an ncRNA listed in Table B, or an ncRNA having at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%%, at least 99.1%, at least 99.2%, at least 99.3%, at least 99.4%, at least 99.5%, at least 99.6%, at least 99.7%, at least 99.8% or at least 99.9% sequence identity to an ncRNA in Table B.
  • the engineered retron of the invention encodes an ncRNA and a reverse transcriptase (RT) or a functional domain thereof, wherein the ncRNA and the RT or functional domain thereof are as described above.
  • the ncRNA may comprise: (I) an ncRNA listed in Table B, or an ncRNA having at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%%, at least 99.1%, at least 99.2%, at least 99.3%, at least 99.4%, at least 99.5%, at least 99.6%, at least 99.7%, at least 99.8% or at least 99.9% sequence identity to an ncRNA listed in Table B.
  • the reverse transcriptase (RT) or functional domain thereof comprises: (A) i) a polypeptide listed in Table A, or a polypeptide having at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%%, at least 99.1%, at least 99.2%, at least 99.3%, at least 99.4%, at least 99.5%, at least 99.6%, at least 99.7%, at least 99.8% or at least 99.9% sequence identity to a polypeptide listed in Table A; and/or ii) a polypeptide listed in Table C; optionally, the RT does not comprise a polypeptide listed in Table X; OR (B) i) a polynucleotide listed
  • the engineered nucleic acid construct comprises: 1) an msr locus (that encodes the msr RNA portion of an msDNA); 2) an msd locus encoding the msd RNA portion of the msDNA; 3) a sequence encoding a retron reverse transcriptase (RT), wherein said msd RNA is capable of being reverse transcribed to form the msDNA by the retron reverse transcriptase (RT); and, 4) a heterologous nucleic acid inserted at or within the msd locus, upstream of the msr locus, upstream or downstream of the msd locus; wherein the engineered nucleic acid construct is engineered based on and/or to resemble a secondary structure of a wild- type or consensus retron encoding a wild-type or consensus retron ncRNA encompassed by: a) any one of the sequences and/or structures as depicted in any one
  • the engineered nucleic acid construct may comprise one or more sequence modifications (e.g., an insertion, deletion, and/or substitution of one or more nucleotide(s)) in the msr locus and/or the msd locus that: a) modulates (e.g., enhances) reverse transcription, processivity, accuracy/fidelity, and/or production of the msDNA (e.g., in the mammalian cell); b) modulates (e.g., reduces) immunogenicity of ncRNA encoded by the engineered retron (e.g., the msr locus and/or the msd locus) in a host (e.g., a host comprising the mammalian cell); c) modulates (e.g., inhibits, either permanently or transiently) a function of the msDNA; and/or d) modulates (e.g., improves) efficiency of
  • the engineered nucleic acid construct (e.g., the engineered retron) is engineered based on and/or to resemble a secondary structure of a wild-type or consensus retron encoding a wild-type or consensus retron ncRNA encompassed by: a) the sequence of any one of Table B ncRNA sequences and/or the structure depicted in any one of FIGs.2-27; or b) a variant of a), having: i) up to 1, 2, or 3 (e.g., up to 1) nucleotide changes per 10 red lettered-nucleotides; ii) up to 4, 5, or 6 (e.g., up to 1 or 2) nucleotide changes per 10 black lettered-nucleotides; and/or iii) up to 7, 8, or 9 (e.g., up to 3 or 4) nucleotide changes per 10 grey lettered-nucleotides; and/or optionally
  • Another aspect of the disclosure provides a vector system comprising a vector comprising the engineered retron described herein.
  • Another aspect of the disclosure provides an isolated host cell comprising the engineered retron described herein, or the vector system described herein.
  • Another aspect of the disclosure provides a pharmaceutical composition comprising the engineered retron described herein, or the vector system described herein.
  • Another aspect of the disclosure provides a delivery vehicle comprising the engineered retron described herein or the ncRNA encoded by the engineered retron described herein, the vector or vector system described herein, the host cell described herein, or the pharmaceutical composition described herein.
  • kits comprising the engineered retron described herein or the ncRNA encoded by the engineered retron described herein, and optionally instructions for genetically modifying a cell using the engineered retron described herein or the ncRNA encoded by the engineered retron described herein.
  • Another aspect of the disclosure provides a method of modifying a target DNA sequence in a host cell (e.g., a mammalian cell), the method comprising introducing into the host cell (e.g., the mammalian cell) the engineered retron of the invention, the ncRNA encoded by the engineered retron of the invention, or the vector / vector system described herein, to allow the production of the msDNA in the host cell (e.g., mammalian cell), wherein at least a part of the heterologous nucleic acid in the msDNA is integrated into the genome of the host (e.g., mammalian) cell at the target DNA sequence.
  • a host cell e.g., a mammalian cell
  • the target sequence is recognized by a suitable nuclease, such as a CRISPR/Cas effector enzyme, a ZFN, a TALEN, a meganuclease, TnpB, IscB, or a restriction endonuclease (RE), and a double-stranded break (DSB) is created by the nuclease to facilitate / promote the insertion of the part of the heterologous nucleic acid into the target sequence.
  • a suitable nuclease such as a CRISPR/Cas effector enzyme, a ZFN, a TALEN, a meganuclease, TnpB, IscB, or a restriction endonuclease (RE)
  • DSB double-stranded break
  • the target sequence modified / inserted by the part of the heterologous nucleic acid can no longer be recognized by the nuclease to re-create a DSB.
  • Another aspect of the disclosure provides a use of the engineered retron in the various methods described herein.
  • nuclease capable of acting at a target site on a genome (e.g. human genome), such as a CRISPR/Cas effector enzyme, a ZFN, a TALEN, a meganuclease, TnpB, IscB, or a restriction endonuclease (RE); and b) an engineered retron described herein, or an ncRNA encoded thereby, or a vector or a vector system comprising or encoding the same.
  • the nuclease may be linked to one or more element(s) of the engineered retron or the encoded ncRNA.
  • the nuclease may be linked (e.g., fused or conjugated) to the reverse transcriptase of the engineered retron described herein.
  • the nuclease may engage/bind to form a complex with a nucleic acid guide sequence (such as a single-guided RNA of a Cas enzyme), wherein the guide sequence is linked to the ncRNA and/or msDNA of the engineered retron described herein.
  • Another aspect of the disclosure provides an enhanced genome editing system, comprising the genome editing system of the disclosure connected to a biomolecule that modulates host DNA repair, in order to, for example, modulate (e.g., enhance) the incorporation of the heterologous nucleic acid sequence into a genome (e.g., human genome).
  • a biomolecule that modulates host DNA repair in order to, for example, modulate (e.g., enhance) the incorporation of the heterologous nucleic acid sequence into a genome (e.g., human genome).
  • Retrons were originally discovered in 1984 in Myxococcus xanthus bacterium when a short, multi-copy single-stranded DNA (msDNA) that is abundantly present in the bacterial cell was identified. Since then, a number of naturally existing retrons have been found in many prokaryotes such as bacteria.
  • msDNA multi-copy single-stranded DNA
  • retrons encode and transcribe as a single RNA, which comprises a non-coding RNA (ncRNA) portion and a portion encoding a specialized reverse transcriptase (RT).
  • ncRNA non-coding RNA
  • RT specialized reverse transcriptase
  • the retron ncRNA is the precursor of the hybrid molecule that eventually forms, and it initially folds into a typical RNA secondary structure that is recognized by the accompanying RT.
  • the translated RT typically recognizes certain secondary structures in the ncRNA, and binds the RNA template downstream from the msd region.
  • the RT initiates reverse transcription of the RNA towards its 5 ⁇ end, starting from the 2’-end of a conserved guanosine (G) residue found immediately after a double-stranded RNA structure (the a1/a2 region) within the ncRNA.
  • G conserved guanosine
  • a portion of the ncRNA serves as a template for reverse transcription, and reverse transcription terminates before reaching the msr locus.
  • cellular RNase H degrades the segment of the ncRNA that serves as template, but not other parts of the ncRNA.
  • the msDNA remains covalently attached to the RNA template via the 2’-5 ⁇ phosphodiester bond, and base-pairs with the RNA template using the 3 ⁇ end of the msDNA.
  • FIG.1A for a general or typical organization of the retron coding sequence, including the RT encoding sequence and the msr and msd loci, as well as the synthesis of the msDNA by reverse transcription of the initial ncRNA transcript.
  • Many retrons also contain an accessory protein (not depicted in FIG.1A), which may have a variable function that may not be fully understood.
  • the engineered retrons described herein do not comprise the accessory protein naturally associated with the wild-type or template retron.
  • Applicant has discovered, analyzed, and phylogenetically classified 7257 previously unknown retrons from nature based on multiple criteria, including sequence homology and conserved predicted secondary structures, and has grouped these retrons into different phylogenetic clades based on sequence homology and/or conserved predicted secondary structures.
  • Type IA_IIA1 (FIG.2), Type 1B1 (FIG.3), Type IB2 (FIG.4), Type 1C (FIG.5), Type IIA1 other (FIG.6), Type IIA2 (FIG.7), Type IIA3 (FIG.8), Type IIA4 (FIG.9), Type IIA5 (FIG.10), Type IIIA1 (FIG.11), Type IIIA2 (FIG.12), Type IIIA3 (FIG. 13), Type IIIA4 (FIG.14), Type IIIA5 (FIG.15), Type IIIunk (FIG.16), Type IV (FIG.
  • FIG.1B.1 depicts an embodiment of a recombinant retron construct (e.g., a nucleotide sequence cloned into an expression vector) contemplated by the present disclosure.
  • the single thin black line represents a double-stranded nucleotide sequence (e.g., as cloned into an expression vector, such as a plasmid).
  • the recombinant retron is constructed by modifying a starting point retron DNA sequence encoding a ncRNA (the msr/msd region) (such as any one of the herein disclosed 7257 newly discovered retron sequences, and specifically any one of the 7257 ncRNA sequences of Table B.
  • a starting point retron DNA sequence encoding an ncRNA may be modified in any number of ways and can including one modification or more than one modification.
  • the retron DNA may modified to contain at least one nucleotide modification, including a single nucleotide substitution, insertion, or deletion, or a substitution, insertion, or deletion of more than one nucleotide, i.e., up to 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, 69, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83,
  • nucleotide of a starting point retron e.g., a wildtype retron
  • the nucleotides may be contiguous or non-contiguous.
  • an engineered retron as a whole is not naturally-occurring, it may include components such as nucleotide sequences that do occur in nature.
  • an engineered retron can have nucleotide sequences from different organisms (e.g., from different bacteria species), or from completely synthetic / artificial / recombinant nucleic acid sequences.
  • an engineered retron can have a bacterial nucleotide sequence, a human nucleotide sequence, a viral nucleotide sequence, and/or a synthetic / artificial / recombinant nucleotide sequence, and/or combinations of such sequences.
  • modifications of the recombinant retrons disclosed herein include the insertion of a heterologous nucleic acid sequence in a retron, for example, inserted into the ncRNA locus, such as in the msr or the msd loci.
  • Linking guide RNA molecules to the 5 ⁇ and/or 3 ⁇ ends also represents another modification contemplated by the recombinant retrons disclosed herein.
  • the guide RNA molecules may also be categorized or referred to more generally as types of heterologous nucleic acid sequences used to modify starting point retrons. These modifications are depicted in FIG.1B.
  • the DNA encoding the RT may also be modified to obtain a recombinant RT.
  • the RT-encoding DNA may modified to contain at least one nucleotide modification, including a single nucleotide substitution, insertion, or deletion, or a substitution, insertion, or deletion of more than one nucleotide, i.e., up to 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, 69, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, 95
  • Such modifications to the DNA encoding ncRNA and/or RT may modulate the function of the ncRNA and/or RT in various ways, including i) modulating (e.g., enhancing) reverse transcription, processivity, accuracy/fidelity, and/or production of the msDNA (e.g., in the mammalian cell); ii) modulating (e.g., reducing) immunogenicity of ncRNA (msr locus and msd locus) encoded by the engineered retron in a host (e.g., a host comprising the mammalian cell); iii) modulating (e.g., inhibits, either permanently or transiently) a function of the msDNA; and/or iv) modulating (e.g., improving) efficiency of targeted genome editing / engineering.
  • modulating e.g., enhancing
  • reverse transcription e.g., processivity, accuracy/fidelity, and/or production of the msDNA
  • the present disclosure provides recombinant retrons having the general structure of: a) an msr locus; b) an msd locus encoding the msd RNA portion of the msDNA; c) a sequence encoding a retron reverse transcriptase (RT) (optionally in trans to the ncRNA), wherein the msd RNA is capable of being reverse transcribed (e.g., in a host cell such as a mammalian cell) to form an msDNA by the retron reverse transcriptase (RT); d) a heterologous nucleic acid (e.g., heterologous DNA) capable of being transcribed with the msr locus and/or the msd locus, optionally, the heterologous nucleic acid is inserted at or within the msd locus, upstream of the msr locus, upstream or downstream of the msd locus.
  • RT retron reverse transcriptas
  • the engineered retrons of the invention are optionally structurally further modified to include one or more heterologous nucleic acids.
  • the engineered retron may be further modified to provide various functional improvements, such as (without limitation), to enhance the production of msDNA in a cell (e.g., a mammalian cell, including a human cell).
  • a cell e.g., a mammalian cell, including a human cell.
  • the disclosure provides engineered retrons based on their conserved predicted secondary structures, such as those in FIGs.2-27.
  • the disclosure provides engineered retrons based on their sequence identity. Exemplary RT amino acid sequences and ret gene nucleic acid sequences are provided in Table A.
  • exemplary RT consensus amino acid sequences and/or ret gene nucleic acid sequences are provided in Table C.
  • Exemplary ncRNA sequences are provided in Table B.
  • Retron sequences provisoed out of the scope of the invention are provided in Table X.
  • exemplary engineered retrons of the invention (1) are engineered based on or engineered to resemble the secondary structures as depicted in any one of FIGs.2-27, and/or (2) are provided in Table B.
  • Sequences with significant sequence percentage identity are also within the scope of the invention.
  • sequence percentage identity e.g., at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%%, at least 99.1%, at least 99.2%, at least 99.3%, at least 99.4%, at least 99.5%, at least 99.6%, at least 99.7%, at least 99.8% or at least 99.9% sequence identity) are also within the scope of the invention.
  • the engineered nucleic acid construct comprises: 1) an msr locus (that encodes the msr RNA portion of an msDNA); 2) an msd locus encoding the msd RNA portion of the msDNA; 3) a sequence encoding a retron reverse transcriptase (RT), wherein said msd RNA is capable of being reverse transcribed to form the msDNA by the retron reverse transcriptase (RT); and, 4) a heterologous nucleic acid inserted at or within the msd locus, upstream of the msr locus, upstream or downstream of the msd locus; wherein the engineered nucleic acid construct is engineered based on and/or to resemble a secondary structure of a wild- type or consensus retron encoding a wild-type or consensus retron ncRNA encompassed by: a) any one of the sequences and/or structures as depicted in any one
  • the engineered retron of the invention encodes a reverse transcriptase (RT) or a functional domain thereof, comprising: i) a polypeptide listed in Table A, or a polypeptide having at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%%, at least 99.1%, at least 99.2%, at least 99.3%, at least 99.4%, at least 99.5%, at least 99.6%, at least 99.7%, at least 99.8% or at least 99.9% sequence identity to a polypeptide listed in Table A.
  • RT reverse transcriptase
  • the RT does not comprise a polypeptide identified in Table X.
  • the engineered retron of the invention encodes a reverse transcriptase (RT) or a functional domain thereof, comprising: i) a polynucleotide listed in Table A, or a polynucleotide having at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%%, at least 99.1%, at least 99.2%, at least 99.3%, at least 99.4%, at least 99.5%, at least 99.6%, at least 99.7%, at least 99.8% or at least 99.9% sequence identity to a polynucleotide of Table A and/or ii) a consensus
  • the polynucleotide encoding the RT does not comprise a polynucleotide identified in Table X.
  • the engineered retron of the invention encodes an ncRNA comprising: (I) an ncRNA listed in Table B, or an ncRNA having at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%%, at least 99.1%, at least 99.2%, at least 99.3%, at least 99.4%, at least 99.5%, at least 99.6%, at least 99.7%, at least 99.8% or at least 99.9% sequence identity to an ncRNA of Table B.
  • the engineered retron of the invention encodes an ncRNA and a reverse transcriptase (RT) or a functional domain thereof, wherein the ncRNA and the RT or functional domain thereof are as described above.
  • the ncRNA may comprise: (I) an ncRNA listed in Table B, or an ncRNA having at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%%, at least 99.1%, at least 99.2%, at least 99.3%, at least 99.4%, at least 99.5%, at least 99.6%, at least 99.7%, at least 99.8% or at least 99.9% sequence identity to an ncRNA listed in Table B
  • the reverse transcriptase (RT) or functional domain thereof comprises: (A) i) a polypeptide listed in Table A, or a polypeptide having at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%%, at least 99.1%, at least 99.2%, at least 99.3%, at least 99.4%, at least 99.5%, at least 99.6%, at least 99.7%, at least 99.8% or at least 99.9% sequence identity to a polypeptide listed in Table A; and/or ii) a polypeptide listed in Table C; optionally, the RT does not comprise a polypeptide identified in Table X; OR (B) i) a polynucleotide listed
  • the heterologous nucleic acid is between >20 nucleotides and about 10,000 nucleotides.
  • the engineered retron may further comprise a sequence modification (e.g., insertion, deletion, and/or substitution of one or more nucleotide(s)) in the msr locus and/or the msd locus that: i) modulates (e.g., enhances) reverse transcription, processivity, accuracy/fidelity, and/or production of the msDNA (e.g., in the mammalian cell); ii) modulates (e.g., reduces) immunogenicity of ncRNA (msr locus and msd locus) encoded by the engineered retron in a host (e.g., a host comprising the mammalian cell); iii) comprises a nucleotide sequence that modulates (e.g., inhibits, either permanently or transiently) a function of the m
  • Retron msr gene, msd gene, and RT nucleic acid sequences e.g., the ret gene
  • the encoded retron reverse transcriptase protein sequences that may serve as the template of the engineered retron described herein may be derived from any source, such as those in Table A, optionally excluding those associated with the sequences of Table X.
  • template or wild-type (wt) sequences of the msr gene, msd gene, and the RT coding sequence (viz., the ret gene) used in the engineered retron are derived from a bacterial retron.
  • representative template/wild-type retrons are from gram negative bacteria.
  • the retron is from a bacterium listed in Table X.
  • the engineered retrons are engineered based on clades defined on retron/retron RTs, in which the retrons are associated with a tripartite system composed of the ncRNA, the RT and an additional protein or RT-fused domain with diverse enzymatic functions.
  • clades are based primarily upon naturally occurring ncRNA and retron/retron RT, and an additional protein or RT-fused domain, the clades, for the purpose of serving as the templates for the subject engineered retrons, are not limited to naturally occurring sequences.
  • retrons may be considered phylogenetically related based on a Neighbor-Joining algorithm of at least 75% (of at least 1000 replicates) and a Poisson correction distance measurement of no more than 0.05, based on alignment of the retron RT.
  • retrons may be considered phylogenetically related when / if the same RT, or closely related RT, can recognize the secondary structures of the ncRNA of the retrons and reserve transcribe the retrons to produce msDNA.
  • sequence alignments between different retron sequences e.g., ncRNA and/or RT (protein and/or nucleic acid) sequences
  • secondary structure generations are based on software known to one of ordinary skill in the art.
  • the retron ncRNA sequences including msr and msd sequences within the same clade may be highly conserved at certain positions, while being less conserved at other positions.
  • Exemplary consensus sequences based on clade members were generated (see, for example, the corresponding FIGs.2-27, respectively) to show these conserved sequences and/or secondary structures, including the highly conserved nucleotides with at least 97% sequence conservation at red lettered-nucleotides, those with between 90-97% sequence conservation at black lettered-nucleotides, and 75-90% nucleotide sequence identity at grey-lettered nucleotides.
  • the template ncRNA based on which the subject engineered retron is modified is a consensus sequence for the various retron ncRNA (including msr and msd nucleic acid sequences) clades, as provided in any one of SEQ ID NOs: of Table B and the corresponding FIGs.2-27, respectively, including the bases that are highly conserved and depicted by a specific-colored letters or circles, and optionally further including bases that may be present at specific locations by specific-colored circles.
  • the engineered retron is engineered, based on and/or to resemble a secondary structure of a wild-type or consensus retron encoding a wild-type or consensus retron ncRNA encompassed by: 1) the sequences and/or structures as depicted in any one of SEQ ID NOs: of Table B and the corresponding FIGs.2-27, respectively; or 2) a variant of 1), having: A) up to 1, 2, or 3 (e.g., up to 1) nucleotide changes per 10 red lettered- nucleotides; B) up to 4, 5, or 6 (e.g., up to 1 or 2) nucleotide changes per 10 black lettered- nucleotides; and/or C) up to 7, 8, or 9 (e.g., up to 3 or 4) nucleotide changes per 10 grey lettered- nucleotides.
  • the variant of 1) further comprises: a) 7, 8, 9, or 10 (e.g., 9 or 10) nucleotides present per 10 red-circled nucleotides; b) 6, 7, 8, 9, or 10 (e.g., 8, 9 or 10) nucleotides present per 10 black-circled nucleotides; c) 4, 5, 6, 7, 8, 9, or 10 (e.g., 6, 7, 8, 9 or 10) nucleotides present per 10 grey-circled nucleotides; and/or d) 2, 3, 4, 5, 6, 7, 8, 9, or 10 (e.g., 4, 5, 6, 7, 8, 9, or 10) nucleotides present per 10 white-circled nucleotides.
  • the engineered retron may be engineered by introducing the sequence modifications (e.g., deletions, additions, or substitutions) into the wild-type retron encoding wild-type retron ncRNA, or into the retron encoding the consensus retron ncRNA.
  • a variant retron may not satisfy the sequence and/or structural requirements of any one of SEQ ID NOs: of Table B and the corresponding FIGs.2-27, respectively, but may still be a suitable template for the engineered retron described herein, so long as one or more of the conditions set forth in A) – C) and/or a)-d) are met.
  • the highly conserved sequences in the template retrons are preserved / conserved or substantially preserved / conserved in the engineered retron described herein.
  • all or substantially all the red lettered-nucleotides i.e., those conserved in about 97% or more of the retrons in the same clade
  • no more than 1, 2, or 3 e.g., up to 1 nucleotide change(s) (e.g., deleted or substituted) occur per 10 red lettered-nucleotides in the engineered retron described herein.
  • red lettered-nucleotides are changed (e.g., deleted or substituted) in the engineered retron described herein.
  • all or substantially all the black lettered-nucleotides i.e., those conserved in about 90-97% of the retrons in the same clade) are preserved / conserved in the engineered retron described herein.
  • nucleotide change(s) e.g., deleted or substituted
  • nucleotide change(s) e.g., deleted or substituted
  • no more than about 3%, 4%, 5% or 10% of the black lettered-nucleotides are changed (e.g., deleted or substituted) in the engineered retron described herein.
  • all or substantially all the grey lettered-nucleotides are preserved / conserved in the engineered retron described herein.
  • no more than 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, or 15 e.g., up to 3 or 4, or up to 7, 8, or 9 nucleotide change(s) (e.g., deleted or substituted) occur per 10 grey lettered-nucleotides are changed in the engineered retron described herein.
  • no more than about 5%, 10%, 15%, 20%, or 25% of the grey lettered-nucleotides are changed (e.g., deleted or substituted) in the engineered retron described herein.
  • all or substantially all the red circled-nucleotides i.e., those with a nucleotide in about 97% or more of the retrons in the same clade
  • no more than 1, 2, or 3 e.g., 0.3, 0.5, or up to 1 nucleotides are absent (e.g., deleted) per 10 red circled-nucleotides in the engineered retron described herein.
  • nucleotides are present per 10 red circled-nucleotides in the engineered retron described herein. In certain embodiments, no more than about 0.3%, 0.5%, 1%, 2%, 3%, 4%, or 5% of the red circled-nucleotides are absent (e.g., deleted) in the engineered retron described herein. [00269] In certain embodiments, all or substantially all the black circled-nucleotides (i.e., those with a nucleotide in about 90-97% of the retrons in the same clade) are present in the engineered retron described herein.
  • nucleotides are absent (e.g., deleted) per 10 black circled-nucleotides in the engineered retron described herein.
  • 6, 7, 8, 9, or 10 e.g., 8, 9 or 10 nucleotides are present per 10 black circled-nucleotides in the engineered retron described herein.
  • no more than about 1%, 2%, 3%, 5% or 10% of the black circled-nucleotides are absent (e.g., deleted) in the engineered retron described herein.
  • all or substantially all the grey circled-nucleotides are present in the engineered retron described herein.
  • no more than 1, 2, 3, 4, or 5 e.g., up to 2, 3, or 4
  • nucleotides are absent (e.g., deleted) per 10 grey circled-nucleotides in the engineered retron described herein.
  • 4, 5, 6, 7, 8, 9, or 10 e.g., 6, 7, 8, 9 or 10 nucleotides are present per 10 grey circled-nucleotides in the engineered retron described herein.
  • no more than about 5%, 10%, 15%, 20%, or 25% of the grey circled-nucleotides are absent (e.g., deleted) in the engineered retron described herein.
  • all or substantially all the white circled-nucleotides i.e., those with a nucleotide in about 50-75% of the retrons in the same clade
  • no more than 1, 2, 3, 4, 5, 6, or 6 are absent (e.g., deleted) per 10 white circled-nucleotides in the engineered retron described herein.
  • 2, 3, 4, 5, 6, 7, 8, 9, or 10 e.g., 4, 5, 6, 7, 8, 9 or 10 (e.g., 4, 5, 6, 7, 8, 9 or 10) nucleotides are present per 10 grey circled-nucleotides in the engineered retron described herein. In certain embodiments, no more than about 5%, 10%, 15%, 20%, 30%, 40%, or 50% of the white circled-nucleotides are absent (e.g., deleted) in the engineered retron described herein. [00272] In some embodiments, the engineered retron is synthetically produced.
  • the synthetically produced engineered retron comprises the sequences and/or secondary structures as depicted in any one of SEQ ID NOs: of Table B and the corresponding FIGs.2-27, respectively, and at least the conserved color lettered nucleotides according to their respective levels of sequence identity (e.g., red, black and gray letters), and/or at least the conserved colored circle nucleotides according to their respective levels of probability of sequence presence (e.g., red, black and gray circles).
  • the sequence modification in the engineered retrons leads to/results in the encoded retron ncRNA having the desired functional improvement.
  • the one or more sequence modifications comprises, in the ncRNA, one or more of: (i) a modified (e.g., mutated, reduced, or eliminated) bulge in a1, a2, or both a1 and a2; (ii) an extension or shortening of a1, a2, or both a1 and a2; (iii) an extension or shortening of a spacer sequence between hairpin loops (e.g., S1, S2, S3, and/or S4 in FIG.2, or any of the S regions in an one of FIGs.2-27); (iv) an additional or modified (e.g., mutated or eliminated) bulge in hairpin loops (e.g., L2 and/or L3 in FIG.2, or any of the L regions in an one of FIGs.2-27 (e.g., by removing unpaired bases in the bulge, or by replacing unpaired bases with an equivalent number of base pairs)); (v) a modified (e) a modified (e.g.
  • the a1 and a2 regions are both single- stranded and substantially reverse complementary to each other, forming a stem with optional interruption by a symmetric or asymmetric bulge, with optional one or more 5 ⁇ and/or 3 ⁇ overhang/unpaired nucleotide(s), wherein the a1 region generally ends before (e.g., ends immediately 5 ⁇ to) the conserved branching guanosine (G) providing the 2’-OH for reverse transcription priming.
  • G conserved branching guanosine
  • the sequence change comprises a mutated, reduced, or eliminated bulge in the a1/a2 stem region, including sequence change(s) in one (i.e., a1 or a2) strand, or both a1 and a2 strands.
  • the sequence change comprises deleting nucleotides from a1, a2, or both a1 and a2, such that the size of the bulge is reduced, or a symmetrical bulge becomes asymmetrical or vice versa, or a bulge is eliminated.
  • the sequence change comprises replacing/substituting nucleotides in a1, a2, or both a1 and a2, such that previously unpaired bases in the bulge become base-paired.
  • the sequence change comprises replacing an unpaired purine base with one or more unpaired pyrimidine base(s).
  • the sequence change comprises replacing an unpaired pyrimidine base with one or more unpaired purine base(s).
  • the sequence change comprises replacing one unpaired purine base (e.g., A or G) with another unpaired purine base (e.g., G or A, respectively).
  • the sequence change comprises replacing one unpaired pyrimidine base (e.g., T/U or C) with another unpaired pyrimidine base (e.g., C or T/U, respectively).
  • the sequence change comprises an extension or shortening of a1, a2, or both a1 and a2.
  • the length of a1 can be shortened by deleting 5 ⁇ overhang, deleting any upstream bulge nucleotides, deleting bases involved in base-pairing.
  • the length of a1 can be extended by adding 5 ⁇ overhang, adding any upstream bulge nucleotides, adding bases involved in base-pairing.
  • the length of a2 can be shortened by deleting 5 ⁇ overhang, deleting any downstream bulge nucleotides, deleting bases involved in base-pairing.
  • the length of a2 can be extended by adding 5 ⁇ overhang, adding any downstream bulge nucleotides, adding bases involved in base-pairing.
  • the spacer sequences between hairpin loops as depicted in any one of FIGs.2-27, can be extended or shortened.
  • the modification can be by inserting a heterologous nucleic acid sequence in spacer sequences between hairpin loops (e.g., S1, S2, S3 and/or S4 in FIG.2).
  • a heterologous nucleic acid sequence is inserted in a spacer sequence in the msd region.
  • the modification of the spacer region can be by interrupting the spacer with additional bulges or hairpin loops.
  • a bulge in the hairpin loops are mutated or eliminated (e.g., by removing unpaired bases) in the bulge, such that, for example, a symmetric bulge becomes an unsymmetrical bulge, or an unsymmetrical bulge becomes a symmetric one or an even more unsymmetrical one.
  • unpaired bases in the bulge is replaced with an equivalent number of base pairs. The additional base pairs may be merged into the stem at one or both ends of the previous bulge, or may bisect a previous bulge to create two bulges.
  • the length of one or more hairpin loops as depicted in any one of FIGs.2-27, can be extended or shortened.
  • the number of unpaired bases within the tip or loop can be increased or decreased.
  • a heterologous nucleic acid sequence of interest can be inserted within the tip or the hairpin loop.
  • the heterologous nucleic acid sequence of interest is inserted within the tip or the hairpin loop in the msd locus.
  • the GC content in the tip or hairpin loops are increased or decreased.
  • a hairpin loop can be deleted.
  • the ncRNA with the 5 ⁇ end and the 3 ⁇ end of the ncRNA can be circularized by being connected either directly, or via a spacer sequence.
  • one or more hairpin loops e.g. L1, L2, L3 and/or L4 of FIG.2 are modified to have complement, reverse, or reverse complement sequences.
  • the branching guanosine (G) capable of initiating reverse transcription priming is repositioned.
  • the G can be placed further downstream of the end of the a1 sequence by, for example, 1, 2, 3, 4, or 5 additional nucleotides.
  • the one or more heterologous nucleic acid sequences comprise: a) a heterologous nucleic acid (such as the coding sequence for an RNA aptamer or a ribozyme) inserted into the msr locus or the msd locus (such as in an S region (e.g., S1, S2, S3 and/or S4 in FIG.2, or any of the S regions in any one of FIGs.2-27), or the tip of an L region (e.g., L1, L2, L3 and/or L4 in FIG.2, or any of the L regions in any one of FIGs.2-27), or upstream or downstream of either the msr locus or the
  • an antisense sequence complementary to a CRISPR/Cas guide RNA (gRNA) sequence encoded by the heterologous nucleic acid can be included, wherein the antisense sequence hybridizes to and inhibits the gRNA in the encoded retron ncRNA, and wherein the antisense sequence is removed upon reverse transcription of the msDNA.
  • gRNA CRISPR/Cas guide RNA
  • said heterologous nucleic acid encodes a protein or peptide of interest, or wherein said heterologous nucleic acid comprises or encodes a donor / template sequence (e.g., a donor that corrects / repairs / removes a mutation at the target genome site, such as a mutated exon in a disease gene; a functional DNA element (such as a promoter, an enhancer, a protein binding sequence, a methylation site, a homology region for assisting gene editing, etc.); or a coding sequence for a functional RNA element (ncRNAs, etc.)).
  • a donor / template sequence e.g., a donor that corrects / repairs / removes a mutation at the target genome site, such as a mutated exon in a disease gene
  • a functional DNA element such as a promoter, an enhancer, a protein binding sequence, a methylation site, a homology region for assisting gene editing, etc.
  • the protein or peptide of interest comprises a therapeutic protein (such as a wildtype protein defective in a disease cell, or a therapeutic antibody or antigen-binding fragment thereof) useful in treating a disease.
  • a therapeutic protein such as a wildtype protein defective in a disease cell, or a therapeutic antibody or antigen-binding fragment thereof.
  • Other heterologous nucleic acids of the invention are described in other section of the specification, all incorporated herein by reference.
  • the template/wild-type retron for the engineered retron encodes a wild-type or consensus retron ncRNA polynucleotide having a consensus secondary structure shown in any one of FIGs.2-27, which are described individually below:
  • Variants of this template include a variant having: A) up to 1, 2, or 3 (e.g., up to 1) nucleotide changes per 10 red lettered-nucleotides; B) up to 4, 5, or 6 (e.g., up to 1 or 2) nucleotide changes per 10 black lettered-nucleotides; and/or C) up to 7, 8, or 9 (e.g., up to 3 or 4) nucleotide changes per 10 grey lettered-nucleotides; and/or optionally further comprising: a) 7, 8, 9, or 10 (e.g., 9 or 10) nucleotides present per 10 red-
  • the engineered retron is entirely synthetically produced and having the conserved nucleotides as denoted by the colored letters as shown in SEQ ID NO. XX and FIGs.2-27.
  • the non-coding RNA (ncRNA) portion of the engineered retron comprises a polynucleotide (e.g., a DNA molecule) encoding an ncRNA listed in Table B, or an ncRNA having at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%%, at least 99.1%, at least 99.2%, at least 99.3%, at least 99.4%, at least 99.5%, at least 99.6%, at least 99.7%, at
  • the ncRNA does not comprise an ncRNA associated with the sequences of Table X.
  • Amplification of an engineered retron described herein may be performed, for example, before transfection of cells or ligation into vectors. Any method for amplifying the engineered retron may be used, including, but not limited to polymerase chain reaction (PCR), isothermal amplification, nucleic acid sequence-based amplification (NASBA), transcription mediated amplification (TMA), strand displacement amplification (SDA), and ligase chain reaction (LCR).
  • PCR polymerase chain reaction
  • NASBA nucleic acid sequence-based amplification
  • TMA transcription mediated amplification
  • SDA strand displacement amplification
  • LCR ligase chain reaction
  • the engineered retron comprise common 5 ⁇ and 3 ⁇ priming sites to allow amplification of retron sequences in parallel with a set of universal primers.
  • a set of selective primers is used to selectively amplify a subset of retron sequences from a pooled mixture.
  • the template/wild-type retron for the engineered retron encodes a wild-type or consensus retron ncRNA polynucleotide having a consensus secondary structure shown in FIG.2-27, and as described individually below: Type IA/IIA1 retron (FIG.2)
  • the template/wt retron for the subject engineered retron encodes a wild-type retron ncRNA polynucleotide having a consensus secondary structure that can be described as: a1-S1-L1-L2-S2-L3-S3-a2-S4-L4, wherein: a1/a2 is a stem 8 bp in length; L1 is a stem of 5 bps with a 10-nt tip; L2 is a stem of 7 bps with a 5-nt tip, and
  • the engineered retron is entirely synthetically produced and having the conserved nucleotides as denoted by the colored letters as shown in FIG.2.
  • Type IB1 retron (FIG.3)
  • the template/wt retron for the subject engineered retron encodes a wild-type retron ncRNA polynucleotide having a consensus secondary structure that can be described as: a1-S1-L1-S2-L2-S3-a2, wherein: a1/a2 is a stem 6 bps in length with a 2/2 bulge 3 bps from the tip, wherein a1 has a 2-nt overhang and a2 has a 6-nt overhang; L1 is a stem of 14 bps with a 3-nt tip, a 1/0 bulge 4 bps from the tip, and a 0/6 bulge 10 bps from the tip; L2 is a stem of 23 bps with a 5-nt tip, a 1
  • the engineered retron is entirely synthetically produced and having the conserved nucleotides as denoted by the colored letters as shown in FIG.3.
  • Type IB2 retron (FIG.4)
  • the template/wt retron for the subject engineered retron encodes a wild-type retron ncRNA polynucleotide having a consensus secondary structure that can be described as: a1-S1-L1-L2-S2-L3-S3-L4-S4-a2, wherein: the a1/a2 stem is 16 bp in length, with a 17-base 5 ⁇ overhang and a 16-base 3 ⁇ overhang; L1 is a stem of 6 bps with a 4-nt tip; L2 is a stem of 4 bps with a 4-nt tip, with a 2/2 bulge 2 nts from the tip; L3 is a stem of 3 bps with a 5-nt tip; L4 is a stem of 9
  • the engineered retron is entirely synthetically produced and having the conserved nucleotides as denoted by the colored letters as shown in FIG.4.
  • Type 1C retron (FIG.5)
  • the template/wt retron for the subject engineered retron encodes a wild-type retron ncRNA polynucleotide having a consensus secondary structure that can be described as: a1-S1-L1-S2-L2-S3-a2, wherein: a1/a2 is a stem 13 bps in length; L1 is a stem of 9 bps with a 3-nt tip; L2 is a stem of 10 bps with a 5-nt tip; S1 is a single-stranded spacer region between a1/a2 and L1; S2 is a single-stranded spacer region between L1 and L2; S3 is a single-stranded spacer region between L2 and a1/a2, the conserved nucleot
  • the engineered retron is entirely synthetically produced and having the conserved nucleotides as denoted by the colored letters as shown in FIG.5.
  • Type IIA1 retron (FIG.6)
  • the template/wt retron for the subject engineered retron encodes a wild-type retron ncRNA polynucleotide having a consensus secondary structure that can be described as: a1-S1-L1-S2-L2-S3-L3-S4-a2, wherein: a1/a2 is a stem 10 bps in length with a 1-nt overhang on a2; L1 is a stem of 10 bps with a 3-nt tip; L2 is a stem of 7 bps with a 5-nt tip; L3 is a stem of 27 bps with a 8-nt tip and a 0/2 bulge 26 bps from the tip; S1 is a single-stranded spacer region between a1/a2 and
  • the engineered retron is entirely synthetically produced and having the conserved nucleotides as denoted by the colored letters as shown in FIG.6.
  • Type IIA2 retron (FIG.7)
  • the template/wt retron for the subject engineered retron encodes a wild-type retron ncRNA polynucleotide having a consensus secondary structure that can be described as: a1-S1-L1-S2-L2-L3-S3-a2, wherein: a1/a2 is a stem 7 bp in length with no overhangs; L1 is a stem of 8 bps with a 3-nt tip; L2 is a stem of 30 bps with a 8-nt tip, a 1/1 bulge 2 bps from the tip, and a 1/1 bulge 27 bps from the tip; L3 is a stem of 8 bps with a 5-nt tip, and a 0/1 bulge 3 nt from the tip;
  • the engineered retron is entirely synthetically produced and having the conserved nucleotides as denoted by the colored letters as shown in FIG.7.
  • Type IIA3 retron (FIG.8)
  • the template/wt retron for the subject engineered retron encodes a wild-type retron ncRNA polynucleotide having a consensus secondary structure that can be described as: a1-S1-L1-L2-S2-L3-S3-a2, wherein: a1/a2 is a stem 6 bps in length; L1 is a stem of 8 bps with a 9-nt tip; L2 is a stem of 8 bps with a 3-nt tip; S1 is a single-stranded spacer region between a1/a2 and L1; S2 is a single-stranded spacer region between L2 and L3; S3 is a single-stranded spacer region between L3 and a1/a2, the
  • the engineered retron is entirely synthetically produced and having the conserved nucleotides as denoted by the colored letters as shown in FIG.8.
  • Type IIA4 retron (FIG.9)
  • the template/wt retron for the subject engineered retron encodes a wild-type retron ncRNA polynucleotide having a consensus secondary structure that can be described as: a1-a2-L1-S1-L2-L3-S2-L4-S3, wherein: a1/a2 is a stem 3 bp in length with no overhangs and a 7-nt tip; L1 is a stem of 7 bps with a 3-nt tip; L2 is a stem of 6 bps with a 4-nt tip; L3 is a stem of 40 bps with a 5-nt tip, and a 2/2 bulge 3 bps from the tip, a 5/4 bulge 10 bps from the tip, and a 12/15 bulge
  • the engineered retron is entirely synthetically produced and having the conserved nucleotides as denoted by the colored letters as shown in FIG.9.
  • Type IIA5 novel retron (FIG.10)
  • the template/wt retron for the subject engineered retron encodes a wild-type retron ncRNA polynucleotide having a consensus secondary structure that can be described as: a1-S1-L1-S2-L2-S3-L3-S4-a2, wherein: a1/a2 is a stem 15 bps in length with a 1-nt overhang on a1, a 13-nt overhang on a2, and a 7/5 bulge 13-nt from the tip; L1 is a stem of 10 bps with a 3-nt tip; L2 is a stem of 35 bps with a 3-nt tip; L3 is a stem of 6 bps with a 5-nt tip; S1 is a single-strand
  • the engineered retron is entirely synthetically produced and having the conserved nucleotides as denoted by the colored letters as shown in FIG.10.
  • Type IIIA1 retron (FIG.11)
  • the template/wt retron for the subject engineered retron encodes a wild-type retron ncRNA polynucleotide having a consensus secondary structure that can be described as: a1-S1-L1-L2-S2-L3-S3-a2, wherein: a1/a2 is a stem 2 bps in length with a 1-nt overhang on a2; L1 is a stem of 8 bps with a 4-nt tip; L2 is a stem of 9 bps with a 3-nt tip and a 1/1 bulge 3 bps from the tip; L3 is a stem of 20 bps with a 3-nt tip and a 1/2 bulge 3 bps from the tip; S1 is a single-stranded spacer
  • the engineered retron is entirely synthetically produced and having the conserved nucleotides as denoted by the colored letters as shown in FIG.11.
  • Type IIIA2 retron (FIG.12)
  • the template/wt retron for the subject engineered retron encodes a wild-type retron ncRNA polynucleotide having a consensus secondary structure that can be described as: a1-S1-L1-S2-L2-S3-L3-S4-L4-S5-a2, wherein: a1/a2 is a stem 15 bp in length; L1 is a stem of 6 bps with a 4-nt tip; L2 is a stem of 13 bps with a 5-nt tip; L3 is a stem of 4 bps with a 8-nt tip; L4 is a stem of 20 bps with a 4-nt tip and a 2/2 bulge 6 bp from the tip; S1 is a single
  • the engineered retron is entirely synthetically produced and having the conserved nucleotides as denoted by the colored letters as shown in FIG.12.
  • Type IIIA3 retron (FIG.13)
  • the template/wt retron for the subject engineered retron encodes a wild-type retron ncRNA polynucleotide having a consensus secondary structure that can be described as: a1-S1-L1-S2-L2-S3-L3-S4-L4-L5-L6-S5-a2, wherein: a1/a2 is a stem 24 bps in length and having a 1/0 bulge 15 bps from the tip, and a 1/1 bulge 19 bps from the tip; L1 is a stem of 7 bps with a 4-nt tip; L2 is a stem of 9 bps with a 8-nt tip; L3 is a stem of 8 bps with a 4-nt tip; L4 is a
  • the engineered retron is entirely synthetically produced and having the conserved nucleotides as denoted by the colored letters as shown in FIG.13.
  • Type IIIA4 retron (FIG.14)
  • the template/wt retron for the subject engineered retron encodes a wild-type retron ncRNA polynucleotide having a consensus secondary structure that can be described as: a1-S1-L1-S2-L2-S3-L3-S4-a2, wherein: a1/a2 is a stem 5 bps in length with a 1/2 bulge 2 bps from the tip; L1 is a stem of 8 bps with a 6-nt tip; L2 is a stem of 8 bps with a 5-nt tip; L3 is a stem of 13 bps with a 14-nt tip and a 1/0 bulge 2 bps from the tip; S1 is a single-stranded spacer region between a1/a2
  • the engineered retron is entirely synthetically produced and having the conserved nucleotides as denoted by the colored letters as shown in FIG.14.
  • Type IIIA5 retron (FIG.15)
  • the template/wt retron for the subject engineered retron encodes a wild-type retron ncRNA polynucleotide having a consensus secondary structure that can be described as: a1-S1-L1-S2-L2-L3-L4-S3-a2, wherein: the a1/a2 stem is 11 bp in length with no overhang; L1 is a stem of 9 bps with a 3-nt tip; L2 is a stem of 14 bps with a 5-nt tip; L3 is a stem of 9 bps with a 7-nt tip; L4 is a stem of 15 bps with a 7-nt tip; S1, S2, and S3 are single-stranded spacer regions between the a1/
  • the engineered retron is entirely synthetically produced and having the conserved nucleotides as denoted by the colored letters as shown in FIG.15.
  • Type IIIunk retron (FIG.16)
  • the template/wt retron for the subject engineered retron encodes a wild-type retron ncRNA polynucleotide having a consensus secondary structure that can be described as: a1-S1-L1-S2-L2-S3-L3-S4-a2, wherein: a1/a2 is a stem 11 bps in length; L1 is a stem of 12 bps with a 2-nt tip; L2 is a stem of 21 bps with a 1-nt tip; L3 is a stem of 20 bps with a 4-nt tip; S1 is a single-stranded spacer region between a1/a2 and L1; S2 is a single-stranded spacer region between L1 and L2; S3 is a
  • the engineered retron is entirely synthetically produced and having the conserved nucleotides as denoted by the colored letters as shown in FIG.16.
  • Type IV retron (FIG.17)
  • the template/wt retron for the subject engineered retron encodes a wild-type retron ncRNA polynucleotide having a consensus secondary structure that can be described as: a1-S1-L1-L2-S2-L3-S3-a2, wherein: a1/a2 is a stem 9 bp in length with no overhang; L1 is a stem of 5 bps with a 6-nt tip; L2 is a stem of 9 bps with a 4-nt tip; L3 is a stem of 26 bps with a 5-nt tip, a 0/1 bulge 7 bps from the tip, and a 0/1 bulge 9 bps from the tip; S1 is a single-stranded spacer region between the
  • the engineered retron is entirely synthetically produced and having the conserved nucleotides as denoted by the colored letters as shown in FIG.17.
  • Type IX retron (FIG.18)
  • the template/wt retron for the subject engineered retron encodes a wild-type retron ncRNA polynucleotide having a consensus secondary structure that can be described as: a1-L1-S1-L2-S2-a2, wherein: a1/a2 is a stem 12 bp in length, wherein a1 has a 14-nt overhang and a2 has a 2-nt overhang; L1 is a stem of 11 bps with a 3-nt tip and a 1/3 bulge 7 bp from the tip; L2 is a stem of 25 bps with a 7-nt tip; S1 is a single-stranded spacer region between L1 and L2; S2 is a single-stranded space
  • the engineered retron is entirely synthetically produced and having the conserved nucleotides as denoted by the colored letters as shown in FIG.18.
  • Type V retron (FIG.19)
  • the template/wt retron for the subject engineered retron encodes a wild-type retron ncRNA polynucleotide having a consensus secondary structure that can be described as: a1-S1-L1-S2-L2-S3-a2, wherein: a1/a2 is a stem 13 bps in length; L1 is a stem of 20 bps with a 4-nt tip and a 6/4 bulge 6 bps from the tip; L2 is a stem of 14 bps with a 4-nt tip and a 1/0 bulge 5 bps from the tip; S1 is a single-stranded spacer region between a1/a2 and L1; S2 is a single-stranded spacer region between L1 and L2; S3
  • the engineered retron is entirely synthetically produced and having the conserved nucleotides as denoted by the colored letters as shown in FIG.19.
  • Type VI retron (FIG.20)
  • the template/wt retron for the subject engineered retron encodes a wild-type retron ncRNA polynucleotide having a consensus secondary structure that can be described as: a1-S1-L1-S2-L2-S3-L3-L4-S4-a2, wherein: a1/a2 is a stem 4 bp in length with a 1 bp 5 ⁇ overhang; L1 is a stem of 7 bps with a 4-nt tip; L2 is a stem of 8 bps with a 4-nt tip; L3 is a stem of 16 bps with a 6-nt tip, a 3/4 bulge 3 bps from the tip, a 2/3 bulge 5 bps from the tip, and a 3/1 bulge
  • the engineered retron is entirely synthetically produced and having the conserved nucleotides as denoted by the colored letters as shown in FIG.20.
  • Type XI Group 1 retron (FIG.21)
  • the template/wt retron for the subject engineered retron encodes a wild-type retron ncRNA polynucleotide having a consensus secondary structure that can be described as: a1-S1-L1-S2-L2-S3-a2, wherein: a1/a2 is a stem 16 bps in length with a 5-nt overhang on a1, and a 3-nt overhang on a2; L1 is a stem of 9 bps with a 3-nt tip; L2 is a stem of 7 bps with a 13-nt tip; S1 is a single-stranded spacer region between a1/a2 and L1; S2 is a single-stranded spacer region between L1 and L2
  • the engineered retron is entirely synthetically produced and having the conserved nucleotides as denoted by the colored letters as shown in FIG.21.
  • Type XI Group 2 retron (FIG.22)
  • the template/wt retron for the subject engineered retron encodes a wild-type retron ncRNA polynucleotide having a consensus secondary structure that can be described as: a1-S1-L1-S2-L2-S3-a2, wherein: a1/a2 is a stem 13 bps in length with a 1-nt overhang on a2; L1 is a stem of 7 bps with a 3-nt tip and a 2/2 bulge 1 bp from the tip; L2 is a stem of 8 bps with a 20-nt tip; S1 is a single-stranded spacer region between a1/a2 and L1; S2 is a single-stranded spacer region between L1 and
  • the engineered retron is entirely synthetically produced and having the conserved nucleotides as denoted by the colored letters as shown in FIG.22.
  • Type XII retron (FIG.23)
  • the template/wt retron for the subject engineered retron encodes a wild-type retron ncRNA polynucleotide having a consensus secondary structure that can be described as: a1-S1-L1-S2-L2-S3-a2, wherein: a1/a2 is a stem 13 bps in length with a 1-nt overhang on a2; L1 is a stem of 7 bps with a 3-nt tip, and a 2/2 bulge 1 bp from the tip; L2 is a stem of 8 bps with a 19-nt tip; S1 is a single-stranded spacer region between a1/a2 and L1; S2 is a single-stranded spacer region between L1 and
  • the engineered retron is entirely synthetically produced and having the conserved nucleotides as denoted by the colored letters as shown in FIG.23.
  • Type XIII retron (FIG.24)
  • the template/wt retron for the subject engineered retron encodes a wild-type retron ncRNA polynucleotide having a consensus secondary structure that can be described as: a1-S1-L1-S2-L2-L3-S3-a2, wherein: a1/a2 is a stem 7 bp in length with no overhangs; L1 is a stem of 8 bps with a 3-nt tip; L2 is a stem of 30 bps with a 8-nt tip, a 1/1 bulge 2 bps from the tip, and a 1/1 bulge 27 bps from the tip; L3 is a stem of 8 bps with a 5-nt tip, and a 0/1 bulge 3 nt from the tip
  • the engineered retron is entirely synthetically produced and having the conserved nucleotides as denoted by the colored letters as shown in FIG.24.
  • Type XIV retron (FIG.25)
  • the template/wt retron for the subject engineered retron encodes a wild-type retron ncRNA polynucleotide having a consensus secondary structure that can be described as: a1-S1-L1-L2-S2-L3-S3-a2, wherein: the a1/a2 stem is 15 bp in length with no overhang, and a 4/2 bulge 7 bps from the 5 ⁇ end of a1; L1 is a stem of 8 bps with a 5-nt tip; L2 is a stem of 7 bps with a 5-nt tip; L3 is a stem of 13 bps with a 2-nt tip, a 5/9 bulge 5 bps from the tip, and a 5/5 bulge 8 bps
  • the engineered retron is entirely synthetically produced and having the conserved nucleotides as denoted by the colored letters as shown in FIG.25.
  • Ec107-like retron (FIG.26)
  • the template/wt retron for the subject engineered retron encodes a wild-type retron ncRNA polynucleotide having a consensus secondary structure that can be described as: a1-S1-L1-S2-L2-S3-L3-S4-a2, wherein: a1/a2 is a stem 12 bp in length; L1 is a stem of 4 bps with a 8-nt tip; L2 is a stem of 8 bps with a 3-nt tip; L3 is a stem of 22 bps with a 3-nt tip, a 4/6 bulge 6 bp from the tip, a 3/3 bulge 13 bp from the tip, and a 1/1 bulge 18 bp from the tip;
  • the engineered retron is entirely synthetically produced and having the conserved nucleotides as denoted by the colored letters as shown in FIG.26.
  • Outgroup A retron (FIG.27)
  • the template/wt retron for the subject engineered retron encodes a wild-type retron ncRNA polynucleotide having a consensus secondary structure that can be described as: a1-S1-L1-S2-L2-S3-L3-S4-a2, wherein: a1/a2 is a stem 5 bps in length with a 2-nt overhang on a2; L1 is a stem of 11 bps with a 4-nt tip; L2 is a stem of 8 bps with a 3-nt tip; L3 is a stem of 18 bps with a 3-nt tip; S1 is a single-stranded spacer region between a1/a2 and L1; S2 is a single-stranded space
  • the engineered retron is entirely synthetically produced and having the conserved nucleotides as denoted by the colored letters as shown in FIG.27.
  • Amplification of an engineered retron described herein may be performed, for example, before transfection of cells or ligation into vectors. Any method for amplifying the engineered retron may be used, including, but not limited to polymerase chain reaction (PCR), isothermal amplification, nucleic acid sequence-based amplification (NASBA), transcription mediated amplification (TMA), strand displacement amplification (SDA), and ligase chain reaction (LCR).
  • PCR polymerase chain reaction
  • NASBA nucleic acid sequence-based amplification
  • TMA transcription mediated amplification
  • SDA strand displacement amplification
  • LCR ligase chain reaction
  • the engineered retron comprise common 5 ⁇ and 3 ⁇ priming sites to allow amplification of retron sequences in parallel with a set of universal primers.
  • a set of selective primers is used to selectively amplify a subset of retron sequences from a pooled mixture.
  • Variants of these templates include a variant having: A) up to 1, 2, or 3 (e.g., up to 1) nucleotide changes per 10 red lettered-nucleotides; B) up to 4, 5, or 6 (e.g., up to 1 or 2) nucleotide changes per 10 black lettered-nucleotides; and/or C) up to 7, 8, or 9 (e.g., up to 3 or 4) nucleotide changes per 10 grey lettered-nucleotides; and/or optionally further comprising: a) 7, 8, 9, or 10 (e.g., 9 or 10) nucleotides present per 10 red-circled nucleotides; b) 6, 7, 8, 9, or 10 (e.g., 8, 9 or 10) nucleotides present per 10 black-circled nucleotides; c) 4, 5, 6, 7, 8, 9, or 10 (e.g., 6, 7, 8, 9
  • the non-coding RNA (ncRNA) portion of the engineered retron comprises a polynucleotide (e.g., a DNA molecule) encoding an ncRNA listed in Table B, or an ncRNA having at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%%, at least 99.1%, at least 99.2%, at least 99.3%, at least 99.4%, at least 99.5%, at least 99.6%, at least 99.7%, at least 99.8% or at least 99.9% sequence identity to an ncRNA listed in Table B.
  • a polynucleotide e.g., a DNA molecule
  • the ncRNA does not comprise an ncRNA associated with the RT sequences of Table X.
  • the engineered retron may comprise or encodes a heterologous nucleic acid (e.g., DNA or RNA) within the msr locus or the msd locus (such as in an S region or the tip of an L region in the consensus structure in any one of FIGs.2-27, and variants thereof), or upstream or downstream of either the msr locus or the msd locus.
  • the heterologous nucleic acid is inserted within the msd locus.
  • the heterologous nucleic acid is inserted upstream of the msr locus. In some embodiments, the heterologous nucleic acid is inserted upstream or downstream of the msd locus. In other embodiments, the heterologous nucleic acid is inserted in spacer sequences (e.g., S1, S2, S3 and/or S4 as depicted in FIG.2) between hairpin loops and/or within hairpin loops (e.g., L1, L2, L3 and/or L4 as depicted in FIG. 2), e.g., the tip or loop region, or within a bulge.
  • spacer sequences e.g., S1, S2, S3 and/or S4 as depicted in FIG.2
  • hairpin loops e.g., L1, L2, L3 and/or L4 as depicted in FIG. 2
  • the heterologous nucleic acid sequence can be inserted in the tip of an L region or hairpin loop (e.g., L1, L2, L3 and/or L4 as depicted in FIG.2).
  • one or more heterologous nucleic acids are inserted into the msd locus.
  • a first heterologous nucleic acid is inserted into the msd locus, and a second heterologous nucleic acid is inserted either upstream of the msr locus or upstream or downstream of the msd locus.
  • the heterologous nucleic acid comprises flanking contiguous nucleotides (also referred to as homologous arms) that are substantially complementary to a target site genomic sequence of a cell to facilitate insertion of at least part of the heterologous nucleic acid into the genome of the cell at the target site via homology directed repair (HDR).
  • the heterologous nucleic acid is between >20 nucleotides and 10,000 nucleotides (e.g., including the flanking homologous arms).
  • one or both of the homology arm(s) on the heterologous nucleic acid are 100% identical to a target genomic sequence.
  • one or both of the homology arm(s) on the heterologous nucleic acid are less than 100% complementary to the target genomic sequence, for example, 99%, 98%, 97%, 96%, 95%, 94%, 93%, 92%, 91%, or 90% identical to a target genomic sequence.
  • the part of the heterologous nucleic acid to be inserted into the target genome sequence is sometimes referred to as a donor sequence.
  • the donor sequence may be partially identical to the full length or portions thereof of a target genomic sequence, or may be unrelated to the target genomic sequence.
  • the donor sequence may be used, for example, to introduce modifications (e.g., substitutions, deletions, insertions, or a combination thereof) such as mutations or other genetic changes (e.g., genetic elements such as stop codons or a shift in an open reading frame on the target polynucleotide) into its target sequence.
  • modifications e.g., substitutions, deletions, insertions, or a combination thereof
  • genetic changes e.g., genetic elements such as stop codons or a shift in an open reading frame on the target polynucleotide
  • the heterologous nucleic acid sequence is or encodes a biologically active molecule such as, but not limited to, a therapeutic protein.
  • Any therapeutic proteins may be encoded by the heterologous nucleic acid.
  • the heterologous nucleic acid sequences encodes one or more prophylactically- or therapeutically-active proteins, polypeptides, or other factors.
  • the heterologous sequences may be or encode an agent that enhances tumor killing activity such as, but not limited to, TRAIL or tumor necrosis factor (TNF), in a cancer.
  • the heterologous sequences may be or encode an agent suitable for the treatment of conditions such as muscular dystrophy (e.g., heterologous sequences are or encode dystrophin or a functional fragment or variant thereof such as the numerous dystrophin minigenes or microdystrophine coding sequences known in the art), cardiovascular disease (e.g., heterologous sequences are or encode SERCA2a, GATA4, Tbx5, Mef2C, Hand2, Myocd, etc.), neurodegenerative disease (e.g., heterologous sequences is or encodes NGF, BDNF, GDNF, NT-3, etc.).
  • the heterologous nucleic acid sequence may be or encode an agent that enhances tumor killing activity such as, but not limited to, TRAIL or tumor necrosis factor (TNF), in a cancer.
  • the heterologous nucleic acid sequence may be or encode an agent suitable for the treatment of conditions such as muscular dystrophy (e.g., heterologous nucleic acid sequence is or encodes Dystrophin), cardiovascular disease (e.g., heterologous nucleic acid sequence is or encodes SERCA2a, GATA4, Tbx5, Mef2C, Hand2, Myocd, etc.), neurodegenerative disease (e.g., heterologous nucleic acid sequence is or encodes NGF, BDNF, GDNF, NT-3, etc.), chronic pain (e.g., heterologous nucleic acid sequence is or encodes GlyRal), an enkephalin, or a glutamate decarboxylase (e.g., heterologous
  • the heterologous nucleic acid sequence is or encodes a factor that can affect the differentiation of a cell.
  • the expression of one or more of Oct4, Klf4, Sox2, c-Myc, L-Myc, dominant-negative p53, Nanog, Glisl, Lin28, TFIID, mir-302/367, or other miRNAs can cause the cell to become an induced pluripotent stem (iPS) cell.
  • iPS induced pluripotent stem
  • the heterologous nucleic acid sequence is or encodes a factor for transdifferentiating cells.
  • Non-limiting examples of factors include: one or more of GATA4, Tbx5, Mef2C, Myocd, Hand2, SRF, Mespl, SMARCD3 for cardiomyocytes; Ascii, Nurrl, LmxlA, Bm2, Mytll, NeuroDl, FoxA2 for neural cells; and Hnf4a, Foxal, Foxa2 or Foxa3 for hepatic cells.
  • the heterologous nucleic acid encodes a therapeutic antibody, or an antigen-binding fragment thereof.
  • the heterologous nucleic acid encodes a protein for replacement therapy.
  • the protein may be defective in a disease cell or a disease organism / individual, and the wild-type protein or a functional fragment or variant thereof, when delivered by the heterologous nucleic acid to the disease cell / tissue / organism / individual, at least partially or fully restores the lost function of the protein the disease cell / tissue / organism / individual.
  • HNS donor DNA template
  • the heterologous nucleic acid sequence is a donor DNA template that can be integrated into a host genome via HDR.
  • the heterologous nucleic acid sequence is a donor DNA that can serve as a template or primer for recombineering during replication.
  • the heterologous nucleic acid comprises or encodes a donor / template sequence, wherein the donor / template corrects / repairs / removes a mutation at the target genome site.
  • the mutation may be a mutated exon in a disease gene.
  • the donor / template may encode or comprises a functional DNA element, such as a promoter, an enhancer, a protein binding sequence, a methylation site, or a homology region for assisting gene editing, etc.
  • donor DNA or “donor DNA template” it is meant a single-stranded DNA to be inserted at a site cleaved by a gene-editing nuclease (e.g., a CRISPR/Cas effector protein; a TALEN; a ZFN) (e.g., after dsDNA cleavage, after nicking a target DNA, after dual nicking a target DNA, and the like).
  • the donor DNA template can contain sufficient homology to a genomic sequence at the target site, e.g., 70%, 80%, 85%, 90%, 95%, or 100% homology with the nucleotide sequences flanking the target site, e.g.
  • Donor DNA template can be of any length, e.g., 50 nucleotides or more, 100 nucleotides or more, 250 nucleotides or more, 500 nucleotides or more, 1000 nucleotides or more, 5000 nucleotides or more, etc.
  • a suitable donor DNA template can be from 50 nucleotides to 100 nucleotides, from 100 nucleotides to 500 nucleotides, from 500 nucleotides to 1000 nucleotides, from 1000 nucleotides to 5000 nucleotides, or from 5000 nucleotides to 10,000 nucleotides, or more than 10,000 nucleotides, in length.
  • the donor DNA template comprises a first homology arm and a second homology arm.
  • the first homology arm is at or near the 5’ end of the donor DNA; and comprises a nucleotide sequence that is at least partially complementary to a first nucleotide sequence in a target nucleic acid.
  • the second homology arm is at or near the 3’ end of the donor DNA; and comprises a nucleotide sequence that is at least partially complementary to a second nucleotide sequence in the target nucleic acid.
  • the first and second homology arms can each independently have a length of from about 10 nucleotides to 400 nucleotides; e.g., from 10 nucleotides (nt) to 15 nt, from 15 nt to 20 nt, from 20 nt to 25 nt, from 25 nt to 30 nt, from 30 nt to 35 nt, from 35 nt to 40 nt, from 40 nt to 45 nt, from 45 nt to 50 nt, from 50 nt to 75 nt, from 75 nt to 100 nt, from 100 nt to 125 nt, from 125 nt to 150 nt, from 150 nt to 175 nt, from 175 nt to 200 nt, from 200 nt to 225 nt, from 225 nt to 250 nt, from 250 nt to 275 nt, from 275 nt to 300 nt, from 325 n
  • the donor DNA template is used for editing the target nucleotide sequence.
  • the donor DNA template comprises one or more mutations to be introduced into the target polynucleotide. Examples of such mutations include substitutions, deletions, insertions, or a combination thereof.
  • the mutation causes a shift in an open reading frame on the target polynucleotide.
  • the donor polynucleotide alters a stop codon in the target polynucleotide. In certain embodiments, the donor polynucleotide corrects a premature stop codon.
  • the correction can be achieved by deleting the stop codon, or by introducing one or more sequence changes to alter the stop codon to a codon.
  • the donor polynucleotide addresses loss of function mutations, deletions, or translocations that may occur, for example, in certain disease contexts by inserting or restoring a functional copy of a gene, or functional fragment thereof, or a functional regulatory sequence or functional fragment of a regulatory sequence.
  • a functional fragment includes a fragment less than the entire copy of a gene but otherwise provides sufficient nucleotide sequence to restore the functionality of a wild type gene or non-coding regulatory sequence (e.g., sequences encoding long non-coding RNA).
  • the donor DNA template may be used to replace a single allele of a defective gene or defective fragment thereof. In another embodiment, the donor DNA template is used to replace both alleles of a defective gene or defective gene fragment.
  • a “defective gene” or “defective gene fragment” is a gene or portion of a gene that when expressed, fails to generate a functioning protein or non-coding RNA with functionality of the corresponding wild-type gene. [00383] In certain example embodiments, these defective genes may be associated with one or more disease phenotypes.
  • the defective gene or gene fragment is not replaced but the heterologous nucleic acid is used to insert donor polynucleotides that encode gene or gene fragments that compensate for or override defective gene expression such that cell phenotypes associated with defective gene expression are eliminated or changed to a different or desired cellular phenotype.
  • This can be achieved by including the coding sequence of a therapeutic protein, such as a therapeutic antibody or functional fragment thereof, or a wild- type version of a defective protein associated with one or more disease phenotypes.
  • the donor may include, but not be limited to, genes or gene fragments, encoding proteins or RNA transcripts to be expressed, regulatory elements, repair templates, and the like.
  • the donor polynucleotides may comprise left end and right end sequence elements that function with transposition components that mediate insertion.
  • the donor DNA template manipulates a splicing site on the target polynucleotide.
  • the donor DNA template disrupts a splicing site. The disruption may be achieved by inserting the polynucleotide to a splicing site and/or introducing one or more mutations to the splicing site.
  • the donor polynucleotide may restore a splicing site.
  • the polynucleotide may comprise a splicing site sequence.
  • the donor DNA template to be inserted has a size from 10 bp to 50 kb in length, e.g., from 50 bp to ⁇ 40kb, from 100 bp to ⁇ 30 kb, from 100 bp to ⁇ 10 kb, from 100 bp to 300 bp, from 200 bp to 400 bp, from 300 bp to 500 bp, from 400 bp to 600 bp, from 500 bp to 700 bp, from 600 bp to 800 bp, from 700 bp to 900 bp, from 800 bp to 1000 bp, from 900 bp to 1100 bp, from 1000 bp to 1200 bp, from 1100 bp to 1300 bp, from 1200 bp to 1400 bp, from 1300 bp to 1500 bp, from 1400 bp to 1600 bp, from 1500 bp to 1700 bp,
  • the homologous arm on one or both ends of the sequence to be inserted is independently about 20 bp, 40 bp, 60 bp, 80 bp, 100 bp, 120 bp, or 150 bp.
  • the first homology arm and the second homology arm of the donor DNA flank a nucleotide sequence (“a nucleotide sequence of interest” or “an intervening nucleotide sequence”) that is to be introduced into a target nucleic acid.
  • the nucleotide sequence of interest can comprise: i) a nucleotide sequence encoding a polypeptide of interest; ii) a nucleotide sequence encoding an exon of a gene; iii) a promoter sequence; iv) an enhancer sequence; v) a nucleotide sequence encoding a non-coding RNA; or vi) any combination of the foregoing.
  • the donor DNA can provide for gene correction, gene replacement, gene tagging, transgene insertion, nucleotide deletion, gene disruption, gene mutation, etc.
  • the donor DNA can be used to add, e.g., insert or replace, nucleic acid material to a target DNA (e.g.
  • a tag e.g., 6xHis, a fluorescent protein (e.g., a green fluorescent protein; a yellow fluorescent protein, etc.), hemagglutinin (HA), FLAG, etc.
  • a regulatory sequence e.g. promoter, polyadenylation signal, internal ribosome entry sequence (IRES), 2A peptide, start codon, stop codon, splice signal, localization signal, enhancer, etc.
  • a nucleic acid sequence e.g., introduce a mutation
  • the donor DNA can be used to modify DNA in a site-specific, i.e. “targeted”, way; for example gene knock-out, gene knock-in, gene editing, gene tagging, etc., as used in, for example, gene therapy, e.g. to treat a disease; or as an antiviral, antipathogenic, or anticancer therapeutic, the production of genetically modified organisms in agriculture, the large scale production of proteins by cells for therapeutic, diagnostic, or research purposes, the induction of pluripotent stem cells, biological research, the targeting of genes of pathogens for deletion or replacement, etc.
  • the donor DNA comprises a nucleotide sequence encoding a polypeptide of interest.
  • Polypeptides of interest include, e.g., a) functional versions of a polypeptide that comprises one or more amino acid substitutions, insertions, and/or deletions and that exhibits reduced function, e.g., where the reduced function is associated with or causes a pathological condition; b) fluorescent polypeptides; c) hormones; d) receptors for ligands; e) ion channels; f) neurotransmitters; g) and the like.
  • the donor DNA comprises a nucleotide sequence that encodes a wild-type protein that is lacking in the recipient cell.
  • the donor DNA encodes a wild type factor (e.g.
  • the donor DNA comprises a nucleotide sequence that encodes a therapeutic antibody.
  • the donor DNA comprises a nucleotide sequence that encodes an engineered protein or receptor.
  • the engineered receptor is a T cell receptor (TCR), a natural killer (NK) receptor (NKR), or a B cell receptor (BCR).
  • TCR T cell receptor
  • NK natural killer
  • BCR B cell receptor
  • the engineered TCR or NKR targets a cancer marker (e.g., a polypeptide that is expressed (e.g., over-expressed) on the surface of a cancer cell).
  • the donor DNA comprises a nucleotide sequence that encodes a chimeric antigen receptor (CAR).
  • CAR targets a cancer marker.
  • Donor DNAs encoding CAR, TCR, and/or NCR proteins may be folded into DNA origami structures (DNA nanostructures) and delivered into T cells or NK cells in vitro or in vivo.
  • Non-limiting examples of polypeptides that can be encoded by a donor DNA include, e.g., IL1B (interleukin 1, beta), XDH (xanthine dehydrogenase), TP53 (tumor protein p53), PTGIS (prostaglandin 12 (prostacyclin) synthase), MB (myoglobin), IL4 (interleukin 4), ANGPT1 (angiopoietin 1), ABCG8 (ATP-binding cassette, sub-family G (WHITE), member 8), CTSK (cathepsin K), PTGIR (prostaglandin 12 (prostacyclin) receptor (IP)), KCNJ11 (potassium inwardly-rectifying channel, subfamily J, member 11), INS (insulin), CRP (C -reactive protein, pentraxin-related), PDGFRB (platelet- derived growth factor receptor, beta polypeptide), CCNA2 (cyclin A2), PDGFB
  • ACE angiotensin I converting enzyme peptidyl-dipeptidase A 1)
  • TNF tumor necrosis factor
  • IL6 interleukin 6 (interferon, beta 2)
  • STN statin
  • SERPINE1 serotonin peptidase inhibitor
  • clade E nonin, plasminogen activator inhibitor type 1
  • ALB albumin
  • ADIPOQ adiponectin, C1Q and collagen domain containing
  • APOB apolipoprotein B (including Ag(x) antigen)
  • APOE apolipoprotein E
  • LEP laeptin
  • MTHFR 5,10- methylenetetrahydrofolate reductase (NADPH)
  • APOA1 apolipoprotein A-I
  • EDN1 endothelin 1
  • NPPB natriuretic peptide precursor B
  • NOS3 nitric oxide synthase 3
  • GNRH1 gonadotropin-releasing hormone 1 (luteinizing- releasing hormone)
  • PAPPA pregnancy-associated plasma protein A, pappalysin 1
  • ARR3 arrestin 3, retinal (X-arrestin)
  • NPPC natriuretic peptide precursor C
  • AHSP alpha hemoglobin stabilizing protein
  • PTK2 PTK2 protein tyrosine kinase 2
  • IL13 interleukin 13
  • MTOR mechanistic target of rapamycin (serine/threonine kinase)
  • ITGB2 integratedin, beta 2 (complement component 3 receptor 3 and 4 subunit)
  • GSTT1 glutthione S-transfcrase theta 1
  • IL6ST interleukin 6 signal transducer (gpl30, oncostatin M receptor)
  • CPB2 carboxypeptidase B2 (plasma)
  • CYP1A2 cytochrome P
  • CAMP cathelicidin antimicrobial peptide
  • ZC3H12A zinc finger CCCH-type containing 12A
  • AKR1B1 aldo-keto reductase family 1, member B1 (aldose reductase)
  • DES desmin
  • MMP7 matrix metallopeptidase 7 (matrilysin, uterine)
  • AHR aryl hydrocarbon receptor
  • CSF1 colony stimulating factor 1 (macrophage)
  • HDAC9 histone deacetylase 9
  • CTGF connective tissue growth factor
  • KCNMA1 potassium large conductance calcium- activated channel, subfamily M, alpha member 1
  • UGT1A UDP glucuronosyltransferase 1 family, polypeptide A complex locus
  • PRKCA protein kinase C, alpha
  • COMT catechol-b- methyltransf erase
  • S100B S100 calcium binding protein B
  • the donor DNA encodes a wild-type version of any of the foregoing polypeptides; i.e., the donor DNA can encode a “normal” version that does not include a mutation(s) that results in reduced function, lack of function, or pathogenesis.
  • the donor DNA comprises a nucleotide sequence encoding a fluorescent polypeptide.
  • Suitable fluorescent proteins include, but are not limited to, green fluorescent protein (GFP) or variants thereof, blue fluorescent variant of GFP (BFP), cyan fluorescent variant of GFP (CFP), yellow fluorescent variant of GFP (YFP), enhanced GFP (EGFP), enhanced CFP (ECFP), enhanced YFP (EYFP), GFPS65T, Emerald, Topaz (TYFP), Venus, Citrine, mCitrine, GFPuv, destabilized EGFP (dEGFP), destabilized ECFP (dECFP), destabilised EYFP (dEYFP), mCFPm, Cerulean, T-Sapphire, CyPet, YPet, mKO, HcRed, t- HcRed, DsRed, DsRed2, DsRed-monomer, J-Red, dimer2, t-dimer2(12), mRFPl, pocilloporin, Renilla GFP, Monster GFP, paGFP, Kae
  • fluorescent proteins include mHoneydew, mBanana, mOrange, dTomato, tdTomato, mTangerine, mStrawberry, mCherry, mGrapel, mRaspberry, mGrape2, m PI urn (Shaner et al. (2005) Nat. Methods 2:905-909), and the like.
  • the donor DNA encodes an RNA, e.g., an siRNA, a microRNA, a short hairpin RNA (shRNA), an anti-sense RNA, a riboswitch, a ribozyme, an aptamer, a ribosomal RNA, a transfer RNA, and the like.
  • a donor DNA can include, in addition to a nucleotide sequence encoding one or more gene products (e.g., an RNA and/or a polypeptide), one or more transcriptional control elements, e.g., a promoter, an enhancer, and the like. In some cases, the transcriptional control element is inducible.
  • the promoter is reversible. In some cases, the transcriptional control element is constitutive. In some cases, the promoter is functional in a eukaryotic cell. In some cases, the promoter is a cell type- specific promoter. In some cases, the promoter is a tissue-specific promoter. [00396]
  • the nucleotide sequence of the donor DNA is typically not identical to the target nucleic acid (e.g., genomic sequence) that it replaces.
  • the donor DNA may contain at least one or more single base changes, insertions, deletions, inversions or rearrangements with respect to the target nucleic acid (e.g., genomic sequence), so long as sufficient homology is present to support homology-directed repair (e.g., for gene correction, e.g., to convert a disease- causing base pair or a non-disease-causing base pair).
  • the donor DNA comprises a non-homologous sequence flanked by two regions of homology, such that homology-directed repair between the target DNA region and the two flanking sequences results in insertion of the non-homologous sequence at the target region.
  • Donor DNA may also comprise a vector backbone containing sequences that are not homologous to the DNA region of interest (the target nucleic acid) and that are not intended for insertion into the DNA region of interest (the target nucleic acid).
  • the homologous region(s) of a donor sequence will have at least 50% sequence identity to a target nucleic acid (e.g., a genomic sequence) with which recombination is desired. In certain cases, 60%, 70%, 80%, 90%, 95%, 98%, 99%, or 99.9% sequence identity is present. Any value between 1% and 100% sequence identity can be present, depending upon the length of the donor polynucleotide.
  • the donor DNA may comprise certain nucleotide sequence differences as compared to the target nucleic acid (e.g., genomic sequence), where such difference include, e.g. restriction sites, nucleotide polymorphisms, selectable markers (e.g., drug resistance genes, fluorescent proteins, enzymes etc.), etc., which may be used to assess for successful insertion of the donor DNA at the cleavage site or in some cases may be used for other purposes (e.g., to signify expression at the targeted genomic locus).
  • nucleotide sequence differences will not change the amino acid sequence, or will make silent amino acid changes (i.e., changes which do not affect the structure or function of the protein).
  • the donor DNA will include one or more nucleotide sequences to aid in localization of the donor to the nucleus of the recipient cell or to aid in the integration of the donor DNA into the target nucleic acid.
  • the donor DNA may comprise one or more nucleotide sequences encoding one or more nuclear localization signals (e.g.
  • the donor DNA will include nucleotide sequences to recruit DNA repair enzymes to increase insertion efficiency.
  • Fiuman enzymes involved in homology directed repair include MRN-CtIP, BLM-DNA2, Exol, ERCC1, Rad51, Rad52, Ligase 1, RoIQ, PARP1, Ligase 3, BRCA2, RecQ/BLM-ToroIIIa, RTEL, Ro ⁇ d, and Ro ⁇ h (Verma and Greenburg (2016) Genes Dev.30 (10): 1138-1154).
  • the donor DNA is delivered as reconstituted chromatin (Cruz- Becerra and Kadonaga (2020) eLife 2020;9:e55780 DOI: 10.7554/eLife.55780).
  • the ends of the donor DNA are protected (e.g., from exonucleolytic degradation) by any convenient method and such methods are known to those of skill in the art.
  • one or more dideoxynucleotide residues can be added to the 3' terminus of a linear molecule and/or self complementary oligonucleotides can be ligated to one or both ends. See, for example, Chang et al. (1987) Proc. Natl. Acad Sci USA 84:4959-4963; Nehls et al. (1996) Science 272:886-889.
  • the donor / template comprises a coding sequence for a functional RNA element (ncRNAs, siRNA, shRNA, sgRNA, etc.).
  • the heterologous nucleic acid sequences encode a functional non-translated RNA.
  • the functional non-translated RNA is an RNA aptamer or a ribozyme.
  • the heterologous nucleic acid of the engineered retrons further includes a unique barcode to facilitate multiplexing. Barcodes may include one or more nucleotide sequences that are used to identify a nucleic acid or cell with which the barcode is associated. Such barcodes may be inserted for example, into the tip/loop region of the msd- encoded DNA.
  • Barcodes can be 3-1000 or more nucleotides in length, 10-250 nucleotides in length, or 10-30 nucleotides in length, including any length within these ranges, such as 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 40, 50, 60, 70, 80, 90, 100, 200, 300, 400, 500, 600, 700, 800, 900, or 1000 nucleotides in length.
  • barcodes are also used to identify the position (viz., positional barcode) of a cell, colony, or sample from which a retron originated, such as the position of a colony in a cellular array, the position of a well in a multi-well plate, the position of a tube in a rack, or the location of a sample in a laboratory.
  • a barcode may be used to identify the position of a genetically modified cell containing a retron. The use of barcodes allows retrons from different cells to be pooled in a single reaction mixture for sequencing while still being able to trace a particular retron back to the colony from which it originated.
  • adapter sequences can be added to engineered retron to facilitate high-throughput amplification or sequencing.
  • a pair of adapter sequences can be added at the 5 ⁇ and 3 ⁇ ends of a retron construct to allow amplification or sequencing of multiple engineered retron simultaneously by the same set of primers.
  • HNS guide RNA
  • the functional non-translated RNA is a CRISPR/Cas guide RNA (gRNA) specific for a target sequence in the mammalian cell.
  • FIG.1G depicts various configurations of a recombinant retron ncRNA which is modified by inserting a guide RNA at the 5’ end or the 3’ of the ncRNA.
  • the guide RNA also may be provided in trans as a separate construct. In addition, guide RNAs may be placed at both ends of a recombinant retron ncRNA. [00405] The skilled person will understand that selection of an appropriate guide RNA is informed by which RNA-guided nuclease is utilized. [00406]
  • a guide RNA provides target specificity to the complex (the RNP complex) by including a targeting segment, which includes a guide sequence (also referred to herein as a targeting sequence), which is a nucleotide sequence that is complementary to a sequence of a target nucleic acid.
  • guide RNA refers to an RNA that comprises: i) an “activator” nucleotide sequence that binds to a CRISPR/Cas effector polypeptide (e.g., a class 2 CRISPR/Cas effector polypeptide such as a type II, type V, or type VI CRISPR/Cas endonuclease) and activates the CRISPR/Cas effector polypeptide; and ii) a “targeter” nucleotide sequence that comprises a nucleotide sequence that hybridizes with a target nucleic acid.
  • a CRISPR/Cas effector polypeptide e.g., a class 2 CRISPR/Cas effector polypeptide such as a type II, type V, or type VI CRISPR/Cas endonuclease
  • the “activator” nucleotide sequence and the “targeter” nucleotide sequence can be on separate RNA molecules (e.g., a “dual-guide RNA”); or can be on the same RNA molecule (a “single-guide RNA”).
  • a guide nucleic acid in some cases includes only ribonucleotides. In some cases, a guide nucleic acid includes both ribonucleotides and deoxyribonucleotides.
  • a CRISPR/Cas guide RNA comprises one or more modifications, e.g., a base modification, a backbone modification, a sugar modification, etc., to provide the nucleic acid with a new or enhanced feature (e.g., improved stability, such as improved in vivo stability).
  • modifications e.g., a base modification, a backbone modification, a sugar modification, etc.
  • Suitable nucleic acid modifications include, but are not limited to: 2’O-methyI modified nucleotides, 2’ fluoro modified nucleotides, locked nucleic acid (LNA) modified nucleotides, peptide nucleic acid (PNA) modified nucleotides, nucleotides with phosphorothioate linkages, and a 5’ cap (e.g., a 7-methyIguanyIate cap (m7G)).
  • LNA locked nucleic acid
  • PNA peptide nucleic acid
  • Suitable modified nucleic acid backbones containing a phosphorus atom therein include, for example, phosphorothioates, chiral phosphorothioates, phosphorodithioates, phosphotriesters, aminoalkylphosphotriesters, methyl and other alkyl phosphonates including 3'-aIkyIene phosphonates, 5'- alkylene phosphonates and chiral phosphonates, phosphinates, phosphoramidates including 3'-amino phosphoramidate and aminoalkylphosphoramidates, phosphorodiamidates, thionophosphoramidates, thionoalkylphosphonates, thionoalkylphosphotriesters, selenophosphates and boranophosphates having normal 3'-5' linkages, 2'-5' linked analogs of these, and those having inverted polarity wherein one or more internucleotide linkages is a 3' to 3', 5' to 5' or
  • Suitable oligonucleotides having inverted polarity comprise a single 3' to 3' linkage at the 3'-most internucleotide linkage i.e. a single inverted nucleoside residue which may be a basic (the nucleobase is missing or has a hydroxyl group in place thereof).
  • Various salts such as, for example, potassium or sodium
  • mixed salts and free acid forms are also included.
  • a CRISPR- Cas guide RNA can also include one or more substituted sugar moieties.
  • Suitable polynucleotides comprise a sugar substituent group selected from: OH; F; 0-, S-, or N-alkyl; 0-, S-, or N-alkenyl; 0-, S- or N-alkynyl; or O-alkyl-O-alkyl, wherein the alkyl, alkenyl and alkynyl may be substituted or unsubstituted Ci to Cio alkyl or C2 to C10 alkenyl and alkynyl.
  • Particularly suitable are 0((CH 2 ) n 0) m CH 3 , 0(CH 2 ) n 0CH 3 , 0(CH 2 ) n NH 2 , 0(CH 2 ) n CH 3 , 0(CH 2 ) n 0NH 2 , and 0(CH 2 ) n 0N((CH 2 ) n CH 3 ) 2 , where n and m are from 1 to about 10.
  • Suitable polynucleotides comprise a sugar substituent group selected from: Ci to Cio lower alkyl, substituted lower alkyl, alkenyl, alkynyl, alkaryl, aralkyl, O-alkaryl or O-aralkyl, SH, SCH 3 , OCN, Cl, Br, CN, CF 3 , OCF 3 , SOCH 3 , S0 2 CH 3 , 0N0 2 , N0 2 , N 3 , NH 2 , heterocycloalkyl, heterocycloalkaryl, aminoalkylamino, polyalkylamino, substituted silyl, an RNA cleaving group, a reporter group, an intercalator, a group for improving the pharmacokinetic properties of an oligonucleotide, or a group for improving the pharmacodynamic properties of an oligonucleotide, and other substituents having similar properties.
  • a sugar substituent group selected from: Ci to Cio lower alky
  • a suitable modification includes 2'-methoxy ethoxy (2'-0-CH 2 CH 2 OCH 3 , also known as 2'-0-(2-methoxyethyl) or 2'- MOE) (Martin et al., Helv. Chim. Acta, 1995, 78, 486-504) i.e., an alkoxyalkoxy group.
  • a further suitable modification includes 2'-dimethylaminooxyethoxy, i.e., a 0(CH 2 ) 2 0N(CH 3 ) 2 group, also known as 2'- DMAOE, as described in examples herein below, and 2'-dimethylaminoethoxyethoxy (also known in the art as 2'-0-dimethyl-amino-ethoxy-ethyl or 2'- DMAEOE), i.e., 2'-0-CH 2 -0-CH 2 -N(CH 3 ) 2 .
  • Examples of various CRISPR/Cas effector proteins and CRISPR/Cas guide RNAs can be found in the art, for example, see Jinek et al., Science.2012 Aug 17;337(6096):816-21 ; Chylinski et al., RNA Biol.2013 May;10(5):726-37; Ma et al., Biomed Res Int.2013;2013:270805; Hou et al., Proc Natl Acad Sci U S A.2013 Sep 24;110(39):15644-9; Jinek et al., Elife.2013;2:e00471; Pattanayak et al., Nat Biotechnol.2013 Sep;31(9):839-43; Qi et al., Cell.2013 Feb 28 ; 152(5): 1173-83; Wang ek et al., Science.2012 Aug 17;337(6096):816-21 ; Chylinski et
  • Reverse transcriptases also known as RNA-directed DNA polymerases
  • RTs are enzymes present in all three domains of life, which are DNA polymerase using RNA as a template.
  • Reverse transcriptases of the present disclosure are used to reverse transcribe template msd RNA into single-stranded msDNA.
  • the reverse transcriptase or a functional domain thereof that may be used in the instant invention includes prokaryotic and eukaryotic RT, provided that the RT functions within the host to generate a donor polynucleotide sequence from the RNA template (e.g., an RNA template from the retron transcript ncRNA).
  • suitable RT sequences are provided in Table A.
  • nucleotide sequence of a native or wild-type RT is modified, for example, using known codon optimization techniques, so that expression within the desired host is optimized.
  • the RT domain of a reverse transcriptase is used in the present invention, so long as it is compatible with the engineered retron of the invention. The domain may include only the RNA-dependent DNA polymerase activity.
  • the RT domain is non-mutagenic, i.e., does not cause mutation in the donor polynucleotide (e.g., during the reverse transcriptase process).
  • the RT domain may be non-retron RT in origin, e.g., a viral RT or a human endogenous RTs.
  • the RT domain is retron RT or DGRs RT.
  • the RT may be less mutagenic than a counterpart wildtype RT. In certain embodiments, the RT is not mutagenic.
  • a reverse transcriptase is encoded by a retron ret gene, which may accompany the cognate msr and msd loci and specifically recognize the secondary structure of the cognate ncRNA transcript.
  • the RT may be obtained from prokaryotic or eukaryotic cells. Most reverse transcriptases (80%) can be phylogenetically clustered into three major lineages: group II introns, diversity-generating retroelements (DGRs), and retrons.
  • RTs include abortive infection (Abi) RTs, CRISPR-Cas-associated RTs, Group II-like (G2L), the unknown groups (UG), and rvt elements.
  • Abi abortive infection
  • CRISPR-Cas-associated RTs Group II-like (G2L), the unknown groups (UG), and rvt elements.
  • the RT gene is a cognate RT, a retron RT from a species within the same species or clade of the cognate RT, or a retron RT not within the same clade of the cognate RT such as an unrelated RT or an engineered RT.
  • the non- retron related RT are RTs from group II introns, diversity-generating retroelements (DGRs), abortive infection (Abi) RTs, CRISPR-Cas-associated RTs, Group II-like (G2L), the unknown groups (UG), and rvt elements.
  • DGRs diversity-generating retroelements
  • Abi abortive infection
  • CRISPR-Cas-associated RTs Group II-like (G2L)
  • G2L Group II-like
  • UG unknown groups
  • the RT are from clades related to retron/retron-like sequences. In some embodiments, the RT are selected from RTs provided in Table A. In some embodiments, the RT is not an RT associated with the sequences identified in Table X. [00419] In prokaryotic retron systems, the RT gene is typically located downstream from the ncRNA (msr and msd) locus. In the engineered retron, the RT position can differ from the natural or wild type retrons. In some embodiments, the RT gene can be provided in cis, such as either upstream or downstream of the msr locus or the msd locus.
  • the RT gene is provided in trans, such as provided separately in a vector of the vector system described herein, wherein the ncRNA coding msr and msd sequences are provided in a different vector of the vector system described herein.
  • the RT is modified (e.g., insertion, deletion, and/or substitution of one or more nucleotide(s)) or codon optimized to enhance activity or processivity.
  • a cryptic stop signal is removed from the RT thereby allowing generation of longer ssDNAs.
  • the RT is from a retron which encodes msDNA as described in US 6,017,737; US5,849,563; US5,780,269; US5,436,141; US5,405,775; US5,320,958; CA2,075,515; all of which are incorporated by reference herein in their entireties.
  • the engineered retron further comprises a polynucleotide (e.g., a DNA molecule) encoding a reverse transcriptase (RT) or a portion thereof.
  • the encoded RT or portion thereof is capable of synthesizing a DNA copy of at least a portion of the msd locus encoding the msDNA.
  • the polynucleotide (e.g., a DNA molecule) encoding the RT comprises a polynucleotide listed in Table A, or a polynucleotide having at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%%, at least 99.1%, at least 99.2%, at least 99.3%, at least 99.4%, at least 99.5%, at least 99.6%, at least 99.7%, at least 99.8% or at least 99.9% sequence identity to a poly
  • the polynucleotide encoding the RT encodes a polypeptide listed in Table A, or a polypeptide having at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%%, at least 99.1%, at least 99.2%, at least 99.3%, at least 99.4%, at least 99.5%, at least 99.6%, at least 99.7%, at least 99.8% or at least 99.9% sequence identity to a polypeptide listed in Table A; and/or a polypeptide of Table C.
  • the polynucleotide encoding the RT does not comprise a polynucleotide listed in Table X.
  • the RT binds the ncRNA template downstream from the msd locus, forming an RT-RNA complex, and initiating reverse transcription of the RNA towards its 5 ⁇ end. Accordingly, in certain aspects the disclosure relates to an engineered nucleic acid- enzyme construct comprising: a.
  • ncRNA non-coding RNA
  • msDNA multi-copy single-stranded DNA
  • msd locus encoding the msd RNA portion of the msDNA
  • a heterologous nucleic acid inserted at or within a location selected from: the msd locus, upstream of the msr locus, upstream of the msd locus, and downstream of the msd locus; and c.
  • a reverse transcriptase or a domain thereof comprising: i) a polypeptide listed in Table A, or a polypeptide having at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%%, at least 99.1%, at least 99.2%, at least 99.3%, at least 99.4%, at least 99.5%, at least 99.6%, at least 99.7%, at least 99.8% or at least 99.9% sequence identity to a polypeptide listed in Table A; and/or ii) a polypeptide listed in Table C.
  • the RT does not comprise a polypeptide listed in Table X.
  • the disclosure relates to an engineered nucleic acid-enzyme construct comprising: a) a non-coding RNA (ncRNA) comprising: i) an msr locus encoding the msr RNA portion of a multi-copy single-stranded DNA (msDNA); and ii) an msd locus encoding the msd RNA portion of the msDNA, b) a heterologous nucleic acid inserted at or within a location selected from: the msd locus; upstream of the msr locus; upstream of the msd locus; and downstream of the msd locus; and c) a reverse transcriptase (RT), or a portion thereof, wherein the RT is capable of synthesizing a DNA copy of at least a portion of the msd locus encoding the
  • the disclosure relates to an engineered nucleic acid-enzyme construct comprising: a) a non-coding RNA (ncRNA) comprising: i) an msr locus encoding the msr RNA portion of a multi-copy single-stranded DNA (msDNA); and ii) an msd locus encoding the msd RNA portion of the msDNA; b) a heterologous nucleic acid inserted at or within a location selected from: the msd locus, upstream of the msr locus, upstream of the msd locus, and downstream of the msd locus; and c) a reverse transcriptase (RT) or a domain thereof: wherein the RT comprises: i) an RT listed in Table A, or an RT having at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at
  • the ncRNA comprises: i) an ncRNA listed in Table B, or an ncRNA having at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%%, at least 99.1%, at least 99.2%, at least 99.3%, at least 99.4%, at least 99.5%, at least 99.6%, at least 99.7%, at least 99.8% or at least 99.9% sequence identity to an ncRNA listed in Table B; and/or optionally wherein the ncRNA is not an ncRNA from the retons of Table X.
  • the RT is linked to components such as RNA-guided and non-RNA guided nucleases.
  • the linked maybe via s peptide bond or a short linker peptide in a fusion protein.
  • Suitable linker peptides include flexible linkers such as those comprising G or S repeats, such as G 4 S repeat units or GS repeat units, with 1-20 repeats, e.g., 1, 2, 3, 4, 5, 6, 7, or 8 repeats.
  • the RT is chemically linked or conjugated to the RNA- guided and non-RNA guided nucleases via non-peptide bonds.
  • Such protein conjugates may be delivered directly to a host cell, either together with the nucleic acid component of the engineered retron described herein, or separately.
  • the RT is linked to a DNA-repair modulating biomolecule (e.g., NHEJ peptide inhibitors.
  • RNA-guided Nucleases RNA-guided Nucleases
  • the engineered retron e.g., an engineered nucleic acid construct or engineered nucleic acid-enzyme construct as described herein
  • gRNA guide RNA
  • the gRNA includes sequences complementary to a genomic sequence, and therefore can mediate binding of the nuclease-gRNA complex to the genomic target site by hybridization between the guide sequence and the target site sequence.
  • the gRNA can be linked to the ncRNA and/or the msDNA encoded by the engineered retron described herein at the 5 ⁇ end of the ncRNA and/or the msDNA.
  • the gRNA can be linked to the ncRNA and/or the msDNA encoded by the engineered retron described herein at the 3 ⁇ end of the RNA of the ncRNA and/or the msDNA after reverse transcription.
  • the nuclease that can form a complex with the gRNA can be any one of the art-recognized clustered regularly interspersed short palindromic repeats (CRISPR) system Cas effector enzymes, which are useful for, e.g., genome editing, including genome editing in mammalian cells or human cells.
  • CRISPR CRISPR
  • a gRNA that can be loaded into a Cas9 or variant thereof may be encoded by the engineered retron, such that the gRNA is transcribed as part of the msDNA.
  • the gRNA may be linked to the 5 ⁇ end of the a1 region of the msr-encoded sequence in the retron ncRNA, as well as the msDNA after reverse transcription.
  • the gRNA may be part of the modified msr-region present in the ncRNA and the msDNA produced after reverse transcription (viz., the encoded gRNA is not degraded by the synthesis of the msDNA through reverse transcription of the ncRNA).
  • Cas enzymes Any art recognized CRISPR/Cas effector enzymes or variants thereof (“Cas enzymes”) known to be useful for CRISPR-based genome editing can be used with the engineered retron, though such Cas enzymes may not necessarily be part of the engineered retron, and can (but is not required to) be provided separately.
  • the Cas enzymes can be provided as part of the vector system described herein.
  • the coding sequence of the suitable Cas enzyme can be present on the same vector that provides the engineered retron, or on different vectors.
  • the engineered retron and the Cas enzymes When the engineered retron and the Cas enzymes are present on the same vector, they may be under the transcriptional control of the same promoter, enhancer, or other transcriptional regulatory elements, or may be separately regulated by different promoters, enhancers, and/or other transcriptional regulatory sequences in the vector system.
  • the Cas enzyme is a Class 2, Type II CRISPR/Cas effector enzyme, such as Cas9.
  • the Cas9 is from Streptococcus pyogenes (SpCas9)
  • the gRNA encoded by the engineered retron comprises both the crisprRNA (crRNA) and the tracrRNA linked together as the single guide RNA (gRNA).
  • the Cas9 is from Staphylococcus aureus Cas9 (SaCas9), or an engineered variant thereof such as SaCas9-HF (a high-fidelity variant with genome-wide activity), KKHSaCas9 (which recognizes a 5 ⁇ -NNGRRT-3 ⁇ PAM, and has a 2-4x broader range of target sites in the human genome than the wildtype SaCas9), and microABE1744 (an engineered SaCas9 variant adapted for use in adenine base editing (ABE), with significantly improved on-target editing compared to other nucleases, with a reduced RNA off-target footprint).
  • SaCas9-HF a high-fidelity variant with genome-wide activity
  • KKHSaCas9 which recognizes a 5 ⁇ -NNGRRT-3 ⁇ PAM, and has a 2-4x broader range of target sites in the human genome than the wildtype SaCas9
  • microABE1744 an engineered SaCas9 variant adapted for
  • the Cas9 is from Streptococcus thermophilus (StCas9), Neisseria meningitidis (NmCas9), Francisella novicida (FnCas9), or Campylobacter jejuni (CjCas9).
  • the Cas9 is from Streptococcus canis (ScCas9), with a less stringent PAM sequence requirement of 5′-NNG-3′ (instead of the more stringent 5′-NGG-3′ for SpCas9).
  • the Cas9 is from Staphylococcus auricularis (SauriCas9) (which recognizes a 5 ⁇ -NNGG-3 ⁇ PAM sequence, has high editing activity).
  • the Cas enzyme is a Cas9 variant with mutated catalytic domain that retains cleavage specificity, but only nicks a single DNA strand at a desired target sequence instead of creating a double-strand break (DSB). Two such Cas9 nickase variants targeting different strands of the same target sequence may be used together to increase fidelity of creating the DSB, each using a different gRNA that can be provided by the engineered retron.
  • the Cas enzyme is a high fidelity Cas9 variant with weakened DNA phosphate backbone interaction (such as SpCas9-HF1) that displays genome- wide specificity and undetectable off-target effects.
  • the Cas enzyme is a Cas9 variant known as eSpCas9, which has weakened interactions between the eSpCas9 and its gRNAs with non-exact complementarity with the target DNA sequence, thus providing improved specificity and lower off-target editing rates.
  • the Cas enzyme is a hyper-accurate Cas9 variant (HypaCas9), which improves proofreading before cleavage, and thus drastically reduces off- target cleavage.
  • the Cas enzyme is a Cas9 variant (FokI-Fused dCas9), which combines the DNA recognition ability by dCas9 with the specificity of an active nuclease FokI. The resulting nuclease cuts the target sequence only after dimerization, which is more difficult to occur at off-target sites, thus resulting in enhanced specificity.
  • the Cas enzyme is a Cas9 variant xCas9, which recognizes a broad range of PAM sequences, thus increasing the target sites to 1 in 4 in the genome.
  • the xCas9 variant also exhibits lower off-target rates than the commonly used SpCas9.
  • the Cas enzyme is a Cas9 variant with altered PAM sequence specificities, including the SpG variant with an expanded target range of PAM sequences, and the SpRY variant that can target almost all PAM sequences.
  • the Cas enzyme is dCas9 having inactivated catalytic nuclease domains while maintaining the recognition domains that allow guide RNA-mediated targeting to specific DNA sequences.
  • the dCas9 may be further linked to a functional domain with a distinct biological function, such as transcriptional activation / depression, DNA methylation, demethylation, endonuclease (such as FokI), or fluorescent dye.
  • Representative (non-limiting) dCas9-linked functional domains include including dCas9-SAM, dCas9-SunTag, dCas9-VPR, and dCas9-KREB.
  • the dCas9 is fused with a catalytic enzyme with cytidine deaminase activity, which converts GC basepair to AT basepair.
  • the dCas9 is fused with an engineered RNA adenosine deaminase, which converts AT basepair to GC basepair.
  • the Cas enzyme is a Class 2, Type V CRISPR/Cas effector enzyme, such as Cas12a (Cpf1) (Type V-A), Cas12b (C2c1) (Type V-B), Cas12c (C2c3) (Type V-C), Cas12d (CasY) (Type V-D), Cas12e (CasX) (Type V-E), Cas12f (Cas14, C2c10) (Type V- F), Cas12g (Type V-G), Cas12h (Type V-H), Cas12i (Type V-I), Cas12k (C2c5) (Type V-K), or C2c4 / C2c8 / C2c9 (Type V-U).
  • the Cas enzyme is Cas12a or Cpf1 (CRISPR from Prevotella and Francisella 1). Unlike Cas9, Cas12a is well-suited for targeting AT-rich DNA sequences because of its AT-rich PAM sequences.
  • the Cas12a is FnCas12a (that recognizes PAM sequence 5′ ⁇ TTN ⁇ 3′), or AsCas12a or LbCas12a (that recognize 5′ ⁇ TTTV ⁇ 3′ PAM sequence), where V is A, G, or C nucleotide.
  • Cas12a creates staggered double-stranded breaks in the target DNA, rather than the blunt-ends generated by SpCas9, thus rendering it more useful for HDR repair.
  • the subject engineered retron encodes a gRNA suitable for use for Cas12a, in that the gRNA does not require a tracer RNA, and only requires the crRNA.
  • the Cas enzyme is a Cas12a variant from Acidaminococcus sp. (enAsCas12a), which has an expanded target range of PAM sequences and significantly higher editing activity compared to wild type Cas12a.
  • the Cas enzyme is a high-fidelity Cas12a variant (enAsCas12a-HF1) that reduces off-target editing.
  • the Cas enzyme is Cas12b or C2c1.
  • the Cas12b is form Alicyclobacillus acidoterrestris (AacCas12b), or from Alicyclobacillus acidiphilus Cas12b (AapCas12b).
  • the Cas enzyme is a Cas12b variant that functions at 37°C, such as one form Bacillus hisashii (BhCas12b).
  • the Cas enzyme is a BhCas12b variant with higher specificity than SpCas9. [00461] In some embodiments, the Cas enzyme is CasX or Cas12d. [00462] In some embodiments, the Cas enzyme is CasY or Cas12e, and the subject engineered retron encodes a short-complementarity untranslated RNA (scoutRNA) together with crRNA (rather than tracrRNA used in other CRISPR-Cas systems). [00463] In some embodiments, the Cas enzyme is Cas14, from archaea bacteria.
  • Cas14 targets single-stranded (ss) DNA target sequences, does not require PAM sequence for activation, and has collateral activity (i.e., cuts other non-target ssDNA strands non-specifically upon binding the target sequence). Unlike Cas12a, Cas14a requires high fidelity complementarity to the target ssDNA, and is very sensitive to internal seed region mismatches of the ssDNA target substrate.
  • the gRNA nuclease is an engineered RNA-guided Fokl nuclease.
  • RNA-guided Fokl nucleases comprise fusions of inactive Cas9 (dCas9) and the Fokl endonuclease (FokI-dCas9), wherein the dCas9 portion confers guide RNA- dependent targeting on Fokl.
  • dCas9 inactive Cas9
  • FokI-dCas9 Fokl endonuclease
  • dCas9 portion confers guide RNA- dependent targeting on Fokl.
  • the RNA-guided nuclease can be a non-CRISPER/Cas related nuclease such as transposon-encoded nucleases, IscBs, IscR, or TnpBs.
  • the Cas enzyme is a Cas9
  • the retron-based editing systems described herein can include any Cas9 equivalent.
  • a Cas9 equivalent is a Cas9-like protein that provides the same or substantially the same function as Cas9.
  • Cas9 refers to a type II enzyme of the CRISPR-Cas system
  • a Cas9 equivalent can refer to a type V or type VI enzyme of the CRISPR- Cas system.
  • Cas12e is a Cas9 equivalent that reportedly has the same function as Cas9 but which evolved through convergent evolution.
  • the Cas12e (CasX) protein is contemplated to be used with the retron-based editor systems described herein.
  • any variant or modification of Cas12e (CasX) is conceivable and within the scope of the present disclosure.
  • Cas9 equivalents may refer to Cas12e (CasX) or Cas12d (CasY), which have been described in, for example, Burstein et al.,“New CRISPR–Cas systems from uncultivated microbes.” Cell Res.2017 Feb 21. doi: 10.1038/cr.2017.21, the entire contents of which is hereby incorporated by reference.
  • CasX Cas12e
  • CasY Cas12d
  • Cas9 refers to Cas12e, or a variant of Cas12e. In some embodiments, Cas9 refers to a Cas12d, or a variant of Cas12d. It should be appreciated that other RNA-guided DNA binding proteins may be used as a nucleic acid programmable DNA binding protein (napDNAbp), and are within the scope of this disclosure.
  • napDNAbp nucleic acid programmable DNA binding protein
  • the Cas9 equivalent comprises an amino acid sequence that is at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or at least 99.5% identical to a naturally-occurring Cas12e (CasX) or Cas12d (CasY) protein.
  • the napDNAbp is a naturally-occurring Cas12e (CasX) or Cas12d (CasY) protein.
  • the napDNAbp comprises an amino acid sequence that is at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or at least 99.5% identical to a wild-type Cas moiety or any Cas moiety provided herein.
  • the nucleic acid programmable DNA binding proteins include, without limitation, Cas9 (e.g., dCas9 and nCas9), Cas12e (CasX), Cas12d (CasY), Cas12a (Cpf1), Cas12b1 (C2c1), Cas13a (C2c2), Cas12c (C2c3), Argonaute, and Cas12b1.
  • Cas9 e.g., dCas9 and nCas9
  • Cas12a (Cpf1) is also a Class 2 CRISPR effector, but it is a member of type V subgroup of enzymes, rather than the type II subgroup. It has been shown that Cas12a (Cpf1) mediates robust DNA interference with features distinct from Cas9.
  • Cas12a (Cpf1) is a single RNA-guided endonuclease lacking tracrRNA, and it utilizes a T-rich protospacer-adjacent motif (TTN, TTTN, or YTN). Moreover, Cpf1 cleaves DNA via a staggered DNA double-stranded break.
  • Cpf1- family proteins Two enzymes from Acidaminococcus and Lachnospiraceae are shown to have efficient genome-editing activity in human cells.
  • Cpf1 proteins are known in the art and have been described previously, for example Yamano et al.,“Crystal structure of Cpf1 in complex with guide RNA and target DNA.” Cell (165) 2016, p.949-962; the entire contents of which is hereby incorporated by reference.
  • the Cas protein may include any CRISPR associated protein, including but not limited to, Cas12a, Cas12b1, Cas1, Cas1B, Cas2, Cas3, Cas4, Cas5, Cas6, Cas7, Cas8, Cas9 (also known as Csn1 and Csx12), Cas10, Csy1, Csy2, Csy3, Cse1, Cse2, Csc1, Csc2, Csa5, Csn2, Csm2, Csm3, Csm4, Csm5, Csm6, Cmr1, Cmr3, Cmr4, Cmr5, Cmr6, Csb1, Csb2, Csb3, Csx17, Csx14, Csx10, Csx16, CsaX, Csx3, Csx1, Csx15, Csf1, Csf2, Csf3, Csf4, homologs thereof, or modified versions thereof.
  • CRISPR associated protein including but not limited to, Cas12a, Cas12b
  • the RNA-guided nuclease can be any of the following proteins: a Cas9, a Cas12a (Cpf1), a Cas12e (CasX), a Cas12d (CasY), a Cas12b1 (C2c1), a Cas13a (C2c2), a Cas12c (C2c3), a GeoCas9, a CjCas9, a Cas12g, a Cas12h, a Cas12i, a Cas13b, a Cas13c, a Cas13d, a Cas14, a Csn2, an xCas9, an SpCas9-NG, or an Argonaute (Ago) domain, or a variant thereof.
  • a Cas9 a Cas12a (Cpf1), a Cas12e (CasX), a Cas12d (CasY), a Cas12b1
  • RNA-guided nucleases are readily available and known in the art. Exemplary RNA-guided nucleases and their amino acid sequences can be found, for example, in WO 2017/070633, US 2020/0010835, US 2022/0204975, US 11071790, WO 2020/191233, US 11447770, US 10858639, and US 10947530, each of which are incorporated herein by reference in their entireties. E.
  • the subject engineered retron can also be used in combination with sequence-specific nucleases that do not use a guide RNA to recognize a target sequence, such as non-CRISPR/Cas sequence-specific nucleases including TALENs, ZFNs, meganucleases, and restriction enzymes, as well as other sequence-specific nucleases that use other RNA guides, such as transposon-encoded IscBs, IscR, or TnpBs.
  • the subject engineered retron may encode or provide a msDNA that can serve as a donor or template sequence for HDR-mediated genome editing.
  • the RT of the engineered retron is fused to such sequence-specific nuclease, such that the msDNA, by way of being generated by the RT close to the site of HDR-mediated genome editing, can be more efficiently participate in the HDR-mediated genome editing.
  • the non-CRISPR/Cas sequence-specific nuclease is or comprises a TALE Nuclease, a TALE nickase, Zinc Finger (ZF) Nuclease, ZF Nickase, meganuclease, or a combination thereof.
  • the non-CRISPR/Cas sequence- specific nuclease is or includes two, three, four, or more of an independently selected TALE Nuclease, TALE nickase, Zinc Finger (ZF) Nuclease, ZF Nickase, Meganuclease, restriction enzymes or a combination thereof.
  • the combination is or comprises a TALE Nuclease/a ZF Nuclease; a TALE Nickase/a ZF nickase.
  • the non-CRISPR/Cas sequence-specific nuclease is or comprises a TALE Nuclease (Transcription Activator-Like Effector Nucleases (TALEN)).
  • TALE Nuclease Transcription Activator-Like Effector Nucleases (TALEN)
  • TALENs are restriction enzymes engineered to cut specific target DNA sequences.
  • TALENs comprise a TAL effector (TALE) DNA-binding domain (which binds at or close to the target DNA), fused to a DNA cleavage domain which cuts target DNA.
  • TALEs are engineered to bind to practically any desired DNA sequence.
  • the TALEN comprises an N-terminal capping region, a DNA binding domain which may comprise at least one or more TALE monomers or half-monomers specifically ordered to target the genomic locus of interest, and a C-terminal capping region, wherein these three parts are arranged in a predetermined N- terminus to C-terminus orientation.
  • the TALEN includes at least one or more regulatory or functional protein domains.
  • the TALE monomers or half monomers may be variant TALE monomers derived from natural or wild type TALE monomers but with altered amino acids at positions usually highly conserved in nature, and in particular have a combination of amino acids as RVDs that do not occur in nature, and which may recognize a nucleotide with a higher activity, specificity, and/or affinity than a naturally occurring RVD.
  • the variants may include deletions, insertions and substitutions at the amino acid level, and transversions, transitions and inversions at the nucleic acid level at one or more locations.
  • the variants may also include truncations.
  • the TALE monomer / half monomer variants include homologous and functional derivatives of the parent molecules.
  • the variants are encoded by polynucleotides capable of hybridizing under high stringency conditions to the parent molecule-encoding wild-type nucleotide sequences.
  • the DNA binding domain of the TALE has at least 5 of more TALE monomers and at least one or more half-monomers specifically ordered or arranged to target a genomic locus of interest.
  • the construction and generation of TALEs or polypeptides of the invention may involve any of the methods known in the art.
  • Naturally occurring TALEs or “wild type TALEs” are nucleic acid binding proteins secreted by numerous species of proteobacteria.
  • TALEs contain a nucleic acid binding domain composed of tandem repeats of highly conserved monomer polypeptides that are predominantly 33, 34 or 35 amino acids in length and that differ from each other mainly in amino acid positions 12 and 13.
  • a general representation of a TALE monomer which is comprised within the DNA binding domain is Xl-11-(X12X13)-X14-33 or 34 or 35, where the subscript indicates the amino acid position and X represents any amino acid.
  • X12X13 indicate the RVDs.
  • the variable amino acid at position 13 is missing or absent and in such monomers, the RVD consists of a single amino acid.
  • the RVD may be alternatively represented as X*, where X represents X12 and (*) indicates that X13 is absent.
  • the DNA binding domain may comprise several repeats of TALE monomers and this may be represented as (Xl-11-(X12X13)-X14-33 or 34 or 35)z, where z is optionally at least 5- 40, such as 10-26.
  • the TALE monomers have a nucleotide binding affinity that is determined by the identity of the amino acids in its RVD.
  • Polypeptide monomers with an RVD of NI preferentially bind to adenine (A), monomers with an RVD of NG preferentially bind to thymine (T), monomers with an RVD of HD preferentially bind to cytosine (C), monomers with an RVD of NN preferentially bind to both adenine (A) and guanine (G), monomers with an RVD of IG preferentially bind to T, monomers with an RVD of NS recognize all four base pairs and may bind to A, T, G or C.
  • the number and order of the polypeptide monomer repeats in the nucleic acid binding domain of a TALE determines its nucleic acid target specificity.
  • TALEs The structure and function of TALEs is further described in, for example, Moscou et al., Science 326:1501 (2009); Boch et al., Science 326:1509-1512 (2009); and Zhang et al., Nature Biotechnology 29:149-153 (2011), each of which is incorporated by reference in its entirety.
  • the TALE is a dTALE (or designerTALE), see Zhang et al., Nature Biotechnology 29:149-153 (2011), incorporated herein by reference.
  • the TALE monomer comprises an RVD of HN or NH that preferentially binds to guanine, and the TALEs have high binding specificity for guanine containing target nucleic acid sequences.
  • polypeptide monomers having RVDs RN, NN, NK, SN, NH, KN, HN, NQ, HH, RG, KH, RH and SS preferentially bind to guanine.
  • polypeptide monomers having RVDs RN, NK, NQ, HH, KH, RH, SS and SN preferentially bind to guanine.
  • polypeptide monomers having RVDs HH, KH, NH, NK, NQ, RH, RN and SS preferentially bind to guanine.
  • the RVDs that have high binding specificity for guanine are RN, NH RH and KH.
  • polypeptide monomers having an RVD of NV preferentially bind to adenine and guanine as do monomers having the RVD HN.
  • Monomers having an RVD of NC preferentially bind to adenine, guanine and cytosine, and monomers having an RVD of S (or S*), bind to adenine, guanine, cytosine and thymine with comparable affinity.
  • monomers having RVDs of H*, HA, KA, N*, NA, NC, NS, RA, and S* bind to adenine, guanine, cytosine and thymine with comparable affinity.
  • Such polypeptide monomers allow for the generation of degenerative TALEs able to bind to a repertoire of related, but not identical, target nucleic acid sequences.
  • the TALE polypeptide has a nucleic acid binding domain containing polypeptide monomers arranged in a predetermined N-terminus to C-terminus order such that each polypeptide monomer binds to a nucleotide of a predetermined target nucleic acid sequence, and where at least one of the polypeptide monomers has an RVD of HN or NH and preferentially binds to guanine, an RVD of NV and preferentially binds to adenine and guanine, an RVD of NC and preferentially binds to adenine, guanine and cytosine or an RVD of S and binds to adenine, guanine, cytosine and thymine.
  • each polypeptide monomer of the nucleic acid binding domain that binds to adenine has an RVD of NI, NN, NV, NC or S.
  • each polypeptide monomer of the nucleic acid binding domain that binds to guanine has an RVD of HN, NH, NN, NV, NC or S.
  • each polypeptide monomer of the nucleic acid binding domain that binds to cytosine has an RVD of HD, NC or S.
  • each polypeptide monomer that binds to thymine has an RVD of NG or S.
  • each polypeptide monomer of the nucleic acid binding domain that binds to adenine has an RVD of NI.
  • each polypeptide monomer of the nucleic acid binding domain that binds to guanine has an RVD of HN or NH.
  • each polypeptide monomer of the nucleic acid binding domain that binds to cytosine has an RVD of HD.
  • each polypeptide monomer that binds to thymine has an RVD of NG.
  • the RVDs that have a specificity for adenine are NI, RI, KI, HI, and SI.
  • the RVDs that have a specificity for adenine are HN, SI and RI, most preferably the RVD for adenine specificity is SI.
  • the RVDs that have a specificity for thymine are NG, HG, RG and KG.
  • the RVDs that have a specificity for thymine are KG, HG and RG, most preferably the RVD for thymine specificity is KG or RG.
  • the RVDs that have a specificity for cytosine are HD, ND, KD, RD, HH, YG and SD.
  • the RVDs that have a specificity for cytosine are SD and RD.
  • FIG.4B of WO 2012/067428 provides representative RVDs and the nucleotides they target, the entire content of which is hereby incorporated herein by reference.
  • the variant TALE monomers may comprise any of the RVDs that exhibit specificity for a nucleotide as depicted in FIG.4A of WO2012/067428.
  • the RVD SH may have a specificity for G
  • the RVD IS may have a specificity for A
  • the RVD IG may have a specificity for T.
  • the RVD NT may bind to G and A.
  • the RVD NP may bind to A, T and C.
  • At least one selected RVD may be NI, HD, NG, NN, KN, RN, NH, NQ, SS, SN, NK, KH, RH, HH, KI, HI, RI, SI, KG, HG, RG, SD, ND, KD, RD, YG, HN, NV, NS, HA, S*, N*, KA, H*, RA, NA or NC.
  • the predetermined N-terminal to C-terminal order of the one or more polypeptide monomers of the nucleic acid or DNA binding domain determines the corresponding predetermined target nucleic acid sequence to which the TALE or polypeptides of the invention may bind.
  • the monomers and at least one or more half monomers are “specifically ordered to target” the genomic locus or gene of interest.
  • the natural TALE-binding sites always begin with a thymine (T), which may be specified by a cryptic signal within the non-repetitive N-terminus of the TALE polypeptide; in some cases this region may be referred to as repeat 0.
  • TALE binding sites do not necessarily have to begin with a thymine (T) and polypeptides of the invention may target DNA sequences that begin with T, A, G or C.
  • tandem repeat of TALE monomers always ends with a half- length repeat or a stretch of sequence that may share identity with only the first 20 amino acids of a repetitive full length TALE monomer and this half repeat may be referred to as a half- monomer (FIG.8 of WO 2012/067428). Therefore, it follows that the length of the nucleic acid or DNA being targeted is equal to the number of full monomers plus two (see FIG.44 of WO 2012/067428).
  • nucleic acid binding domains are engineered to contain 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, or more polypeptide monomers arranged in a N-terminal to C-terminal direction to bind to a predetermined 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25 nucleotide length nucleic acid sequence.
  • nucleic acid binding domains are engineered to contain 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26 or more full length polypeptide monomers that are specifically ordered or arranged to target nucleic acid sequences of length 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27 and 28 nucleotides, respectively.
  • the polypeptide monomers are contiguous.
  • half-monomers may be used in the place of one or more monomers, particularly if they are present at the C-terminus of the TALE.
  • Polypeptide monomers are generally 33, 34 or 35 amino acids in length.
  • the amino acid sequences of polypeptide monomers are highly conserved or as described herein, the amino acids in a polypeptide monomer, with the exception of the RVD, exhibit patterns that effect TALE activity, the identification of which may be used in preferred embodiments of the invention.
  • the DNA binding domain may comprise (Xl-11- X12X13-X14-33 or 34 or 35)z, wherein Xl-11 is a chain of 11 contiguous amino acids, wherein X12X13 is a repeat variable diresidue (RVD), wherein X14-33 or 34 or 35 is a chain of 21, 22 or 23 contiguous amino acids, wherein z is at least 5 to 26, then the preferred combinations of amino acids are LTLD or LTLA or LTQV at Xl-4, or EQHG or RDHG at positions X30-33 or X31-34 or X32-35.
  • RVD repeat variable diresidue
  • amino acid combinations of interest in the monomers are LTPD at Xl-4 and NQALE at XI 6-20 and DHG at X32-34 when the monomer is 34 amino acids in length.
  • the monomer is 33 or 35 amino acids long, then the corresponding shift occurs in the positions of the contiguous amino acids NQALE and DHG.
  • NQALE is at X15-19 or X17-21 and DHG is at X31-33 or X33-35.
  • amino acid combinations of interest in the monomers are LTPD at Xl-4 and KRALE at X16-20 and AHG at X32-34 or LTPE at Xl-4 and KRALE at XI 6- 20 and DHG at X32-34 when the monomer is 34 amino acids in length.
  • the monomer is 33 or 35 amino acids long, the corresponding shift occurs in the positions of the contiguous amino acids KRALE, AHG and DHG.
  • the positions of the contiguous amino acids may be (LTPD at Xl-4 and KRALE at X15-19 and AHG at X31-33) or (LTPE at Xl-4 and KRALE at X15-19 and DHG at X31-33) or (LTPD at Xl-4 and KRALE at X17-21 and AHG at X33-35) or (LTPE at Xl-4 and KRALE at X17-21 and DHG at X33-35).
  • contiguous amino acids [NGKQALE] are present at positions X14-20 or X13-19 or X15-21.
  • TALE polypeptide binding efficiency is increased by including amino acid sequences from the “capping regions” that are directly N-terminal or C- terminal of the DNA binding region of naturally occurring TALEs into the engineered TALEs at positions N-terminal or C-terminal of the engineered TALE DNA binding region.
  • the TALE polypeptides described herein further comprise an N-terminal capping region and/or a C-terminal capping region.
  • An exemplary amino acid sequence of a N-terminal capping region is: MDPIRSRTPSPARELLSGPQPDGVQPTADRGVSPPAGGPLDGLPARRTMSRTRLPSPPAPSPAF SADSFSDLLRQFDPSLFNTSLFDSLPPFGAHHTEAATGEWDEVQSGLRAADAPPPTMRVAVTAA RPPRAKPAPRRRAAQPSDASPAAQVDLRTLGYSQQQQEKIKPKVRSTVAQHHEALVGHGFTHAH IVALSQHPAALGTVAVKYQDMIAALPEATHEAIVGVGKQWSGARALEALLTVAGELRGPPLQLD TGQLLKIAKRGGVTAVEAVHAWRNALTGAPLN [00517] An exemplary amino acid sequence of a C-terminal capping region is: RPALESIVAQLSRPDPALAALTNDHLVALACLGGRPALDAVKKGLPHAPALIKRTNRRIPERTS HRVADHAQVVRVLGFFQCHSHPAQAFDDAMTQFGMSR
  • N-terminal and/or C-terminal capping regions are not necessary to enhance the binding activity of the DNA binding region. Therefore, in certain embodiments, fragments of the N-terminal and/or C-terminal capping regions are included in the TALE polypeptides described herein.
  • the TALE (including TALEs) polypeptides described herein contain a N-terminal capping region fragment that included at least 10, 20, 30, 40, 50, 54, 60, 70, 80, 87, 90, 94, 100, 102, 110, 117, 120, 130, 140, 147, 150, 160, 170, 180, 190, 200, 210, 220, 230, 240, 250, 260 or 270 amino acids of an N-terminal capping region.
  • the N-terminal capping region fragment amino acids are of the C-terminus (the DNA-binding region proximal end) of an N-terminal capping region.
  • N-terminal capping region fragments that include the C-terminal 240 amino acids enhance binding activity equal to the full length capping region, while fragments that include the C-terminal 147 amino acids retain greater than 80% of the efficacy of the full length capping region, and fragments that include the C-terminal 117 amino acids retain greater than 50% of the activity of the full-length capping region.
  • the TALE polypeptides described herein contain a C- terminal capping region fragment that included at least 6, 10, 20, 30, 37, 40, 50, 60, 68, 70, 80, 90, 100, 110, 120, 127, 130, 140, 150, 155, 160, 170, 180 amino acids of a C-terminal capping region.
  • the C-terminal capping region fragment amino acids are of the N-terminus (the DNA-binding region proximal end) of a C-terminal capping region.
  • C-terminal capping region fragments that include the C-terminal 68 amino acids enhance binding activity equal to the full length capping region, while fragments that include the C-terminal 20 amino acids retain greater than 50% of the efficacy of the full length capping region.
  • the capping regions of the TALE polypeptides described herein do not need to have identical sequences to the capping region sequences provided herein.
  • the capping region of the TALE polypeptides described herein have sequences that are at least 50%, 60%, 70%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% identical or share identity to the capping region amino acid sequences provided herein.
  • Sequence identity is related to sequence homology. Homology comparisons may be conducted by eye, or more usually, with the aid of readily available sequence comparison programs. These commercially available computer programs may calculate percent (%) homology between two or more sequences and may also calculate the sequence identity shared by two or more amino acid or nucleic acid sequences.
  • the capping region of the TALE polypeptides described herein have sequences that are at least 95% identical or share identity to the capping region amino acid sequences provided herein.
  • Sequence homologies may be generated by any of a number of computer programs known in the art, which include but are not limited to BLAST or FASTA. Suitable computer program for carrying out alignments like the GCG Wisconsin Bestfit package may also be used. Once the software has produced an optimal alignment, it is possible to calculate % homology, preferably % sequence identity. The software typically does this as part of the sequence comparison and generates a numerical result.
  • % homology may be calculated over contiguous sequences, i.e., one sequence is aligned with the other sequence and each amino acid or nucleotide in one sequence is directly compared with the corresponding amino acid or nucleotide in the other sequence, one residue at a time. This is called an “ungapped” alignment. Typically, such ungapped alignments are performed only over a relatively short number of residues.
  • the TALEs described herein also include a nuclear localization signal and/or cellular uptake signal. Such signals are known in the art and may target a TALE to the nucleus and/or intracellular compartment of a cell. Such cellular uptake signals include, but are not limited to, the minimal Tat protein transduction domain which spans residues 47-57 of the human immunodeficiency virus Tat protein: YGRKKRRQRRR. [00526] In some embodiments, the TALEs described herein include a nucleic acid or DNA binding domain that is a non-TALE nucleic acid or a non-TALE DNA binding domain.
  • non-TALE DNA binding domain refers to a DNA binding domain that has a nucleic acid sequence corresponding to a nucleic acid sequence which is not substantially homologous to a nucleic acid that encodes for a TALE protein or fragment thereof, e.g., a nucleic acid sequence which is different from a nucleic acid that encodes for a TALE protein and which is derived from the same or a different organism.
  • the TALEs described herein include a nucleic acid or DNA binding domain that is linked to a non-TALE polypeptide.
  • a “non-TALE polypeptide” refers to a polypeptide having an amino acid sequence corresponding to a protein which is not substantially homologous to a TALE protein or fragment thereof, e.g., a protein which is different from a TALE protein and which is derived from the same or a different organism.
  • the term “linked” is intended include any manner by which the nucleic acid binding domain and the non-TALE polypeptide could be connected to each other, including, for example, through peptide bonds by being part of the same polypeptide chain or through other covalent interactions, such as a chemical linker.
  • the non- TALE polypeptide may be linked, for example to the N-terminus and/or C-terminus of the nucleic acid binding domain, may be linked to a C-terminal or N-terminal cap region, or may be connected to the nucleic acid binding domain indirectly.
  • the TALEs or polypeptides of the invention comprise chimeric DNA binding domains.
  • Chimeric DNA binding domains may be generated by fusing a full TALE (including the N- and C- terminal capping regions) with another TALE or non-TALE DNA binding domain such as zinc finger (ZF), helix-loop-helix, or catalytically-inactivated DNA endonucleases (e.g., EcoRI, meganucleases, etc.), or parts of TALE may be fused to other DNA binding domains.
  • the chimeric domain may have novel DNA binding specificity that combines the specificity of both domains.
  • the TALE polypeptides of the invention include a nucleic acid binding domain linked to the one or more effector domains.
  • the effector domain is a nickase or nuclease.
  • the sequence-specific nuclease is a zinc finger nuclease (ZFN), such as an artificial zinc-finger nuclease having arrays of zinc-finger (ZF) modules to target new DNA-binding sites in a target sequence (e.g., target sequence or target site in the genome).
  • ZFN zinc finger nuclease
  • ZF zinc-finger
  • Each zinc finger module in a ZF array targets three DNA bases.
  • a customized array of individual zinc finger domains is assembled into a ZF protein (ZFP). The resulting ZFP can be linked to a functional domain such as a nuclease.
  • ZF nucleases may be used as alternative programmable nucleases for use in retron-based editing in place of RNA-guide nucleases.
  • ZFN proteins have been extensively described in the art, for example, in Carroll et al.,“Genome Engineering with Zinc-Finger Nucleases,” Genetics, Aug 2011, Vol.188: 773-782; Durai et al.,“Zinc finger nucleases: custom- designed molecular scissors for genome engineering of plant and mammalian cells,” Nucleic Acids Res, 2005, Vol.33: 5978-90; and Gaj et al., “ZFN, TALEN, and CRISPR/Cas-based methods for genome engineering,” Trends Biotechnol.2013, Vol.31: 397-405, each of which are incorporated herein by reference in their entireties.
  • the ZF-linked nuclease is a catalytic domain of the Type IIS restriction enzyme FokI (see Kim et al., PNAS U.S.A.91:883-887, 1994; Kim et al., PNAS U.S.A.93:1156-1160, 1996, both incorporated herein by reference).
  • the ZFN comprises paired ZFN heterodimers, resulting in increased cleavage specificity and/or decreased off-target activity.
  • each ZFN in the heterodimer targets different nucleotide sequences separated by a short spacer (see Doyon et al., Nat.
  • the ZFN comprises a polynucleotide-binding domain (comprising multiple sequence-specific ZF modules) and a polynucleotide cleavage nickase domain.
  • the ZFs are engineered using libraries of two finger modules.
  • strings of two-finger units are used in ZFNs to improve DNA binding specificity from polyzinc finger peptides (see PNAS USA 98: 1437-1441, incorporated herein by reference).
  • the ZFN has more than 3 fingers.
  • the ZFN has 4, 5, or 6 fingers.
  • the ZF modules in the ZFN are separated by one or more linkers to improve specificity.
  • the ZF of the ZFN includes substitutions in the dimer interface of the cleavage domain that prevent homodimerization between ZFs, but allow heterodimers to form.
  • the ZF of the ZFN has a design that retains activity while suppressing homodimerization.
  • the ZFN is any one of the ZF nucleases in Table 1 of Carroll et al., Genetics 188(4):773-782, 2011, incorporated herein by reference.
  • Additional, non-limiting ZFs and AFNz that can be adapted for use in the instant invention include those described in WO2010/065123, WO2000/041566, WO2003/080809, WO2015/143046, WO2016/183298, WO2013/044008, WO2015/031619, WO2017/136049, WO2016/014794, WO2017/091512, WO1995/009233, WO2000/023464, WO2000/042219, WO2002/026960, WO2001/083793; US9428756, US9145565, US8846578, US8524874, US6777185, US6599692, US7235354, US6503717, US7491531, US7943553, US7262054, US8680021, US7705139, US7273923, US6780590, US6785613, US7788044, US7177766, US6453242, US6794136, US7358085, US838376
  • Polynucleotides and vectors capable of expressing one or more of the ZFNs are also provided herein, which can be part of the vector system of the invention.
  • the polynucleotides and vectors can be expressed in a cell, such as a eukaryotic cell, a mammalian cell, or a human cell.
  • a cell such as a eukaryotic cell, a mammalian cell, or a human cell.
  • Suitable vectors, cells and expression systems are described in greater detail elsewhere herein, and can be suitable for use with the TALEs, the meganucleases, and the CRISPR-Cas nucleases.
  • the sequence-specific nuclease is a meganuclease.
  • Meganucleases are a class of sequence-specific endonucleases that recognize large DNA target sites (>12 bp). These proteins can cleave a unique chromosomal sequence without affecting overall genome integrity. Meganucleases create site specific DNA DSBs, and, in the presence of donor DNA, such as one present in the heterologous nucleic acid encompassed by or encoded by the engineered retron of the invention, promotes the integration of the donor DNA at the cleavage site through homologous recombination (HR).
  • HR homologous recombination
  • the meganuclease is a homing endonuclease, which is a widespread class of proteins found in eukaryotes, bacteria and archaea.
  • the meganuclease is of the LAGLIDADG family of homing endonucleases.
  • the meganuclease is I-Scel, I-Cre-I, I-Dmol, or an engineered or a naturally occurring variant thereof. The hallmark of these proteins is a well conserved LAGLIDADG peptide motif, termed (do)decapeptide, found in one or two copies.
  • Additional homing nucleases are found at the website of homingendonuclease.net, which provides a database listing basic properties of known LAGLIDADG homing endonucleases. See also Taylor et al., Nucleic Acids Research 40 (Wl): W110-W116, 2012 (all incorporated herein by reference).
  • specificity (or polynucleotide recognition) of the meganuclease is modified by altering the amino acids within the meganuclease, and/or by fusing other effector domains with the meganuclease.
  • the meganuclease is a megaTAL, which includes a DNA binding domain from a TALE.
  • the meganuclease is engineered to have nickase activity.
  • the sequence-specific nuclease is TnpB, which is a programmable RNA-guided DNA endonuclease. It is believed that TnpB is a functional progenitor of the CRISPR-Cas nucleases.
  • TnpB is a functional progenitor of the CRISPR-Cas nucleases.
  • Transposons are mobile genetic elements that contain only the genes required for their transposition and its regulation. These elements encode the tnpA transposase, which is essential for mobilization, and often carry an accessory tnpB gene, which is dispensable for transposition.
  • TnpB has been shown to be a nuclease that is guided by an RNA, derived from the right-end element of a transposon, to cleave DNA next to the 5′-TTGAT transposon-associated motif, and TnpB can be reprogrammed to cleave DNA target sites in human cells.
  • tnpB is from D. radiodurans ISDra2 of the IS200/IS605 family.
  • tnpB is from transposon PsiTn554.
  • the sequence-specific nuclease is a TnpB-like protein, such as Fanzor1 or Fanzor2.
  • Fanzor and TnpB proteins share the same conserved amino acid motif in their C-terminal half regions: D-X(125, 275)-[TS]-[TS]-X-X-[C4 zinc finger]-X(5,50)-RD, but are highly variable in their N-terminal regions.
  • Fanzor1 proteins are frequently captured by DNA transposons from different superfamilies including Helitron, Mariner, IS4-like, Sola and MuDr. In contrast, Fanzor2 proteins appear only in some IS607-type elements.
  • the sequence-specific nuclease is IscB.
  • ISC insertion sequences Cas9-like
  • ISC transposon-encoded two nuclease domain-containing proteins are the likely ancestors of the CRISPR-associated Cas9.
  • the homology region includes the arginine-rich helix and the HNH nuclease domain that is inserted into the RuvC-like nuclease domain.
  • ISC genes are not linked to Cas genes or CRISPR. They represent a distinct group of nonautonomous transposons, with many diverse families of full-length ISC transposons.
  • terminal sequences are similar to those of IS605 superfamily transposons that are mobilized by the Y1 tyrosine transposase encoded by the TnpA gene, and often also encode the TnpB protein containing the RuvC-like endonuclease domain.
  • the terminal regions of the ISC and IS605 transposons contain palindromic structures that are likely recognized by the Y1 transposase. The transposons from these two groups are inserted either exactly in the middle or upstream of specific 4-bp target sites, without target site duplication.
  • the sequence-specific nuclease is a restriction endonuclease (RE), such as an RE with stringent / long recognition sequence of at least 8 nts.
  • RE restriction endonuclease
  • the RE is a rare-cutter RE with seven and eight base pair recognition sequences.
  • Exemplary rare-cutter RE enzyme include NotI, which cuts after the first GC of a 5 ⁇ -GCGGCCGC-3 ⁇ sequence.
  • the components of the system – e.g., the retron encoded ncRNA or msDNA in complex with the RT, the sequence-specific nuclease, and the DNA-repair modulating biomolecule, may form multiple complexes in a so-called split complex configuration.
  • the multiple complexes may be brought together to form a functional complex.
  • a first component in the system may be a split protein or domain.
  • a fragment of the split protein or domain may associate with a second component of the system, while another fragment of the split protein or domain may associate with a third component of the system.
  • the split protein or domain is the sequence-specific nuclease, e.g., a CRISPR/Cas effector enzyme (e.g., Cas protein such as Cas9 or Cas12), a ZFN, a TALEN, a meganuclease, TnpB, IscB, or a restriction endonuclease (RE).
  • a CRISPR/Cas effector enzyme e.g., Cas protein such as Cas9 or Cas12
  • a ZFN e.g., Cas protein such as Cas9 or Cas12
  • TALEN e.g., TALEN, a meganuclease, TnpB, IscB, or a restriction endonuclease (RE).
  • the split protein or domain is the reverse transcriptase domain.
  • the split protein or domain is the DNA-repair modulating biomolecule.
  • a first fragment of the sequence-specific nuclease may associate with the reverse transcriptase domain and a second fragment of the sequence-specific nuclease may associate with the DNA-repair modulating biomolecule.
  • the two fragments of the split protein or domain may be brought together (e.g., along with the reverse transcriptase domain and the DNA- repair modulating biomolecule) to form a functional complex.
  • the associations between the parts of the split protein or domain may be through adaptor proteins or linkers described herein (e.g., those used for associating Cas proteins with function domains).
  • the split protein or domain is split in the sense that the two parts of the split protein or domain substantially comprise a functioning split protein or domain. Ideally, the split should always be so that the catalytic domain(s) are unaffected. That split protein or domain may function as a sequence-specific nuclease or it may be a dead-Cas which is essentially an RNA-binding protein with very little or no catalytic activity, due to typically mutation(s) in its catalytic domains. [00571] Each fragment of the split protein or domain may be fused to a dimerization partner. For example, rapamycin sensitive dimerization domains enables a chemically inducible split protein or domain for temporal control of the split protein or domain’s activity.
  • the split protein or domain can thus be rendered chemically inducible by being split into two fragments and that rapamycin-sensitive dimerization domains may be used for controlled reassembly of the split protein or domain.
  • the two parts of the split protein or domain can be thought of as the N’ terminal part and the C’ terminal part of the split protein or domain.
  • the fusion is typically at the split point of the split protein or domain.
  • the C’ terminal of the N’ terminal part of the split protein or domain is fused to one of the dimer halves, whilst the N’ terminal of the C’ terminal part is fused to the other dimer half.
  • the split protein or domain does not have to be split in the sense that the break is newly created.
  • the split point is typically designed in silico and cloned into the constructs. Together, the two parts of the split protein or domain, the N’ terminal and C’ terminal parts, form a full split protein or domain, comprising preferably at least 70% or more of the wildtype amino acids (or nucleotides encoding them), at least 80% or more, at least 90% or more, at least 95% or more, and at least 99% or more of the wildtype amino acids (or nucleotides encoding them). When the two parts are brought together, the desired split protein or domain function is restored or reconstituted.
  • the dimer may be a homodimer or a heterodimer.
  • the protein components of the system – e.g., the RT, the sequence-specific nuclease (a CRISPR/Cas effector enzyme, a ZFN, a TALEN, a meganuclease, TnpB, IscB, or a restriction endonuclease (RE)), and the DNA-repair modulating biomolecule may further comprise one or more additional functional domains.
  • the functional domain comprises a nuclear localization signal (NLS).
  • NLS nuclear localization signal
  • one or more C-terminal or N-terminal NLSs are attached.
  • a C-terminal NLS is attached for expression and nuclear targeting in eukaryotic cells, e.g., human cells.
  • the NLS(s) may be at a location that is not at the C-terminus or N-terminus, for example, the NLS(s) may be between two polypeptides.
  • Non-limiting examples of NLSs include an NLS sequence derived from: the NLS of the SV40 virus large T-antigen; the NLS from nucleoplasmin (e.g., the nucleoplasmin bipartite NLS); the c-myc NLS; the hRNPAl M9 NLS; the NLS of the IBB domain from importin-alpha; the NLS of the myoma T protein; the NLS of human p53; the NLS of mouse c- abl IV; the NLS of the influenza virus NS1; the NLS of the Hepatitis virus delta antigen; the NLS of the mouse Mxl protein; the NLS of the human poly(ADP-ribose) polymerase; and the NLS of the steroid hormone receptors (human) glucocorticoid.
  • nucleoplasmin e.g., the nucleoplasmin bipartite NLS
  • the c-myc NLS e.g., the nucleoplasm
  • the functional domain comprises at least two NLS domains.
  • the one or more NLS domain(s) may be positioned at or near or in proximity to a terminus of a polypeptide and, if two or more NLSs, each of the two may be positioned at or near or in proximity to a terminus of the polypeptide.
  • the fusion between the two domains (such as the RT and Cas enzyme, or the Cas and the DNA-repair modulating biomolecule) may be linked through a linker.
  • a “linker” as used herein includes a peptide which joins two proteins or domains to form a fusion protein. Generally, such molecules have no specific biological activity other than to join or to preserve some minimum distance or other spatial relationship between the proteins / domains. However, in certain embodiments, the linker may be selected to influence some property of the linker and/or the fusion protein such as the folding, net charge, or hydrophobicity of the linker. Suitable linkers for use in the present disclosure are well-known to those of skill in the art and include, but are not limited to, straight or branched-chain carbon linkers, heterocyclic carbon linkers, or peptide linkers.
  • the linker may also be a covalent bond (carbon-carbon bond or carbon-heteroatom bond).
  • the linker is used to separate the sequence-specific nuclease (a CRISPR/Cas effector enzyme, a ZFN, a TALEN, a meganuclease, TnpB, IscB, or a restriction endonuclease (RE)) and the RT and/or the DNA-repair modulating biomolecule by a distance sufficient to ensure that each protein domain retains its required functional property.
  • sequence-specific nuclease a CRISPR/Cas effector enzyme, a ZFN, a TALEN, a meganuclease, TnpB, IscB, or a restriction endonuclease (RE)
  • RE restriction endonuclease
  • Preferred peptide linker sequences adopt a flexible extended conformation and do not exhibit a propensity for developing an ordered secondary structure.
  • the linker can be a chemical moiety which can be monomeric, dimeric, multimeric or polymeric.
  • the linker comprises amino acids. Typical amino acids in flexible linkers include Gly, Asn and Ser. Accordingly, in particular embodiments, the linker comprises a combination of one or more of Gly, Asn and Ser amino acids. Other near neutral amino acids, such as Thr and Ala, also may be used in the linker sequence.
  • the linker comprises a G 4 S linker with 3, 6, 9, or 12 repeats.
  • the linker is one disclosed in Maratea et al., Gene 40: 39- 46, 1985; Murphy et al., PNAS USA 83: 8258-62, 1986; US4,935,233; or US4,751,180, all incorporated by reference.
  • the linker comprises a GlySer linker such as GGS, GGGS, GSG, GGGGS, optionally with repeats of 3 (such as (GGS) 3 , (GGGGS) 3 ), 4, 5, 6, 7, 8, 9, 10, 11, or 12 or more, to provide suitable lengths.
  • the linker comprises (GGGGS) 3-15 , such as (GGGGS) 3-11 , e.g., GGGGS with 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, or 11 repeats.
  • the linker comprises LEPGEKPYKCPECGKSFSQSGALTRHQR THTR.
  • the linker is an XTEN linker.
  • N-and/or C-terminal NLSs also function as linker (e.g., PKKKRKVEASSPKKRKVEAS).
  • the gRNA and the various nucleases and fusions thereof can be provided in the form of a protein, optionally where the nuclease is complexed with a gRNA, or provided by a nucleic acid encoding the RNA-guided nuclease, such as an RNA (e.g., messenger RNA) or DNA (expression vector).
  • a nucleic acid encoding the RNA-guided nuclease such as an RNA (e.g., messenger RNA) or DNA (expression vector).
  • the RNA-guided nuclease and the gRNA are both provided by vectors. Both can be expressed by a single vector or separately on different vectors.
  • the vectors encoding the RNA-guided nuclease and gRNA may be included in the vector system comprising the engineered retron msr gene, msd gene and ret gene sequences.
  • Codon usage may be optimized to improve production of the engineered retron e.g., retron reverse transcriptase, ncRNA and/or RNA-guided nuclease in a particular cell or organism.
  • a nucleic acid encoding an ncRNA, RNA-guided nuclease or reverse transcriptase can be modified to substitute codons having a higher frequency of usage in a the particular cell such as a eukaryotic cell (e.g., yeast cell, a human cell, a non-human cell, a mammalian cell, a rodent cell, a mouse cell, a rat cell), or any other host cell of interest, as compared to the naturally occurring polynucleotide sequence.
  • a nucleic acid encoding the reverse transcriptase or ncRNA is introduced into cells, the protein can be transiently, conditionally, or constitutively expressed in the cell.
  • the recombinant retron-based editing system described herein contemplates fusion proteins comprising a programmable nuclease (PN) and a RT, optionally joined by a linker.
  • PN programmable nuclease
  • the application contemplates any suitable programmable nuclease and RT (e.g., retron RTs of Table A) to be combined in a single fusion protein.
  • the RT is joined to the N-terminus of the PN.
  • the RT is joined to the C-terminus of the PN.
  • the Examples of PNs and RTs are each defined herein.
  • the fusion proteins may comprise any suitable structural configuration.
  • the fusion protein may comprise from the N-terminus to the C- terminus direction, a PN fused to a RT.
  • the fusion protein may comprise from the N-terminus to the C-terminus direction, a RT fused to a NP.
  • the fused domain may optionally be joined by a linker, e.g., an amino acid sequence.
  • a linker e.g., an amino acid sequence.
  • any of the polypeptide components of the retron-based editing systems may be engineered with one or more nuclear locatlization signals, which help promote translocation of a protein into the cell nucleus.
  • the polypeptides of the retron-based editing system may comprise any known NLS sequence, including any of those described in Cokol et al.,“Finding nuclear localization signals,” EMBO Rep., 2000, 1(5): 411-415 and Freitas et al.,“Mechanisms and Signals for the Nuclear Import of Proteins,” Current Genomics, 2009, 10(8): 550-7, each of which are incorporated herein by reference.
  • the polypeptides disclosed herein may include one or more, preferably, at least two nuclear localization signals.
  • the NLSs may be any known NLS sequence in the art.
  • the NLSs may also be any future-discovered NLSs for nuclear localization.
  • the NLSs also may be any naturally-occurring NLS, or any non-naturally occurring NLS (e.g., an NLS with one or more desired mutations).
  • the term“nuclear localization sequence” or“NLS” refers to an amino acid sequence that promotes import of a protein into the cell nucleus, for example, by nuclear transport. Nuclear localization sequences are known in the art and would be apparent to the skilled artisan. For example, NLS sequences are described in Plank et al., International PCT application PCT/EP2000/011690, filed November 23, 2000, published as WO/2001/038547 on May 31, 2001, the contents of which are incorporated herein by reference.
  • an NLS comprises the amino acid sequence PKKKRKV.
  • a representative nuclear localization signal is a peptide sequence that directs the protein to the nucleus of the cell in which the sequence is expressed.
  • a nuclear localization signal is predominantly basic, can be positioned almost anywhere in a protein's amino acid sequence, generally comprises a short sequence of four amino acids (Autieri & Agrawal, (1998) J. Biol. Chem.273: 14731-37, incorporated herein by reference) to eight amino acids, and is typically rich in lysine and arginine residues (Magin et al., (2000) Virology 274: 11-16, incorporated herein by reference). Nuclear localization signals often comprise proline residues.
  • nuclear localization signals have been identified and have been used to effect transport of biological molecules from the cytoplasm to the nucleus of a cell. See, e.g., Tinland et al., (1992) Proc. Natl. Acad. Sci. U.S.A.89:7442-46; Moede et al., (1999) FEBS Lett.461:229-34, which is incorporated by reference. Translocation is currently thought to involve nuclear pore proteins.
  • NLSs can be classified in three general groups: (i) a monopartite NLS exemplified by the SV40 large T antigen NLS (PKKKRKV); (ii) a bipartite motif consisting of two basic domains separated by a variable number of spacer amino acids and exemplified by the Xenopus nucleoplasmin NLS (KRXXXXXXXXKKKL); and (iii) noncanonical sequences such as M9 of the hnRNP Al protein, the influenza virus nucleoprotein NLS, and the yeast Gal4 protein NLS (Dingwall and Laskey 1991). Nuclear localization signals appear at various points in the amino acid sequences of proteins.
  • NLS NLS’s have been identified at the N-terminus, the C- terminus and in the central region of proteins.
  • the disclosure provides polypeptides that may be modified with one or more NLSs at the C-terminus, the N-terminus, as well as at in internal region of a polypeptide (including a fusion protein).
  • the present disclosure contemplates any suitable means by which to modify a polypeptide to include one or more NLSs.
  • a polypeptide e.g., a programmable nuclease
  • the NLSs may include various amino acid linkers or spacer regions encoded between a polypeptide and the N-terminally, C-terminally, or internally-attached NLS amino acid sequence, e.g, and in the central region of proteins.
  • the present disclosure also provides for nucleotide constructs, vectors, and host cells for expressing fusion proteins that comprise a polypeptide and one or more NLSs. H.
  • the engineered retron described herein e.g., an engineered nucleic acid construct or engineered nucleic acid-enzyme construct described herein
  • the engineered retron described herein further comprises or encodes a DNA-repair modulating biomolecule, which may further enhance the efficiency of integration of a transgene on the heterologous nucleic acid by homology dependent repair (HDR).
  • HDR homology dependent repair
  • the DNA-repair modulating biomolecule comprises a Nonhomologous end joining (NHEJ) inhibitor.
  • NHEJ Nonhomologous end joining
  • the DNA-repair modulating biomolecule comprises a homologous directed repair (HDR) promoter.
  • the DNA-repair modulating biomolecule comprises a NHEJ inhibitor and an HDR promoter.
  • the DNA-repair modulating biomolecule enhances or improves more precise genome editing and/or the efficiency of homologous recombination, compared to the otherwise identical embodiment without the DNA-repair modulating biomolecule.
  • HDR promoters and/or NHEJ inhibitors can, in some embodiments, comprise one or more small molecules. Systems bearing recombination enhancers such as small molecules that activate HDR and suppress NHEJ locally at the genomic site of the DNA damage can be tailored in their placement on the engineered systems to further enhance their efficiency.
  • the small molecule recombination enhancers can be synthesized to bear linkers and a functional group, such as maleimide for reacting with a thiol group on a Cys residue of a protein, for chemical conjugation to the engineered systems.
  • a functional group such as maleimide
  • PEG linkers alkyne, azide, cyclooctyne etc.
  • orthogonal conjugation chemistries can be utilized for the multivalent display.
  • multivalent display of one or more DNA-repair modulating biomolecule can be affected, including multiple moieties of NHEJ inhibitors, HDR promoters, or a combination thereof. See, for example, “Genomic targeting of epigenetic probes using a chemically tailored Cas9 system” by Liszczak et al., Proc Natl Acad Sci U.S.A.114: 681- 686, 2017 (incorporated herein by reference).
  • multivalent display of small molecule compounds can be achieved through sortase loop proteins used as a scaffold for their display.
  • the DNA-repair modulating biomolecule may comprise an HDR promoter.
  • the HDR promoter may comprise small molecules, such as RSI or analogs thereof.
  • the HDR promoter stimulates RAD51 activity or RAD52 motif protein 1 (RDMl) activity.
  • the HDR promoter comprises Nocodazole, which can result in higher HDR selection.
  • the HDR promoter may be administered prior to the delivery of the engineered retron described herein.
  • the HDR promoter locally enhances HDR without NHEJ inhibition.
  • RAD5l is a protein involved in strand exchange and the search for homology regions during HDR repair.
  • the HDR promoter is phenylbenzamide RSI, identified as a small-molecule RAD51-stimulator (see WO2019/135816 at [0200]-[0204], specifically incorporated herein by reference).
  • the DNA-repair modulating biomolecule comprises C- terminal binding protein interacting protein (CtIP) or a functional fragment or homolog thereof.
  • CtIP is a key protein in early steps of homologous recombination.
  • the CtIP or the functional fragment or homolog thereof can be linked (e.g., fused) to the RT or the sequence-specific nuclease (e.g., a CRISPR/Cas effector enzyme, a ZFN, a TALEN, a meganuclease, TnpB, IscB, or a restriction endonuclease (RE)), and stimulates transgene integration by HDR.
  • sequence-specific nuclease e.g., a CRISPR/Cas effector enzyme, a ZFN, a TALEN, a meganuclease, TnpB, IscB, or a restriction endonuclease (RE)
  • the CtIP fragment is a minimal N-terminal fragment of the wild-type CtIP, such as the N-terminal fragment comprising residues 1-296 of the full-length CtIP (the HE for HDR enhancer), as described in Charpentier et al. (Nature Comm., DOI: 10.1038/s41467-018-03475-7, incorporated herein by reference), shown to be sufficient to stimulate HDR.
  • the activity of the fragment depends on CDK phosphorylation sites (e.g., S233, T245, and S276) and the multimerization domain essential for CtIP activity in homologous recombination.
  • the DNA-repair modulating biomolecule comprises a dominant negative 53BP1.
  • the DNA-repair modulating biomolecule comprises a cell cycle-specific degradation tag, such as the degradation domain of the (human) Geminin, and the (murine) CyclinB2.
  • the DNA-repair modulating biomolecule comprises CyclinB2, a member of the B-type cyclins that associate with p34cdc2, and an essential component of the cell cycle regulatory machinery.
  • the CRISPR-mediated knock-in efficiency may be increased by promoting the relative increase in Cas9 activity in G2 phase of the cell cycle, when HDR is more active.
  • the degradation domains of the (human) Geminin and (murine) CyclinB2 can be used as either N- or C-terminal fusion (e.g., fusions with a Cas, such as Cas9, or the retron RT) to serve as the DNA-repair modulating biomolecule.
  • N- or C-terminal fusion e.g., fusions with a Cas, such as Cas9, or the retron RT
  • These domains are known to determine a cell-cycle specific profile of chimeric proteins, namely an increase in their relative concentration in S and G2 compared to G1, high-jacking the conventional CyclinB2 and Geminin degradation pathways.
  • the DNA-repair modulating biomolecule comprises a Rad family member protein, such as Rad50, Rad51, Rad52, etc., which functions to promote foreign DNA integration into a host chromosome.
  • Rad52 is an important homologous recombinant protein, and its complex with Rad51 plays a key role in HDR, mainly involved in the regulation of foreign DNA in eukaryotes. Key steps in the process of HR include repair mediated by Rad51 and strand exchange. Co-expression of Rad52 as a DNA-repair modulating biomolecule significantly enhances the likelihood of HDR by, e.g., three-fold.
  • the DNA-repair modulating biomolecule comprises a RAD52 protein as, e.g., either an N- or a C-terminal fusion.
  • the DNA-repair modulating biomolecule comprises a RAD52 motif protein 1 (RDMl) that functions similarly as RAD52.
  • RDM1 has been shown to be able to repair DSBs caused by DNA replication, prevent G2 or M cell cycle arrest, and improve HDR selection.
  • the DNA-repair modulating biomolecule comprises a dominant negative version of the tumor suppressor p53-binding protein 1 (53BP1).
  • the wild- type protein 53BP1 is a key regulator of the choice between NHEJ and HDR – it is a pro-NHEJ factor which limits HDR by blocking DNA end resection, and also by inhibiting BRCA1 recruitment to DSB sites.
  • the dominant negative (DN) version of the 53BP1 comprises the minimal focus forming region, but lacks domains outside this region, e.g., towards the N-terminus and tandem C-terminal BRCT repeats that recruit key effectors involved in NHEJ, such as RIFl-PTIP and EXPAND, respectively.
  • the 53BP1 adapter protein is recruited to specific histone marks at sites of DSBs via this minimal focus forming region, which comprises several conserved domains including an oligomerization domain (OD), a glycine-arginine rich (GAR) motif, a Vietnamese domain, and an adjacent ubiquitin-dependent recruitment (UDR) motif.
  • the Tudor domain mediates interactions with histone H4 dimethylated at K2023.
  • a dominant negative version of 53BP1 suppresses the accumulation of endogenous 53BP1 and downstream NHEJ proteins at sites of DNA damage, while upregulating the recruitment of the BRCA1 HDR protein.
  • the DNA-repair modulating biomolecule can be used as the DNA-repair modulating biomolecule, either as an N- or a C-terminal fusion (such as a Cas9 fusion, to locally inhibit NHEJ at the Cas9-target site defined by its gRNA, while promoting an increase in HDR, and does not globally affect NHEJ, thereby improving cell viability).
  • the DNA-repair modulating biomolecule comprises an NHEJ inhibitor, such as an inhibitor of DNA ligase IV, a KU inhibitor (e.g., KU70 or KU80), a DNA-PKc inhibitor, or an artemis inhibitor.
  • the NHEJ inhibitor inhibits the NHEJ pathway, enhances HDR, or modulates both.
  • the NHEJ inhibitor is a small molecule inhibitor.
  • the small molecule inhibitor of the NHEJ pathway comprises an SCR7 analog, for example, PK66, PK76, PK409.
  • the NHEJ inhibitor comprises a KU inhibitor, for example, KU5788, and KU0060648.
  • a small molecule NHEJ inhibitor is linked to a polyglycine tripeptide through PEG for sortase-mediated ligation, as described in WO2019/135816, Guimaraes et al., Nat Protoc 8:1787-99, 2013; Theile et al., Nat Protoc 8:1800-7, 2013; and Schmohl et al., Curr Opin Chem Biol 22:122-8, 2014 (all incorporated herein by reference). The same means can also be used for attaching small molecule HDR enhancers to protein.
  • a nucleic acid targeting moiety conjugates based on small molecule inhibitor of DNA-dependent protein kinase (DNA-PK) or heterodimeric Ku (KU70/KU80) can be utilized.
  • KU-0060648 is one potent KU-inhibitors, which can also be functionalized with poly-glycine and used for recombination enhancement.
  • the DNA-repair modulating biomolecule comprises the Tumor Suppressor p53. p53 plays a direct role in DNA repair, including HR regulation, where it affects the extension of new DNA, thereby affecting HDR selection.
  • p53 In vivo, p53 binds to the nuclear matrix and is a rate-limiting factor in repairing DNA structure. p53 regulates DNA repair processes in almost all eukaryotes via transactivation-dependent and -independent pathways, but only the transactivation-independent function of p53 is involved in HR regulation. Wild-type p53 protein can link double stranded breaks to form intact DNA, as well as also playing a role in inhibiting NHEJ. p53 interacts with HR-related proteins, including Rad51, where it controls HR through direct interaction with Rad51.
  • Delivery of an engineered retron to a cell can generally be accomplished with or without vectors.
  • Delivery of the ncRNA encoded by the engineered retron generally does not require a vector used to produce the ncRNA from the engineered retron.
  • the ncRNA can be packaged directly into a delivery vehicle such as a lipid nanoparticle and delivered into a host cell, as described in other sections.
  • the engineered retrons may be introduced into any type of cell, including any cell from a prokaryotic, eukaryotic, or archaeon organism, including bacteria, archaea, fungi, protists, plants (e.g., monocotyledonous and dicotyledonous plants); and animals (e.g., vertebrates and invertebrates).
  • Examples of animals that may be transfected with an engineered retron include, without limitation, vertebrates such as fish, birds, mammals (e.g., human and non-human primates, farm animals, pets, and laboratory animals), reptiles, and amphibians.
  • the engineered retrons can be introduced into a single cell or a population of cells. Cells from tissues, organs, and biopsies, as well as recombinant cells, genetically modified cells, cells from cell lines cultured in vitro, and artificial cells (e.g., nanoparticles, liposomes, polymersomes, or microcapsules encapsulating nucleic acids) may all be transfected with the engineered retrons.
  • the engineered retrons can be introduced into cellular fragments, cell components, or organelles (e.g., mitochondria in animal and plant cells, plastids (e.g., chloroplasts) in plant cells and algae).
  • organelles e.g., mitochondria in animal and plant cells, plastids (e.g., chloroplasts) in plant cells and algae.
  • Cells may be cultured or expanded after transfection with the engineered retron.
  • Methods of introducing nucleic acids into a host cell are well known in the art.
  • Commonly used methods include chemically induced transformation, typically using divalent cations (e.g., CaCl 2 ), dextran-mediated transfection, polybrene mediated transfection, lipofectamine and LT-1 mediated transfection, electroporation, protoplast fusion, encapsulation of nucleic acids in liposomes, and direct microinjection of the nucleic acids comprising engineered retrons into nuclei.
  • divalent cations e.g., CaCl 2
  • the plant cells that have been transformed may be grown into a transgenic organism, such as a plant, in accordance with conventional methods.
  • Plant material that may be transformed with the engineered retrons described herein includes plant cells, plant protoplasts, plant cell tissue cultures from which plants can be regenerated, plant calli, plant clumps, and plant cells that are intact in plants or parts of plants such as embryos, pollen, ovules, seeds, leaves, flowers, branches, fruit, kernels, ears, cobs, husks, stalks, roots, root tips, anthers, and the like. Progeny, variants, and mutants of the regenerated plants are also included within the scope of the disclosure, provided that these parts comprise the genetic modification introduced by the engineered retron.
  • the engineered retrons described herein may be used to produce transgenic plants with desired phenotypes, including but not limited to, increased disease resistance (e.g., increased viral, bacterial of fungal resistance), increased insect resistance, increased drought resistance, increased yield, and altered fruit ripening characteristics, sugar and oil composition, and color.
  • the retron msr gene, msd gene, and/or ret gene are expressed in vitro from a vector, such as in an in vitro transcription system.
  • the resulting ncRNA or msDNA can be isolated before being packaged and/or formulated for direct delivery into a host cell.
  • the isolated ncRNA or msDNA can be packaged/formulated in a delivery vehicle such as lipid nanoparticles as described in other sections.
  • a delivery vehicle such as lipid nanoparticles as described in other sections.
  • the retron msr gene, msd gene, and/or ret gene are expressed in vivo from a vector within a cell.
  • the retron msr gene, msd gene, and/or ret gene can be introduced into a cell with a single vector or in multiple separate vectors to produce msDNA in a host subject.
  • the retron msr gene, msd gene, and/or ret gene, and any other components of the retron-based genome editing systems described herein may be expressed in vivo from RNA delivered to the cell.
  • the retron msr gene, msd gene, and/or ret gene can be introduced into a cell with a single vector or in multiple separate vectors to produce msDNA in a host subject.
  • Vectors and/or nucleic acid molecules encoding the recombinant retron-based genome editing system or components thereof can include control elements operably linked to the retron sequences, which allow for the production of msDNA either in vitro, or in vivo in the subject species.
  • the retron msr gene, msd gene, and/or ret gene can be operably linked to a promoter to allow expression of the retron reverse transcriptase and/or the msDNA product.
  • heterologous sequences encoding desired products of interest may be inserted in the msr gene and/or msd gene.
  • desired products of interest e.g., polynucleotide encoding polypeptide or regulatory RNA, donor polynucleotide for gene editing, or protospacer DNA for molecular recording
  • desired products of interest e.g., polynucleotide encoding polypeptide or regulatory RNA, donor polynucleotide for gene editing, or protospacer DNA for molecular recording
  • desired products of interest e.g., polynucleotide encoding polypeptide or regulatory RNA, donor polynucleotide for gene editing, or protospacer DNA for molecular recording
  • Any eukaryotic, archaeal, or prokaryotic cell capable of being transfected with a vector or retron delivery system comprising the engineered retron sequences, may be used to produce the msDNA in vivo.
  • the transfected cell can be assayed either through phenotypic changes that occur due to the introduced sequences or by direct DNA sequencing.
  • the engineered retron is produced by a vector system comprising one or more vectors.
  • the msr gene, the msd gene, and/or the ret gene may be provided by the same vector (i.e., cis arrangement of all such retron elements), wherein the vector comprises a promoter operably linked to the msr gene and/or the msd gene.
  • the promoter is further operably linked to the ret gene.
  • the vector further comprises a second promoter operably linked to the ret gene.
  • the ret gene may be provided by a second vector that does not include the msr gene and/or the msd gene (i.e., trans arrangement of msr-msd and ret).
  • the msr gene, the msd gene, and the ret gene are each provided by different vectors (i.e., trans arrangement of all retron elements).
  • Numerous vectors are available for use in the vector or vector system, including but not limited to, linear polynucleotides, polynucleotides associated with ionic or amphiphilic compounds, plasmids, and viruses.
  • viral vectors include, but are not limited to, adenoviral vectors, adeno-associated virus (AAV) vectors, retroviral vectors, lentiviral vectors, and the like.
  • An expression construct can be replicated in a living cell, or it can be made synthetically.
  • the nucleic acid comprising an engineered retron sequence is under transcriptional control of a promoter.
  • the promoter is competent for initiating transcription of an operably linked coding sequence by a RNA polymerase I, II, or III.
  • Exemplary promoters for mammalian cell expression include the SV40 early promoter, a CMV promoter such as the CMV immediate early promoter (see, U. S. Patent Nos. 5,168,062 and 5,385,839, incorporated herein by reference in their entireties), the mouse mammary tumor virus LTR promoter, the adenovirus major late promoter (Ad MLP), and the herpes simplex virus promoter, among others.
  • Other nonviral promoters such as a promoter derived from the murine metallothionein gene, will also find use for mammalian expression.
  • Exemplary promoters for plant cell expression include the CaMV 35S promoter (Odell et al., 1985, Nature 313:810-812); the rice actin promoter (McElroy et al., 1990, Plant Cell 2:163-171); the ubiquitin promoter (Christensen et al., 1989, Plant Mol. Biol.12:619-632; and Christensen et al., 1992, Plant Mol. Biol.18:675-689); the pEMU promoter (Last et al., 1991, Theor. Appl. Genet.81:581-588); and the MAS promoter (Velten et al., 1984, EMBO J.
  • the retron-based vectors may also comprise tissue-specific promoters to start expression only after it is delivered into a specific tissue.
  • tissue-specific promoters include B29 promoter, CD14 promoter, CD43 promoter, CD45 promoter, CD68 promoter, desmin promoter, elastase- 1 promoter, endoglin promoter, fibronectin promoter, Flt-1 promoter, GFAP promoter, GPIIb promoter, ICAM- 2 promoter, INF-b promoter, Mb promoter, Nphsl promoter, OG-2 promoter, SP-B promoter, SYN1 promoter, and WASP promoter.
  • promoters can be obtained from or incorporated into commercially available plasmids, using techniques well known in the art. See, e.g., Sambrook et al., supra.
  • one or more enhancer elements is/are used in association with the promoter to increase expression levels of the constructs. Examples include the SV40 early gene enhancer, as described in Dijkema et al., EMBOJ (1985) 4:761, the enhancer/promoter derived from the long terminal repeat (LTR) of the Rous Sarcoma Virus, as described in Gorman et al., Proc. Natl. Acad. Sci.
  • LTR long terminal repeat
  • an expression vector for expressing an engineered retron including the msr gene, msd gene, and/or ret gene comprises a promoter operably linked to a polynucleotide encoding the msr gene, msd gene, and/or ret gene.
  • the vector or vector system also comprises a transcription terminator/polyadenylation signal.
  • sequences include, but are not limited to, those derived from SV40, as described in Sambrook et al., supra, as well as a bovine growth hormone terminator sequence (see, e.g., U.S. Patent No.5,122,458).
  • 5 ⁇ - UTR sequences can be placed adjacent to the coding sequence to further enhance the expression.
  • Such sequences may include UTRs comprising an internal ribosome entry site (IRES).
  • IRES internal ribosome entry site
  • IRES internal ribosome entry site
  • IRES permits the translation of one or more open reading frames from a vector.
  • the IRES element attracts a eukaryotic ribosomal translation initiation complex and promotes translation initiation. See, e.g., Kaufman et al., Nuc. Acids Res.
  • IRES sequences are known and include sequences derived from a wide variety of viruses, such as from leader sequences of picomaviruses such as the encephalomyocarditis virus (EMCV) UTR (Jang et al.. Virol. (1989) 63:1651-1660).
  • EMCV encephalomyocarditis virus
  • the polio leader sequence the hepatitis A virus leader, the hepatitis C virus IRES, human rhinovirus type 2 IRES (Dobrikova et al., Proc. Natl. Acad. Sci. (2003) 100(251:15125-151301)).
  • an IRES element from the foot and mouth disease virus (Ramesh et al., Nucl. Acid Res. (1996) 24:2697-2700), a giardiavirus IRES (Garlapati et al., J Biol. Chem. (2004) 279(51):3389-33971) and the like.
  • IRES sequences will also find use herein, including, but not limited to IRES sequences from yeast, as well as the human angiotensin II type 1 receptor IRES (Martin et al., Mol. Cell Endocrinol. (2003) 212:51-61), fibroblast growth factor IRESs (FGF-1 IRES and FGF-2 IRES, Martineau et al. (2004) Mol. Cell. Biol.24( 17): 7622-7635), vascular endothelial growth factor IRES (Baranick et al. (2008) Proc. Natl. Acad Sci. U.S.A.105(12):4733-4738, Stein et al. (1998) Mol. Cell.
  • An IRES sequence may be included in a vector, for example, to express multiple bacteriophage recombination proteins for recombineering or an RNA-guided nuclease (e.g., Cas9) for HDR in combination with a retron reverse transcriptase from an expression cassette.
  • a polynucleotide encoding a viral self-cleaving 2A peptide, such as a T2A peptide can be used to allow production of multiple protein products (e.g., Cas9, bacteriophage recombination proteins, retron reverse transcriptase) from a single vector or a single transcription unit under one promoter.
  • One or more 2A linker peptides can be inserted between the coding sequences in the multicistronic construct.
  • the 2A peptide which is self- cleaving, allows co-expressed proteins from the multicistronic construct to be produced at equimolar levels.
  • 2A peptides from various viruses may be used, including, but not limited to 2A peptides derived from the foot-and-mouth disease virus, equine rhinitis A virus, Jhosea asigna virus and porcine teschovirus-1. See, e.g., Kim et al. (2011) PLoS One 6(4): el8556, Trichas et al. (2008) BMC Biol.6:40, Provost et al.
  • the expression construct comprises a plasmid suitable for transforming a bacterial host.
  • Numerous bacterial expression vectors are known to those of skill in the art, and the selection of an appropriate vector is a matter of choice.
  • Bacterial expression vectors include, but are not limited to, pACYC177, pASK75, pBAD, pBADM, pBAT, pCal, pET, pETM, pGAT, pGEX, pHAT, pKK223, pMal, pProEx, pQE, and pZA31
  • Bacterial plasmids may contain antibiotic selection markers (e.g., ampicillin, kanamycin, erythromycin, carbenicillin, streptomycin, or tetracycline resistance), a lacZ gene (b-galactosidase produces blue pigment from x-gal substrate), fluorescent markers (e.g., GFP.
  • the expression construct comprises a plasmid suitable for transforming a yeast cell.
  • Yeast expression plasmids typically contain a yeast-specific origin of replication (ORI) and nutritional selection markers (e.g., HIS3, URA3, LYS2, LEU2, TRP1, METIS, ura4+, leul+, ade6+), antibiotic selection markers (e.g., kanamycin resistance), fluorescent markers (e.g., mCherry), or other markers for selection of transformed yeast cells.
  • ORI yeast-specific origin of replication
  • nutritional selection markers e.g., HIS3, URA3, LYS2, LEU2, TRP1, METIS, ura4+, leul+, ade6+
  • antibiotic selection markers e.g., kanamycin resistance
  • fluorescent markers e.g., mCherry
  • the yeast plasmid may further contain components to allow shuttling between a bacterial host (e.g., E coif) and yeast cells.
  • yeast plasmids include yeast integrating plasmids (Yip), which lack an ORI and are integrated into host chromosomes by homologous recombination; yeast replicating plasmids (YRp), which contain an autonomously replicating sequence (ARS) and can replicate independently; yeast centromere plasmids (YCp), which are low copy vectors containing a part of an ARS and part of a centromere sequence (CEN); and yeast episomal plasmids (YEp), which are high copy number plasmids comprising a fragment from a 2 micron circle (a natural yeast plasmid) that allows for 50 or more copies to be stably propagated per cell.
  • Yip yeast integrating plasmids
  • ARS autonomously replicating sequence
  • YCp yeast centromere plasmids
  • the expression construct does not comprise a plasmid suitable for transforming a yeast cell.
  • the expression construct comprises a virus or engineered construct derived from a viral genome.
  • viral based systems have been developed for gene transfer into mammalian cells. These include adenoviruses, retroviruses (g-retroviruses and lentiviruses), poxviruses, adeno-associated viruses, baculoviruses, and herpes simplex viruses (see e.g., Wamock et al. (2011) Methods Mol. Biol.737:1-25; Walther et al.
  • retroviruses provide a convenient platform for gene delivery systems. Selected sequences can be inserted into a vector and packaged in retroviral particles using techniques known in the art. The recombinant virus can then be isolated and delivered to cells of the subject either in vivo or ex vivo. A number of retroviral systems have been described (U.S. Pat.
  • Lentiviruses are a class of retroviruses that are particularly useful for delivering polynucleotides to mammalian cells because they are able to infect both dividing and nondividing cells (see e.g., Lois et al. (2002) Science 295:868-872; Durand et al. (2011) Viruses 3(2): 132-159; herein incorporated by reference).
  • a number of adenoviral vectors have also been described. Unlike retroviruses which integrate into the host genome, adenoviruses persist extrachromosomally thus minimizing the risks associated with insertional mutagenesis.
  • AAV vectors can be readily constructed using techniques well known in the art. See, e.g., U.S. Pat. Nos.5,173,414 and 5,139,941; International Publication Nos. WO 92/01070 (published 23 January 1992) and WO 93/03769 (published 4 March 1993); Lebkowski et al., Molec. Cell. Biol. (1988) 8:3988-3996; Vincent et al., Vaccines 90 (1990) (Cold Spring Harbor LaboratoryPress); Carter, B. J. Current Opinion in Biotechnology (1992) 3:533-539; Muzyczka, N.
  • Another vector system useful for delivering nucleic acids encoding the engineered retrons is the enterically administered recombinant poxvirus vaccines described by Small, Jr., P. A., et al. (U.S. Pat. No.5,676,950, issued Oct.14, 1997, herein incorporated by reference).
  • viral vectors include those derived from the pox family of viruses, including vaccinia virus and avian poxvirus.
  • vaccinia virus recombinants expressing a nucleic acid molecule of interest can be constructed as follows. The DNA encoding the particular nucleic acid sequence is first inserted into an appropriate vector so that it is adjacent to a vaccinia promoter and flanking vaccinia DNA sequences, such as the sequence encoding thymidine kinase (TK). This vector is then used to transfect cells which are simultaneously infected with vaccinia.
  • TK thymidine kinase
  • avipoxviruses such as the fowlpox and canarypox viruses, can also be used to deliver the nucleic acid molecules of interest.
  • the use of an avipox vector is particularly desirable in human and other mammalian species since members of the avipox genus can only productively replicate in susceptible avian species and therefore are not infective in mammalian cells.
  • Molecular conjugate vectors such as the adenovirus chimeric vectors described in Michael et al., J. Biol. Chem. (1993) 268:6866-6869 and Wagner et al., Proc. Natl. Acad. Sci. USA (1992) 89:6099-6103, can also be used for gene delivery.
  • Sindbis-virus derived vectors useful for the practice of the instant methods, see, Dubensky et al. (1996) J. Virol.70:508-519; and International Publication Nos. WO 95/07995, WO 96/17072; as well as, Dubensky, Jr., T. W., et al., U.S. Pat.
  • chimeric alphavirus vectors comprised of sequences derived from Sindbis virus and Venezuelan equine encephalitis virus. See, e.g., Perri et al. (2003) J. Virol.77: 10394-10403 and International Publication Nos. WO 02/099035, WO 02/080982, WO 01/81609, and WO 00/61772; herein incorporated by reference in their entireties.
  • a vaccinia-based infection/transfection system can be conveniently used to provide for inducible, transient expression of the nucleic acids of interest (e.g., engineered retron) in a host cell.
  • cells are first infected in vitro with a vaccinia virus recombinant that encodes the bacteriophage T7 RNA polymerase.
  • This polymerase displays extraordinar specificity in that it only transcribes templates bearing T7 promoters.
  • cells are transfected with the nucleic acid of interest, driven by a T7 promoter.
  • the polymerase expressed in the cytoplasm from the vaccinia virus recombinant transcribes the transfected DNA into RNA.
  • RNA messenger RNA
  • Elroy-Stein and Moss Proc. Natl. Acad. Sci. USA (1990) 87:6743- 6747; Fuerst et al., Proc. Natl. Acad. Sci. USA (1986) 83:8122-8126.
  • an amplification system can be used that will lead to high level expression following introduction into host cells.
  • T7 RNA polymerase promoter preceding the coding region for T7 RNA polymerase can be engineered. Translation of RNA derived from this template will generate T7 RNA polymerase which in turn will transcribe more templates. Concomitantly, there will be a cDNA whose expression is under the control of the T7 promoter. Thus, some of the T7 RNA polymerase generated from translation of the amplification template RNA will lead to transcription of the desired gene. Because some T7 RNA polymerase is required to initiate the amplification, T7 RNA polymerase can be introduced into cells along with the template(s) to prime the transcription reaction.
  • the polymerase can be introduced as a protein or on a plasmid encoding the RNA polymerase.
  • T7 systems and their use for transforming cells see, e.g., International Publication No. WO 94/26911; Studier and Moffatt, J. Mol. Biol. (1986) 189:113-130; Deng and Wolff, Gene (1994) 143:245-249; Gao et al., Biochem. Biophys. Res. Commun. (1994) 200:1201-1206; Gao and Huang, Nuc. Acids Res. (1993) 21:2867-2872; Chen et al., Nuc. Acids Res. (1994) 22:2114-2120; and U.S.
  • Insect cell expression systems such as baculovirus systems, can also be used and are known to those of skill in the art and described in, e.g., Baculovirus and Insect Cell Expression Protocols (Methods in Molecular Biology, D.W. Murhammer ed., Humana Press, 2nd edition, 2007) and L. King The Baculovirus Expression System: A laboratory guide (Springer, 1992). Materials and methods for baculovirus/insect cell expression systems are commercially available in kit form from, inter alia, Thermo Fisher Scientific (Waltham, MA) and Clontech (Mountain View, CA).
  • Plant expression systems can also be used for transforming plant cells. Generally, such systems use virus-based vectors to transfect plant cells with heterologous genes. For a description of such systems see, e.g., Porta et al., Mol. Biotech. (1996) 5:209-221; andhackland et al., Arch. Virol. (1994) 139:1-22.
  • the expression construct or the ncRNA must be delivered into a cell. This delivery may be accomplished in vitro, as in laboratory procedures for transforming cells lines, or in vivo or ex vivo, as in the treatment of certain disease states.
  • One mechanism for delivery is via viral infection where the expression construct is encapsulated in an infectious viral particle.
  • Several non-viral methods for the transfer of expression constructs into cultured cells also are contemplated. These include the use of calcium phosphate precipitation, DEAE- dextran, electroporation, direct microinjection, DNA-loaded liposomes, lipofectamine-DNA complexes, cell sonication, gene bombardment using high velocity microprojectiles, and receptor-mediated transfection (see, e.g., Graham and Van Der Eb (1973) Virology 52:456-467; Chen and Okayama (1987) Mol. Cell Biol.7:2745-2752; Rippe et al. (1990) Mol. Cell Biol.
  • nucleic acid comprising the engineered retron sequence may be positioned and expressed at different sites.
  • the nucleic acid comprising the engineered retron sequence may be stably integrated into the genome of the cell.
  • the nucleic acid may be stably maintained in the cell as a separate, episomal segment of DNA. Such nucleic acid segments or episomes encode sequences sufficient to permit maintenance and replication independent of or in synchronization with the host cell cycle. How the expression construct is delivered to a cell and where in the cell the nucleic acid remains is dependent on the type of expression construct employed. [00678] In some embodiments, the expression construct may simply consist of naked recombinant DNA or plasmids comprising the engineered retron.
  • Transfer of the construct may be performed by any of the methods mentioned above which physically or chemically permeabilize the cell membrane. This is particularly applicable for transfer in vitro but it may be applied to in vivo use as well.
  • Dubensky et al. Proc. Natl. Acad. Sci. USA (1984) 81:7529-7533
  • Benvenisty & Neshif Proc. Natl. Acad. Sci. USA (1986) 83:9551-9555
  • direct intraperitoneal injection of calcium phosphate- precipitated plasmids results in expression of the transfected genes.
  • DNA encoding an engineered retron of interest may also be transferred in a similar manner in vivo and express retron products.
  • a naked DNA expression construct may be transferred into cells by particle bombardment. This method depends on the ability to accelerate DNA-coated microprojectiles to a high velocity allowing them to pierce cell membranes and enter cells without killing them (Klein et al. (1987) Nature 327:70-73).
  • Several devices for accelerating small particles have been developed. One such device relies on a high voltage discharge to generate an electrical current, which in turn provides the motive force (Yang et al. (1990) Proc. Natl. Acad. Sci. USA 87:9568-9572).
  • the microprojectiles may consist of biologically inert substances, such as tungsten or gold beads.
  • the expression construct may be delivered using liposomes.
  • Liposomes are vesicular structures characterized by a phospholipid bilayer membrane and an inner aqueous medium. Multilamellar liposomes have multiple lipid layers separated by aqueous medium. They form spontaneously when phospholipids are suspended in an excess of aqueous solution.
  • the liposome may be complexed with a hemagglutinating virus (HVJ). This has been shown to facilitate fusion with the cell membrane and promote cell entry of liposome-encapsulated DNA (Kaneda et al.
  • the liposome may be complexed or employed in conjunction with nuclear non- histone chromosomal proteins (HMG-I) (Kato et al. (1991) J. Biol. Chem.266(6):3361-3364).
  • HMG-I nuclear non- histone chromosomal proteins
  • the liposome may be complexed or employed in conjunction with both HVJ and HMG-I.
  • a bacterial promoter is employed in the DNA construct, it also will be desirable to include within the liposome an appropriate bacterial polymerase.
  • Other expression constructs which can be employed to deliver a nucleic acid into cells are receptor-mediated delivery vehicles.
  • Receptor-mediated gene targeting vehicles generally consist of two components: a cell receptor-specific ligand and a DNA-binding agent.
  • ligands have been used for receptor-mediated gene transfer. The most extensively characterized ligands are asialoorosomucoid (ASOR) and transferrin (see, e.g., Wu and Wu (1987), supra; Wagner et al. (1990) Proc. Natl. Acad. Sci.
  • the delivery vehicle may comprise a ligand and a liposome.
  • Nicolau et al. Methods Enzymol.
  • the promoters that may be used in the retron delivery systems described herein may be constitutive, inducible, or tissue-specific. In some embodiments, the promoters may be a constitutive promoters.
  • Non-limiting exemplary constitutive promoters include cytomegalovirus immediate early promoter (CMV), simian virus (SV40) promoter, adenovirus major late (MLP) promoter, Rous sarcoma virus (RSV) promoter, mouse mammary tumor virus (MMTV) promoter, phosphoglycerate kinase (PGK) promoter, elongation factor-alpha (EFla) promoter, ubiquitin promoters, actin promoters, tubulin promoters, immunoglobulin promoters, a functional fragment thereof, or a combination of any of the foregoing.
  • the promoter may be a CMV promoter.
  • the promoter may be a truncated CMV promoter. In other embodiments, the promoter may be an EFla promoter. In some embodiments, the promoter may be an inducible promoter. Non-limiting exemplary inducible promoters include those inducible by heat shock, light, chemicals, peptides, metals, steroids, antibiotics, or alcohol. In some embodiments, the inducible promoter may be one that has a low basal (non-induced) expression level, such as, e.g., the Tet-On® promoter (Clontech). In some embodiments, the promoter may be a tissue-specific promoter. In some embodiments, the tissue-specific promoter is exclusively or predominantly expressed in liver tissue.
  • Non-limiting exemplary tissue-specific promoters include B29 promoter, CD14 promoter, CD43 promoter, CD45 promoter, CD68 promoter, desmin promoter, elastase- 1 promoter, endoglin promoter, fibronectin promoter, Flt-1 promoter, GFAP promoter, GPIIb promoter, ICAM- 2 promoter, INF-b promoter, Mb promoter, Nphsl promoter, OG-2 promoter, SP-B promoter, SYN1 promoter, and WASP promoter.
  • the engineered retrons can be delivered by any known delivery system such as those described above.
  • Non-limiting examples of delivery vehicles include lipid particles (e.g.
  • LNPs Lipid nanoparticles
  • SNALPs Stable nucleic-acid-lipid particles
  • SLO Stable nucleic-acid-lipid particles
  • MEND multifunctional envelope-type nanodevice
  • lipid-coated mesoporous silica particles inorganic nanoparticles, and polymeric delivery technology (e.g., polymer-based particles).
  • All-RNA delivery system and RNA ratios [00687] In various embodiments, the retron editing systems disclosed herein may be delivered in an “all-RNA” format.
  • the term “all-RNA” format refers to the fact that each of the components of a retron editing system (e.g., the retron RT, the programmable nuclease, the sgRNA, and the ncRNA) are delivered and/or administered as RNA.
  • the RNA components may be delivered to cells and/or tissues by direct means, such as electroporation or transfection.
  • the RNA components may be delivered to cells and/or tissues by way of a delivery vehicle, such as an LNP or liposome.
  • the retron editing systems described herein may comprise a coding RNA (e.g., linear or circular mRNA) that encodes a retron reverse transcriptase (e.g., any RT from Table X or Table A), a coding RNA (e.g., linear or circular mRNA) that encodes a programmable nuclease, a retron ncRNA (e.g., a ncRNA from Table B), and a guide RNA.
  • RT and nuclease components may be encoded on the same coding RNA molecule.
  • the proteins may also be expressed from separate coding RNA molecules.
  • the RT and the nuclease components can be fused together as a singular fusion polypeptide having an RT domain and a nuclease domain optionally joined by a linker.
  • the ncRNA and the guide RNA may be fused together as a single RNA molecule.
  • the guide RNA may be located at the 5’ end of the ncRNA.
  • the guide RNA may be located at the 3’ end of the ncRNA.
  • the ncRNA may comprise a guide RNA at both the 3’ and the 5’ ends of the ncRNA.
  • the ncRNA and the guide RNA may be separate molecules, i.e., delivered separately.
  • the retron editing system may include both a ncRNA- guide RNA fusion and an additional guide RNA provided as a separate molecule.
  • the different RNA components of the all-RNA retron editing system can be combined and administered (e.g., directly or within a delivery vehicle) in different ratios. In some embodiments, the ratios of such RNA components or species can be expressed as molar ratios.
  • the molar ratio of RT coding RNA to nuclease coding RNA can be about 1:1, about 1:1.5, about 1:2, about 1:2.5, about 1:3, about 1:4, about 1:5, about 1:6, about 1:7, about 1:8, about 1:9, about 1:10, about 1:12, about 1:15, about 1:20.
  • Useful ranges include from 1:1 to 1:2, from 1:1.5 to 1:4, from 1:2 to 1:4, from 1:2 to 1:8, from 1:2 to 1:10, from 1:3 to 1:9, from 1:3 to 1:12, from 1:3 to 1:15, from 1:4 to 1:8, from 1:4 to 1:12, from 1:4 to 1:20, from 1:5 to 1:10, from 1:5 to 1:15, from 1:5 to 1:20, from 1:10 to 1:20, or from 1:10 to 1:40.
  • the molar ratio of nuclease coding RNA to RT coding RNA can be about 1:1, about 1:1.5, about 1:2, about 1:2.5, about 1:3, about 1:4, about 1:5, about 1:6, about 1:7, about 1:8, about 1:9, about 1:10, about 1:12, about 1:15, about 1:20.
  • Useful ranges include from 1:1 to 1:2, from 1:1.5 to 1:4, from 1:2 to 1:4, from 1:2 to 1:8, from 1:2 to 1:10, from 1:3 to 1:9, from 1:3 to 1:12, from 1:3 to 1:15, from 1:4 to 1:8, from 1:4 to 1:12, from 1:4 to 1:20, from 1:5 to 1:10, from 1:5 to 1:15, from 1:5 to 1:20, from 1:10 to 1:20, or from 1:10 to 1:40.
  • the molar ratio of ncRNA or ncRNA-guide RNA fusion to separate guide RNA can be about 1:1, about 1:1.5, about 1:2, about 1:2.5, about 1:3, about 1:4, about 1:5, about 1:6, about 1:7, about 1:8, about 1:9, about 1:10, about 1:12, about 1:15, about 1:20.
  • Useful ranges include from 1:1 to 1:2, from 1:1.5 to 1:4, from 1:2 to 1:4, from 1:2 to 1:8, from 1:2 to 1:10, from 1:3 to 1:9, from 1:3 to 1:12, from 1:3 to 1:15, from 1:4 to 1:8, from 1:4 to 1:12, from 1:4 to 1:20, from 1:5 to 1:10, from 1:5 to 1:15, from 1:5 to 1:20, from 1:10 to 1:20, or from 1:10 to 1:40.
  • the molar ratio of separate guide RNA to ncRNA or ncRNA-guide RNA fusion can be about 1:1, about 1:1.5, about 1:2, about 1:2.5, about 1:3, about 1:4, about 1:5, about 1:6, about 1:7, about 1:8, about 1:9, about 1:10, about 1:12, about 1:15, about 1:20.
  • Useful ranges include from 1:1 to 1:2, from 1:1.5 to 1:4, from 1:2 to 1:4, from 1:2 to 1:8, from 1:2 to 1:10, from 1:3 to 1:9, from 1:3 to 1:12, from 1:3 to 1:15, from 1:4 to 1:8, from 1:4 to 1:12, from 1:4 to 1:20, from 1:5 to 1:10, from 1:5 to 1:15, from 1:5 to 1:20, from 1:10 to 1:20, or from 1:10 to 1:40.
  • the molar ratio of ncRNA to separate guide RNA can be about 1:1, about 1:1.5, about 1:2, about 1:2.5, about 1:3, about 1:4, about 1:5, about 1:6, about 1:7, about 1:8, about 1:9, about 1:10, about 1:12, about 1:15, about 1:20.
  • Useful ranges include from 1:1 to 1:2, from 1:1.5 to 1:4, from 1:2 to 1:4, from 1:2 to 1:8, from 1:2 to 1:10, from 1:3 to 1:9, from 1:3 to 1:12, from 1:3 to 1:15, from 1:4 to 1:8, from 1:4 to 1:12, from 1:4 to 1:20, from 1:5 to 1:10, from 1:5 to 1:15, from 1:5 to 1:20, from 1:10 to 1:20, or from 1:10 to 1:40.
  • the molar ratio of separate guide RNA to ncRNA can be about 1:1, about 1:1.5, about 1:2, about 1:2.5, about 1:3, about 1:4, about 1:5, about 1:6, about 1:7, about 1:8, about 1:9, about 1:10, about 1:12, about 1:15, about 1:20.
  • Useful ranges include from 1:1 to 1:2, from 1:1.5 to 1:4, from 1:2 to 1:4, from 1:2 to 1:8, from 1:2 to 1:10, from 1:3 to 1:9, from 1:3 to 1:12, from 1:3 to 1:15, from 1:4 to 1:8, from 1:4 to 1:12, from 1:4 to 1:20, from 1:5 to 1:10, from 1:5 to 1:15, from 1:5 to 1:20, from 1:10 to 1:20, or from 1:10 to 1:40.
  • the molar ratio of a coding RNA (e.g., encoding RT and/or nuclease) to ncRNA or ncRNA-guide RNA fusion, as the case may be, can be about 1:1, about 1:1.5, about 1:2, about 1:2.5, about 1:3, about 1:4, about 1:5, about 1:6, about 1:7, about 1:8, about 1:9, about 1:10, about 1:12, about 1:15, about 1:20.
  • Useful ranges include from 1:1 to 1:2, from 1:1.5 to 1:4, from 1:2 to 1:4, from 1:2 to 1:8, from 1:2 to 1:10, from 1:3 to 1:9, from 1:3 to 1:12, from 1:3 to 1:15, from 1:4 to 1:8, from 1:4 to 1:12, from 1:4 to 1:20, from 1:5 to 1:10, from 1:5 to 1:15, from 1:5 to 1:20, from 1:10 to 1:20, or from 1:10 to 1:40.
  • the molar ratio of a coding RNA encoding a retron RT to ncRNA or ncRNA-guide RNA fusion can be about 1:1, about 1:1.5, about 1:2, about 1:2.5, about 1:3, about 1:4, about 1:5, about 1:6, about 1:7, about 1:8, about 1:9, about 1:10, about 1:12, about 1:15, about 1:20.
  • Useful ranges include from 1:1 to 1:2, from 1:1.5 to 1:4, from 1:2 to 1:4, from 1:2 to 1:8, from 1:2 to 1:10, from 1:3 to 1:9, from 1:3 to 1:12, from 1:3 to 1:15, from 1:4 to 1:8, from 1:4 to 1:12, from 1:4 to 1:20, from 1:5 to 1:10, from 1:5 to 1:15, from 1:5 to 1:20, from 1:10 to 1:20, or from 1:10 to 1:40.
  • the molar ratio of a coding RNA encoding a programmable nuclease to ncRNA or ncRNA-guide RNA fusion can be about 1:1, about 1:1.5, about 1:2, about 1:2.5, about 1:3, about 1:4, about 1:5, about 1:6, about 1:7, about 1:8, about 1:9, about 1:10, about 1:12, about 1:15, about 1:20.
  • Useful ranges include from 1:1 to 1:2, from 1:1.5 to 1:4, from 1:2 to 1:4, from 1:2 to 1:8, from 1:2 to 1:10, from 1:3 to 1:9, from 1:3 to 1:12, from 1:3 to 1:15, from 1:4 to 1:8, from 1:4 to 1:12, from 1:4 to 1:20, from 1:5 to 1:10, from 1:5 to 1:15, from 1:5 to 1:20, from 1:10 to 1:20, or from 1:10 to 1:40.
  • the molar ratio of a coding RNA encoding a retron RT or a nuclease to a separate guide RNA can be about 1:1, about 1:1.5, about 1:2, about 1:2.5, about 1:3, about 1:4, about 1:5, about 1:6, about 1:7, about 1:8, about 1:9, about 1:10, about 1:12, about 1:15, about 1:20.
  • Useful ranges include from 1:1 to 1:2, from 1:1.5 to 1:4, from 1:2 to 1:4, from 1:2 to 1:8, from 1:2 to 1:10, from 1:3 to 1:9, from 1:3 to 1:12, from 1:3 to 1:15, from 1:4 to 1:8, from 1:4 to 1:12, from 1:4 to 1:20, from 1:5 to 1:10, from 1:5 to 1:15, from 1:5 to 1:20, from 1:10 to 1:20, or from 1:10 to 1:40.
  • the molar ratio of a separate guide RNA to a coding RNA encoding a retron RT or a nuclease can be about 1:1, about 1:1.5, about 1:2, about 1:2.5, about 1:3, about 1:4, about 1:5, about 1:6, about 1:7, about 1:8, about 1:9, about 1:10, about 1:12, about 1:15, about 1:20.
  • Useful ranges include from 1:1 to 1:2, from 1:1.5 to 1:4, from 1:2 to 1:4, from 1:2 to 1:8, from 1:2 to 1:10, from 1:3 to 1:9, from 1:3 to 1:12, from 1:3 to 1:15, from 1:4 to 1:8, from 1:4 to 1:12, from 1:4 to 1:20, from 1:5 to 1:10, from 1:5 to 1:15, from 1:5 to 1:20, from 1:10 to 1:20, or from 1:10 to 1:40. [00705] In certain embodiments, the amount of ncRNA-sgRNA relative to RT mRNA is augmented.
  • the RT mRNA: ncRNA-sgRNA ratio is about 1:1.5, about 1:2, about 1:2.5, about 1:3, about 1:4, about 1:5, about 1:6, about 1:7, about 1:8, about 1:9, about 1:10, about 1:12, about 1:15, about 1:20.
  • Useful ranges include from 1:1 to 1:2, from 1:1.5 to 1:4, from 1:2 to 1:4, from 1:2 to 1:8, from 1:2 to 1:10, from 1:3 to 1:9, from 1:3 to 1:12, from 1:3 to 1:15, from 1:4 to 1:8, from 1:4 to 1:12, from 1:4 to 1:20, from 1:5 to 1:10, from 1:5 to 1:15, from 1:5 to 1:20, from 1:10 to 1:20, or from 1:10 to 1:40.
  • an RT-Cas9 (or Cas9-RT) fusion is encoded by an mRNA.
  • the RT-Cas9 mRNA: ncRNA-sgRNA ratio is about 1:1.5, about 1:2, about 1:2.5, about 1:3, about 1:4, about 1:5, about 1:6, about 1:7, about 1:8, about 1:9, about 1:10, about 1:12, about 1:15, about 1:20.
  • Useful ranges include from 1:1 to 1:2, from 1:1.5 to 1:4, from 1:2 to 1:4, from 1:2 to 1:8, from 1:2 to 1:10, from 1:3 to 1:9, from 1:3 to 1:12, from 1:3 to 1:15, from 1:4 to 1:8, from 1:4 to 1:12, from 1:4 to 1:20, from 1:5 to 1:10, from 1:5 to 1:15, from 1:5 to 1:20, from 1:10 to 1:20, or from 1:10 to 1:40.
  • multiple genetic loci are targeted hence the ncRNA-sgRNA includes a mixture of ncRNA-sgRNA species and the same ratios and ranges are applicable.
  • the ncRNA and the guide RNA can be delivered as a single molecule, i.e., with the guide RNA fused to the 5’ and/or 3’ end of the ncRNA.
  • the ncRNA may have guide RNAs located at both ends in some embodiments.
  • the guide RNA and ncRNA may be provided and/or delivered as separate components. As shown in Example 4, separation of the guide RNA from the ncRNA can result in increased editing efficiency.
  • a ncRNA-gRNA fusion may be co-delivered with a separate guide RNA.
  • the ncRNAs disclosed herein may be modified by introducing additional RNA motifs into the ncRNAs, e.g., at the 5 ⁇ and 3 ⁇ termini of the ncRNAs, or even at positions therein between (e.g., in the msr or msd regions) to improve transcriptional production and/or stability and/or function (e.g., RT-DNA production).
  • Such structures may include, but are not limited to RNA hairpins, RNA step-loops, RNA quadruplexes, cap structures, and poly(A) tails, or ribozyme functions and the like.
  • ncRNAs could also be modified to include one or more nuclear localization sequences.
  • Additional RNA motifs could also improve RT processivity of the ncRNA or enhance ncRNA activity by enhancing RT binding. Addition of dimerization motifs - such as kissing loops or a GNRA tetraloop/tetraloop receptor pair - at the 5 ⁇ and 3 ⁇ termini of the ncRNA could also result in effective circularization of the ncRNA, improving stability. Additionally, it is envisioned that addition of these motifs could enable the physical separation of ncRNA components, e.g., separation of the msr and msd regions.
  • ncRNAs could be further improved via directed evolution, in an analogous fashion to how protein function can be improved. Directed evolution could enhance ncRNA recognition by RT and/or reduce off-site targeting and/or indels and/or improve precise editing efficiency.
  • the present disclosure contemplates any such ways to further improve the stability and/or functionality of the ncRNAs disclosed here.
  • the RNAs (including the guide RNAs and the ncRNAs) used in the compositions of the disclosure have undergone a chemical or biological modification to render them more stable.
  • Exemplary modifications to an RNA include the depletion of a base (e.g., by deletion or by the substitution of one nucleotide for another) or modification of a base, for example, the chemical modification of a base.
  • RNA modifications includes modifications which introduce chemistries which differ from those seen in naturally occurring RNA, for example, covalent modifications such as the introduction of modified nucleotides, (e.g., nucleotide analogs, or the inclusion of pendant groups which are not naturally found in such mRNA molecules).
  • RNAs used in the compositions of the disclosure include, but are not limited to, 4'- thio-modified bases: 4'-thio-adenosine, 4'-thio-guanosine, 4'-thio-cytidine, 4'-thio-uridine, 4'- thio-5-methyl- cytidine, 4'-thio-pseudouridine, and 4'-thio-2-thiouridine, pyridin-4-one ribonucleoside, 5-aza- uridine, 2-thio-5-aza-uridine, 2-thiouridine, 4-thio-pseudouridine, 2- thio-pseudouridine, 5- hydroxyuridine, 3-methyluridine, 5-carboxymethyl-uridine, 1- carboxymethyl-pseudouridine, 5- propynyl-uridine, 1-propynyl-pseudour
  • modification also includes, for example, the incorporation of non-nucleotide linkages or modified nucleotides into the mRNA sequences of the present invention (e.g., modifications to one or both of the 3' and 5' ends of an mRNA molecule encoding a functional protein or enzyme).
  • modifications include the addition of bases to an mRNA sequence (e.g., the inclusion of a poly A tail or a longer poly A tail), the alteration of the 3' UTR or the 5' UTR, complexing the mRNA with an agent (e.g., a protein or a complementary nucleic acid molecule), and inclusion of elements which change the structure of an RNA molecule (e.g., which form secondary structures).
  • RNAs include a 5' cap structure.
  • a 5' cap is typically added as follows: first, an RNA terminal phosphatase removes one of the terminal phosphate groups from the 5' nucleotide, leaving two terminal phosphates; guanosine triphosphate (GTP) is then added to the terminal phosphates via a guanylyl transferase, producing a 5'5'5 triphosphate linkage; and the 7-nitrogen of guanine is then methylated by a methyltransferase.
  • GTP guanosine triphosphate
  • cap structures include, but are not limited to, m7G(5')ppp (5'(A,G(5')ppp(5')A and G(5')ppp(5')G.
  • Naturally occurring cap structures comprise a 7- methyl guanosine that is linked via a triphosphate bridge to the 5'-end of the first transcribed nucleotide, resulting in a dinucleotide cap of m7G(5')ppp(5')N, where N is any nucleoside.
  • the cap is added enzymatically. The cap is added in the nucleus and is catalyzed by the enzyme guanylyl transferase.
  • the addition of the cap to the 5' terminal end of RNA occurs immediately after initiation of transcription.
  • the terminal nucleoside is typically a guanosine, and is in the reverse orientation to all the other nucleotides, i.e., G(5')ppp(5')GpNpNp.
  • Additional cap analogs include, but are not limited to, a chemical structures selected from the group consisting of m7GpppG, m7GpppA, m7GpppC; unmethylated cap analogs (e.g., GpppG); dimethylated cap analog (e.g., m2,7GpppG), trimethylated cap analog (e.g., m2,2,7GpppG), dimethylated symmetrical cap analogs (e.g., m7Gpppm7G), or anti reverse cap analogs (e.g., ARCA; m7,2'OmeGpppG, m72'dGpppG, m7,3'OmeGpppG, m7,3'dGpppG and their tetraphosphate derivatives) (see, e.g., Jemielity, J.
  • RNA e.g., ncRNA
  • a poly A or poly U tail is thought to stabilize natural messengers and synthetic sense RNA. Therefore, in certain embodiments a long poly A or poly U tail can be added to an RNA molecule thus rendering the RNA more stable. Poly A or poly U tails can be added using a variety of art-recognized techniques.
  • long poly A tails can be added to synthetic or in vitro transcribed RNA using poly A polymerase (Yokoe, et al. Nature Biotechnology.1996; 14: 1252-1256).
  • a transcription vector can also encode long poly A tails.
  • poly A tails can be added by transcription directly from PCR products.
  • Poly A may also be ligated to the 3' end of a sense RNA with RNA ligase (see, e.g., Molecular Cloning A Laboratory Manual, 2nd Ed., ed. by Sambrook, Fritsch and Maniatis (Cold Spring Harbor Laboratory Press: 1991 edition)).
  • the length of a poly A or poly U tail can be at least about 10, 50, 100, 200, 300, 400 at least 500 nucleotides.
  • a poly-A tail on the 3' terminus of mRNA typically includes about 10 to 300 adenosine nucleotides (e.g., about 10 to 200 adenosine nucleotides, about 10 to 150 adenosine nucleotides, about 10 to 100 adenosine nucleotides, about 20 to 70 adenosine nucleotides, or about 20 to 60 adenosine nucleotides).
  • mRNAs include a 3' poly(C) tail structure.
  • a suitable poly-C tail on the 3' terminus of mRNA typically include about 10 to 200 cytosine nucleotides (e.g., about 10 to 150 cytosine nucleotides, about 10 to 100 cytosine nucleotides, about 20 to 70 cytosine nucleotides, about 20 to 60 cytosine nucleotides, or about 10 to 40 cytosine nucleotides).
  • the poly-C tail may be added to the poly-A or poly U tail or may substitute the poly-A or poly U tail.
  • RNAs according to the present disclosure e.g., ncRNAs
  • RNAs according to the present invention may be synthesized via in vitro transcription (IVT).
  • IVT in vitro transcription
  • a linear or circular DNA template containing a promoter, a pool of ribonucleotide triphosphates, a buffer system that may include DTT and magnesium ions, and an appropriate RNA polymerase (e.g., T3, T7 or SP6 RNA polymerase), DNAse I, pyrophosphatase, and/or RNAse inhibitor.
  • an appropriate RNA polymerase e.g., T3, T7 or SP6 RNA polymerase
  • the ncRNAs can comprise an MS2 modification, as specific RNA hairpin structure recognized in nature by a certain MS2-binding protein. This domain can help to stabilize the ncRNA and improve the editing efficiency.
  • MS2 modification as specific RNA hairpin structure recognized in nature by a certain MS2-binding protein. This domain can help to stabilize the ncRNA and improve the editing efficiency.
  • the disclosure contemplates other similar modifications.
  • the lipid delivery sytem includes lipid nanoparticles (LNP).
  • LNP lipid nanoparticles
  • the LNP are small solid or semi-solid particles possessing an exterior lipid layer with a hydrophilic exterior surface that is exposed to the non-LNP environment, an interior space which may aqueous (vesicle like) or non-aqueous (micelle like), and at least one hydrophobic inter-membrane space.
  • LNP membranes may be lamellar or non- lamellar and may be comprised of 1, 2, 3, 4, 5 or more layers.
  • LNPs may comprise a nucleic acid (e.g.
  • an LNP of the present disclosure comprises an ionizable lipid, a structural lipid, a PEGylated lipid (aka PEG lipid), and a phospholipid.
  • an LNP comprises an ionizable lipid, a structural lipid, a PEGylated lipid (aka PEG lipid), and a zwitterionic amino acid lipid.
  • an LNP further comprises a 5 th lipid, besides any of the aforementioned lipid components.
  • the LNP encapsulates one or more elements of the active agent of the present disclosure.
  • an LNP further comprises a targeting moiety covalently or non-covalently bound to the outer surface of the LNP.
  • the targeting moiety is a targeting moiety that binds to, or otherwise facilitates uptake by, cells of a particular organ system.
  • an LNP has a diameter of at least about 20nm, 30 nm, 40nm, 50nm, 60nm, 70nm, 80nm, or 90nm.
  • an LNP has a diameter of less than about 100nm, 110nm, 120nm, 130nm, 140nm, 150nm, or 160nm. In some embodiments, an LNP has a diameter of less than about 100nm.
  • an LNP has a diameter of less than about 90nm. In some embodiments, an LNP has a diameter of less than about 80nm. In some embodiments, an LNP has a diameter of about 60-100nm. In some embodiments, an LNP has a diameter of about 75-80nm.
  • the lipid nanoparticle compositions of the present disclosure are described according to the respective molar ratios of the component lipids in the formulation. As a non-limiting example, the mol-% of the ionizable lipid may be from about 10 mol-% to about 80 mol-%.
  • the mol-% of the ionizable lipid may be from about 20 mol-% to about 70 mol-%. As a non-limiting example, the mol-% of the ionizable lipid may be from about 30 mol-% to about 60 mol-%. As a non-limiting example, the mol-% of the ionizable lipid may be from about 35 mol-% to about 55 mol-%. As a non-limiting example, the mol-% of the ionizable lipid may be from about 40 mol-% to about 50 mol-%.
  • the mol-% of the phospholipid may be from about 1 mol- % to about 50 mol-%. In some embodiments, the mol-% of the phospholipid may be from about 2 mol-% to about 45 mol-%. In some embodiments, the mol-% of the phospholipid may be from about 3 mol-% to about 40 mol-%. In some embodiments, the mol-% of the phospholipid may be from about 4 mol-% to about 35 mol-%. In some embodiments, the mol-% of the phospholipid may be from about 5 mol-% to about 30 mol-%.
  • the mol-% of the phospholipid may be from about 10 mol-% to about 20 mol-%. In some embodiments, the mol- % of the phospholipid may be from about 5 mol-% to about 20 mol-%. [00726] In some embodiments, the mol-% of the structural lipid may be from about 10 mol-% to about 80 mol-%. In some embodiments, the mol-% of the structural lipid may be from about 20 mol-% to about 70 mol-%. In some embodiments, the mol-% of the structural lipid may be from about 30 mol-% to about 60 mol-%.
  • the mol-% of the structural lipid may be from about 35 mol-% to about 55 mol-%. In some embodiments, the mol-% of the structural lipid may be from about 40 mol-% to about 50 mol-%. [00727] In some embodiments, the mol-% of the PEG lipid may be from about 0.1 mol-% to about 10 mol-%. In some embodiments, the mol-% of the PEG lipid may be from about 0.2 mol-% to about 5 mol-%. In some embodiments, the mol-% of the PEG lipid may be from about 0.5 mol-% to about 3 mol-%.
  • the mol-% of the PEG lipid may be from about 1 mol-% to about 2 mol-%. In some embodiments, the mol-% of the PEG lipid may be about 1.5 mol-%.
  • an LNP disclosed herein comprises an ionizable lipid. In some embodiments, an LNP comprises two or more ionizable lipids. [00729] In some embodiments, an ionizable lipid has a dimethylamine or an ethanolamine head. In some embodiments, an ionizable lipid has an alkyl tail. In some embodiments, a tail has one or more ester linkages, which may enhance biodegradability.
  • a tail is branched, such as with 3 or more branches. In some embodiments, a branched tail may enhance endosomal escape.
  • an ionizable lipid has a pKa between 6 and 7, which may be measured, for example, by TNS assay. [00730] In some embodiments, an ionizable lipid has a structure of any of the formulas disclosed below, and all formulas disclosed in a reference publication and patent application publication cited below. In some embodiments, an ionizable lipid comprises a head group of any structure or formula disclosed below. In some embodiments, an ionizable lipid comprises a bridging moiety of any structure or formula disclosed below.
  • an ionizable lipid comprises any tail group, or combination of tail groups disclosed below.
  • the present disclosure contemplates all permutations and combinations of head group, bridging moiety and tail group, or tail groups, disclosed herein.
  • a head, tail, or structure of an ionizable lipid is described in US patent application US20170210697A1.
  • a compound has a structure according to formula 1: wherein: R 1 is selected from the group consisting of C 5-30 alkyl, C 5-20 alkenyl, - R*YR", YR", and - R"M'R'; R 2 and R 3 are independently selected from the group consisting of H, C1-14 alkyl, C2-14 alkenyl, —R*YR", —YR", and —R*OR", or R 2 and R 3 , together with the atom to which they are attached, form a heterocycle or carbocycle; R 4 is selected from the group consisting of a C 3-6 carbocycle, —(CH 2 )nQ, —(CH2)nCHQR, — CHQR, CO(R) 2 , and unsubstituted C 1-6 alkyl, where Q is selected from a carbocycle, heterocycle, —OR, —O(CH 2 )nN(R) 2 , —C(O)
  • each R is independently selected from the group consisting of C1-3 alkyl, C2-3 alkenyl, and H; each R’ is independently selected from the group consisting of C1-18 alkyl, C2-18 alkenyl, — R*YR", —YR", and H; each R” is independently selected from the group consisting of C3-14 alkyl, C3-14 alkenyl, and H; each R* is independently selected from the group consisting of C1-12 alkyl and C2-12 alkenyl: each Y is independently a C3-6 carbocycle; each X is independently selected from the group consisting of F, Cl, Br, and I; each Q is is -OH, -NHC(S)N(R) 2 , -NHC(O)N(R) 2 ,
  • R 4 is in Table 1.
  • R 4 in formula 1 is selected from head groups 1-47.
  • Table 1 - Ionizable lipid head groups [00735]
  • a 1 and a 2 are, at each occurrence, independently an integer from 3 to 12; b 1 and b 2 are, at each occurrence, independently 0 or 1; c 1 and c 2 are, at each occurrence, independently an integer from 5 to 10; d 1 and d 2 are, at each occurrence, independently an integer from 5 to 10; y is, at each occurrence, independently an integer from 0 to 2; and n is an integer from 1 to 6, wherein each alkyl, alkylene, hydroxylalkyl, aminoalkyl, alkylaminylalkyl, alkoxyalkyl, alkoxycarbonyl, alkylcarbonyloxy, alkylcarbonyloxyalkyl and alkylcarbonyl is optionally substituted with one or more substituent.
  • an ionizable lipid has a disulfide tail.
  • an ionizable lipid includes short peptides of 12-15 mer length as head groups.
  • the head of an ionizable lipid comprises the structure of Vitamin A, D, E, or K as described in the published Patent Application WO2019232095A1, which is incorporated by herein by reference in its entirety.
  • a lipid is described in international patent applications WO2021077067, or WO2019152557, each of which is incorporated herein by reference in its entirety.
  • an LNP described herein comprises a lipid, e.g., an ionizable lipid, disclosed in US 2019/0240354, which is incorporated herein by reference in its entirety.
  • the lipids disclosed in US 2019/0240354 are of Formula I: or salts thereof, wherein: R 1 and R 2 are either the same or different and are independently hydrogen (H) or an optionally substituted C 1 -C 6 alkyl, C 2 -C 6 alkenyl, or C 2 -C 6 alkynyl, or R 1 and R 2 may join to form an optionally substituted heterocyclic ring of 4 to 6 carbon atoms and 1 or 2 heteroatoms selected from the group consisting of nitrogen (N), oxygen (O), and mixtures thereof; R 3 is either absent or is hydrogen (H) or a C 1 -C 6 alkyl to provide a quaternary amine; R 4 and R 5 are either the same or different and are independently an optionally substituted C 10 -C 24 alkyl, C 10 -C 24 alkenyl, C 10 -C 24 alkynyl, or C 10 -C 24 acyl, wherein at least one of R 4 and
  • the lipids disclosed in US 2019/0240354 are of Formula II: wherein R 1 and R 2 are either the same or different and are independently an optionally substituted C 12 -C 24 alkyl, C 12 -C 24 alkenyl, C 12 -C 24 alkynyl, or C 12 -C 24 acyl; R 3 and R 4 are either the same or different and are independently an optionally substituted C 1 -C 6 alkyl, C 2 -C 6 alkenyl, or C 2 - C 6 alkynyl, or R 3 and R 4 may join to form an optionally substituted heterocyclic ring of 4 to 6 carbon atoms and 1 or 2 heteroatoms chosen from nitrogen and oxygen; R 5 is either absent or is hydrogen (H) or a C 1 -C 6 alkyl to provide a quaternary amine; m, n, and p are either the same or different and are independently either 0, 1, or 2, with the proviso that m, n
  • the cationic lipid of Formula II is 2,2-dilinoleyl-4-(2- dimethylaminoethyl)-[1,3]-dioxolane, 2,2-dilinoleyl-4-(3-dimethylaminopropyl)-[1,3]-dioxolane, 2,2-dilinoleyl-4-(4-dimethylaminobutyl)-[1,3]-dioxolane, 2,2-dilinoleyl-5-dimethylaminomethyl- [1,3]-dioxane, 2,2-dilinoleyl-4-N-methylpepiazino-[1,3]-dioxolane, 2,2-dilinoleyl-4- dimethylaminomethyl-[1,3]-dioxolane, 2,2-dioleoyl-4-dimethylaminomethyl-[1,3]-dioxolane, 2,2-2-dilinoleyl-4- dimethylamin
  • the cationic lipid of Formula II is 2,2-dilinoleyl-4-(2- dimethylaminoethyl)-[1,3]-dioxolane.
  • the lipids disclosed in US 2019/0240354 are of Formula III: or salts thereof, wherein: R 1 and R 2 are either the same or different and are independently an optionally substituted C 1 -C 6 alkyl, C 2 -C 6 alkenyl, or C 2 -C 6 alkynyl, or R 1 and R 2 may join to form an optionally substituted heterocyclic ring of 4 to 6 carbon atoms and 1 or 2 heteroatoms selected from the group consisting of nitrogen (N), oxygen (O), and mixtures thereof; R 3 is either absent or is hydrogen (H) or a C 1 -C 6 alkyl to provide a quaternary amine; R 4 and R 5 are either absent or present and when present are either the same or different and are independently an optionally substituted
  • the lipids disclosed in US 2019/0240354 are of Formula C: X-A-Y—Z 1 ; (Formula C) or salts thereof, wherein: X is —N(H)R or —NR 2 ; A is absent, C 1 to C 6 alkyl, C 2 to C 6 alkenyl, or C 2 to C 6 alkynyl, which C 1 to C 6 alkyl, C 2 to C 6 alkenyl, and C 2 to C 6 alkynyl is optionally substituted with one or more groups independently selected from oxo, halogen, heterocycle, —CN, —OR x , —NR x R y , —NR x C( ⁇ O)R y , — NR x SO 2 R y , —C( ⁇ O)R x , —C( ⁇ O)OR x , —C( ⁇ O)NR x R y , —SO n R
  • an LNP described herein comprises a lipid, e.g., an ionizable lipid, disclosed in US 2010/0130588, which is incorporated herein by reference in its entirety.
  • the lipids disclosed in US 2010/0130588 are of Formula I: wherein R 1 and R 2 are independently selected and are H or C 1 -C 3 alkyls, R 3 and R 4 are independently selected and are alkyl groups having from about 10 to about 20 carbon atoms, and at least one of R 3 and R 4 comprises at least two sites of unsaturation.
  • R 3 and R 4 are both the same, i.e., R 3 and R 4 are both linoleyl (C 18 ), etc. In some embodiments, R 3 and R 4 are different, i.e., R 3 is tetradectrienyl (C 14 ) and R 4 is linoleyl (C 18 ).
  • the lipid of Formula I is 1,2-dilinoleyloxy-N,N- dimethylaminopropane (DLinDMA) or 1,2-dilinolenyloxy-N,N-dimethylaminopropane (DLenDMA).
  • the lipids disclosed in US 2010/0130588 are of Formula II: wherein R 1 and R 2 are independently selected and are H or C 1 -C 3 alkyls, R 3 and R 4 are independently selected and are alkyl groups having from about 10 to about 20 carbon atoms, and at least one of R 3 and R 4 comprises at least two sites of unsaturation.
  • an LNP described herein comprises a lipid, e.g., an ionizable lipid, disclosed in US 2021/0087135, which is incorporated herein by reference in its entirety.
  • the lipids disclosed in US 2021/0087135 are of Formula (A): or its N-oxide, or a salt or isomer thereof, wherein R ′a is R ′branched or R ′cyclic ; wherein R ′branched is: R ′cyclic is: wherein: denotes a point of attachment; wherein R a ⁇ is H, and R a ⁇ , R a ⁇ , and R a ⁇ are each independently selected from the group consisting of H, C 2-12 alkyl, and C 2-12 alkenyl, wherein at least one of R a ⁇ , R a ⁇ , and R a ⁇ is selected from the group consisting of C 2-12 alkyl and C 2-12 alkenyl; R 2 and R 3 are each C 1-14 alkyl; R 4 is selected from the group consisting of —(CH 2 ) 2 OH, —(CH 2 ) 3 OH, —(CH 2 ) 4 OH,
  • an LNP described herein comprises a lipid, e.g., an ionizable lipid, disclosed in US 2021/0128488, which is incorporated herein by reference in its entirety
  • the lipids disclosed in US 2021/0128488 are of structure (I): or a pharmaceutically acceptable salt, prodrug or stereoisomer thereof, wherein: L 1 is —O(C ⁇ O)R′, —(C ⁇ O)OR 1 , —C( ⁇ O)R 1 , —OR 1 , —S(O) x R 1 , —S—SR 1 , —C(′O)SR′, — SC( ⁇ O)R′, —NR a C( ⁇ O)R 1 , —C( ⁇ O)NR b R c , —NR a C( ⁇ O)NR b R c , —OC( ⁇ O)NR b R c or —
  • an LNP described herein comprises a lipid, e.g., an ionizable lipid, disclosed in US 2020/0121809, which is incorporated herein by reference in its entirety.
  • the lipids disclosed in US 2020/0121809 have a structure of Formula II: or a pharmaceutically acceptable salt, tautomer, prodrug or stereoisomer thereof, wherein: one of L 1 or L 2 is —O(C ⁇ O)—, —(C ⁇ O)O—, —C( ⁇ O)—, —O—, —S(O) x —, —S—S—, — C( ⁇ O)S—, SC( ⁇ O)—, —NR a C( ⁇ O)—, —C( ⁇ O)NR a —, NR a C( ⁇ O)NR a —, —OC( ⁇ O)NR a — or —NR a C( ⁇ O)
  • the lipids disclosed in US 2020/0121809 have a structure of Formula III: or a pharmaceutically acceptable salt, prodrug or stereoisomer thereof, wherein: one of L 1 or L 2 is —O(C ⁇ O)—, —(C ⁇ O)O—, —C( ⁇ O)—, —O—, —S(O) x —, —S—S—, — C( ⁇ O)S—, SC( ⁇ O)—, —NR a C( ⁇ O)—, —C( ⁇ O)NR a —, NR a C( ⁇ O)NR a —, —OC( ⁇ O)NR a — or —NR a C( ⁇ O)O—, and the other of L 1 or L 2 is —O(C ⁇ O)—, —(C ⁇ O)O—, —C( ⁇ O)—, — O—, —S(O) x —, —S—S
  • the lipids disclosed in US 2020/0121809 have a structure of Formula (IV): or a pharmaceutically acceptable salt, prodrug or stereoisomer thereof, wherein: one of G 1 or G 2 is, at each occurrence, —O(C ⁇ O)—, —(C ⁇ O)O—, —C( ⁇ O)—, —O—, — S(O) y —, —S—S—, —C( ⁇ O)S—, SC( ⁇ O)—, —N(R a )C( ⁇ O)—, —C( ⁇ O)N(R a )—, — N(R a )C( ⁇ O)N(R a )—, —OC( ⁇ O)N(R a )— or —N(R a )C( ⁇ O)O—, and the other of G 1 or G 2 is, at each occurrence, —O(C ⁇ O)—, —(C)O—, —(C(
  • an LNP described herein comprises a lipid, e.g., an ionizable lipid, disclosed in US 2013/0108685, which is incorporated herein by reference in its entirety.
  • the lipids disclosed in US 2013/0108685 are represented by the following formula (I): wherein: R 1 and R 2 are, the same or different, each linear or branched alkyl, alkenyl or alkynyl having 12 to 24 carbon atoms, or R 1 and R 2 are combined together to form dialkylmethylene, dialkenylmethylene, dialkynylmethylene or alkylalkenylmethylene, X 1 and X 3 are hydrogen atoms, or are combined together to form a single bond or alkylene, X 3 is absent or represents alkyl having 1 to 6 carbon atoms, or alkenyl having 3 to 6 carbon atoms, when X 3 is absent, Y is absent, a and b are
  • an LNP described herein comprises a lipid, e.g., an ionizable lipid, disclosed in US 2013/0195920, which is incorporated herein by reference in its entirety.
  • the lipids disclosed in US 2013/0195920 are of formula (I), which has a branched alkyl at the alpha position adjacent to the biodegradable group (between the biodegradable group and the teriary carbon): or a salt thereof (e.g., a pharmaceutically acceptable salt thereof), wherein R′ is absent, hydrogen, or alkyl (e.g., C 1 -C 4 alkyl); with respect to R 1 and R 2 , (i) R 1 and R 2 are each, independently, optionally substituted alkyl, alkenyl, alkynyl, cycloalkylalkyl, heterocycle, or R 10 ; (ii) R 1 and R 2 , together with the nitrogen atom to which they are
  • X and Y are each, independently, alkylene or alkenylene (e.g., C 4 to C 20 alkylene or C 4 to C 20 alkenylene); M 1 and M 2 are each, independently, a biodegradable group (e.g., —OC(O)—, —C(O)O—, — SC(O)—, —C(O)S—, —OC(S)—, —C(S)O, —S—S—, C(R 5 ) ⁇ N—, —N ⁇ C(R 5 )—, — C(R 5 ) ⁇ N—O—, —O—N ⁇ C(R 5 )—, —C(O)(NR 5 )—, —N(R 5 )C(O)—, —C(S)(NR 5 )—, — N(R 5 )C(O)—, —N(R 5 )C(O)N(R 5 )
  • the lipids disclosed in US 2013/0195920 are of formula (II), which has a branched alkyl at the alpha position adjacent to the biodegradable group (between the biodegradable group and the terminus of the tail, i.e., Z 1 or Z 2 ): or a salt thereof (e.g., a pharmaceutically acceptable salt thereof), wherein R′ is absent, hydrogen, or alkyl (e.g., C 1 -C 4 alkyl); with respect to R 1 and R 2 , (i) R 1 and R 2 are each, independently, optionally substituted alkyl, alkenyl, alkynyl, cycloalkylalkyl, heterocycle, or R 10 ; (ii) R 1 and R 2 , together with the nitrogen atom to which they are attached, form an optionally substituted heterocylic ring; or (iii) one of R 1 and R 2 is optionally substituted alkyl, alkenyl, alky
  • the lipids disclosed in US 2013/0195920 are of formula (III), which has a branching point at a position that is 2-6 carbon atoms (i.e., at the beta ( ⁇ ), gamma ( ⁇ ), delta ( ⁇ ), epsilon ( ⁇ ) or zeta position ( ⁇ ) adjacent to the biodegradable group (between the biodegradable group and the terminus of the tail, i.e., Z 1 or Z 2 ): or a salt thereof (e.g., a pharmaceutically acceptable salt thereof), wherein R′, R 1 , R 2 , R, R 3 , R 4 , R 10 , Q, R 5 , M 1 , M 2 , R z , a, and b are defined as in formula (I); L 1 and L 2 are each, independently, C 1 -C 5 alkylene or C 2 -C 5 alkenylene; X and Y are each, independently, alkylene (e.g.,
  • the cationic lipid disclosed in US 2013/0195920 is a compound of formula (IV), which has a branching point at a position that is 2-6 carbon atoms (i.e., at beta ( ⁇ ), gamma ( ⁇ ), delta ( ⁇ ), epsilon ( ⁇ ) or zeta position ( ⁇ ) adjacent to the biodegradable group (between the biodegradable group and the terminus of the tail, i.e., Z 1 or Z 2 ): or a salt thereof (e.g., a pharmaceutically acceptable salt thereof), wherein R′, R 1 , R 2 , R, R 3 , R 4 , R 10 , Q, R 5 , M 2 , R z , a, and b are defined as in formula (I); L 1 and L 2 and are each, independently,
  • -L 1 -C(Z) 3 is —CH 2 C(CH 3 ) 3 .
  • -L 1 -C(Z) 3 is —CH 2 CH 2 C(CH 3 ) 3 .
  • the lipids disclosed in US 2013/0195920 are of formula (V), which has an alkoxy or thioalkoxy (i.e., —S-alkyl) group substitution on at least one tail: or a salt thereof (e.g., a pharmaceutically acceptable salt thereof), wherein R′, R 1 , R 2 , R, R 3 , R 4 , R 10 , Q, R 5 , M 1 , M 2 , a, and b are defined as in formula (I);
  • X and Y are each, independently, alkylene (e.g., C 6 -C 8 alkylene) or alkenylene, wherein the alkylene or alkenylene group is optionally substituted with one or two fluorine atoms at the alpha position to the M 1 or M 2 group;
  • Z 1 and Z 2 are each, independently, C 8 -C 14 alkyl or C 8 -C 14 alkenyl, wherein (i) the C 8 -C
  • the lipids disclosed in US 2013/0195920 are of formula (VIA), which has one or more fluoro substituents on at least one tail at a position that is either alpha to a double bond or alpha to a biodegradable group: or a salt thereof (e.g., a pharmaceutically acceptable salt thereof), wherein R 1 , R 2 , R, a, and b are as defined with respect to formula (I); Q is absent or is —O—, —NH—, —S—, —C(O)—, —C(O)O—, —OC(O)—, —C(O)N(R 4 )—, —N(R 5 )C(O)—, —S—S—, —OC(O)O—, —O—N ⁇ C(R 5 )—, —C(R 5 ) ⁇ N—O—, — OC(O)N(R 5 )—, —N(R
  • the terminus of R 9 and R 10 is separated from the tertiary carbon atom marked with an asterisk (*) by a chain of 18-22 carbon atoms (e.g., 18-20 carbon atoms).
  • the lipids disclosed in US 2013/0195920 are of formula (VIB), which has one or more fluoro substituents on at least one tail at a position that is either alpha to a double bond or alpha to a biodegradable group: or a salt thereof (e.g., a pharmaceutically acceptable salt thereof), wherein R′, R 1 , R 2 , R, R 3 , R 4 , R 10 , Q, R 5 , M 1 , M 2 , a, and b are defined as in formula (I); X and Y are each, independently, alkylene (e.g., C 6 -C 8 alkylene) or alkenylene, wherein the alkylene or alkenylene group is optionally substituted with one or two fluorine atoms at the alpha position to the M 1 or M 2 group; and Z 1 and Z 2 are each, independently, C 8 -C 14 alkyl or C 8 -C 14 alkenyl, wherein said
  • the lipids disclosed in US 2013/0195920 are of formula (VII), which has an acetal group as a biodegradable group in at least one tail: or a salt thereof (e.g., a pharmaceutically acceptable salt thereof), wherein R′, R 1 , R 2 , R, R 3 , R 4 , R 10 , Q, R 5 , a, and b are defined as in formula (I);
  • X and Y are each, independently, alkylene (e.g., C 6 -C 8 alkylene) or alkenylene, wherein the alkylene or alkenylene group is optionally substituted with one or two fluorine atoms at the alpha position to the M 1 or M 2 group
  • M 1 and M 2 are each, independently, a biodegradable group (e.g., —OC(O)—, —C(O)O—, — SC(O)—, —C(O)S—, —OC(S)—
  • an LNP described herein comprises a lipid, e.g., an ionizable lipid, disclosed in US 2015/0005363, which is incorporated herein by reference in its entirety.
  • an LNP described herein comprises a lipid, e.g., an ionizable lipid, disclosed in US 2014/0308304, which is incorporated herein by reference in its entirety.
  • the lipid disclosed in US 2014/0308304 is a compound of formula (I): or a salt thereof (e.g., a pharmaceutically acceptable salt thereof), wherein Xaa is a D- or L-amino acid residue having the formula —NR N —CR 1 R 2 —(C ⁇ O)—, or a peptide of amino acid residues having the formula — ⁇ NR N —CR 1 R 2 —(C ⁇ O) ⁇ n —, wherein n is 2 to 20; R 1 is independently, for each occurrence, a non-hydrogen, substituted or unsubstituted side chain of an amino acid; R 2 and R N are independently, for each occurrence, hydrogen, an organic group consisting of carbon, oxygen, nitrogen, sulfur, and hydrogen atoms, or any combination of the foregoing, and having from 1 to 20 carbon atoms, C (1-5) alkyl, cycloalkyl, cycloalkylalkyl, C (3-5)
  • the lipid disclosed in US 2014/0308304 is a compound of formula (IA): or a salt thereof (e.g., a pharmaceutically acceptable salt thereof), wherein Z and Xaa are as defined with respect to formula (I) (the variables which are used in the definition of Xaa, namely R N , R 1 and R 2 , are also as defined in formula (I)); each occurrence of R is, independently, —(CR 3 R 4 )—; each occurrence of R 3 and R 4 are, independently H, halogen, OH, alkyl, alkoxy, —NH 2 , alkylamino, or dialkylamino (in some embodiments, each occurrence of R 3 and R 4 are, independently H or C 1 -C 4 alkyl); or R 3 and R 4 , together with the carbon atom to which they are directly attached, form a cycloalkyl group, wherein no more than three R groups in each chain between the —Z-Xaa- C(O)
  • the lipids disclosed in US 2014/0308304 are of the formula (IC): or a salt thereof (e.g., a pharmaceutically acceptable salt thereof), wherein Z and Xaa are as defined with respect to formula (I) (the variables which are used in the definition of Xaa, namely R N , R 1 and R 2 , are also as defined in formula (I)); each of R 9 and R 10 are, independently, alkylene or alkenylene; each of R 11 and R 12 are, independently, alkyl or alkenyl, optionally terminated by COOR 13 wherein each R 13 is independently unsubstituted alkyl (e.g., C 1 -C 4 alkyl such as methyl or ethyl), substituted alkyl (such as benzyl), or cycloalkyl; M 1 and M 2 are each, independently, a biodegradable group (e.g., —OC(O)—, —C(O)O—, — SC(O
  • R 7 is selected from lysyl, ornithyl, 2,3-diaminobutyryl, histidyl and an acyl moiety of the formula: t is 1, 2 or 3; the NH 3 + moiety in the acyl moiety in R 7 is optionally absent; each occurrence of Y ⁇ is independently a pharmaceutically acceptable anion (e.g., halide, such as chloride);
  • R 5 and R 6 are each, independently a lipophilic tail derived from a naturally-occurring or synthetic lipid, phospholipid, glycolipid, triacylglycerol, glycerophospholipid, sphingolipid, ceramide, sphingomyelin, cerebroside, or ganglioside, wherein the tail may contain a steroid; or a substituted or unsubstituted C (3-22) alkyl
  • the lipids disclosed in US 2014/0308304 are of the formula (IIA): or a salt thereof (e.g., a pharmaceutically acceptable salt thereof), wherein: R 7 and s are as defined with respect to formula (II); each occurrence of R is, independently, —(CR 3 R 4 )—; each occurrence of R 3 and R 4 are, independently H, halogen, OH, alkyl, alkoxy, —NH 2 , alkylamino, or dialkylamino (in some embodiments, each occurrence of R 3 and R 4 are, independently H or C 1 -C 4 alkyl); or R 3 and R 4 , together with the carbon atom to which they are directly attached, form a cycloalkyl group, wherein no more than three R groups in each chain attached to the nitrogen N* are cycloalkyl (e.g., cyclopropyl); Q 1 and Q 2 are each, independently, absent, —O—, —
  • the lipid disclosed in US 2014/0308304 are of the formula (IIC): or a salt thereof (e.g., a pharmaceutically acceptable salt thereof), wherein: R 7 and s are as defined with respect to formula (II); each of R 9 and R 10 are independently alkyl (e.g., C 12 -C 24 alkyl) or alkenyl (e.g., C 12 -C 24 alkenyl); each of R 11 and R 12 are independently alkyl or alkenyl, optionally terminated by COOR 13 where each R 13 is independently alkyl (e.g., C 1 -C 4 alkyl such as methyl or ethyl); M 1 and M 2 are each, independently, a biodegradable group (e.g., —OC(O)—, —C(O)O—, — SC(O)—, —C(O)S—, —OC(S)—, —C(S)O—, —S—
  • the lipid disclosed in US 2014/0308304 is a compound of the formula (4): wherein: X is N or P; R 1 , R 2 , R, a, b, M 1 , and M 2 are as defined with respect to formula (I); Q is absent or is —O—, —NH—, —S—, —C(O)O—, —OC(O)—, —C(O)N(R 4 )—, — N(R 5 )C(O)—, —S—S—, —OC(O)O—, —O—N ⁇ C(R 5 )—, —C(R 5 ) ⁇ N—O—, — OC(O)N(R 5 )—, —N(R 5 )C(O)N(R 5 )—, —N(R 5 )C(O)O—, —C(O)S—, —C(S)O— or — C(
  • the lipid disclosed in US 2014/0308304 is a compound of the formula (5) wherein: X is N or P; R 1 , R 2 , R, a, and b are as defined with respect to formula (I); Q is absent or is —O—, —NH—, —S—, —C(O)O—, —OC(O)—, —C(O)N(R 4 )—, — N(R 5 )C(O)—, —S—S—, —OC(O)O—, —O—N ⁇ C(R 5 )—, —C(R 5 ) ⁇ N—O—, — OC(O)N(R 5 )—, —N(R 5 )C(O)N(R 5 )—, —N(R 5 )C(O)O—, —C(O)S—, —C(S)O— or — C(R 5 ) ⁇ N—O
  • the lipids disclosed in US 2014/0308304 are of Formula A: or a pharmaceutically acceptable salt or stereoisomer thereof, wherein: n is 0-6 (e.g., n is 0, 1 or 2); R 1 and R 2 are independently selected from H, (C 1 -C 6 )alkyl, heterocyclyl, and a polyamine, wherein said alkyl, heterocyclyl and polyamine are optionally substituted with one or more sub stituents selected from R′, or R 1 and R 2 can be taken together with the nitrogen to which they are attached to form a monocyclic heterocycle with 3-7 (e.g., 4-7) members optionally containing, in addition to the nitrogen, one or two additional heteroatoms selected from N, O and S, said monocyclic heterocycle is optionally substituted with one or more substituents selected from R′; R 3 is selected from H and (C 1 -C 6 )alkyl, wherein said alkyl is optionally substituted with one or
  • the lipids disclosed in US 2014/0308304 are of formula B: or a pharmaceutically acceptable salt or stereoisomer thereof, wherein: n is 0, 1, 2, 3, 4, or 5; R 6 and R 7 are each independently (i) C 1 -C 4 linear or branched alkyl (e.g., methyl or ethyl) optionally substituted with 1-4 R′, or (ii) C 3 -C 8 cycloalkyl (e.g., C 3 -C 6 cycloalkyl); or R 6 and R 7 together with the nitrogen atom adjacent to them form a 3-6 membered ring; L 1 is a C 4 -C 22 alkyl or C 4 -C 22 alkenyl, said alkyl or alkenyl optionally interrupted by or terminated with one or more biodegradable groups; and said alkyl or alkenyl is optionally substituted with 1-5 sub stituents selected from R′; and L 2 is a C
  • lipids disclosed in US 2014/0308304 are of formula C: or a pharmaceutically acceptable salt or stereoisomer thereof, wherein: n is 0, 1, 2, 3, 4, or 5; L 1 is a C 4 -C 22 alkyl or C 4 -C 22 alkenyl, said alkyl or alkenyl optionally has one or more biodegradable groups; each biodegradable group independently interrupts the alkyl or alkenyl group or is substituted at the terminus of the alkyl or alkenyl group, and said alkyl or alkenyl is optionally substituted with 1-5 sub stituents selected from R′; and L 2 is a C 4 -C 22 alkyl or C 4 -C 22 alkenyl, said alkyl or alkenyl optionally interrupted by or terminated with one or more biodegradable groups; and said alkyl or alkenyl is optionally substituted with 1-5 sub stituents selected from R′; each
  • the lipid disclosed in US 2014/0308304 are of formula D: or a pharmaceutically acceptable salt or stereoisomer thereof, wherein m is 0, 1, 2, or 3; n is 0, 1, 2, 3, 4, or 5; R 6 and R 7 are each independently (i) C 1 -C 4 linear or branched alkyl (e.g., methyl or ethyl) optionally substituted with 1-4 R′, or (ii) C 3 -C 8 cycloalkyl (e.g., C 3 -C 6 cycloalkyl); or R 6 and R 7 together with the nitrogen atom adjacent to them form a 3-6 membered ring; L 1 is a C 4 -C 22 alkyl or C 4 -C 22 alkenyl, said alkyl or alkenyl optionally interrupted by or terminated with one or more biodegradable groups; and said alkyl or alkenyl is optionally substituted with 1-5 substituents selected from R′; and L 2
  • lipid disclosed in US 2014/0308304 are of formula E: or a pharmaceutically acceptable salt or stereoisomer thereof, wherein n is 0, 1, 2, 3, 4, or 5; the group “amino acid” is an amino acid residue; L 1 is a C 4 -C 22 alkyl or C 4 -C 22 alkenyl, said alkyl or alkenyl optionally interrupted by or terminated with one or more biodegradable groups, and said alkyl or alkenyl is optionally substituted with 1-5 sub stituents selected from R′; and L 2 is a C 4 -C 22 alkyl or C 4 -C 22 alkenyl, said alkyl or alkenyl optionally interrupted by or terminated with one or more biodegradable groups, and said alkyl or alkenyl is optionally substituted with 1-5 sub stituents selected from R′; each occurrence of R′ is independently selected from halogen, R′′, OR′′, SR′′,
  • the amino acid residue in formula E may have the formula —C(O)—C(R 9 )(NH 2 ), where R 9 is an amino acid side chain.
  • the lipid disclosed in US 2014/0308304 are of formula F: or a pharmaceutically acceptable salt or stereoisomer thereof, wherein: R 6 and R 7 are independently (i) C 1 -C 4 linear or branched alkyl (e.g., methyl or ethyl) optionally substituted with 1-4 R′, or (ii) C 3 -C 8 cycloalkyl (e.g., C 3 -C 6 cycloalkyl); or R 6 and R 7 together with the nitrogen atom adjacent to them form a 3-6 membered ring; L 1 is a C 4 -C 22 alkyl or C 4 -C 22 alkenyl optionally interrupted by or terminated with one or more biodegradable groups, and said alkyl or alkenyl is optionally substituted
  • the lipid disclosed in US 2014/0308304 are of formula G: or a pharmaceutically acceptable salt or stereoisomer thereof, wherein: n is 0, 1, 2, 3, 4, or 5; q is 1, 2, 3, or 4 R 6 and R 7 are independently (i) C 1 -C 4 linear or branched alkyl (e.g., methyl or ethyl) optionally substituted with 1-4 R′, or (ii) C 3 -C 8 cycloalkyl (e.g., C 3 -C 6 cycloalkyl); L 1 is a C 4 -C 22 alkyl or C 4 -C 22 alkenyl optionally interrupted by or terminated with one or more biodegradable groups, and said alkyl or alkenyl is optionally substituted with 1-5 substituents selected from R′; and L 2 is a C 4 -C 22 alkyl or C 4 -C 22 alkenyl optionally interrupted by or terminated with one or more biodegrad
  • an LNP described herein comprises a lipid, e.g., an ionizable lipid, disclosed in US 2013/0053572, which is incorporated herein by reference in its entirety.
  • the lipids disclosed in US 2013/0053572 are of Formula A: wherein: n is 0, 1 or 2; R 1 and R 2 are independently selected from H, (C 1 -C 6 )alkyl, heterocyclyl, and a polyamine, wherein said alkyl, heterocyclyl and polyamine are optionally substituted with one or more substituents selected from R′, or R 1 , and R 2 can be taken together with the nitrogen to which they are attached to form a monocyclic heterocycle with 4-7 members optionally containing, in addition to the nitrogen, one or two additional heteroatoms selected from N, O and S, said monocyclic heterocycle is optionally substituted with one or more substituents selected from R′; R 3 is selected from H and (C 1
  • an LNP described herein comprises a lipid, e.g., an ionizable lipid, disclosed in US Application publication US2017/0119904, which is incorporated by reference herein, in its entirety.
  • an LNP described herein comprises a lipid, e.g., an ionizable lipid, disclosed in PCT Application publication WO2021/204179, which is incorporated by reference herein, in its entirety.
  • an LNP described herein comprises a lipid, e.g., an ionizable lipid, disclosed in PCT Application PCT/US2022/031383, which is incorporated by reference herein, in its entirety.
  • an LNP described herein comprises an ionizable lipid of Table 2: Table 2 – Exemplary Ionizable Lipids
  • an LNP comprises a structural lipid.
  • Structural lipids can be selected from the group consisting of, but are not limited to, cholesterol, fecosterol, fucosterol, beta sitosterol, sitosterol, ergosterol, campesterol, stigmasterol, brassicasterol, tomatidine, cholic acid, sitostanol, litocholic acid, tomatine, ursolic acid, alpha-tocopherol, and mixtures thereof.
  • the structural lipid is cholesterol.
  • the structural lipid includes cholesterol and a corticosteroid (such as prednisolone, dexamethasone, prednisone, and hydrocortisone), or any combinations thereof.
  • a structural lipid is described in international patent application WO2019152557A1, which is incorporated herein by reference in its entirety.
  • a structural lipid is a cholesterol analog.
  • a cholesterol analog may enhance endosomal escape as described in Patel et al., Naturally- occuring cholesterol analogues in lipid nanoparticles induce polymorphic shape and enhance intracellular delivery of mRNA, Nature Communications (2020), which is incorporated herein by reference.
  • a structural lipid is a phytosterol. Using a phytosterol may enhance endosomal escape as described in Herrera et al., Illuminating endosomal escape of polymorphic lipid nanoparticles that boost mRNA delivery, Biomaterials Science (2020), which is incorporated herein by reference. [00797] In some embodiments, a structural lipid contains plant sterol mimetics for enhanced endosomal release. iii. PEGylated lipids [00798] A PEGylated lipid is a lipid modified with polyethylene glycol. In some embodiments, the LNP comprises a compound of Formula I or a pharmaceutically acceptable salt thereof, as described herein above.
  • the LNP comprises a compound of Formula II or a pharmaceutically acceptable salt thereof, as described herein above.
  • an LNP comprises an additional PEGylated lipid or PEG- modified lipid.
  • a PEGylated lipid may be selected from the non-limiting group consisting of PEG-modified phosphatidylethanolamines, PEG-modified phosphatidic acids, PEG-modified ceramides, PEG-modified dialkylamines, PEG-modified diacylglycerols, PEG-modified dialkylglycerols, and mixtures thereof.
  • a PEG lipid may be PEG-c-DOMG, PEG- DMG, PEG-DLPE, PEG-DMPE, PEG-DPPC, or a PEG-DSPE lipid.
  • the LNP comprises a PEGylated lipid disclosed in one of US 2019/0240354; US 2010/0130588; US 2021/0087135; WO 2021/204179; US 2021/0128488; US 2020/0121809; US 2017/0119904; US 2013/0108685; US 2013/0195920; US 2015/0005363; US 2014/0308304; US 2013/0053572; WO 2019/232095A1; WO 2021/077067; WO 2019/152557; US 2015/0203446; US 2017/0210697; US 2014/0200257; or WO 2019/089828A1, each of which is incorporated by reference herein in their entirety.
  • an LNP of the present disclosure comprises a phospholipid.
  • Phospholipids useful in the compositions and methods may be selected from the non-limiting group consisting of 1,2-distearoyl-sn-glycero-3-phosphocholine (DSPC), 1,2- dioleoyl-sn-glycero-3-phosphoethanolamine (DOPE), 1,2-dilinoleoyl-sn-glycero-3- phosphocholine (DLPC), 1,2-dimyristoyl-sn-glycero-phosphocholine (DMPC), 1.2-dioleoyl-sn- glycero-3-phosphocholine (DOPC), 1,2-dipalmitoyl-sn-glycero-3-phosphocholine (DPPC), 1,2- diundecanoyl-sn-glycero-phosphocholine (DUPC), 1-palmitoyl-2-oleoyl-sn-glycero-3-phosphocholine (DSPC), 1,2- dio
  • an LNP includes DSPC. In certain embodiments, an LNP includes DOPE. In some embodiments, an LNP includes both DSPC and DOPE. [00802] In some embodiments, a phospholipid tail may be modified in order to promote endosomal escape as described in U.S.2021/0121411, which is incorporated herein by reference.
  • the LNP comprises a phospholipid disclosed in one of US 2019/0240354; US 2010/0130588; US 2021/0087135; WO 2021/204179; US 2021/0128488; US 2020/0121809; US 2017/0119904; US 2013/0108685; US 2013/0195920; US 2015/0005363; US 2014/0308304; US 2013/0053572; WO 2019/232095A1; WO 2021/077067; WO 2019/152557; US 2017/0210697; or WO 2019/089828A1, each of which is incorporated by reference herein in their entirety.
  • phospholipids disclosed in US 2020/0121809 have the following structure: wherein R 1 and R 2 are each independently a branched or straight, saturated or unsaturated carbon chain (e.g., alkyl, alkenyl, alkynyl).
  • Targeting moieties [00805]
  • the lipid nanoparticle further comprises a targeting moiety.
  • the targeting moiety may be an antibody or a fragment thereof.
  • the targeting moiety may be capable of binding to a target antigen.
  • the pharmaceutical composition comprises a targeting moiety that is operably connected to a lipid nanoparticle.
  • the targeting moiety is capable of binding to a target antigen.
  • the target antigen is expressed in a target organ.
  • the target antigen is expressed more in the target organ than it is in the liver.
  • the targeting moiety is an antibody as described in WO2016189532A1, which is incorporated herein by reference.
  • the targeted particles are conjugated to a specific anti-CD38 monoclonal antibody (mAb), which allows specific delivery of the siRNAs encapsulated within the particles at a greater percentage to B-cell lymphocytes malignancies (such as MCL) than to other subtypes of leukocytes.
  • mAb specific anti-CD38 monoclonal antibody
  • the lipid nanoparticles may be targeted when conjugated/attached/associated with a targeting moiety such as an antibody.
  • a targeting moiety such as an antibody.
  • Zwitterionic amino lipids [00810] In some embodiments, an LNP comprises a zwitterionic lipid. In some embodiments, an LNP comprising a zwitterionic lipid does not comprise a phospholipid. [00811] Zwitterionic amino lipids have been shown to be able to self-assemble into LNPs without phospholipids to load, stabilize, and release mRNAs intracellular as described in U.S. Patent Application 20210121411, which is incorporated herein by reference in its entirety.
  • Zwitterionic, ionizable cationic and permanently cationic helper lipids enable tissue-selective mRNA delivery and CRISPR-Cas9 gene editing in spleen, liver and lungs as described in Liu et al., Membrane-destablizing ionizable phospholipids for organ-selective mRNA delivery and CRISPR-Cas gene editing, Nat Mater. (2021), which is incorporated herein by reference in its entirety.
  • the zwitterionic lipids may have head groups containing a cationic amine and an anionic carboxylate as described in Walsh et al., Synthesis, Characterization and Evaluation of Ionizable Lysine-Based Lipids for siRNA Delivery, Bioconjug Chem. (2013), which is incorporated herein by reference in its entirety.
  • Ionizable lysine-based lipids containing a lysine head group linked to a long-chain dialkylamine through an amide linkage at the lysine ⁇ -amine may reduce immunogenicity as described in Walsh et al., Synthesis, Characterization and Evaluation of Ionizable Lysine-Based Lipids for siRNA Delivery, Bioconjug Chem. (2013).
  • the LNP compositions of the present disclosure further comprise one or more additional lipid components capable of influencing the tropism of the LNP.
  • the LNP further comprises at least one lipid selected from DDAB, EPC, 14PA, 18BMP, DODAP, DOTAP, and C12-200 (see Cheng, et al. Nat Nanotechnol.2020 April; 15(4): 313–320.; Dillard, et al. PNAS 2021 Vol.118 No.52.).
  • a nanoparticle includes an ionizable lipid, a phospholipid, a PEG lipid, and a structural lipid.
  • the lipid component of the nanoparticle composition includes about 30 mol % to about 60 mol % ionizable lipid, about 0 mol % to about 30 mol % phospholipid, about 18.5 mol % to about 48.5 mol % structural lipid, and about 0 mol% to about 10 mol% of PEG lipid, provided that the total mol % does not exceed 100%.
  • the lipid component of the nanoparticle composition includes about 35 mol % to about 55 mol % ionizable lipid, about 5 mol % to about 25 mol % phospholipid, about 30 mol % to about 40 mol % structural lipid, and about 0 mol % to about 10 mol % of PEG lipid.
  • the lipid component includes about 50 mol % ionizable lipid, about 10 mol % phospholipid, about 38.5 mol % structural lipid, and about 1.5 mol% of PEG lipid.
  • the lipid component includes about 40 mol % ionizable lipid, about 20 mol % phospholipid, about 38.5 mol % structural lipid, and about 1.5 mol % of PEG lipid. In another particular embodiment, the lipid component includes about 48.5 mol % ionizable lipid, about 10 mol % phospholipid, about 40 mol % structural lipid, and about 1.5 mol % of PEG lipid. In another particular embodiment, the lipid component includes about 48.5 mol % ionizable lipid, about 10 mol % phospholipid, about 39 mol % structural lipid, and about 2.5 mol % of PEG lipid. In some embodiments, the phospholipid may be DOPE or DSPC.
  • the PEG lipid may be PEG-DMG and/or the structural lipid may be cholesterol.
  • the amount of active agent in a nanoparticle composition may depend on the size, composition, desired target and/or application, or other properties of the nanoparticle composition as well as on the properties of the active agent.
  • the amount of active agent useful in a nanoparticle composition may depend on the size, sequence, and other characteristics of the active agent.
  • the relative amounts of active agent and other elements (e.g., lipids) in a nanoparticle composition may also vary.
  • the wt/wt ratio of the lipid component to an enzyme in a nanoparticle composition may be from about 5:1 to about 60:1, such as 5:1, 6:1, 7:1, 8:1, 9:1, 10:1, 11:1, 12:1, 13:1, 14:1, 15:1, 16:1, 17:1, 18:1, 19:1, 20:1, 25:1, 30:1, 35:1, 40:1, 45:1, 50:1, and 60:1.
  • the amount of a enzyme in a nanoparticle composition may, for example, be measured using absorption spectroscopy (e.g., ultraviolet-visible spectroscopy).
  • absorption spectroscopy e.g., ultraviolet-visible spectroscopy
  • the E:P ratio of the composition refers to the molar ratio of nitrogen atoms in one or more lipids to the number of phosphate groups in an RNA active agent. In general, a lower E:P ratio is preferred.
  • the one or more enzymes, lipids, and amounts thereof may be selected to provide an E:P ratio from about 2:1 to about 30:1, such as 2:1, 3:1, 4:1, 5:1, 6:1, 7:1, 8:1, 9:1, 10:1, 12:1, 14:1, 16:1, 18:1, 20:1, 22:1, 24:1, 26:1, 28:1, or 30:1.
  • the E:P ratio may be from about 2:1 to about 8:1. In other embodiments, the E:P ratio is from about 5:1 to about 8:1.
  • the E:P ratio may be about 5.0:1, about 5.5:1, about 5.67:1, about 6.0:1, about 6.5:1, or about 7.0:1.
  • the characteristics of a nanoparticle composition may depend on the components thereof.
  • a nanoparticle composition including cholesterol as a structural lipid may have different characteristics than a nanoparticle composition that includes a different structural lipid.
  • the characteristics of a nanoparticle composition may depend on the absolute or relative amounts of its components. For instance, a nanoparticle composition including a higher molar fraction of a phospholipid may have different characteristics than a nanoparticle composi tion including a lower molar fraction of a phospholipid.
  • Nanoparticle compositions may be characterized by a variety of methods. For example, microscopy (e.g., transmission electron microscopy or scanning electron microscopy) may be used to examine the morphology and size distribution of a nanoparticle composition. Dynamic light scattering or potentiometry (e.g., potentiometric titrations) may be used to measure Zeta potentials. Dynamic light scattering may also be utilized to determine particle sizes.
  • microscopy e.g., transmission electron microscopy or scanning electron microscopy
  • Dynamic light scattering or potentiometry e.g., potentiometric titrations
  • Dynamic light scattering may also be utilized to determine particle sizes.
  • the mean size of a nanoparticle composition may be between 10s of nm and 100s of nm, e.g., measured by dynamic light scattering (DLS).
  • DLS dynamic light scattering
  • the mean size may be from about 40 nm to about 150 nm, such as about 40 nm, 45 nm, 50 nm, 55 nm, 60 nm, 65 nm, 70 nm, 75 nm, 80 nm, 85 nm, 90 nm, 95 nm, 100 nm, 105 nm, 110 nm, 115nm, 120 nm, 125 nm, 130 nm, 135 nm, 140 nm, 145 nm, or 150 nm.
  • the mean size of a nanoparticle composition may be from about 50 nm to about 100 nm, from about 50 nm to about 90 nm, from about 50 nm to about 80 nm, from about 50 nm to about 70 nm, from about 50 nm to about 60 nm, from about 60 nm to about 100 nm, from about 60 nm to about 90 nm, from about 60 nm to about 80 nm, from about 60 nm to about 70 nm, from about 70 nm to about 100 nm, from about 70 nm to about 90 nm, from about 70 nm to about 80 nm, from about 80 nm to about 100 nm, from about 80 nm to about 90 nm, or from about 90 nm to about 100 nm.
  • the mean size of a nanoparticle composition may be from about 70 nm to about 100 nm. In a particular embodiment, the mean size may be about 80 nm. In other embodiments, the mean size may be about 100 nm.
  • a nanoparticle composition may be relatively homogenous.
  • a polydispersity index may be used to indicate the homogeneity of a nanoparticle composition, e.g., the particle size distribution of the nanoparticle compositions. A small (e.g., less than 0.3) polydispersity index generally indicates a narrow particle size distribution.
  • a nanoparticle composition may have a polydispersity index from about 0 to about 0.25, such as 0.01, 0.02, 0.03, 0.04, 0.05, 0.06, 0.07, 0.08, 0.09, 0.10, 0.11, 0.12, 0.13, 0.14, 0.15, 0.16, 0.17, 0.18, 0.19, 0.20, 0.21, 0.22, 0.23, 0.24, or 0.25.
  • the Zeta potential of a nanoparticle composition may be used to indicate the electrokinetic potential of the composition.
  • the Zeta potential may describe the surface charge of a nanoparticle composition.
  • Nanoparticle compositions with relatively low charges, positive or negative, are generally desirable, as more highly charged species may interact undesirably with cells, tissues, and other elements in the body.
  • the Zeta potential of a nanoparticle composition may be from about -10 mV to about +20 mV, from about -10 mV to about +15 mV, from about -10 mV to about +10 mV, from about -10 mV to about +5 mV, from about -10 mV to about 0 mV, from about -10 mV to about -5 mV, from about -5 mV to about +20 mV, from about -5 mV to about +15 mV, from about -5 mV to about +10 mV, from about -5 mV to about +5 mV, from about -5 mV to about 0 mV, from about 0 mV, to about +20 mV, from about 0 mV to about +15 m
  • the efficiency of encapsulation of a payload describes the amount of payload that is encapsulated or otherwise associated with a nanoparticle composition after preparation, relative to the initial amount provided.
  • the encapsulation efficiency is desirably high (e.g., close to 100%).
  • the encapsulation efficiency may be measured, for example, by comparing the amount of payload in a solution containing the nanoparticle composition before and after breaking up the nanoparticle composition with one or more organic solvents or detergents. Fluorescence may be used to measure the amount of free payload in a solution.
  • the encapsulation efficiency of a therapeutic and/or prophylactic may be at least 50%, for example 50%, 55%, 60%.65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100%. In some embodiments, the encapsulation efficiency may be at least 80%. In certain embodiments, the encapsulation efficiency may be at least 90%. [00821] Lipids and their method of preparation are disclosed in, e.g., U.S. Patent Nos. 8,569,256, 5,965,542 and U.S.
  • a nanoparticle composition may include any substance useful in pharmaceutical compositions.
  • the nanoparticle composition may include one or more pharmaceutically acceptable excipients or accessory ingredients such as, but not limited to, one or more solvents, dispersion media, diluents, dispersion aids, suspension aids, granulating aids, disintegrants, fillers, glidants, liquid vehicles, binders, surface active agents, isotonic agents, thickening or emulsifying agents, buffering agents, lubricating agents, oils, preservatives, and other species.
  • Excipients such as waxes, butters, coloring agents, coating agents, flavorings, and perfuming agents may also be included.
  • lipids or liposomal formulations including nanoparticles and methods of administration include, but are not limited to, U.S. Patent Publication 20030203865, 20020150626, 20030032615, and 20040048787, which are specifically incorporated by reference to the extent they disclose formulations and other related aspects of administration and delivery of nucleic acids. Methods used for forming particles are also disclosed in U.S. Pat.
  • the LNP encapsulates the engineered retron, e.g., an engineered nucleic acid construct, ncRNA, vector system, RNA molecule, and/or engineered nucleic acid-enzyme construct as described herein.
  • the lipid nanoparticle comprises: one or more ionizable lipids; one or more structural lipids; one or more PEGylated lipids; and one or more phospholipids.
  • the one or more ionizable lipids is selected from the group consisting of those disclosed in Table X.
  • the one or more structural lipids are selected from the group consisting of cholesterol, fecosterol, beta sitosterol, sitosterol, ergosterol, campesterol, stigmasterol, brassicasterol, tomatidine, tomatine, ursolic acid, alpha-tocopherol, prednisolone, dexamethasone, prednisone, and hydrocortisone.
  • the one or more PEGylated lipids are selected from the group consisting of PEG-c-DOMG, PEG-DMG, PEG- DLPE, PEG-DMPE, PEG-DPPC, and PEG-DSPE.
  • the one or more phospholipids are selected from the group consisting of 1,2-distearoyl-sn-glycero-3-phosphocholine (DSPC), 1,2-dioleoyl-sn-glycero-3- phosphoethanolamine (DOPE), 1,2-dilinoleoyl-sn-glycero-3-phosphocholine (DLPC), 1,2- dimyristoyl-sn-glycero-phosphocholine (DMPC), 1.2-dioleoyl-sn-glycero-3-phosphocholine (DOPC), 1,2-dipalmitoyl-sn-glycero-3-phosphocholine (DPPC), 1,2-diundecanoyl-sn-glycero- phosphocholine (DUPC), 1-palmitoyl-2-oleoyl-sn-glycero-3-phosphocho line (POPC), 1,2-di-O- octadecenyl-
  • DOPE 1,2-diste
  • the lipid nanoparticle comprises about 48.5 mol % ionizable lipid, about 10 mol % phospholipid, about 40 mol % structural lipid, and about 1.5 mol% of PEG lipid. [00828] In some embodiments, the lipid nanoparticle comprises about 48.5 mol % ionizable lipid, about 10 mol % phospholipid, about 39 mol % structural lipid, and about 2.5 mol% of PEG lipid. In some embodiments, the LNP further comprises a targeting moiety operably connected to the LNP.
  • the LNP further comprises one or more additional components selected from the group consisting of DDAB, EPC, 14PA, 18BMP, DODAP, DOTAP, and C12-200.
  • the engineered retron can be used for gene transfer, which may be performed under ex vivo or in vivo conditions.
  • Ex vivo gene therapy refers to the isolation of cells from a subject, the delivery of a nucleic acid into cells in vitro, and the return of the modified cells back into the subject. This may involve the collection of a biological sample comprising cells from the subject. For example, blood can be obtained by venipuncture, and solid tissue samples can be obtained by surgical techniques according to methods well known in the art.
  • the subject who receives the cells is also the subject from whom the cells are harvested or obtained, which provides the advantage that the donated cells are autologous.
  • cells can be obtained from another subject (e.g., a donor), a culture of cells from a donor, or from established cell culture lines. Accordingly, in some embodiments the cells are allogeneic to the recipient.
  • Cells may be obtained from the same or a different species than the subject to be treated, but preferably are of the same species, and more preferably of the same immunological profile as the subject.
  • Such cells can be obtained, for example, from a biological sample comprising cells from a close relative or matched donor, then transfected with nucleic acids (e.g., comprising an engineered retron), and administered to a subject in need of genome modification, for example, for treatment of a disease or condition.
  • nucleic acids e.g., comprising an engineered retron
  • the engineered retron can be introduced in vivo (e.g., used in gene therapy) by physically delivering the engineered retron to a subject. Examples of physically introducing the engineered retron includes via injections, electroporation and transfection (e.g., calcium-mediated or liposome tranfection, or the like). K.
  • kits comprising engineered retrons (e.g., engineered nucleic acid constructs, or engineered nucleic acid-enzyme constructs) as described herein.
  • the kit provides an engineered retron construct or a vector system comprising such a retron construct.
  • the engineered retron construct included in the kit, comprises a heterologous sequence capable of providing a cell with a nucleic acid encoding a protein or regulatory RNA of interest, a cellular barcode, a donor polynucleotide suitable for use in gene editing, e.g., by homology directed repair (HDR) or recombination-mediated genetic engineering (recombineering), or a CRISPR protospacer DNA sequence for use in molecular recording.
  • HDR homology directed repair
  • recombination-mediated genetic engineering recombineering
  • CRISPR protospacer DNA sequence for use in molecular recording.
  • Other agents may also be included in the kit such as transfection agents, host cells, suitable media for culturing cells, buffers, and the like.
  • agents can be provided in liquid or solid form in any convenient packaging (e.g., stick pack, dose pack, etc.).
  • the agents of a kit can be present in the same or separate containers.
  • the kits may contain any one or more of the components described herein in one or more containers.
  • the components may be prepared sterilely, packaged in a syringe and shipped refrigerated. Alternatively it may be housed in a vial or other container for storage. A second container may have other components prepared sterilely.
  • the kits may include the active agents premixed and shipped in a vial, tube, or other container.
  • kits may have a variety of forms, such as a blister pouch, a shrink wrapped pouch, a vacuum sealable pouch, a sealable thermoformed tray, or a similar pouch or tray form, with the accessories loosely packed within the pouch, one or more tubes, containers, a box or a bag.
  • the kits may be sterilized after the accessories are added, thereby allowing the individual accessories in the container to be otherwise unwrapped.
  • the kits can be sterilized using any appropriate sterilization techniques, such as radiation sterilization, heat sterilization, or other sterilization methods known in the art.
  • kits may also include other components, depending on the specific application, for example, containers, cell media, salts, buffers, reagents, syringes, needles, a fabric, such as gauze, for applying or removing a disinfecting agent, disposable gloves, a support for the agents prior to administration, etc.
  • kits comprising a nucleic acid construct comprising a nucleotide sequence encoding the various components of the retron-based editing system described herein.
  • the subject kits may further include (in some embodiments) instructions for practicing the subject methods. These instructions may be present in the subject kits in a variety of forms, one or more of which may be present in the kit.
  • these instructions may be present in printed information on a suitable medium or substrate, e.g., a piece or pieces of paper on which the information is printed, in the packaging of the kit, in a package insert, and the like.
  • a suitable medium or substrate e.g., a piece or pieces of paper on which the information is printed, in the packaging of the kit, in a package insert, and the like.
  • a computer readable medium e.g., diskette, compact disk (CD), flash drive, and the like, on which the information has been recorded.
  • Yet another form of these instructions that may be present is a website address which may be used via the internet to access the information at a remote site. In some embodiments, information provided on the website is periodically updated to provide, for example, the most up-to-date information.
  • the written instructions may be in a form prescribed by a governmental agency regulating the manufacture, use, or sale of pharmaceuticals or biological products, which can also reflect approval by the agency of manufacture, use or sale for animal administration.
  • “promoted” includes all methods of doing business including methods of education, hospital and other clinical instruction, scientific inquiry, drug discovery or development, academic research, pharmaceutical industry activity including pharmaceutical sales, and any advertising or other promotional activity including written, oral and electronic communication of any form, associated with the disclosure.
  • kits comprising one or more nucleic acid constructs (e.g., one or more mRNA or circular RNA molecules encoding the components of the retron-based genome editing system)
  • all nucleic acid constructs can be based on RNA molecules, i.e., and “all-RNA system.”
  • each of the components of the editing system could be expressed from a mRNA molecule, which would be delivered to a target cell by one more more delivery methods (e.g., LNP delivery).
  • One aspect of the disclosure provides an isolated host cell that includes one or more of the compositions described herein, including, but not limited to, engineered retrons and/or retron components, engineered ncRNAs, engineered msDNA, engineered RT, nucleic acid molecules encoding the engineered retrons and/or retron components, and vector or vector systems encoding the engineered retrons and/or retron components, and any combinations thereof.
  • the host cell is a prokaryotic cell, an archaeal cell, or a eukaryotic host cell.
  • the eukaryotic host cell is a mammalian cell, such as a human cell, a non-human cell, or a non-human mammalian cell.
  • the host cell is an artificial cell or genetically modified cell.
  • the host cell is in vitro, such as a tissue culture cell.
  • the host cell is within a living host organism.
  • Cells that may contain any of the compositions described herein. The methods described herein are used to deliver recombinant retrons or components thereof into a eukaryotic cell (e.g., a mammalian cell, such as a human cell).
  • the cell is in vitro (e.g., cultured cell.
  • the cell is in vivo (e.g., in a subject such as a human subject).
  • the cell is ex vivo (e.g., isolated from a subject and may be administered back to the same or a different subject).
  • the present disclosure contemplates the use of any suitable host cell.
  • the cell host can be a mammalian cell. Mammalian cells of the present disclosure include human cells, primate cells (e.g., vero cells), rat cells (e.g., GH3 cells, OC23 cells) or mouse cells (e.g., MC3T3 cells).
  • human cell lines including, without limitation, human embryonic kidney (HEK) cells, HeLa cells, cancer cells from the National Cancer Institute's 60 cancer cell lines (NCI60), DU145 (prostate cancer) cells, Lncap (prostate cancer) cells, MCF-7 (breast cancer) cells, MDA-MB-438 (breast cancer) cells, PC3 (prostate cancer) cells, T47D (breast cancer) cells, THP-1 (acute myeloid leukemia) cells, U87 (glioblastoma) cells, SHSY5Y human neuroblastoma cells (cloned from a myeloma) and Saos-2 (bone cancer) cells.
  • HEK human embryonic kidney
  • HeLa cells cancer cells from the National Cancer Institute's 60 cancer cell lines (NCI60)
  • DU145 (prostate cancer) cells Lncap (prostate cancer) cells
  • MCF-7 breast cancer
  • MDA-MB-438 breast cancer
  • PC3 prostate cancer
  • T47D
  • the cells can be human embryonic kidney (HEK) cells (e.g., HEK 293 or HEK 293T cells).
  • the cells can be stem cells (e.g., human stem cells) such as, for example, pluripotent stem cells (e.g., human pluripotent stem cells including human induced pluripotent stem cells (hiPSCs)).
  • stem cell refers to a cell with the ability to divide for indefinite periods in culture and to give rise to specialized cells.
  • a pluripotent stem cell refers to a type of stem cell that is capable of differentiating into all tissues of an organism, but not alone capable of sustaining full organismal development.
  • a human induced pluripotent stem cell refers to a somatic (e.g., mature or adult) cell that has been reprogrammed to an embryonic stem cell-like state by being forced to express genes and factors important for maintaining the defining properties of embryonic stem cells (see, e.g., Takahashi and Yamanaka, Cell 126 (4): 663–76, 2006, incorporated by reference herein).
  • Human induced pluripotent stem cells express stem cell markers and are capable of generating cells characteristic of all three germ layers (ectoderm, endoderm, mesoderm).
  • a host cell is transiently or non-transiently transfected with one or more delivery systems described herein, including virus-based systems, virus-like particle systems, and non- virus-base delivery, including LNPs and liposomes.
  • a cell is transfected as it naturally occurs in a subject.
  • a cell that is transfected is taken from a subject, i.e., ex vivo transfection.
  • the cell is derived from cells taken from a subject, such as a cell line.
  • a wide variety of cell lines for tissue culture are known in the art.
  • cell lines include, but are not limited to, C8161, CCRF-CEM, MOLT, mIMCD- 3, NHDF, HeLa-S3, Huh1, Huh4, Huh7, HUVEC, HASMC, HEKn, HEKa, MiaPaCell, Panc1, PC- 3, TF1, CTLL-2, C1R, Rat6, CV1, RPTE, A10, T24, J82, A375, ARH-77, Calu1, SW480, SW620, SKOV3, SK-UT, CaCo2, P388D1, SEM-K2, WEHI-231, HB56, TIB55, Jurkat, J45.01, LRMB, Bcl-1, BC-3, IC21, DLD2, Raw264.7, NRK, NRK-52E, MRC5, MEF, Hep G2, HeLa B, HeLa T4, COS, COS-1, COS-6, COS-M6A, BS-C-1 monkey kidney epithelial, BALB
  • Cell lines are available from a variety of sources known to those with skill in the art (see, e.g., the American Type Culture Collection (ATCC) (Manassus, Va.)).
  • ATCC American Type Culture Collection
  • a cell transfected with one or more retron delivery systems described herein is used to establish a new cell line comprising one or more nucleic acid molecules encoding the recombinant retron-based gene editing systems described herein, or encoding at last a component of said systems (e.g., a recombinant ncRNA or a recombinant retron RT).
  • ATCC American Type Culture Collection
  • a cell transfected with one or more retron delivery systems described herein is used to establish a new cell line comprising one or more nucleic acid molecules encoding the recombinant retron-based gene editing systems described herein, or encoding at last a component of said systems (e.g., a recombinant ncRNA or a
  • compositions [00843]
  • the engineered retron-based genome editing systems described herein, or one or more components thereof may be provided as pharmaceutical compositions.
  • one or more LNPs or other non-virus-based delivery system comprising one or more circular or linear RNA molecules encoding each of the components of the retron-based genome editing system may be formulated as a pharmaceutical composition for administering to a subject in need (e.g., a human in need of gene editing).
  • Formulations can include, without limitation, saline, liposomes, lipid nanoparticles, polymers, peptides, proteins, cells transfected with viral vectors (e.g., for transfer or transplantation into a subject) and combinations thereof.
  • Formulations of the pharmaceutical compositions described herein may be prepared by any method known or hereafter developed in the art of pharmacology.
  • pharmaceutical composition refers to compositions comprising at least one active ingredient and optionally one or more pharmaceutically acceptable excipients.
  • preparatory methods include the step of associating the active ingredient with an excipient and/or one or more other accessory ingredients.
  • the phrase “active ingredient” generally refers an engineered retron as described herein.
  • a pharmaceutical composition in accordance with the present disclosure may be prepared, packaged, and/or sold in bulk, as a single unit dose, and/or as a plurality of single unit doses.
  • a “unit dose” refers to a discrete amount of the pharmaceutical composition comprising a predetermined amount of the active ingredient.
  • the amount of the active ingredient is generally equal to the dosage of the active ingredient which would be administered to a subject and/or a convenient fraction of such a dosage such as, for example, one-half or one-third of such a dosage.
  • compositions comprising any of the various components of the recombinant retron-based genome editing systems described herein, including, but not limited to, engineered retrons and/or retron components, engineered ncRNAs, engineered msDNA, engineered RT, nucleic acid molecules encoding the engineered retrons and/or retron components, programmable nucleases (e.g., RNA- guided nucleases), guide RNAs, and vector or vector systems encoding the engineered retrons and/or retron components, and any combinations thereof.
  • programmable nucleases e.g., RNA- guided nucleases
  • guide RNAs e.g., RNA- guided nucleases
  • vector or vector systems encoding the engineered retrons and/or retron components, and any combinations thereof.
  • pharmaceutical composition refers to a composition formulated for pharmaceutical use.
  • the pharmaceutical composition further comprises a pharmaceutically acceptable carrier.
  • the pharmaceutical composition comprises additional agents (e.g. for specific delivery, increasing half-life, or other therapeutic compounds).
  • pharmaceutically-acceptable carrier means a pharmaceutically-acceptable material, composition or vehicle, such as a liquid or solid filler, diluent, excipient, manufacturing aid (e.g., lubricant, talc magnesium, calcium or zinc stearate, or steric acid), or solvent encapsulating material, involved in carrying or transporting the compound from one site (e.g., the delivery site) of the body, to another site (e.g., organ, tissue or portion of the body).
  • manufacturing aid e.g., lubricant, talc magnesium, calcium or zinc stearate, or steric acid
  • solvent encapsulating material involved in carrying or transporting the compound from one site (e.g., the delivery site) of the body, to another site (e.g., organ, tissue or portion of the body).
  • a pharmaceutically acceptable carrier is“acceptable” in the sense of being compatible with the other ingredients of the formulation and not injurious to the tissue of the subject (e.g., physiologically compatible, sterile, physiologic pH, etc.).
  • materials which can serve as pharmaceutically-acceptable carriers include: (1) sugars, such as lactose, glucose and sucrose; (2) starches, such as corn starch and potato starch; (3) cellulose, and its derivatives, such as sodium carboxymethyl cellulose, methylcellulose, ethyl cellulose, microcrystalline cellulose and cellulose acetate; (4) powdered tragacanth; (5) malt; (6) gelatin; (7) lubricating agents, such as magnesium stearate, sodium lauryl sulfate and talc; (8) excipients, such as cocoa butter and suppository waxes; (9) oils, such as peanut oil, cottonseed oil, safflower oil, sesame oil, olive oil, corn oil and soybean oil; (9) oils, such
  • the pharmaceutical composition is formulated for delivery to a subject, e.g., for gene editing.
  • Suitable routes of administrating the pharmaceutical composition described herein include, without limitation: topical, subcutaneous, transdermal, intradermal, intralesional, intraarticular, intraperitoneal, intravesical, transmucosal, gingival, intradental, intracochlear, transtympanic, intraorgan, epidural, intrathecal, intramuscular, intravenous, intravascular, intraosseus, periocular, intratumoral, intracerebral, and intracerebroventricular administration.
  • the pharmaceutical composition described herein is administered locally to a diseased site (e.g., tumor site).
  • the pharmaceutical composition described herein is administered to a subject by injection, by means of a catheter, by means of a suppository, or by means of an implant, the implant being of a porous, non-porous, or gelatinous material, including a membrane, such as a sialastic membrane, or a fiber.
  • the pharmaceutical composition described herein is delivered in a controlled release system.
  • a pump may be used (see, e.g., Langer, 1990, Science 249:1527-1533; Sefton, 1989, CRC Crit. Ref. Biomed.
  • polymeric materials can be used.
  • Polymeric materials See, e.g., Medical Applications of Controlled Release (Langer and Wise eds., CRC Press, Boca Raton, Fla., 1974); Controlled Drug Bioavailability, Drug Product Design and Performance (Smolen and Ball eds., Wiley, New York, 1984); Ranger and Peppas, 1983, Macromol. Sci. Rev. Macromol. Chem.23:61.
  • the pharmaceutical composition is formulated in accordance with routine procedures as a composition adapted for intravenous or subcutaneous administration to a subject, e.g., a human.
  • pharmaceutical composition for administration by injection are solutions in sterile isotonic aqueous buffer.
  • the pharmaceutical can also include a solubilizing agent and a local anesthetic such as lignocaine to ease pain at the site of the injection.
  • the ingredients are supplied either separately or mixed together in unit dosage form, for example, as a dry lyophilized powder or water free concentrate in a hermetically sealed container such as an ampoule or sachette indicating the quantity of active agent.
  • a hermetically sealed container such as an ampoule or sachette indicating the quantity of active agent.
  • the pharmaceutical is to be administered by infusion, it can be dispensed with an infusion bottle containing sterile pharmaceutical grade water or saline.
  • an ampoule of sterile water for injection or saline can be provided so that the ingredients can be mixed prior to administration.
  • a pharmaceutical composition for systemic administration may be a liquid, e.g., sterile saline, lactated Ringer’s or Hank’s solution.
  • the pharmaceutical composition can be in solid forms and re-dissolved or suspended immediately prior to use. Lyophilized forms are also contemplated.
  • the pharmaceutical composition can be contained within a lipid particle or vesicle, such as a liposome or microcrystal or LNP, which is also suitable for parenteral administration.
  • the particles can be of any suitable structure, such as unilamellar or plurilamellar, so long as compositions are contained therein.
  • SPLP stabilized plasmid- lipid particles
  • DOPE fusogenic lipid dioleoylphosphatidylethanolamine
  • PEG polyethyleneglycol
  • Positively charged lipids such as N-[1-(2,3-dioleoyloxi)propyl]-N,N,N-trimethyl- amoniummethylsulfate, or “DOTAP,” are particularly preferred for such particles and vesicles.
  • DOTAP N-[1-(2,3-dioleoyloxi)propyl]-N,N,N-trimethyl- amoniummethylsulfate
  • the pharmaceutical composition can be provided as a pharmaceutical kit comprising (a) a container containing a recombinant retron-based genome editing system or one or more components thereof in lyophilized form and (b) a second container containing a pharmaceutically acceptable diluent (e.g., sterile water) for injection.
  • a pharmaceutically acceptable diluent e.g., sterile water
  • the pharmaceutically acceptable diluent can be used for reconstitution or dilution of the lyophilized system of the invention.
  • Optionally associated with such container(s) can be a notice in the form prescribed by a governmental agency regulating the manufacture, use or sale of pharmaceuticals or biological products, which notice reflects approval by the agency of manufacture, use or sale for human administration.
  • an article of manufacture containing materials useful for the treatment of the diseases described above is included.
  • the article of manufacture comprises a container and a label.
  • Suitable containers include, for example, bottles, vials, syringes, and test tubes.
  • the containers may be formed from a variety of materials such as glass or plastic.
  • the container holds a composition that is effective for treating a disease described herein and may have a sterile access port.
  • the container may be an intravenous solution bag or a vial having a stopper pierce- able by a hypodermic injection needle.
  • the active agent in the composition is a compound of the invention.
  • the label on or associated with the container indicates that the composition is used for treating the disease of choice.
  • the article of manufacture may further comprise a second container comprising a pharmaceutically-acceptable buffer, such as phosphate-buffered saline, Ringer's solution, or dextrose solution. It may further include other materials desirable from a commercial and user standpoint, including other buffers, diluents, filters, needles, syringes, and package inserts with instructions for use. N.
  • the engineered retron comprising the heterologous nucleic acid sequence can be used in a variety of applications, several non-limiting examples of which are described herein. In general, the engineered retron can be used in any suitable organism. In some embodiments, the organism is a eukaryote. [00860] In some embodiments, the organism is an animal. In some embodiments, the animal is a fish, an amphibian, a reptile, a mammal, or a bird. In some embodiments, the animal is a farm animal or agriculture animal.
  • Non-limiting examples of farm and agriculture animals include horses, goats, sheep, swine, cattle, llamas, alpacas, and birds, e.g., chickens, turkeys, ducks, and geese.
  • the animal is a non-human primate, e.g., baboons, capuchin monkeys, chimpanzees, lemurs, macaques, marmosets, tamarins, spider monkeys, squirrel monkeys, and vervet monkeys.
  • the animal is a pet.
  • Non-limiting examples of pets include dogs, cats horses, wolfs, rabbits, ferrets, gerbils, hamsters, chinchillas, fancy rats, guinea pigs, canaries, parakeets, and parrots.
  • the organism is a plant. Plants that may be transfected with an engineered retron include monocots and dicots.
  • Particular examples include, but are not limited to, corn (maize), sorghum, wheat, sunflower, potato, cotton, rice, soybean, sugarbeet, sugarcane, tobacco, barley, and oilseed rape, Brassica sp., alfalfa, rye, millet, safflower, peanuts, sweet potato, cassava, coffee, coconut, pineapple, citrus trees, cocoa, tea, banana, avocado, fig, guava, mango, olive, papaya, cashew, macadamia, almond, oats, vegetables, ornamentals, and conifers.
  • Vegetables include, but are not limited to, crucifers, peppers, tomatoes, lettuce, green beans, lima beans, peas, and members of the genus Cucumis such as cucumber, cantaloupe, and musk melon.
  • Ornamentals include, but are not limited to, azalea, hydrangea, hibiscus, roses, tulips, daffodils, petunias, carnation, poinsettia, and chrysanthemum.
  • heterologous nucleic acid sequences can be added to the subject engineered retron to provide a cell with a heterologous nucleic acid encoding a protein or regulatory RNA of interest, a cellular barcode, a donor polynucleotide suitable for use in gene editing, e.g., by homology directed repair (HDR) or recombination-mediated genetic engineering (recombineering), or a CRISPR protospacer DNA sequence for use in molecular recording, as discussed further below.
  • HDR homology directed repair
  • recombination-mediated genetic engineering recombineering
  • CRISPR protospacer DNA sequence for use in molecular recording, as discussed further below.
  • the engineered retrons described herein may be used for research tools, such as kits, functional genomics assays, and generating engineered cell lines and animal models for research and drug screening.
  • the kit may comprise one or more reagents in addition to the engineered retron, such as a buffer, a control reagent, a control vector, a control RNA polynucleotide, a reagent for in vitro production of the polypeptide from DNA, and adaptors for sequencing.
  • a buffer can be, for example, a stabilization buffer, a reconstituting buffer, a diluting buffer, a wash buffer, or a buffer for introducing a polypeptide and/or polynucleotide of the kit into a cell.
  • a kit can comprise one or more additional reagents specific for plants.
  • One or more additional reagents for plants can include, for example, soil, nutrients, plants, seeds, spores, Agrobacterium, a T-DNA vector, and a pBINAR vector.
  • Production of Protein or RNA [00864]
  • the single-stranded msDNA generated by the engineered retron of the invention can be used to produce a desired product of interest in cells.
  • the retron is engineered with a heterologous sequence encoding a polypeptide of interest to allow production of the polypeptide from the retron msDNA generated in a cell.
  • the polypeptide of interest may be any type of protein/peptide including, without limitation, an enzyme, an extracellular matrix protein, a receptor, transporter, ion channel, or other membrane protein, a hormone, a neuropeptide, an antibody, or a cytoskeletal protein, a functional fragment thereof, or a biologically active domain of interest.
  • the protein is a therapeutic protein, therapeutic antibody for use in treatment of a disease, or a template to fix a mutation or mutated exon in the genome.
  • Non-limiting examples of polypeptides of interest include: growth hormones, insulin-like growth factors (IGF-1), Fat-1, Phytase, xylanase, beta-glucanase, Lysozyme or lysostaphin, Histone deacetylase such as HDAC6, CD163, etc.
  • the retron is engineered with a heterologous sequence encoding an RNA of interest to allow production of the RNA from the retron in a cell.
  • RNA of interest may be any type of RNA including, without limitation, a RNA interference (RNAi) nucleic acid or regulatory RNA such as, but not limited to, a microRNA (miRNA), a small interfering RNA (siRNA), a short hairpin RNA (shRNA), a small nuclear RNA (snRNA), a long non-coding RNA (IncRNA), an antisense nucleic acid, and the like.
  • RNAi RNA interference
  • miRNA microRNA
  • siRNA small interfering RNA
  • shRNA short hairpin RNA
  • snRNA small nuclear RNA
  • IncRNA long non-coding RNA
  • an antisense nucleic acid and the like.
  • Gene Editing [00868]
  • the retron is used for genome editing a desired site.
  • a retron is engineered with a heterologous nucleic acid sequence encoding a donor polynucleotide suitable for use with nuclease genome editing system.
  • the nuclease is designed to specifically target a location proximal to the desired edit (the nuclease should be designed such that it will not cut the target once the edit is properly installed).
  • the nuclease e.g., CAS or non-CAS
  • the nuclease is linked to the retron, either by direct fusion to the RT or by fusion of the msDNA to the gRNA (only applicable for RNA-guided nucleases).
  • a heterologous nucleic acid sequence is inserted into the retron msd. See for example FIG.3, which shows a marker representing the edit.
  • the heterologous nucleic acid sequence has 10-100 or more bp of homologous nucleic acid sequence to the genome on both sides of the desired edit.
  • the desired edit (insertion, deletion, or mutation) is in between the homologous sequence.
  • donor polynucleotides comprise a sequence comprising an intended genome edit flanked by a pair of homology arms responsible for targeting the donor polynucleotide to the target locus to be edited in a cell.
  • the donor polynucleotide typically comprises a 5 ⁇ homology arm that hybridizes to a 5 ⁇ genomic target sequence and a 3 ⁇ homology arm that hybridizes to a 3 ⁇ genomic target sequence.
  • the homology arms are referred to herein as 5 ⁇ and 3 ⁇ (i.e., upstream and downstream) homology arms, which relate to the relative position of the homology arms to the nucleotide sequence comprising the intended edit within the donor polynucleotide.
  • the 5 ⁇ and 3 ⁇ homology arms hybridize to regions within the target locus in the genomic DNA to be modified, which are referred to herein as the “5 ⁇ target sequence” and “3 ⁇ target sequence,” respectively.
  • the homology arm must be sufficiently complementary for hybridization to the target sequence to mediate homologous recombination between the donor polynucleotide and genomic DNA at the target locus.
  • a homology arm may comprise a nucleotide sequence having at least about 80-100% sequence identity to the corresponding genomic target sequence, including any percent identity within this range, such as at least 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% sequence identity thereto, wherein the nucleotide sequence comprising the intended edit can be integrated into the genomic DNA by HDR at the genomic target locus recognized (i.e., having sufficient complementary for hybridization) by the 5 ⁇ and 3 ⁇ homology arms.
  • the corresponding homologous nucleotide sequences in the genomic target sequence flank a specific site for cleavage and/or a specific site for introducing the intended edit.
  • the distance between the specific cleavage site and the homologous nucleotide sequences can be several hundred nucleotides. In some embodiments, the distance between a homology arm and the cleavage site is 200 nucleotides or less (e.g., 0, 10, 20, 30, 50, 75, 100, 125, 150, 175, and 200 nucleotides).
  • the donor polynucleotide is substantially identical to the target genomic sequence, across its entire length except for the sequence changes to be introduced to a portion of the genome that encompasses both the specific cleavage site and the portions of the genomic target sequence to be altered.
  • a homology arm can be of any length, e.g.10 nucleotides or more, 15 nucleotides or more, 20 nucleotides or more, 50 nucleotides or more, 100 nucleotides or more, 250 nucleotides or more, 300 nucleotides or more, 350 nucleotides or more, 400 nucleotides or more, 450 nucleotides or more, 500 nucleotides or more, 1000 nucleotides (1 kb) or more, 5000 nucleotides (5 kb) or more, 10000 nucleotides (10 kb) or more, etc.
  • the 5 ⁇ and 3 ⁇ homology arms are substantially equal in length to one another.
  • the 5 ⁇ and 3 ⁇ homology arms are not necessarily equal in length to one another.
  • one homology arm may be 30% shorter or less than the other homology arm, 20% shorter or less than the other homology arm, 10% shorter or less than the other homology arm, 5% shorter or less than the other homology arm, 2% shorter or less than the other homology arm, or only a few nucleotides less than the other homology arm.
  • the 5 ⁇ and 3 ⁇ homology arms are substantially different in length from one another, e.g. one may be 40% shorter or more, 50% shorter or more, sometimes 60% shorter or more, 70% shorter or more, 80% shorter or more, 90% shorter or more, or 95% shorter or more than the other homology arm.
  • the donor polynucleotide may be used in combination with an RNA-guided nuclease, which is targeted to a particular genomic sequence (i.e., genomic target sequence to be modified) by a guide RNA.
  • a target-specific guide RNA comprises a nucleotide sequence that is complementary to a genomic target sequence, and thereby mediates binding of the nuclease- gRNA complex by hybridization at the target site.
  • the gRNA can be designed with a sequence complementary to the sequence of a minor allele to target the nuclease-gRNA complex to the site of a mutation.
  • the mutation may comprise an insertion, a deletion, or a substitution.
  • the mutation may include a single nucleotide variation, gene fusion, translocation, inversion, duplication, frameshift, missense, nonsense, or other mutation associated with a phenotype or disease of interest.
  • the targeted minor allele may be a common genetic variant or a rare genetic variant.
  • the gRNA is designed to selectively bind to a minor allele with single base-pair discrimination, for example, to allow binding of the nuclease-gRNA complex to a single nucleotide polymorphism (SNP).
  • SNP single nucleotide polymorphism
  • the gRNA may be designed to target disease-relevant mutations of interest for the purpose of genome editing to remove the mutation from a gene.
  • the gRNA can be designed with a sequence complementary to the sequence of a major or wild-type allele to target the nuclease-gRNA complex to the allele for the purpose of genome editing to introduces a mutation into a gene in the genomic DNA of the cell, such as an insertion, deletion, or substitution.
  • Such genetically modified cells can be used, for example, to alter phenotype, confer new properties, or produce disease models for drug screening.
  • the RNA-guided nuclease used for genome modification is a clustered regularly interspersed short palindromic repeats (CRISPR) system Cas nuclease.
  • CRISPR clustered regularly interspersed short palindromic repeats
  • RNA-guided Cas nuclease capable of catalyzing site- directed cleavage of DNA to allow integration of donor polynucleotides by the HDR mechanism can be used in genome editing, including CRISPR system Class 1, Type I, II, or III Cas nucleases; Class 2, Type II nuclease (such as Cas9); a Class 2, Type V nuclease (such as Cpfl), or a Class 2, Type VI nuclease (such as C2c2).
  • Cas proteins include Casl, CaslB, Cas2, Cas3, Cas4, Cas5, Cas5e (CasD), Cas6, Cas6e, Cas6f, Cas7, Cas8al, Cas8a2, Cas8b, Cas8c, Cas9 (Csnl or Csxl2), CaslO, CaslOd, CasF, CasG, CasH, Csyl, Csy2, Csy3, Csel (CasA), Cse2 (CasB), Cse3 (CasE), Cse4 (CasC), Cscl, Csc2, Csa5, Csn2, Csm2, Csm3, Csm4, Csm5, Csm6, Cmrl, Cmr3, Cmr4, Cmr5, Cmr6, Csbl, Csb2, Csb3, Csxl7, Csxl4, CsxlO, Cs
  • a Class 1, type II CRISPR system Cas9 endonuclease is used.
  • Cas9 nucleases from any species, or biologically active fragments, variants, analogs, or derivatives thereof that retain Cas9 endonuclease activity i.e., catalyze site-directed cleavage of DNA to generate double-strand breaks
  • the Cas9 need not be physically derived from an organism but may be synthetically or recombinantly produced.
  • Cas9 sequences from a number of bacterial species are well known in the art and listed in the National Center for Biotechnology Information (NCBI) database.
  • sequences or a variant thereof comprising a sequence having at least about 70-100% sequence identity thereto, including any percent identity within this range, such as 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98, or 99% sequence identity thereto, can be used for genome editing, as described herein. See also Fonfara et al. (2014) Nucleic Acids Res.42(4):2577-90; Kapitonov et al. (2015) J.
  • the genomic target site will typically comprise a nucleotide sequence that is complementary to the gRNA and may further comprise a protospacer adjacent motif (PAM).
  • PAM protospacer adjacent motif
  • the target site comprises 20-30 base pairs in addition to a 3 or more base pair PAM.
  • the first nucleotide of a PAM can be any nucleotide, while the two or more other nucleotides will depend on the specific Cas9 protein that is chosen.
  • Exemplary PAM sequences are known to those of skill in the art and include, without limitation, NNG, NGN, NAG, and NGG, wherein N represents any nucleotide.
  • the allele targeted by a gRNA comprises a mutation that creates a PAM within the allele, wherein the PAM promotes binding of the Cas9-gRNA complex to the allele.
  • the gRNA is 5-50 nucleotides, 10-30 nucleotides, 15- 25 nucleotides, 18-22 nucleotides, or 19-21 nucleotides in length, or any length between the stated ranges, including, for example, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, or 35 nucleotides in length.
  • the guide RNA may be a single guide RNA comprising crRNA and tracrRNA sequences in a single RNA molecule, or the guide RNA may comprise two RNA molecules with crRNA and tracrRNA sequences residing in separate RNA molecules.
  • Cpfl is another class II CRISPR/Cas system RNA-guided nuclease with similarities to Cas9 and may be used analogously. Unlike Cas9, Cpfl does not require a tracrRNA and only depends on a crRNA in its guide RNA, which provides the advantage that shorter guide RNAs can be used with Cpfl for targeting than Cas9. Cpfl is capable of cleaving either DNA or RNA.
  • the PAM sites recognized by Cpfl have the sequences 5 ⁇ -YTN-3 ⁇ (where “Y” is a pyrimidine and “N” is any nucleobase) or 5 ⁇ -TTN-3 ⁇ , in contrast to the G-rich PAM site recognized by Cas9.
  • Cpfl cleavage of DNA produces double-stranded breaks with a sticky-ends having a 4 or 5 nucleotide overhang.
  • C2c1 (Cas12b) is another class II CRISPR/Cas system RNA-guided nuclease that may be used.
  • C2cl similarly to Cas9, depends on both a crRNA and tracrRNA for guidance to target sites. See, e.g., Shmakov et al. (2015) Mol Cell.60(3):385-397, Zhang et al. (2017) Front Plant Sci.8:177; herein incorporated by reference.
  • RNA-guided Fokl nucleases comprise fusions of inactive Cas9 (dCas9) and the Fokl endonuclease (FokI-dCas9), wherein the dCas9 portion confers guide RNA-dependent targeting on Fokl.
  • dCas9 inactive Cas9
  • FokI-dCas9 Fokl endonuclease
  • dCas9 portion confers guide RNA-dependent targeting on Fokl.
  • the RNA-guided nuclease is provided in the form of a protein, optionally where the nuclease is complexed with a gRNA to form a ribonucleoprotein (RNP) complex.
  • the RNA-guided nuclease is provided by a nucleic acid encoding the RNA-guided nuclease, such as an RNA (e.g., messenger RNA) or DNA (expression vector).
  • the RNA-guided nuclease and the gRNA are both provided by vectors, such as the vectors and the vector system described in other parts of the application (all incorporated herein by reference). Both can be expressed by a single vector or separately on different vectors.
  • the vectors encoding the RNA-guided nuclease and gRNA may be included in the vector system comprising the engineered retron msr gene, msd gene and ret gene sequences.
  • the RNA-guided nuclease is fused to the RT and/or the msDNA.
  • the RNP complex may be administered to a subject or delivered into a cell by methods known in the art, such as those described in U.S. Pat. No.11,390,884, which is incorporated by reference herein in its entirety.
  • the endonuclease/gRNA ribonucleoprotein (RNP) complexes are delivered to cells by electroporation. Direct delivery of the RNP complex to a subject or cell eliminates the need for expression from nucleic acids (e.g., transfection of plasmids encoding Cas9 and gRNA). It also eliminates unwanted integration of DNA segments derived from nucleic acid delivery (e.g., transfection of plasmids encoding Cas9 and gRNA).
  • Codon usage may be optimized to further improve production of an RNA-guided nuclease and/or reverse transcriptase (RT) in a particular cell or organism.
  • a nucleic acid encoding an RNA-guided nuclease or reverse transcriptase can be modified to substitute codons having a higher frequency of usage in a yeast cell, a bacterial cell, a human cell, a non-human cell, a mammalian cell, a rodent cell, a mouse cell, a rat cell, or any other host cell of interest, as compared to the naturally occurring polynucleotide sequence.
  • the protein can be transiently, conditionally, or constitutively expressed in the cell.
  • the engineered retron used for genome editing with nuclease genome editing systems can further include accessory or enhancer proteins for recombination.
  • recombination enhancers can include nonhomologous end joining (NHEJ) inhibitors (e.g., inhibitor of DNA ligase IV, a KU inhibitor (e.g., KU70 or KU80), a DNA-PKc inhibitor, or an artemis inhibitor) and homologous directed repair (HDR) promoters, or both, that can enhance or improve more precise genome editing and/or the efficiency of homologous recombination.
  • NHEJ nonhomologous end joining
  • KU inhibitor e.g., KU70 or KU80
  • HDR homologous directed repair
  • the recombination accessory or enhancers can comprise C-terminal binding protein interacting protein (CtIP), cyclinB2, Rad family members (e.g. Rad50, Rad51, Rad52, etc).
  • CtIP C-terminal binding protein interacting protein
  • cyclinB2 Rad family members
  • Rad50, Rad51, Rad52, etc C-terminal binding protein interacting protein
  • HDR may be enhanced by using Cas9 nuclease associated (e.g.
  • an N-terminal fragment of CtIP called HE for HDR enhancer, may be sufficient for HDR stimulation and requires the CtIP multimerization domain and CDK phosphorylation sites to be active.
  • HDR stimulation by the Cas9-HE fusion depends on the guide RNA used, and therefore the guide RNA will be designed accordingly.
  • any target gene or sequence in a host cell can be edited or modified for a desired trait, including but not limited to: Myostatin (e.g., GDF8) to increase muscle growth; Pc POLLED to induce hairlessness; KISS1R to induce bore taint; Dead end protein (dnd) to induce sterility; Nano2 and DDX to induce sterility; CD163 to induce PRRSV resistance; RELA to induce ASFV resilience; CD18 to induce Mannheimia (Pasteurella) haemolytica resilience; NRAMPl to induce tuberculosis resilience; Negative regulators of muscle mass (e.g., Myostatin) to increase muscle mass.
  • Myostatin e.g., GDF8
  • Pc POLLED to induce hairlessness
  • KISS1R to induce bore taint
  • Dead end protein (dnd) to induce sterility
  • Nano2 and DDX to induce sterility
  • CD163 to induce PRRSV resistance
  • RELA
  • Recombineering (recombination-mediated genetic engineering) can be used in modifying chromosomal as well as episomal replicons in cells, for example, to create gene replacements, gene knockouts, deletions, insertions, inversions, or point mutations. Recombineering can also be used to modify a plasmid or bacterial artificial chromosome (BAC), for example, to clone a gene or insert markers or tags.
  • BAC bacterial artificial chromosome
  • the engineered retrons described herein can be used in recombineering applications to provide linear single-stranded or double-stranded DNA for recombination.
  • Homologous recombination may be mediated by bacteriophage proteins such as RecE/RecT from Rac prophage or Redobd from bacteriophage lambda.
  • the linear DNA should have sufficient homology at the 5 ⁇ and 3 ⁇ ends to a target DNA molecule present in a cell (e.g., plasmid, BAC, or chromosome) to allow recombination.
  • a target DNA molecule e.g., plasmid, BAC, or chromosome
  • the linear double-stranded or single-stranded DNA molecule used in recombineering comprises a sequence having the intended edit to be inserted flanked by two homology arms that target the linear DNA molecule to a target site for homologous recombination.
  • Homology arms for recombineering typically range in length from 13-300 nucleotides, or 20 to 200 nucleotides, including any length within this range such as 13, 14, 15, 16, 17, 18, 19, 20, 22, 24, 26, 28, 30, 32, 34, 36, 38, 40, 42, 44, 46, 48, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, 100, 105, 110, 115, 120, 125, 130, 135, 140, 145, 150, 155, 160, 165, 170, 175, 180, 185, 190, 195, or 200 nucleotides in length.
  • a homology arm is at least 15, at least 20, at least 30, at least 40, or at least 50 or more nucleotides in length.
  • Homology arms ranging from 40-50 nucleotides in length generally have sufficient targeting efficiency for recombination; however, longer homology arms ranging from 150 to 200 bases or more may further improve targeting efficiency.
  • the 5 ⁇ homology arm and the 3 ⁇ homology arm differ in length.
  • the linear DNA may have about 50 bases at the 5 ⁇ end and about 20 bases at the 3 ⁇ end with homology to the region to be targeted.
  • the bacteriophage homologous recombination proteins can be provided to a cell as proteins or by one or more vectors encoding the recombination proteins, such as the vector or vector system.
  • one or more vectors encoding the bacteriophage recombination proteins are included in the vector system comprising the engineered retron msr gene, msd gene, and/or ret gene sequences.
  • a number of bacterial strains containing prophage recombination systems are available for recombineering, including, without limitation, DY380, containing a defective l prophage with recombination proteins exo, bet, and gam; EL250, derived from DY380, which in addition to the recombination genes found in DY380, also contains a tightly controlled arabinose-inducible flpe gene (flpe mediates recombination between two identical frt sites); EL350, also derived from DY380, which in addition to the recombination genes found in DY380, also contains a tightly controlled arabinose-inducible ere gene (ere mediates recombination between two identical loxP sites;
  • Recombineering can be carried out by transfecting bacterial cells of such strains with an engineered retron comprising a heterologous sequence encoding a linear DNA suitable for recombineering.
  • an engineered retron comprising a heterologous sequence encoding a linear DNA suitable for recombineering.
  • the heterologous sequence in the engineered retron construct comprises a synthetic CRISPR protospacer DNA sequence to allow molecular recording.
  • the endogenous CRISPR Cas1-Cas2 system is normally utilized by bacteria and archaea to keep track of foreign DNA sequences originating from viral infections by storing short sequences (i.e., protospacers) that confer sequence-specific resistance to invading viral nucleic acids within genome-based arrays. These arrays not only preserve the spacer sequences but also record the order in which the sequences are acquired, generating a temporal record of acquisition events.
  • This system can be adapted to record arbitrary DNA sequences into a genomic CRISPR array in the form of “synthetic protospacers” that are introduced into cells using engineered retrons.
  • Engineered retrons carrying the protospacer sequences can be used for integration of synthetic CRISPR protospacer sequences at a specific genomic locus by utilizing the CRISPR system Cas1-Cas2 complex.
  • Molecular recording can be used to keep track of certain biological events by producing a stable genetic memory tracking code. See, e.g., Shipman et al. (2016) Science 353(6298): aafl 175 and International Patent Application Publication No. WO/2018/191525; herein incorporated by reference in their entireties.
  • the CRISPR-Cas system is harnessed to record specific and arbitrary DNA sequences into a bacterial genome.
  • the DNA sequences can be produced by an engineered retron within the cell.
  • the engineered retron can be used to produce the protospacers within the cell, which are inserted into a CRISPR array within the cell.
  • the cell may be modified to include one or more engineered returns (or vector systems encoding them) that can produce one or more synthetic protospacers in the cell, wherein the synthetic protospacers are added to the CRISPR array.
  • a record of defined sequences, recorded over many days, and in multiple modalities can be generated.
  • the engineered retron comprises an msd protospacer nucleic acid region or an msr protospacer nucleic acid region.
  • the protospacer sequence is first incorporated into the msr RNA, which is reverse transcribed into protospacer DNA.
  • Double stranded protospacer DNA is produced when two complementary protospacer DNA sequences having complementary sequences hybridize, or when a double-stranded structure (such as a hairpin) is formed in a single stranded protospacer DNA (e.g., a single msDNA can form an appropriate hairpin structure to provide the double stranded DNA protospacer).
  • a single stranded DNA produced in vivo from a first engineered retron may be hybridized with a complementary single-stranded DNA produced in vivo from the same retron or a second engineered retron or may form a hairpin structure and then used as a protospacer sequence to be inserted into a CRISPR array as a spacer sequence.
  • the engineered retron(s) should provide sufficient levels of the protospacer sequence within a cell for incorporation into the CRISPR array.
  • the use of protospacers generated within the cell extends the in vivo molecular recording system from only capturing information known to a user, to capturing biological or environmental information that may be previously unknown to a user.
  • an msDNA protospacer sequence in an engineered retron construct may be driven by a promoter that is downstream of a sensor pathway for a biological phenomenon or environmental toxin.
  • the capture and storage of the protospacer sequence in the CRISPR array records the event. If multiple msDNA protospacers are driven by different promoters, the activity of those promoters is recorded (along with anything that may be upstream of the promoters) as well as the relative order of promoter activity (based on the relative position of spacer sequences in the CRISPR array).
  • the CRISPR array may be sequenced to determine whether a given biological or environmental event has taken place and the order of multiple events, given by the presence and relative position of msDNA-derived spacers in the CRISPR array.
  • the synthetic protospacer further comprises an AAG PAM sequence at its 5 ⁇ end. Protospacers including the 5 ⁇ AAG PAM are acquired by the CRISPR array with greater efficiency than those that do not include a PAM sequence.

Abstract

Disclosed are engineered retrons and methods of use such as to modify the genome of a host (e.g., mammalian) cell by delivering the engineered retron or the encoded ncRNA in vitro or in vivo to the host (e.g., mammalian) cell.

Description

ENGINEERED RETRONS AND METHODS OF USE RELATED APPLICATIONS [0001] This application claims priority under 35 U.S.C. § 119(e) to U.S. Provisional Application Serial No.63/301,936, filed January 21, 2022, U.S. Provisional Application Serial No. 63/370,880, filed August 9, 2022, U.S. Provisional Application Serial No.63/373,545, filed August 25, 2022, and U.S. Provisional Application Serial No.63/476,900, filed December 22, 2022, each of which are incorporated herein by reference in their entireties. [0002] All documents cited herein or during their prosecution (“appln cited documents”) and all documents cited or referenced in the appln cited documents, and all documents cited or referenced herein (“herein cited documents”), and all documents cited or referenced in herein cited documents, together with any manufacturer’s instructions, descriptions, product specifications, and product sheets for any products mentioned herein or in any document incorporated by reference herein, are hereby incorporated herein by reference, and may be employed in the practice of the invention. More specifically, all referenced documents are incorporated by reference to the same extent as if each individual document was specifically and individually indicated to be incorporated by reference. SEQUENCE LISTING [0003] This application contains a sequence listing filed in electronic form in eXtensible Markup Language (XML) format entitled RTX006_Sequence_Listing.xml, created on January 20, 2023 and having a size of 32,261,680 bytes. The content of the sequence listing is incorporated herein in its entirety. TECHNICAL FIELD [0004] The present disclosure generally relates to systems, methods and compositions used for precise genome editing, including nucleic acid insertions, replacements, and deletions at targeted and precise genome sites, wherein said systems, methods, and compositions are based on novel and/or engineered retrons. BACKGROUND OF THE INVENTION [0005] Precise genome editing by programmable nucleases (e.g., RNA-guided nucleases (e.g., CRISPR nucleases), zinc-finger nucleases (ZFN), and transcription activator-like effector nucleases (TALENS)) typically relies on homology-directed repair (HDR) and the presence of a donor DNA template at the site of a double-strand break (DSB) induced by the programmable nuclease. It is generally accepted that a limiting step for HDR-dependent precise genome editing is the delivery of donor DNA template to the nuclease-induced DSB (e.g., see Ling et al., “Improving the efficiency of precise genome editing with site-specific Cas9-oligonucleotide conjugates,” Science Advances, 2020, Vol.6, No.15, pp.1-8). Various methods aimed at boosting the efficiency of HDR-dependent editing have been reported, many of which involve the physical tethering of the DNA donor to a component of the precise editing system. Exemplary methods have been discussed in: K. Lee et al., “Synthetically modified guide RNA and donor DNA are a versatile platform for CRISPR-Cas9 engineering,” eLife 6, e25312 (2017); J. Carlson-Stevermer et al., “Assembly of CRISPR ribonucleoproteins with biotinylated oligonucleotides via an RNA aptamer for precise gene editing,” Nat. Commun.8, 1711 (2017); N. Savic, et al., “Covalent linkage of the DNA repair template to the CRISPR-Cas9 nuclease enhances homology-directed repair,” eLife 7, e33761 (2018); and E. J. Aird et al., “Increasing Cas9-mediated homology-directed repair efficiency through covalent tethering of DNA repair template.,” Commun. Biol.1, 54 (2018), each of which are incorporated herein by reference. Despite these efforts, efficiency of HDR-dependent precise editing remains unsatisfactory. [0006] Retrons are defined by their unique ability to produce an unusual satellite DNA known as msDNA (multicopy single-stranded DNA). DNA encoding retrons includes a reverse trancriptase (RT)-coding gene (ret) and a nucleic acid sequence encoding the non-coding RNA (ncRNA), which contains two contiguous and inverted non-coding sequences referred to as the msr and msd. The ret gene and the non-coding RNA (including the msr and msd) are transcribed as a single RNA transcript, which becomes folded into a specific secondary structure following post-transcriptional processing. Once translated, the RT binds the RNA template downstream from the msd locus, initiating reverse transcription of the RNA towards its 5ʹ end, assisted by the 2’OH group present in a conserved branching guanosine residue that acts as a primer. Reverse transcription halts before reaching the msr locus, and the resulting DNA, the msDNA, remains covalently attached to the RNA template via a 2’-5ʹ phosphodiester bond and base-pairing between the 3ʹ ends of the msDNA and the RNA template. The external regions, at the 5ʹ and 3ʹ ends of the msd/msr transcript (a1 and a2, respectively) are complementary and can hybridize, leaving the structures located in the msr and msd regions in internal positions (see FIG.1A). The msr locus, which is not reverse transcribed, forms one to three short stem-loops of variable size, ranging from 3 to 10 base pairs, whereas the msd locus folds into a single/double long hairpin with a highly variable long stem of 10-50 bp in length that is also present in the final msDNA form. [0007] It has recently been reported that retrons may be utilized as a means to provide donor DNA template for HDR-dependent genome editing (e.g., see Lopez et al., “Precise genome editing across kingdoms of life using retron-derived DNA,” Nature Chemical Biology, December 12, 2021, 18, pages199–206 (2022)), however, producing sufficient levels of donor DNA template intracellularly to sufficiently support efficient HDR-dependent editing remains a significant challenge. Improved retron-based genome modification systems are highly desirous in the art. SUMMARY OF THE INVENTION [0008] In one aspect, the present disclosure provides recombinant retrons comprising one or more genetic modifications which improves the functionality and/or properties of a retron. Such genetic modifications can include a mutation, insertion, deletion, inversion, replacement, substitution, or translocation of one or more contiguous or non-contiguous nucleobases in a nucleic acid molecule encoding a retron or a component of a retron, such as an ncRNA or a reverse transcriptase. In various aspects, the retron that becomes modifed with the one or more genetic modifications (i.e., the “pre-modified” or “unmodified” retron or retron component) is a naturally occurring retron or retron component (e.g., naturally occuring ncRNA of Table A or RT) ability to facilitate homology-dependent recombination (or HDR) in a cell, thereby resulting in a relative increase in the concentrations or amounts of msDNA comprising a DNA donor template. In particular embodiments, the recombinant retrons are based on and/or derived from a naturally-occurring retron, such as any retron-related sequence provided by Table X (the introduction of the one or more genetic modifications into a set of 7257 previously unknown retrons discovered through computational methods described herein (e.g., see Examples). In other embodiments, the recombinant retrons are based on introducing the one or more genetic modifications into previously available retron sequences (e.g., the “Mestre et al., Systematic Prediction of Genes Functionally Associated with Bacterial Retrons and Classification of The Encoded Tripartite Systems, Nucleic Acids Research, Volume 48, Issue 22, 16 December 2020, Pages 12632-12647” (incorporated herein by reference) to achieve recombinant retrons with the enhanced ability to produce increased concentrations or amounts of msDNA comprising a DNA donor template. [0009] In another aspect, the present disclosure further provides nucleic acid molecules encoding the recombinant retrons and/or recombinant retron components (e.g., a recombinant ncRNA and/or a recombinant retron RT). In still another aspect, the present disclosure provides genome editing systems comprising recombinant retron components (e.g., recombinant ncRNA and/or recombinant RT), programmable nucleases (e.g., RNA-guided nucleases, such as CRISPR-Cas proteins, ZFPs, and TALENS), and guide RNAs (in the case where RNA-guide nucleases are used in said genome editing systems). In a further aspect, the disclosure provides nucleic acid molecules encoding the described genome editing systems and said components thereof, as well as polypeptides making up the components of said genome editing systems. In yet another aspect, the disclosure provides vectors for transferring and/or expressing said genome editing systems, e.g., under in vitro, ex vivo, and in vivo conditions. In still another aspect, the disclosure provides cell-delivery compositions and methods, including compositions for passive and/or active transport to cells (e.g., plasmids), delivery by virus-based recombinant vectors (e.g., AAV and/or lentivirus vectors), delivery by non-virus-based systems (e.g., liposomes and LNPs), and delivery by virus-like particles. Depending on the delivery system employed, the retron-based genome editing systems described herein may be delivered in the form of DNA (e.g., plasmids or DNA-based virus vectors), RNA (e.g., ncRNA and mRNA delivered by LNPs), a mixture of DNA and RNA, protein (e.g., virus-like particles), and ribonucleoprotein (RNP) complexes. Any suitable combinations of approaches for delivering the components of the herein disclosed retron-based genome editing systems may be employed. In one embodiment, each of the components of the retron-based genome editing system is delivered by an all-RNA system, e.g., the delivery of one or more RNA molecules (e.g., mRNA and/or ncRNA) by one or more LNPs, wherein the one or more RNA molecules form the ncRNA and guide RNA (as needed) and/or are translated into the polypeptide components (e.g., the RT and a programmable nuclease). In yet another aspect, the disclosure provides methods for genome editing by introducing a retron-based genome editing system described herein into a cell (e.g., under in vitro, in vivo, or ex vivo conditions) comprising a target edit site, thereby resulting in an edit at the target edit. In other aspects, the disclosure provides formulations comprising any of the aforementioned components for delivery to cells and/or tissues, including in vitro, in vivo, and ex vivo delivery, recombinant cells and/or tissues modified by the recombinant retron-based genome modification systems and methods described herein, and methods of modifying cells by conducting genome editing and related DNA donor-dependent methods, such as recombineering, or cell recording, using the herein disclosed retron-based genome modification systems. The disclosure also provides methods of making the recombinant retrons, retron-based genome modification systems, vectors, compositions and formulations described herein, as well as to pharmaceutical compositions and kits for modifying cells under in vitro, in vivo, and ex vivo conditions that comprise the herein disclosed genome editing and/or modification systems. [0010] In an embodiment, this disclosure or the inventions herein provide a gene editing system comprising one or more delivery vehicles, wherein: the delivery vehicle(s) comprise RNA cargo; the RNA cargo comprises (a) at least one mRNA molecule encoding (i) a nucleic acid programmable nuclease and (ii) a retron reverse transcriptase, (b) an engineered retron ncRNA, and (c) guide RNA for the programmable nuclease; and each delivery vehicle contains (a)(i) and/or (a)(ii) and/or (b) and/or (c); whereby one delivery vehicle or more than one delivery vehicle delivers (a)(i), (a)(ii), (b), and (c). [0011] In an embodiment, in the gene editing system, (a)(i) and (a)(ii) comprise a single mRNA molecule encoding the nucleic acid programmable nuclease and the retron reverse transcriptase. [0012] In an embodiment, in the gene editing system, (a)(i) and (a)(ii) are encoded and expressed as a fusion protein. [0013] In an embodiment, in the gene editing system (a)(i) and (a)(ii) are encoded and expressed as a fusion protein and the fusion protein comprises the C-terminal end of the nucleic acid programmable nuclease fused to the N-terminal end of the retron reverse transcriptase (nuclease:RT fusion); or the fusion protein comprises the N-terminal end of the nucleic acid programmable nuclease fused to the C-terminal end of the retron reverse transcriptase (RT:nuclease fusion). [0014] In an embodiment, in the gene editing system, (a)(i) and (a)(ii) comprise a first mRNA molecule encoding the nucleic acid programmable nuclease and a second mRNA molecule encoding the retron reverse transcriptase. [0015] In an embodiment, in the gene editing system, (c) is separate from (a)(i), (a)(ii) and (b) or is provided in trans. [0016] In an embodiment, in the gene editing system, (b) the engineered retron ncRNA, and (c) the guide RNA are fused or are provided in cis. [0017] In an embodiment, in the gene editing system, (b) the engineered retron ncRNA, and (c) the guide RNA are fused or are provided in cis and the guide RNA is fused to the 5’ end of the retron ncRNA. [0018] In an embodiment, in the gene editing system, (b) the engineered retron ncRNA, and (c) the guide RNA are fused or are provided in cis and the guide RNA is fused to the 3’ end of the retron ncRNA. [0019] In an embodiment, in the gene editing system, (b) the engineered retron ncRNA, and (c) the guide RNA are fused or are provided in cis and the engineered ncRNA comprises a first guide RNA fused to the 5’ end of the retron ncRNA, and a second guide RNA fused to the 3’ end of the retron ncRNA, and the first and second guide RNAs target different sequences. Thus, on a broader scale, in an embodiment, in the gene editing system, (c) guide RNA for the programmable nuclease, can comprise one or more guides that target the same or different target sequences. Such guide RNA(s) in an embodiment, can be single guide RNA(s) or sgRNA(s); for instance, when the nucleic acid programmable nuclease comprises a Cas9. [0020] In an embodiment, in the gene editing system, the one or more delivery vehicles comprise a liposome or a lipid nanoparticle (LNP). [0021] In an embodiment, in the gene editing system, (a) the at least one mRNA molecule encoding (i) the nucleic acid programmable nuclease and (ii) the retron reverse transcriptase, and (b) the engineered retron ncRNA, are in the same delivery vehicle. [0022] In an embodiment, in the gene editing system, (a) the at least one mRNA molecule encoding (i) the nucleic acid programmable nuclease and (ii) the retron reverse transcriptase, and (b) the engineered retron ncRNA, are in separate delivery vehicles. [0023] In an embodiment, in the gene editing system, the nucleic acid programmable nuclease and the retron reverse transcriptase are encoded on separate mRNA molecules and those separate mRNA molecules of (a)(i) and (a)(ii) are contained in the same delivery vehicle. [0024] In an embodiment, in the gene editing system, the nucleic acid programmable nuclease and the retron reverse transcriptase are encoded on separate mRNA molecules and those separate mRNA molecules of (a)(i) and (a)(ii) are contained in different delivery vehicles. [0025] In an embodiment, in the gene editing system, the engineered retron ncRNA includes a sequence of interest encoding a donor polynucleotide comprising an intended edit to be integrated at a target sequence in a cell, and wherein the donor polynucleotide is flanked by a 5' homology arm that hybridizes to a sequence 5’ to the target sequence and a 3' homology arm that hybridizes to a sequence 3' to the target sequence. In an embodiment, the donor polynucleotide can be heterologous to the cell. In an embodiment, the donor polynucleotide can be endogenous to the cell; for instance, the cell can contain a sequence that is typical for those in a population having a disease state and the donor polynucleotide can be a sequence that is typical for those in the population not having a non-disease state (e.g., the donor can be for a genetic correction or repair of a cell to modify the cell from having a mutation or modification that gives rise to a disease state to having a sequence typical of not having the disease state). Such can be done in an animal cell, or a mammalian cell (e.g., a primate, a non-human primate, or a domesticated mammal such as a cat or dog or horse) or a human cell; for instance to correct, address, treat, mitigate a genetic condition in the animal, mammal, domesticated mammal, cat, dog, horse or human. Such can be done in plant cells to introduce mutations that give rise to favorable phenotypic characteristics such as disease resistance or other favorable plant trait(s). [0026] In an embodiment, in the gene editing system, the nucleic acid programmable nuclease comprises a Cas9 nuclease, a TnpB nuclease, or a Cas12a nuclease. [0027] In an embodiment, in the gene editing system, the engineered retron ncRNA comprises: A) a pre-msr sequence having a first complementary region of the retron ncRNA; B) an msr sequence including an msr stem-loop structure; C) an msd sequence including an msd stem-loop structure and a sequence of interest, wherein said msd sequence templates a single strand DNA product (RT-DNA) in the presence of the retron reverse transcriptase; and D) a post- msd sequence having a second complementary region, wherein the first and second complementary regions form an a1/a2 duplex region of the retron ncRNA, wherein the msr stem- loop structure, the msd stem-loop structure, or the a1/a2 duplex comprise a modification which result in increased editing efficiency in the presence of a nucleic acid programmable nuclease that associates with the one or more guide RNAs, and wherein optionally one or more of the guide RNAs of (c) are coupled to the pre-msr sequence, the post-msd sequence, or both the pre- msr sequence and the post-msd sequence. In such an embodiment where the engineered retron ncRNA comprises A), B), C) and D), wherein the sequence of interest can encode a donor polynucleotide comprising an intended edit to be integrated at a target sequence of a cell, wherein the donor polynucleotide is flanked by a 5' homology arm that hybridizes to a sequence 5’ to the target sequence and a 3' homology arm that hybridizes to a sequence 3' to the target sequence. In such an embodiment where the engineered retron ncRNA comprises A), B), C) and D) (either with the sequence of interest encoding a donor polynucleotide or simply being a sequence of interest), the ncRNA has a nucleotide sequence of Table B, or a nucleotide sequence having at least 75%, 80%, 85%, 90%, 95%, 99%, or 100% sequence identity with a sequence from Table B. The donor polynucleotide can be heterologous to a cell. Alternatively, the donor polynucleotide can be endogenous to the cell. For instance, the cell can contain a sequence that is typical for those in a population having a disease state and the donor polynucleotide can be a sequence that is typical for those in the population not having a non-disease state (e.g., the donor can be for a genetic correction or repair of a cell to modify the cell from having a mutation or modification that gives rise to a disease state to having a sequence typical of not having the disease state). [0028] In an embodiment of the gene editing system, the gene editing system can comprise any combination(s) of the foregoing embodiments of the gene editing system. [0029] In an embodiment, this disclosure or the inventions herein provide a cell, such as an isolated cell comprising the gene editing system disclosed herein, such as in any of the foregoing paragraphs. In an embodiment the cell, e.g., isolated cell, can be a eukaryotic cell. In an embodiment the eukaryotic cell can be a plant cell or an animal cell or a mammalian cell, e.g., an isolated plant cell or an isolated animal cell or an isolated mammalian cell. In an embodiment, the mammalian cell, e.g., an isolated mammalian cell, can be a human cell. In an embodiment, the cell can be a prokaryotic cell, e.g., a bacterial cell. In such an embodiment where the cell is a bacterial cell, the donor polynucleotide can code for antibiotic susceptibility; and thus, the invention can involve a means for addressing antibiotic resistant bacteria by rendering such bacteria susceptible to antibiotics (and a subject to whom the gene editing system is administered can also then receive antibiotics to which the bacteria are rendered susceptible by the gene editing system). [0030] In an embodiment, this disclosure or the inventions herein provide a composition comprising: a) the gene editing system disclosed herein, such as in any of the foregoing paragraphs; and b)a pharmaceutically or veterinarily acceptable carrier. In an embodiment, in the composition the delivery vehicle can comprise a lipid nanoparticle comprising: a) one or more ionizable lipids; b) one or more structural lipids; c) one or more PEGylated lipids; and d)one or more phospholipids. In an embodiment, in the composition the one or more ionizable lipids comprises an ionizable lipid set forth in Table 2. [0031] In an embodiment, this disclosure or the inventions herein provide uses of the gene editing system embodiments and/or the compositions disclosed herein, such as in any of the foregoing paragraphs; for instance, use in modifying a cell or genetically modifying a cell, e.g., a eukaryotic or a prokaryotic cell and/or an animal cell and/or a mammalian and/or a human cell and/or a bacterial cell and/or a plant cell, in vivo, in vitro or ex vivo (e.g., any cell discussed herein wherein the cell comprises an isolated cell). In an embodiment this disclosure or the inventions herein provide uses of the gene editing system embodiments and/or the compositions disclosed herein, such as in any of the foregoing paragraphs; for instance, use in treating or addressing a genetic condition of a subject, [0032] In an embodiment, this disclosure or the inventions herein provide methods of genetically modifying a cell comprising: contacting a gene editing system as herein discussed, such as in any of the foregoing paragraphs, or a composition as herein discussed, such as in any of the foregoing paragraphs (which comprises a gene editing system as herein discussed, such as in any of the foregoing paragraphs), advantageously a gene editing system that includes a sequence of interest encoding a donor polynucleotide comprising an intended edit to be integrated at a target sequence in a cell, said method comprising contacting the composition or the gene editing system with the cell, thereby delivering the RNA cargo to the cell, wherein: the nucleic acid programmable nuclease forms a complex with the guide RNA, wherein said guide RNA directs the complex to the target sequence; the nucleic acid programmable nuclease creates a double-stranded break in in the target sequence; the retron reverse transcriptase and engineered retron ncRNA create RT DNA that comprises the donor polynucleotide; and the donor polynucleotide becomes integrated at the target sequence; whereby editing the cell is genetically modified. In an embodiment, the cell can be a eukaryotic or a prokaryotic cell or an animal cell or a mammalian cell or a human cell or a bacterial cell or a plant cell. [0033] Additional exemplary and non-limiting aspects and embodiments of the disclosure are summarized as follows in the form of numbered paragraphs. 1. An engineered nucleic acid construct comprising: a) a first polynucleotide encoding a non-coding RNA (ncRNA), said first polynucleotide comprising: 1) an msr locus encoding the msr RNA portion of a multi-copy single- stranded DNA (msDNA); and 2) an msd locus encoding the msd RNA portion of the msDNA; and b) one or more heterologous nucleic acids inserted at or within a location selected from: the msd locus, upstream of the msr locus, upstream of the msd locus, and downstream of the msd locus, wherein the ncRNA comprises: (I) an ncRNA listed in Table B, or an ncRNA having at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%%, at least 99.1%, at least 99.2%, at least 99.3%, at least 99.4%, at least 99.5%, at least 99.6%, at least 99.7%, at least 99.8% or at least 99.9% sequence identity with an ncRNA listed in Table B; and/or (II) an ncRNA having a conserved structure of any one of the ncRNA structures of FIGs.2-27; and wherein the ncRNA optionally excludes any ncRNA associated in nature with any one of the retron reverse transcriptases of Table X. 2. The engineered nucleic acid construct of paragraph 1, further comprising a second polynucleotide encoding a reverse transcriptase (RT), or a portion thereof, wherein the encoded RT or portion thereof is capable of synthesizing a DNA copy of at least a portion of the msd locus encoding the msDNA. 3. The engineered nucleic acid construct of paragraph 2, wherein the second polynucleotide comprises: III) a polynucleotide listed in Table A, or a polynucleotide having at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%%, at least 99.1%, at least 99.2%, at least 99.3%, at least 99.4%, at least 99.5%, at least 99.6%, at least 99.7%, at least 99.8% or at least 99.9% sequence identity to a polynucleotide listed in Table A; and/or IV) encodes a consensus amino acid sequence of Table C, or encodes an amino acid sequence having at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%%, at least 99.1%, at least 99.2%, at least 99.3%, at least 99.4%, at least 99.5%, at least 99.6%, at least 99.7%, at least 99.8% or at least 99.9% sequence identity to an amino acid sequence listed in Table C; and/or wherein the second polynucleotide encodes: V) a polypeptide listed in Table A, or a polypeptide having at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%%, at least 99.1%, at least 99.2%, at least 99.3%, at least 99.4%, at least 99.5%, at least 99.6%, at least 99.7%, at least 99.8% or at least 99.9% sequence identity to a polypeptide listed in Table A; and/or VI) a polypeptide comprising a polypeptide consensus sequence listed in Table C, or a polypeptide having at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%%, at least 99.1%, at least 99.2%, at least 99.3%, at least 99.4%, at least 99.5%, at least 99.6%, at least 99.7%, at least 99.8% or at least 99.9% sequence identity to an amino acid sequence listed in Table C; and/or wherein the second polynucleotide optionally does not encode an amino acid sequence listed in Table X. 4. An engineered nucleic acid construct comprising: a) a first polynucleotide encoding a non-coding RNA (ncRNA), said first polynucleotide comprising: 1) an msr locus encoding the msr RNA portion of a multi-copy single- stranded DNA (msDNA); and 2) an msd locus encoding the msd RNA portion of the msDNA; b) one or more heterologous nucleic acids inserted at or within a location selected from: the msd locus, upstream of the msr locus, upstream of the msd locus, and downstream of the msd locus; and c) a second polynucleotide encoding a reverse transcriptase (RT), or a portion thereof, wherein the encoded RT or portion thereof is capable of synthesizing a DNA copy of at least a portion of the msd locus encoding the msDNA, and, wherein the non-coding RNA (ncRNA) of the first polynucleotide optionally has a conserved structure of any one of the ncRNA structures of FIGs.2-27; wherein the second polynucleotide comprises: I) a polynucleotide listed in Table A, or a polynucleotide having at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%%, at least 99.1%, at least 99.2%, at least 99.3%, at least 99.4%, at least 99.5%, at least 99.6%, at least 99.7%, at least 99.8% or at least 99.9% sequence identity to a polynucleotide listed in Table A; and/or wherein the second polynucleotide encodes: II) a polypeptide listed in Table A, or a polypeptide having at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%%, at least 99.1%, at least 99.2%, at least 99.3%, at least 99.4%, at least 99.5%, at least 99.6%, at least 99.7%, at least 99.8% or at least 99.9% sequence identity to a polypeptide listed in Table A; and/or IV) a polypeptide comprising a polypeptide consensus sequence listed in Table C, or a polypeptide having at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%%, at least 99.1%, at least 99.2%, at least 99.3%, at least 99.4%, at least 99.5%, at least 99.6%, at least 99.7%, at least 99.8% or at least 99.9% sequence identity to a polypeptide listed in Table C; and wherein the second polynucleotide optionally does not encode an amino acid sequence of Table X. 4a. An engineered nucleic acid construct, comprising: 1) an msr locus (that encodes the msr RNA portion of an msDNA); 2) an msd locus encoding the msd RNA portion of the msDNA; 3) a sequence encoding a retron reverse transcriptase (RT), wherein said msd RNA is capable of being reverse transcribed to form the msDNA by the retron reverse transcriptase (RT); and, 4) a heterologous nucleic acid inserted at or within the msd locus, upstream of the msr locus, upstream or downstream of the msd locus; wherein the engineered nucleic acid construct optionally has (a) a secondary structure of a wild-type ncRNA of any one of FIGs.2-27 or b) a variant of a), having: i) up to 1, 2, or 3 (e.g., up to 1) nucleotide changes per 10 red lettered- nucleotides; ii) up to 4, 5, or 6 (e.g., up to 1 or 2) nucleotide changes per 10 black lettered- nucleotides; and/or iii) up to 7, 8, or 9 (e.g., up to 3 or 4) nucleotide changes per 10 grey lettered- nucleotides; and/or optionally further comprising: i) 7, 8, 9, or 10 (e.g., 9 or 10) nucleotides present per 10 red-circled nucleotides; ii) 6, 7, 8, 9, or 10 (e.g., 8, 9 or 10) nucleotides present per 10 black-circled nucleotides; iii) 4, 5, 6, 7, 8, 9, or 10 (e.g., 6, 7, 8, 9 or 10) nucleotides present per 10 grey- circled nucleotides; and/or iv) 2, 3, 4, 5, 6, 7, 8, 9, or 10 (e.g., 4, 5, 6, 7, 8, 9, or 10) nucleotides present per 10 white-circled nucleotides. 5. The engineered nucleic acid construct of any one of paragraphs 1 to 4a, comprising one or more sequence modifications (e.g., an insertion, deletion, and/or substitution of one or more nucleotide(s)) in the msr locus and/or the msd locus that: a) modulates (e.g., enhances) reverse transcription, processivity, accuracy/fidelity, and/or production of the msDNA (e.g., in the mammalian cell); b) modulates (e.g., reduces) immunogenicity of ncRNA encoded by the engineered retron (e.g., the msr locus and/or the msd locus) in a host (e.g., a host comprising the mammalian cell); c) modulates (e.g., inhibits, either permanently or transiently) a function of the msDNA; and/or d) modulates (e.g., improves) efficiency of targeted genome editing / engineering. 6. The engineered nucleic acid construct of any one of paragraphs 1 to 4, wherein said engineered nucleic acid construct has a secondary structure of a wild-type retron encoding a wild-type retron ncRNA encompassed by: a) any one of the structures as depicted in FIGs.2-27, or b) a variant of a), having: i) up to 1, 2, or 3 (e.g., up to 1) nucleotide changes per 10 red lettered- nucleotides; ii) up to 4, 5, or 6 (e.g., up to 1 or 2) nucleotide changes per 10 black lettered- nucleotides; and/or iii) up to 7, 8, or 9 (e.g., up to 3 or 4) nucleotide changes per 10 grey lettered- nucleotides; and/or optionally further comprising: i) 7, 8, 9, or 10 (e.g., 9 or 10) nucleotides present per 10 red-circled nucleotides; ii) 6, 7, 8, 9, or 10 (e.g., 8, 9 or 10) nucleotides present per 10 black-circled nucleotides; iii) 4, 5, 6, 7, 8, 9, or 10 (e.g., 6, 7, 8, 9 or 10) nucleotides present per 10 grey- circled nucleotides; and/or iv) 2, 3, 4, 5, 6, 7, 8, 9, or 10 (e.g., 4, 5, 6, 7, 8, 9, or 10) nucleotides present per 10 white-circled nucleotides. 7. The engineered nucleic acid construct of any one of paragraphs 1-6, wherein the nucleic acid construct is engineered by introducing the one or more sequence modifications into a wild-type retron encoding a wild-type ncRNA listed in Table B. 8. The engineered nucleic acid construct of any one of paragraphs 1-7, wherein the one or more sequence modifications in the ncRNA comprises one or more of: (i) a modified (e.g., mutated, reduced, or eliminated) bulge in a1, a2, or both a1 and a2; (ii) an extension or shortening of a1, a2, or both a1 and a2; (iii) an extension or shortening of a spacer sequence between hairpin loops (e.g., S1, S2, S3, and/or S4); (iv) an additional or modified (e.g., mutated or eliminated) bulge in hairpin loops (e.g., L2 and/or L3 (e.g., by removing unpaired bases in the bulge, or by replacing unpaired bases with an equivalent number of base pairs)); (v) a modified (e.g., extended or shortened) length of hairpin loops (e.g., L1, L2, L3, and/or L4); (vi) an alternative L1 and/or L2 having complement, reverse, or reverse complement sequences; (vii) a modified (e.g., increased) number of unpaired bases at the tip of hairpin loops (e.g., L1, L2, L3, and/or L4); (viii) a modified (e.g., increased or decreased) GC content in hairpin loops (e.g., L1, L2, L3, and/or L4); (ix) an insertion of the heterologous nucleic acid in spacer sequences between hairpin loops (e.g., S1, S2, S3 and/or S4), or at the tip of hairpin loops (e.g., L1, L2, L3, and/or L4); (x) a deletion of one or more hairpin loops (e.g., L1, L2, L3 and/or L4); (xi) an addition of a new loop in a spacer sequence between hairpin loops (e.g., S1, S2, S3, and/or S4); (xii) circularization of the ncRNA with the 5ʹ end and the 3ʹ end of the ncRNA being connected either directly, or via a spacer sequence; (xiii) a repositioned branching guanosine capable of initiating reverse transcription priming; (xiv) a staggered end sequence that reduces immunogenicity of the retron ncRNA, created by, e.g., adding or removing the 5ʹ a1 nucleotides and/or the 3ʹ a2 nucleotides; and/or, (xv) an antisense sequence complementary to a CRISPR/Cas guide RNA (gRNA) sequence encoded by the heterologous nucleic acid, wherein the antisense sequence hybridizes to and inhibits said gRNA in the encoded retron ncRNA, and wherein said antisense sequence is removed upon reverse transcription of the msDNA. 9. The engineered nucleic acid construct of any one of paragraphs 1-8, wherein the one or more heterologous nucleic acid sequences comprise: a) a heterologous nucleic acid (such as the coding sequence for an RNA aptamer or a ribozyme) inserted into the msr locus or the msd locus (such as in an S region (e.g., S1, S2, S3 and/or S4), or the tip of an L region (e.g., L1, L2, L3 and/or L4), or upstream or downstream of either the msr locus or the msd locus; or b) a first heterologous nucleic acid inserted into the msd locus, and a second heterologous nucleic acid inserted either upstream of the msr locus or downstream of the msd locus, wherein the second heterologous nucleic acid encodes a guide RNA. 10. The engineered nucleic acid construct of any one of paragraphs 1-9, wherein said heterologous nucleic acid encodes: (a) a protein or peptide of interest, or wherein said heterologous nucleic acid comprises; (b) a DNA donor template sequence; (c) a functional DNA element selected from a promoter, an enhancer, a protein binding sequence, a methylation site, a homology region for assisting gene editing, and the like; or (d) a coding sequence for a functional RNA element selected from a guide RNA and a ncRNA. 11. The engineered nucleic acid construct of paragraph 10, wherein said protein or peptide of interest comprises a therapeutic protein useful in treating a disease. 12. The engineered nucleic acid construct of paragraph 10, wherein said DNA donor template sequence corrects/repairs/removes a mutation at the target genome site. 13. The engineered nucleic acid construct of any one of paragraphs 1-12, further comprising or encoding a sequence-specific nuclease (such as a CRISPR/Cas effector enzyme, a ZFN, a TALEN, a meganuclease, TnpB, IscB, or a restriction endonuclease (RE)), and/or a DNA-repair modulating biomolecule. 13b. The engineered nucleic acid construct of paragraphs 1-13 wherein the engineered nucleic acid is an all-RNA component system. 13c. The engineered nucleic acid construct of paragraphs 1-13 wherein the engineered nucleic acid is an all-DNA molecule system. 14. The engineered nucleic acid construct of paragraph 13, wherein the sequence-specific nuclease is fused to the RT, optionally via a flexible linker (e.g., a flexible linker comprising Gly and Ser rich sequences such as G4S repeats or GS repeats) or by a generally disordered protein sequence (such as unstructured hydrophilic, biodegradable protein polymer, e.g., an XTEN peptide polymer). 15. The engineered nucleic acid construct of paragraph 13 or 14, wherein the nuclease is a CRISPR/Cas effector enzyme that forms a complex with a guide RNA (gRNA) recognizing a target sequence, wherein the gRNA is linked to the ncRNA and/or the msDNA, either directly or through a linker / spacer polynucleotide. 16. The engineered nucleic acid construct of paragraph 13, wherein the DNA-repair modulating biomolecule is a regulatory protein that modulates (e.g., enhances) HDR, and the regulatory protein is fused to the RT or to the sequence-specific nuclease, optionally via a flexible linker (e.g., the flexible linker comprising Gly and Ser rich sequences such as G4S repeats or GS repeats) or by a generally disordered protein sequence (such as unstructured hydrophilic, biodegradable protein polymer, e.g., an XTEN peptide polymer). 17. A vector system comprising one or more vectors comprising the engineered nucleic acid construct of any one of paragraphs 1-16, wherein the vector system is optionally all- RNA. 18. The vector system of paragraph 17, wherein the msr locus, the msd locus, and the polynucleotide encoding the RT are comprised within the same vector. 19. The vector system of paragraph 17 or 18, wherein the same vector further comprises a promoter operably linked to the msr locus and/or the msd locus. 20. The vector system of paragraph 19, wherein the promoter is further operably linked to the polynucleotide encoding the RT. 21. A vector system comprising one or more vectors, comprising the engineered nucleic acid construct of paragraph 1 or 2, wherein the vector system further comprises a second polynucleotide encoding a reverse transcriptase (RT), or a portion thereof, wherein the encoded RT is capable of synthesizing a DNA copy of at least a portion of the msd locus encoding the msDNA, and wherein the msr locus, the msd locus, and the second polynucleotide encoding the RT are provided by at least two different vectors. 22. The vector system of paragraph 21, wherein: a) the second polynucleotide comprises: i) a polynucleotide listed in Table A, or a polynucleotide having at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%%, at least 99.1%, at least 99.2%, at least 99.3%, at least 99.4%, at least 99.5%, at least 99.6%, at least 99.7%, at least 99.8% or at least 99.9% sequence identity to a polynucleotide listed in Table A; and/or b) the second polynucleotide encodes: i) a polypeptide listed in Table A, or a polypeptide having at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%%, at least 99.1%, at least 99.2%, at least 99.3%, at least 99.4%, at least 99.5%, at least 99.6%, at least 99.7%, at least 99.8% or at least 99.9% sequence identity to a polypeptide listed in Table A; and/or ii) a polypeptide listed in Table C, or a polypeptide having at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%%, at least 99.1%, at least 99.2%, at least 99.3%, at least 99.4%, at least 99.5%, at least 99.6%, at least 99.7%, at least 99.8% or at least 99.9% sequence identity to a polypeptide listed in Table C; and wherein the second polynucleotide optionally does not encode a polypeptide listed in Table X. 23. The vector system of paragraph 21 or 22, wherein the polynucleotide encoding the RT is provided in trans with respect to the msr gene and/or the msd gene. 24. The vector system of any one of paragraphs 17-23, wherein the one or more vectors comprise a viral vector. 25. The vector system of paragraph 24, wherein the viral vector is a retroviral vector, a lentiviral vector, an adenoviral vector, an adeno-associated viral vector, a vaccinia viral vector, a poxviral vector, or a herpes simplex viral vector. 26. The vector system of any one of paragraphs 17-23, wherein the one or more vectors comprise a non-viral vector. 27. The vector system of paragraph 26, wherein the non-viral vector comprises a plasmid. 28. The vector system of paragraph 26, wherein the non-viral vector comprises a liposome, a lipid nanoparticle (LNP), a cationic polymer, a vesicle, or a gold nanoparticle. 29. The vector system of any one of paragraphs 17-28, comprising a vector encoding a sequence-specific nuclease. 30. The vector system of paragraph 29, wherein the sequence-specific nuclease comprises an RNA-guided sequence-specific nuclease (e.g., a CRISPR/Cas effector enzyme, an engineered RNA-guided FokI-nuclease (e.g., dCas-FokI), an RNA-guided DNA endonuclease, TnpB, IscB, or a transposon-associated nuclease), or a non-RNA-guided sequence-specific nuclease (e.g., a meganuclease, a zinc finger nuclease (ZFN), a TALE nuclease (TALEN), or a restriction endonuclease (RE)). 31. The vector system of paragraph 30, wherein the Cas effector enzyme is a Class 1, Type I, II, or III Cas; a Class 2, Type II Cas (e.g., Cas9); or a Class 2, Type V Cas (e.g., Cpfl). 32. The vector system of paragraph 30, wherein: 1) the RNA-guided sequence-specific nuclease comprises the CRISPR/Cas effector enzyme, the engineered RNA-guided FokI-nuclease (e.g., dCas-FokI), the RNA- guided DNA endonuclease, TnpB, IscB, IsrB, or the transposon-associated nuclease; or, 2) non-RNA-guided sequence-specific nuclease comprises the meganuclease, the zinc finger nuclease (ZFN), the TALE nuclease (TALEN), or the restriction endonuclease (RE). 33. The vector system of any one of paragraphs 17-32, further comprising a vector encoding a homologous recombination enhancer protein. 34. An RNA molecule encoded by the engineered nucleic acid construct of any one of paragraphs 1-16. 35. An engineered nucleic acid-enzyme construct comprising: a) a non-coding RNA (ncRNA) comprising: i) an msr locus encoding the msr RNA portion of a multi-copy single- stranded DNA (msDNA); and ii) an msd locus encoding the msd RNA portion of the msDNA; b) a heterologous nucleic acid inserted at or within a location selected from: the msd locus, upstream of the msr locus, upstream of the msd locus, and downstream of the msd locus; and c) a sequence encoding a reverse transcriptase (RT), or a domain thereof comprising: i) a polypeptide listed in Table A, or a polypeptide having at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%%, at least 99.1%, at least 99.2%, at least 99.3%, at least 99.4%, at least 99.5%, at least 99.6%, at least 99.7%, at least 99.8% or at least 99.9% sequence identity to a polypeptide listed in Table A; and/or ii) a polypeptide listed in Table C, or a polypeptide having at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%%, at least 99.1%, at least 99.2%, at least 99.3%, at least 99.4%, at least 99.5%, at least 99.6%, at least 99.7%, at least 99.8% or at least 99.9% sequence identity to a polypeptide listed in Table C; and wherein, the RT optionally does not comprise a polypeptide listed in Table X. 36. An engineered nucleic acid-enzyme construct comprising: a) a non-coding RNA (ncRNA) comprising: i) an msr locus encoding the msr RNA portion of a multi-copy single- stranded DNA (msDNA); and ii) an msd locus encoding the msd RNA portion of the msDNA; wherein the ncRNA comprises: i) an ncRNA listed in Table B, or an ncRNA having at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%%, at least 99.1%, at least 99.2%, at least 99.3%, at least 99.4%, at least 99.5%, at least 99.6%, at least 99.7%, at least 99.8% or at least 99.9% sequence identity to an ncRNA listed in Table B; and/or ii) an ncRNA having a conserved structure of any one of the ncRNA structures of FIGs.2-27; and wherein the ncRNA optionally excludes any ncRNA associated in nature with any one of the retron reverse transcriptases of Table X; b) a heterologous nucleic acid inserted at or within a location selected from: the msd locus; upstream of the msr locus; upstream of the msd locus; and downstream of the msd locus; and c) a reverse transcriptase (RT), or a portion thereof, wherein the RT is capable of synthesizing a DNA copy of at least a portion of the msd locus encoding the msDNA. 37. An engineered nucleic acid-enzyme construct comprising: a) a non-coding RNA (ncRNA) comprising: i) an msr locus encoding the msr RNA portion of a multi-copy single- stranded DNA (msDNA); and ii) an msd locus encoding the msd RNA portion of the msDNA; wherein, the ncRNA comprises: i) an ncRNA listed in Table B, or an ncRNA having at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%%, at least 99.1%, at least 99.2%, at least 99.3%, at least 99.4%, at least 99.5%, at least 99.6%, at least 99.7%, at least 99.8% or at least 99.9% sequence identity to an ncRNA listed in Table B; and/or ii) an ncRNA having a conserved structure of any one of the ncRNA structures of FIGs.2-27; and wherein the ncRNA optionally excludes any ncRNA associated in nature with any one of the retron reverse transcriptases of Table X; b) a heterologous nucleic acid inserted at or within a location selected from: the msd locus, upstream of the msr locus, upstream of the msd locus, and downstream of the msd locus; and c) a reverse transcriptase (RT) or a domain thereof: wherein the RT comprises: i) an RT listed in Table A, or an RT having at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%%, at least 99.1%, at least 99.2%, at least 99.3%, at least 99.4%, at least 99.5%, at least 99.6%, at least 99.7%, at least 99.8% or at least 99.9% sequence identity to an RT listed in Table A; and/or ii) a consensus sequence listed in Table C, or a polypeptide having at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%%, at least 99.1%, at least 99.2%, at least 99.3%, at least 99.4%, at least 99.5%, at least 99.6%, at least 99.7%, at least 99.8% or at least 99.9% sequence identity to an amino acid sequence listed in Table C; and wherein the RT does not optionally comprise an RT listed in Table X. 38. An isolated host cell comprising the engineered nucleic acid construct of any one of paragraphs 1-16, the vector system of any of paragraphs 17-33, the RNA molecule of paragraph 34, or the engineered nucleic acid-enzyme construct of any one of paragraphs 35-37. 39. The isolated host cell of paragraph 38, wherein the host cell is a prokaryotic, archeon, or eukaryotic host cell. 40. The isolated host cell of paragraph 38, wherein the eukaryotic host cell is a mammalian host cell. 41. The isolated host cell of paragraph 39, wherein the eukaryotic host cell is a non-human host cell. 42. The isolated host cell of paragraph 40, wherein the mammalian host cell is a human host cell. 43. The isolated host cell of any one of paragraphs 38-42, wherein the host cell is an artificial cell or genetically modified cell. 44. A pharmaceutical composition comprising: a) the engineered nucleic acid construct of any one of paragraphs 1-16, ncRNA encoded by the engineered nucleic acid construct of any one of paragraphs 1-16, the vector system of any one of paragraphs 17-33, the RNA molecule of paragraph 34, the engineered nucleic acid-enzyme construct of any one of paragraphs 35-37, and/or the isolated host cell of any one of paragraphs 38-43; and b) a pharmaceutically acceptable carrier. 45. A pharmaceutical composition comprising: a) a lipid nanoparticle (LNP); and b) the engineered nucleic acid construct of any one of paragraphs 1-16, ncRNA encoded by the engineered nucleic acid construct of any one of paragraphs 1-16, the vector system of any one of paragraphs 17-33, the RNA molecule of paragraph 34, and/or the engineered nucleic acid-enzyme construct of any one of paragraphs 35-37. 46. The pharmaceutical composition of paragraph 45, wherein the LNP encapsulates the engineered nucleic acid construct, ncRNA, vector system, RNA molecule, and/or engineered nucleic acid-enzyme construct. 47. The pharmaceutical composition of paragraph 45 or 46, wherein the lipid nanoparticle comprises: a) one or more ionizable lipids; b) one or more structural lipids; c) one or more PEGylated lipids; and d) one or more phospholipids. 48. The pharmaceutical composition of paragraph 47, wherein the one or more ionizable lipids is selected from the group consisting of those disclosed in Table 2. 49. The pharmaceutical composition of paragraph 47 or 48, wherein the one or more structural lipids are selected from the group consisting of cholesterol, fecosterol, beta sitosterol, sitosterol, ergosterol, campesterol, stigmasterol, brassicasterol, tomatidine, tomatine, ursolic acid, alpha-tocopherol, prednisolone, dexamethasone, prednisone, and hydrocortisone. 50. The pharmaceutical composition of any one of paragraphs 47-49, wherein the one or more PEGylated lipids are selected from the group consisting of PEG-c-DOMG, PEG- DMG, PEG-DLPE, PEG-DMPE, PEG-DPPC, and PEG-DSPE. 51. The pharmaceutical composition of any one of paragraphs 47-50, wherein the one or more phospholipids are selected from the group consisting of 1,2-distearoyl-sn-glycero-3- phosphocholine (DSPC), 1,2-dioleoyl-sn-glycero-3-phosphoethanolamine (DOPE), 1,2- dilinoleoyl-sn-glycero-3-phosphocholine (DLPC), 1,2-dimyristoyl-sn-glycero- phosphocholine (DMPC), 1.2-dioleoyl-sn-glycero-3-phosphocholine (DOPC), 1,2- dipalmitoyl-sn-glycero-3-phosphocholine (DPPC), 1,2-diundecanoyl-sn-glycero- phosphocholine (DUPC), 1-palmitoyl-2-oleoyl-sn-glycero-3-phosphocho line (POPC), 1,2-di-O-octadecenyl-sn-glycero-3-phosphocholine (18:0 Diether PC), 1-oleoyl-2- cholesterylhemisuc cinoyl-sn-glycero-3-phosphocholine (OChemsPC), 1-hexadecyl-sn- glycero-3-phosphocholine (C16 Lyso PC), 1,2-dilinolenoyl-sn-glycero-3- phosphocholine, 1,2-diarachidonoyl-sn-glycero-3-phosphocholine, 1,2- didocosahexaenoyl-sn-glycero-3-phosphocholine, 1,2-diphytanoylsn-glycero-3- phosphoethanolamine (ME 16.0 PE), 1,2-distearoyl-sn-glycero-3-phosphoethanolamine, 1,2-dilinoleoyl-sn-glycero-3-phosphoethanolamine, 1,2-dilinolenoyl-sn-glycero-3- phosphoethanolamine, 1,2-diarachidonoyl-sn-glycero-3-phosphoethanolamine, 1,2- didocosahexaenoyl-sn-glycero-3-phosphoethanolamine, 1,2-dioleoyl-sn-glycero-3- phospho-rac-(1-glycerol) sodium salt (DOPG), and sphingomyelin. 52. The pharmaceutical composition of any one of paragraphs 47-51, wherein the lipid nanoparticle comprises about 48.5 mol % ionizable lipid, about 10 mol % phospholipid, about 40 mol % structural lipid, and about 1.5 mol% of PEG lipid. 53. The pharmaceutical composition of any one of paragraphs 47-52, wherein the lipid nanoparticle comprises about 48.5 mol % ionizable lipid, about 10 mol % phospholipid, about 39 mol % structural lipid, and about 2.5 mol% of PEG lipid. 54. The pharmaceutical composition of any one of paragraphs 47-53, wherein the LNP further comprises a targeting moiety operably connected to the LNP. 55. The pharmaceutical composition of any one of paragraphs 47-54, wherein the LNP further comprises one or more additional components selected from the group consisting of DDAB, EPC, 14PA, 18BMP, DODAP, DOTAP, and C12-200. 56. The pharmaceutical composition of paragraph 45, wherein the lipid nanoparticle comprises at least one cationic lipid selected from the group consisting of: a lipid in Table 2, a lipid having a structure of Formula (I), a lipid having a structure of Formula (II), a lipid having a structure of Formula (III), a lipid having a structure of Formula (IV), a lipid having a structure of Formula (V), a lipid having a structure of Formula (VI), and combinations thereof. 57. A kit comprising the engineered nucleic acid construct of any one of paragraphs 1-16, ncRNA encoded by the engineered nucleic acid construct of any one of paragraphs 1-16, the vector system of any one of paragraphs 17-33, the RNA molecule of paragraph 34, the engineered nucleic acid-enzyme construct of any one of paragraphs 35-37, the host cell of any one of paragraphs 38-43, or the pharmaceutical composition of any one of paragraphs 44-56, and instructions for genetically modifying a cell with said engineered nucleic acid construct, ncRNA, vector system, host cell, or pharmaceutical composition. 58. A method of modifying a target DNA sequence in a host (e.g., mammalian) cell, the method comprising introducing into the mammalian cell the engineered nucleic acid construct of any one of paragraphs 1-16, ncRNA encoded by the engineered nucleic acid construct of any one of paragraphs 1-16, the vector system of any one of paragraphs 17- 33, the RNA molecule of paragraph 34, the engineered nucleic acid-enzyme construct of any one of paragraphs 35-37, or the pharmaceutical composition of any one of paragraphs 44-56, to allow production of the msDNA in the host (e.g., mammalian) cell, wherein the heterologous nucleic acid in the msDNA is integrated into the genome of the host (e.g., mammalian) cell at the target DNA sequence by homology-dependent recombination. 59. The method of paragraph 58, wherein the modifying comprises introducing an insertion, deletion and/or substitution into the target DNA sequence. 60. A method of treating a disease or condition in a subject in need thereof, the method comprising administering a therapeutically effective amount of the engineered nucleic acid construct of any one of paragraphs 1-16, ncRNA encoded by the engineered nucleic acid construct of any one of paragraphs 1-16, the vector system of any one of paragraphs 17-33, the RNA molecule of paragraph 34, the engineered nucleic acid-enzyme construct of any one of paragraphs 35-37, the host cell of any one of paragraphs 38-43, or the pharmaceutical composition of any one of paragraphs 44-56 to the subject, thereby treating the disease or condition in the subject. 61. A method of treating a disease or condition in a subject in need thereof, the method comprising administering a therapeutically effective amount of the host cell of any one of paragraphs 38-43 to the subject, thereby treating the disease or condition in the subject. 62. The method of paragraph 61, wherein the host cell is autologous to the subject. 63. The method of paragraph 61, wherein the host cell is allogeneic to the subject. [0034] Additional exemplary and non-limiting aspects and embodiments of the disclosure are summarized as follows in the form of numbered paragraphs. 1. A gene editing system comprising one or more delivery vehicles, wherein: the delivery vehicle(s) comprise RNA cargo, said RNA cargo comprises (a) at least one mRNA molecule encoding (i) a nucleic acid programmable nuclease and (ii) a retron reverse transcriptase, (b) an engineered retron ncRNA, and (c) guide RNA for the programmable nuclease, each delivery vehicle contains (a)(i) and/or (a)(ii) and/or (b) and/or (c), whereby one delivery vehicle or more than one delivery vehicle delivers (a)(i), (a)(ii), (b), and (c). 2. The gene editing system of paragraph 1, wherein (a)(i) and (a)(ii) comprise a single mRNA molecule encoding the nucleic acid programmable nuclease and the retron reverse transcriptase. 3. The gene editing system of paragraph 2, wherein (a)(i) and (a)(ii) are encoded and expressed as a fusion protein. 4. The gene editing system of paragraph 3, wherein the fusion protein comprises the C- terminal end of the nucleic acid programmable nuclease fused to the N-terminal end of the retron reverse transcriptase (nuclease:RT fusion). 5. The gene editing system of paragraph 3, wherein the fusion protein comprises the N- terminal end of the nucleic acid programmable nuclease fused to the C-terminal end of the retron reverse transcriptase (RT:nuclease fusion). 6. The gene editing system of paragraph 1, wherein (a)(i) and (a)(ii) comprise a first mRNA molecule encoding the nucleic acid programmable nuclease and a second mRNA molecule encoding the retron reverse transcriptase. 7. The gene editing system of paragraph 1, wherein (c) is separate from (a)(i), (a)(ii) and (b) or is provided in trans. 8. The gene editing system of paragraph 1, wherein (b) the engineered retron ncRNA, and (c) the guide RNA are fused or are provided in cis. 9. The gene editing system of paragraph 8, wherein the guide RNA is fused to the 5’ end of the retron ncRNA. 10. The gene editing system of paragraph 8, wherein the guide RNA is fused to the 3’ end of the retron ncRNA. 11. The gene editing system of paragraph 8, wherein the engineered ncRNA comprises a first guide RNA fused to the 5’ end of the retron ncRNA, and a second guide RNA fused to the 3’ end of the retron ncRNA, and the first and second guide RNAs target different sequences. 12. The gene editing system of paragraph 1, wherein the one or more delivery vehicles comprise a liposome or a lipid nanoparticle (LNP). 13. The gene editing system of paragraph 1, wherein (a) the at least one mRNA molecule encoding (i) the nucleic acid programmable nuclease and (ii) the retron reverse transcriptase, and (b) the engineered retron ncRNA are in the same delivery vehicle. 14. The gene editing system of paragraph 1, wherein the (a) the at least one mRNA molecule encoding (i) the nucleic acid programmable nuclease and (ii) the retron reverse transcriptase, and (b) the engineered retron ncRNA are in separate delivery vehicles. 15. The gene editing system of paragraph 1, wherein the nucleic acid programmable nuclease and the retron reverse transcriptase are encoded on separate mRNA molecules and those separate mRNA molecules of (a)(i) and (a)(ii) are contained in the same delivery vehicle. 16. The gene editing system of paragraph 1, wherein the nucleic acid programmable nuclease and the retron reverse transcriptase are encoded on separate mRNA molecules and those separate mRNA molecules of (a)(i) and (a)(ii) are contained in different delivery vehicles. 17. The gene editing system of paragraph 1, wherein the engineered retron ncRNA includes a sequence of interest encoding a donor polynucleotide comprising an intended edit to be integrated at a target sequence in a cell, and wherein the donor polynucleotide is flanked by a 5' homology arm that hybridizes to a sequence 5’ to the target sequence and a 3' homology arm that hybridizes to a sequence 3' to the target sequence. 18. The gene editing system of paragraph 1, wherein the nucleic acid programmable nuclease comprises a Cas9 nuclease, a TnpB nuclease, or a Cas12a nuclease. 19. The gene editing system of paragraph 18, wherein the nucleic acid programmable nuclease comprises a Cas9 nuclease. 20. The gene editing system of paragraph 1, wherein the retron reverse transcriptase has an amino acid sequence of Table A, or an amino acid sequence having at least 90%, 95%, 99%, or 100% sequence identity with any amino acid sequence of Table A. 21. The gene editing system of paragraph 1, wherein the engineered retron ncRNA comprises: A) a pre-msr sequence having a first complementary region of the retron ncRNA; B) an msr sequence including an msr stem-loop structure; C) an msd sequence including an msd stem-loop structure and a sequence of interest, wherein said msd sequence templates a single strand DNA product (RT-DNA) in the presence of the retron reverse transcriptase; and D) a post-msd sequence having a second complementary region, wherein the first and second complementary regions form an a1/a2 duplex region of the retron ncRNA, wherein the msr stem-loop structure, the msd stem-loop structure, or the a1/a2 duplex comprise a modification which result in increased editing efficiency in the presence of a nucleic acid programmable nuclease that associates with the one or more guide RNAs, and wherein optionally one or more of the guide RNAs of (c) are coupled to the pre-msr sequence, the post-msd sequence, or both the pre-msr sequence and the post-msd sequence. 22. The gene editing system of paragraph 21, wherein the sequence of interest encodes a donor polynucleotide comprising an intended edit to be integrated at a target sequence of a cell, wherein the donor polynucleotide is flanked by a 5' homology arm that hybridizes to a sequence 5’ to the target sequence and a 3' homology arm that hybridizes to a sequence 3' to the target sequence. 23. The gene editing system of paragraph 21, wherein the ncRNA has a nucleotide sequence of any sequences of Table B, or a nucleotide sequence having at least 90%, 95%, 99%, or 100% sequence identity with any sequence of Table B. 24. An isolated cell comprising the gene editing system of paragraph 1. 25. The isolated cell of paragraph 24, wherein the isolated cell is a mammalian cell. 26. The isolated cell of paragraph 25, wherein the mammalian cell is a human cell. 27. A composition comprising: a) the gene editing system of paragraph 1; and b) a pharmaceutically or veterinarily acceptable carrier. 28. The composition of paragraph 27, wherein the delivery vehicle is a lipid nanoparticle comprising: a) one or more ionizable lipids; b) one or more structural lipids; c) one or more PEGylated lipids; and d) one or more phospholipids. 29. The composition of paragraph 28, wherein the one or more ionizable lipids comprises an ionizable lipid set forth in Table 2. 30. A method of genetically modifying a cell comprising: contacting the gene editing system of paragraph 17 with the cell, thereby delivering the RNA cargo to the cell, wherein: the nucleic acid programmable nuclease forms a complex with the guide RNA, wherein said guide RNA directs the complex to the target sequence, the nucleic acid programmable nuclease creates a double-stranded break in in the target sequence, the retron reverse transcriptase and engineered retron ncRNA create RT DNA that comprises the donor polynucleotide, and the donor polynucleotide becomes integrated at the target sequence. [0035] Additional exemplary and non-limiting aspects and embodiments of the disclosure are summarized as follows in the form of numbered paragraphs. 1. An engineered retron, comprising: a) an msr locus (that encodes the msr RNA portion of an msDNA); b) an msd locus encoding the msd RNA portion of the msDNA; c) a retron reverse transcriptase (RT), wherein said msd RNA is capable of being reverse transcribed to form the msDNA by the retron reverse transcriptase (RT); d) a heterologous nucleic acid inserted at or within the msd locus, upstream of the msr locus, upstream or downstream of the msd locus; and, e) a sequence modification (e.g., insertion, deletion, and/or substitution of one or more nucleotide(s)) in the msr locus and/or the msd locus that: i) modulates (e.g., enhances) reverse transcription, processivity, acuracy/fidelity, and/or production of the msDNA (e.g., in the mammalian cell); ii) modulates (e.g., reduces) immunogenicity of ncRNA encoded by the engineered retron (e.g., the msr locus and/or the msd locus) in a host (e.g., a host comprising the mammalian cell); and/or iii) comprises a nucleotide sequence that modulates (e.g., inhibits, either permanently or transiently) a function of the msDNA; and/or iv) modulates (e.g., improves) efficiency of targeted genome editing / engineering. 2. The engineered retron of paragraph 1, wherein said engineered retron is engineered based on and/or to resemble a secondary structure of a wild-type or consensus retron encoding a wild-type or consensus retron ncRNA encompassed by: 1) any one of the sequences and/or structures as depicted in SEQ ID NOs: 1-21 and/or FIGs.2-22; or 2) a variant of 1), having: A) up to 1, 2, or 3 (e.g., up to 1) nucleotide changes per 10 red lettered- nucleotides; B) up to 4, 5, or 6 (e.g., up to 1 or 2) nucleotide changes per 10 black lettered- nucleotides; and/or C) up to 7, 8, or 9 (e.g., up to 3 or 4) nucleotide changes per 10 grey lettered- nucleotides; and/or optionally further comprising: a) 7, 8, 9, or 10 (e.g., 9 or 10) nucleotides present per 10 red-circled nucleotides; b) 6, 7, 8, 9, or 10 (e.g., 8, 9 or 10) nucleotides present per 10 black-circled nucleotides; c) 4, 5, 6, 7, 8, 9, or 10 (e.g., 6, 7, 8, 9 or 10) nucleotides present per 10 grey- circled nucleotides; and/or d) 2, 3, 4, 5, 6, 7, 8, 9, or 10 (e.g., 4, 5, 6, 7, 8, 9, or 10) nucleotides present per 10 white-circled nucleotides. 3. The engineered retron of any one of paragraphs 1-2, wherein said engineered retron is engineered by introducing said sequence modification into a wild-type retron encoding a wild-type ncRNA, or into a synthetic/artificial retron encoding the consensus retron ncRNA. 4. The engineered retron of any one of paragraphs 1-3, wherein the sequence modification comprises, in the encoded retron ncRNA, one or more of: (i) a modified (e.g., mutated, reduced, or eliminated) bulge in a1, a2 or both a1 and a2; (ii) an extension or shortening of a1, a2 or both a1 and a2; (iii) an extension or shortening of a spacer sequence between hairpin loops (e.g., S1, S2, S3, S4, S5 and/or S6); (iv) a modified (e.g., mutated or eliminated) bulge in hairpin loops (e.g., L1, L2, L3, L4, L5 and/or L6 (e.g., by removing unpaired bases in the bulge, or by replacing unpaired bases with an equivalent number of base pairs)); (v) a modified (e.g., extended or shortened) length of hairpin loops (e.g., L1, L2, L3, L4, L5 and/or L6); (vi) an alternative L1 and/or L2 having complement, reverse, or reverse complement sequences; (vii) a modified (e.g., increased) number of unpaired bases at the tip of hairpin loops (e.g., L1, L2, L3, L4, L5 and/or L6); (viii) a modified (e.g., increased or decreased) GC content in hairpin loops (e.g., L1, L2, L3, L4, L5 and/or L6); (ix) an insertion of the heterologous nucleic acid in spacer sequences between hairpin loops (e.g., S1, S2, S3, S4, S5 and/or S6), or at the tip of hairpin loops (e.g., L1, L2, L3, L4, L5 and/or L6); (x) a deletion of hairpin loops (e.g., L1, L2, L3, L4, L5 and/or L6); (xi) an addition of a new loop in spacer sequences between hairpin loops (e.g., S1, S2, S3, S4, S5 and/or S6); (xii) circularization of the ncRNA with the 5’ end and the 3’ end of the ncRNA being connected either directly, or via a spacer sequence; (xiii) a repositioned branching guanosine capable of initiating reverse transcription priming; (xiv) a staggered end sequence that reduces immunogenicity of the retron ncRNA, created by, e.g., adding or removing the 5’ a1 nucleotides and/or the 3’ a2 nucleotides; and/or, (xv) an antisense sequence complementary to a CRISPR/Cas guide RNA (gRNA) sequence encoded by the heterologous nucleic acid, wherein the antisense sequence hybridizes to and inhibits said gRNA in the encoded retron ncRNA, and wherein said antisense sequence is removed upon reverse transcription of the msDNA. 5. The engineered retron of any one of paragraphs 1-4, comprising: (i) a heterologous nucleic acid (such as the coding sequence for an RNA aptamer or a ribozyme) inserted into the msr locus or the msd locus (such as in an S region (e.g., S1, S2, S3, S4, S5 and/or S6), or the tip of an L region (e.g., L1, L2, L3, L4, L5 and/or L6)), or upstream or downstream of either the msr locus or the msd locus; or, (ii) a first heterologous nucleic acid inserted into the msd locus, and a second heterologous nucleic acid inserted either upstream of the msr locus or downstream of the msd locus, wherein the second heterologous nucleic acid encodes a CRISPR/Cas guide RNA (gRNA). 6. The engineered retron of any one of paragraphs 1-5, wherein said retron reverse transcriptase (RT) is a cognate RT, a retron RT from a species within the same clade of the cognate RT, or a retron RT not within the same clade of the cognate RT (such as an unrelated RT or an engineered RT). 7. The engineered retron of any one of paragraphs 1-6, wherein said heterologous nucleic acid encodes a protein or peptide of interest, or wherein said heterologous nucleic acid comprises or encodes a donor / template sequence (e.g., a donor that corrects / repairs / removes a mutation at the target genome site, such as a mutated exon in a disease gene; a functional DNA element (such as a promoter, an enhancer, a protein binding sequence, a methylation site, a homology region for assiting gene editing, etc.); or a coding sequence for a functional RNA element (ncRNAs, etc)). 8. The engineered retron paragraph 7, wherein said protein or peptide of interest comprises a therapeutic protein (such as a wt protein defective in a disease cell, or a therapeutic antibody or antigen-binding fragment thereof) useful in treating a disease. 9. The engineered retron of paragraph 7, wherein said donor / template sequence corrects/repairs/removes a mutation at the target genome site (such as a mutated exon in a disease gene). 10. The engineered retron of any one of paragraphs 1-9, further comprising or encoding a sequence-specific nuclease (such as a CRISPR/Cas effector enzyme, a ZFN, a TALEN, a meganuclease, TnpB, IscB, or a restriction endonuclease (RE)), and/or a DNA-repair modulating biomolecule. 11. The engineered retron of paragraph 10, wherein the sequence-specific nuclease is fused to the RT, optionally via a flexible linker (e.g., a flexible linker comprising Gly and Ser rich sequences such as G4S repeats or GS repeats) or by a generally disordered protein sequence (such as unstructured hydrophilic, biodegradable protein polymer, e.g., an XTEN peptide polymer). 12. The engineered retron of paragraph 10 or 11, wherein the nuclease is a CRISPR/Cas effector enzyme that forms a complex with a guide RNA (gRNA) recognizing a target sequence, wherein the gRNA is linked to the ncRNA and/or the msDNA, either directly or through a linker / spacer polynucleotide. 13. The engineered retron of paragraph 10, wherein the DNA-repair modulating biomolecule is a regulatory protein that modulates (e.g., enhances) HDR, and the regulatory protein is fused to the RT or to the sequence-specific nuclease, optionally via a flexible linker (e.g., the flexible linker comprising Gly and Ser rich sequences such as G4S repeats or GS repeats) or by a generally disordered protein sequence (such as unstructured hydrophilic, biodegradable protein polymer, e.g., an XTEN peptide polymer). 14. A vector system comprising a vector comprising the engineered retron of any one of paragraphs 1-13. 15. The vector system of paragraph 14, wherein the msr locus, the msd locus, and the RT gene are provided by the same vector. 16. The vector system of paragraph 14 or 15, wherein the same vector comprises a promoter operably linked to the msr locus and/or the msd locus. 17. The vector system of any one of paragraphs 14-16, wherein the promoter is further operably linked to the RT gene. 18. The vector system of paragraph 14, wherein the msr locus, the msd locus, and the RT gene are provided by at least two different vectors. 19. The vector system of paragraph 14 or 18, wherein said RT gene is provided in trans with respect to the msr gene and/or the msd gene. 20. The vector system of any one of paragraphs 14-19, wherein the one or more vectors comprise a viral vector and/or a nonviral vector. 21. The vector system of paragraph 20, wherein the nonviral vector comprises a plasmid. 22. The vector system of any one of paragraphs 14-21, comprising a vector encoding a sequence-specific nuclease. 23. The vector system of paragraph 22, wherein the sequence-specific nuclease comprises an RNA-guided sequence-specific nuclease (e.g., a CRISPR/Cas effector enzyme, an engineered RNA-guided FokI-nuclease (e.g., dCas-FokI), an RNA-guided DNA endonuclease, TnpB, IscB, or a transposon-associated nuclease), or a non-RNA-guided sequence-specific nuclease (e.g., a meganuclease, a zinc finger nuclease (ZFN), a TALE nuclease (TALEN), or a restriction endonuclease (RE)). 24. The vector system of paragraph 23, wherein the Cas effector enzyme is a Class 1, Type I, II, or III Cas; a Class 2, Type II Cas (e.g., Cas9); or a Class 2, Type V Cas (e.g., Cpfl). 25. The vector system of paragraph 23, wherein: 1) the RNA-guided sequence-specific nuclease comprises the CRISPR/Cas effector enzyme, the engineered RNA-guided FokI-nuclease (e.g., dCas-FokI), the RNA- guided DNA endonuclease, TnpB, IscB, IsrB, or the transposon-associated nuclease; or, 2) non-RNA-guided sequence-specific nuclease comprises the meganuclease, the zinc finger nuclease (ZFN), the TALE nuclease (TALEN), or the restriction endonuclease (RE). 26. The vector system of any one of paragraphs 14-25, further comprising a vector encoding a homologous recombination enhancer protein. 27. An isolated host cell comprising the engineered retron of any one of paragraphs 1-13, or the vector system of any of paragraphs 14-26. 28. The host cell of paragraph 27, wherein the host cell is a prokaryotic, archeon, or eukaryotic host cell. 29. The host cell of paragraph 28, wherein the eukaryotic host cell is a mammalian host cell. 30. The host cell of paragraph 28, wherein the eukaryotic host cell is a non-human host cell. 31. The host cell of paragraph 29, wherein the mammalian host cell is a human host cell. 32. The host cell of any of paragraphs 28-31, wherein the host cell is an artificial cell or genetically modified cell. 33. A pharmaceutical composition comprising the engineered retron of any one of paragraphs 1-13. 34. A delivery vehicle comprising the engineered retron of any one of paragraphs 1-13 or ncRNA encoded by the engineered retron of any one of paragraphs 1-13, the vector of any one of paragraphs 14-26, the host cell of any one of paragraphs 27-32, or the pharmaceutical composition of paragraph 33. 35. The delivery vehicle of paragraph 34, which is a lipid nanoparticle. 36. The delivery vehicle of paragraph 35, wherein the lipid nanoparticle comprises at least one cationic lipid selected from the group consists of: a lipid in Table (I), a lipid having a structure of Formula (I), a lipid having a structure of Formula (II), a lipid having a structure of Formula (III), a lipid having a structure of Formula (IV), a lipid having a structure of Formula (V), a lipid having a structure of Formula (VI), and combinations thereof. 37. A kit comprising the engineered retron of any one of paragraphs 1-13 or ncRNA encoded by the engineered retron of any one of paragraphs 1-13, the vector or vector system of any one of claims 14-26, the host cell of any one of claims 27-32, the pharmaceutical composition of claim 33, or the delivery vehicle of any one of claims 34-36, and instructions for genetically modifying a cell with said engineered retron, ncRNA, vector / vector system, host cell, pharmaceutical composition, or delivery vehicle. 38. A method of modifying a target DNA sequence in a host (e.g., mammalian) cell, the method comprising introducing into the mammalian cell the engineered retron of any one of claims 1-13 or ncRNA encoded by the engineered retron of any one of claims 1-13, the vector or vector system of any one of claims 14-26, the pharmaceutical composition of claim 33, or the delivery vehicle of any one of claims 34-36, to allow the production of the msDNA in the host (e.g., mammalian) cell, wherein the encoded heterologous nucleic acid in the msDNA is integrated into the genome of the host (e.g., mammalian) cell at the target DNA sequence. 39. Use of the engineered retron of any one of claims 1-13 or ncRNA encoded by the engineered retron of any one of claims 1-13, the vector or vector system of any one of claims 14-26, the pharmaceutical composition of claim 33, or the delivery vehicle of any one of claims 34-36, to effect modification of a target DNA sequence in a host (e.g., mammalian) cell. [0036] Additional exemplary and non-limiting aspects and embodiments of the disclosure are summarized as follows in the form of numbered paragraphs. 1. An engineered retron coding for a retron reverse transcriptase (rRT) and a retron DNA/RNA hybrid (msRNA), said engineered retron comprising: a) a msr locus comprising a sequence encoding msr RNA of the msDNA, b) a msd locus comprising a sequence encoding msd RNA corresponding to DNA of the msDNA, wherein a non-coding retron RNA (ncRNA) comprises the msr RNA and the msd RNA transcribed from the msr locus and the msd locus, c) a ret locus comprising a sequence encoding the rRT, wherein the rRT processes ncRNA into the msDNA, wherein the engineered retron is engineered by further comprising a sequence modification comprising: (i) a nucleic acid heterologous to the retron of the rRT and to the retron of the msRNA inserted at or within the msd locus, or upstream of the msr locus, or upstream or downstream of the msd locus; and/or, (ii) an insertion, deletion, and/or substitution of one or more nucleotides in the msr locus and/or the msd locus that: in a mammalian cell enhances reverse transcription, processivity, accuracy or fidelity, or enhances production of the msDNA; or in a mammalian cell or mammalian host reduces immunogenicity of ncRNA; or comprises a nucleotide sequence that inhibits a function of the msDNA; or improves targeted genome editing / engineering of a gene editing system comprising the engineered retron. 2. The engineered retron of paragraph 1, wherein: a) the engineered retron is engineered from a type IA/IIA1 retron, the ncRNA of which has a secondary structure of comprising a consensus secondary structure illustrated in FIG.2 and comprising a1-S1-L1-L2-S2-L3-S3-a2-S4-L4, wherein: a1/a2 is a stem 8 bp in length; L1 is a stem of 5 bps with a 10-nt tip; L2 is a stem of 7 bps with a 5-nt tip, and a 1/1 bulge 3 nt from the tip; L3 is a stem of 23 bps with a 22-nt tip, and a 2/2 bulge 21 bps from the tip; L4 is a stem of 11 bps with a 5-nt tip; S1 is a single-stranded spacer region between the a1/a2 stem and L1, with no spacer between L1 and L2; S2 is a single-stranded spacer region between L2 and L3; S3 is a single-stranded spacer region between L3 and the a1/a2 stem; and S4 is a single-stranded spacer region between the a1/a2 stem and L4, and conserved nucleotides are as shown in SEQ ID NO: 1 or FIG.2; or b) the engineered retron is engineered from a type IB1 retron, the ncRNA of which has a secondary structure comprising a consensus secondary structure illustrated in FIG. 3 and comprising a1-S1-L1-S2-L2-S3-a2, wherein: a1/a2 is a stem 6 bps in length with a 2/2 bulge 3 bps from the tip, wherein a1 has a 2-nt overhang and a2 has a 6-nt overhang; L1 is a stem of 14 bps with a 3-nt tip, a 1/0 bulge 4 bps from the tip, and a 0/6 bulge 10 bps from the tip; L2 is a stem of 23 bps with a 5-nt tip, a 1/1 bulge 4 bps from the tip, and a 0/1 bulge 18 bps from the tip; S1 is a single-stranded spacer region between a1/a2 and L1; S2 is a single-stranded spacer region between L1 and L2; S3 is a single-stranded spacer region between L2 and a1/a2; and conserved nucleotides are as shown in FIG.3; or c) the engineered retron is engineered from a type IB2 retron, the ncRNA of which has a secondary structure comprising a consensus secondary structure illustrated in FIG. 4 and comprising a1-S1-L1-L2-S2-L3-S3-L4-S4-a2, wherein: the a1/a2 stem is 16 bp in length, with a 17-base 5ʹ overhang and a 16-base 3ʹ overhang; L1 is a stem of 6 bps with a 4-nt tip; L2 is a stem of 4 bps with a 4-nt tip, with a 2/2 bulge 2 nts from the tip; L3 is a stem of 3 bps with a 5-nt tip; L4 is a stem of 9 bps with a 5-nt tip, and a 1/1 bulge 4 nts from the tip; S1, S2, S3, and S4 are single-stranded spacer regions between the a1/a2 stem and L1, L2 and L3, L3 and L4, and L4 and the a1/a2 stem, respectively, with no spacer between L1 and L2; wherein the last 5 nts of S1 and the 5th-9th nts of S2 form a 5-bp stem, and, conserved nucleotides are as shown in FIG.4; or d) the engineered retron is engineered from a type 1C retron, the ncRNA of which has a secondary structure comprising a consensus secondary structure illustrated in FIG. 5 and comprising a1-S1-L1-S2-L2-S3-a2, wherein: a1/a2 is a stem 13 bps in length; L1 is a stem of 9 bps with a 3-nt tip; L2 is a stem of 10 bps with a 5-nt tip; S1 is a single-stranded spacer region between a1/a2 and L1; S2 is a single-stranded spacer region between L1 and L2; S3 is a single-stranded spacer region between L2 and a1/a2; and conserved nucleotides are as shown in FIG.5; or e) the engineered retron is engineered from a type IIA1 retron, the ncRNA of which has a secondary structure comprising a consensus secondary structure illustrated in FIG. 6 and comprising a1-S1-L1-S2-L2-S3-L3-S4-a2, wherein: a1/a2 is a stem 10 bps in length with a 1-nt overhang on a2; L1 is a stem of 10 bps with a 3-nt tip; L2 is a stem of 7 bps with a 5-nt tip; L3 is a stem of 27 bps with a 8-nt tip and a 0/2 bulge 26 bps from the tip; S1 is a single-stranded spacer region between a1/a2 and L1; S2 is a single-stranded spacer region between L1 and L2; S3 is a single-stranded spacer region between L2 and L3; S4 is a single-stranded spacer region between L3 and a1/a2; and conserved nucleotides are as shown in FIG.6; or f) the engineered retron is engineered from a type IIA2 retron, the ncRNA of which has a secondary structure comprising a consensus secondary structure illustrated in FIG. 7 and comprising a1-S1-L1-S2-L2-L3-S3-a2, wherein: a1/a2 is a stem 7 bp in length with no overhangs; L1 is a stem of 8 bps with a 3-nt tip; L2 is a stem of 30 bps with a 8-nt tip, a 1/1 bulge 2 bps from the tip, and a 1/1 bulge 27 bps from the tip; L3 is a stem of 8 bps with a 5-nt tip, and a 0/1 bulge 3 nt from the tip; S1 is a single-stranded spacer region between the a1/a2 stem and L1; S2 is a single-stranded spacer region between L1 and L2; S3 is a single-stranded spacer region between L3 and the a1/a2 stem; and conserved nucleotides are as shown in FIG.7; or g) the engineered retron is engineered from a type IIA3 retron, the ncRNA of which has a secondary structure comprising a consensus secondary structure illustrated in FIG. 8 and comprising a1-S1-L1-L2-S2-L3-S3-a2, wherein: a1/a2 is a stem 6 bps in length; L1 is a stem of 8 bps with a 9-nt tip; L2 is a stem of 8 bps with a 3-nt tip; S1 is a single-stranded spacer region between a1/a2 and L1; S2 is a single-stranded spacer region between L2 and L3; S3 is a single-stranded spacer region between L3 and a1/a2; and conserved nucleotides are as shown in FIG.8 h) the engineered retron is engineered from a type IIA4 retron, the ncRNA of which has a secondary structure comprising a consensus secondary structure illustrated in FIG. 9 and comprising a1-a2-L1-S1-L2-L3-S2-L4-S3, wherein: a1/a2 is a stem 3 bp in length with no overhangs and a 7-nt tip; L1 is a stem of 7 bps with a 3-nt tip; L2 is a stem of 6 bps with a 4-nt tip; L3 is a stem of 40 bps with a 5-nt tip, and a 2/2 bulge 3 bps from the tip, a 5/4 bulge 10 bps from the tip, and a 12/15 bulge 30 bps from the tip; L4 is a stem of 4 bps with a 9-nt tip; S1 is a single-stranded spacer region between L1 and L2/L3; S2 is a single-stranded spacer region between L2/L3 and L4; S3 is a single-stranded spacer region between L4 and the 3ʹ end of the ncRNA; and conserved nucleotides are as shown in FIG.9; or i) the engineered retron is engineered from a type IIA5 retron, the ncRNA of which has a secondary structure comprising a consensus secondary structure illustrated in FIG. 10 and comprising a1-S1-L1-S2-L2-S3-L3-S4-a2, wherein: a1/a2 is a stem 15 bps in length with a 1-nt overhang on a1, a 13-nt overhang on a2, and a 7/5 bulge 13-nt from the tip; L1 is a stem of 10 bps with a 3-nt tip; L2 is a stem of 35 bps with a 3-nt tip; L3 is a stem of 6 bps with a 5-nt tip; S1 is a single-stranded spacer region between a1/a2 and L1; S2 is a single-stranded spacer region between L1 and L2; S3 is a single-stranded spacer region between L2 and L3; S4 is a single-stranded spacer region between L3 and a1/a2; and conserved nucleotides are as shown in FIG.10; or j) the engineered retron is engineered from a type IIIA1 retron, the ncRNA of which has a secondary structure comprising a consensus secondary structure illustrated in FIG. 11 and comprising a1-S1-L1-L2-S2-L3-S3-a2, wherein: a1/a2 is a stem 2 bps in length with a 1-nt overhang on a2; L1 is a stem of 8 bps with a 4-nt tip; L2 is a stem of 9 bps with a 3-nt tip and a 1/1 bulge 3 bps from the tip; L3 is a stem of 20 bps with a 3-nt tip and a 1/2 bulge 3 bps from the tip; S1 is a single-stranded spacer region between a1/a2 and L1; S2 is a single-stranded spacer region between L2 and L3; S3 is a single-stranded spacer region between L3 and a1/a2; and conserved nucleotides are as shown in FIG.11; or k) the engineered retron is engineered from a type IIIA2 retron, the ncRNA of which has a secondary structure comprising a consensus secondary structure illustrated in FIG. 12 and comprising a1-S1-L1-S2-L2-S3-L3-S4-L4-S5-a2, wherein: a1/a2 is a stem 15 bp in length; L1 is a stem of 6 bps with a 4-nt tip; L2 is a stem of 13 bps with a 5-nt tip; L3 is a stem of 4 bps with a 8-nt tip; L4 is a stem of 20 bps with a 4-nt tip and a 2/2 bulge 6 bp from the tip; S1 is a single-stranded spacer region between a1/a2 and L1; S2 is a single-stranded spacer region between L1 and L2; S3 is a single-stranded spacer region between L2 and L3; S4 is a single-stranded spacer region between L3 and L4; S5 is a single-stranded spacer region between L4 and a1/a2; and conserved nucleotides are as shown in FIG.12; or l) the engineered retron is engineered from a type IIIA3 retron, the ncRNA of which has a secondary structure comprising a consensus secondary structure illustrated in FIG. 13 and comprising a1-S1-L1-S2-L2-S3-L3-S4-L4-L5-L6-S5-a2, wherein: a1/a2 is a stem 24 bps in length and having a 1/0 bulge 15 bps from the tip, and a 1/1 bulge 19 bps from the tip; L1 is a stem of 7 bps with a 4-nt tip; L2 is a stem of 9 bps with a 8-nt tip; L3 is a stem of 8 bps with a 4-nt tip; L4 is a stem of 4 bps with a 9-nt tip, and a 2/2 bulge 3 bps from the tip; L5 is a stem of 19 bps with a 18-nt tip; L6 is a stem of 5 bps with a 3-nt tip; S1 is a single-stranded spacer region between a1/a2 and L1; S2 is a single-stranded spacer region between L1 and L2; S3 is a single-stranded spacer region between L2 and L3; S4 is a single-stranded spacer region between L3 and L4; S5 is a single-stranded spacer region between L6 and a1/a2; and conserved nucleotides are as shown in FIG.13; or m) the engineered retron is engineered from a type IIIA4 retron, the ncRNA of which has a secondary structure comprising a consensus secondary structure illustrated in FIG. 14 and comprising a1-S1-L1-S2-L2-S3-L3-S4-a2, wherein: a1/a2 is a stem 5 bps in length with a 1/2 bulge 2 bps from the tip; L1 is a stem of 8 bps with a 6-nt tip; L2 is a stem of 8 bps with a 5-nt tip; L3 is a stem of 13 bps with a 14-nt tip and a 1/0 bulge 2 bps from the tip; S1 is a single-stranded spacer region between a1/a2 and L1; S2 is a single-stranded spacer region between L1 and L2; S3 is a single-stranded spacer region between L2 and L3: S4 is a single-stranded spacer region between L3 and a1/a2; and conserved nucleotides are as shown in SEQ ID NO: 19 or FIG.20 or FIG.14; or n) the engineered retron is engineered from a type IIIA5 retron, the ncRNA of which has a secondary structure comprising a consensus secondary structure illustrated in FIG. 15 and comprising a1-S1-L1-S2-L2-L3-L4-S3-a2, wherein: the a1/a2 stem is 11 bp in length with no overhang; L1 is a stem of 9 bps with a 3-nt tip; L2 is a stem of 14 bps with a 5-nt tip; L3 is a stem of 9 bps with a 7-nt tip; L4 is a stem of 15 bps with a 7-nt tip; S1, S2, and S3 are single-stranded spacer regions between the a1/a2 stem and L1, L2 and L3, and L4 and the a1/a2 stem, respectively, with no spacer between L1 and L2, and no spacer between L2 and L3; wherein the 5th-2nd last nts of S2 and the 3rd-6th nts of S3 forms a 4-bp stem, and, conserved nucleotides are as shown in FIG.15; or o) the engineered retron is engineered from a type IIIunk retron, the ncRNA of which has a secondary structure comprising a consensus secondary structure illustrated in FIG.16 and comprising a1-S1-L1-S2-L2-S3-L3-S4-a2, wherein: a1/a2 is a stem 11 bps in length; L1 is a stem of 12 bps with a 2-nt tip; L2 is a stem of 21 bps with a 1-nt tip; L3 is a stem of 20 bps with a 4-nt tip; S1 is a single-stranded spacer region between a1/a2 and L1; S2 is a single-stranded spacer region between L1 and L2; S3 is a single-stranded spacer region between L2 and L3; S4 is a single-stranded spacer region between L3 and a1/a2; and conserved nucleotides are as shown in FIG.16; or p) the engineered retron is engineered from a type IV retron, the ncRNA of which has a secondary structure comprising a consensus secondary structure illustrated in FIG. 17 and comprising a1-S1-L1-L2-S2-L3-S3-a2, wherein: a1/a2 is a stem 9 bp in length with no overhang; L1 is a stem of 5 bps with a 6-nt tip; L2 is a stem of 9 bps with a 4-nt tip; L3 is a stem of 26 bps with a 5-nt tip, a 0/1 bulge 7 bps from the tip, and a 0/1 bulge 9 bps from the tip; S1 is a single-stranded spacer region between the a1/a2 stem and L1, with no spacer region between L1 and L2; S2 is a single-stranded spacer region between L2 and L3; S3 is a single-stranded spacer region between L3 and the a1/a2 stem; and conserved nucleotides are as shown in FIG.17; or q) the engineered retron is engineered from a type IX retron, the ncRNA of which has a secondary structure comprising a consensus secondary structure illustrated in FIG. 18 and comprising a1-L1-S1-L2-S2-a2, wherein: a1/a2 is a stem 12 bp in length, wherein a1 has a 14-nt overhang and a2 has a 2-nt overhang; L1 is a stem of 11 bps with a 3-nt tip and a 1/3 bulge 7 bp from the tip; L2 is a stem of 25 bps with a 7-nt tip; S1 is a single-stranded spacer region between L1 and L2; S2 is a single-stranded spacer region between L2 and a1/a2; and conserved nucleotides are as shown in FIG.18; or r) the engineered retron is engineered from a type V retron, the ncRNA of which has a secondary structure comprising a consensus secondary structure illustrated in FIG.19 and comprising a1-S1-L1-S2-L2-S3-a2, wherein: a1/a2 is a stem 13 bps in length; L1 is a stem of 20 bps with a 4-nt tip and a 6/4 bulge 6 bps from the tip; L2 is a stem of 14 bps with a 4-nt tip and a 1/0 bulge 5 bps from the tip; S1 is a single-stranded spacer region between a1/a2 and L1; S2 is a single-stranded spacer region between L1 and L2; S3 is a single-stranded spacer region between L2 and a1/a2; and conserved nucleotides are as shown in FIG.19; or s) the engineered retron is engineered from a type VI retron, the ncRNA of which has a secondary structure comprising a consensus secondary structure illustrated in FIG. 20 and comprising a1-S1-L1-S2-L2-S3-L3-L4-S4-a2, wherein: a1/a2 is a stem 4 bp in length with a 1 bp 5ʹ overhang; L1 is a stem of 7 bps with a 4-nt tip; L2 is a stem of 8 bps with a 4-nt tip; L3 is a stem of 16 bps with a 6-nt tip, a 3/4 bulge 3 bps from the tip, a 2/3 bulge 5 bps from the tip, and a 3/1 bulge 8 bps from the tip; S1 is a single-stranded spacer region between the a1/a2 stem and L1; S2 is a single-stranded spacer region between L1 and L2; S3 is a single-stranded spacer region between L2 and L3, with no spacer region between L3 and L4; S4 is a single-stranded spacer region between L4 and the a1/a2 stem; and conserved nucleotides are as shown in FIG.20; or t) the engineered retron is engineered from a type XI Group 1 retron, the ncRNA of which has a secondary structure comprising a consensus secondary structure illustrated in FIG.21 and comprising a1-S1-L1-S2-L2-S3-a2, wherein: a1/a2 is a stem 16 bps in length with a 5-nt overhang on a1, and a 3-nt overhang on a2; L1 is a stem of 9 bps with a 3-nt tip; L2 is a stem of 7 bps with a 13-nt tip; S1 is a single-stranded spacer region between a1/a2 and L1; S2 is a single-stranded spacer region between L1 and L2; S3 is a single-stranded spacer region between L2 and a1/a2; and conserved nucleotides are as shown in FIG.21; or u) the engineered retron is engineered from a type XI Group 2 retron, the ncRNA of which has a secondary structure comprising a consensus secondary structure illustrated in FIG.22 and comprising a1-S1-L1-S2-L2-S3-a2, wherein: a1/a2 is a stem 13 bps in length with a 1-nt overhang on a2; L1 is a stem of 7 bps with a 3-nt tip and a 2/2 bulge 1 bp from the tip; L2 is a stem of 8 bps with a 20-nt tip; S1 is a single-stranded spacer region between a1/a2 and L1; S2 is a single-stranded spacer region between L1 and L2; S3 is a single-stranded spacer region between L2 and a1/a2; and conserved nucleotides are as shown in FIG.22; or v) the engineered retron is engineered from a type XII retron, the ncRNA of which has a secondary structure comprising a consensus secondary structure illustrated in FIG. 23 and comprising a1-S1-L1-S2-L2-S3-a2, wherein: a1/a2 is a stem 13 bps in length with a 1-nt overhang on a2; L1 is a stem of 7 bps with a 3-nt tip, and a 2/2 bulge 1 bp from the tip; L2 is a stem of 8 bps with a 19-nt tip; S1 is a single-stranded spacer region between a1/a2 and L1; S2 is a single-stranded spacer region between L1 and L2; S3 is a single-stranded spacer region between L2 and a1/a2; and conserved nucleotides are as shown in FIG.23; or w) the engineered retron is engineered from a type XIII retron, the ncRNA of which has a secondary structure comprising a consensus secondary structure illustrated in FIG. 24 and comprising a1-S1-L1-S2-L2-L3-S3-a2, wherein: a1/a2 is a stem 7 bp in length with no overhangs; L1 is a stem of 8 bps with a 3-nt tip; L2 is a stem of 30 bps with a 8-nt tip, a 1/1 bulge 2 bps from the tip, and a 1/1 bulge 27 bps from the tip; L3 is a stem of 8 bps with a 5-nt tip, and a 0/1 bulge 3 nt from the tip; S1 is a single-stranded spacer region between the a1/a2 stem and L1; S2 is a single-stranded spacer region between L1 and L2; and, S3 is a single-stranded spacer region between L3 and the a1/a2 stem; and conserved nucleotides are as shown in FIG.24; or x) the engineered retron is engineered from a type XIV retron, the ncRNA of which has a secondary structure comprising a consensus secondary structure illustrated in FIG. 25 and comprising a1-S1-L1-L2-S2-L3-S3-a2, wherein: the a1/a2 stem is 15 bp in length with no overhang, and a 4/2 bulge 7 bps from the 5ʹ end of a1; L1 is a stem of 8 bps with a 5-nt tip; L2 is a stem of 7 bps with a 5-nt tip; L3 is a stem of 13 bps with a 2-nt tip, a 5/9 bulge 5 bps from the tip, and a 5/5 bulge 8 bps from the tip; S1, S2, and S3 are single-stranded spacer regions between the a1/a2 stem and L1, L2 and L3, and L3 and the a1/a2 stem, respectively, with no spacer between L1 and L2; and wherein the 5th-3rd last nts of S1 and the 2nd-5th 4 nts of S2 forms a 3-bp stem; and conserved nucleotides are as shown in FIG.25; or y) the engineered retron is engineered from an Ec107-like retron, the ncRNA of which has a secondary structure comprising a consensus secondary structure illustrated in FIG.26 and comprising a1-S1-L1-S2-L2-S3-L3-S4-a2, wherein: a1/a2 is a stem 12 bp in length; L1 is a stem of 4 bps with a 8-nt tip; L2 is a stem of 8 bps with a 3-nt tip; L3 is a stem of 22 bps with a 3-nt tip, a 4/6 bulge 6 bp from the tip, a 3/3 bulge 13 bp from the tip, and a 1/1 bulge 18 bp from the tip; S1 is a single-stranded spacer region between a1/a2 and L1; S2 is a single-stranded spacer region between L1 and L2; S3 is a single-stranded spacer region between L2 and L3; S4 is a single-stranded spacer region between L3 and a1/a2; and conserved nucleotides are as shown in FIG.26; or z) the engineered retron is engineered from an Outgroup A retron, the ncRNA of which has a secondary structure comprising a consensus secondary structure illustrated in FIG.27 and comprising a1-S1-L1-S2-L2-S3-L3-S4-a2, wherein: a1/a2 is a stem 5 bps in length with a 2-nt overhang on a2; L1 is a stem of 11 bps with a 4-nt tip; L2 is a stem of 8 bps with a 3-nt tip; L3 is a stem of 18 bps with a 3-nt tip; S1 is a single-stranded spacer region between a1/a2 and L1; S2 is a single-stranded spacer region between L1 and L2; S3 is a single-stranded spacer region between L2 and L3; S4 is a single-stranded spacer region between L3 and a1/a2; and conserved nucleotides are as shown in FIG.27; or aa) the engineered retron is engineered from any retron set out in Table X; or bb) the ret locus encodes a rRT as set forth in Table A; or cc) the rRT comprises a consensus amino acid sequence of Table C; or dd) the ncRNA comprises a sequence set forth in Table B; or ee) the rRT and the ncRNA are cognate; or ff) the rRT and the ncRNA are not cognate; or gg) the engineered retron is engineered from any one of the sequences set forth in any one of SEQ ID NOs: 1-21. 3. The engineered retron of any one of paragraphs 1-2, wherein said engineered retron is engineered by introducing said sequence modification into a wild-type retron encoding a wild-type ncRNA, or into a synthetic/artificial retron encoding consensus ncRNA. 4. The engineered retron of any one of paragraphs 2-3, wherein the sequence modification comprises, in the ncRNA, one or more of: (i) a mutated, reduced, or eliminated bulge in a1, a2 or both a1 and a2; (ii) an extension or shortening of a1, a2 or both a1 and a2; (iii) an extension or shortening of a spacer sequence between hairpin loops S1, S2, S3, S4, S5 and/or S6; (iv) a mutated or eliminated bulge in hairpin loops L1, L2, L3, L4, L5 and/or L6 from removal of unpaired bases in the bulge, or by replacing unpaired bases with an equivalent number of base pairs; or (v) extended or shortened length of hairpin loops L1, L2, L3, L4, L5 and/or L6; (vi) an alternative L1 and/or L2 having complement, reverse, or reverse complement sequences; (vii) an increased number of unpaired bases at the tip of hairpin loops L1, L2, L3, L4, L5 and/or L6; (viii) an increased or decreased GC content in hairpin loops L1, L2, L3, L4, L5 and/or L6; (ix) an insertion of the heterologous nucleic acid in spacer sequences between hairpin loops S1, S2, S3, S4, S5 and/or S6, or at the tip of hairpin loops L1, L2, L3, L4, L5 and/or L6; (x) a deletion of hairpin loop(s) L1, L2, L3, L4, L5 and/or L6; (xi) an addition of a new loop in spacer sequences between hairpin loops S1, S2, S3, S4, S5 and/or S6; (xii) circularization of the ncRNA with the 5’ end and the 3’ end of the ncRNA being connected either directly, or via a spacer sequence; (xiii) a repositioned branching guanosine capable of initiating reverse transcription priming; (xiv) a staggered end sequence that reduces immunogenicity of the ncRNA, created byadding or removing the 5’ a1 nucleotides and/or the 3’ a2 nucleotides; and/or, (xv) insertion of the heterologous nucleic acid and an antisense sequence complementary to a CRISPR-Cas guide RNA (gRNA) sequence encoded by the heterologous nucleic acid, wherein the antisense sequence hybridizes to and inhibits said gRNA in encoded ncRNA, and wherein said antisense sequence is removed upon reverse transcription resulting in the msDNA. 5. The engineered retron of any one of paragraphs 1-4, comprising: (i) a heterologous to the retron of the rRT and to the retron of the msRNA nucleic acid inserted into the msr locus or the msd locus in S1, S2, S3, S4, S5 and/or S6, or the tip of an L1, L2, L3, L4, L5 and/or L6, or upstream or downstream of either the msr locus or the msd locus; or, (ii) a first heterologous to the retron of the rRT and to the retron of the msRNA nucleic acid inserted into the msd locus, and a second heterologous to the retron of the rRT and to the retron of the msRNA nucleic acid inserted either upstream of the msr locus or downstream of the msd locus, wherein the second heterologous nucleic acid encodes a CRISPR-Cas guide RNA (gRNA). 6. The engineered retron of any one of paragraphs 1-5, wherein the rRT and the ncRNA are cognate. 7. The engineered retron of any one of paragraphs 1-5, wherein the rRT and the ncRNA are not cognate. 8. The engineered retron of any one of paragraphs 1-7, wherein said heterologous nucleic acid encodes a sequence for an RNA aptamer or a ribozyme, or a protein or peptide of interest, or wherein said heterologous nucleic acid comprises or encodes a donor sequence. 9. The engineered retron of paragraph 8 wherein the donor sequence that corrects or repairs or removes a mutation at a target genome site, or repairs or removes a mutation in an exon in a disease gene; or provides a functional DNA element comprising a promoter, an enhancer, a protein binding sequence, a methylation site, or a homology region for gene editing. 10. The engineered retron paragraph 8, wherein said protein or peptide of interest comprises a therapeutic protein. 11. The engineered protein of paragraph 10, wherein the therapeutic protein comprises a wild type protein that is expressed in a defective manner in a disease cell, or a therapeutic antibody or an antigen-binding fragment thereof. 12. A system comprising: a) a retron, or the rRT and the ncRNA of the retron, or RNA encoding the rRT and the ncRNA of the retron, wherein the retron comprises: i) an engineered retron of any one of paragraphs 1-11, or ii) a retron set forth in Table X or a retron having a sequence having at least 85%, or 90%, or 95% or 99% sequence identity with the sequence of a retron set forth in Table X, or iii) a retron comprising a rRT sequence set forth in Table A, or a sequence having at least 85%, or 90%, or 95% or 99% sequence identity with a rRT sequence set forth in Table A, or iv) a retron encoding an ncRNA comprising a sequence set forth in Table B, or a sequence having at least 85%, or 90%, or 95% or 99% sequence identity with a ncRNA sequence set forth in Table b, or v) a retron wherein the rRT comprises a consensus sequence as set forth in Table C, or a sequence having at least 85%, or 90%, or 95% or 99% sequence identity with a consensus sequence set forth in Table C; and b) sequence-specific nuclease(s) or a DNA-repair modulating biomolecule(s) or nucleic acid sequence(s) encoding the sequence-specific nuclease(s) or the DNA- repair modulating biomolecule(s). 13. The system of paragraph 12, wherein b) comprises a programmable nuclease or a CRISPR-Cas enzyme and guide RNA for the programmable nuclease or the CRISPR-Cas enzyme, or a Class 1, Type I, II, or III Cas, or a Class 2, Type II Cas and guide RNA therefor, or a Cas9 and guide RNA therefor, or a Class 2, Type V Cas and guide RNA therefor, or a Cpfl and guide RNA therefor, or a Zinc Finger Nuclease, or a transcription activator-like effector nuclease (TALEN), or a meganuclease, or TnpB, or IscB, or a restriction endonuclease (RE), or an engineered RNA-guided FokI-nuclease, or a dCas- FokI, or a transposon-associated nuclease; or b) comprises nucleic acid sequence(s) encoding a CRISPR-Cas enzyme and guide RNA for the programmable nuclease or the CRISPR-Cas enzyme, or a Class 1, Type I, II, or III Cas, or a Class 2, Type II Cas and guide RNA therefor, or a Cas9 and guide RNA therefor, or a Class 2, Type V Cas and guide RNA therefor, or a Cpfl and guide RNA therefor, or a Zinc Finger Nuclease, or a transcription activator-like effector nuclease (TALEN), or a meganuclease, or TnpB, or IscB, or a restriction endonuclease (RE), or an engineered RNA-guided FokI-nuclease, or a dCas-FokI, or a transposon-associated nuclease. 14. The system of paragraph 13, wherein when expressed, b) is fused to the rRT, or b) is provided in cis with the rRT, optionally via a flexible linker, or a generally disordered protein sequence. 15. The system of paragraph 14, wherein b) is fused to the rRT via the flexible linker and the flexible linker comprises Gly and/or Ser rich sequences, or G4S repeats, or GS repeats, or a generally disordered protein sequence; or b) is fused to the rRT via the generally disordered protein sequence comprising an unstructured hydrophilic, or a biodegradable protein polymer, or an XTEN peptide polymer. 16. The system of paragraph 12, wherein b) and the rRT are provided in trans. 17. The system of any one of paragraphs 12-16, wherein b) comprises a CRISPR-Cas enzyme and gRNA that recognizes a target sequence. 18. The system of paragraph 17, wherein the gRNA is linked to the ncRNA and/or the msDNA, either directly or through a linker / spacer polynucleotide. 19. The system of paragraph 18, wherein the gRNA is fused to the 5’ end of the ncRNA. 20. The system of paragraph 18, wherein the gRNA is fused to the 3’ end of the ncRNA. 21. The system of any one of paragraphs 12-20, wherein b) includes a regulatory protein that enhances homology directed repair (HDR). 22. The system of paragraph 21, wherein the regulatory protein is fused to the rRT or to the sequence-specific nuclease, optionally via a flexible linker or by a generally disordered protein sequence. 23. The system of any one of paragraphs 12-22, further comprising one or more delivery vehicles. 24. The system of paragraph 23, wherein the one or more delivery vehicles comprise a liposome or a lipid nanoparticle (LNP); or, wherein the LNP comprises at least one cationic lipid comprising a lipid in Table (I), a lipid having a structure of Formula (I), a lipid having a structure of Formula (II), a lipid having a structure of Formula (III), a lipid having a structure of Formula (IV), a lipid having a structure of Formula (V), a lipid having a structure of Formula (VI), or combinations thereof; or wherein the LNP comprises a) one or more ionizable lipids, b) one or more structural lipids, c) one or more PEGylated lipids, and d) one or more phospholipids; or wherein or wherein the LNP comprises a) one or more ionizable lipids, b) one or more structural lipids, c) one or more PEGylated lipids, and d) one or more phospholipids and the one or more ionizable lipids comprises an ionizable lipid set forth in Table 2. 25. The system of paragraph 23 or 24, wherein: the delivery vehicle(s) comprise RNA cargo, said RNA cargo comprises (a) at least one mRNA molecule encoding (i) the nucleic acid programmable nuclease or CRISPR-Cas enzyme and (ii) the rRT, (b) and the ncRNA, and (c) guide RNA for the programmable nuclease, each delivery vehicle contains (a)(i) and/or (a)(ii) and/or (b) and/or (c), whereby one delivery vehicle or more than one delivery vehicle delivers (a)(i), (a)(ii), (b), and (c). 26. The system of paragraph 25, wherein (a)(i) and (a)(ii) comprise a single mRNA molecule encoding the nucleic acid programmable nuclease or CRISPR-Cas enzyme and the retron reverse transcriptase. 27. The system of paragraph 25 or 26, wherein (a)(i) and (a)(ii) are encoded and expressed as a fusion protein. 28. The system of paragraph 27, wherein the fusion protein comprises the C-terminal end of the nucleic acid programmable nuclease or CRISPR-Cas enzyme fused to the N-terminal end of the retron reverse transcriptase (nuclease:RT fusion). 29. The system of paragraph 27, wherein the fusion protein comprises the N-terminal end of the nucleic acid programmable nuclease or CRISPR-Cas enzyme fused to the C-terminal end of the retron reverse transcriptase (RT:nuclease fusion). 30. The system of paragraph 25, wherein (a)(i) and (a)(ii) comprise a first mRNA molecule encoding the nucleic acid programmable nuclease or CRISPR-Cas enzyme and a second mRNA molecule encoding the retron reverse transcriptase. 31. The system of paragraph 25, wherein (c) is separate from (a)(i), (a)(ii) and (b) or is provided in trans. 32. The system of paragraph 25, wherein (b) the ncRNA, and (c) the guide RNA are fused or are provided in cis. 33. The system of paragraph 32, wherein the guide RNA is fused to the 5’ end of the ncRNA. 34. The system of paragraph 32, wherein the guide RNA is fused to the 3’ end of the ncRNA. 35. The system of paragraph 32, wherein the ncRNA comprises a first guide RNA fused to the 5’ end of the ncRNA, and a second guide RNA fused to the 3’ end of the ncRNA, and the first and second guide RNAs target different sequences. 36. The system of paragraph 25, wherein (a) the at least one mRNA molecule encoding (i) the nucleic acid programmable nuclease or CRISPR-Cas enzyme and (ii) the retron reverse transcriptase, and (b) the ncRNA are in the same delivery vehicle. 37. The system of paragraph 25, wherein the (a) the at least one mRNA molecule encoding (i) the nucleic acid programmable nuclease or CRISPR-Cas enzyme and (ii) the retron reverse transcriptase, and (b) the ncRNA are in separate delivery vehicles. 38. The system of paragraph 25, wherein the nucleic acid programmable nuclease or CRISPR-Cas enzyme and the retron reverse transcriptase are encoded on separate mRNA molecules and those separate mRNA molecules of (a)(i) and (a)(ii) are contained in the same delivery vehicle. 39. The system of paragraph 25, wherein the nucleic acid programmable nuclease or CRISPR-Cas enzyme and the retron reverse transcriptase are encoded on separate mRNA molecules and those separate mRNA molecules of (a)(i) and (a)(ii) are contained in different delivery vehicles. 40. The system of paragraph 25, wherein the ncRNA includes a sequence of interest encoding a donor polynucleotide comprising an intended edit to be integrated at a target sequence in a cell, and wherein the donor polynucleotide is flanked by a 5' homology arm that hybridizes to a sequence 5’ to the target sequence and a 3' homology arm that hybridizes to a sequence 3' to the target sequence. 41. The system of any one of paragraphs 25-40, wherein the nucleic acid programmable nuclease or CRISPR-Cas enzyme comprises a Cas9 nuclease, a TnpB nuclease, or a Cas12a nuclease. 42. The system of paragraph 41, wherein the nucleic acid programmable nuclease comprises a Cas9 nuclease. 43. The system of paragraph any one of paragraphs 25-42, wherein the ncRNA comprises: A) a pre-msr sequence having a first complementary region of the ncRNA; B) an msr sequence including an msr stem-loop structure; C) an msd sequence including an msd stem-loop structure and a sequence of interest, wherein said msd sequence templates a single strand DNA product (RT-DNA) in the presence of the retron reverse transcriptase; and D) a post-msd sequence having a second complementary region, wherein the first and second complementary regions form an a1/a2 duplex region of the retron ncRNA, wherein the msr stem-loop structure, the msd stem-loop structure, or the a1/a2 duplex comprise a modification which result in increased editing efficiency in the presence of a nucleic acid programmable nuclease that associates with the one or more guide RNAs, and wherein optionally one or more of the guide RNAs of (c) are coupled to the pre-msr sequence, the post-msd sequence, or both the pre-msr sequence and the post-msd sequence. 44. The system of paragraph 43, wherein the sequence of interest encodes a donor polynucleotide comprising an intended edit to be integrated at a target sequence of a cell, wherein the donor polynucleotide is flanked by a 5' homology arm that hybridizes to a sequence 5’ to the target sequence and a 3' homology arm that hybridizes to a sequence 3' to the target sequence. 45. The system of any one of paragraphs 12-44, wherein the system is a gene editing system. 46. A vector system comprising vector(s) comprising the engineered retron of any one of paragraphs 1-11, or comprising the system of any one of paragraphs 12-24 or the system of paragraph 45 as dependent on paragraphs 12-24. 47. The vector system of paragraph 46, wherein the msr locus, the msd locus, and the RT gene are provided by the same vector. 48. The vector system of paragraph 46 or 47, wherein the same vector comprises a promoter operably linked to the msr locus and/or the msd locus. 49. The vector system of any one of paragraphs 46-48, wherein a promoter is operably linked to the ret locus. 50. The vector system of paragraph 46, wherein the msr locus, the msd locus, and the ret locus are provided by at least two different vectors. 51. The vector system of paragraph 46 or 50, wherein said ret locus is provided in trans with respect to the msr gene and/or the msd gene. 52. The vector system of any one of paragraphs 46-51, wherein the one or more vectors comprise a viral vector and/or a nonviral vector. 53. The vector system of paragraph 52, wherein the nonviral vector comprises a DNA plasmid or mRNA. 54. The vector system of paragraph 52, wherein the viral vector comprises an adenovirus or a lentivirus or an adeno-associated virus. 55. The vector system of any one of paragraphs 46-54, further comprising a vector encoding a homologous recombination enhancer protein, or additionally expressing a homologous recombination enhancer protein. 56. An isolated cell comprising an engineered retron or system of any of the preceding paragraphs. 57. The cell of paragraph 56, wherein the cell is a prokaryotic, archaeon, or eukaryotic cell. 58. The cell of paragraph 57, wherein the eukaryotic cell is a mammalian cell. 59. The cell of paragraph 58, wherein the mammalian cell is a non-human cell. 60. The cell of paragraph 58, wherein the mammalian cell is a human cell. 61. The cell of any of paragraphs 56-60, wherein the cell is an artificial cell or genetically modified cell. 62. A composition comprising an engineered retron or system or cell of any of the preceding paragraphs, and a pharmaceutical or veterinarily acceptable carrier. 63. A kit comprising an engineered retron or system or cell or composition of any of the preceding paragraphs, and instructions for genetically modifying a cell with the engineered retron or the system or the composition, or for use of the cell. 64. An engineered retron or system or cell or composition or kit of any of the preceding paragraphs for use in modifying a cell or for genetically modifying a cell, or for treatment of a subject or for effecting modification of a target DNA sequence in a host or subject; or use of an engineered retron or system or cell or composition or kit of any of the preceding paragraphs for modifying or genetically modifying a cell, or in treatment of a subject, or for effecting modification of a target DNA sequence in a host or subject. 65. A method of modifying a cell comprising: contacting the engineered retron or system or cell or composition of any of the preceding paragraphs with the cell. 66. A method of genetically modifying a cell comprising: contacting the cell with a system as described in any one of paragraphs 12-45, wherein: a complex is formed and guided to a target sequence, the complex provides a break in the target sequence, the rRT and ncRNA create msDNA that comprises a donor polynucleotide, and the donor polynucleotide becomes integrated at the target sequence. [0037] Accordingly, it is an object of the invention not to encompass within the invention any previously known product, process of making the product, or method of using the product such that Applicants reserve the right and hereby disclose a disclaimer of any previously known product, process, or method. It is further noted that the invention does not intend to encompass within the scope of the invention any product, process, or making of the product or method of using the product, which does not meet the written description and enablement requirements of the USPTO (35 U.S.C. §112, first paragraph) or the EPO (Article 83 of the EPC), such that Applicants reserve the right and hereby disclose a disclaimer of any previously described product, process of making the product, or method of using the product. It may be advantageous in the practice of the invention to be in compliance with Art.53(c) EPC and Rule 28(b) and (c) EPC. All rights to explicitly disclaim any embodiments that are the subject of any granted patent(s) of applicant in the lineage of this application or in any other lineage or in any prior filed application of any third party is explicitly reserved. Nothing herein is to be construed as a promise. [0038] It is noted that in this disclosure and particularly in the claims and/or paragraphs, terms such as "comprises", "comprised", "comprising" and the like can have the meaning attributed to it in U.S. Patent law; e.g., they can mean "includes", "included", "including", and the like; and that terms such as "consisting essentially of" and "consists essentially of" have the meaning ascribed to them in U.S. Patent law, e.g., they allow for elements not explicitly recited, but exclude elements that are found in the prior art or that affect a basic or novel characteristic of the invention. [0039] These and other embodiments are disclosed or are obvious from and encompassed by, the following Detailed Description. BRIEF DESCRIPTION OF THE DRAWINGS [0040] The following drawings form part of the present specification and are included to further demonstrate certain aspects of the present disclosure, which can be better understood by reference to one or more of these drawings in combination with the detailed description of specific embodiments presented herein. [0041] FIG.1A is a schematic that depicts naturally occurring retrons from genomic DNA stage through production of the msDNA chimeric microsatellite molecule. Retrons are encoded in the bacterial genome and comprise a non-coding RNA (ncRNA) portion and a portion encoding a specialized reverse transcriptase (RT). The ncRNA and the RT initially are transcribed from the retron DNA as a single polycistronic message. The initial transcript is processed resulting in the removal or separation of the transcript encoding the retron RT. The remaining transcript is the ncRNA, which undergoes folding to form a secondary structure having several characteristic stem-loops and a duplex formed between the 5ʹ and 3ʹ regions of the ncRNA (i.e., the a1/a2 duplex). The folded ncRNA is recognized by the accompanying RT which is separately translated and provided in trans. The translated RT typically recognizes certain secondary structures in the ncRNA, and binds the RNA template downstream from the msd region. The RT initiates reverse transcription of the RNA towards its 5ʹ end, starting from the 2’-end of a conserved guanosine (G) residue found immediately after a double-stranded RNA structure (the a1/a2 region) within the ncRNA. A portion (i.e., the msd region) of the ncRNA serves as a template for reverse transcription, and reverse transcription terminates before reaching the msr locus. During reverse transcription, cellular RNase H degrades the segment of the ncRNA that serves as template, but not other parts of the ncRNA. The result of the reverse transcription, the msDNA (lower right of schematic), remains covalently attached to the RNA template via the 2’- 5ʹ phosphodiester bond, and base-pairs with the RNA template using the 3ʹ end of the msDNA. [0042] FIG.1B is a schematic depicting an embodiment of a recombinant retron contemplated by this disclosure. In this embodiment, a nucleic acid molecule comprises a nucleotide sequence encoding a retron ncRNA region (the msr/msd region) is depicted at the top left. The msr region has been modified by introducing one or more nucleotide modifications (e.g., a nucleotide substitution, deletion, or insertion). For example, it may be desirable to introduce one or more nucleotide substitutions in the msr to enhance functionality (e.g., binding of the corresponding ncRNA to RT, improved stability, improved folding, etc.). The modified msr is referred to as msr’. In addition, the msd has been modified by introducing a heterologous nucleotide sequence encoding an HDR donor template. Lastly, the retron DNA has been modifed to introduce a nucleotide sequence encoding a guide RNA at the 3ʹ end of the retron DNA sequence. is configured on a DNA vector (e.g., a plasmid). The DNA is shown to be transcribed as a polycistronic message that includes the msr’/msd’ region (forming the ncRNA) which is fused at its 3ʹ end to the guide RNA (in other embodiments, the guide RNA could be fused to the 5ʹ end of the retron ncRNA. This intermediate is shown to form a complex with a reverse transcriptase provided in trans (e.g., by way of a separate expression vector or delivered mRNA). The top right schematic shows the formation of a complex between the recombinant ncRNA and the RT and the beginning of reverse transcription from the covalently-linked conserved guanosine (G) (i.e., the “priming G” or “priming guanosine”) using the msd RNA as a template sequence. Following the completion of reverse transcription and RNaseH degradation of the template RNA sequence, a recombinant msDNA is formed which comprises three modifications, as shown: (a) a guide RNA linked to the 3ʹ end of the msDNA, (b) a nucleotide change in the msr’, and (c) the reverse-transcribed single-strand DNA comprises a region that is an HDR donor template. Such a recombinant msDNA could then facilitate various genome modification applications in the cell, including genome editing with an RNA-guided nuclease provided to the cell in trans. [0043] FIG.1C is a schematic depicting a recombinant retron-based genome editing system described herein. In the case of genome editing involving an RNA-guided nuclease, the components of such a system may include (a) a guide RNA provided in cis (e.g., fused to the recombinant retron msDNA) and/or in trans (e.g., separately expressed in a cell), (b) a recombinant ncRNA (including at least a sequence encoding an HDR donor template and optionally a guide RNA fused ot the ncRNA), (c) a reverse transcriptase, and (d) a programmable nuclease. These components are provided to a cell in the form of DNA and/or RNA and/or protein by a delivery means (e.g., LNPs, liposomes, virus-based delivery, or passive/active transport). Once inside the cell, the recombinant msDNA is formed. The msDNA and the programmable nuclease translocate to the nuclease to conduct gene editing at a target DNA site, thereby producing an edited DNA target. [0044] FIG.1D provides a simplified schematic of a the natural lifecycle of a retron. Retrons typically comprise a reverse transcriptase (RT) and two non-coding contiguous inverted sequences (msr and msd) transcribed as a single RNA that is folded into a specific secondary structure. The conserved NAXXH motif and VTG triplet in retron RTs are indicated. The RT binds downstream from the msd locus in the RNA, initiating reverse transcription of the RNA template towards its 5ʹ end, assisted by the 2ʹOH group present in a conserved branching G residue acting as a primer. Reverse transcription halts before the msr region is reached, and the resulting msDNA remains covalently attached to the RNA template via a 2ʹ-5ʹ phosphodiester bond and base-pairing of the 3ʹ ends of the molecules. [0045] FIG.1E provides a detailed representation of the natural biological pathway of a retron in a cell, concluding with the generation of the msDNA satellite molecule. This figure parallels FIG.1D but more completely depicts the stages of msDNA production. (1) depicts the retron locus which includes an ncRNA locus having an msr locus and a msd locus (both of which are non-coding) and a reverse transcriptase (RT) locus. The ncDNA locus and the RT locus are transcribed as a single RNA transcript, which is depicted in (2). The colors representing each component in (1) are carried through each of stages (2) through (6). Stages (3) and (4) depict the folding of the ncRNA portion into a series of stem loops, wherein the 5ʹ end and the 3ʹ ends of the ncRNA form a duplex. In addition, the position of the conserved branching guanosine residue having a 2’OH group is show. The branching guanosine serves as a future priming site for the reverse transcriptase. Stage (4) further shows that the region of the transcript encoding the reverse transcriptase is removed, separately being translated to produce the reverse transcriptas enzyme. In stage (5), the reverse transcriptase associates with the folded ncRNA and begins polymerization of a single strand of DNA (i.e., the reverse transcription product) from the primer site (i.e., the conserved branching guanosine residue having the 2’OH end) and using the msd RNA sequence as a template. Reverse transcription terminates at the msr region. The msd RNA template is exonucleotically removed, thereby resulting in a chimeric molecule comprising the msr RNA region which is covalently joined to the ssDNA transcription product through covalent linkage to the conserved guanosine primer residue. There is also a short duplex region that forms between the 3ʹ end of the msr RNA and the 3ʹ of the ssDNA reverse transcript product. The complete molecule is referred to as the “msDNA.” [0046] FIG.1F is a schematic depicting that the herein disclosed recombinant retron-based genome modification systems may be implemented as (a) cell recorder systems, (b) genome editing systems, and (c) recombineering systems. These uses are not intended to be limiting. [0047] FIG.1G is a schematic depicting various configurations contemplated for the recombinant retron disclosed herein. (1) shows the operonic structure of a wild type retron; (2) shows the operonic structure of a recombinant retron configured to encode an HDR donor template in the final msDNA molecule; (3) shows (2) but further modified to encode a guide RNA at the 3ʹ end of the retron; (4) shows (2) but further modified to encode a guide RNA in trans. [0048] FIG.1H is a schematic that emphasizes that any suitable configuration for presenting the components of the recombinant retron-based genome modification systems disclosed herein to a cell are contemplated, including where the RT and/or the programmable nuclease are provided in trans relative to the retron ncRNA. In some embodiments, the RT and the programmable nuclease may be provided as a fusion protein. [0049] FIG.1I depicts that the RT and programmable nuclease may be provided as fusion proteins (top, middle) or provided separate from one another. [0050] FIG.1J depicts that nuclear localization signals may be engineered into the polypeptides of the disclosure (e.g., a RNA-guide nuclease) to facilitate translocation into the nuclease of cell where editing occurs. [0051] FIG.1K is a schematic (not to scale) representation depicting an embodiment of genome editing, in which double-stranded break (DSB) created by a suitable nuclease (such as a CRISPR/Cas effector enzyme, a ZFN, TALEN, meganuclease, TnpB, IscB, or restriction enzymes (Res)) promotes the insertion of a donor or template sequence (shown here as a “marker” flanked by homologous sequences matching those flanking the DSB, provided on a “donor vector”). [0052] FIG.2 is a schematic representation of a consensus secondary structure of a retron ncRNA msr/msd of Type IA/IIA1 retron produced by computational structural alignment of ncRNA sequences from Table B as described in Example 3. [0053] The colored dots represents the probability a base is at that location (e.g., red circle represents the presence of a base in 97% of the cases, black represents the presence of a base in 90-97% of the cases, grey represents the presence of a base in 75-90% of the cases, and white represents the presence of a base in 50-75% of the cases), as opposed to a gap (no base), whereas the colored letters represent bases that are conserved to different degrees (e.g., with red representing 97%+ conserved, black being 90%+ conserved, and grey at least 75% conserved). Each highlighted base-pair represents a significantly covarying basepair. [0054] FIG.3 is a schematic representation of a consensus secondary structure of a retron ncRNA msr/msd of Type IB1 retrons produced by computational structural alignment of ncRNA sequences from Table B as described in Example 3. [0055] The colored dots represents the probability a base is at that location (e.g., red circle represents the presence of a base in 97% of the cases, black represents the presence of a base in 90-97% of the cases, grey represents the presence of a base in 75-90% of the cases, and white represents the presence of a base in 50-75% of the cases), as opposed to a gap (no base), whereas the colored letters represent bases that are conserved to different degrees (e.g., with red representing 97%+ conserved, black being 90%+ conserved, and grey at least 75% conserved). Each highlighted base-pair represents a significantly covarying basepair. [0056] FIG.4 is a schematic representation of a consensus secondary structure of a retron ncRNA msr/msd of Type IB2 retrons produced by computational structural alignment of ncRNA sequences from Table B as described in Example 3. [0057] The colored dots represents the probability a base is at that location (e.g., red circle represents the presence of a base in 97% of the cases, black represents the presence of a base in 90-97% of the cases, grey represents the presence of a base in 75-90% of the cases, and white represents the presence of a base in 50-75% of the cases), as opposed to a gap (no base), whereas the colored letters represent bases that are conserved to different degrees (e.g., with red representing 97%+ conserved, black being 90%+ conserved, and grey at least 75% conserved). Each highlighted base-pair represents a significantly covarying basepair. [0058] FIG.5 is a schematic representation of a consensus secondary structure of a retron ncRNA msr/msd of Type IC retrons produced by computational structural alignment of ncRNA sequences from Table B as described in Example 3. [0059] The colored dots represents the probability a base is at that location (e.g., red circle represents the presence of a base in 97% of the cases, black represents the presence of a base in 90-97% of the cases, grey represents the presence of a base in 75-90% of the cases, and white represents the presence of a base in 50-75% of the cases), as opposed to a gap (no base), whereas the colored letters represent bases that are conserved to different degrees (e.g., with red representing 97%+ conserved, black being 90%+ conserved, and grey at least 75% conserved). Each highlighted base-pair represents a significantly covarying basepair. [0060] FIG.6 is a schematic representation of a consensus secondary structure of a retron ncRNA msr/msd of Type IIA other retrons produced by computational structural alignment of ncRNA sequences from Table B as described in Example 3. [0061] The colored dots represents the probability a base is at that location (e.g., red circle represents the presence of a base in 97% of the cases, black represents the presence of a base in 90-97% of the cases, grey represents the presence of a base in 75-90% of the cases, and white represents the presence of a base in 50-75% of the cases), as opposed to a gap (no base), whereas the colored letters represent bases that are conserved to different degrees (e.g., with red representing 97%+ conserved, black being 90%+ conserved, and grey at least 75% conserved). Each highlighted base-pair represents a significantly covarying basepair. [0062] FIG.7 is a schematic representation of a consensus secondary structure of a retron ncRNA msr/msd of Type IIA2 retrons produced by computational structural alignment of ncRNA sequences from Table B as described in Example 3. [0063] The colored dots represents the probability a base is at that location (e.g., red circle represents the presence of a base in 97% of the cases, black represents the presence of a base in 90-97% of the cases, grey represents the presence of a base in 75-90% of the cases, and white represents the presence of a base in 50-75% of the cases), as opposed to a gap (no base), whereas the colored letters represent bases that are conserved to different degrees (e.g., with red representing 97%+ conserved, black being 90%+ conserved, and grey at least 75% conserved). Each highlighted base-pair represents a significantly covarying basepair. [0064] FIG.8 is a schematic representation of a consensus secondary structure of a retron ncRNA msr/msd of Type IIA3 retrons produced by computational structural alignment of ncRNA sequences from Table B as described in Example 3. [0065] The colored dots represents the probability a base is at that location (e.g., red circle represents the presence of a base in 97% of the cases, black represents the presence of a base in 90-97% of the cases, grey represents the presence of a base in 75-90% of the cases, and white represents the presence of a base in 50-75% of the cases), as opposed to a gap (no base), whereas the colored letters represent bases that are conserved to different degrees (e.g., with red representing 97%+ conserved, black being 90%+ conserved, and grey at least 75% conserved). Each highlighted base-pair represents a significantly covarying basepair. [0066] FIG.9 is a schematic representation of a consensus secondary structure of a retron ncRNA msr/msd of Type IIA4 retrons produced by computational structural alignment of ncRNA sequences from Table B as described in Example 3. [0067] The colored dots represents the probability a base is at that location (e.g., red circle represents the presence of a base in 97% of the cases, black represents the presence of a base in 90-97% of the cases, grey represents the presence of a base in 75-90% of the cases, and white represents the presence of a base in 50-75% of the cases), as opposed to a gap (no base), whereas the colored letters represent bases that are conserved to different degrees (e.g., with red representing 97%+ conserved, black being 90%+ conserved, and grey at least 75% conserved). Each highlighted base-pair represents a significantly covarying basepair. [0068] FIG.10 is a schematic representation of a consensus secondary structure of a retron ncRNA msr/msd of Type IIA5 retrons produced by computational structural alignment of ncRNA sequences from Table B as described in Example 3. [0069] The colored dots represents the probability a base is at that location (e.g., red circle represents the presence of a base in 97% of the cases, black represents the presence of a base in 90-97% of the cases, grey represents the presence of a base in 75-90% of the cases, and white represents the presence of a base in 50-75% of the cases), as opposed to a gap (no base), whereas the colored letters represent bases that are conserved to different degrees (e.g., with red representing 97%+ conserved, black being 90%+ conserved, and grey at least 75% conserved). Each highlighted base-pair represents a significantly covarying basepair. [0070] FIG.11 is a schematic representation of a consensus secondary structure of a retron ncRNA msr/msd of Type IIIA1 retrons produced by computational structural alignment of ncRNA sequences from Table B as described in Example 3. [0071] The colored dots represents the probability a base is at that location (e.g., red circle represents the presence of a base in 97% of the cases, black represents the presence of a base in 90-97% of the cases, grey represents the presence of a base in 75-90% of the cases, and white represents the presence of a base in 50-75% of the cases), as opposed to a gap (no base), whereas the colored letters represent bases that are conserved to different degrees (e.g., with red representing 97%+ conserved, black being 90%+ conserved, and grey at least 75% conserved). Each highlighted base-pair represents a significantly covarying basepair. [0072] FIG.12 is a schematic representation of a consensus secondary structure of a retron ncRNA msr/msd of Type IIIA2 retrons produced by computational structural alignment of ncRNA sequences from Table B as described in Example 3. [0073] The colored dots represents the probability a base is at that location (e.g., red circle represents the presence of a base in 97% of the cases, black represents the presence of a base in 90-97% of the cases, grey represents the presence of a base in 75-90% of the cases, and white represents the presence of a base in 50-75% of the cases), as opposed to a gap (no base), whereas the colored letters represent bases that are conserved to different degrees (e.g., with red representing 97%+ conserved, black being 90%+ conserved, and grey at least 75% conserved). Each highlighted base-pair represents a significantly covarying basepair. [0074] FIG.13 is a schematic representation of a consensus secondary structure of a retron ncRNA msr/msd of IIIA3 retrons produced by computational structural alignment of ncRNA sequences from Table B as described in Example 3. [0075] The colored dots represents the probability a base is at that location (e.g., red circle represents the presence of a base in 97% of the cases, black represents the presence of a base in 90-97% of the cases, grey represents the presence of a base in 75-90% of the cases, and white represents the presence of a base in 50-75% of the cases), as opposed to a gap (no base), whereas the colored letters represent bases that are conserved to different degrees (e.g., with red representing 97%+ conserved, black being 90%+ conserved, and grey at least 75% conserved). Each highlighted base-pair represents a significantly covarying basepair. [0076] FIG.14 is a schematic representation of a consensus secondary structure of a retron ncRNA msr/msd of Type IIIA4 retrons produced by computational structural alignment of ncRNA sequences from Table B as described in Example 3. [0077] The colored dots represents the probability a base is at that location (e.g., red circle represents the presence of a base in 97% of the cases, black represents the presence of a base in 90-97% of the cases, grey represents the presence of a base in 75-90% of the cases, and white represents the presence of a base in 50-75% of the cases), as opposed to a gap (no base), whereas the colored letters represent bases that are conserved to different degrees (e.g., with red representing 97%+ conserved, black being 90%+ conserved, and grey at least 75% conserved). Each highlighted base-pair represents a significantly covarying basepair. [0078] FIG.15 is a schematic representation of a consensus secondary structure of a retron ncRNA msr/msd of Type IIIA5 retrons produced by computational structural alignment of ncRNA sequences from Table B as described in Example 3. [0079] The colored dots represents the probability a base is at that location (e.g., red circle represents the presence of a base in 97% of the cases, black represents the presence of a base in 90-97% of the cases, grey represents the presence of a base in 75-90% of the cases, and white represents the presence of a base in 50-75% of the cases), as opposed to a gap (no base), whereas the colored letters represent bases that are conserved to different degrees (e.g., with red representing 97%+ conserved, black being 90%+ conserved, and grey at least 75% conserved). Each highlighted base-pair represents a significantly covarying basepair. [0080] FIG.16 is a schematic representation of a consensus secondary structure of a retron ncRNA msr/msd of Type IIIunk retrons produced by computational structural alignment of ncRNA sequences from Table B as described in Example 3. [0081] The colored dots represents the probability a base is at that location (e.g., red circle represents the presence of a base in 97% of the cases, black represents the presence of a base in 90-97% of the cases, grey represents the presence of a base in 75-90% of the cases, and white represents the presence of a base in 50-75% of the cases), as opposed to a gap (no base), whereas the colored letters represent bases that are conserved to different degrees (e.g., with red representing 97%+ conserved, black being 90%+ conserved, and grey at least 75% conserved). Each highlighted base-pair represents a significantly covarying basepair. [0082] FIG.17 is a schematic representation of a consensus secondary structure of a retron ncRNA msr/msd of Type IV retrons produced by computational structural alignment of ncRNA sequences from Table B as described in Example 3. [0083] The colored dots represents the probability a base is at that location (e.g., red circle represents the presence of a base in 97% of the cases, black represents the presence of a base in 90-97% of the cases, grey represents the presence of a base in 75-90% of the cases, and white represents the presence of a base in 50-75% of the cases), as opposed to a gap (no base), whereas the colored letters represent bases that are conserved to different degrees (e.g., with red representing 97%+ conserved, black being 90%+ conserved, and grey at least 75% conserved). Each highlighted base-pair represents a significantly covarying basepair. [0084] FIG.18 is a schematic representation of a consensus secondary structure of a retron ncRNA msr/msd of Type IX retrons produced by computational structural alignment of ncRNA sequences from Table B as described in Example 3. [0085] The colored dots represents the probability a base is at that location (e.g., red circle represents the presence of a base in 97% of the cases, black represents the presence of a base in 90-97% of the cases, grey represents the presence of a base in 75-90% of the cases, and white represents the presence of a base in 50-75% of the cases), as opposed to a gap (no base), whereas the colored letters represent bases that are conserved to different degrees (e.g., with red representing 97%+ conserved, black being 90%+ conserved, and grey at least 75% conserved). Each highlighted base-pair represents a significantly covarying basepair. [0086] FIG.19 is a schematic representation of a consensus secondary structure of a retron ncRNA msr/msd of Type V retrons produced by computational structural alignment of ncRNA sequences from Table B as described in Example 3. [0087] The colored dots represents the probability a base is at that location (e.g., red circle represents the presence of a base in 97% of the cases, black represents the presence of a base in 90-97% of the cases, grey represents the presence of a base in 75-90% of the cases, and white represents the presence of a base in 50-75% of the cases), as opposed to a gap (no base), whereas the colored letters represent bases that are conserved to different degrees (e.g., with red representing 97%+ conserved, black being 90%+ conserved, and grey at least 75% conserved). Each highlighted base-pair represents a significantly covarying basepair. [0088] FIG.20 is a schematic representation of a consensus secondary structure of a retron ncRNA msr/msd of Type VI retrons produced by computational structural alignment of ncRNA sequences from Table B as described in Example 3. [0089] The colored dots represents the probability a base is at that location (e.g., red circle represents the presence of a base in 97% of the cases, black represents the presence of a base in 90-97% of the cases, grey represents the presence of a base in 75-90% of the cases, and white represents the presence of a base in 50-75% of the cases), as opposed to a gap (no base), whereas the colored letters represent bases that are conserved to different degrees (e.g., with red representing 97%+ conserved, black being 90%+ conserved, and grey at least 75% conserved). Each highlighted base-pair represents a significantly covarying basepair. [0090] FIG.21 is a schematic representation of a consensus secondary structure of a retron ncRNA msr/msd of Type XI Group 1 retrons produced by computational structural alignment of ncRNA sequences from Table B as described in Example 3. [0091] The colored dots represents the probability a base is at that location (e.g., red circle represents the presence of a base in 97% of the cases, black represents the presence of a base in 90-97% of the cases, grey represents the presence of a base in 75-90% of the cases, and white represents the presence of a base in 50-75% of the cases), as opposed to a gap (no base), whereas the colored letters represent bases that are conserved to different degrees (e.g., with red representing 97%+ conserved, black being 90%+ conserved, and grey at least 75% conserved). Each highlighted base-pair represents a significantly covarying basepair. [0092] FIG.22 is a schematic representation of a consensus secondary structure of a retron ncRNA msr/msd of Type XI Group 2 retrons produced by computational structural alignment of ncRNA sequences from Table B as described in Example 3. [0093] The colored dots represents the probability a base is at that location (e.g., red circle represents the presence of a base in 97% of the cases, black represents the presence of a base in 90-97% of the cases, grey represents the presence of a base in 75-90% of the cases, and white represents the presence of a base in 50-75% of the cases), as opposed to a gap (no base), whereas the colored letters represent bases that are conserved to different degrees (e.g., with red representing 97%+ conserved, black being 90%+ conserved, and grey at least 75% conserved). Each highlighted base-pair represents a significantly covarying basepair. [0094] FIG.23 is a schematic representation of a consensus secondary structure of a retron ncRNA msr/msd of Type XII retrons produced by computational structural alignment of ncRNA sequences from Table B as described in Example 3. [0095] The colored dots represents the probability a base is at that location (e.g., red circle represents the presence of a base in 97% of the cases, black represents the presence of a base in 90-97% of the cases, grey represents the presence of a base in 75-90% of the cases, and white represents the presence of a base in 50-75% of the cases), as opposed to a gap (no base), whereas the colored letters represent bases that are conserved to different degrees (e.g., with red representing 97%+ conserved, black being 90%+ conserved, and grey at least 75% conserved). Each highlighted base-pair represents a significantly covarying basepair. [0096] FIG.24 is a schematic representation of a consensus secondary structure of a retron ncRNA msr/msd of Type XIII retrons produced by computational structural alignment of ncRNA sequences from Table B as described in Example 3. [0097] The colored dots represents the probability a base is at that location (e.g., red circle represents the presence of a base in 97% of the cases, black represents the presence of a base in 90-97% of the cases, grey represents the presence of a base in 75-90% of the cases, and white represents the presence of a base in 50-75% of the cases), as opposed to a gap (no base), whereas the colored letters represent bases that are conserved to different degrees (e.g., with red representing 97%+ conserved, black being 90%+ conserved, and grey at least 75% conserved). Each highlighted base-pair represents a significantly covarying basepair. [0098] FIG.25 is a schematic representation of a consensus secondary structure of a retron ncRNA msr/msd of Type XIV retrons produced by computational structural alignment of ncRNA sequences from Table B as described in Example 3. [0099] The colored dots represents the probability a base is at that location (e.g., red circle represents the presence of a base in 97% of the cases, black represents the presence of a base in 90-97% of the cases, grey represents the presence of a base in 75-90% of the cases, and white represents the presence of a base in 50-75% of the cases), as opposed to a gap (no base), whereas the colored letters represent bases that are conserved to different degrees (e.g., with red representing 97%+ conserved, black being 90%+ conserved, and grey at least 75% conserved). Each highlighted base-pair represents a significantly covarying basepair. [00100] FIG.26 is a schematic representation of a consensus secondary structure of a retron ncRNA msr/msd of Ec107 retrons produced by computational structural alignment of ncRNA sequences from Table B as described in Example 3. [00101] The colored dots represents the probability a base is at that location (e.g., red circle represents the presence of a base in 97% of the cases, black represents the presence of a base in 90-97% of the cases, grey represents the presence of a base in 75-90% of the cases, and white represents the presence of a base in 50-75% of the cases), as opposed to a gap (no base), whereas the colored letters represent bases that are conserved to different degrees (e.g., with red representing 97%+ conserved, black being 90%+ conserved, and grey at least 75% conserved). Each highlighted base-pair represents a significantly covarying basepair. [00102] FIG.27 is a schematic representation of a consensus secondary structure of a retron ncRNA msr/msd of Outgroup A retrons produced by computational structural alignment of ncRNA sequences from Table B as described in Example 3. [00103] The colored dots represents the probability a base is at that location (e.g., red circle represents the presence of a base in 97% of the cases, black represents the presence of a base in 90-97% of the cases, grey represents the presence of a base in 75-90% of the cases, and white represents the presence of a base in 50-75% of the cases), as opposed to a gap (no base), whereas the colored letters represent bases that are conserved to different degrees (e.g., with red representing 97%+ conserved, black being 90%+ conserved, and grey at least 75% conserved). Each highlighted base-pair represents a significantly covarying basepair. [00104] FIG.28 is a phylogenetic tree of RT sequences constructed in accordance with Example 3. [00105] FIG.29 is a structural representation of the retron loci associated with each retron type in FIG.28. [00106] FIG.30 shows the position of certain retrons (EcoI, Eco3, Eco5, AcoI, RTX003_2042, RTX003_6083v1, and RTX003_6943) within the phylogenetic retron tree of FIG.28. [00107] FIG.31A is a plasmid map of an exemplary retron (EcoI) tested in the Examples herein. [00108] FIG.31B is a linear representation in 5’ to 3’ direction of the plasmid map of FIG.31A. [00109] FIG.31C is a plasmid map of an exemplary retron (RTX3_6083v1) tested in the Examples herein. [00110] FIG.31D is a linear representation in 5’ to 3’ direction of the plasmid map of FIG.31C. [00111] FIG.32 is a representation of a plasmid-based assay used to measure retron precise edits and indels as performed in the Examples. In Step 1, a plasmid (e.g., that of FIG. 31A or FIG.31C) is transfected into human cells (e.g., HEK293t cells) which are engineered to express Cas9. Editing is allowed to occur for 72 hours at 37°C. In Step 2, the genomic DNA is extracted from the cells and used to prepare a next-generation sequencing (NGS) library for sequencing. The library is sequenced over the target site (e.g., EMX1) of editing to generate sequence reads. In Step 3, the sequencing reads are analyzed to obtain a frequency of sequence reads containing the desired edit (percentage of precision editing) and a frequence of indels at the desired edit site (percentage of indels). [00112] FIG.33 is an equivalent representation of FIG.32. [00113] FIG.34 is a representation of a methodology for transfecting HEK293T cells. Cells were seeded in 24 well plate one day prior to transfection. Appropriate amount of plasmid and transfection reagent (e.g., Lipofectamine 3000) were mixed and transferred to cells. After 72 hours incubation, genomic DNA was extracted and the target edit region was amplified into sequencing libraries. Sequencing data was analyzed by CRISPResso2 and percentage of precise edit and indels were calculated. [00114] FIG.35 is an example of reference sequence and desired editing outcome for Eco3 retron at he EMX1 genomic site. Analysis of editing outcomes is performed using CRISPresso2 pipeline. In this example, the editing template inserts a 10 bp insertion into the EMX1 gene (TTACGTCTGC) along with a 6 bp substitution to mutate the PAM sequence (GAAGGG>AAAGTT). [00115] FIG.36 shows the results of plasmid-based assay (e.g., according to FIG.33) demonstrating up to about 0.3% precise edits and as low as 40% indels with Eco1 retron in Cas9 expressing HEK293T cells. The plasmid that encodes Eco1 RT and Eco1 ncRNA-sgRNA fusion targeting EMX1 was transfected via lipofection using two different amounts of Lipofectamine. [00116] FIG.37 shows the results of plasmid-based assay (e.g., according to FIG.33) demonstrating up to about 0.1% precise edits and as low as 3% indels with AcoI. Aco1 retron has not been experimentally validated to produce msDNA. Precise editing activity observed in this experiment strongly support that Aco1 retron is capable of generating msDNA inside human cells. [00117] FIG.38 shows the results of plasmid-based assay (e.g., according to FIG.33) demonstrating up to about 0.3% precise edits and as low as 5% indels with RTX003_2042. This retron can achieve a comparable precise editing to Eco1 but with significantly lower indels (10- fold). RTX003_2042 is a novel retron and precise editing activity observed in this experiment strongly support that RTX003_2042 retron could generate msDNA inside human cells. [00118] FIG.39A shows the results of plasmid-based assay demonstrating up to about 0.05~0.08% precise edits and as low as 2.5~4% indels with RTX003_6083v1 and 6943. Both are novel retrons and precise editing activity observed in this experiment strongly support that RTX003_6083v1 and 6943 retron could generate msDNA inside human cells. [00119] FIG.39B shows follow up experiments using the same assay of FIG.39A indicated that RTX3_6083v1 and RTX3_6943 generated 3-4 fold more precise edits than Eco1 while indels generated from these two retrons were 2-3 fold lower. RTX3_2042 showed precise editing at similar frequencies to Eco1 but had more variability than other samples. [00120] FIG.39C shows follow up experiments using the same assay of FIG.39A indicated that RTX3_6083v1 and RTX3_6943 generated 3-4 fold more precise edits than Eco1 while indels generated from these two retrons were 2-3 fold lower. RTX3_2042 showed precise editing at similar frequencies to Eco1 but had more variability than other samples. [00121] FIG.39D shows the results of plasmid-based assay demonstrating up to about 0.7% precise edits and as low as ~4% indels with RTX003_0637, RTX003_1262, and RTX003_6342 compared to empty vector and EcoI. RTX003_0637, RTX003_1262, and RTX003_6342 are novel retrons and precise editing activity observed in this experiment strongly support that these retrons could generate msDNA inside cells. [00122] FIG.39E shows the results of plasmid-based assay demonstrating precise editing and indel generation for an array of retrons, including EcoI, Eco3, RTX3_2042_RT_inactivated, RTX3_2042, RTX3_6083v1, RTX3_6943, RTX3_6943, RTX3_1262, RTX3_6342S, and RTX3_6342L. up to about 2.5% precise edits and as low as ~4% indels with EcoI, Eco3, RTX3_2042_RT_inactivated, RTX3_2042, RTX3_6083v1, RTX3_6943, RTX3_6943, RTX3_1262, RTX3_6342S, and RTX3_6342L compared to empty vector and EcoI as controls. [00123] FIG.40 is a representation of an two-RNA editing assay used in the Examples to measure the relative editing efficiency of exemplary retrons using electroporation-based delivery of two RNA components into HEK293T cells. Appropriate amount of RT mRNA and ncRNA- sgRNA fusion were mixed and electroporated to cells. After 72 hours incubation, genomic DNA was extracted and the targeting region was amplified into sequencing libraries. Sequencing data was analyzed by CRISPResso2 and precise edit and indels were calculated. [00124] FIG.41 shows the results of two-RNA system (RT mRNA + ncRNA-sgRNA fusion) delivered to Cas9 expressing HEK293T cells by electroporation. Eco1, Eco3 and Eco5 retrons were tested. Results showed precise edits (left graph) up to 0.4% for Eco3 and as low as 10% indels (right graph) for Eco3. Precise edits mediated by Eco3 increased with augmenting amount of ncRNA-sgRNA fusion from 1:2 to 1:4 ratio between RT mRNA and ncRNA-sgRNA fusion. [00125] FIG.42 shows the results of titration of two-component Eco3 RNA system (RT mRNA + ncRNA-sgRNA fusion) delivered to Cas9 expressing 293T cells by electroporation. The RT mRNA and the ncRNA were mixed at ratios of 1:2, 1:3, 1:4, 1:5, 1:8, 1:10, respectively, and delivered at two different amounts of RT mRNA (0.2 or 0.5 µg). On left, data showed that Eco3 at 0.5 µg produced highest percentage of precise edits at a 1:3 and 1:5 ratio of RT mRNA to ncRNA. On right, data further showed that a more equivalent ratio of RT mRNA to ncRNA resulted in a trend of lower percentage of indels. [00126] FIG.43 is a representation of a three-RNA retron editing system which involves delivery by electroporation of three RNA components (RT mRNA, retron ncRNA-sgRNA fusion, and Cas9 mRNA) into HEK293T cells. Appropriate amount of RT mRNA, ncRNA- sgRNA fusion, and Cas9 mRNA were mixed and electroporated to cells. After 72 hours incubation, genomic DNA was extracted and targeting region was amplified into sequencing libraries. Sequencing data was analyzed by CRISPResso2 and precise edit and indels were calculated. [00127] FIG.44 shows the results of Cas9 mRNA titration of three-component Eco3 RNA system (RT mRNA + ncRNA-sgRNA fusion + Cas9 mRNA) delivered to 293T cells by electroporation. The RT mRNA and the ncRNA-sgRNA fusion were mixed at given amounts on the graph and the amount Cas9 mRNA was titrated. At 0.2 µg of Cas9 mRNA, up to 0.1% of precise editing was observed. While the editing efficiency is an order of magnitude lower than two RNA system, the editing occurred by specific action of Cas9 and retron, since absence of either abrogated the editing. [00128] FIG.45 depicts a process of lipofection using three RNA system in HEK293T cells. Cells were seeded in 96 well plate one day prior to transfection. Appropriate amount of RT mRNA, ncRNA-sgRNA fusion, Cas9 mRNA and Lipofectamine reagent were mixed and transferred to cells. After 72 hours incubation, genomic DNA was extracted and targeting region was amplified into sequencing libraries. Sequencing data was analyzed by CRISPResso2 and precise edit and indels were calculated. [00129] FIG.46 shows the results of three-component Aco1 RNA system (RT mRNA + ncRNA +Cas9 mRNA) delivered to HEK293T cells by lipofection. The RT mRNA, the ncRNA and the Cas9 mRNA were mixed at amounts indicated in the graph and transfected to HEK293T cells.56 bp insertion and 6 bp deletion at EMX1 locus was scored as precise edits and ~0.1% of cell population has undergone precise editing on the left graph. The editing was dependent on Cas9 nuclease since its absence abrogated the editing. The frequency of indels was about 1.5% on the right graph. [00130] FIG.47 shows the results of minimal Cas9 nuclease activity when sgRNA is fused to ncRNA of Eco3 retron. Cas9 activity was evaluated by frequency of indels.1 µg of ncRNA-sgRNA fusion shows 20-fold lower activity than equimolar separated sgRNA alone. In parallel, activity of chemical modified vs unmodified sgRNA was compared and the former shows 6-fold higher activity than the latter at the condition described in the graph. [00131] FIG.48 is a representation of the all-RNA editing assay used in the Examples to measure the relative editing efficiency of the sample retrons in an all-RNA format, modified with a step of in trans guide RNA spike-in. Electroporation using three RNA system + sgRNA trans spike-in in HEK293T cells. Appropriate amount of RT mRNA, ncRNA-sgRNA fusion, Cas9 mRNA, and sgRNA were mixed and electroporated to cells. After 72 hours incubation, genomic DNA was extracted and targeting region was amplified into sequencing libraries. Sequencing data was analyzed by CRISPResso2 and precise edit and indels were calculated. [00132] FIG.49 shows the results of guide RNA spike-in in all RNA system (RT mRNA + ncRNA-sgRNA fusion + Cas9 mRNA + sgRNA) delivered to HEK293T cells by electroporation. At given amount of Cas9 and RT mRNA on the graph, the amount of guide RNA spike-in is titrated at 50, 100 and 200 ng. The titration was done at two different ratios of RT mRNA: ncRNA-sgRNA fusion=1:6 or 1:8. The guide RNA spike-in in all RNA system increased precise editing up to ~50 fold. The increasing amount of guide RNA gradually increased precise editing and 1:8 of RT mRNA:ncRNA-sgRNA fusion performed slightly better than 1:6, reaching 13% of precise editing. On the right graph, frequency of indels is shown for respective conditions. [00133] FIG.50 represents a lipofection process using three RNA system + gRNA trans spike-in in HEK293T cells. Cells were seeded in 96 well plate one day prior to transfection. Appropriate amount of RT mRNA, ncRNA-sgRNA fusion, Cas9 mRNA, sgRNA and Lipofectamine reagent were mixed and transferred to cells. After 72 hours incubation, genomic DNA was extracted and targeting region was amplified into sequencing libraries. Sequencing data was analyzed by CRISPResso2 and precise edit and indels were calculated. [00134] FIG.51 shows the results of guide RNA spike-in (i.e., delivering a separate molecule bolus of guide RNA which in all RNA system (RT mRNA + ncRNA-sgRNA fusion + Cas9 mRNA + sgRNA) delivered to HEK293T cells by lipofection. At given amount of RT mRNA, ncRNA-sgRNA fusion, and Cas9 mRNA on the graph, the amount of guide RNA spike- in is titrated at 2, 5 and 10 ng. The guide RNA spike-in in all RNA system increased precise editing up to 3.5 fold, 12% of efficiency. The increasing amount of guide RNA at this range did not further increase precise editing. The precise editing is completely dependent on the presence of Retron machinery. On the right graph, frequency of indels is shown for respective conditions. [00135] FIG.52 shows the results of ncRNA-sgRNA fusion separation in all RNA system (RT mRNA + Cas9 mRNA + either ncRNA-sgRNA fusion OR separate ncRNA + sgRNA) delivered to HEK293T cells by lipofection. At given amount of RT mRNA and Cas9 mRNA on the graph, the amount of guide RNA spike-in is titrated at 0, 2, 5, 10, 50 and 100 ng. At 10 ng guide RNA, precise editing peaked at 2.23% compared to 1.78% with ncRNA-sgRNA fusion. The increasing amount of guide RNA at this range did not further increase precise editing. On the right graph, frequency of indels is shown for respective conditions. [00136] FIG.53 is a schematic of improved templates used for in vitro transcription to produce ncRNA (left or A) and ncRNA modifications (right or B). (A) relates to the optimization of RNA production by in vitro transcription. Previously made in vitro transcription experiments to produce RNA used a double-stranded DNA template containing a 3’ overhang (on same strand as T7 promoter sequence). A new template with a blunt end was designed and tested and as shown in FIG.54 results in increased precise editing efficiency. (B) relates to modified ncRNAs which are modified by addition of an MS2 stem loop hairpin at the 3’ end of the ncRNA. Without being bound by theory, the MS2 loop helps stabilize the ncRNA and results in significantly improved precise editing efficiency, as shown in FIG.54. [00137] FIG.54 shows the results of ncRNA-sgRNA fusion separation in 4 component all-RNA system (RT mRNA + Cas9 mRNA + ncRNA + sgRNA) delivered to HEK293T cells by Lipofectamine MessengerMAX. All RNA was transfected at a fixed amount as shown on the graphs. Using RNA generated from linearized plasmid template containing a 3’ overhang produced 1.35% precise edits. Using RNA generated from the improved linearized plasmid template containing a blunt end increased precise editing to 5.94%. Adding an MS2 stem loop to the 3’ end of the ncRNA (blunt end) further increased precise editing to 12.39%. On the right graph, frequency of indels is shown for respective conditions. [00138] FIG.55 provides schematics for end protection of RNA from cellular nuclease activity by capping and tailing. In (A), a 7-methylguanosine cap0 was added to 5’ triphosphate of RNA. In (B), a poly-A tail was added to the 3’ end by enzymatic addition. Tail length is estimated over 50 nucleotides. In (C), RNA containing both a 5’ cap and a 3’ tail is shown. Results are shown in FIG.56. [00139] FIG.56 shows the result of end protection of ncRNA-sgRNA fusion by cap and tail in 4 component all-RNA system (RT mRNA + Cas9 mRNA + ncRNA + sgRNA) delivered to HEK293T cells by Lipofectamine MessengerMAX. All RNA was transfected at a fixed amount RT mRNA 100 ng, ncRNA-sgRNA 400ng, Cas9 mRNA 100 ng, and sgRNA 5 ng. ncRNA-gRNA fusion was either capped (+cap –tail) or poly-A tailed (-cap +tail) or both capped and poly-A tailed (+cap +tail). Using RNA without end protection (-cap –tail) produced ~4.5 % precise edits and the editing was dependent on retron since the absence of RT abrogated precise editing. Using RNA with either or both protection by cap and tail produced lower precise editing (left graph) but lowered indels (right graph) than without cap and tail. [00140] FIG.57 shows the result that shortening msd stems can modulate precising editing in a retron specific manner. Modified retrons include RTX3_4536 (long (L) and short (S) versions), RTX3_6279 (long (L) and short (S) versions), RTX3_6342 (long (L) and short (S) versions), RTX3_6438 (long (L) and short (S) versions), RTX3_6549 (long (L) and short (S) versions), and RTX3_6605 (long (L) and short (S) versions). [00141] FIG.58 shows the result that shortening msd stems can modulate precising editing in a retron specific manner. Modified retrons include RTX3_5752 (long (L) and short (S) versions), RTX3_6221 (long (L) and short (S) versions), and RTX3_6034 (long (L) and short (S) versions). [00142] FIG.59A Demonstrates that four retrons from various clades are capable of insertions of up to 100 bps to EMX1 locus. Figure shows the results of testing different template lengths for four different retrons in all RNA system delivered to HEK293T cells by lipofection. At given amount of RT mRNA and Cas9 mRNA, ncRNA-sgRNA fusion with different template lengths ranging from 10 to 100 bp were added. For 100 bp insertions, precise editing was 1.04% for Eco3, 1.52% for Aco1, 1.37% for R2042, and 0.05% for R6943. On the bottom row of graphs, frequency of indels is shown for respective conditions. [00143] FIG.59B Demonstrates that four retrons from various clades are capable of insertions of up to 100 bps to EMX1 locus. Figure shows indel results corresponding with the constructs of FIG.59A, graphing frequency of indels for respective conditions. [00144] FIG.60A Demonstrates that additional sgRNA targeting the same genomic locus increases the indel and insert frequencies. This figure reports same conditions as FIGs.59A-59B but with sgRNA added, which increases the frequency of precise editing and indels. For 100 bp insertions, precise editing was 1.82% for Eco3, 4.50% for Aco1, 4.40% for R2042 and 0.38% for R6943. On the bottom row of graphs, frequency of indels is shown for respective conditions. [00145] FIG.60B Demonstrates that additional sgRNA targeting the same genomic locus increases the indel and insert frequencies. This figure reports the frequency of indels associated with the retrons of FIG.60A. [00146] FIG.61A is a schematic showing unedited and edited allele by GFP gene insertion at EMX1 locus using retron editing system. Primer pair EMX1 Forward and Reverse hybridizes just outside of 5’ and 3’ homology arms and amplifies 169 bp on the unedited allele. On the edited allele, they amplify 1433 bp spanning GFP gene insertion. Primers to detect 5’ and 3’ junctions are indicated on the edited allele .5’ junction on the edited allele is amplified by primer pair: EMX1 Forward and 5’ Junction GFP Reverse and 3’ junction is amplified by primer pair: 3’ Junction GFP forward and EMX1 Reverse. Neither 5’ Junction GFP Reverse nor 3’ Junction GFP Forward primer binds to unedited allele. [00147] FIG.61B Demonstrates three retrons from various clades are capable of GFP integration at EMX1 locus. The figure provides qPCR data depicting GFP insertion at 5’ or 3’ junctions at EMX1 locus by three retrons. ncRNA-sgRNA engineered to contain GFP gene insert was transfected to HEK293T cells, together with RT mRNA of each retron, Cas9 mRNA, with/without additional sgRNA. Genomic DNA was analyzed by indicated primer pairs to detect GFP gene insertion at 5’ or 3’ junctions on the edited allele. ΔCt was obtained by subtracting +RT sample’s Ct value from –RT sample’s Ct value for relative quantification of GFP insertion at EMX1 locus. The bigger value of ΔCt indicates the higher frequency of insert in the sample. With all three retrons, amplicons of both 5’ and 3’ junctions are detectable significantly above background level. Amplicon of 3’ junction is more abundant than that of 5’ junction probably because insertion at 3’ is facilitated by more enriched 5’ end of msDNA from reverse transcription. [00148] FIG.61C Demonstrates that two retrons are capable of GFP integration at EMX1 locus. The figure provides qPCR data depicting GFP insertion at 5’ or 3’ junctions at EMX1 locus by two retrons. ncRNA-sgRNA engineered to contain GFP gene insert was transfected to HEK293T cells, together with RT mRNA of each retron, Cas9 mRNA, with/without additional sgRNA. Genomic DNA was analyzed by indicated primer pairs in the table to detect GFP gene insertion at 5’ or 3’ junctions on the edited allele. ΔCt was obtained by subtracting junction Ct value from EMX1 Ct value for relative quantification of GFP insertion at EMX1 locus. With all two retrons, amplicons of both 5’ and 3’ junctions are detectable significantly above background level. Amplicon of 3’ junction is slightly more abundant than that of 5’ junction probably because insertion at 3’ is facilitated by more enriched 5’ end of msDNA from reverse transcription. [00149] FIG.61D shows PCR products for Aco1 were run on Agilent Tapestation High Sensitivity D1000 screentape. In the presence of RT (+RT sample, left on the gel), the expected size of amplicons were detected at both 5’ and 3’ junctions of edited alleles as indicated by arrows. Edited alleles were not detected in the absence of RT (-RT sample, right on the gel). Triangles indicate the expected amplicon for unedited alleles, which is present in both -RT and +RT samples. [00150] FIG.62A Demonstrates that order of sgRNA and ncRNA within fused RNA wherein sgRNA is positioned upstream of ncRNA is more active in precise editing and indels. Results of testing different ncRNA-sgRNA fusion orders for three different retrons in all RNA system delivered to HEK293T cells by lipofection. At given amount of RT mRNA and Cas9 mRNA, ncRNA fusions with different order were added. For Eco3, Aco1 and R2042, precise editing was increased was higher with sgRNA-ncRNA compared to ncRNA-sgRNA. On the bottom row of graphs, frequency of indels is shown for respective conditions. [00151] FIG.62B Demonstrates that where sgRNA is positioned downstream of ncRNA, additional sgRNA compensates for lower editing activity of configuration. Same as previous figure but with the addition of sgRNA, In the presence of additional of sgRNA, the differences between ncRNA-sgRNA and sgRNA-ncRNA depends on each retron. For Eco3 and R2042, sgRNA-ncRNA performed better than ncRNA-sgRNA. For Aco1, ncRNA-sgRNA performed slightly better than sgRNA-ncRNA. On the bottom row of graphs, frequency of indels is shown for respective conditions. [00152] FIG.63 Demonstrates that RT and Cas9 are required for precise editing. Results of testing negative controls in all RNA system delivered to HEK293T cells by lipofection. RT and Cas9 are required for precise editing. When any one of these components is removed, precise editing is undetectable or at background levels. On the bottom graphs, frequency of indels is shown for respective conditions. [00153] FIG.64 Demonstrates that separation of ncRNA and gRNA does not result in the decreased frequency of precise edits. Results of testing ncRNA-sgRNA fusions versus separate ncRNA for four different retrons in all RNA system delivered to HEK293T cells by lipofection. At given amount of RT mRNA, Cas9 mRNA and additional sgRNA described underneath of graph, ncRNA/ncRNA-sgRNA/sgRNA-ncRNA were added. All insertion templates tested were 25 bp. The best ncRNA format depends on each retron. For Eco3, the highest precise editing was observed with separate ncRNA + additional sgRNA (9.88%). For Aco1, the highest precise editing was observed with ncRNA-sgRNA + additional sgRNA (2.96%). For R2042, the highest precise editing was observed with separate ncRNA + additional sgRNA (7.4%). For R6943, the highest precise editing was observed with ncRNA-sgRNA fusion + additional sgRNA (0.39%). On the bottom row of graphs, frequency of indels is shown for respective conditions. [00154] FIG.65 Demonstrates that retrons can achieve precise editing at AAVS1 gene locus. Results of testing four different retrons to insert 25 bp template into AAVS1 locus. Retrons were delivered as all RNA to HEK293T cells by lipofection. For all four retrons, the best precise editing was observed in the presence of additional sgRNA and ranged from 0.73% to 2.05% depending on the retron used. On the right, frequency of indels is shown for respective conditions. [00155] FIG.66 Demonstrates that retrons can achieve precise editing at AAVS1 gene locus. Results of negative controls for AAVS1 experiments in previous figure. Removing either RT, ncRNA-sgRNA or Cas9 mRNA abrogates precise editing. On the bottom row of graphs, frequency of indels is shown for respective conditions. [00156] FIG.67 Demonstrates that template for either target or non-target strand can be used for precise editing. Results of testing templates on different strands relative to Cas9 sgRNA target strand for two different retrons in all RNA system delivered to HEK293T cells by lipofection. At given amount of RT mRNA, Cas9 mRNA and additional sgRNA, ncRNA- sgRNA was added. All insertion templates tested were 25 bp. All previous figures encoded the insertion template on the same strand targeted by the Cas9 sgRNA denoted by the letter T. For Eco3, in the presence of additional sgRNA, precise editing was 1.67% when template was on the target strand and 0.85% when the template was on the non-target strand. For Aco1, in the presence of additional sgRNA, precise editing was 2.96% when template was on the target strand and 3.40% when the template was on the non-target strand. On the bottom row of graphs, frequency of indels is shown for respective conditions. [00157] FIG.68 Demonstrates that retron mediated Precise editing is observed with Cas9 nickase in 4 component all-RNA system. Cas9 nickase activity in 4 component all-RNA system (RT mRNA + Cas9 mRNA + ncRNA/ncRNA-sgRNA + sgRNA) delivered to HEK293T cells by Lipofectamine MessengerMAX. All RNA was transfected at a fixed amount RT mRNA 100 ng, ncRNA/ncRNA-sgRNA 400ng, Cas9 variant mRNA 100 ng, and sgRNA 5 ng. Either separated ncRNA or ncRNA-sgRNA fusion was used. Precise editing by Cas9 WT (Wild Type) was ~9% when either ncRNA or ncRNA-sgRNA was used with additional sgRNA. With Cas9 H480A mutant that nicks non-target strand, activity was barely seen above background in any condition. With Cas9 D10A mutant that nicks target strand, precise editing was observed ~0.1% frequency in both separated and fused ncRNA and sgRNA format. Indels are shown at the right graph at respective conditions of left graph. As expected, indels are extremely low for either nickases with or without additional sgRNA and only WT Cas9 generated significant indels. [00158] FIG.69. Demonstrates that retron mediated precise editing by Cas9 nickase is dependent on each component of all RNA system. Same as previous figure but testing negative controls that lack one of components (Cas9 or ncRNA/ncRNA-sgRNA or RT) of all RNA system with or without additional sgRNA. All control conditions tested show background precise editing activity in the top graph suggesting that precise editing achieved by Cas9 WT or Cas9 nickase is dependent on each component of all RNA system. Significant indels are detectable only with Cas9 WT in the bottom graph. [00159] FIG.70 demonstrates that dual sgRNAs increases precise editing, but not indels. The figure shows the results of testing dual sgRNAs for R2042 in all RNA system delivered to HEK293T cells by lipofection. At given amount of RT mRNA, Cas9 mRNA and ncRNA- sgRNA, additional sgRNA was added. The same ncRNA-sgRNA with template encoded a 25 bp insertion on the Cas9 sgRNA’s non-targeting strand was used for all conditions. With one additional sgRNA (#1), precise editing was 0.27%, which increased to either 1.33% (#2) or 1.13% (#3) in the presence of a second sgRNA. On the bottom graph, frequency of indels is shown for respective conditions. See Example 9. DEFINITIONS [00160] All technical and scientific terms used herein have the meaning commonly understood by a person skilled in the art to which this invention belongs. The following references provide one of skill with a general definition of many of the terms used in this invention: Singleton et al., Dictionary of Microbiology and Molecular Biology (2nd ed.1994); The Cambridge Dictionary of Science and Technology (Walker ed., 1988); The Glossary of Genetics, 5th Ed., R. Rieger et al. (eds.), Springer Verlag (1991); and Hale & 62/1005 Marham, The Harper Collins Dictionary of Biology (1991). [00161] General methods in molecular and cellular biochemistry can be found in such standard textbooks as Molecular Cloning: A Laboratory Manual, 3rd Ed. (Sambrook et al., HaRBor Laboratory Press 2001); Short Protocols in Molecular Biology, 4th Ed. (Ausubel et al. eds., John Wiley & Sons 1999); Protein Methods (Bollag et al., John Wiley & Sons 1996); Nonviral Vectors for Gene Therapy (Wagner et al. eds., Academic Press 1999); Viral Vectors (Kaplift & Loewy eds., Academic Press 1995); Immunology Methods Manual (I. Lefkovits ed., Academic Press 1997); and Cell and Tissue Culture: Laboratory Procedures in Biotechnology (Doyle & Griffiths, John Wiley & Sons 1998), the disclosures of which are incorporated herein by reference. [00162] Where a range of values is provided, it is understood that each intervening value, to the tenth of the unit of the lower limit unless the context clearly dictates otherwise, between the upper and lower limit of that range and any other stated or intervening value in that stated range, is encompassed within the disclosure. The upper and lower limits of these smaller ranges may independently be included in the smaller ranges, and are also encompassed within the disclosure, subject to any specifically excluded limit in the stated range. Where the stated range includes one or both of the limits, ranges excluding either or both of those included limits are also included in the disclosure. [00163] It must be noted that as used herein and in the appended claims, the singular forms “a,” “an,” and “the” include plural referents unless the context clearly dictates otherwise. Thus, for example, reference to “a ncRNA” includes a plurality of ncRNAs and reference to “the reverse transcriptase” includes reference to one or more RTs and equivalents thereof known to those skilled in the art, and so forth. It is further noted that the claims may be drafted to exclude any optional element. As such, this statement is intended to serve as antecedent basis for use of such exclusive terminology as “solely,” “only” and the like in connection with the recitation of claim elements, or use of a “negative” limitation. For example, claims may be drafted to exclude certain RT sequences. [00164] It is appreciated that certain features of the invention, which are, for clarity, described in the context of separate embodiments, may also be provided in combination in a single embodiment. Conversely, various features of the invention, which are, for brevity, described in the context of a single embodiment, may also be provided separately or in any suitable sub-combination. All combinations of the embodiments pertaining to the invention are specifically embraced by the present invention and are disclosed herein just as if each and every combination was individually and explicitly disclosed. In addition, all sub-combinations of the various embodiments and elements thereof are also specifically embraced by the present invention and are disclosed herein just as if each and every such sub combination was individually and explicitly disclosed herein. Biologically active [00165] As used herein, the term “biologically active” refers to a characteristic of an agent (e.g., DNA, RNA, or protein) that has activity in a biological system (including in vitro and in vivo biological system), and particularly in a living organism, such as in a mammal, including human and non-human mammals. For instance, an agent when administered to an organism has a biological effect on that organism, is considered to be biologically active. Bulge [00166] As used herein, the term “bulge” refers to a small region of unpaired base(s) that interrupts a “stem” of base-paired nucleotides. The bulge may comprise one or two single- stranded or unbase-paired nucleotides joined at both ends by base-paired nucleotides of the stem. The bulge can be symmetrical (viz., the two unbase-paired single-stranded regions have the same number of nucleotides), or asymmetrical (viz., the unbase-paired single stranded region(s) have different or unequal numbers of nucleotides), or there is only one unbase-paired nucleotide on one strand. A bulge can be described as A/B (such as a “2/2 bulge,” or a “1/0 bulge”) wherein A represents the number of unpaired nucleotides on the upstream strand of the stem, and B represents the number of unpaired nucleotides on the downstream strand of the stem. An upstream strand of a bulge is more 5ʹ to a downstream strand of the bulge in the primary nucleotide sequence. cDNA [00167] As used hereing, the term “cDNA” refers to a strand of DNA copied from an RNA template, e.g., by a reverse transcriptase. Complementary [00168] As used herein, the terms “complementary” or “substantially complementary" are meant to refer to a nucleic acid (e.g., RNA, DNA) that comprises a sequence of nucleotides that enables it to non-covalently bind, i.e., form Watson-Crick base pairs and/or G/U base pairs, “anneal”, or “hybridize,” to another nucleic acid in a sequence-specific, antiparallel, manner (i.e., a nucleic acid specifically binds to a complementary nucleic acid) under the appropriate in vitro and/or in vivo conditions of temperature and solution ionic strength. Standard Watson-Crick base-pairing includes: adenine (A) pairing with thymidine (T), adenine (A) pairing with uracil (U), and guanine (G) pairing with cytosine (C) [DNA, RNA]. In addition, for hybridization between two RNA molecules (e.g., dsRNA), and for hybridization of a DNA molecule with an RNA molecule (e.g., when a DNA target nucleic acid base pairs with a guide RNA, etc.): guanine (G) can also base pair with uracil (U). For example, G/U base-pairing is at least partially responsible for the degeneracy (i.e., redundancy) of the genetic code in the context of tRNA anti- codon base-pairing with codons in mRNA. Thus, in the context of this disclosure, a guanine (G) is considered complementary to both a uracil (U) and to an adenine (A). For example, when a G/U base-pair can be made at a given nucleotide position of a dsRNA duplex of a guide RNA molecule, the position is not considered to be non-complementary, but is instead considered to be complementary. [00169] It is understood that the sequence of a polynucleotide need not be 100% complementary to that of its target nucleic acid to be specifically hybridizable or hybridizable. Moreover, a polynucleotide may hybridize over one or more segments such that intervening or adjacent segments are not involved in the hybridization event (e.g., a bulge, a loop structure or hairpin structure, etc.). A polynucleotide can comprise 60% or more, 65% or more, 70% or more, 75% or more, 80% or more, 85% or more, 90% or more, 95% or more, 98% or more, 99% or more, 99.5% or more, or 100% sequence complementarity to a target region within the target nucleic acid sequence to which it will hybridize. For example, an antisense nucleic acid in which 18 of 20 nucleotides of the antisense compound are complementary to a target region, and would therefore specifically hybridize, would represent 90 percent complementarity. In this example, the remaining noncomplementary nucleotides may be clustered or interspersed with complementary nucleotides and need not be contiguous to each other or to complementary nucleotides. Percent complementarity between particular stretches of nucleic acid sequences within nucleic acids can be determined using any convenient method. Example methods include BLAST programs (basic local alignment search tools) and PowerBLAST programs (Altschul et al., J. Mol. Biol., 1990, 215, 403-410; Zhang and Madden, Genome Res., 1997, 7, 649-656), the Gap program (Wisconsin Sequence Analysis Package, Version 8 for Unix, Genetics Computer Group, University Research Park, Madison Wis.), e.g., using default settings, which uses the algorithm of Smith and Waterman (Adv. Appl. Math., 1981, 2, 482-489), and the like. DNA-guided nuclease [00170] As used herein, an “DNA-guided nuclease” is a type of “programmable nuclease,” and a specific type of “nucleic acid-guided nuclease.” An example of a DNA-guided nuclease is reported in Varshney et al., DNA-guided genome editing using structure-guided endonucleases, Genome Biology, 2016, 17(1), 187, which may be used in the context of the present disclosure and is incorporated herein by reference. As used herein, the term “DNA- guided nuclease” or “DNA-guided endonuclease” refers to a nuclease that associates covalently or non-covalently with a guide RNA thereby forming a complex between the guide RNA and the DNA-guided nuclease. The guide RNA comprises a spacer sequence which comprises a nucleotide sequence having complementarity with a strand of a target DNA sequence. Thus, the DNA-guided nuclease is indirectly guided or programmed to localize to a specific site in a DNA molecule through its association with the guide RNA, which directly binds or anneals to a strand of the target DNA through its complementarity region via Watson-Crick base-pairing. DNA regulatory sequences [00171] As used herein, the terms “DNA regulatory sequences,” “control elements,” and “regulatory elements,” can be used interchangeably herein to refer to transcriptional and translational control sequences, such as promoters, enhancers, polyadenylation signals, terminators, protein degradation signals, and the like, that provide for and/or regulate transcription of a non-coding sequence (e.g., guide RNA) or a coding sequence and/or regulate translation of a mRNA into an encoded polypeptide. Donor nucleic acid [00172] By a “donor nucleic acid” or “donor polynucleotide” or “donor DNA” or “HDR donor DNA” it is meant a single-stranded DNA to be inserted at a site cleaved by a programmable nuclease (e.g., a CRISPR/Cas effector protein; a TALEN; a ZFN; a meganuclease) (e.g., after dsDNA cleavage, after nicking a target DNA, after dual nicking a target DNA, and the like). The donor polynucleotide can contain sufficient homology to a genomic sequence at the target site, e.g.70%, 80%, 85%, 90%, 95%, or 100% homology with the nucleotide sequences flanking the target site, e.g., within about 200 bases or less of the target site, e.g., within about 190 bases or less of the target site, e.g., within about 180 bases or less of the target site, e.g., within about 170 bases or less of the target site, e.g., within about 160 bases or less of the target site, e.g., within about 150 bases or less of the target site, e.g., within about 140 bases or less of the target site, e.g., within about 130 bases or less of the target site, e.g., within about 120 bases or less of the target site, e.g., within about 110 bases or less of the target site, e.g., within about 100 bases or less of the target site, e.g., within about 90 bases or less of the target site, e.g., within about 80 bases or less of the target site, e.g., within about 70 bases or less of the target site, e.g., within about 60 bases or less of the target site, e.g., 50 bases or less of the target site, e.g., within about 30 bases, within about 15 bases, within about 10 bases, within about 5 bases, or immediately flanking the target site, to support homology-directed repair between it and the genomic sequence to which it bears homology. Encodes [00173] As used herein, a DNA sequence that “encodes” a particular RNA is a DNA nucleotide sequence that is transcribed into RNA. A DNA polynucleotide may encode an RNA (mRNA) that is translated into protein (and therefore the DNA and the mRNA both encode the protein), or a DNA polynucleotide may encode an RNA that is not translated into protein (e.g. tRNA, rRNA, microRNA (miRNA), a “non-coding” RNA (ncRNA), a guide RNA, etc.). In the case of retrons, the retron DNA may encode the ncRNA loci (which includes the msr and msd regions) as well as a retron RT. Engineered retron [00174] As used herein, the term “engineered retron” or equivalently, “recombinant retron,” refers to a retron that does not occur in nature. In one embodiment, engineered retrons can include wildtype or naturally-occurring retrons that are modified to contain at least one modification, including a single nucleotide substitution, insertion, or deletion, or a substitution, insertion, or deletion of more than one nucleotide, i.e., up to 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, 69, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, or up to 100, or up to 200, 300, 400, 500, 600, 700, 800, 900, 1000, 1100, 1200, 1300, 1400, 1500, 1600, 1700, 1800, 1900, or up to 2000 nucleotides substituted, inserted, or deleted from a starting point retron (e.g., a wildtype retron). Where more than one nucleotide of a starting point retron (e.g., a wildtype retron) is substituted, deleted, or inserted, the nucleotides may be contiguous or non-contiguous. While an engineered retron as a whole is not naturally-occurring, it may include components such as nucleotide sequences that do occur in nature. For example, an engineered retron can have nucleotide sequences from different organisms (e.g., from different bacteria species), or from completely synthetic / artificial / recombinant nucleic acid sequences. Thus, an engineered retron can have a bacterial nucleotide sequence, a human nucleotide sequence, a viral nucleotide sequence, and/or a synthetic / artificial / recombinant nucleotide sequence, and/or combinations of such sequences. An example of modifications of the recombinant retrons disclosed herein include the insertion of a heterologous nucleic acid sequence in a retron, for example, inserted into the ncRNA locus, such as in the msr or the msd loci. Linking guide RNA molecules to the 5ʹ and/or 3ʹ ends (i.e., linking one at the 5ʹ end of a ncRNA and/or one at the 3ʹ end of a ncRNA) also represent another modification envisioned by the recombinant retrons disclosed herein. In such embodiments, the guide RNA molecules may also be categorized or referred to more generally as types of heterologous nucleic acid sequences used to modify starting point retrons. Exosomes [0001] As used herein, the term “exosomes” refer to small membrane bound vesicles with an endocytic origin. Without wishing to be bound by theory, exosomes are generally released into an extracellular environment from host/progenitor cells post fusion of multivesicular bodies the cellular plasma membrane. As such, exosomes can include components of the progenitor membrane in addition to designed components (e.g. engineered retron). Exosome membranes are generally lamellar, composed of a bilayer of lipids, with an aqueous inter-nanoparticle space. Expression vector [0002] As used herein, the term “expression vector” or “expression construct” refers to a vector that includes one or more expression control sequences, and an “expression control sequence” is a DNA sequence that controls and regulates the transcription and/or translation of another DNA sequence. Suitable expression vectors include, without limitation, plasmids and viral vectors derived from, for example, bacteriophage, baculoviruses, tobacco mosaic virus, herpes viruses, cytomegalovirus, retroviruses, vaccinia viruses, adenoviruses, and adeno- associated viruses. Numerous vectors and expression systems are commercially available, such as from Novagen (Madison, WI), Clontech (Palo Alto, CA), Stratagene (La Jolla, CA), and Invitrogen/Life Technologies (Carlsbad, CA). The present invention comprehends recombinant vectors that may include viral vectors, bacterial vectors, protozoan vectors, DNA vectors, or recombinants thereof. Heterologous nucleic acid sequence [00175] As used herein, the term “heterologous nucleic acid” refers to a genotypically distinct entity from that of the rest of the entity to which it is compared or into which it is introduced or incorporated. For example, a polynucleotide introduced by genetic engineering techniques into a different cell type is a heterologous polynucleotide (e.g., DNA or RNA) and, if expressed, can encode a heterologous polypeptide. Similarly, a cellular sequence (e.g., a gene or portion thereof) that is incorporated into a viral vector is a heterologous nucleotide sequence with respect to the vector. In some embodiments, the heterologous sequence inserted into the wild- type retron regions does not naturally insert into such regions (e.g., the engineered retron with the inserted heterologous sequence is not naturally existing). For example, the heterologous sequence can be from the same species of bacteria in which the wild-type retron is normally found, so long as the heterologous sequence is not naturally inserted in the wild-type retron at the location in which the heterologous sequence is inserted. In certain embodiments, the heterologous sequence is a mammalian sequence (e.g., a human sequence), or a reverse complement thereof. Heterologous nucleic acid sequences introduced into retrons can including without limitation guide RNA sequences, HDR donor templates, protein-encoding genes, or non- coding functional RNA elements (e.g., stem-loops, hairpins, and bulges). Lipid nanoparticle (LNP) [00176] As used herein, the term “lipid nanoparticle” or LNP refers to a type of lipid particle delivery system formed of small solid or semi-solid particles possessing an exterior lipid layer with a hydrophilic exterior surface that is exposed to the non-LNP environment, an interior space which may aqueous (vesicle like) or non-aqueous (micelle like), and at least one hydrophobic inter-membrane space. LNP membranes may be lamellar or non-lamellar and may be comprised of 1, 2, 3, 4, 5 or more layers. In some embodiments, LNPs may comprise a nucleic acid (e.g. engineered retron) into their interior space, into the inter membrane space, onto their exterior surface, or any combination thereof. In some embodiments, an LNP of the present disclosure comprises an ionizable lipid, a structural lipid, a PEGylated lipid (aka PEG lipid), and a phospholipid. In alternative embodiments, an LNP comprises an ionizable lipid, a structural lipid, a PEGylated lipid (aka PEG lipid), and a zwitterionic amino acid lipid. [00177] Further discuss of liposomes can be found, for example, in Tenchov et al., “Lipid Nanoparticles – From Liposomes to mRNA Vaccine Delivery, a Landscape of Diversity and Advancement,” ACS Nano, 2021, 15, pp.16982-17015 (the contents of which are incorporated by reference). Linker [00178] As used herein, the term“linker” refers to a molecule linking or joining two other molecules or moieties. The linker can be an amino acid sequence in the case of a linker joining two fusion proteins. For example, an RNA-guided nuclease (e.g., Cas12a) can be fused to a retron reverse transcriptase by an amino acid linker sequence. The linker can also be a nucleotide sequence in the case of joining two nucleotide sequences together. For example, in the instant case, a ncRNA at its 5ʹ and/or 3ʹ ends may be linked by a nucleotide sequence linker to one or more guide RNAs. In other embodiments, the linker is an organic molecule, group, polymer, or chemical moiety. In some embodiments, the linker is 5-100 amino acids in length, for example, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 30- 35, 35-40, 40- 45, 45-50, 50-60, 60-70, 70-80, 80-90, 90-100, 100-150, or 150-200 amino acids in length. Longer or shorter linkers are also contemplated. Liposomes [00179] As used herein, the term “liposomes” refer to a type of lipid particle delivery system comprising small vesicles that contain at least one lipid membrane surrounding an aqueous inner-nanoparticle space that is generally not derived from a progenitor/host cell. Liposomes are a versatile carrier platform in that they are capable of transporting hydrophobic or hydrophilic molecules, including small molecules, proteins, and nucleic acids into cells. They were the earliest developed generation of nanoscale medicine delivery platform. Numerous liposomal drug formulations have been approved for human medicines, e.g., Doxil, a lipid nanoparticle formulation of the antitumor agent doxorubicin. Further discuss of liposomes can be found, for example, in Tenchov et al., “Lipid Nanoparticles – From Liposomes to mRNA Vaccine Delivery, a Landscape of Diversity and Advancement,” ACS Nano, 2021, 15, pp. 16982-17015 (the contents of which are incorporated by reference). Loop [0001] As used herein, the term “loop” in a polynucleotide refers to a single stranded stretch of one or more nucleotides, such as 2, 3, 4, 5, 6, 7, 8, 9, or 10 nucleotides, wherein the most 5ʹ nucleotide and the most 3ʹ nucleotide of the loop are each linked to a base- paired nucleotide in a stem. Micelles [00180] As used herein, the term “micelles” refer to small particles which do not have an aqueous intra-particle space. Nanoparticle [00181] As used herein, the term “nanoparticle” refers to any nanoscale particle typically ranging in size from about 1 nm to 1000 nm. Nuclear localization sequence (NLS) [00182] As used herein, the term“nuclear localization sequence” or“NLS” refers to an amino acid sequence that promotes import of a protein (e.g., a RNA-guided nuclease) into the cell nucleus, for example, by nuclear transport. Nuclear localization sequences are known in the art. For example, NLS sequences are described in Plank et al., international PCT application, PCT/EP2000/011690, filed November 23, 2000, published as WO/2001/038547 on May 31, 2001, the contents of which are incorporated herein by reference for its disclosure of exemplary nuclear localization sequences. Nucleic acid [00183] As used herein, the term “nucleic acid” or “nucleic acid molecule” or “nucleic acid sequence” or “polynucleotide” generally refer to deoxyribonucleic or ribonucleic oligonucleotides in either single- or double-stranded form. The term may also encompass oligonucleotides containing known analogues of natural nucleotides. The term also may also encompass nucleic acid-like structures with synthetic backbones, see, e.g., Eckstein, 1991; Baserga et al., 1992; Milligan, 1993; WO 97/03211; WO 96/39154; Mata, 1997; Strauss-Soukup, 1997; and Samstag, 1996. The term encompasses both ribonucleic acid (RNA) and DNA, including cDNA (including RT DNA), genomic DNA, synthetic, synthesized (e.g., chemically synthesized) DNA, and/or DNA (or RNA) containing nucleic acid analogs. The nucleotides Adenine (A), Thymine (T), Guanine (G) and Cytosine (C) also may (or may not) encompass nucleotide modifications, e.g., methylated and/or hydroxylated nucleotides, e.g., Cytosine (C) encompasses 5-methylcytosine and 5- hydroxymethylcytosine. Nucleic acid-guided nuclease [00184] As used herein, the term “nucleic acid-guided nuclease” or “nucleic acid-guided endonuclease” refers to a nuclease that associates covalently or non-covalently with a guide nucleic acid (e.g., a guide RNA or a guide DNA) thereby forming a complex between the guide nucleic acid and the nucleic acid-guided nuclease. The guide nucleic acid comprises a spacer sequence which comprises a nucleotide sequence having complementarity with a strand of a target DNA sequence. Thus, the nucleic acid-guided nuclease is indirectly guided or programmed to localize to a specific site in a DNA molecule through its association with the guide nucleic acid, which directly binds or anneals to a strand of the target DNA through its complementarity region via Watson-Crick base-pairing. In some embodiments, the nucleic acid- guided nuclease will include a DNA-binding activity (e.g., as in the case for CRISPR Cas9). Most commonly, the nucleic acid-guided nuclease is programmed by associating with a guide RNA molecule and in such cases the nuclease may be called “RNA-guided nuclease.” When programmed by a guide DNA, the nuclease may be called a “DNA-guided nuclease.” Nucleic acid-guided, RNA-guided, or DNA-guided nucleases may also be referred to as “programmable nucleases,” which also include other classes of programmable nucleases which associate with specific DNA sequences through amino acid / nucleotide sequence recognition (e.g., zinc fingers nucleases (ZFN) and transcription activator like effector nucleases (TALEN)) rather than through guide RNAs. In addition, any nuclease contemplated herein may also be engineered to remove, inactivate, or otherwise eliminate one or more nuclease activities (e.g., by introducing a nuclease-inactiving mutation in the active site(s) of a nuclease). A nuclease that has been modified to remove, inactivate, or otherwise eliminate all nuclease activity may be referred to as a “dead” nuclease. A dead nuclease is not able to cut either strand of a double-stranded DNA molecule. A nuclease that has been modified to remove, inactivate, or otherwise eliminate at least one nuclease activity but which still retains at least one nuclease activity may be referred to as a “nickase” nuclease. A nickase nuclease cuts one strand of a double-stranded DNA molecule, but not both strands. For example, a CRISPR Cas9 naturally comprises two distinct nuclease activity domains, namely, the HNH domain and the RuvC domain. The HNH domain cuts the strand of DNA bound to the guide RNA and the RuvC domain cuts the protospacer strand. One can obtain a nickase Cas9 by inactivating either the HNH domain or the RuvC domain. One can obtain a dead Cas9 by inactivating both the HNH domain and the RuvC domain. Other RNA-guided nuclease may be similarly converted to nickases and/or dead nucleases by inactivating one or more of the existing nuclease domains.
Operably linked
[00185] As used herein, the term “operably linked” or “under transcriptional control,” when used in conjunction with the description of a promoter, refers to the correct location and orientation in relation to a polynucleotide (e.g., a coding sequence) to control the initiation of transcription by RNA polymerase and expression of the coding sequence, such as one for the msr gene, msd gene, and/or the ret gene. Other transcriptional control regulatory elements (e.g., enhancer sequences, transcription factor binding sites) may also be operably linked to a gene if their location relative to a gene controls or regulates the expression of the gene.
Programmable nuclease
[00186] As used herein, the term “programmable nuclease” is meant to refer to a polypeptide that has the property of selective localization to a specific desired nucleotide sequence target in a nucleic acid molecule (e.g., to a specific gene target) due to one or more targeting functions. Such targeting functions can include one or more DNA-binding domains, such as zinc finger domains characteristic of many different types of DNA binding proteins or TALE domains characteristic of TALEN proteins. Such targeting function may also include the ability to associate and/or form a complex with a guide RNA, which then localizes to a specific site on the DNA which bears a sequence that is complementary to a portion of the guide RNA (i.e., the spacer of the guide RNA). In some embodiments, the programmable nuclease may be a single protein which comprises both a domain that binds directly (e.g., a ZF protein) or indirectly (e.g., an RNA-guided protein) to a target DNA site, as well as a nuclease domain. In other embodiments, the programmable nuclease may be a composite of two or more separate proteins or domains (from different proteins) which together provide the necessary functions of selective DNA binding and nuclease activity. For example, the programmable nuclease may comprise a (a) nuclease-inactive RNA-guided nuclease (which still is capable of binding a guide RNA, localizing to a target DNA, and binding to the target DNA, but not capable of cutting or nicking the strands) fused to a (b) nuclease protein or domain, such as a FokI nuclease.
Promoter [00187] As used herein, the term“promoter” is art-recognized and refers to a nucleic acid molecule with a sequence recognized by the cellular transcription machinery and which is able to initiate transcription of a downstream gene. A promoter can be constitutively active, meaning that the promoter is always active in a given cellular context, or conditionally active, meaning that the promoter is only active in the presence of a specific condition. For example, a conditional promoter may only be active in the presence of a specific protein that connects a protein associated with a regulatory element in the promoter to the basic transcriptional machinery, or only in the absence of an inhibitory molecule. Within the promoter sequence will be found a transcription initiation site, as well as protein binding domains responsible for the binding of RNA polymerase. Eukaryotic promoters will often, but not always, contain “TATA” boxes and “CAT” boxes. Various promoters, including inducible promoters, may be used to drive expression by the various vectors of the present disclosure. Recombinant nucleic acid [00188] A “recombinant nucleic acid” or “recombinant nucleotide” refers to a molecule that is constructed by joining nucleic acid molecules, which optionally may self-replicate in a live cell. Retron [00189] As used herein, the term “retron” refers to a specific type of naturally-occuring and distinct DNA sequence found in the genome of many bacteria which typically encodes three distinct components, namely, (a) a non-coding RNA (“ncRNA”) (comprising continguous inverted sequences (msr and msd), (b) a reverse transcriptase (RT)-coding gene (ret), and (c) in many cases, a retron-associated gene of unknown function. Retrons are particularly defined by their unique ability to produce a satellite DNA known as msDNA (multicopy single-stranded DNA). The ncRNA (comprising the msr and msd elements) and the ret gene are transcribed as a single polycitronic RNA transcript which processed into the ncRNA transcript and a transcript encoding the ret gene. The ncRNA then becomes folded into a specific secondary structure. Once translated, the RT then binds the folded ncRNA and reverse transcribes the msd region to form a single strand of cDNA (the msDNA) that remains covalently attached to the RNA template via a 2’-5ʹ phophodiester bond and base-pairing between the 3ʹ ends of the msDNA and the RNA template. See FIG.1A which provides a schematic of the production of an msDNA from a naturally-occurring retron. Retron component [00190] As used herein, the term “retron component” refers to a distinct element or feature of a retron, namely (a) a non-coding RNA (“ncRNA”) (comprising continguous inverted sequences (msr and msd), (b) a reverse transcriptase (RT)-coding gene (ret), and (c) in many cases, a retron-associated gene of unknown function. RNA-guided nuclease [00191] As used herein, an “RNA-guided nuclease” is a type of “programmable nuclease,” and a specific type of “nucleic acid-guided nuclease.” As used herein, the term “RNA-guided nuclease” or “RNA-guided endonuclease” refers to a nuclease that associates covalently or non- covalently with a guide RNA thereby forming a complex between the guide RNA and the RNA- guided nuclease. The guide RNA comprises a spacer sequence which comprises a nucleotide sequence having complementarity with a strand of a target DNA sequence. Thus, the RNA- guided nuclease is indirectly guided or programmed to localize to a specific site in a DNA molecule through its association with the guide RNA, which directly binds or anneals to a strand of the target DNA through its complementarity region via Watson-Crick base-pairing. Sequence identity [00192] As used herein, the term “sequence identity” refers to the overall relatedness between polymeric molecules, e.g., between polynucleotide molecules (e.g., DNA molecules and/or RNA molecules) and/or between polypeptide molecules. Calculation of the percent identity of two polynucleotide sequences, for example, can be performed by aligning the two sequences for optimal comparison purposes (e.g., gaps can be introduced in one or both of a first and a second nucleic acid sequences for optimal alignment and non-identical sequences can be disregarded for comparison purposes). For example, the length of a sequence aligned for comparison purposes is at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, at least 95%, or 100% of the length of the reference sequence. The nucleotides at corresponding nucleotide positions are then compared. When a position in the first sequence is occupied by the same nucleotide as the corresponding position in the second sequence, then the molecules are identical at that position. The percent identity between the two sequences is a function of the number of identical positions shared by the sequences, taking into account the number of gaps, and the length of each gap, which needs to be introduced for optimal alignment of the two sequences. The comparison of sequences and determination of percent identity between two sequences can be accomplished using a mathematical algorithm. For example, the percent identity between two nucleotide sequences can be determined using methods such as those described in Computational Molecular Biology, Lesk, A. M., ed., Oxford University Press, New York, 1988; Biocomputing: Informatics and Genome Projects, Smith, D. W., ed., Academic Press, New York, 1993; Sequence Analysis in Molecular Biology, von Heinje, G., Academic Press, 1987; Computer Analysis of Sequence Data, Part I, Griffin, A. M., and Griffin, H. G., eds., Humana Press, New Jersey, 1994; and Sequence Analysis Primer, Gribskov, M. and Devereux, J., eds., M Stockton Press, New York, 1991; each of which is incorporated herein by reference. For example, the percent identity between two nucleotide sequences can be determined using the algorithm of Meyers and Miller (CABIOS, 1989, 4:11- 17), which has been incorporated into the ALIGN program (version 2.0) using a PAM120 weight residue table, a gap length penalty of 12 and a gap penalty of 4. The percent identity between two nucleotide sequences can, alternatively, be determined using the GAP program in the GCG software package using an NWSgapdna. CMP matrix. Methods commonly employed to determine percent identity between sequences include, but are not limited to those disclosed in Carillo, H. and Lipman, D., SIAM J Applied Math., 48:1073 (1988); incorporated herein by reference. Techniques for determining identity are codified in publicly available computer programs. Exemplary computer software to determine homology between two sequences include, but are not limited to, GCG program package, Devereux, J., et al., Nucleic Acids Research, 12(1), 387 (1984)), BLASTP, BLASTN, and FASTA Altschul, S. F. et al., J. Molec. Biol., 215, 403 (1990). Subject [00193] As used herein, the term“subject” refers to an individual organism, for example, an individual mammal. In some embodiments, the subject is a human. In some embodiments, the subject is a non-human mammal. In some embodiments, the subject is a non-human primate. In some embodiments, the subject is a rodent. In some embodiments, the subject is a sheep, a goat, a cattle, a cat, or a dog. In some embodiments, the subject is a vertebrate, an amphibian, a reptile, a fish, an insect, a fly, or a nematode. In some embodiments, the subject is a research animal. In some embodiments, the subject is genetically engineered, e.g., a genetically engineered non- human subject. The subject may be of either sex and at any stage of development. The terms “individual,” “subject,” “host,” and “patient,” used interchangeably herein. Stem [00194] As used herein, the term “stem” refers to two or more base pairs, such as 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20 or more base pairs, formed by inverted repeat sequences connected at a “tip,” where the more 5ʹ or “upstream” strand of the stem bends to allows the more 3ʹ or “downstream” strand to base-pair with the upstream strand. The number of base pairs in a stem is the “length” of the stem. The tip of the stem is typically at least 3 nucleotides, but can be 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15 or more nucleotides. Larger tips with more than 5 nucleotides are also referred to as a “loop.” An otherwise continuous stem may be interrupted by one or more bulges as defined herein. The number of unpaired nucleotides in the bulge(s) are not included in the length of the stem. The position of a bulge closest to the tip can be described by the number of base pairs between the bulge and the tip (e.g., the bulge is 4 bps from the tip). The position of the other bulges (if any) further away from the tip can be described by the number of base pairs in the stem between the bulge in question and the tip, excluding any unpaired bases of other bulges in between. Synthetic or artificial nucleic acid [00195] A “synthetic or artificial nucleic acid” refers nucleic acids that are non-naturally occurring sequences. Such sequences do not originate from, or are not known to be present in any living organism (e.g., based on sequence search in existing sequence databases). Recombinant nucleic acids and synthetic nucleic acids also include those molecules that result from the replication of either of the foregoing. Engineered nucleic acid constructs of the present disclosure, such as the engineer ed retron described herein, may be encoded by a single molecule (e.g., encoded by or present on the same plasmid or other suitable vector) or by multiple different molecules (e.g., multiple independently-replicating vectors). Target site [00196] As used herein, a “target site” as used herein is a polynucleotide (e.g., DNA such as genomic DNA) that includes a site or specific locus (“target site” or “target sequence”) targeted by a recombinant retron genome modification system disclosed herein. In the context of retron genome modification systems disclosed herein that comprise an RNA-guided nuclease, a target sequence is the sequence to which the guide sequence of a guide nucleic acid (e.g., guide RNA) will hybridize. For example, the target site (or target sequence) 5ʹ-GTCAATGGACC-3ʹ within a target nucleic acid is targeted by (or is bound by, or hybridizes with, or is complementary to) the sequence 5ʹ-GGTCCATTGAC-3ʹ. Suitable hybridization conditions include physiological conditions normally present in a cell. For a double stranded target nucleic acid, the strand of the target nucleic acid that is complementary to and hybridizes with the guide RNA is referred to as the “complementary strand” or “target strand”; while the strand of the target nucleic acid that is complementary to the “target strand” (and is therefore not complementary to the guide RNA) is referred to as the “non-target strand” or “non- complementary strand.” Treatment [00197] As used herein, the terms“treatment,” “treat,” and“treating,” refer to a clinical intervention aimed to reverse, alleviate, delay the onset of, or inhibit the progress of a disease or disorder, or one or more symptoms thereof, as described herein. In some embodiments, treatment may be administered after one or more symptoms have developed and/or after a disease has been diagnosed. In other embodiments, treatment may be administered in the absence of symptoms, e.g., to prevent or delay onset of a symptom or inhibit onset or progression of a disease. For example, treatment may be administered to a susceptible individual prior to the onset of symptoms (e.g., in light of a history of symptoms and/or in light of genetic or other susceptibility factors). Treatment may also be continued after symptoms have resolved, for example, to prevent or delay their recurrence. Upstream and downstream [00198] As used herein, the terms “upstream” and “downstream” are terms of relativity that define the linear position of at least two elements located in a nucleic acid molecule (whether single or double-stranded) that is orientated in a 5ʹ-to-3ʹ direction. A first element is said to be upstream of a second element in a nucleic acid molecule where the first element is positioned somewhere that is 5ʹ to the second element. Conversely, a first element is downstream of a second element in a nucleic acid molecule where the first element is positioned somewhere that is 3ʹ to the second element. Variant [00199] As used herein the term“variant” should be taken to mean the exhibition of qualities that have a pattern that deviates from what occurs in nature, e.g., a variant retron RT is retron RT comprising one or more changes in amino acid residues as compared to a wild type retron RT amino acid sequence. The term“variant” encompasses homologous proteins having at least 75%, or at least 80%, or at least 85%, or at least 90%, or at least 95%, or at least 99% percent identity with a reference sequence and having the same or substantially the same functional activity or activities as the reference sequence. The term also encompasses mutants, truncations, or domains of a reference sequence, and which display the same or substantially the same functional activity or activities as the reference sequence. Vector [00200] As used herein, the term “vector” permits or facilitates the transfer of a polynucleotide from one environment to another. It is a replicon such as a plasmid, phage, or cosmid into which another DNA segment may be inserted so as to bring about the replication of the inserted segment (e.g., the subject engineered retron). Generally, a vector is capable of replication when associated with the proper control elements. The term “vector” may include cloning and expression vectors, as well as viral vectors and integrating vectors. Wild type [00201] As used herein the term “wild type” is a term of the art understood by skilled persons and means the typical form of an organism, strain, gene, protein, or characteristic as it occurs in nature as distinguished from mutant or variant forms. DETAILED DESCRIPTION [00202] The present disclosure provides systems, methods and compositions used for precise genome editing, including installing nucleic acid insertions, replacements, and deletions at targeted and precise genome sites, wherein said systems, methods, and compositions are based on novel and/or modified retrons or components thereof, such as modified versions of the retron RTs of Table X, modified versions of the ncRNAs of Table A, and modified versions of the RTs of Table B. [00203] In one aspect, the present disclosure provides recombinant retrons comprising one or more genetic modifications which improves the functionality and/or properties of a retron. Such genetic modifications can include a mutation, insertion, deletion, inversion, replacement, substitution, or translocation of one or more contiguous or non-contiguous nucleobases in a nucleic acid molecule encoding a retron or a component of a retron, such as an ncRNA or a reverse transcriptase. In various aspects, the retron that becomes modifed with the one or more genetic modifications (i.e., the “pre-modified” or “unmodified” retron or retron component) is a naturally occurring retron or retron component (e.g., naturally occuring ncRNA of Table A or RT) ability to facilitate homology-dependent recombination (or HDR) in a cell, thereby resulting in a relative increase in the concentrations or amounts of msDNA comprising a DNA donor template. In particular embodiments, the recombinant retrons are based on and/or derived from a naturally-occurring retron, such as any retron-related sequence provided by Table X (the introduction of the one or more genetic modifications into a set of 7257 previously unknown retrons discovered through computational methods described herein (e.g., see Examples). In other embodiments, the recombinant retrons are based on introducing the one or more genetic modifications into previously available retron sequences (e.g., the “Mestre et al., Systematic Prediction of Genes Functionally Associated with Bacterial Retrons and Classification of The Encoded Tripartite Systems, Nucleic Acids Research, Volume 48, Issue 22, 16 December 2020, Pages 12632-12647” (incorporated herein by reference) to achieve recombinant retrons with the enhanced ability to produce increased concentrations or amounts of msDNA comprising a DNA donor template. [00204] In another aspect, the present disclosure further provides nucleic acid molecules encoding the recombinant retrons and/or recombinant retron components (e.g., a recombinant ncRNA and/or a recombinant retron RT). In still another aspect, the present disclosure provides genome editing systems comprising recombinant retron components (e.g., recombinant ncRNA and/or recombinant RT), programmable nucleases (e.g., RNA-guided nucleases, such as CRISPR-Cas proteins, ZFPs, and TALENS), and guide RNAs (in the case where RNA-guide nucleases are used in said genome editing systems). In a further aspect, the disclosure provides nucleic acid molecules encoding the described genome editing systems and said components thereof, as well as polypeptides making up the components of said genome editing systems. In yet another aspect, the disclosure provides vectors for transferring and/or expressing said genome editing systems, e.g., under in vitro, ex vivo, and in vivo conditions. In still another aspect, the disclosure provides cell-delivery compositions and methods, including compositions for passive and/or active transport to cells (e.g., plasmids), delivery by virus-based recombinant vectors (e.g., AAV and/or lentivirus vectors), delivery by non-virus-based systems (e.g., liposomes and LNPs), and delivery by virus-like particles. Depending on the delivery system employed, the retron-based genome editing systems described herein may be delivered in the form of DNA (e.g., plasmids or DNA-based virus vectors), RNA (e.g., ncRNA and mRNA delivered by LNPs), a mixture of DNA and RNA, protein (e.g., virus-like particles), and ribonucleoprotein (RNP) complexes. Any suitable combinations of approaches for delivering the components of the herein disclosed retron-based genome editing systems may be employed. In one embodiment, each of the components of the retron-based genome editing system is delivered by an all-RNA system, e.g., the delivery of one or more RNA molecules (e.g., mRNA and/or ncRNA) by one or more LNPs, wherein the one or more RNA molecules form the ncRNA and guide RNA (as needed) and/or are translated into the polypeptide components (e.g., the RT and a programmable nuclease). In yet another aspect, the disclosure provides methods for genome editing by introducing a retron-based genome editing system described herein into a cell (e.g., under in vitro, in vivo, or ex vivo conditions) comprising a target edit site, thereby resulting in an edit at the target edit. In other aspects, the disclosure provides formulations comprising any of the aforementioned components for delivery to cells and/or tissues, including in vitro, in vivo, and ex vivo delivery, recombinant cells and/or tissues modified by the recombinant retron-based genome modification systems and methods described herein, and methods of modifying cells by conducting genome editing and related DNA donor-dependent methods, such as recombineering, or cell recording, using the herein disclosed retron-based genome modification systems. The disclosure also provides methods of making the recombinant retrons, retron-based genome modification systems, vectors, compositions and formulations described herein, as well as to pharmaceutical compositions and kits for modifying cells under in vitro, in vivo, and ex vivo conditions that comprise the herein disclosed genome editing and/or modification systems. [00205] Described herein are engineered retrons comprising one or more heterologous nucleic acids. The one or more heterologous nucleic acids may be inserted, for example, at or within a location selected from: the msd locus, upstream of the msr locus, upstream of the msd locus, and downstream of the msd locus. In some embodiments, the engineered retrons have structural improvements over their naturally existing counterparts or wild-type retrons at least with respect to the encoded ncRNA and/or the reverse transcriptase (RT), such that the engineered retron or the encoded ncRNA thereof, when delivered to a host cell, such as a mammalian host cell, exhibits various functional improvements over its naturally existing/wild- type retron elements. [00206] Exemplary (non-limiting functional improvements) may include any one or more of the features described herein. For example, in some embodiments, the engineered retron may comprise a sequence modification (e.g., insertion, deletion, and/or substitution of one or more nucleotide(s)) in the msr locus and/or the msd locus that: i) modulates (e.g., enhances) reverse transcription, processivity, accuracy/fidelity, and/or production of the msDNA (e.g., in the mammalian cell); ii) modulates (e.g., reduces) immunogenicity of the ncRNA encoded by the engineered retron (e.g., encoded by the msr locus and/or the msd locus) in a host (e.g., a host comprising the mammalian cell); iii) comprises a nucleotide sequence that modulates (e.g., inhibits or antagonizes) a function of the msDNA; and/or iv) modulates (e.g., improves) efficiency of targeted genomic engineering. [00207] Thus, in general, the engineered retron is an engineered nucleic acid construct comprising: a) a first polynucleotide encoding a non-coding RNA (ncRNA), said first polynucleotide comprising: i) an msr locus encoding the msr RNA portion of a multi-copy single-stranded DNA (msDNA); and ii) an msd locus encoding the msd RNA portion of the msDNA; and [00208] b) one or more heterologous nucleic acids inserted at or within a location selected from: the msd locus, upstream of the msr locus, upstream of the msd locus, and downstream of the msd locus. [00209] The engineered nucleic acid construct (e.g., the engineered retron) may further comprise a second polynucleotide encoding a reverse transcriptase (RT), or a portion thereof, wherein the encoded RT is capable of synthesizing a DNA copy of at least a portion of the msd locus encoding the msDNA. [00210] In certain embodiments, the engineered retron of the invention encodes a reverse transcriptase (RT) or a functional domain thereof, comprising: i) a polypeptide listed in Table A, or a polypeptide having at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%%, at least 99.1%, at least 99.2%, at least 99.3%, at least 99.4%, at least 99.5%, at least 99.6%, at least 99.7%, at least 99.8% or at least 99.9% sequence identity to a polypeptide listed in Table A; and/or ii) a polypeptide listed in any one of Table C. In some embodiments, the RT does not comprise a polypeptide listed in Table X. [00211] In certain embodiments, the engineered retron of the invention encodes a reverse transcriptase (RT) or a functional domain thereof, comprising: i) a polynucleotide listed in Table A, or a polynucleotide having at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%%, at least 99.1%, at least 99.2%, at least 99.3%, at least 99.4%, at least 99.5%, at least 99.6%, at least 99.7%, at least 99.8% or at least 99.9% sequence identity to a polynucleotide listed in Table A; and/or ii) a consensus polynucleotide sequence listed in Table C. In some embodiments, the polynucleotide encoding the RT does not comprise a polynucleotide of Table X. [00212] In certain embodiments, the engineered retron of the invention encodes an ncRNA comprising: (I) an ncRNA listed in Table B, or an ncRNA having at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%%, at least 99.1%, at least 99.2%, at least 99.3%, at least 99.4%, at least 99.5%, at least 99.6%, at least 99.7%, at least 99.8% or at least 99.9% sequence identity to an ncRNA in Table B. [00213] In certain embodiments, the engineered retron of the invention encodes an ncRNA and a reverse transcriptase (RT) or a functional domain thereof, wherein the ncRNA and the RT or functional domain thereof are as described above. [00214] Specifically, in such embodiment, the ncRNA may comprise: (I) an ncRNA listed in Table B, or an ncRNA having at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%%, at least 99.1%, at least 99.2%, at least 99.3%, at least 99.4%, at least 99.5%, at least 99.6%, at least 99.7%, at least 99.8% or at least 99.9% sequence identity to an ncRNA listed in Table B. [00215] Also in such embodiment, the reverse transcriptase (RT) or functional domain thereof comprises: (A) i) a polypeptide listed in Table A, or a polypeptide having at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%%, at least 99.1%, at least 99.2%, at least 99.3%, at least 99.4%, at least 99.5%, at least 99.6%, at least 99.7%, at least 99.8% or at least 99.9% sequence identity to a polypeptide listed in Table A; and/or ii) a polypeptide listed in Table C; optionally, the RT does not comprise a polypeptide listed in Table X; OR (B) i) a polynucleotide listed in Table A, or a polynucleotide having at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%%, at least 99.1%, at least 99.2%, at least 99.3%, at least 99.4%, at least 99.5%, at least 99.6%, at least 99.7%, at least 99.8% or at least 99.9% sequence identity to a polynucleotide in Table A; and/or optionally, the polynucleotide encoding the RT does not comprise a polynucleotide in Table X. [00216] In certain embodiments, the engineered nucleic acid construct comprises: 1) an msr locus (that encodes the msr RNA portion of an msDNA); 2) an msd locus encoding the msd RNA portion of the msDNA; 3) a sequence encoding a retron reverse transcriptase (RT), wherein said msd RNA is capable of being reverse transcribed to form the msDNA by the retron reverse transcriptase (RT); and, 4) a heterologous nucleic acid inserted at or within the msd locus, upstream of the msr locus, upstream or downstream of the msd locus; wherein the engineered nucleic acid construct is engineered based on and/or to resemble a secondary structure of a wild- type or consensus retron encoding a wild-type or consensus retron ncRNA encompassed by: a) any one of the sequences and/or structures as depicted in any one of SEQ ID NOs: of Table B and/or FIGs.2-27; or b) a variant of a), having: i) up to 1, 2, or 3 (e.g., up to 1) nucleotide changes per 10 red lettered-nucleotides; ii) up to 4, 5, or 6 (e.g., up to 1 or 2) nucleotide changes per 10 black lettered-nucleotides; and/or iii) up to 7, 8, or 9 (e.g., up to 3 or 4) nucleotide changes per 10 grey lettered-nucleotides; and/or optionally further comprising: i) 7, 8, 9, or 10 (e.g., 9 or 10) nucleotides present per 10 red-circled nucleotides; ii) 6, 7, 8, 9, or 10 (e.g., 8, 9 or 10) nucleotides present per 10 black-circled nucleotides; iii) 4, 5, 6, 7, 8, 9, or 10 (e.g., 6, 7, 8, 9 or 10) nucleotides present per 10 grey-circled nucleotides; and/or iv) 2, 3, 4, 5, 6, 7, 8, 9, or 10 (e.g., 4, 5, 6, 7, 8, 9, or 10) nucleotides present per 10 white-circled nucleotides; wherein the ncRNA does not comprise an ncRNA associated with the sequences of Table X. [00217] The engineered nucleic acid construct (e.g., the engineered retron) may comprise one or more sequence modifications (e.g., an insertion, deletion, and/or substitution of one or more nucleotide(s)) in the msr locus and/or the msd locus that: a) modulates (e.g., enhances) reverse transcription, processivity, accuracy/fidelity, and/or production of the msDNA (e.g., in the mammalian cell); b) modulates (e.g., reduces) immunogenicity of ncRNA encoded by the engineered retron (e.g., the msr locus and/or the msd locus) in a host (e.g., a host comprising the mammalian cell); c) modulates (e.g., inhibits, either permanently or transiently) a function of the msDNA; and/or d) modulates (e.g., improves) efficiency of targeted genome editing / engineering. [00218] In some embodiments, the engineered nucleic acid construct (e.g., the engineered retron) is engineered based on and/or to resemble a secondary structure of a wild-type or consensus retron encoding a wild-type or consensus retron ncRNA encompassed by: a) the sequence of any one of Table B ncRNA sequences and/or the structure depicted in any one of FIGs.2-27; or b) a variant of a), having: i) up to 1, 2, or 3 (e.g., up to 1) nucleotide changes per 10 red lettered-nucleotides; ii) up to 4, 5, or 6 (e.g., up to 1 or 2) nucleotide changes per 10 black lettered-nucleotides; and/or iii) up to 7, 8, or 9 (e.g., up to 3 or 4) nucleotide changes per 10 grey lettered-nucleotides; and/or optionally further comprising: i) 7, 8, 9, or 10 (e.g., 9 or 10) nucleotides present per 10 red-circled nucleotides; ii) 6, 7, 8, 9, or 10 (e.g., 8, 9 or 10) nucleotides present per 10 black-circled nucleotides; iii) 4, 5, 6, 7, 8, 9, or 10 (e.g., 6, 7, 8, 9 or 10) nucleotides present per 10 grey-circled nucleotides; and/or iv) 2, 3, 4, 5, 6, 7, 8, 9, or 10 (e.g., 4, 5, 6, 7, 8, 9, or 10) nucleotides present per 10 white-circled nucleotides. [00219] Another aspect of the disclosure provides a vector system comprising a vector comprising the engineered retron described herein. [00220] Another aspect of the disclosure provides an isolated host cell comprising the engineered retron described herein, or the vector system described herein. [00221] Another aspect of the disclosure provides a pharmaceutical composition comprising the engineered retron described herein, or the vector system described herein. [00222] Another aspect of the disclosure provides a delivery vehicle comprising the engineered retron described herein or the ncRNA encoded by the engineered retron described herein, the vector or vector system described herein, the host cell described herein, or the pharmaceutical composition described herein. [00223] Another aspect of the disclosure provides a kit comprising the engineered retron described herein or the ncRNA encoded by the engineered retron described herein, and optionally instructions for genetically modifying a cell using the engineered retron described herein or the ncRNA encoded by the engineered retron described herein.
[00224] Another aspect of the disclosure provides a method of modifying a target DNA sequence in a host cell (e.g., a mammalian cell), the method comprising introducing into the host cell (e.g., the mammalian cell) the engineered retron of the invention, the ncRNA encoded by the engineered retron of the invention, or the vector / vector system described herein, to allow the production of the msDNA in the host cell (e.g., mammalian cell), wherein at least a part of the heterologous nucleic acid in the msDNA is integrated into the genome of the host (e.g., mammalian) cell at the target DNA sequence. Optionally, the target sequence is recognized by a suitable nuclease, such as a CRISPR/Cas effector enzyme, a ZFN, a TALEN, a meganuclease, TnpB, IscB, or a restriction endonuclease (RE), and a double-stranded break (DSB) is created by the nuclease to facilitate / promote the insertion of the part of the heterologous nucleic acid into the target sequence. Further optionally, the target sequence modified / inserted by the part of the heterologous nucleic acid can no longer be recognized by the nuclease to re-create a DSB.
[00225] Another aspect of the disclosure provides a use of the engineered retron in the various methods described herein.
[00226] Another aspect of the disclosure provides a genome editing system comprising: a) nuclease capable of acting at a target site on a genome (e.g. human genome), such as a CRISPR/Cas effector enzyme, a ZFN, a TALEN, a meganuclease, TnpB, IscB, or a restriction endonuclease (RE); and b) an engineered retron described herein, or an ncRNA encoded thereby, or a vector or a vector system comprising or encoding the same. Optionally, the nuclease may be linked to one or more element(s) of the engineered retron or the encoded ncRNA. For example, in one embodiment, the nuclease may be linked (e.g., fused or conjugated) to the reverse transcriptase of the engineered retron described herein. In another embodiment, the nuclease may engage/bind to form a complex with a nucleic acid guide sequence (such as a single-guided RNA of a Cas enzyme), wherein the guide sequence is linked to the ncRNA and/or msDNA of the engineered retron described herein. [00227] Another aspect of the disclosure provides an enhanced genome editing system, comprising the genome editing system of the disclosure connected to a biomolecule that modulates host DNA repair, in order to, for example, modulate (e.g., enhance) the incorporation of the heterologous nucleic acid sequence into a genome (e.g., human genome). [00228] With the general aspect of the disclosure described herein, specific aspects and embodiments of the disclosure are further described in the sections below. It should be understood that any one embodiment of the disclosure, including those described only in the examples or the claims, or only in one section herein below, can be combined with any one or more additional embodiments of the invention, unless such combination is expressly disclaimed or are improper. A. Recombinant Retrons [00229] The present disclosure provides engineered retrons, as well as compositions, systems, and methods that include or utilize the engineered retrons for genome modification, such as genome editing, cell recording, and recombineering. [00230] Retrons were originally discovered in 1984 in Myxococcus xanthus bacterium when a short, multi-copy single-stranded DNA (msDNA) that is abundantly present in the bacterial cell was identified. Since then, a number of naturally existing retrons have been found in many prokaryotes such as bacteria. [00231] As depicted in FIG.1A, retrons encode and transcribe as a single RNA, which comprises a non-coding RNA (ncRNA) portion and a portion encoding a specialized reverse transcriptase (RT). The retron ncRNA (msr and msd) is the precursor of the hybrid molecule that eventually forms, and it initially folds into a typical RNA secondary structure that is recognized by the accompanying RT. The translated RT typically recognizes certain secondary structures in the ncRNA, and binds the RNA template downstream from the msd region. The RT initiates reverse transcription of the RNA towards its 5ʹ end, starting from the 2’-end of a conserved guanosine (G) residue found immediately after a double-stranded RNA structure (the a1/a2 region) within the ncRNA. A portion of the ncRNA serves as a template for reverse transcription, and reverse transcription terminates before reaching the msr locus. During reverse transcription, cellular RNase H degrades the segment of the ncRNA that serves as template, but not other parts of the ncRNA. The result of the reverse transcription, the msDNA, remains covalently attached to the RNA template via the 2’-5ʹ phosphodiester bond, and base-pairs with the RNA template using the 3ʹ end of the msDNA. See FIG.1A for a general or typical organization of the retron coding sequence, including the RT encoding sequence and the msr and msd loci, as well as the synthesis of the msDNA by reverse transcription of the initial ncRNA transcript. [00232] Many retrons also contain an accessory protein (not depicted in FIG.1A), which may have a variable function that may not be fully understood. In certain embodiments, the engineered retrons described herein do not comprise the accessory protein naturally associated with the wild-type or template retron. [00233] Applicant has discovered, analyzed, and phylogenetically classified 7257 previously unknown retrons from nature based on multiple criteria, including sequence homology and conserved predicted secondary structures, and has grouped these retrons into different phylogenetic clades based on sequence homology and/or conserved predicted secondary structures. These clades include Type IA_IIA1 (FIG.2), Type 1B1 (FIG.3), Type IB2 (FIG.4), Type 1C (FIG.5), Type IIA1 other (FIG.6), Type IIA2 (FIG.7), Type IIA3 (FIG.8), Type IIA4 (FIG.9), Type IIA5 (FIG.10), Type IIIA1 (FIG.11), Type IIIA2 (FIG.12), Type IIIA3 (FIG. 13), Type IIIA4 (FIG.14), Type IIIA5 (FIG.15), Type IIIunk (FIG.16), Type IV (FIG. IX), Type V (FIG.19), Type VI (FIG.20), Type XI Group 1 (FIG.21), Type XI (Group 2) (FIG.22), Type XII (FIG.23), Type XIII (FIG.24), Type XIV (FIG.25), Eco107-like (FIG.26), and Outgroup A (FIG.27). The disclosure further describes the engineering and/or modification of these newly discovered retron sequences as a starting point to obtain useful recombinant retrons, such as those depicted in FIG.1B. [00234] FIG.1B.1 depicts an embodiment of a recombinant retron construct (e.g., a nucleotide sequence cloned into an expression vector) contemplated by the present disclosure. In the top left schematic, the single thin black line represents a double-stranded nucleotide sequence (e.g., as cloned into an expression vector, such as a plasmid). The recombinant retron is constructed by modifying a starting point retron DNA sequence encoding a ncRNA (the msr/msd region) (such as any one of the herein disclosed 7257 newly discovered retron sequences, and specifically any one of the 7257 ncRNA sequences of Table B. A starting point retron DNA sequence encoding an ncRNA may be modified in any number of ways and can including one modification or more than one modification. For example, the retron DNA may modified to contain at least one nucleotide modification, including a single nucleotide substitution, insertion, or deletion, or a substitution, insertion, or deletion of more than one nucleotide, i.e., up to 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, 69, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, or up to 100, or up to 200, 300, 400, 500, 600, 700, 800, 900, 1000, 1100, 1200, 1300, 1400, 1500, 1600, 1700, 1800, 1900, or up to 2000 nucleotides substituted, inserted, or deleted from a starting point retron (e.g., a wildtype retron). Where more than one nucleotide of a starting point retron (e.g., a wildtype retron) is substituted, deleted, or inserted, the nucleotides may be contiguous or non-contiguous. While an engineered retron as a whole is not naturally-occurring, it may include components such as nucleotide sequences that do occur in nature. For example, an engineered retron can have nucleotide sequences from different organisms (e.g., from different bacteria species), or from completely synthetic / artificial / recombinant nucleic acid sequences. Thus, an engineered retron can have a bacterial nucleotide sequence, a human nucleotide sequence, a viral nucleotide sequence, and/or a synthetic / artificial / recombinant nucleotide sequence, and/or combinations of such sequences. An example of modifications of the recombinant retrons disclosed herein include the insertion of a heterologous nucleic acid sequence in a retron, for example, inserted into the ncRNA locus, such as in the msr or the msd loci. Linking guide RNA molecules to the 5ʹ and/or 3ʹ ends (i.e., linking one at the 5ʹ end of a ncRNA and/or one at the 3ʹ end of a ncRNA) also represents another modification contemplated by the recombinant retrons disclosed herein. In such embodiments, the guide RNA molecules may also be categorized or referred to more generally as types of heterologous nucleic acid sequences used to modify starting point retrons. These modifications are depicted in FIG.1B. [00235] In addition to the DNA encoding the ncRNA, the DNA encoding the RT may also be modified to obtain a recombinant RT. For example, the RT-encoding DNA may modified to contain at least one nucleotide modification, including a single nucleotide substitution, insertion, or deletion, or a substitution, insertion, or deletion of more than one nucleotide, i.e., up to 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, 69, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, or up to 100, or up to 200, 300, 400, 500, 600, 700, 800, 900, 1000, 1100, 1200, 1300, 1400, 1500, 1600, 1700, 1800, 1900, or up to 2000 nucleotides substituted, inserted, or deleted from a starting point retron (e.g., a wildtype retron) within the RT gene. [00236] Such modifications to the DNA encoding ncRNA and/or RT may modulate the function of the ncRNA and/or RT in various ways, including i) modulating (e.g., enhancing) reverse transcription, processivity, accuracy/fidelity, and/or production of the msDNA (e.g., in the mammalian cell); ii) modulating (e.g., reducing) immunogenicity of ncRNA (msr locus and msd locus) encoded by the engineered retron in a host (e.g., a host comprising the mammalian cell); iii) modulating (e.g., inhibits, either permanently or transiently) a function of the msDNA; and/or iv) modulating (e.g., improving) efficiency of targeted genome editing / engineering. [00237] In one embodiment, the present disclosure provides recombinant retrons having the general structure of: a) an msr locus; b) an msd locus encoding the msd RNA portion of the msDNA; c) a sequence encoding a retron reverse transcriptase (RT) (optionally in trans to the ncRNA), wherein the msd RNA is capable of being reverse transcribed (e.g., in a host cell such as a mammalian cell) to form an msDNA by the retron reverse transcriptase (RT); d) a heterologous nucleic acid (e.g., heterologous DNA) capable of being transcribed with the msr locus and/or the msd locus, optionally, the heterologous nucleic acid is inserted at or within the msd locus, upstream of the msr locus, upstream or downstream of the msd locus. [00238] The engineered retrons of the invention are optionally structurally further modified to include one or more heterologous nucleic acids. The engineered retron may be further modified to provide various functional improvements, such as (without limitation), to enhance the production of msDNA in a cell (e.g., a mammalian cell, including a human cell). [00239] In certain embodiments, the disclosure provides engineered retrons based on their conserved predicted secondary structures, such as those in FIGs.2-27. [00240] In other embodiments, the disclosure provides engineered retrons based on their sequence identity. Exemplary RT amino acid sequences and ret gene nucleic acid sequences are provided in Table A. Exemplary RT consensus amino acid sequences and/or ret gene nucleic acid sequences are provided in Table C. Exemplary ncRNA sequences are provided in Table B. [00241] Retron sequences provisoed out of the scope of the invention are provided in Table X. [00242] In certain embodiments, exemplary engineered retrons of the invention (1) are engineered based on or engineered to resemble the secondary structures as depicted in any one of FIGs.2-27, and/or (2) are provided in Table B. Sequences with significant sequence percentage identity (e.g., at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%%, at least 99.1%, at least 99.2%, at least 99.3%, at least 99.4%, at least 99.5%, at least 99.6%, at least 99.7%, at least 99.8% or at least 99.9% sequence identity) are also within the scope of the invention. [00243] In certain embodiments, the engineered nucleic acid construct comprises: 1) an msr locus (that encodes the msr RNA portion of an msDNA); 2) an msd locus encoding the msd RNA portion of the msDNA; 3) a sequence encoding a retron reverse transcriptase (RT), wherein said msd RNA is capable of being reverse transcribed to form the msDNA by the retron reverse transcriptase (RT); and, 4) a heterologous nucleic acid inserted at or within the msd locus, upstream of the msr locus, upstream or downstream of the msd locus; wherein the engineered nucleic acid construct is engineered based on and/or to resemble a secondary structure of a wild- type or consensus retron encoding a wild-type or consensus retron ncRNA encompassed by: a) any one of the sequences and/or structures as depicted in any one of SEQ ID Nos of Table B and/or FIGs.2 -27; or b) a variant of a), having: i) up to 1, 2, or 3 (e.g., up to 1) nucleotide changes per 10 red lettered-nucleotides; ii) up to 4, 5, or 6 (e.g., up to 1 or 2) nucleotide changes per 10 black lettered-nucleotides; and/or iii) up to 7, 8, or 9 (e.g., up to 3 or 4) nucleotide changes per 10 grey lettered-nucleotides; and/or optionally further comprising: i) 7, 8, 9, or 10 (e.g., 9 or 10) nucleotides present per 10 red-circled nucleotides; ii) 6, 7, 8, 9, or 10 (e.g., 8, 9 or 10) nucleotides present per 10 black-circled nucleotides; iii) 4, 5, 6, 7, 8, 9, or 10 (e.g., 6, 7, 8, 9 or 10) nucleotides present per 10 grey-circled nucleotides; and/or iv) 2, 3, 4, 5, 6, 7, 8, 9, or 10 (e.g., 4, 5, 6, 7, 8, 9, or 10) nucleotides present per 10 white-circled nucleotides; wherein the ncRNA does not comprise an ncRNA associated with the sequences of Table X. [00244] In certain embodiments, the engineered retron of the invention encodes a reverse transcriptase (RT) or a functional domain thereof, comprising: i) a polypeptide listed in Table A, or a polypeptide having at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%%, at least 99.1%, at least 99.2%, at least 99.3%, at least 99.4%, at least 99.5%, at least 99.6%, at least 99.7%, at least 99.8% or at least 99.9% sequence identity to a polypeptide listed in Table A. In some embodiments, the RT does not comprise a polypeptide identified in Table X. [00245] In certain embodiments, the engineered retron of the invention encodes a reverse transcriptase (RT) or a functional domain thereof, comprising: i) a polynucleotide listed in Table A, or a polynucleotide having at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%%, at least 99.1%, at least 99.2%, at least 99.3%, at least 99.4%, at least 99.5%, at least 99.6%, at least 99.7%, at least 99.8% or at least 99.9% sequence identity to a polynucleotide of Table A and/or ii) a consensus polynucleotide sequence listed in Table A. In some embodiments, the polynucleotide encoding the RT does not comprise a polynucleotide identified in Table X. [00246] In certain embodiments, the engineered retron of the invention encodes an ncRNA comprising: (I) an ncRNA listed in Table B, or an ncRNA having at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%%, at least 99.1%, at least 99.2%, at least 99.3%, at least 99.4%, at least 99.5%, at least 99.6%, at least 99.7%, at least 99.8% or at least 99.9% sequence identity to an ncRNA of Table B. [00247] In certain embodiments, the engineered retron of the invention encodes an ncRNA and a reverse transcriptase (RT) or a functional domain thereof, wherein the ncRNA and the RT or functional domain thereof are as described above. [00248] Specifically, in such embodiments, the ncRNA may comprise: (I) an ncRNA listed in Table B, or an ncRNA having at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%%, at least 99.1%, at least 99.2%, at least 99.3%, at least 99.4%, at least 99.5%, at least 99.6%, at least 99.7%, at least 99.8% or at least 99.9% sequence identity to an ncRNA listed in Table B; and wherein the ncRNA optionally excludes the ncRNA associated with the sequences identified in Table X. [00249] Also in such embodiment, the reverse transcriptase (RT) or functional domain thereof comprises: (A) i) a polypeptide listed in Table A, or a polypeptide having at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%%, at least 99.1%, at least 99.2%, at least 99.3%, at least 99.4%, at least 99.5%, at least 99.6%, at least 99.7%, at least 99.8% or at least 99.9% sequence identity to a polypeptide listed in Table A; and/or ii) a polypeptide listed in Table C; optionally, the RT does not comprise a polypeptide identified in Table X; OR (B) i) a polynucleotide listed in Table A, or a polynucleotide having at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%%, at least 99.1%, at least 99.2%, at least 99.3%, at least 99.4%, at least 99.5%, at least 99.6%, at least 99.7%, at least 99.8% or at least 99.9% sequence identity to a polynucleotide listed in Table A; optionally, the polynucleotide encoding the RT does not comprise a polynucleotide associated with the sequences identified in Table X. [00250] In certain embodiments, the heterologous nucleic acid is between >20 nucleotides and about 10,000 nucleotides. [00251] The engineered retron may further comprise a sequence modification (e.g., insertion, deletion, and/or substitution of one or more nucleotide(s)) in the msr locus and/or the msd locus that: i) modulates (e.g., enhances) reverse transcription, processivity, accuracy/fidelity, and/or production of the msDNA (e.g., in the mammalian cell); ii) modulates (e.g., reduces) immunogenicity of ncRNA (msr locus and msd locus) encoded by the engineered retron in a host (e.g., a host comprising the mammalian cell); iii) comprises a nucleotide sequence that modulates (e.g., inhibits, either permanently or transiently) a function of the msDNA; and/or iv) modulates (e.g., improves) efficiency of targeted genome editing / engineering. [00252] Retron msr gene, msd gene, and RT nucleic acid sequences (e.g., the ret gene) as well as the encoded retron reverse transcriptase protein sequences that may serve as the template of the engineered retron described herein may be derived from any source, such as those in Table A, optionally excluding those associated with the sequences of Table X. [00253] In some embodiments, template or wild-type (wt) sequences of the msr gene, msd gene, and the RT coding sequence (viz., the ret gene) used in the engineered retron are derived from a bacterial retron. [00254] In some embodiments, representative template/wild-type retrons are from gram negative bacteria. In some embodiments, the retron is from a bacterium listed in Table X. [00255] In some embodiments, the engineered retrons are engineered based on clades defined on retron/retron RTs, in which the retrons are associated with a tripartite system composed of the ncRNA, the RT and an additional protein or RT-fused domain with diverse enzymatic functions. See, for example, “Mestre et al., Systematic Prediction of Genes Functionally Associated with Bacterial Retrons and Classification of The Encoded Tripartite Systems, Nucleic Acids Research, Volume 48, Issue 22, 16 December 2020, Pages 12632- 12647” (incorporated herein by reference). While the clades are based primarily upon naturally occurring ncRNA and retron/retron RT, and an additional protein or RT-fused domain, the clades, for the purpose of serving as the templates for the subject engineered retrons, are not limited to naturally occurring sequences. Rather, the clades can also encompass non-naturally occurring ncRNA and RT, including, without limitation, recombinant, modified or altered, chimeric, hybrid, synthetic, artificial, etc. [00256] Thus, according to the instant disclosure, retrons may be considered phylogenetically related based on a Neighbor-Joining algorithm of at least 75% (of at least 1000 replicates) and a Poisson correction distance measurement of no more than 0.05, based on alignment of the retron RT. Alternatively or in addition, retrons may be considered phylogenetically related when / if the same RT, or closely related RT, can recognize the secondary structures of the ncRNA of the retrons and reserve transcribe the retrons to produce msDNA. [00257] In certain embodiments, sequence alignments between different retron sequences (e.g., ncRNA and/or RT (protein and/or nucleic acid) sequences) or secondary structure generations are based on software known to one of ordinary skill in the art. [00258] The retron ncRNA sequences including msr and msd sequences within the same clade may be highly conserved at certain positions, while being less conserved at other positions. [00259] Exemplary consensus sequences based on clade members were generated (see, for example, the corresponding FIGs.2-27, respectively) to show these conserved sequences and/or secondary structures, including the highly conserved nucleotides with at least 97% sequence conservation at red lettered-nucleotides, those with between 90-97% sequence conservation at black lettered-nucleotides, and 75-90% nucleotide sequence identity at grey-lettered nucleotides. Further structural limitations of the consensus sequences for the clades are provided as colored circles indicating the probability of having a base at that specific position, including red circles representing a base in 97% of the cases, black circles representing a base in 90-97% of the cases, and grey circles representing a base in 75-90% of the cases. [00260] In some embodiments, the template ncRNA based on which the subject engineered retron is modified (including the msr and msd region sequences) is a consensus sequence for the various retron ncRNA (including msr and msd nucleic acid sequences) clades, as provided in any one of SEQ ID NOs: of Table B and the corresponding FIGs.2-27, respectively, including the bases that are highly conserved and depicted by a specific-colored letters or circles, and optionally further including bases that may be present at specific locations by specific-colored circles. [00261] In some embodiments, the engineered retron is engineered, based on and/or to resemble a secondary structure of a wild-type or consensus retron encoding a wild-type or consensus retron ncRNA encompassed by: 1) the sequences and/or structures as depicted in any one of SEQ ID NOs: of Table B and the corresponding FIGs.2-27, respectively; or 2) a variant of 1), having: A) up to 1, 2, or 3 (e.g., up to 1) nucleotide changes per 10 red lettered- nucleotides; B) up to 4, 5, or 6 (e.g., up to 1 or 2) nucleotide changes per 10 black lettered- nucleotides; and/or C) up to 7, 8, or 9 (e.g., up to 3 or 4) nucleotide changes per 10 grey lettered- nucleotides. Optionally, the variant of 1) further comprises: a) 7, 8, 9, or 10 (e.g., 9 or 10) nucleotides present per 10 red-circled nucleotides; b) 6, 7, 8, 9, or 10 (e.g., 8, 9 or 10) nucleotides present per 10 black-circled nucleotides; c) 4, 5, 6, 7, 8, 9, or 10 (e.g., 6, 7, 8, 9 or 10) nucleotides present per 10 grey-circled nucleotides; and/or d) 2, 3, 4, 5, 6, 7, 8, 9, or 10 (e.g., 4, 5, 6, 7, 8, 9, or 10) nucleotides present per 10 white-circled nucleotides. [00262] The engineered retron may be engineered by introducing the sequence modifications (e.g., deletions, additions, or substitutions) into the wild-type retron encoding wild-type retron ncRNA, or into the retron encoding the consensus retron ncRNA. [00263] For example, a variant retron may not satisfy the sequence and/or structural requirements of any one of SEQ ID NOs: of Table B and the corresponding FIGs.2-27, respectively, but may still be a suitable template for the engineered retron described herein, so long as one or more of the conditions set forth in A) – C) and/or a)-d) are met. [00264] In certain embodiments, the highly conserved sequences in the template retrons are preserved / conserved or substantially preserved / conserved in the engineered retron described herein. [00265] In certain embodiments, all or substantially all the red lettered-nucleotides (i.e., those conserved in about 97% or more of the retrons in the same clade) are preserved / conserved in the engineered retron described herein. In certain embodiments, no more than 1, 2, or 3 (e.g., up to 1) nucleotide change(s) (e.g., deleted or substituted) occur per 10 red lettered-nucleotides in the engineered retron described herein. In certain embodiments, no more than about 0.3%, 0.5%, 1%, 2%, 3%, 4%, or 5% of the red lettered-nucleotides are changed (e.g., deleted or substituted) in the engineered retron described herein. [00266] In certain embodiments, all or substantially all the black lettered-nucleotides (i.e., those conserved in about 90-97% of the retrons in the same clade) are preserved / conserved in the engineered retron described herein. In certain embodiments, no more than 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 (e.g., up to 1 or 2) nucleotide change(s) (e.g., deleted or substituted) occur per 10 black lettered-nucleotides are changed in the engineered retron described herein. In certain embodiments, no more than about 3%, 4%, 5% or 10% of the black lettered-nucleotides are changed (e.g., deleted or substituted) in the engineered retron described herein. [00267] In certain embodiments, all or substantially all the grey lettered-nucleotides (i.e., those conserved in about 75-90% of the retrons in the same clade) are preserved / conserved in the engineered retron described herein. In certain embodiments, no more than 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, or 15 (e.g., up to 3 or 4, or up to 7, 8, or 9) nucleotide change(s) (e.g., deleted or substituted) occur per 10 grey lettered-nucleotides are changed in the engineered retron described herein. In certain embodiments, no more than about 5%, 10%, 15%, 20%, or 25% of the grey lettered-nucleotides are changed (e.g., deleted or substituted) in the engineered retron described herein. [00268] In certain embodiments, all or substantially all the red circled-nucleotides (i.e., those with a nucleotide in about 97% or more of the retrons in the same clade) are present in the engineered retron described herein. In certain embodiments, no more than 1, 2, or 3 (e.g., 0.3, 0.5, or up to 1) nucleotides are absent (e.g., deleted) per 10 red circled-nucleotides in the engineered retron described herein. In certain embodiments, 7, 8, 9, or 10 (e.g., 9 or 10) nucleotides are present per 10 red circled-nucleotides in the engineered retron described herein. In certain embodiments, no more than about 0.3%, 0.5%, 1%, 2%, 3%, 4%, or 5% of the red circled-nucleotides are absent (e.g., deleted) in the engineered retron described herein. [00269] In certain embodiments, all or substantially all the black circled-nucleotides (i.e., those with a nucleotide in about 90-97% of the retrons in the same clade) are present in the engineered retron described herein. In certain embodiments, no more than 1, 2, 3, or 4 (e.g., up to 1 or 2) nucleotides are absent (e.g., deleted) per 10 black circled-nucleotides in the engineered retron described herein. In certain embodiments, 6, 7, 8, 9, or 10 (e.g., 8, 9 or 10) nucleotides are present per 10 black circled-nucleotides in the engineered retron described herein. In certain embodiments, no more than about 1%, 2%, 3%, 5% or 10% of the black circled-nucleotides are absent (e.g., deleted) in the engineered retron described herein. [00270] In certain embodiments, all or substantially all the grey circled-nucleotides (i.e., those with a nucleotide in about 75-90% of the retrons in the same clade) are present in the engineered retron described herein. In certain embodiments, no more than 1, 2, 3, 4, or 5 (e.g., up to 2, 3, or 4) nucleotides are absent (e.g., deleted) per 10 grey circled-nucleotides in the engineered retron described herein. In certain embodiments, 4, 5, 6, 7, 8, 9, or 10 (e.g., 6, 7, 8, 9 or 10) nucleotides are present per 10 grey circled-nucleotides in the engineered retron described herein. In certain embodiments, no more than about 5%, 10%, 15%, 20%, or 25% of the grey circled-nucleotides are absent (e.g., deleted) in the engineered retron described herein. [00271] In certain embodiments, all or substantially all the white circled-nucleotides (i.e., those with a nucleotide in about 50-75% of the retrons in the same clade) are present in the engineered retron described herein. In certain embodiments, no more than 1, 2, 3, 4, 5, 6, or 6 (e.g., up to 2, 3, 4, 5, 6) nucleotide are absent (e.g., deleted) per 10 white circled-nucleotides in the engineered retron described herein. In certain embodiments, 2, 3, 4, 5, 6, 7, 8, 9, or 10 (e.g., 4, 5, 6, 7, 8, 9 or 10) nucleotides are present per 10 grey circled-nucleotides in the engineered retron described herein. In certain embodiments, no more than about 5%, 10%, 15%, 20%, 30%, 40%, or 50% of the white circled-nucleotides are absent (e.g., deleted) in the engineered retron described herein. [00272] In some embodiments, the engineered retron is synthetically produced. In other embodiments, the synthetically produced engineered retron comprises the sequences and/or secondary structures as depicted in any one of SEQ ID NOs: of Table B and the corresponding FIGs.2-27, respectively, and at least the conserved color lettered nucleotides according to their respective levels of sequence identity (e.g., red, black and gray letters), and/or at least the conserved colored circle nucleotides according to their respective levels of probability of sequence presence (e.g., red, black and gray circles). [00273] In some embodiments, the sequence modification in the engineered retrons leads to/results in the encoded retron ncRNA having the desired functional improvement. [00274] In certain embodiments, the one or more sequence modifications comprises, in the ncRNA, one or more of: (i) a modified (e.g., mutated, reduced, or eliminated) bulge in a1, a2, or both a1 and a2; (ii) an extension or shortening of a1, a2, or both a1 and a2; (iii) an extension or shortening of a spacer sequence between hairpin loops (e.g., S1, S2, S3, and/or S4 in FIG.2, or any of the S regions in an one of FIGs.2-27); (iv) an additional or modified (e.g., mutated or eliminated) bulge in hairpin loops (e.g., L2 and/or L3 in FIG.2, or any of the L regions in an one of FIGs.2-27 (e.g., by removing unpaired bases in the bulge, or by replacing unpaired bases with an equivalent number of base pairs)); (v) a modified (e.g., extended or shortened) length of hairpin loops (e.g., L1, L2, L3, and/or L4 in FIG.2, or any of the L regions in an one of FIGs.2- 27); (vi) an alternative L1 and/or L2 (in FIG.2, or any of the L regions in an one of FIGs.2-27) having complement, reverse, or reverse complement sequences; (vii) a modified (e.g., increased) number of unpaired bases at the tip of hairpin loops (e.g., L1, L2, L3, and/or L4 in FIG.2, or any of the L regions in an one of FIGs.2-27); (viii) a modified (e.g., increased or decreased) GC content in hairpin loops (e.g., L1, L2, L3, and/or L4 in FIG.2, or any of the L regions in an one of FIGs.2-27); (ix) an insertion of the heterologous nucleic acid in spacer sequences between hairpin loops (e.g., S1, S2, S3 and/or S4 in FIG.2, or any of the S regions in an one of FIGs.2- 27), or at the tip of hairpin loops (e.g., L1, L2, L3, and/or L4 in FIG.2, or any of the L regions in an one of FIGs.2-27); (x) a deletion of one or more hairpin loops (e.g., L1, L2, L3 and/or L4 in FIG.2, or any of the L regions in an one of FIGs.2-27); (xi) an addition of a new loop in a spacer sequence between hairpin loops (e.g., S1, S2, S3, and/or S4 in FIG.2, or any of the S regions in an one of FIGs.2-27); (xii) circularization of the ncRNA with the 5ʹ end and the 3ʹ end of the ncRNA being connected either directly, or via a spacer sequence; (xiii) a repositioned branching guanosine capable of initiating reverse transcription priming; (xiv) a staggered end sequence that reduces immunogenicity of the retron ncRNA, created by, e.g., adding or removing the 5ʹ a1 nucleotides and/or the 3ʹ a2 nucleotides; and/or, (xv) an antisense sequence complementary to a CRISPR/Cas guide RNA (gRNA) sequence encoded by the heterologous nucleic acid, wherein the antisense sequence hybridizes to and inhibits said gRNA in the encoded retron ncRNA, and wherein said antisense sequence is removed upon reverse transcription of the msDNA. [00275] Unless specifically indicated otherwise, the a1 and a2 regions are both single- stranded and substantially reverse complementary to each other, forming a stem with optional interruption by a symmetric or asymmetric bulge, with optional one or more 5ʹ and/or 3ʹ overhang/unpaired nucleotide(s), wherein the a1 region generally ends before (e.g., ends immediately 5ʹ to) the conserved branching guanosine (G) providing the 2’-OH for reverse transcription priming. [00276] In some embodiments, the sequence change comprises a mutated, reduced, or eliminated bulge in the a1/a2 stem region, including sequence change(s) in one (i.e., a1 or a2) strand, or both a1 and a2 strands. [00277] For example, in some embodiments, the sequence change comprises deleting nucleotides from a1, a2, or both a1 and a2, such that the size of the bulge is reduced, or a symmetrical bulge becomes asymmetrical or vice versa, or a bulge is eliminated. [00278] In some embodiments, the sequence change comprises replacing/substituting nucleotides in a1, a2, or both a1 and a2, such that previously unpaired bases in the bulge become base-paired. [00279] In some embodiments, the sequence change comprises replacing an unpaired purine base with one or more unpaired pyrimidine base(s). [00280] In some embodiments, the sequence change comprises replacing an unpaired pyrimidine base with one or more unpaired purine base(s). [00281] In some embodiments, the sequence change comprises replacing one unpaired purine base (e.g., A or G) with another unpaired purine base (e.g., G or A, respectively). [00282] In some embodiments, the sequence change comprises replacing one unpaired pyrimidine base (e.g., T/U or C) with another unpaired pyrimidine base (e.g., C or T/U, respectively). [00283] In some embodiments, the sequence change comprises an extension or shortening of a1, a2, or both a1 and a2. [00284] For example, the length of a1 can be shortened by deleting 5ʹ overhang, deleting any upstream bulge nucleotides, deleting bases involved in base-pairing. Likewise, the length of a1 can be extended by adding 5ʹ overhang, adding any upstream bulge nucleotides, adding bases involved in base-pairing. [00285] In some embodiments, the length of a2 can be shortened by deleting 5ʹ overhang, deleting any downstream bulge nucleotides, deleting bases involved in base-pairing. Likewise, the length of a2 can be extended by adding 5ʹ overhang, adding any downstream bulge nucleotides, adding bases involved in base-pairing. [00286] In some embodiments, the spacer sequences between hairpin loops as depicted in any one of FIGs.2-27, (e.g., S1, S2, S3 and/or S4 in FIG.2) can be extended or shortened. In some embodiments the modification can be by inserting a heterologous nucleic acid sequence in spacer sequences between hairpin loops (e.g., S1, S2, S3 and/or S4 in FIG.2). In certain embodiments, one or more heterologous nucleic acid sequences is inserted in a spacer sequence in the msd region. In some embodiments, the modification of the spacer region can be by interrupting the spacer with additional bulges or hairpin loops. [00287] In other embodiments, a bulge in the hairpin loops are mutated or eliminated (e.g., by removing unpaired bases) in the bulge, such that, for example, a symmetric bulge becomes an unsymmetrical bulge, or an unsymmetrical bulge becomes a symmetric one or an even more unsymmetrical one. In certain embodiments, unpaired bases in the bulge is replaced with an equivalent number of base pairs. The additional base pairs may be merged into the stem at one or both ends of the previous bulge, or may bisect a previous bulge to create two bulges. [00288] In some embodiments, the length of one or more hairpin loops as depicted in any one of FIGs.2-27, (e.g., L1, L2, L3 and/or L4 of FIG.2) can be extended or shortened. For example, the number of unpaired bases within the tip or loop can be increased or decreased. Further, a heterologous nucleic acid sequence of interest can be inserted within the tip or the hairpin loop. In certain embodiments, the heterologous nucleic acid sequence of interest is inserted within the tip or the hairpin loop in the msd locus. [00289] In other embodiments, the GC content in the tip or hairpin loops are increased or decreased. [00290] In still other embodiments, a hairpin loop can be deleted. [00291] In some embodiments, the ncRNA with the 5ʹ end and the 3ʹ end of the ncRNA can be circularized by being connected either directly, or via a spacer sequence. [00292] In some embodiments, one or more hairpin loops (e.g. L1, L2, L3 and/or L4 of FIG.2) are modified to have complement, reverse, or reverse complement sequences. [00293] In certain embodiments, the branching guanosine (G) capable of initiating reverse transcription priming is repositioned. For example, the G can be placed further downstream of the end of the a1 sequence by, for example, 1, 2, 3, 4, or 5 additional nucleotides. [00294] In certain embodiments, immunogenicity of the retron ncRNA is reduced by, e.g., adding or removing the 5ʹ a1 nucleotides and/or the 3ʹ a2 nucleotides. [00295] In certain embodiments, the one or more heterologous nucleic acid sequences (inserted into the subject engineered retron) comprise: a) a heterologous nucleic acid (such as the coding sequence for an RNA aptamer or a ribozyme) inserted into the msr locus or the msd locus (such as in an S region (e.g., S1, S2, S3 and/or S4 in FIG.2, or any of the S regions in any one of FIGs.2-27), or the tip of an L region (e.g., L1, L2, L3 and/or L4 in FIG.2, or any of the L regions in any one of FIGs.2-27), or upstream or downstream of either the msr locus or the msd locus; or b) a first heterologous nucleic acid inserted into the msd locus, and a second heterologous nucleic acid inserted either upstream of the msr locus or downstream of the msd locus, wherein the second heterologous nucleic acid encodes a CRISPR/Cas guide RNA (gRNA). [00296] In certain embodiments, an antisense sequence complementary to a CRISPR/Cas guide RNA (gRNA) sequence encoded by the heterologous nucleic acid can be included, wherein the antisense sequence hybridizes to and inhibits the gRNA in the encoded retron ncRNA, and wherein the antisense sequence is removed upon reverse transcription of the msDNA. [00297] In certain embodiments, said heterologous nucleic acid encodes a protein or peptide of interest, or wherein said heterologous nucleic acid comprises or encodes a donor / template sequence (e.g., a donor that corrects / repairs / removes a mutation at the target genome site, such as a mutated exon in a disease gene; a functional DNA element (such as a promoter, an enhancer, a protein binding sequence, a methylation site, a homology region for assisting gene editing, etc.); or a coding sequence for a functional RNA element (ncRNAs, etc.)). [00298] In certain embodiments, the protein or peptide of interest comprises a therapeutic protein (such as a wildtype protein defective in a disease cell, or a therapeutic antibody or antigen-binding fragment thereof) useful in treating a disease. [00299] Other heterologous nucleic acids of the invention are described in other section of the specification, all incorporated herein by reference. [00300] In some embodiments, the template/wild-type retron for the engineered retron encodes a wild-type or consensus retron ncRNA polynucleotide having a consensus secondary structure shown in any one of FIGs.2-27, which are described individually below: [00301] Variants of this template, which can also be used in the engineered retron of the invention, include a variant having: A) up to 1, 2, or 3 (e.g., up to 1) nucleotide changes per 10 red lettered-nucleotides; B) up to 4, 5, or 6 (e.g., up to 1 or 2) nucleotide changes per 10 black lettered-nucleotides; and/or C) up to 7, 8, or 9 (e.g., up to 3 or 4) nucleotide changes per 10 grey lettered-nucleotides; and/or optionally further comprising: a) 7, 8, 9, or 10 (e.g., 9 or 10) nucleotides present per 10 red-circled nucleotides; b) 6, 7, 8, 9, or 10 (e.g., 8, 9 or 10) nucleotides present per 10 black-circled nucleotides; c) 4, 5, 6, 7, 8, 9, or 10 (e.g., 6, 7, 8, 9 or 10) nucleotides present per 10 grey-circled nucleotides; and/or d) 2, 3, 4, 5, 6, 7, 8, 9, or 10 (e.g., 4, 5, 6, 7, 8, 9, or 10) nucleotides present per 10 white-circled nucleotides. [00302] In some embodiments, the engineered retron is entirely synthetically produced and having the conserved nucleotides as denoted by the colored letters as shown in SEQ ID NO. XX and FIGs.2-27. [00303] In some embodiments, the non-coding RNA (ncRNA) portion of the engineered retron comprises a polynucleotide (e.g., a DNA molecule) encoding an ncRNA listed in Table B, or an ncRNA having at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%%, at least 99.1%, at least 99.2%, at least 99.3%, at least 99.4%, at least 99.5%, at least 99.6%, at least 99.7%, at least 99.8% or at least 99.9% sequence identity to an ncRNA listed in Table B. In some embodiments, the ncRNA does not comprise an ncRNA associated with the sequences of Table X. [00304] Amplification of an engineered retron described herein may be performed, for example, before transfection of cells or ligation into vectors. Any method for amplifying the engineered retron may be used, including, but not limited to polymerase chain reaction (PCR), isothermal amplification, nucleic acid sequence-based amplification (NASBA), transcription mediated amplification (TMA), strand displacement amplification (SDA), and ligase chain reaction (LCR). In one embodiment, the engineered retron comprise common 5ʹ and 3ʹ priming sites to allow amplification of retron sequences in parallel with a set of universal primers. In another embodiment, a set of selective primers is used to selectively amplify a subset of retron sequences from a pooled mixture. [00305] In some embodiments, the template/wild-type retron for the engineered retron encodes a wild-type or consensus retron ncRNA polynucleotide having a consensus secondary structure shown in FIG.2-27, and as described individually below: Type IA/IIA1 retron (FIG.2) [00306] In some embodiments, the template/wt retron for the subject engineered retron encodes a wild-type retron ncRNA polynucleotide having a consensus secondary structure that can be described as: a1-S1-L1-L2-S2-L3-S3-a2-S4-L4, wherein: a1/a2 is a stem 8 bp in length; L1 is a stem of 5 bps with a 10-nt tip; L2 is a stem of 7 bps with a 5-nt tip, and a 1/1 bulge 3 nt from the tip; L3 is a stem of 23 bps with a 22-nt tip, and a 2/2 bulge 21 bps from the tip; L4 is a stem of 11 bps with a 5-nt tip; S1 is a single-stranded spacer region between the a1/a2 stem and L1, with no spacer between L1 and L2; S2 is a single-stranded spacer region between L2 and L3; S3 is a single-stranded spacer region between L3 and the a1/a2 stem; and S4 is a single-stranded spacer region between the a1/a2 stem and L4, and the conserved nucleotides are as shown in SEQ ID NO: 1 and FIG.2, and wherein the colored circled nucleotides are present at the respective levels of certainty (e.g., at least about 97% of the red-circled nucleotides, at least about 90-97% of the black-circled nucleotides, at least about 75- 90% of the grey-circled nucleotides, and at least about 50% of the white-circled nucleotides are present). [00307] In some embodiments, the engineered retron is entirely synthetically produced and having the conserved nucleotides as denoted by the colored letters as shown in FIG.2. Type IB1 retron (FIG.3) [00308] In some embodiments, the template/wt retron for the subject engineered retron encodes a wild-type retron ncRNA polynucleotide having a consensus secondary structure that can be described as: a1-S1-L1-S2-L2-S3-a2, wherein: a1/a2 is a stem 6 bps in length with a 2/2 bulge 3 bps from the tip, wherein a1 has a 2-nt overhang and a2 has a 6-nt overhang; L1 is a stem of 14 bps with a 3-nt tip, a 1/0 bulge 4 bps from the tip, and a 0/6 bulge 10 bps from the tip; L2 is a stem of 23 bps with a 5-nt tip, a 1/1 bulge 4 bps from the tip, and a 0/1 bulge 18 bps from the tip; S1 is a single-stranded spacer region between a1/a2 and L1; S2 is a single-stranded spacer region between L1 and L2; S3 is a single-stranded spacer region between L2 and a1/a2, the conserved nucleotides are as shown in FIG.3, and wherein the colored circled nucleotides are present at the respective levels of certainty (e.g., at least about 97% of the red-circled nucleotides, at least about 90-97% of the black-circled nucleotides, at least about 75-90% of the grey-circled nucleotides, and at least about 50% of the white-circled nucleotides are present). [00309] In some embodiments, the engineered retron is entirely synthetically produced and having the conserved nucleotides as denoted by the colored letters as shown in FIG.3. Type IB2 retron (FIG.4) [00310] In some embodiments, the template/wt retron for the subject engineered retron encodes a wild-type retron ncRNA polynucleotide having a consensus secondary structure that can be described as: a1-S1-L1-L2-S2-L3-S3-L4-S4-a2, wherein: the a1/a2 stem is 16 bp in length, with a 17-base 5ʹ overhang and a 16-base 3ʹ overhang; L1 is a stem of 6 bps with a 4-nt tip; L2 is a stem of 4 bps with a 4-nt tip, with a 2/2 bulge 2 nts from the tip; L3 is a stem of 3 bps with a 5-nt tip; L4 is a stem of 9 bps with a 5-nt tip, and a 1/1 bulge 4 nts from the tip; S1, S2, S3, and S4 are single-stranded spacer regions between the a1/a2 stem and L1, L2 and L3, L3 and L4, and L4 and the a1/a2 stem, respectively, with no spacer between L1 and L2; wherein the last 5 nts of S1 and the 5th-9th nts of S2 form a 5-bp stem, and, the conserved nucleotides are as shown in FIG.4, and wherein the colored circled nucleotides are present at the respective levels of certainty (e.g., at least about 97% of the red-circled nucleotides, at least about 90-97% of the black-circled nucleotides, at least about 75-90% of the grey-circled nucleotides, and at least about 50% of the white-circled nucleotides are present). [00311] In some embodiments, the engineered retron is entirely synthetically produced and having the conserved nucleotides as denoted by the colored letters as shown in FIG.4. Type 1C retron (FIG.5) [00312] In some embodiments, the template/wt retron for the subject engineered retron encodes a wild-type retron ncRNA polynucleotide having a consensus secondary structure that can be described as: a1-S1-L1-S2-L2-S3-a2, wherein: a1/a2 is a stem 13 bps in length; L1 is a stem of 9 bps with a 3-nt tip; L2 is a stem of 10 bps with a 5-nt tip; S1 is a single-stranded spacer region between a1/a2 and L1; S2 is a single-stranded spacer region between L1 and L2; S3 is a single-stranded spacer region between L2 and a1/a2, the conserved nucleotides are as shown in FIG.5, and wherein the colored circled nucleotides are present at the respective levels of certainty (e.g., at least about 97% of the red-circled nucleotides, at least about 90-97% of the black-circled nucleotides, at least about 75-90% of the grey-circled nucleotides, and at least about 50% of the white-circled nucleotides are present). [00313] In some embodiments, the engineered retron is entirely synthetically produced and having the conserved nucleotides as denoted by the colored letters as shown in FIG.5. Type IIA1 retron (FIG.6) [00314] In some embodiments, the template/wt retron for the subject engineered retron encodes a wild-type retron ncRNA polynucleotide having a consensus secondary structure that can be described as: a1-S1-L1-S2-L2-S3-L3-S4-a2, wherein: a1/a2 is a stem 10 bps in length with a 1-nt overhang on a2; L1 is a stem of 10 bps with a 3-nt tip; L2 is a stem of 7 bps with a 5-nt tip; L3 is a stem of 27 bps with a 8-nt tip and a 0/2 bulge 26 bps from the tip; S1 is a single-stranded spacer region between a1/a2 and L1; S2 is a single-stranded spacer region between L1 and L2; S3 is a single-stranded spacer region between L2 and L3; S4 is a single-stranded spacer region between L3 and a1/a2, the conserved nucleotides are as shown in FIG.6, and wherein the colored circled nucleotides are present at the respective levels of certainty (e.g., at least about 97% of the red-circled nucleotides, at least about 90-97% of the black-circled nucleotides, at least about 75-90% of the grey-circled nucleotides, and at least about 50% of the white-circled nucleotides are present). [00315] In some embodiments, the engineered retron is entirely synthetically produced and having the conserved nucleotides as denoted by the colored letters as shown in FIG.6. Type IIA2 retron (FIG.7) [00316] In some embodiments, the template/wt retron for the subject engineered retron encodes a wild-type retron ncRNA polynucleotide having a consensus secondary structure that can be described as: a1-S1-L1-S2-L2-L3-S3-a2, wherein: a1/a2 is a stem 7 bp in length with no overhangs; L1 is a stem of 8 bps with a 3-nt tip; L2 is a stem of 30 bps with a 8-nt tip, a 1/1 bulge 2 bps from the tip, and a 1/1 bulge 27 bps from the tip; L3 is a stem of 8 bps with a 5-nt tip, and a 0/1 bulge 3 nt from the tip; S1 is a single-stranded spacer region between the a1/a2 stem and L1; S2 is a single-stranded spacer region between L1 and L2; and, S3 is a single-stranded spacer region between L3 and the a1/a2 stem; the conserved nucleotides are as shown in FIG.7, and wherein the colored circled nucleotides are present at the respective levels of certainty (e.g., at least about 97% of the red-circled nucleotides, at least about 90-97% of the black-circled nucleotides, at least about 75-90% of the grey-circled nucleotides, and at least about 50% of the white-circled nucleotides are present). [00317] In some embodiments, the engineered retron is entirely synthetically produced and having the conserved nucleotides as denoted by the colored letters as shown in FIG.7. Type IIA3 retron (FIG.8) [00318] In some embodiments, the template/wt retron for the subject engineered retron encodes a wild-type retron ncRNA polynucleotide having a consensus secondary structure that can be described as: a1-S1-L1-L2-S2-L3-S3-a2, wherein: a1/a2 is a stem 6 bps in length; L1 is a stem of 8 bps with a 9-nt tip; L2 is a stem of 8 bps with a 3-nt tip; S1 is a single-stranded spacer region between a1/a2 and L1; S2 is a single-stranded spacer region between L2 and L3; S3 is a single-stranded spacer region between L3 and a1/a2, the conserved nucleotides are as shown in FIG.8, and wherein the colored circled nucleotides are present at the respective levels of certainty (e.g., at least about 97% of the red-circled nucleotides, at least about 90-97% of the black-circled nucleotides, at least about 75-90% of the grey-circled nucleotides, and at least about 50% of the white-circled nucleotides are present). [00319] In some embodiments, the engineered retron is entirely synthetically produced and having the conserved nucleotides as denoted by the colored letters as shown in FIG.8. Type IIA4 retron (FIG.9) [00320] In some embodiments, the template/wt retron for the subject engineered retron encodes a wild-type retron ncRNA polynucleotide having a consensus secondary structure that can be described as: a1-a2-L1-S1-L2-L3-S2-L4-S3, wherein: a1/a2 is a stem 3 bp in length with no overhangs and a 7-nt tip; L1 is a stem of 7 bps with a 3-nt tip; L2 is a stem of 6 bps with a 4-nt tip; L3 is a stem of 40 bps with a 5-nt tip, and a 2/2 bulge 3 bps from the tip, a 5/4 bulge 10 bps from the tip, and a 12/15 bulge 30 bps from the tip; L4 is a stem of 4 bps with a 9-nt tip; S1 is a single-stranded spacer region between L1 and L2/L3; S2 is a single-stranded spacer region between L2/L3 and L4; S3 is a single-stranded spacer region between L4 and the 3ʹ end of the ncRNA; and the conserved nucleotides are as shown in FIG.9, and wherein the colored circled nucleotides are present at the respective levels of certainty (e.g., at least about 97% of the red-circled nucleotides, at least about 90-97% of the black-circled nucleotides, at least about 75-90% of the grey-circled nucleotides, and at least about 50% of the white-circled nucleotides are present). [00321] In some embodiments, the engineered retron is entirely synthetically produced and having the conserved nucleotides as denoted by the colored letters as shown in FIG.9. Type IIA5 novel retron (FIG.10) [00322] In some embodiments, the template/wt retron for the subject engineered retron encodes a wild-type retron ncRNA polynucleotide having a consensus secondary structure that can be described as: a1-S1-L1-S2-L2-S3-L3-S4-a2, wherein: a1/a2 is a stem 15 bps in length with a 1-nt overhang on a1, a 13-nt overhang on a2, and a 7/5 bulge 13-nt from the tip; L1 is a stem of 10 bps with a 3-nt tip; L2 is a stem of 35 bps with a 3-nt tip; L3 is a stem of 6 bps with a 5-nt tip; S1 is a single-stranded spacer region between a1/a2 and L1; S2 is a single-stranded spacer region between L1 and L2; S3 is a single-stranded spacer region between L2 and L3; S4 is a single-stranded spacer region between L3 and a1/a2, the conserved nucleotides are as shown in FIG.10, and wherein the colored circled nucleotides are present at the respective levels of certainty (e.g., at least about 97% of the red-circled nucleotides, at least about 90-97% of the black-circled nucleotides, at least about 75-90% of the grey-circled nucleotides, and at least about 50% of the white-circled nucleotides are present). [00323] In some embodiments, the engineered retron is entirely synthetically produced and having the conserved nucleotides as denoted by the colored letters as shown in FIG.10. Type IIIA1 retron (FIG.11) [00324] In some embodiments, the template/wt retron for the subject engineered retron encodes a wild-type retron ncRNA polynucleotide having a consensus secondary structure that can be described as: a1-S1-L1-L2-S2-L3-S3-a2, wherein: a1/a2 is a stem 2 bps in length with a 1-nt overhang on a2; L1 is a stem of 8 bps with a 4-nt tip; L2 is a stem of 9 bps with a 3-nt tip and a 1/1 bulge 3 bps from the tip; L3 is a stem of 20 bps with a 3-nt tip and a 1/2 bulge 3 bps from the tip; S1 is a single-stranded spacer region between a1/a2 and L1; S2 is a single-stranded spacer region between L2 and L3; S3 is a single-stranded spacer region between L3 and a1/a2, the conserved nucleotides are as shown in FIG.11, and wherein the colored circled nucleotides are present at the respective levels of certainty (e.g., at least about 97% of the red-circled nucleotides, at least about 90-97% of the black-circled nucleotides, at least about 75-90% of the grey-circled nucleotides, and at least about 50% of the white-circled nucleotides are present). [00325] In some embodiments, the engineered retron is entirely synthetically produced and having the conserved nucleotides as denoted by the colored letters as shown in FIG.11. Type IIIA2 retron (FIG.12) [00326] In some embodiments, the template/wt retron for the subject engineered retron encodes a wild-type retron ncRNA polynucleotide having a consensus secondary structure that can be described as: a1-S1-L1-S2-L2-S3-L3-S4-L4-S5-a2, wherein: a1/a2 is a stem 15 bp in length; L1 is a stem of 6 bps with a 4-nt tip; L2 is a stem of 13 bps with a 5-nt tip; L3 is a stem of 4 bps with a 8-nt tip; L4 is a stem of 20 bps with a 4-nt tip and a 2/2 bulge 6 bp from the tip; S1 is a single-stranded spacer region between a1/a2 and L1; S2 is a single-stranded spacer region between L1 and L2; S3 is a single-stranded spacer region between L2 and L3; S4 is a single-stranded spacer region between L3 and L4; S5 is a single-stranded spacer region between L4 and a1/a2; the conserved nucleotides are as shown in FIG.12, and wherein the colored circled nucleotides are present at the respective levels of certainty (e.g., at least about 97% of the red-circled nucleotides, at least about 90-97% of the black-circled nucleotides, at least about 75-90% of the grey-circled nucleotides, and at least about 50% of the white-circled nucleotides are present). [00327] In some embodiments, the engineered retron is entirely synthetically produced and having the conserved nucleotides as denoted by the colored letters as shown in FIG.12. Type IIIA3 retron (FIG.13) [00328] In some embodiments, the template/wt retron for the subject engineered retron encodes a wild-type retron ncRNA polynucleotide having a consensus secondary structure that can be described as: a1-S1-L1-S2-L2-S3-L3-S4-L4-L5-L6-S5-a2, wherein: a1/a2 is a stem 24 bps in length and having a 1/0 bulge 15 bps from the tip, and a 1/1 bulge 19 bps from the tip; L1 is a stem of 7 bps with a 4-nt tip; L2 is a stem of 9 bps with a 8-nt tip; L3 is a stem of 8 bps with a 4-nt tip; L4 is a stem of 4 bps with a 9-nt tip, and a 2/2 bulge 3 bps from the tip; L5 is a stem of 19 bps with a 18-nt tip; L6 is a stem of 5 bps with a 3-nt tip; S1 is a single-stranded spacer region between a1/a2 and L1; S2 is a single-stranded spacer region between L1 and L2; S3 is a single-stranded spacer region between L2 and L3; S4 is a single-stranded spacer region between L3 and L4; S5 is a single-stranded spacer region between L6 and a1/a2; the conserved nucleotides are as shown in FIG.13, and wherein the colored circled nucleotides are present at the respective levels of certainty (e.g., at least about 97% of the red-circled nucleotides, at least about 90-97% of the black-circled nucleotides, at least about 75-90% of the grey-circled nucleotides, and at least about 50% of the white-circled nucleotides are present). [00329] In some embodiments, the engineered retron is entirely synthetically produced and having the conserved nucleotides as denoted by the colored letters as shown in FIG.13. Type IIIA4 retron (FIG.14) [00330] In some embodiments, the template/wt retron for the subject engineered retron encodes a wild-type retron ncRNA polynucleotide having a consensus secondary structure that can be described as: a1-S1-L1-S2-L2-S3-L3-S4-a2, wherein: a1/a2 is a stem 5 bps in length with a 1/2 bulge 2 bps from the tip; L1 is a stem of 8 bps with a 6-nt tip; L2 is a stem of 8 bps with a 5-nt tip; L3 is a stem of 13 bps with a 14-nt tip and a 1/0 bulge 2 bps from the tip; S1 is a single-stranded spacer region between a1/a2 and L1; S2 is a single-stranded spacer region between L1 and L2; S3 is a single-stranded spacer region between L2 and L3: S4 is a single-stranded spacer region between L3 and a1/a2, the conserved nucleotides are as shown in SEQ ID NO: 19 and FIG.20, and wherein the colored circled nucleotides are present at the respective levels of certainty (e.g., at least about 97% of the red-circled nucleotides, at least about 90-97% of the black-circled nucleotides, at least about 75- 90% of the grey-circled nucleotides, and at least about 50% of the white-circled nucleotides are present). [00331] In some embodiments, the engineered retron is entirely synthetically produced and having the conserved nucleotides as denoted by the colored letters as shown in FIG.14. Type IIIA5 retron (FIG.15) [00332] In some embodiments, the template/wt retron for the subject engineered retron encodes a wild-type retron ncRNA polynucleotide having a consensus secondary structure that can be described as: a1-S1-L1-S2-L2-L3-L4-S3-a2, wherein: the a1/a2 stem is 11 bp in length with no overhang; L1 is a stem of 9 bps with a 3-nt tip; L2 is a stem of 14 bps with a 5-nt tip; L3 is a stem of 9 bps with a 7-nt tip; L4 is a stem of 15 bps with a 7-nt tip; S1, S2, and S3 are single-stranded spacer regions between the a1/a2 stem and L1, L2 and L3, and L4 and the a1/a2 stem, respectively, with no spacer between L1 and L2, and no spacer between L2 and L3; wherein the 5th-2nd last nts of S2 and the 3rd-6th nts of S3 forms a 4-bp stem, and, the conserved nucleotides are as shown in FIG.15, and wherein the colored circled nucleotides are present at the respective levels of certainty (e.g., at least about 97% of the red-circled nucleotides, at least about 90-97% of the black-circled nucleotides, at least about 75-90% of the grey-circled nucleotides, and at least about 50% of the white-circled nucleotides are present). [00333] In some embodiments, the engineered retron is entirely synthetically produced and having the conserved nucleotides as denoted by the colored letters as shown in FIG.15. Type IIIunk retron (FIG.16) [00334] In some embodiments, the template/wt retron for the subject engineered retron encodes a wild-type retron ncRNA polynucleotide having a consensus secondary structure that can be described as: a1-S1-L1-S2-L2-S3-L3-S4-a2, wherein: a1/a2 is a stem 11 bps in length; L1 is a stem of 12 bps with a 2-nt tip; L2 is a stem of 21 bps with a 1-nt tip; L3 is a stem of 20 bps with a 4-nt tip; S1 is a single-stranded spacer region between a1/a2 and L1; S2 is a single-stranded spacer region between L1 and L2; S3 is a single-stranded spacer region between L2 and L3; S4 is a single-stranded spacer region between L3 and a1/a2, the conserved nucleotides are as shown in FIG.16, and wherein the colored circled nucleotides are present at the respective levels of certainty (e.g., at least about 97% of the red-circled nucleotides, at least about 90-97% of the black-circled nucleotides, at least about 75-90% of the grey-circled nucleotides, and at least about 50% of the white-circled nucleotides are present). [00335] In some embodiments, the engineered retron is entirely synthetically produced and having the conserved nucleotides as denoted by the colored letters as shown in FIG.16. Type IV retron (FIG.17) [00336] In some embodiments, the template/wt retron for the subject engineered retron encodes a wild-type retron ncRNA polynucleotide having a consensus secondary structure that can be described as: a1-S1-L1-L2-S2-L3-S3-a2, wherein: a1/a2 is a stem 9 bp in length with no overhang; L1 is a stem of 5 bps with a 6-nt tip; L2 is a stem of 9 bps with a 4-nt tip; L3 is a stem of 26 bps with a 5-nt tip, a 0/1 bulge 7 bps from the tip, and a 0/1 bulge 9 bps from the tip; S1 is a single-stranded spacer region between the a1/a2 stem and L1, with no spacer region between L1 and L2; S2 is a single-stranded spacer region between L2 and L3; and, S3 is a single-stranded spacer region between L3 and the a1/a2 stem; the conserved nucleotides are as shown in FIG.17, and wherein the colored circled nucleotides are present at the respective levels of certainty (e.g., at least about 97% of the red-circled nucleotides, at least about 90-97% of the black-circled nucleotides, at least about 75-90% of the grey-circled nucleotides, and at least about 50% of the white-circled nucleotides are present). [00337] In some embodiments, the engineered retron is entirely synthetically produced and having the conserved nucleotides as denoted by the colored letters as shown in FIG.17. Type IX retron (FIG.18) [00338] In some embodiments, the template/wt retron for the subject engineered retron encodes a wild-type retron ncRNA polynucleotide having a consensus secondary structure that can be described as: a1-L1-S1-L2-S2-a2, wherein: a1/a2 is a stem 12 bp in length, wherein a1 has a 14-nt overhang and a2 has a 2-nt overhang; L1 is a stem of 11 bps with a 3-nt tip and a 1/3 bulge 7 bp from the tip; L2 is a stem of 25 bps with a 7-nt tip; S1 is a single-stranded spacer region between L1 and L2; S2 is a single-stranded spacer region between L2 and a1/a2; the conserved nucleotides are as shown in FIG.18, and wherein the colored circled nucleotides are present at the respective levels of certainty (e.g., at least about 97% of the red-circled nucleotides, at least about 90-97% of the black-circled nucleotides, at least about 75-90% of the grey-circled nucleotides, and at least about 50% of the white-circled nucleotides are present). [00339] In some embodiments, the engineered retron is entirely synthetically produced and having the conserved nucleotides as denoted by the colored letters as shown in FIG.18. Type V retron (FIG.19) [00340] In some embodiments, the template/wt retron for the subject engineered retron encodes a wild-type retron ncRNA polynucleotide having a consensus secondary structure that can be described as: a1-S1-L1-S2-L2-S3-a2, wherein: a1/a2 is a stem 13 bps in length; L1 is a stem of 20 bps with a 4-nt tip and a 6/4 bulge 6 bps from the tip; L2 is a stem of 14 bps with a 4-nt tip and a 1/0 bulge 5 bps from the tip; S1 is a single-stranded spacer region between a1/a2 and L1; S2 is a single-stranded spacer region between L1 and L2; S3 is a single-stranded spacer region between L2 and a1/a2, the conserved nucleotides are as shown in FIG.19, and wherein the colored circled nucleotides are present at the respective levels of certainty (e.g., at least about 97% of the red-circled nucleotides, at least about 90-97% of the black-circled nucleotides, at least about 75-90% of the grey-circled nucleotides, and at least about 50% of the white-circled nucleotides are present). [00341] In some embodiments, the engineered retron is entirely synthetically produced and having the conserved nucleotides as denoted by the colored letters as shown in FIG.19. Type VI retron (FIG.20) [00342] In some embodiments, the template/wt retron for the subject engineered retron encodes a wild-type retron ncRNA polynucleotide having a consensus secondary structure that can be described as: a1-S1-L1-S2-L2-S3-L3-L4-S4-a2, wherein: a1/a2 is a stem 4 bp in length with a 1 bp 5ʹ overhang; L1 is a stem of 7 bps with a 4-nt tip; L2 is a stem of 8 bps with a 4-nt tip; L3 is a stem of 16 bps with a 6-nt tip, a 3/4 bulge 3 bps from the tip, a 2/3 bulge 5 bps from the tip, and a 3/1 bulge 8 bps from the tip; S1 is a single-stranded spacer region between the a1/a2 stem and L1; S2 is a single-stranded spacer region between L1 and L2; S3 is a single-stranded spacer region between L2 and L3, with no spacer region between L3 and L4; S4 is a single-stranded spacer region between L4 and the a1/a2 stem; and, the conserved nucleotides are as shown in FIG.20, and wherein the colored circled nucleotides are present at the respective levels of certainty (e.g., at least about 97% of the red-circled nucleotides, at least about 90-97% of the black-circled nucleotides, at least about 75-90% of the grey-circled nucleotides, and at least about 50% of the white-circled nucleotides are present). [00343] In some embodiments, the engineered retron is entirely synthetically produced and having the conserved nucleotides as denoted by the colored letters as shown in FIG.20. Type XI Group 1 retron (FIG.21) [00344] In some embodiments, the template/wt retron for the subject engineered retron encodes a wild-type retron ncRNA polynucleotide having a consensus secondary structure that can be described as: a1-S1-L1-S2-L2-S3-a2, wherein: a1/a2 is a stem 16 bps in length with a 5-nt overhang on a1, and a 3-nt overhang on a2; L1 is a stem of 9 bps with a 3-nt tip; L2 is a stem of 7 bps with a 13-nt tip; S1 is a single-stranded spacer region between a1/a2 and L1; S2 is a single-stranded spacer region between L1 and L2; S3 is a single-stranded spacer region between L2 and a1/a2, the conserved nucleotides are as shown in FIG.21, and wherein the colored circled nucleotides are present at the respective levels of certainty (e.g., at least about 97% of the red-circled nucleotides, at least about 90-97% of the black-circled nucleotides, at least about 75-90% of the grey-circled nucleotides, and at least about 50% of the white-circled nucleotides are present). [00345] In some embodiments, the engineered retron is entirely synthetically produced and having the conserved nucleotides as denoted by the colored letters as shown in FIG.21. Type XI Group 2 retron (FIG.22) [00346] In some embodiments, the template/wt retron for the subject engineered retron encodes a wild-type retron ncRNA polynucleotide having a consensus secondary structure that can be described as: a1-S1-L1-S2-L2-S3-a2, wherein: a1/a2 is a stem 13 bps in length with a 1-nt overhang on a2; L1 is a stem of 7 bps with a 3-nt tip and a 2/2 bulge 1 bp from the tip; L2 is a stem of 8 bps with a 20-nt tip; S1 is a single-stranded spacer region between a1/a2 and L1; S2 is a single-stranded spacer region between L1 and L2; S3 is a single-stranded spacer region between L2 and a1/a2, the conserved nucleotides are as shown in FIG.22, and wherein the colored circled nucleotides are present at the respective levels of certainty (e.g., at least about 97% of the red-circled nucleotides, at least about 90-97% of the black-circled nucleotides, at least about 75-90% of the grey-circled nucleotides, and at least about 50% of the white-circled nucleotides are present). [00347] In some embodiments, the engineered retron is entirely synthetically produced and having the conserved nucleotides as denoted by the colored letters as shown in FIG.22. Type XII retron (FIG.23) [00348] In some embodiments, the template/wt retron for the subject engineered retron encodes a wild-type retron ncRNA polynucleotide having a consensus secondary structure that can be described as: a1-S1-L1-S2-L2-S3-a2, wherein: a1/a2 is a stem 13 bps in length with a 1-nt overhang on a2; L1 is a stem of 7 bps with a 3-nt tip, and a 2/2 bulge 1 bp from the tip; L2 is a stem of 8 bps with a 19-nt tip; S1 is a single-stranded spacer region between a1/a2 and L1; S2 is a single-stranded spacer region between L1 and L2; S3 is a single-stranded spacer region between L2 and a1/a2, the conserved nucleotides are as shown in FIG.23, and wherein the colored circled nucleotides are present at the respective levels of certainty (e.g., at least about 97% of the red-circled nucleotides, at least about 90-97% of the black-circled nucleotides, at least about 75-90% of the grey-circled nucleotides, and at least about 50% of the white-circled nucleotides are present). [00349] In some embodiments, the engineered retron is entirely synthetically produced and having the conserved nucleotides as denoted by the colored letters as shown in FIG.23. Type XIII retron (FIG.24) [00350] In some embodiments, the template/wt retron for the subject engineered retron encodes a wild-type retron ncRNA polynucleotide having a consensus secondary structure that can be described as: a1-S1-L1-S2-L2-L3-S3-a2, wherein: a1/a2 is a stem 7 bp in length with no overhangs; L1 is a stem of 8 bps with a 3-nt tip; L2 is a stem of 30 bps with a 8-nt tip, a 1/1 bulge 2 bps from the tip, and a 1/1 bulge 27 bps from the tip; L3 is a stem of 8 bps with a 5-nt tip, and a 0/1 bulge 3 nt from the tip; S1 is a single-stranded spacer region between the a1/a2 stem and L1; S2 is a single-stranded spacer region between L1 and L2; and, S3 is a single-stranded spacer region between L3 and the a1/a2 stem; the conserved nucleotides are as shown in FIG.24, and wherein the colored circled nucleotides are present at the respective levels of certainty (e.g., at least about 97% of the red-circled nucleotides, at least about 90-97% of the black-circled nucleotides, at least about 75-90% of the grey-circled nucleotides, and at least about 50% of the white-circled nucleotides are present). [00351] In some embodiments, the engineered retron is entirely synthetically produced and having the conserved nucleotides as denoted by the colored letters as shown in FIG.24. Type XIV retron (FIG.25) [00352] In some embodiments, the template/wt retron for the subject engineered retron encodes a wild-type retron ncRNA polynucleotide having a consensus secondary structure that can be described as: a1-S1-L1-L2-S2-L3-S3-a2, wherein: the a1/a2 stem is 15 bp in length with no overhang, and a 4/2 bulge 7 bps from the 5ʹ end of a1; L1 is a stem of 8 bps with a 5-nt tip; L2 is a stem of 7 bps with a 5-nt tip; L3 is a stem of 13 bps with a 2-nt tip, a 5/9 bulge 5 bps from the tip, and a 5/5 bulge 8 bps from the tip; S1, S2, and S3 are single-stranded spacer regions between the a1/a2 stem and L1, L2 and L3, and L3 and the a1/a2 stem, respectively, with no spacer between L1 and L2; and wherein the 5th-3rd last nts of S1 and the 2nd-5th 4 nts of S2 forms a 3-bp stem; and, the conserved nucleotides are as shown in FIG.25, and wherein the colored circled nucleotides are present at the respective levels of certainty (e.g., at least about 97% of the red-circled nucleotides, at least about 90-97% of the black-circled nucleotides, at least about 75-90% of the grey-circled nucleotides, and at least about 50% of the white-circled nucleotides are present). [00353] In some embodiments, the engineered retron is entirely synthetically produced and having the conserved nucleotides as denoted by the colored letters as shown in FIG.25. Ec107-like retron (FIG.26) [00354] In some embodiments, the template/wt retron for the subject engineered retron encodes a wild-type retron ncRNA polynucleotide having a consensus secondary structure that can be described as: a1-S1-L1-S2-L2-S3-L3-S4-a2, wherein: a1/a2 is a stem 12 bp in length; L1 is a stem of 4 bps with a 8-nt tip; L2 is a stem of 8 bps with a 3-nt tip; L3 is a stem of 22 bps with a 3-nt tip, a 4/6 bulge 6 bp from the tip, a 3/3 bulge 13 bp from the tip, and a 1/1 bulge 18 bp from the tip; S1 is a single-stranded spacer region between a1/a2 and L1; S2 is a single-stranded spacer region between L1 and L2; S3 is a single-stranded spacer region between L2 and L3; and S4 is a single-stranded spacer region between L3 and a1/a2, the conserved nucleotides are as shown in FIG.26, and wherein the colored circled nucleotides are present at the respective levels of certainty (e.g., at least about 97% of the red-circled nucleotides, at least about 90-97% of the black-circled nucleotides, at least about 75-90% of the grey-circled nucleotides, and at least about 50% of the white-circled nucleotides are present). [00355] In some embodiments, the engineered retron is entirely synthetically produced and having the conserved nucleotides as denoted by the colored letters as shown in FIG.26. Outgroup A retron (FIG.27) [00356] In some embodiments, the template/wt retron for the subject engineered retron encodes a wild-type retron ncRNA polynucleotide having a consensus secondary structure that can be described as: a1-S1-L1-S2-L2-S3-L3-S4-a2, wherein: a1/a2 is a stem 5 bps in length with a 2-nt overhang on a2; L1 is a stem of 11 bps with a 4-nt tip; L2 is a stem of 8 bps with a 3-nt tip; L3 is a stem of 18 bps with a 3-nt tip; S1 is a single-stranded spacer region between a1/a2 and L1; S2 is a single-stranded spacer region between L1 and L2; S3 is a single-stranded spacer region between L2 and L3; S4 is a single-stranded spacer region between L3 and a1/a2, the conserved nucleotides are as shown in FIG.27, and wherein the colored circled nucleotides are present at the respective levels of certainty (e.g., at least about 97% of the red-circled nucleotides, at least about 90-97% of the black-circled nucleotides, at least about 75-90% of the grey-circled nucleotides, and at least about 50% of the white-circled nucleotides are present). [00357] In some embodiments, the engineered retron is entirely synthetically produced and having the conserved nucleotides as denoted by the colored letters as shown in FIG.27. [00358] Amplification of an engineered retron described herein (e.g., in FIGs.2-27) may be performed, for example, before transfection of cells or ligation into vectors. Any method for amplifying the engineered retron may be used, including, but not limited to polymerase chain reaction (PCR), isothermal amplification, nucleic acid sequence-based amplification (NASBA), transcription mediated amplification (TMA), strand displacement amplification (SDA), and ligase chain reaction (LCR). In one embodiment, the engineered retron comprise common 5ʹ and 3ʹ priming sites to allow amplification of retron sequences in parallel with a set of universal primers. In another embodiment, a set of selective primers is used to selectively amplify a subset of retron sequences from a pooled mixture. [00359] Variants of these templates, which can also be used in the engineered retron of the invention, include a variant having: A) up to 1, 2, or 3 (e.g., up to 1) nucleotide changes per 10 red lettered-nucleotides; B) up to 4, 5, or 6 (e.g., up to 1 or 2) nucleotide changes per 10 black lettered-nucleotides; and/or C) up to 7, 8, or 9 (e.g., up to 3 or 4) nucleotide changes per 10 grey lettered-nucleotides; and/or optionally further comprising: a) 7, 8, 9, or 10 (e.g., 9 or 10) nucleotides present per 10 red-circled nucleotides; b) 6, 7, 8, 9, or 10 (e.g., 8, 9 or 10) nucleotides present per 10 black-circled nucleotides; c) 4, 5, 6, 7, 8, 9, or 10 (e.g., 6, 7, 8, 9 or 10) nucleotides present per 10 grey-circled nucleotides; and/or d) 2, 3, 4, 5, 6, 7, 8, 9, or 10 (e.g., 4, 5, 6, 7, 8, 9, or 10) nucleotides present per 10 white-circled nucleotides. [00360] In some embodiments, the non-coding RNA (ncRNA) portion of the engineered retron comprises a polynucleotide (e.g., a DNA molecule) encoding an ncRNA listed in Table B, or an ncRNA having at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%%, at least 99.1%, at least 99.2%, at least 99.3%, at least 99.4%, at least 99.5%, at least 99.6%, at least 99.7%, at least 99.8% or at least 99.9% sequence identity to an ncRNA listed in Table B. In some embodiments, the ncRNA does not comprise an ncRNA associated with the RT sequences of Table X. B. Heterologous nucleotide sequence (HNS) [00361] The engineered retron may comprise or encodes a heterologous nucleic acid (e.g., DNA or RNA) within the msr locus or the msd locus (such as in an S region or the tip of an L region in the consensus structure in any one of FIGs.2-27, and variants thereof), or upstream or downstream of either the msr locus or the msd locus. In some embodiments, the heterologous nucleic acid is inserted within the msd locus. In some embodiments, the heterologous nucleic acid is inserted upstream of the msr locus. In some embodiments, the heterologous nucleic acid is inserted upstream or downstream of the msd locus. In other embodiments, the heterologous nucleic acid is inserted in spacer sequences (e.g., S1, S2, S3 and/or S4 as depicted in FIG.2) between hairpin loops and/or within hairpin loops (e.g., L1, L2, L3 and/or L4 as depicted in FIG. 2), e.g., the tip or loop region, or within a bulge. In some embodiments, the heterologous nucleic acid sequence can be inserted in the tip of an L region or hairpin loop (e.g., L1, L2, L3 and/or L4 as depicted in FIG.2). In some embodiments, one or more heterologous nucleic acids are inserted into the msd locus. In some embodiments, a first heterologous nucleic acid is inserted into the msd locus, and a second heterologous nucleic acid is inserted either upstream of the msr locus or upstream or downstream of the msd locus. [00362] In some embodiments, the heterologous nucleic acid comprises flanking contiguous nucleotides (also referred to as homologous arms) that are substantially complementary to a target site genomic sequence of a cell to facilitate insertion of at least part of the heterologous nucleic acid into the genome of the cell at the target site via homology directed repair (HDR). In some embodiments, the heterologous nucleic acid is between >20 nucleotides and 10,000 nucleotides (e.g., including the flanking homologous arms). [00363] In some embodiments, one or both of the homology arm(s) on the heterologous nucleic acid are 100% identical to a target genomic sequence. In some embodiments, one or both of the homology arm(s) on the heterologous nucleic acid are less than 100% complementary to the target genomic sequence, for example, 99%, 98%, 97%, 96%, 95%, 94%, 93%, 92%, 91%, or 90% identical to a target genomic sequence. [00364] The part of the heterologous nucleic acid to be inserted into the target genome sequence is sometimes referred to as a donor sequence. The donor sequence may be partially identical to the full length or portions thereof of a target genomic sequence, or may be unrelated to the target genomic sequence. The donor sequence may be used, for example, to introduce modifications (e.g., substitutions, deletions, insertions, or a combination thereof) such as mutations or other genetic changes (e.g., genetic elements such as stop codons or a shift in an open reading frame on the target polynucleotide) into its target sequence. [00365] In some embodiments, the heterologous nucleic acid sequence is or encodes a biologically active molecule such as, but not limited to, a therapeutic protein. [00366] Any therapeutic proteins may be encoded by the heterologous nucleic acid. [00367] In some embodiments, the heterologous nucleic acid sequences encodes one or more prophylactically- or therapeutically-active proteins, polypeptides, or other factors. [00368] As a non-limiting example, the heterologous sequences may be or encode an agent that enhances tumor killing activity such as, but not limited to, TRAIL or tumor necrosis factor (TNF), in a cancer. As another non-limiting example, the heterologous sequences may be or encode an agent suitable for the treatment of conditions such as muscular dystrophy (e.g., heterologous sequences are or encode dystrophin or a functional fragment or variant thereof such as the numerous dystrophin minigenes or microdystrophine coding sequences known in the art), cardiovascular disease (e.g., heterologous sequences are or encode SERCA2a, GATA4, Tbx5, Mef2C, Hand2, Myocd, etc.), neurodegenerative disease (e.g., heterologous sequences is or encodes NGF, BDNF, GDNF, NT-3, etc.). [00369] As additional non-limiting example, the heterologous nucleic acid sequence may be or encode an agent that enhances tumor killing activity such as, but not limited to, TRAIL or tumor necrosis factor (TNF), in a cancer. As another non-limiting example, the heterologous nucleic acid sequence may be or encode an agent suitable for the treatment of conditions such as muscular dystrophy (e.g., heterologous nucleic acid sequence is or encodes Dystrophin), cardiovascular disease (e.g., heterologous nucleic acid sequence is or encodes SERCA2a, GATA4, Tbx5, Mef2C, Hand2, Myocd, etc.), neurodegenerative disease (e.g., heterologous nucleic acid sequence is or encodes NGF, BDNF, GDNF, NT-3, etc.), chronic pain (e.g., heterologous nucleic acid sequence is or encodes GlyRal), an enkephalin, or a glutamate decarboxylase (e.g., heterologous nucleic acid sequence is or encodes GAD65, GAD67, or another isoform), lung disease (e.g., heterologous nucleic acid sequence is or encodes CFTR), hemophilia (e.g., heterologous nucleic acid sequence is or encodes Factor VIII or Factor IX), neoplasia (e.g., heterologous nucleic acid sequence is or encodes PTEN, ATM, ATR, EGFR, ERBB2, ERBB3, ERBB4, Notchl, Notch2, Notch3, Notch4, AKT, AKT2, AKT3, HIF, HI Fla, HIF3a, Met, HRG, Bcl2, PPARalpha, PPAR gamma, WT1 (Wilms Tumor), FGF Receptor Family members (5 members: 1, 2, 3, 4, 5), CDKN2a, APC, RB (retinoblastoma), MEN1, VHL, BRCA1, BRCA2, AR (Androgen Receptor), TSG101, IGF, IGF Receptor, Igfl (4 variants), Igf2 (3 variants), Igfl Receptor, Igf2 Receptor, Bax, Bcl2, caspases family (9 members: 1, 2, 3, 4, 6, 7, 8, 9, 12), Kras, Ape), age-related macular degeneration (e.g., heterologous nucleic acid sequence is or encodes Aber, Ccl2, Cc2, cp (ceruloplasmin), Timp3, cathepsin D, Vldlr), schizophrenia (e.g., Neuregulin (Nrgl), Erb4 (receptor for Neuregulin), Complexin-l (Cplxl), Tphl Tryptophan hydroxylase, Tph2 Tryptophan hydroxylase 2, Neurexin 1, GSK3, GSK3a, GSK3b, 5-HIT (Slc6a4), COMT, DRD (Drdla), SLC6A3, DAOA, DTNBPI, Dao (Daol)), trinucleotide repeat disorders (e.g., HTT (Huntington's Dx), SBMA/SMAXI/AR (Kennedy's Dx), FXN/X25 (Friedrich's Ataxia), ATX3 (Machado-Joseph's Dx), ATXNI and ATXN2 (spinocerebellar ataxias), DMPK (myotonic dystrophy), Atrophin-1 and Atnl(DRPLA Dx), CBP (Creb-BP-global instability), VLDLR (Alzheimer's), Atxn7, Atxn10), fragile X syndrome (e.g., heterologous nucleic acid sequence is or encodes FMR2, FXRI, FXR2, mGLUR5), secretase related disorders (e.g., heterologous nucleic acid sequence is or encodes APH-1 (alpha and beta), Presenilin (Psenl), nicastrin (Ncstn), PEN-2), ALS (e.g., heterologous nucleic acid sequence is or encodes SOD1, ALS2, STEX, FUS, TARD BP, VEGF (VEGF-a, VEGF-b, VEGF-c)), autism (e.g., heterologous nucleic acid sequence is or encodes Mecp2, BZRAP1, MDGA2, Sema5A, Neurexin 1), Alzheimer's disease (e.g., heterologous nucleic acid sequence is or encodes El, CHIP, UCH, UBB, Tau, LRP, PICALM, Clusterin, PS1, SORL1, CR1, Vldlr, Ubal, Uba3, CHIP28 (Aqpl, Aquaporin 1), Uchll, Uchl3, APP), inflammation (e.g., heterologous nucleic acid sequence is or encodes IL-10, IL-1 (IL-Ia, IL-Ib), IL-13, IL-17 (IL-17a (CTLA8), IL-17b, IL- 17c, IL-17d, IL-171), 11-23, Cx3crl, ptpn22, TNFa, NOD2/CARD15 for IBD, IL-6, IL-12 (IL- 12a, IL-12b), CTLA4, Cx3cll), Parkinson's Disease (e.g., x-Synuclein, DJ-1, LRRK2, Parkin, PINK1), blood and coagulation disorders, such as, e.g., anemia, bare lymphocyte syndrome, bleeding disorders, hemophagocytic lymphohistiocytosis disorders, hemophilia A, hemophilia B, hemorrhagic disorders, leukocyte deficiencies and disorders, sickle cell anemia, and thalassemia (e.g., heterologous nucleic acid sequence is or encodes CRAN1, CDA1, RPS19, DBA, PKLR, PK1, NT5C3, UMPH1, PSNI, RHAG, RH50A, NRAMP2, SPTB, ALAS2, ANH1, ASB, ABCB7, ABC7, ASAT, TAPBP, TPSN, TAP2, ABCB3, PSF2, RING11, MHC2TA, C2TA, RFX5, RFXAP, RFX5, TBXA2R, P2RX1, P2X1, HF1, CFH, HUS, MCFD2, FANCA, FAC A, FA1, FA, FA A, FAAP95, FAAP90, FLJ34064, FANCB, FANCC, FACC, BRCA2, FANCDI, FANCD2, FANCD, FACD, FAD, FANCE, FACE, FANCF, XRCC9, FANCG, BR1PI, BACH1, FANCJ, PHF9, FANCL, FANCM, KIAA1596, PRF1, HPLH2, UNC13D, MUNC13-4, HPLH3, HLH3, FHL3, F8, FSC, PI, ATT, F5, ITGB2, CD18, LCAMB, LAD, EIF2B1, EIF2BA, EIF2B2, EIF2B3, EIF2B5, LVWM, CACH, CLE, EIF2B4, HBB, HBA2, HBB, HBD, LCRB, HBA1), B- cell non-Hodgkin lymphoma or leukemia (e.g., heterologous nucleic acid sequence is or encodes BCL7A, BCL7, ALI, TCL5, SCL, TAL2, FLT3, NBS1, NBS, ZNFN1AI, 1KI, LYF1, HOXD4, HOX4B, BCR, CML, PHL, ALL, ARNT, KRAS2, RASK2, GMPS, AFIO, ARHGEF12, LARG, KIAA0382, CALM, CLTH, CEBPA, CEBP, CHIC2, BTL, FLT3, KIT, PBT, LPP, NPMI, NUP214, D9S46E, CAN, CAIN, RUNXI, CBFA2, AML1, WHSC1LI, NSD3, FLT3, AF1Q, NPMI, NUMA1, ZNF145, PLZF, PML, MYL, STAT5B, AF1Q, CALM, CLTH, ARL11, ARLTS1, P2RX7, P2X7, BCR, CML, PHL, ALL, GRAF, NF1, VRNF, WSS, NFNS, PTPNII, PTP2C, SHP2, NS1, BCL2, CCND1, PRAD1, BCL1, TCRA, GATA1, GF1, ERYF1, NFE1, ABLI, NQO1, DIA4, NMOR1, NUP214, D9S46E, CAN, CAIN), inflammation and immune related diseases and disorders (e.g., heterologous nucleic acid sequence is or encodes KIR3DL1, NKAT3, NKB1, AMB11, K1R3DS1, IFNG, CXCL12, TNFRSF6, APT1, FAS, CD95, ALPS1A, IL2RG, SCIDX1, SCIDX, IMD4, CCL5, SCYA5, D17S136E, TCP228, IL10, CSIF, CMKBR2, CCR2, CMKBR5, CCCKR5 (CCR5), CD3E, CD3G, AICDA, AID, HIGM2, TNFRSF5, CD40, UNG, DGU, HIGM4, TNFSFS, CD40LG, HIGM1, IGM, FOXP3, IPEX, AIID, XPID, PIDX, TNFRSF14B, TACI), inflammation (e.g., heterologous nucleic acid sequence is or encodes IL-10, IL-1 (IL-IA, IL-IB), IL-13, IL-17 (IL-17a (CTLA8), IL-17b, IL- 17c, IL-17d, IL-171), 11-23, Cx3crl, ptpn22, TNFa, NOD2/CARD15 for IBD, IL-6, IL-12 (IL- 12a, IL-12b), CTLA4, Cx3cII), JAK3, JAKL, DCLREIC, ARTEMIS, SCIDA, RAG1, RAG2, ADA, PTPRC, CD45, LCA, IL7R, CD3D, T3D, IL2RG, SCIDXI, SCIDX, IMD4), metabolic, liver, kidney and protein diseases and disorders (e.g., heterologous nucleic acid sequence is or encodes TTR, PALB, APOA1, APP, AAA, CVAP, ADI, GSN, FGA, LYZ, TTR, PALB, KRT18, KRT8, CIRH1A, NAIC, TEX292, KIAA1988, CFTR, ABCC7, CF, MRP7, SLC2A2, GLUT2, G6PC, G6PT, G6PT1, GAA, LAMP2, LAMPB, AGL, GDE, GBE1, GYS2, PYGL, PFKM, TCF1, HNF1A, MODY3, SCOD1, SCOl, CTNNB1, PDGFRL, PDGRL, PRLTS, AX1NI, AXIN, CTNNB1, TP53, P53, LFS1, IGF2R, MPRI, MET, CASP8, MCH5, UMOD, HNFJ, FJHN, MCKD2, ADMCKD2, PAH, PKU1, QDPR, DHPR, PTS, FCYT, PKHD1, ARPKD, PKD1, PKD2, PKD4, PKDTS, PRKCSH, G19P1, PCLD, SEC63), muscular/skeletal diseases and disorders (e.g., heterologous nucleic acid sequence is or encodes DMD, BMD, MYF6, LMNA, LMN1, EMD2, FPLD, CMDIA, HGPS, LGMDIB, LMNA, LMNI, EMD2, FPLD, CMDIA, FSHMD1A, FSHD1A, FKRP, MDC1C, LGMD2I, LAMA2, LAMM, LARGE, KIAA0609, MDC1D, FCMD, TTID, MYOT, CAPN3, CANP3, DYSF, LGMD2B, SGCG, LGMD2C, DMDA1, SCG3, SGCA, ADL, DAG2, LGMD2D, DMDA2, SGCB, LGMD2E, SGCD, SGD, LGMD2F, CMD1L, TCAP, LGMD2G, CMD1N, TRIM32, HT2A, LGMD2H, FKRP, MDCIC, LGMD21, TTN, CMD1G, TMD, LGMD2J, POMT1, CAV3, LGMD1C, SEPN1, SELN, RSMD1, PLEC1, PLTN, EBS1, LRP5, BMNDl, LRP7, LR3, OPPG, VBCH2, CLCN7, CLC7, OPTA2, OSTMI, GL, TCIRG1, TIRC7, OC116, OPTB1, VAPB, VAPC, ALS8, SMN1, SMA1, SMA2, SMA3, SMA4, BSCL2, SPG17, GARS, SMAD1, CMT2D, HEXB, IGHMBP2, SMUBP2, CATF1, SMARD1), neurological and neuronal diseases and disorders (e.g., heterologous nucleic acid sequence is or encodes SOD1, ALS2, STEX, FUS, TARDBP, VEGF (VEGF-a, VEGF-b, VEGF-c), APP, AAA, CVAP, ADI, APOE, AD2, PSEN2, AD4, STM2, APBB2, FE65LI, NOS3, PLAU, URK, ACE, DCPI, ACEI, MPO, PAC1PI, PAXIPIL, PTIP, A2M, BLMH, BMH, PSEN1, AD3, Mecp2, BZRAP1, MDGA2, Sema5A, Neurexin 1, GLOl, MECP2, RTT, PPMX, MRX16, MRX79, NLGN3, NLGN4, KIAA1260, AUTSX2, FMR2, FXR1, FXR2, mGLUR5, HD, IT15, PRNP, PRIP, JPH3, JP3, HDL2, TBP, SCA17, NR4A2, NURR1, NOT, TINUR, SNCAIP, TBP, SCA17, SNCA, NACP, PARK1, PARK4, DJI, PARK7, LRRK2, PARK8, PINK1, PARK6, UCHL1, PARK5, SNCA, NACP, PARKl, PARK4, PRKN, PARK2, PDJ, DBH, NDUFV2, MECP2, RTT, PPMX, MRX16, MRX79, CDKL5, STK9, MECP2, RTT, PPMX, MRX16,MRX79, x-Synuclein, DJ-1, Neuregulin-l (Nrgl), Erb4 (receptor for Neuregulin), Complexin-l (Cplxl), Tphl Tryptophan hydroxylase, Tph2, Tryptophan hydroxylase 2, Neurexin 1, GSK3, GSK3a, GSK3b, 5-HTT (Slc6a4), CONT, DRD (Drdla), SLC6A , DAOA, DTNBP1, Dao (Daol), APH-l(alpha and beta), Presenilin (Psenl), Nicastrin, (Ncstn), PEN-2, Nosl, Parpl, Natl, Nat2, HTT, SBMA/SMAX1/AR, FXN/X25, ATX3, TXN, ATXN2, DMPK, Atrophin-1, Atnl, CBP, VLDLR, Atxn7, and AtxnlO), and ocular diseases and disorders (e.g., Aber, Ccl2, Cc2, cp (ceruloplasmin), Timp3, cathepsin-D, Vldlr, Ccr2, CRYAA, CRYA1, CRYBB2, CRYB2, PITX3, BFSP2, CP49, CP47, CRYAA, CRYAI, PAX6, AN2, MGDA, CRYBA1, CRYB1, CRYGC, CRYG3, CCL, LIM2, MP19, CRYGD, CRYG4, BFSP2, CP49, CP47, HSF4, CTM, HSF4, CTM, MIP, AQPO, CRYAB, CRYA2, CTPP2, CRYBB1, CRYGD, CRYG4, CRYBB2, CRYB2, CRYGC, CRYG3, CCL, CRYAA, CRYAI, GJA8, CX50, CAE1, GJA3, CX46, CZP3, CAE3, CCM1, CAM, KRIT1, APOA1, TGFBI, CSD2, CDGG1, CSD, BIGH3, CDG2, TACSTD2, TROP2, M1SI, VSX1, RINX, PPCD, PPD, KTCN, COL8A2, FECD, PPCD2, PIP5K3, CFD, KERA, CNA2, MYOC, TIGR, GLCIA, JO AG, GPOA, OPTN, GLC1E, FIP2, HYPL, NRP, CYP1BI, GLC3A, OPA1, NTG, NPG, CYP1BI, GLC3A, CRB1, RP12, CRX, CORD2, CRD, RPGRIPI, LCA6, CORD9, RPE65, RP20, AIPL1, LCA4, GUCY2D, GUC2D, LCA1, CORD6, RDH12, LCA3, ELOVL4, ADMD, STGD2, STGD3, RDS, RP7, PRPH2, PRPH, AVMD, AOFMD, and VMD2). [00370] In some embodiments, the heterologous nucleic acid sequence is or encodes a factor that can affect the differentiation of a cell. As a non-limiting example, the expression of one or more of Oct4, Klf4, Sox2, c-Myc, L-Myc, dominant-negative p53, Nanog, Glisl, Lin28, TFIID, mir-302/367, or other miRNAs can cause the cell to become an induced pluripotent stem (iPS) cell. [00371] In some embodiments, the heterologous nucleic acid sequence is or encodes a factor for transdifferentiating cells. Non-limiting examples of factors include: one or more of GATA4, Tbx5, Mef2C, Myocd, Hand2, SRF, Mespl, SMARCD3 for cardiomyocytes; Ascii, Nurrl, LmxlA, Bm2, Mytll, NeuroDl, FoxA2 for neural cells; and Hnf4a, Foxal, Foxa2 or Foxa3 for hepatic cells. [00372] In certain embodiments, the heterologous nucleic acid encodes a therapeutic antibody, or an antigen-binding fragment thereof. [00373] In certain embodiments, the heterologous nucleic acid encodes a protein for replacement therapy. The protein may be defective in a disease cell or a disease organism / individual, and the wild-type protein or a functional fragment or variant thereof, when delivered by the heterologous nucleic acid to the disease cell / tissue / organism / individual, at least partially or fully restores the lost function of the protein the disease cell / tissue / organism / individual. HNS = donor DNA template [00374] In some embodiments, the heterologous nucleic acid sequence is a donor DNA template that can be integrated into a host genome via HDR. [00375] In some embodiments, the heterologous nucleic acid sequence is a donor DNA that can serve as a template or primer for recombineering during replication. [00376] In certain embodiments, the heterologous nucleic acid comprises or encodes a donor / template sequence, wherein the donor / template corrects / repairs / removes a mutation at the target genome site. For example, the mutation may be a mutated exon in a disease gene. [00377] In certain embodiments, the donor / template may encode or comprises a functional DNA element, such as a promoter, an enhancer, a protein binding sequence, a methylation site, or a homology region for assisting gene editing, etc. [00378] By “donor DNA” or “donor DNA template” it is meant a single-stranded DNA to be inserted at a site cleaved by a gene-editing nuclease (e.g., a CRISPR/Cas effector protein; a TALEN; a ZFN) (e.g., after dsDNA cleavage, after nicking a target DNA, after dual nicking a target DNA, and the like). The donor DNA template can contain sufficient homology to a genomic sequence at the target site, e.g., 70%, 80%, 85%, 90%, 95%, or 100% homology with the nucleotide sequences flanking the target site, e.g. within about 50 bases or less of the target site, e.g. within about 30 bases, within about 15 bases, within about 10 bases, within about 5 bases, or immediately flanking the target site, to support homology-directed repair between it and the genomic sequence to which it bears homology. [00379] Approximately 25, 50, 100, or 200 nucleotides, or more than 200 nucleotides, of sequence homology between a donor DNA template and a genomic sequence (or any integral value between 10 and 200 nucleotides, or more) can support homology-directed repair. Donor DNA template can be of any length, e.g., 50 nucleotides or more, 100 nucleotides or more, 250 nucleotides or more, 500 nucleotides or more, 1000 nucleotides or more, 5000 nucleotides or more, etc. A suitable donor DNA template can be from 50 nucleotides to 100 nucleotides, from 100 nucleotides to 500 nucleotides, from 500 nucleotides to 1000 nucleotides, from 1000 nucleotides to 5000 nucleotides, or from 5000 nucleotides to 10,000 nucleotides, or more than 10,000 nucleotides, in length. [00380] As noted above, the donor DNA template comprises a first homology arm and a second homology arm. The first homology arm is at or near the 5’ end of the donor DNA; and comprises a nucleotide sequence that is at least partially complementary to a first nucleotide sequence in a target nucleic acid. The second homology arm is at or near the 3’ end of the donor DNA; and comprises a nucleotide sequence that is at least partially complementary to a second nucleotide sequence in the target nucleic acid. The first and second homology arms can each independently have a length of from about 10 nucleotides to 400 nucleotides; e.g., from 10 nucleotides (nt) to 15 nt, from 15 nt to 20 nt, from 20 nt to 25 nt, from 25 nt to 30 nt, from 30 nt to 35 nt, from 35 nt to 40 nt, from 40 nt to 45 nt, from 45 nt to 50 nt, from 50 nt to 75 nt, from 75 nt to 100 nt, from 100 nt to 125 nt, from 125 nt to 150 nt, from 150 nt to 175 nt, from 175 nt to 200 nt, from 200 nt to 225 nt, from 225 nt to 250 nt, from 250 nt to 275 nt, from 275 nt to 300 nt, from 325 nt to 350 nt, from 350 nt to 375 nt, or from 375 nt to 400 nt. [00381] In certain embodiments, the donor DNA template is used for editing the target nucleotide sequence. In certain embodiments, the donor DNA template comprises one or more mutations to be introduced into the target polynucleotide. Examples of such mutations include substitutions, deletions, insertions, or a combination thereof. In certain embodiments, the mutation causes a shift in an open reading frame on the target polynucleotide. In certain embodiments, the donor polynucleotide alters a stop codon in the target polynucleotide. In certain embodiments, the donor polynucleotide corrects a premature stop codon. The correction can be achieved by deleting the stop codon, or by introducing one or more sequence changes to alter the stop codon to a codon. In certain embodiments, the donor polynucleotide addresses loss of function mutations, deletions, or translocations that may occur, for example, in certain disease contexts by inserting or restoring a functional copy of a gene, or functional fragment thereof, or a functional regulatory sequence or functional fragment of a regulatory sequence. A functional fragment includes a fragment less than the entire copy of a gene but otherwise provides sufficient nucleotide sequence to restore the functionality of a wild type gene or non-coding regulatory sequence (e.g., sequences encoding long non-coding RNA). [00382] In certain embodiments, the donor DNA template may be used to replace a single allele of a defective gene or defective fragment thereof. In another embodiment, the donor DNA template is used to replace both alleles of a defective gene or defective gene fragment. A “defective gene” or “defective gene fragment” is a gene or portion of a gene that when expressed, fails to generate a functioning protein or non-coding RNA with functionality of the corresponding wild-type gene. [00383] In certain example embodiments, these defective genes may be associated with one or more disease phenotypes. In certain example embodiments, the defective gene or gene fragment is not replaced but the heterologous nucleic acid is used to insert donor polynucleotides that encode gene or gene fragments that compensate for or override defective gene expression such that cell phenotypes associated with defective gene expression are eliminated or changed to a different or desired cellular phenotype. This can be achieved by including the coding sequence of a therapeutic protein, such as a therapeutic antibody or functional fragment thereof, or a wild- type version of a defective protein associated with one or more disease phenotypes. [00384] In certain embodiments, the donor may include, but not be limited to, genes or gene fragments, encoding proteins or RNA transcripts to be expressed, regulatory elements, repair templates, and the like. According to the invention, the donor polynucleotides may comprise left end and right end sequence elements that function with transposition components that mediate insertion. [00385] In certain embodiments, the donor DNA template manipulates a splicing site on the target polynucleotide. In certain embodiments, the donor DNA template disrupts a splicing site. The disruption may be achieved by inserting the polynucleotide to a splicing site and/or introducing one or more mutations to the splicing site. In certain embodiments, the donor polynucleotide may restore a splicing site. For example, the polynucleotide may comprise a splicing site sequence. [00386] In certain embodiments, the donor DNA template to be inserted has a size from 10 bp to 50 kb in length, e.g., from 50 bp to ~40kb, from 100 bp to ~30 kb, from 100 bp to ~10 kb, from 100 bp to 300 bp, from 200 bp to 400 bp, from 300 bp to 500 bp, from 400 bp to 600 bp, from 500 bp to 700 bp, from 600 bp to 800 bp, from 700 bp to 900 bp, from 800 bp to 1000 bp, from 900 bp to 1100 bp, from 1000 bp to 1200 bp, from 1100 bp to 1300 bp, from 1200 bp to 1400 bp, from 1300 bp to 1500 bp, from 1400 bp to 1600 bp, from 1500 bp to 1700 bp, from 1600 bp to 1800 bp, from 1700 bp to 1900 bp, from 1800 bp to 2000 bp nucleotides in length. [00387] In certain embodiments, the homologous arm on one or both ends of the sequence to be inserted is independently about 20 bp, 40 bp, 60 bp, 80 bp, 100 bp, 120 bp, or 150 bp. [00388] The first homology arm and the second homology arm of the donor DNA flank a nucleotide sequence (“a nucleotide sequence of interest” or “an intervening nucleotide sequence”) that is to be introduced into a target nucleic acid. The nucleotide sequence of interest can comprise: i) a nucleotide sequence encoding a polypeptide of interest; ii) a nucleotide sequence encoding an exon of a gene; iii) a promoter sequence; iv) an enhancer sequence; v) a nucleotide sequence encoding a non-coding RNA; or vi) any combination of the foregoing. [00389] The donor DNA can provide for gene correction, gene replacement, gene tagging, transgene insertion, nucleotide deletion, gene disruption, gene mutation, etc. For example, the donor DNA can be used to add, e.g., insert or replace, nucleic acid material to a target DNA (e.g. to “knock in” a nucleic acid that encodes a protein, an siRNA, an miRNA, etc.), to add a tag (e.g., 6xHis, a fluorescent protein (e.g., a green fluorescent protein; a yellow fluorescent protein, etc.), hemagglutinin (HA), FLAG, etc.), to add a regulatory sequence to a gene (e.g. promoter, polyadenylation signal, internal ribosome entry sequence (IRES), 2A peptide, start codon, stop codon, splice signal, localization signal, enhancer, etc.), to modify a nucleic acid sequence (e.g., introduce a mutation), and the like. For example, the donor DNA can be used to modify DNA in a site-specific, i.e. “targeted”, way; for example gene knock-out, gene knock-in, gene editing, gene tagging, etc., as used in, for example, gene therapy, e.g. to treat a disease; or as an antiviral, antipathogenic, or anticancer therapeutic, the production of genetically modified organisms in agriculture, the large scale production of proteins by cells for therapeutic, diagnostic, or research purposes, the induction of pluripotent stem cells, biological research, the targeting of genes of pathogens for deletion or replacement, etc. [00390] In some cases, the donor DNA comprises a nucleotide sequence encoding a polypeptide of interest. Polypeptides of interest include, e.g., a) functional versions of a polypeptide that comprises one or more amino acid substitutions, insertions, and/or deletions and that exhibits reduced function, e.g., where the reduced function is associated with or causes a pathological condition; b) fluorescent polypeptides; c) hormones; d) receptors for ligands; e) ion channels; f) neurotransmitters; g) and the like. [00391] In some cases, the donor DNA comprises a nucleotide sequence that encodes a wild-type protein that is lacking in the recipient cell. In some cases, the donor DNA encodes a wild type factor (e.g. Factor VII, Factor VIII, Factor IX and the like) involved in coagulation. In some cases, the donor DNA comprises a nucleotide sequence that encodes a therapeutic antibody. In some cases, the donor DNA comprises a nucleotide sequence that encodes an engineered protein or receptor. In some cases, the engineered receptor is a T cell receptor (TCR), a natural killer (NK) receptor (NKR), or a B cell receptor (BCR). In some cases, the engineered TCR or NKR targets a cancer marker (e.g., a polypeptide that is expressed (e.g., over-expressed) on the surface of a cancer cell). In some cases, the donor DNA comprises a nucleotide sequence that encodes a chimeric antigen receptor (CAR). In some cases, the CAR targets a cancer marker. Donor DNAs encoding CAR, TCR, and/or NCR proteins may be folded into DNA origami structures (DNA nanostructures) and delivered into T cells or NK cells in vitro or in vivo. [00392] Non-limiting examples of polypeptides that can be encoded by a donor DNA include, e.g., IL1B (interleukin 1, beta), XDH (xanthine dehydrogenase), TP53 (tumor protein p53), PTGIS (prostaglandin 12 (prostacyclin) synthase), MB (myoglobin), IL4 (interleukin 4), ANGPT1 (angiopoietin 1), ABCG8 (ATP-binding cassette, sub-family G (WHITE), member 8), CTSK (cathepsin K), PTGIR (prostaglandin 12 (prostacyclin) receptor (IP)), KCNJ11 (potassium inwardly-rectifying channel, subfamily J, member 11), INS (insulin), CRP (C -reactive protein, pentraxin-related), PDGFRB (platelet- derived growth factor receptor, beta polypeptide), CCNA2 (cyclin A2), PDGFB (platelet-derived growth factor beta polypeptide (simian sarcoma viral (v-sis) oncogene homolog)), KCNJ5 (potassium inwardly- rectifying channel, subfamily J, member 5), KCNN3 (potassium intermediate/small conductance calcium-activated channel, subfamily N, member 3), CAPN10 (calpain 10), PTGES (prostaglandin E synthase), ADRA2B (adrenergic, alpha-2B-, receptor), ABCG5 (ATP-binding cassette, sub-family G (WHITE), member 5), PRDX2 (peroxiredoxin 2), CAPN5 (calpain 5), PARP14 (poly (ADP-ribose) polymerase family, member 14), MEX3C (mex-3 homolog C (C. elegans)), ACE angiotensin I converting enzyme (peptidyl-dipeptidase A) 1), TNF (tumor necrosis factor (TNF superfamily, member 2)), IL6 (interleukin 6 (interferon, beta 2)), STN (statin), SERPINE1 (serpin peptidase inhibitor, clade E (nexin, plasminogen activator inhibitor type 1), member 1), ALB (albumin), ADIPOQ (adiponectin, C1Q and collagen domain containing), APOB (apolipoprotein B (including Ag(x) antigen)), APOE (apolipoprotein E), LEP (leptin), MTHFR (5,10- methylenetetrahydrofolate reductase (NADPH)), APOA1 (apolipoprotein A-I), EDN1 (endothelin 1), NPPB (natriuretic peptide precursor B), NOS3 (nitric oxide synthase 3 (endothelial cell)), PPARG (peroxisome proliferator-activated receptor gamma), PLAT (plasminogen activator, tissue), PTGS2 (prostaglandin-endoperoxide synthase 2 (prostaglandin G/H synthase and cyclooxygenase)), CETP (cholesteryl ester transfer protein, plasma), AGTR1 (angiotensin II receptor, type 1), HMGCR (3-hydroxy-3-methylglutaryl-Coenzyme A reductase), IGF1 (insulin-like growth factor 1 (somatomedin C)), SELE (selectin E), REN (renin), PPARA (peroxisome proliferator-activated receptor alpha), PON1 (paraoxonase 1), KNG1 (kininogen 1), CCL2 (chemokine (C-C motif) ligand 2), LPL (lipoprotein lipase), vWF (von Willebrand factor), F2 (coagulation factor II (thrombin)), ICAM1 (intercellular adhesion molecule 1), TGFB1 (transforming growth factor, beta 1), NPPA (natriuretic peptide precursor A), IL10 (interleukin 10), EPO (erythropoietin), SOD1 (superoxide dismutase 1, soluble), VCAM1 (vascular cell adhesion molecule 1), IFNG (interferon, gamma), LPA (lipoprotein, Lp(a)), MPO (myeloperoxidase), ESR1 (estrogen receptor 1), MAPK1 (mitogen-activated protein kinase 1), HP (haptoglobin), F3 (coagulation factor III (thromboplastin, tissue factor)), CST3 (cystatin C), COG2 (component of oligomeric Golgi complex 2), MMP9 (matrix metallopeptidase 9 (gelatinase B, 92 kDa gelatinase, 92 kDa type IV collagenase)), SERPINC1 (serpin peptidase inhibitor, clade C (antithrombin), member 1), F8 (coagulation factor VIII, procoagulant component), HMOX1 (heme oxygenase (decycling) 1), APOC3 (apolipoprotein C-III), IL8 (interleukin 8), PROK1 (prokineticin 1), CBS (cystathionine-beta-synthase), NOS2 (nitric oxide synthase 2, inducible), TLR4 (toll-like receptor 4), SELP (selectin P (granule membrane protein 140 kDa, antigen CD62)), ABCA1 (ATP-binding cassette, sub-family A (ABC1), member 1), AGT (angiotensinogen (serpin peptidase inhibitor, clade A, member 8)), LDLR (low density lipoprotein receptor), GPT (glutamic -pyruvate transaminase (alanine aminotransferase)), VEGFA (vascular endothelial growth factor A), NR3C2 (nuclear receptor subfamily 3, group C, member 2), IL18 (interleukin 18 (interferon-gamma-inducing factor)), NOS1 (nitric oxide synthase 1 (neuronal)), NR3C1 (nuclear receptor subfamily 3, group C, member 1 (glucocorticoid receptor)), FGB (fibrinogen beta chain), HGF (hepatocyte growth factor (hepapoietin A; scatter factor)), ILIA (interleukin 1, alpha), RETN (resistin), AKT1 (v-akt murine thymoma viral oncogene homolog 1), LIPC (lipase, hepatic), HSPD1 (heat shock 60 kDa protein 1 (chaperonin)), MAPK14 (mitogen-activated protein kinase 14), SPP1 (secreted phosphoprotein 1), ITGB3 (integrin, beta 3 (platelet glycoprotein 111a, antigen CD61)), CAT (catalase), UTS2 (urotensin 2), THBD (thrombomodulin), F10 (coagulation factor X), CP (ceruloplasmin (ferroxidase)), TNFRSF11B (tumor necrosis factor receptor superfamily, member lib), EDNRA (endothelin receptor type A), EGFR (epidermal growth factor receptor (erythroblastic leukemia viral (v-erb-b) oncogene homolog, avian)), MMP2 (matrix metallopeptidase 2 (gelatinase A, 72 kDa gelatinase, 72 kDa type IV collagenase)), PLG (plasminogen), NPY (neuropeptide Y), RHOD (ras homolog gene family, member D), MAPK8 (mitogen-activated protein kinase 8), MYC (v-myc myelocytomatosis viral oncogene homolog (avian)), FN1 (fibronectin 1), CMA1 (chymase 1, mast cell), PLAU (plasminogen activator, urokinase), GNB3 (guanine nucleotide binding protein (G protein), beta polypeptide 3), ADRB2 (adrenergic, beta-2-, receptor, surface), APOA5 (apolipoprotein A-V), SOD2 (superoxide dismutase 2, mitochondrial), F5 (coagulation factor V (proaccelerin, labile factor)), VDR (vitamin D (1,25- dihydroxyvitamin D3) receptor), ALOX5 (arachidonate 5 -lipoxygenase), HLA-DRB1 (major histocompatibility complex, class II, DR beta 1), PARP1 (poly (ADP-ribose) polymerase 1), CD40LG (CD40 ligand), PON2 (paraoxonase 2), AGER (advanced glycosylation end product-specific receptor), IRS1 (insulin receptor substrate 1), PTGS1 (prostaglandin- endoperoxide synthase 1 (prostaglandin G/H synthase and cyclooxygenase)), ECE1 (endothelin converting enzyme 1), F7 (coagulation factor VII (serum prothrombin conversion accelerator)), URN (interleukin 1 receptor antagonist), EPHX2 (epoxide hydrolase 2, cytoplasmic), IGFBP1 (insulin-like growth factor binding protein 1), MAPK10 (mitogen- activated protein kinase 10), FAS (Fas (TNF receptor superfamily, member 6)), ABCB1 (ATP-binding cassette, sub-family B (MDR/TAP), member 1), JUN (jun oncogene), IGFBP3 (insulin-like growth factor binding protein 3), CD14 (CD14 molecule), PDE5A (phosphodiesterase 5A, cGMP-specific), AGTR2 (angiotensin II receptor, type 2), CD40 (CD40 molecule, TNF receptor superfamily member 5), LCAT (lecithin-cholesterol acyltransferase), CCR5 (chemokine (C-C motif) receptor 5), MMP1 (matrix metallopeptidase 1 (interstitial collagenase)), TIMP1 (TIMP metallopeptidase inhibitor 1), ADM (adrenomedullin), DYT10 (dystonia 10), STAT3 (signal transducer and activator of transcription 3 (acute-phase response factor)), MMP3 (matrix metallopeptidase 3 (stromelysin 1, progelatinase)), ELN (elastin), USF1 (upstream transcription factor 1), CFH (complement factor H), HSPA4 (heat shock 70 kDa protein 4), MMP12 (matrix metallopeptidase 12 (macrophage elastase)), MME (membrane metallo- endopeptidase), F2R (coagulation factor II (thrombin) receptor), SELL (selectin L), CTSB (cathepsin B), ANXA5 (annexin A5), ADRB1 (adrenergic, beta-1-, receptor), CYBA (cytochrome b-245, alpha polypeptide), FGA (fibrinogen alpha chain), GGT1 (gamma-glutamyltransferase 1), LIPG (lipase, endothelial), HIF1A (hypoxia inducible factor 1, alpha subunit (basic helix-loop-helix transcription factor)), CXCR4 (chemokine (C-X-C motif) receptor 4), PROC (protein C (inactivator of coagulation factors Va and Villa)), SCARB1 (scavenger receptor class B, member 1), CD79A (CD79a molecule, immunoglobulin-associated alpha), PLTP (phospholipid transfer protein), ADD1 (adducin 1 (alpha)), FGG (fibrinogen gamma chain), SAA1 (serum amyloid Al), KCNH2 (potassium voltage-gated channel, subfamily H (eag-related), member 2), DPP4 (dipeptidyl-peptidase 4), G6PD (glucose-6-phosphate dehydrogenase), NPR1 (natriuretic peptide receptor A/guanylate cyclase A (atrionatriuretic peptide receptor A)), VTN (vitronectin), KIAA0101 (KIAA0101), FOS (FBJ murine osteosarcoma viral oncogene homolog), TLR2 (toll-like receptor 2), PPIG (peptidylprolyl isomer ase G (cyclophilin G)), IL1R1 (interleukin 1 receptor, type I), AR (androgen receptor), CYP1A1 (cytochrome P450, family 1, subfamily A, polypeptide 1), SERPINA1 (serpin peptidase inhibitor, clade A (alpha-1 antiproteinase, antitrypsin), member 1), MTR (5- methyltetrahydrofolate-homocysteine methyltransferase), RBP4 (retinol binding protein 4, plasma), APOA4 (apolipoprotein A-IV), CDKN2A (cyclin-dependent kinase inhibitor 2A (melanoma, pl6, inhibits CDK4)), FGF2 (fibroblast growth factor 2 (basic)), EDNRB (endothelin receptor type B), ITGA2 (integrin, alpha 2 (CD49B, alpha 2 subunit of VLA-2 receptor)), CAB INI (calcineurin binding protein 1), SHBG (sex hormone-binding globulin), HMGB1 (high- mobility group box 1), HSP90B2P (heat shock protein 90 kDa beta (Grp94), member 2 (pseudogene)), CYP3A4 (cytochrome P450, family 3, subfamily A, polypeptide 4), GJA1 (gap junction protein, alpha 1, 43 kDa), CAV1 (caveolin 1, caveolae protein, 22 kDa), ESR2 (estrogen receptor 2 (ER beta)), LTA (lymphotoxin alpha (TNF superfamily, member 1)), GDF15 (growth differentiation factor 15), BDNF (brain-derived neurotrophic factor), CYP2D6 (cytochrome P450, family 2, subfamily D, polypeptide 6), NGF (nerve growth factor (beta polypeptide)), SP1 (Sp 1 transcription factor), TGIF1 (TGFB-induced factor homeobox 1), SRC (v-src sarcoma (Schmidt-Ruppin A-2) viral oncogene homolog (avian)), EGF (epidermal growth factor (beta- urogastrone)), PIK3CG (phosphoinositide-3-kinase, catalytic, gamma polypeptide), HLA-A (major histocompatibility complex, class I, A), KCNQ1 (potassium voltage-gated channel, KQT- like subfamily, member 1), CNR1 (cannabinoid receptor 1 (brain)), FBN1 (fibrillin 1), CHKA (choline kinase alpha), BEST1 (bestrophin 1), APP (amyloid beta (A4) precursor protein), CTNNB1 (catenin (cadherin-associated protein), beta 1, 88 kDa), IL2 (interleukin 2), CD36 (CD36 molecule (thrombospondin receptor)), PRKAB1 (protein kinase, AMP-activated, beta 1 non-catalytic subunit), TPO (thyroid peroxidase), ALDH7A1 (aldehyde dehydrogenase 7 family, member Al), CX3CR1 (chemokine (C-X3-C motif) receptor 1), TH (tyrosine hydroxylase), F9 (coagulation factor IX), GH1 (growth hormone 1), TF (transferrin), HFE (hemochromatosis), IE17A (interleukin 17A), PTEN (phosphatase and tensin homolog), GSTM1 (glutathione S - transferase mu 1), DMD (dystrophin), GATA4 (GATA binding protein 4), F13A1 (coagulation factor XIII, Al polypeptide), TTR (transthyretin), FABP4 (fatty acid binding protein 4, adipocyte), PON3 (paraoxonase 3), APOC1 (apolipoprotein C-I), INSR (insulin receptor), TNFRSF1B (tumor necrosis factor receptor superfamily, member IB), HTR2A (5- hydroxytryptamine (serotonin) receptor 2A), CSF3 (colony stimulating factor 3 (granulocyte)), CYP2C9 (cytochrome P450, family 2, subfamily C, polypeptide 9), TXN (thioredoxin), CYP11B2 (cytochrome P450, family 11, subfamily B, polypeptide 2), PTH (parathyroid hormone), CSF2 (colony stimulating factor 2 (granulocyte-macrophage)), KDR (kinase insert domain receptor (a type III receptor tyrosine kinase)), PLA2G2A (phospholipase A2, group IIA (platelets, synovial fluid)), B2M (beta-2-microglobulin), THBS1 (thrombospondin 1), GCG (glucagon), RHOA (ras homolog gene family, member A), ALDH2 (aldehyde dehydrogenase 2 family (mitochondrial)), TCF7L2 (transcription factor 7-like 2 (T-cell specific, HMG-box)), BDKRB2 (bradykinin receptor B2), NFE2L2 (nuclear factor (erythroid-derived 2)-like 2), NOTCH1 (Notch homolog 1, translocation-associated (Drosophila)), UGT1A1 (UDP glucuronosyltransferase 1 family, polypeptide Al), IFNA1 (interferon, alpha 1), PPARD (peroxisome proliferator-activated receptor delta), SIRT1 (sirtuin (silent mating type information regulation 2 homolog) 1 (S. cerevisiae)), GNRH1 (gonadotropin-releasing hormone 1 (luteinizing- releasing hormone)), PAPPA (pregnancy-associated plasma protein A, pappalysin 1), ARR3 (arrestin 3, retinal (X-arrestin)), NPPC (natriuretic peptide precursor C), AHSP (alpha hemoglobin stabilizing protein), PTK2 (PTK2 protein tyrosine kinase 2), IL13 (interleukin 13), MTOR (mechanistic target of rapamycin (serine/threonine kinase)), ITGB2 (integrin, beta 2 (complement component 3 receptor 3 and 4 subunit)), GSTT1 (glutathione S-transfcrase theta 1), IL6ST (interleukin 6 signal transducer (gpl30, oncostatin M receptor)), CPB2 (carboxypeptidase B2 (plasma)), CYP1A2 (cytochrome P450, family 1, subfamily A, polypeptide 2), HNF4A (hepatocyte nuclear factor 4, alpha), SLC6A4 (solute carrier family 6 (neurotransmitter transporter, serotonin), member 4), PLA2G6 (phospholipase A2, group VI (cytosolic, calcium- independent)), TNFSF11 (tumor necrosis factor (ligand) superfamily, member 11), SLC8A1 (solute carrier family 8 (sodium/calcium exchanger), member 1), F2RL1 (coagulation factor II (thrombin) receptor-like 1), AKR1A1 (aldo-keto reductase family 1, member A1 (aldehyde reductase)), ALDH9A1 (aldehyde dehydrogenase 9 family, member Al), BGLAP (bone gamma- carboxyglutamate (gla) protein), MTTP (microsomal triglyceride transfer protein), MTRR (5- methyltetrahydrofolate- homocysteine methyltransferase reductase), SULT1A3 (sulfotransferase family, cytosolic, 1A, phenol- preferring, member 3), RAGE (renal tumor antigen), C4B (complement component 4B (Chido blood group), P2RY12 (purinergic receptor P2Y, G-protein coupled, 12), RNLS (renalase, FAD-dependent amine oxidase), CREB1 (cAMP responsive element binding protein 1), POMC (proopiomelanocortin), RAC1 (ras-related C3 botulinum toxin substrate 1 (rho family, small GTP binding protein Racl)), LMNA (lamin NC), CD59 (CD59 molecule, complement regulatory protein), SCN5A (sodium channel, voltage-gated, type V, alpha subunit), CYP1B1 (cytochrome P450, family 1, subfamily B, polypeptide 1), MIF (macrophage migration inhibitory factor (glycosylation-inhibiting factor)), MMP13 (matrix metallopeptidase 13 (collagenase 3)), TIMP2 (TIMP metallopeptidase inhibitor 2), CYP19A1 (cytochrome P450, family 19, subfamily A, polypeptide 1), CYP21A2 (cytochrome P450, family 21, subfamily A, polypeptide 2), PTPN22 (protein tyrosine phosphatase, non-receptor type 22 (lymphoid)), MYH14 (myosin, heavy chain 14, non-muscle), MBL2 (mannose-binding lectin (protein C) 2, soluble (opsonic defect)), SELPLG (selectin P ligand), AOC3 (amine oxidase, copper containing 3 (vascular adhesion protein 1)), CTSL1 (cathepsin LI), PCNA (proliferating cell nuclear antigen), IGF2 (insulin like growth factor 2 (somatomedin A)), ITGB1 (integrin, beta 1 (fibronectin receptor, beta polypeptide, antigen CD29 includes MDF2, MSK12)), CAST (calpastatin), CXCL12 (chemokine (C-X-C motif) ligand 12 (stromal cell-derived factor 1)), IGHE (immunoglobulin heavy constant epsilon), KCNE1 (potassium voltage-gated channel, Isk- related family, member 1), TFRC (transferrin receptor (p90, CD71)), COL1A1 (collagen, type I, alpha 1), COL1A2 (collagen, type I, alpha 2), IL2RB (interleukin 2 receptor, beta), PLA2G10 (phospholipase A2, group X), ANGPT2 (angiopoietin 2), PROCR (protein C receptor, endothelial (EPCR)), NOX4 (NADPH oxidase 4), HAMP (hepcidin antimicrobial peptide), PTPN11 (protein tyrosine phosphatase, non-receptor type 11), SLC2A1 (solute carrier family 2 (facilitated glucose transporter), member 1), IL2RA (interleukin 2 receptor, alpha), CCL5 (chemokine (C-C motif) ligand 5), IRF1 (interferon regulatory factor 1), CFLAR (CASP8 and FADD-like apoptosis regulator), CALC A (calcitonin-related polypeptide alpha), EIF4E (eukaryotic translation initiation factor 4E), GSTP1 (glutathione S-transferase pi 1), JAK2 (Janus kinase 2), CYP3A5 (cytochrome P450, family 3, subfamily A, polypeptide 5), HSPG2 (heparan sulfate proteoglycan 2), CCL3 (chemokine (C-C motif) ligand 3), MYD88 (myeloid differentiation primary response gene (88)), VIP (vasoactive intestinal peptide), SOAT1 (sterol O-acyltransferase 1), ADRBK1 (adrenergic, beta, receptor kinase 1), NR4A2 (nuclear receptor subfamily 4, group A, member 2), MMP8 (matrix metallopeptidase 8 (neutrophil collagenase)), NPR2 (natriuretic peptide receptor B/guanylate cyclase B (atrionatriuretic peptide receptor B)), GCH1 (GTP cyclohydrolase 1), EPRS (glutamyl-prolyl-tRNA synthetase), PPARGC1A (peroxisome proliferator-activated receptor gamma, coactivator 1 alpha), F12 (coagulation factor XII (Hageman factor)), PEC AMI (platelet/endothelial cell adhesion molecule), CCL4 (chemokine (C-C motif) ligand 4), SERPINA3 (serpin peptidase inhibitor, clade A (alpha- 1 antiproteinase, antitrypsin), member 3), CASR (calcium-sensing receptor), GJA5 (gap junction protein, alpha 5, 40 kDa), FABP2 (fatty acid binding protein 2, intestinal), TTF2 (transcription termination factor, RNA polymerase II), PROS1 (protein S (alpha)), CTF1 (cardiotrophin 1), SGCB (sarcoglycan, beta (43 kDa dystrophin- associated glycoprotein)), YME1L1 (YMEl-like 1 (S. cerevisiae)), CAMP (cathelicidin antimicrobial peptide), ZC3H12A (zinc finger CCCH-type containing 12A), AKR1B1 (aldo-keto reductase family 1, member B1 (aldose reductase)), DES (desmin), MMP7 (matrix metallopeptidase 7 (matrilysin, uterine)), AHR (aryl hydrocarbon receptor), CSF1 (colony stimulating factor 1 (macrophage)), HDAC9 (histone deacetylase 9), CTGF (connective tissue growth factor), KCNMA1 (potassium large conductance calcium- activated channel, subfamily M, alpha member 1), UGT1A (UDP glucuronosyltransferase 1 family, polypeptide A complex locus), PRKCA (protein kinase C, alpha), COMT (catechol-b- methyltransf erase), S100B (S100 calcium binding protein B), EGR1 (early growth response 1), PRL (prolactin), IL15 (interleukin 15), DRD4 (dopamine receptor D4), CAMK2G (calcium/calmodulin- dependent protein kinase II gamma), SLC22A2 (solute carrier family 22 (organic cation transporter), member 2), CCL11 (chemokine (C-C motif) ligand 11), PGF (placental growth factor), THPO (thrombopoietin), GP6 (glycoprotein VI (platelet)), TACR1 (tachykinin receptor 1), NTS (neurotensin), HNF1A (HNF1 homeobox A), SST (somatostatin), KCND1 (potassium voltage-gated channel, Shal- related subfamily, member 1), LOC646627 (phospholipase inhibitor), TBXAS1 (thromboxane A synthase 1 (platelet)), CYP2J2 (cytochrome P450, family 2, subfamily J, polypeptide 2), TBXA2R (thromboxane A2 receptor), ADH1C (alcohol dehydrogenase 1C (class I), gamma polypeptide), ALOX12 (arachidonate 12- lipoxygenase), AHSG (alpha-2-HS-glycoprotein), BHMT (betaine- homocysteine methyltransferase), GJA4 (gap junction protein, alpha 4, 37 kDa), SLC25A4 (solute carrier family 25 (mitochondrial carrier; adenine nucleotide translocator), member 4), ACLY (ATP citrate lyase), ALOX5AP (arachidonate 5-lipoxygenase-activating protein), NUMA1 (nuclear mitotic apparatus protein 1), CYP27B1 (cytochrome P450, family 27, subfamily B, polypeptide 1), CYSLTR2 (cysteinyl leukotriene receptor 2), SOD3 (superoxide dismutase 3, extracellular), LTC4S (leukotriene C4 synthase), UCN (urocortin), GHRL (ghrelin/obestatin prepropeptide), APOC2 (apolipoprotein C-II), CLEC4A (C-type lectin domain family 4, member A), KBTBD10 (kelch repeat and BTB (POZ) domain containing 10), TNC (tenascin C), TYMS (thymidylate synthetase), SHC1 (SHC (Src homology 2 domain containing) transforming protein 1), LRP1 (low density lipoprotein receptor-related protein 1), SOCS3 (suppressor of cytokine signaling 3), ADH1B (alcohol dehydrogenase IB (class I), beta polypeptide), KLK3 (kallikrein-related peptidase 3), HSD11B1 (hydroxysteroid (11 -beta) dehydrogenase 1), VKORC1 (vitamin K epoxide reductase complex, subunit 1), SERPINB2 (serpin peptidase inhibitor, clade B (ovalbumin), member 2), TNS1 (tensin 1), RNF19A (ring finger protein 19A), EPOR (erythropoietin receptor), ITGAM (integrin, alpha M (complement component 3 receptor 3 subunit)), PITX2 (paired-like homeodomain 2), MAPK7 (mitogen-activated protein kinase 7), FCGR3A (Fc fragment of IgG, low affinity 111a, receptor (CD16a)), LEPR (leptin receptor), ENG (endoglin), GPX1 (glutathione peroxidase 1), GOT2 (glutamic-oxaloacetic transaminase 2, mitochondrial (aspartate aminotransferase 2)), HRH1 (histamine receptor HI), NR112 (nuclear receptor subfamily 1, group I, member 2), CRH (corticotropin releasing hormone), HTR1A (5- hydroxytryptamine (serotonin) receptor 1A), VDAC1 (voltage-dependent anion channel 1), HPSE (heparanase), SFTPD (surfactant protein D), TAP2 (transporter 2, ATP-binding cassette, sub-family B (MDR/TAP)), RNF123 (ring finger protein 123), PTK2B (PTK2B protein tyrosine kinase 2 beta), NTRK2 (neurotrophic tyrosine kinase, receptor, type 2), IL6R (interleukin 6 receptor), ACHE (acetylcholinesterase (Yt blood group)), GLP1R (glucagon-like peptide 1 receptor), GHR (growth hormone receptor), GSR (glutathione reductase), NQOl (NAD(P)H dehydrogenase, quinone 1), NR5A1 (nuclear receptor subfamily 5, group A, member 1), GJB2 (gap junction protein, beta 2, 26 kDa), SLC9A1 (solute carrier family 9 (sodium/hydrogen exchanger), member 1), MAOA (monoamine oxidase A), PCSK9 (proprotein convertase subtilisin/kexin type 9), FCGR2A (Fc fragment of IgG, low affinity Ila, receptor (CD32)), SERPINF1 (serpin peptidase inhibitor, clade F (alpha-2 antiplasmin, pigment epithelium derived factor), member 1), EDN3 (endothelin 3), DHFR (dihydrofolate reductase), GAS6 (growth arrest-specific 6), SMPD1 (sphingomyelin phosphodiesterase 1, acid lysosomal), UCP2 (uncoupling protein 2 (mitochondrial, proton carrier)), TFAP2A (transcription factor AP-2 alpha (activating enhancer binding protein 2 alpha)), C4BPA (complement component 4 binding protein, alpha), SERPINF2 (serpin peptidase inhibitor, clade F (alpha-2 antiplasmin, pigment epithelium derived factor), member 2), TYMP (thymidine phosphorylase), ALPP (alkaline phosphatase, placental (Regan isozyme)), CXCR2 (chemokine (C-X-C motif) receptor 2), SLC39A3 (solute carrier family 39 (zinc transporter), member 3), ABCG2 (ATP- binding cassette, sub-family G (WHITE), member 2), ADA (adenosine deaminase), JAK3 (Janus kinase 3), HSPA1A (heat shock 70 kDa protein 1A), FASN (fatty acid synthase), FGF1 (fibroblast growth factor 1 (acidic)), Fll (coagulation factor XI), ATP7A (ATPase, Cu++ transporting, alpha polypeptide), CR1 (complement component (3b/4b) receptor 1 (Knops blood group)), GFAP (glial fibrillary acidic protein), ROCK1 (Rho-associated, coiled-coil containing protein kinase 1), MECP2 (methyl CpG binding protein 2 (Rett syndrome)), MYLK (myosin light chain kinase), BCF1E (butyrylcholinesterase), LIPE (lipase, hormone-sensitive), PRDX5 (peroxiredoxin 5), ADORA1 (adenosine A1 receptor), WRN (Werner syndrome, RecQ helicase-like), CXCR3 (chemokine (C-X-C motif) receptor 3), CD81 (CD81 molecule), SMAD7 (SMAD family member 7), LAMC2 (laminin, gamma 2), MAP3K5 (mitogen- activated protein kinase kinase kinase 5), CF1GA (chromogranin A (parathyroid secretory protein 1)), IAPP (islet amyloid polypeptide), RFIO (rhodopsin), ENPP1 (ectonucleotide pyrophosphatase/phosphodiesterase 1), PTF1LF1 (parathyroid hormone-like hormone), NRG1 (neuregulin 1), VEGFC (vascular endothelial growth factor C), ENPEP (glutamyl aminopeptidase (aminopeptidase A)), CEBPB (CCAAT/enhancer binding protein (C/EBP), beta), NAGLU (N-acetylglucosaminidase, alpha), F2RL3 (coagulation factor II (thrombin) receptor-like 3), CX3CL1 (chemokine (C-X3-C motif) ligand 1), BDKRB1 (bradykinin receptor Bl), ADAMTS13 (ADAM metallopeptidase with thrombospondin type 1 motif, 13), ELANE (elastase, neutrophil expressed), ENPP2 (ectonucleotide pyrophosphatase/phosphodiesterase 2), CISFl (cytokine inducible SF12- containing protein), GAST (gastrin), MYOC (myocilin, trabecular mesh work inducible glucocorticoid response), ATP1A2 (ATPase, Na+/K+ transporting, alpha 2 polypeptide), NF1 (neurofibromin 1), GJB1 (gap junction protein, beta 1, 32 kDa), MEF2A (myocyte enhancer factor 2A), VCL (vinculin), BMPR2 (bone morphogenetic protein receptor, type II (serine/threonine kinase)), TUBB (tubulin, beta), CDC42 (cell division cycle 42 (GTP binding protein, 25 kDa)), KRT18 (keratin 18), F1SF1 (heat shock transcription factor 1), MYB (v-myb myeloblastosis viral oncogene homolog (avian)), PRKAA2 (protein kinase, AMP-activated, alpha 2 catalytic subunit), ROCK2 (Rho-associated, coiled-coil containing protein kinase 2), TFPI (tissue factor pathway inhibitor (lipoprotein-associated coagulation inhibitor)), PRKG1 (protein kinase, cGMP- dependent, type I), BMP2 (bone morphogenetic protein 2), CTNND1 (catenin (cadherin-associated protein), delta 1), CTF1 (cystathionase (cystathionine gamma- lyase)), CTSS (cathepsin S), VAV2 (vav 2 guanine nucleotide exchange factor), NPY2R (neuropeptide Y receptor Y2), IGFBP2 (insulin-like growth factor binding protein 2, 36 kDa), CD28 (CD28 molecule), GSTA1 (glutathione S-transferase alpha 1), PPIA (peptidylprolyl isomerase A (cyclophilin A)), APOF1 (apolipoprotein FI (beta-2- glycoprotein I)), S100A8 (S100 calcium binding protein A8), IL11 (interleukin 11), ALOX15 (arachidonate 15 - lipoxygenase), FBLN1 (fibulin 1), NR1F13 (nuclear receptor subfamily 1, group FI, member 3), SCD (stearoyl-CoA desaturase (delta-9-desaturase)), GIP (gastric inhibitory polypeptide), CF1GB (chromogranin B (secretogranin 1)), PRKCB (protein kinase C, beta), SRD5A1 (steroid- 5-alpha- reductase, alpha polypeptide 1 (3-oxo-5 alpha-steroid delta 4-dehydrogenase alpha 1)), F1SD11B2 (hydroxy steroid (11-beta) dehydrogenase 2), CALCRL (calcitonin receptor-like), GALNT2 (UDP-N- acetyl-alpha-D-galactosamine:polypeptide N- acetylgalactosaminyltransferase 2 (GalNAc-T2)), ANGPTL4 (angiopoietin-like 4), KCNN4 (potassium intermediate/small conductance calcium-activated channel, subfamily N, member 4), PIK3C2A (phosphoinositide-3-kinase, class 2, alpha polypeptide), HBEGF (heparin-binding EGF-like growth factor), CYP7A1 (cytochrome P450, family 7, subfamily A, polypeptide 1), HLA-DRB5 (major histocompatibility complex, class II, DR beta 5), BNIP3 (BCL2/adeno virus E1B 19 kDa interacting protein 3), GCKR (glucokinase (hexokinase 4) regulator), S100A12 (S100 calcium binding protein A 12), PADI4 (peptidyl arginine deaminase, type IV), HSPA14 (heat shock 70 kDa protein 14), CXCR1 (chemokine (C-X-C motif) receptor 1), H19 (H19, imprinted maternally expressed transcript (non-protein coding)), KRTAP19-3 (keratin associated protein 19-3), insulin, RAC2 (ras-related C3 botulinum toxin substrate 2 (rho family, small GTP binding protein Rac2)), RYR1 (ryanodine receptor 1 (skeletal)), CLOCK (clock homolog (mouse)), NGFR (nerve growth factor receptor (TNFR superfamily, member 16)), DBH (dopamine beta-hydroxylase (dopamine beta-monooxygenase)), CHRNA4 (cholinergic receptor, nicotinic, alpha 4), CACNA1C (calcium channel, voltage-dependent, L type, alpha 1C subunit), PRKAG2 (protein kinase, AMP-activated, gamma 2 non-catalytic subunit), CHAT (choline acetyltransferase), PTGDS (prostaglandin D2 synthase 21 kDa (brain)), NR1H2 (nuclear receptor subfamily 1, group H, member 2), TEK (TEK tyrosine kinase, endothelial), VEGFB (vascular endothelial growth factor B), MEF2C (myocyte enhancer factor 2C), MAPKAPK2 (mitogen-activated protein kinase-activated protein kinase 2), TNFRSF11 A (tumor necrosis factor receptor superfamily, member 11a, NFKB activator), HSPA9 (heat shock 70 kDa protein 9 (mortalin)), CYSLTR1 (cysteinyl leukotriene receptor 1), MAT1A (methionine adenosyltransferase I, alpha), OPRL1 (opiate receptor-like 1), IMPA1 (inositol(myo)-l(or 4) - monophosphatase 1), CLCN2 (chloride channel 2), DLD (dihydrolipoamide dehydrogenase), PSMA6 (proteasome (prosome, macropain) subunit, alpha type, 6), PSMB8 (proteasome (prosome, macropain) subunit, beta type, 8 (large multifunctional peptidase 7)), CHI3L1 (chitinase 3-like 1 (cartilage glycoprotein-39)), ALDH1B1 (aldehyde dehydrogenase 1 family, member Bl), PARP2 (poly (ADP-ribose) polymerase 2), STAR (steroidogenic acute regulatory protein), LBP (lipopolysaccharide binding protein), ABCC6 (ATP- binding cassette, sub-family C(CFTR/MRP), member 6), RGS2 (regulator of G-protein signaling 2, 24 kDa), EFNB2 (ephrin- B2), cystic fibrosis transmembrane conductance regulator (CFTR), GJB6 (gap junction protein, beta 6, 30 kDa), APOA2 (apolipoprotein A-II), AMPD1 (adenosine monophosphate deaminase 1), DYSF (dysferlin, limb girdle muscular dystrophy 2B (autosomal recessive)), FDFT1 (farnesyl-diphosphate farnesyltransferase 1), EDN2 (endothelin 2), CCR6 (chemokine (C-C motif) receptor 6), GJB3 (gap junction protein, beta 3, 31 kDa), IL1RL1 (interleukin 1 receptor- like 1), ENTPD1 (ectonucleoside triphosphate diphosphohydrolase 1), BBS4 (Bardet-Biedl syndrome 4), CELSR2 (cadherin, EGF LAG seven-pass G-type receptor 2 (flamingo homolog, Drosophila)), F11R (Fll receptor), RAPGEF3 (Rap guanine nucleotide exchange factor (GEF) 3), HYAL1 (hyaluronoglucosaminidase 1), ZNF259 (zinc finger protein 259), ATOX1 (ATX1 antioxidant protein 1 homolog (yeast)), ATF6 (activating transcription factor 6), KΉK (ketohexokinase (fructokinase)), SAT1 (spermidine/spermine Nl-acetyltransferase 1), GGFI (gamma-glutamyl hydrolase (conjugase, folylpolygammaglutamyl hydrolase)), TIMP4 (TIMP metallopeptidase inhibitor 4), SLC4A4 (solute carrier family 4, sodium bicarbonate cotransporter, member 4), PDE2A (phosphodiesterase 2 A, cGMP- stimulated), PDE3B (phosphodiesterase 3B, cGMP-inhibited), FADS1 (fatty acid desaturase 1), FADS2 (fatty acid desaturase 2), TMSB4X (thymosin beta 4, X-linked), TXNIP (thioredoxin interacting protein), LIMS1 (LIM and senescent cell antigen-like domains 1), RFIOB (ras homolog gene family, member B), LY96 (lymphocyte antigen 96), FOXOl (forkhead box 01), PNPLA2 (patatin-like phospholipase domain containing 2), TRH (thyrotropin-releasing hormone), GJC1 (gap junction protein, gamma 1, 45 kDa), SLC17A5 (solute carrier family 17 (anion/sugar transporter), member 5), FTO (fat mass and obesity associated), GJD2 (gap junction protein, delta 2, 36 kDa), PSRC1 (proline/serine-rich coiled-coil 1), CASP12 (caspase 12 (gene/pseudogene)), GPBAR1 (G protein-coupled bile acid receptor 1), PXK (PX domain containing serine/threonine kinase), IL33 (interleukin 33), TRIB1 (tribbles homolog 1 (Drosophila)), PBX4 (pre-B-cell leukemia homeobox 4), NUPR1 (nuclear protein, transcriptional regulator, 1), 15-Sep(15 kDa selenoprotein), CILP2 (cartilage intermediate layer protein 2), TERC (telomerase RNA component), GGT2 (gamma-glutamyltransf erase 2), MT-COl (mitochondrially encoded cytochrome c oxidase I), UOX (urate oxidase, pseudogene), a CRISPR/Cas effector polypeptide, an enzymatically active CRISPR/Cas effector polypeptide (e.g., is capable of cleaving a target nucleic acid) and a CRISPR/Cas effector polypeptide that is not enzymatically active (e.g., does not cleave a target nucleic acid, but retains binding to the target nucleic acid). In some cases, the donor DNA encodes a wild-type version of any of the foregoing polypeptides; i.e., the donor DNA can encode a “normal” version that does not include a mutation(s) that results in reduced function, lack of function, or pathogenesis. [00393] In some cases, the donor DNA comprises a nucleotide sequence encoding a fluorescent polypeptide. Suitable fluorescent proteins include, but are not limited to, green fluorescent protein (GFP) or variants thereof, blue fluorescent variant of GFP (BFP), cyan fluorescent variant of GFP (CFP), yellow fluorescent variant of GFP (YFP), enhanced GFP (EGFP), enhanced CFP (ECFP), enhanced YFP (EYFP), GFPS65T, Emerald, Topaz (TYFP), Venus, Citrine, mCitrine, GFPuv, destabilized EGFP (dEGFP), destabilized ECFP (dECFP), destabilised EYFP (dEYFP), mCFPm, Cerulean, T-Sapphire, CyPet, YPet, mKO, HcRed, t- HcRed, DsRed, DsRed2, DsRed-monomer, J-Red, dimer2, t-dimer2(12), mRFPl, pocilloporin, Renilla GFP, Monster GFP, paGFP, Kaede protein and kindling protein, Phycobiliproteins and Phycobiliprotein conjugates including B-Phycoerythrin, R-Phycoerythrin and Allophycocyanin. Other examples of fluorescent proteins include mHoneydew, mBanana, mOrange, dTomato, tdTomato, mTangerine, mStrawberry, mCherry, mGrapel, mRaspberry, mGrape2, m PI urn (Shaner et al. (2005) Nat. Methods 2:905-909), and the like. Any of a variety of fluorescent and colored proteins from Anthozoan species, as described in, e.g., Matz et al. (1999) Nature Biotechnol.17:969-973, can be encoded. [00394] In some cases, the donor DNA encodes an RNA, e.g., an siRNA, a microRNA, a short hairpin RNA (shRNA), an anti-sense RNA, a riboswitch, a ribozyme, an aptamer, a ribosomal RNA, a transfer RNA, and the like. [00395] A donor DNA can include, in addition to a nucleotide sequence encoding one or more gene products (e.g., an RNA and/or a polypeptide), one or more transcriptional control elements, e.g., a promoter, an enhancer, and the like. In some cases, the transcriptional control element is inducible. In some cases, the promoter is reversible. In some cases, the transcriptional control element is constitutive. In some cases, the promoter is functional in a eukaryotic cell. In some cases, the promoter is a cell type- specific promoter. In some cases, the promoter is a tissue-specific promoter. [00396] The nucleotide sequence of the donor DNA is typically not identical to the target nucleic acid (e.g., genomic sequence) that it replaces. Rather, the donor DNA may contain at least one or more single base changes, insertions, deletions, inversions or rearrangements with respect to the target nucleic acid (e.g., genomic sequence), so long as sufficient homology is present to support homology-directed repair (e.g., for gene correction, e.g., to convert a disease- causing base pair or a non-disease-causing base pair). In some cases, the donor DNA comprises a non-homologous sequence flanked by two regions of homology, such that homology-directed repair between the target DNA region and the two flanking sequences results in insertion of the non-homologous sequence at the target region. Donor DNA may also comprise a vector backbone containing sequences that are not homologous to the DNA region of interest (the target nucleic acid) and that are not intended for insertion into the DNA region of interest (the target nucleic acid). Generally, the homologous region(s) of a donor sequence will have at least 50% sequence identity to a target nucleic acid (e.g., a genomic sequence) with which recombination is desired. In certain cases, 60%, 70%, 80%, 90%, 95%, 98%, 99%, or 99.9% sequence identity is present. Any value between 1% and 100% sequence identity can be present, depending upon the length of the donor polynucleotide. [00397] The donor DNA may comprise certain nucleotide sequence differences as compared to the target nucleic acid (e.g., genomic sequence), where such difference include, e.g. restriction sites, nucleotide polymorphisms, selectable markers (e.g., drug resistance genes, fluorescent proteins, enzymes etc.), etc., which may be used to assess for successful insertion of the donor DNA at the cleavage site or in some cases may be used for other purposes (e.g., to signify expression at the targeted genomic locus). In some cases, if located in a coding region, such nucleotide sequence differences will not change the amino acid sequence, or will make silent amino acid changes (i.e., changes which do not affect the structure or function of the protein). Alternatively, these sequences differences may include flanking recombination sequences such as FLPs, loxP sequences, or the like, that can be activated at a later time for removal of the marker sequence. In some cases, the donor DNA will include one or more nucleotide sequences to aid in localization of the donor to the nucleus of the recipient cell or to aid in the integration of the donor DNA into the target nucleic acid. For example, in some case, the donor DNA may comprise one or more nucleotide sequences encoding one or more nuclear localization signals (e.g. PKKKRKV (SEQ ID NO:l), VSRKRPRP (SEQ ID NO:2), QRKRKQ (SEQ ID NO:3), and the like (Frietas et al (2009) Cun- Genomics 10:550-7). In some cases, the donor DNA will include nucleotide sequences to recruit DNA repair enzymes to increase insertion efficiency. Fiuman enzymes involved in homology directed repair include MRN-CtIP, BLM-DNA2, Exol, ERCC1, Rad51, Rad52, Ligase 1, RoIQ, PARP1, Ligase 3, BRCA2, RecQ/BLM-ToroIIIa, RTEL, Roΐd, and Roΐh (Verma and Greenburg (2016) Genes Dev.30 (10): 1138-1154). In some cases, the donor DNA is delivered as reconstituted chromatin (Cruz- Becerra and Kadonaga (2020) eLife 2020;9:e55780 DOI: 10.7554/eLife.55780). [00398] In some cases, the ends of the donor DNA are protected (e.g., from exonucleolytic degradation) by any convenient method and such methods are known to those of skill in the art. For example, one or more dideoxynucleotide residues can be added to the 3' terminus of a linear molecule and/or self complementary oligonucleotides can be ligated to one or both ends. See, for example, Chang et al. (1987) Proc. Natl. Acad Sci USA 84:4959-4963; Nehls et al. (1996) Science 272:886-889. Additional methods for protecting exogenous polynucleotides from degradation include, but are not limited to, addition of terminal amino group(s) and the use of modified internucleotide linkages such as, for example, phosphorothioates, phosphoramidates, and O-methyl ribose or deoxyribose residues. As an alternative to protecting the termini of a linear donor DNA, additional lengths of sequence may be included outside of the regions of homology that can be degraded without impacting recombination. HNS = functional RNA element [00399] In certain embodiments, the donor / template comprises a coding sequence for a functional RNA element (ncRNAs, siRNA, shRNA, sgRNA, etc.). [00400] In some embodiments, the heterologous nucleic acid sequences encode a functional non-translated RNA. In some embodiments, the functional non-translated RNA is an RNA aptamer or a ribozyme. [00401] In some embodiments, the heterologous nucleic acid of the engineered retrons further includes a unique barcode to facilitate multiplexing. Barcodes may include one or more nucleotide sequences that are used to identify a nucleic acid or cell with which the barcode is associated. Such barcodes may be inserted for example, into the tip/loop region of the msd- encoded DNA. Barcodes can be 3-1000 or more nucleotides in length, 10-250 nucleotides in length, or 10-30 nucleotides in length, including any length within these ranges, such as 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 40, 50, 60, 70, 80, 90, 100, 200, 300, 400, 500, 600, 700, 800, 900, or 1000 nucleotides in length. [00402] In some embodiments, barcodes are also used to identify the position (viz., positional barcode) of a cell, colony, or sample from which a retron originated, such as the position of a colony in a cellular array, the position of a well in a multi-well plate, the position of a tube in a rack, or the location of a sample in a laboratory. In particular, a barcode may be used to identify the position of a genetically modified cell containing a retron. The use of barcodes allows retrons from different cells to be pooled in a single reaction mixture for sequencing while still being able to trace a particular retron back to the colony from which it originated. [00403] In addition, adapter sequences can be added to engineered retron to facilitate high-throughput amplification or sequencing. For example, a pair of adapter sequences can be added at the 5ʹ and 3ʹ ends of a retron construct to allow amplification or sequencing of multiple engineered retron simultaneously by the same set of primers. HNS = guide RNA [00404] In some embodiments, the functional non-translated RNA is a CRISPR/Cas guide RNA (gRNA) specific for a target sequence in the mammalian cell. FIG.1G depicts various configurations of a recombinant retron ncRNA which is modified by inserting a guide RNA at the 5’ end or the 3’ of the ncRNA. The guide RNA also may be provided in trans as a separate construct. In addition, guide RNAs may be placed at both ends of a recombinant retron ncRNA. [00405] The skilled person will understand that selection of an appropriate guide RNA is informed by which RNA-guided nuclease is utilized. [00406] A guide RNA provides target specificity to the complex (the RNP complex) by including a targeting segment, which includes a guide sequence (also referred to herein as a targeting sequence), which is a nucleotide sequence that is complementary to a sequence of a target nucleic acid. The term “guide RNA”, as used herein, refers to an RNA that comprises: i) an “activator” nucleotide sequence that binds to a CRISPR/Cas effector polypeptide (e.g., a class 2 CRISPR/Cas effector polypeptide such as a type II, type V, or type VI CRISPR/Cas endonuclease) and activates the CRISPR/Cas effector polypeptide; and ii) a “targeter” nucleotide sequence that comprises a nucleotide sequence that hybridizes with a target nucleic acid. The “activator” nucleotide sequence and the “targeter” nucleotide sequence can be on separate RNA molecules (e.g., a “dual-guide RNA”); or can be on the same RNA molecule (a “single-guide RNA”). A guide nucleic acid in some cases includes only ribonucleotides. In some cases, a guide nucleic acid includes both ribonucleotides and deoxyribonucleotides. [00407] In some cases, a CRISPR/Cas guide RNA comprises one or more modifications, e.g., a base modification, a backbone modification, a sugar modification, etc., to provide the nucleic acid with a new or enhanced feature (e.g., improved stability, such as improved in vivo stability). Suitable nucleic acid modifications include, but are not limited to: 2’O-methyI modified nucleotides, 2’ fluoro modified nucleotides, locked nucleic acid (LNA) modified nucleotides, peptide nucleic acid (PNA) modified nucleotides, nucleotides with phosphorothioate linkages, and a 5’ cap (e.g., a 7-methyIguanyIate cap (m7G)). Suitable modified nucleic acid backbones containing a phosphorus atom therein include, for example, phosphorothioates, chiral phosphorothioates, phosphorodithioates, phosphotriesters, aminoalkylphosphotriesters, methyl and other alkyl phosphonates including 3'-aIkyIene phosphonates, 5'- alkylene phosphonates and chiral phosphonates, phosphinates, phosphoramidates including 3'-amino phosphoramidate and aminoalkylphosphoramidates, phosphorodiamidates, thionophosphoramidates, thionoalkylphosphonates, thionoalkylphosphotriesters, selenophosphates and boranophosphates having normal 3'-5' linkages, 2'-5' linked analogs of these, and those having inverted polarity wherein one or more internucleotide linkages is a 3' to 3', 5' to 5' or 2' to 2' linkage. Suitable oligonucleotides having inverted polarity comprise a single 3' to 3' linkage at the 3'-most internucleotide linkage i.e. a single inverted nucleoside residue which may be a basic (the nucleobase is missing or has a hydroxyl group in place thereof). Various salts (such as, for example, potassium or sodium), mixed salts and free acid forms are also included. A CRISPR- Cas guide RNA can also include one or more substituted sugar moieties. Suitable polynucleotides comprise a sugar substituent group selected from: OH; F; 0-, S-, or N-alkyl; 0-, S-, or N-alkenyl; 0-, S- or N-alkynyl; or O-alkyl-O-alkyl, wherein the alkyl, alkenyl and alkynyl may be substituted or unsubstituted Ci to Cio alkyl or C2 to C10 alkenyl and alkynyl. Particularly suitable are 0((CH2)n0) mCH3, 0(CH2)n0CH3, 0(CH2)nNH2, 0(CH2)nCH3, 0(CH2)n0NH2, and 0(CH2)n0N((CH2)nCH3)2, where n and m are from 1 to about 10. Other suitable polynucleotides comprise a sugar substituent group selected from: Ci to Cio lower alkyl, substituted lower alkyl, alkenyl, alkynyl, alkaryl, aralkyl, O-alkaryl or O-aralkyl, SH, SCH3, OCN, Cl, Br, CN, CF3, OCF3, SOCH3, S02CH3, 0N02, N02, N3, NH2, heterocycloalkyl, heterocycloalkaryl, aminoalkylamino, polyalkylamino, substituted silyl, an RNA cleaving group, a reporter group, an intercalator, a group for improving the pharmacokinetic properties of an oligonucleotide, or a group for improving the pharmacodynamic properties of an oligonucleotide, and other substituents having similar properties. A suitable modification includes 2'-methoxy ethoxy (2'-0-CH2 CH2OCH3, also known as 2'-0-(2-methoxyethyl) or 2'- MOE) (Martin et al., Helv. Chim. Acta, 1995, 78, 486-504) i.e., an alkoxyalkoxy group. A further suitable modification includes 2'-dimethylaminooxyethoxy, i.e., a 0(CH2)20N(CH3)2 group, also known as 2'- DMAOE, as described in examples herein below, and 2'-dimethylaminoethoxyethoxy (also known in the art as 2'-0-dimethyl-amino-ethoxy-ethyl or 2'- DMAEOE), i.e., 2'-0-CH2-0-CH2-N(CH3)2. [00408] Examples of various CRISPR/Cas effector proteins and CRISPR/Cas guide RNAs (as well as information regarding requirements related to protospacer adjacent motif (PAM) sequences present in targeted nucleic acids) can be found in the art, for example, see Jinek et al., Science.2012 Aug 17;337(6096):816-21 ; Chylinski et al., RNA Biol.2013 May;10(5):726-37; Ma et al., Biomed Res Int.2013;2013:270805; Hou et al., Proc Natl Acad Sci U S A.2013 Sep 24;110(39):15644-9; Jinek et al., Elife.2013;2:e00471; Pattanayak et al., Nat Biotechnol.2013 Sep;31(9):839-43; Qi et al., Cell.2013 Feb 28 ; 152(5): 1173-83; Wang et al., Cell.2013 May 9;153(4):910-8; Auer et al., Genome Res.2013 Oct 31; Chen et al., Nucleic Acids Res.2013 Nov l;41(20):el9; Cheng et al., Cell Res.2013 Oct;23(10):1163- 71; Cho et al., Genetics.2013 Nov;195(3):1177-80; DiCarlo et al., Nucleic Acids Res.2013 Apr;41(7):4336-43; Dickinson et al., Nat Methods.2013 Oct;10(10):1028-34; Ebina et al., Sci Rep.2013;3:2510; Fujii et al., Nucleic Acids Res.2013 Nov l;41(20):el87; Hu et al., Cell Res.2013 Nov;23(ll):1322-5; Jiang et al., Nucleic Acids Res.2013 Nov l;41(20):el88; Larson et al., Nat Protoc.2013 Nov;8(ll):2180- 96; Mali et al., Nat Methods.2013 Oct;10(10):957-63; Nakayama et al.,Genesis.2013 Dec;51(12):835-43; Ran et al., Nat Protoc.2013 Nov;8(ll):2281-308; Ran et al., Cell.2013 Sep 12;154(6):1380-9; Upadhyay et al., G3 (Bethesda).2013 Dec 9;3(12):2233-8; Walsh et al.,Proc Natl Acad Sci U S A.2013 Sep 24;110(39):15514-5; Xie et al., Mol Plant.2013 Oct 9; Yang et al., Cell.2013 Sep 12;154(6):1370-9; Briner et al., Mol Cell.2014 Oct 23;56(2):333-9; and U.S. patents and patent applications: 8,906,616; 8,895,308; 8,889,418; 8,889,356; 8,871,445; 8,865,406; 8,795,965; 8,771,945; 8,697,359; 20140068797; 20140170753; 20140179006; 20140179770; 20140186843; 20140186919; 20140186958; 20140189896; 20140227787; 20140234972; 20140242664; 20140242699; 20140242700; 20140242702; 20140248702; 20140256046; 20140273037; 20140273226; 20140273230; 20140273231; 20140273232; 20140273233; 20140273234; 20140273235; 20140287938; 20140295556; 20140295557; 20140298547; 20140304853; 20140309487; 20140310828; 20140310830; 20140315985; 20140335063; 20140335620; 20140342456; 20140342457; 20140342458; 20140349400; 20140349405; 20140356867; 20140356956; 20140356958; 20140356959; 20140357523; 20140357530; 20140364333; and 20140377868; all of which are hereby incorporated by reference in their entirety. [00409] Examples and guidance related to type V or type VI CRISPR/Cas endonucleases and guide RNAs (as well as information regarding requirements related to protospacer adjacent motif (PAM) sequences present in targeted nucleic acids) can be found in the art, for example, see Zetsche et al., Cell.2015 Oct 22;163(3):759-71; Makarova et al., Nat Rev Microbiol.2015 Nov;13(ll):722-36; and Shmakov et al., Mol Cell.2015 Nov 5;60(3):385-97. C. Reverse Transcriptases (RTs) [00410] Reverse transcriptases (RTs, also known as RNA-directed DNA polymerases) are enzymes present in all three domains of life, which are DNA polymerase using RNA as a template. Reverse transcriptases of the present disclosure are used to reverse transcribe template msd RNA into single-stranded msDNA. [00411] The reverse transcriptase or a functional domain thereof that may be used in the instant invention includes prokaryotic and eukaryotic RT, provided that the RT functions within the host to generate a donor polynucleotide sequence from the RNA template (e.g., an RNA template from the retron transcript ncRNA). [00412] In certain embodiments, suitable RT sequences (including amino acid sequences and the encoding polynucleotide sequences) are provided in Table A. [00413] In certain embodiments, the nucleotide sequence of a native or wild-type RT is modified, for example, using known codon optimization techniques, so that expression within the desired host is optimized. [00414] In certain embodiments, the RT domain of a reverse transcriptase is used in the present invention, so long as it is compatible with the engineered retron of the invention. The domain may include only the RNA-dependent DNA polymerase activity. In certain embodiments, the RT domain is non-mutagenic, i.e., does not cause mutation in the donor polynucleotide (e.g., during the reverse transcriptase process). In certain embodiments, the RT domain may be non-retron RT in origin, e.g., a viral RT or a human endogenous RTs. In certain embodiments, the RT domain is retron RT or DGRs RT. In certain embodiments, the RT may be less mutagenic than a counterpart wildtype RT. In certain embodiments, the RT is not mutagenic. [00415] In some embodiments, a reverse transcriptase is encoded by a retron ret gene, which may accompany the cognate msr and msd loci and specifically recognize the secondary structure of the cognate ncRNA transcript. [00416] In some embodiments, the RT may be obtained from prokaryotic or eukaryotic cells. Most reverse transcriptases (80%) can be phylogenetically clustered into three major lineages: group II introns, diversity-generating retroelements (DGRs), and retrons. Other clades of RTs include abortive infection (Abi) RTs, CRISPR-Cas-associated RTs, Group II-like (G2L), the unknown groups (UG), and rvt elements. [00417] In some embodiments, the RT gene is a cognate RT, a retron RT from a species within the same species or clade of the cognate RT, or a retron RT not within the same clade of the cognate RT such as an unrelated RT or an engineered RT. In some embodiments, the non- retron related RT are RTs from group II introns, diversity-generating retroelements (DGRs), abortive infection (Abi) RTs, CRISPR-Cas-associated RTs, Group II-like (G2L), the unknown groups (UG), and rvt elements. See Mestre et al., Nucleic Acids Research, Volume 48, Issue 22, 16 December 2020, Pages 12632-12647; and Mestere et al., UG/Abi: “A Highly Diverse Family of Prokaryotic Reverse Transcriptases Associated With Defense Functions,” doi.org/10.1101/2021.12.02.470933 (incorporated herein by reference). [00418] In some embodiments, the RT are from clades related to retron/retron-like sequences. In some embodiments, the RT are selected from RTs provided in Table A. In some embodiments, the RT is not an RT associated with the sequences identified in Table X. [00419] In prokaryotic retron systems, the RT gene is typically located downstream from the ncRNA (msr and msd) locus. In the engineered retron, the RT position can differ from the natural or wild type retrons. In some embodiments, the RT gene can be provided in cis, such as either upstream or downstream of the msr locus or the msd locus. In certain embodiments, the RT gene is provided in trans, such as provided separately in a vector of the vector system described herein, wherein the ncRNA coding msr and msd sequences are provided in a different vector of the vector system described herein. [00420] In some embodiments, the RT is modified (e.g., insertion, deletion, and/or substitution of one or more nucleotide(s)) or codon optimized to enhance activity or processivity. [00421] In certain embodiments, a cryptic stop signal is removed from the RT thereby allowing generation of longer ssDNAs. [00422] In certain embodiments, the RT is from a retron which encodes msDNA as described in US 6,017,737; US5,849,563; US5,780,269; US5,436,141; US5,405,775; US5,320,958; CA2,075,515; all of which are incorporated by reference herein in their entireties. [00423] In some embodiments, the engineered retron further comprises a polynucleotide (e.g., a DNA molecule) encoding a reverse transcriptase (RT) or a portion thereof. In some embodiments, the encoded RT or portion thereof is capable of synthesizing a DNA copy of at least a portion of the msd locus encoding the msDNA. [00424] In some embodiments, the polynucleotide (e.g., a DNA molecule) encoding the RT comprises a polynucleotide listed in Table A, or a polynucleotide having at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%%, at least 99.1%, at least 99.2%, at least 99.3%, at least 99.4%, at least 99.5%, at least 99.6%, at least 99.7%, at least 99.8% or at least 99.9% sequence identity to a polynucleotide listed in Table A. [00425] In some embodiments, the polynucleotide encoding the RT encodes a polypeptide listed in Table A, or a polypeptide having at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%%, at least 99.1%, at least 99.2%, at least 99.3%, at least 99.4%, at least 99.5%, at least 99.6%, at least 99.7%, at least 99.8% or at least 99.9% sequence identity to a polypeptide listed in Table A; and/or a polypeptide of Table C. [00426] In some embodiments, the polynucleotide encoding the RT does not comprise a polynucleotide listed in Table X. [00427] Once translated, the RT binds the ncRNA template downstream from the msd locus, forming an RT-RNA complex, and initiating reverse transcription of the RNA towards its 5ʹ end. Accordingly, in certain aspects the disclosure relates to an engineered nucleic acid- enzyme construct comprising: a. a non-coding RNA (ncRNA) comprising: i) an msr locus encoding the msr RNA portion of a multi-copy single-stranded DNA (msDNA); and ii) an msd locus encoding the msd RNA portion of the msDNA; b. a heterologous nucleic acid inserted at or within a location selected from: the msd locus, upstream of the msr locus, upstream of the msd locus, and downstream of the msd locus; and c. a reverse transcriptase (RT), or a domain thereof comprising: i) a polypeptide listed in Table A, or a polypeptide having at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%%, at least 99.1%, at least 99.2%, at least 99.3%, at least 99.4%, at least 99.5%, at least 99.6%, at least 99.7%, at least 99.8% or at least 99.9% sequence identity to a polypeptide listed in Table A; and/or ii) a polypeptide listed in Table C. In some embodiments, the RT does not comprise a polypeptide listed in Table X. [00428] In certain aspects, the disclosure relates to an engineered nucleic acid-enzyme construct comprising: a) a non-coding RNA (ncRNA) comprising: i) an msr locus encoding the msr RNA portion of a multi-copy single-stranded DNA (msDNA); and ii) an msd locus encoding the msd RNA portion of the msDNA, b) a heterologous nucleic acid inserted at or within a location selected from: the msd locus; upstream of the msr locus; upstream of the msd locus; and downstream of the msd locus; and c) a reverse transcriptase (RT), or a portion thereof, wherein the RT is capable of synthesizing a DNA copy of at least a portion of the msd locus encoding the msDNA, and wherein the ncRNA and/or the RT is any one of the invention described herein. [00429] In certain aspects, the disclosure relates to an engineered nucleic acid-enzyme construct comprising: a) a non-coding RNA (ncRNA) comprising: i) an msr locus encoding the msr RNA portion of a multi-copy single-stranded DNA (msDNA); and ii) an msd locus encoding the msd RNA portion of the msDNA; b) a heterologous nucleic acid inserted at or within a location selected from: the msd locus, upstream of the msr locus, upstream of the msd locus, and downstream of the msd locus; and c) a reverse transcriptase (RT) or a domain thereof: wherein the RT comprises: i) an RT listed in Table A, or an RT having at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%%, at least 99.1%, at least 99.2%, at least 99.3%, at least 99.4%, at least 99.5%, at least 99.6%, at least 99.7%, at least 99.8% or at least 99.9% sequence identity to an RT listed in Table A; and/or ii) a consensus sequence listed in Table C; and wherein, the RT does not comprise a sequence from Table X. [00430] In some embodiments of the nucleic-acid enzyme constructs described herein, the ncRNA comprises: i) an ncRNA listed in Table B, or an ncRNA having at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%%, at least 99.1%, at least 99.2%, at least 99.3%, at least 99.4%, at least 99.5%, at least 99.6%, at least 99.7%, at least 99.8% or at least 99.9% sequence identity to an ncRNA listed in Table B; and/or optionally wherein the ncRNA is not an ncRNA from the retons of Table X. [00431] In some embodiments, the RT is linked to components such as RNA-guided and non-RNA guided nucleases. The linked maybe via s peptide bond or a short linker peptide in a fusion protein. Suitable linker peptides include flexible linkers such as those comprising G or S repeats, such as G4S repeat units or GS repeat units, with 1-20 repeats, e.g., 1, 2, 3, 4, 5, 6, 7, or 8 repeats. [00432] In certain embodiments, the RT is chemically linked or conjugated to the RNA- guided and non-RNA guided nucleases via non-peptide bonds. Such protein conjugates may be delivered directly to a host cell, either together with the nucleic acid component of the engineered retron described herein, or separately. [00433] In some embodiments, the RT is linked to a DNA-repair modulating biomolecule (e.g., NHEJ peptide inhibitors. D. Programmable Nucleases (RNA-guided Nucleases) [00434] In certain embodiments, the engineered retron (e.g., an engineered nucleic acid construct or engineered nucleic acid-enzyme construct as described herein) may comprise or encode, as one heterologous nucleic acid, a guide RNA (gRNA) suitable for guiding a nuclease to target a particular genomic sequence to be modified. The gRNA includes sequences complementary to a genomic sequence, and therefore can mediate binding of the nuclease-gRNA complex to the genomic target site by hybridization between the guide sequence and the target site sequence. [00435] In certain embodiments, the gRNA can be linked to the ncRNA and/or the msDNA encoded by the engineered retron described herein at the 5ʹ end of the ncRNA and/or the msDNA. In certain embodiments, the gRNA can be linked to the ncRNA and/or the msDNA encoded by the engineered retron described herein at the 3ʹ end of the RNA of the ncRNA and/or the msDNA after reverse transcription. [00436] In some embodiments, the nuclease that can form a complex with the gRNA can be any one of the art-recognized clustered regularly interspersed short palindromic repeats (CRISPR) system Cas effector enzymes, which are useful for, e.g., genome editing, including genome editing in mammalian cells or human cells. [00437] For example, a gRNA that can be loaded into a Cas9 or variant thereof may be encoded by the engineered retron, such that the gRNA is transcribed as part of the msDNA. In some embodiments, the gRNA may be linked to the 5ʹ end of the a1 region of the msr-encoded sequence in the retron ncRNA, as well as the msDNA after reverse transcription. In some embodiments, the gRNA may be part of the modified msr-region present in the ncRNA and the msDNA produced after reverse transcription (viz., the encoded gRNA is not degraded by the synthesis of the msDNA through reverse transcription of the ncRNA). [00438] Any art recognized CRISPR/Cas effector enzymes or variants thereof (“Cas enzymes”) known to be useful for CRISPR-based genome editing can be used with the engineered retron, though such Cas enzymes may not necessarily be part of the engineered retron, and can (but is not required to) be provided separately. For example, the Cas enzymes can be provided as part of the vector system described herein. The coding sequence of the suitable Cas enzyme can be present on the same vector that provides the engineered retron, or on different vectors. When the engineered retron and the Cas enzymes are present on the same vector, they may be under the transcriptional control of the same promoter, enhancer, or other transcriptional regulatory elements, or may be separately regulated by different promoters, enhancers, and/or other transcriptional regulatory sequences in the vector system. [00439] In some embodiments, the Cas enzyme is a Class 2, Type II CRISPR/Cas effector enzyme, such as Cas9. In some embodiments, the Cas9 is from Streptococcus pyogenes (SpCas9), and the gRNA encoded by the engineered retron comprises both the crisprRNA (crRNA) and the tracrRNA linked together as the single guide RNA (gRNA). [00440] In some embodiments, the Cas9 is from Staphylococcus aureus Cas9 (SaCas9), or an engineered variant thereof such as SaCas9-HF (a high-fidelity variant with genome-wide activity), KKHSaCas9 (which recognizes a 5ʹ-NNGRRT-3ʹ PAM, and has a 2-4x broader range of target sites in the human genome than the wildtype SaCas9), and microABE1744 (an engineered SaCas9 variant adapted for use in adenine base editing (ABE), with significantly improved on-target editing compared to other nucleases, with a reduced RNA off-target footprint). [00441] In some embodiments, the Cas9 is from Streptococcus thermophilus (StCas9), Neisseria meningitidis (NmCas9), Francisella novicida (FnCas9), or Campylobacter jejuni (CjCas9). [00442] In some embodiments, the Cas9 is from Streptococcus canis (ScCas9), with a less stringent PAM sequence requirement of 5′-NNG-3′ (instead of the more stringent 5′-NGG-3′ for SpCas9). [00443] In some embodiments, the Cas9 is from Staphylococcus auricularis (SauriCas9) (which recognizes a 5ʹ-NNGG-3ʹ PAM sequence, has high editing activity). [00444] In some embodiments, the Cas enzyme is a Cas9 variant with mutated catalytic domain that retains cleavage specificity, but only nicks a single DNA strand at a desired target sequence instead of creating a double-strand break (DSB). Two such Cas9 nickase variants targeting different strands of the same target sequence may be used together to increase fidelity of creating the DSB, each using a different gRNA that can be provided by the engineered retron. [00445] In some embodiments, the Cas enzyme is a high fidelity Cas9 variant with weakened DNA phosphate backbone interaction (such as SpCas9-HF1) that displays genome- wide specificity and undetectable off-target effects. [00446] In some embodiments, the Cas enzyme is a Cas9 variant known as eSpCas9, which has weakened interactions between the eSpCas9 and its gRNAs with non-exact complementarity with the target DNA sequence, thus providing improved specificity and lower off-target editing rates. [00447] In some embodiments, the Cas enzyme is a hyper-accurate Cas9 variant (HypaCas9), which improves proofreading before cleavage, and thus drastically reduces off- target cleavage. [00448] In some embodiments, the Cas enzyme is a Cas9 variant (FokI-Fused dCas9), which combines the DNA recognition ability by dCas9 with the specificity of an active nuclease FokI. The resulting nuclease cuts the target sequence only after dimerization, which is more difficult to occur at off-target sites, thus resulting in enhanced specificity. [00449] In some embodiments, the Cas enzyme is a Cas9 variant xCas9, which recognizes a broad range of PAM sequences, thus increasing the target sites to 1 in 4 in the genome. In addition, the xCas9 variant also exhibits lower off-target rates than the commonly used SpCas9. [00450] In some embodiments, the Cas enzyme is a Cas9 variant with altered PAM sequence specificities, including the SpG variant with an expanded target range of PAM sequences, and the SpRY variant that can target almost all PAM sequences. [00451] In some embodiments, the Cas enzyme is dCas9 having inactivated catalytic nuclease domains while maintaining the recognition domains that allow guide RNA-mediated targeting to specific DNA sequences. The dCas9 may be further linked to a functional domain with a distinct biological function, such as transcriptional activation / depression, DNA methylation, demethylation, endonuclease (such as FokI), or fluorescent dye. Representative (non-limiting) dCas9-linked functional domains include including dCas9-SAM, dCas9-SunTag, dCas9-VPR, and dCas9-KREB. [00452] In some embodiments, the dCas9 is fused with a catalytic enzyme with cytidine deaminase activity, which converts GC basepair to AT basepair. [00453] In some embodiments, the dCas9 is fused with an engineered RNA adenosine deaminase, which converts AT basepair to GC basepair. [00454] In some embodiments, the Cas enzyme is a Class 2, Type V CRISPR/Cas effector enzyme, such as Cas12a (Cpf1) (Type V-A), Cas12b (C2c1) (Type V-B), Cas12c (C2c3) (Type V-C), Cas12d (CasY) (Type V-D), Cas12e (CasX) (Type V-E), Cas12f (Cas14, C2c10) (Type V- F), Cas12g (Type V-G), Cas12h (Type V-H), Cas12i (Type V-I), Cas12k (C2c5) (Type V-K), or C2c4 / C2c8 / C2c9 (Type V-U). [00455] In some embodiments, the Cas enzyme is Cas12a or Cpf1 (CRISPR from Prevotella and Francisella 1). Unlike Cas9, Cas12a is well-suited for targeting AT-rich DNA sequences because of its AT-rich PAM sequences. In some embodiments, the Cas12a is FnCas12a (that recognizes PAM sequence 5′‐TTN‐3′), or AsCas12a or LbCas12a (that recognize 5′‐TTTV‐3′ PAM sequence), where V is A, G, or C nucleotide. Further, Cas12a creates staggered double-stranded breaks in the target DNA, rather than the blunt-ends generated by SpCas9, thus rendering it more useful for HDR repair. In this embodiment, the subject engineered retron encodes a gRNA suitable for use for Cas12a, in that the gRNA does not require a tracer RNA, and only requires the crRNA. [00456] In some embodiments, the Cas enzyme is a Cas12a variant from Acidaminococcus sp. (enAsCas12a), which has an expanded target range of PAM sequences and significantly higher editing activity compared to wild type Cas12a. [00457] In some embodiments, the Cas enzyme is a high-fidelity Cas12a variant (enAsCas12a-HF1) that reduces off-target editing. [00458] In some embodiments, the Cas enzyme is Cas12b or C2c1. In some embodiments, the Cas12b is form Alicyclobacillus acidoterrestris (AacCas12b), or from Alicyclobacillus acidiphilus Cas12b (AapCas12b). [00459] In some embodiments, the Cas enzyme is a Cas12b variant that functions at 37°C, such as one form Bacillus hisashii (BhCas12b). [00460] In some embodiments, the Cas enzyme is a BhCas12b variant with higher specificity than SpCas9. [00461] In some embodiments, the Cas enzyme is CasX or Cas12d. [00462] In some embodiments, the Cas enzyme is CasY or Cas12e, and the subject engineered retron encodes a short-complementarity untranslated RNA (scoutRNA) together with crRNA (rather than tracrRNA used in other CRISPR-Cas systems). [00463] In some embodiments, the Cas enzyme is Cas14, from archaea bacteria. Cas14 targets single-stranded (ss) DNA target sequences, does not require PAM sequence for activation, and has collateral activity (i.e., cuts other non-target ssDNA strands non-specifically upon binding the target sequence). Unlike Cas12a, Cas14a requires high fidelity complementarity to the target ssDNA, and is very sensitive to internal seed region mismatches of the ssDNA target substrate. [00464] In some embodiments, the gRNA nuclease is an engineered RNA-guided Fokl nuclease. RNA-guided Fokl nucleases comprise fusions of inactive Cas9 (dCas9) and the Fokl endonuclease (FokI-dCas9), wherein the dCas9 portion confers guide RNA- dependent targeting on Fokl. For a description of engineered RNA-guided Fold nucleases, see, e.g., Havlicek et al. (2017) Mol. Ther.25(2):342-355, Pan et al. (2016) Sci Rep.6:35794, Tsai et al. (2014) Nat Biotechnol.32(6):569-576; herein incorporated by reference. [00465] In other embodiments, the RNA-guided nuclease can be a non-CRISPER/Cas related nuclease such as transposon-encoded nucleases, IscBs, IscR, or TnpBs. [00466] In some embodiments, the Cas enzyme is a Cas9 [00467] In some embodiments, the retron-based editing systems described herein can include any Cas9 equivalent. A Cas9 equivalent is a Cas9-like protein that provides the same or substantially the same function as Cas9. For instance, if Cas9 refers to a type II enzyme of the CRISPR-Cas system, a Cas9 equivalent can refer to a type V or type VI enzyme of the CRISPR- Cas system. [00468] For example, Cas12e (CasX) is a Cas9 equivalent that reportedly has the same function as Cas9 but which evolved through convergent evolution. Thus, the Cas12e (CasX) protein is contemplated to be used with the retron-based editor systems described herein. In addition, any variant or modification of Cas12e (CasX) is conceivable and within the scope of the present disclosure. [00469] In some embodiments, Cas9 equivalents may refer to Cas12e (CasX) or Cas12d (CasY), which have been described in, for example, Burstein et al.,“New CRISPR–Cas systems from uncultivated microbes.” Cell Res.2017 Feb 21. doi: 10.1038/cr.2017.21, the entire contents of which is hereby incorporated by reference. Using genome-resolved metagenomics, a number of CRISPR–Cas systems were identified, including the first reported Cas9 in the archaeal domain of life. This divergent Cas9 protein was found in little- studied nanoarchaea as part of an active CRISPR–Cas system. In bacteria, two previously unknown systems were discovered, CRISPR– Cas12e and CRISPR– Cas12d, which are among the most compact systems yet discovered. In some embodiments, Cas9 refers to Cas12e, or a variant of Cas12e. In some embodiments, Cas9 refers to a Cas12d, or a variant of Cas12d. It should be appreciated that other RNA-guided DNA binding proteins may be used as a nucleic acid programmable DNA binding protein (napDNAbp), and are within the scope of this disclosure. [00470] In some embodiments, the Cas9 equivalent comprises an amino acid sequence that is at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or at least 99.5% identical to a naturally-occurring Cas12e (CasX) or Cas12d (CasY) protein. In some embodiments, the napDNAbp is a naturally-occurring Cas12e (CasX) or Cas12d (CasY) protein. In some embodiments, the napDNAbp comprises an amino acid sequence that is at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or at least 99.5% identical to a wild-type Cas moiety or any Cas moiety provided herein. [00471] In various embodiments, the nucleic acid programmable DNA binding proteins include, without limitation, Cas9 (e.g., dCas9 and nCas9), Cas12e (CasX), Cas12d (CasY), Cas12a (Cpf1), Cas12b1 (C2c1), Cas13a (C2c2), Cas12c (C2c3), Argonaute, and Cas12b1. One example of a nucleic acid programmable DNA-binding protein that has different PAM specificity than Cas9 is Clustered Regularly Interspaced Short Palindromic Repeats from Prevotella and Francisella 1 (i.e, Cas12a (Cpf1)). Similar to Cas9, Cas12a (Cpf1) is also a Class 2 CRISPR effector, but it is a member of type V subgroup of enzymes, rather than the type II subgroup. It has been shown that Cas12a (Cpf1) mediates robust DNA interference with features distinct from Cas9. Cas12a (Cpf1) is a single RNA-guided endonuclease lacking tracrRNA, and it utilizes a T-rich protospacer-adjacent motif (TTN, TTTN, or YTN). Moreover, Cpf1 cleaves DNA via a staggered DNA double-stranded break. Out of 16 Cpf1- family proteins, two enzymes from Acidaminococcus and Lachnospiraceae are shown to have efficient genome-editing activity in human cells. Cpf1 proteins are known in the art and have been described previously, for example Yamano et al.,“Crystal structure of Cpf1 in complex with guide RNA and target DNA.” Cell (165) 2016, p.949-962; the entire contents of which is hereby incorporated by reference. [00472] In still other embodiments, the Cas protein may include any CRISPR associated protein, including but not limited to, Cas12a, Cas12b1, Cas1, Cas1B, Cas2, Cas3, Cas4, Cas5, Cas6, Cas7, Cas8, Cas9 (also known as Csn1 and Csx12), Cas10, Csy1, Csy2, Csy3, Cse1, Cse2, Csc1, Csc2, Csa5, Csn2, Csm2, Csm3, Csm4, Csm5, Csm6, Cmr1, Cmr3, Cmr4, Cmr5, Cmr6, Csb1, Csb2, Csb3, Csx17, Csx14, Csx10, Csx16, CsaX, Csx3, Csx1, Csx15, Csf1, Csf2, Csf3, Csf4, homologs thereof, or modified versions thereof. [00473] In various other embodiments, the RNA-guided nuclease can be any of the following proteins: a Cas9, a Cas12a (Cpf1), a Cas12e (CasX), a Cas12d (CasY), a Cas12b1 (C2c1), a Cas13a (C2c2), a Cas12c (C2c3), a GeoCas9, a CjCas9, a Cas12g, a Cas12h, a Cas12i, a Cas13b, a Cas13c, a Cas13d, a Cas14, a Csn2, an xCas9, an SpCas9-NG, or an Argonaute (Ago) domain, or a variant thereof. [00474] Amino acid sequences of RNA-guided nucleases are readily available and known in the art. Exemplary RNA-guided nucleases and their amino acid sequences can be found, for example, in WO 2017/070633, US 2020/0010835, US 2022/0204975, US 11071790, WO 2020/191233, US 11447770, US 10858639, and US 10947530, each of which are incorporated herein by reference in their entireties. E. Programmable Nucleases (Other) [00475] In addition to the CRISPR/Cas system, the subject engineered retron can also be used in combination with sequence-specific nucleases that do not use a guide RNA to recognize a target sequence, such as non-CRISPR/Cas sequence-specific nucleases including TALENs, ZFNs, meganucleases, and restriction enzymes, as well as other sequence-specific nucleases that use other RNA guides, such as transposon-encoded IscBs, IscR, or TnpBs. [00476] For example, the subject engineered retron may encode or provide a msDNA that can serve as a donor or template sequence for HDR-mediated genome editing. Optionally, the RT of the engineered retron is fused to such sequence-specific nuclease, such that the msDNA, by way of being generated by the RT close to the site of HDR-mediated genome editing, can be more efficiently participate in the HDR-mediated genome editing. [00477] In some embodiments, the non-CRISPR/Cas sequence-specific nuclease is or comprises a TALE Nuclease, a TALE nickase, Zinc Finger (ZF) Nuclease, ZF Nickase, meganuclease, or a combination thereof. In some embodiments, the non-CRISPR/Cas sequence- specific nuclease is or includes two, three, four, or more of an independently selected TALE Nuclease, TALE nickase, Zinc Finger (ZF) Nuclease, ZF Nickase, Meganuclease, restriction enzymes or a combination thereof. In some embodiments, the combination is or comprises a TALE Nuclease/a ZF Nuclease; a TALE Nickase/a ZF nickase. [00478] In some embodiments, the non-CRISPR/Cas sequence-specific nuclease is or comprises a TALE Nuclease (Transcription Activator-Like Effector Nucleases (TALEN)). TALENs are restriction enzymes engineered to cut specific target DNA sequences. TALENs comprise a TAL effector (TALE) DNA-binding domain (which binds at or close to the target DNA), fused to a DNA cleavage domain which cuts target DNA. TALEs are engineered to bind to practically any desired DNA sequence. Thus in some embodiments, the TALEN comprises an N-terminal capping region, a DNA binding domain which may comprise at least one or more TALE monomers or half-monomers specifically ordered to target the genomic locus of interest, and a C-terminal capping region, wherein these three parts are arranged in a predetermined N- terminus to C-terminus orientation. Optionally, the TALEN includes at least one or more regulatory or functional protein domains. [00479] In some embodiments, the TALE monomers or half monomers may be variant TALE monomers derived from natural or wild type TALE monomers but with altered amino acids at positions usually highly conserved in nature, and in particular have a combination of amino acids as RVDs that do not occur in nature, and which may recognize a nucleotide with a higher activity, specificity, and/or affinity than a naturally occurring RVD. The variants may include deletions, insertions and substitutions at the amino acid level, and transversions, transitions and inversions at the nucleic acid level at one or more locations. The variants may also include truncations. [00480] In some embodiments, the TALE monomer / half monomer variants include homologous and functional derivatives of the parent molecules. In some embodiments, the variants are encoded by polynucleotides capable of hybridizing under high stringency conditions to the parent molecule-encoding wild-type nucleotide sequences. [00481] In some embodiments, the DNA binding domain of the TALE has at least 5 of more TALE monomers and at least one or more half-monomers specifically ordered or arranged to target a genomic locus of interest. The construction and generation of TALEs or polypeptides of the invention may involve any of the methods known in the art. [00482] Naturally occurring TALEs or “wild type TALEs” are nucleic acid binding proteins secreted by numerous species of proteobacteria. TALEs contain a nucleic acid binding domain composed of tandem repeats of highly conserved monomer polypeptides that are predominantly 33, 34 or 35 amino acids in length and that differ from each other mainly in amino acid positions 12 and 13. A general representation of a TALE monomer which is comprised within the DNA binding domain is Xl-11-(X12X13)-X14-33 or 34 or 35, where the subscript indicates the amino acid position and X represents any amino acid. X12X13 indicate the RVDs. In some polypeptide monomers, the variable amino acid at position 13 is missing or absent and in such monomers, the RVD consists of a single amino acid. In such cases the RVD may be alternatively represented as X*, where X represents X12 and (*) indicates that X13 is absent. The DNA binding domain may comprise several repeats of TALE monomers and this may be represented as (Xl-11-(X12X13)-X14-33 or 34 or 35)z, where z is optionally at least 5- 40, such as 10-26. [00483] The TALE monomers have a nucleotide binding affinity that is determined by the identity of the amino acids in its RVD. Polypeptide monomers with an RVD of NI preferentially bind to adenine (A), monomers with an RVD of NG preferentially bind to thymine (T), monomers with an RVD of HD preferentially bind to cytosine (C), monomers with an RVD of NN preferentially bind to both adenine (A) and guanine (G), monomers with an RVD of IG preferentially bind to T, monomers with an RVD of NS recognize all four base pairs and may bind to A, T, G or C. Thus, the number and order of the polypeptide monomer repeats in the nucleic acid binding domain of a TALE determines its nucleic acid target specificity. The structure and function of TALEs is further described in, for example, Moscou et al., Science 326:1501 (2009); Boch et al., Science 326:1509-1512 (2009); and Zhang et al., Nature Biotechnology 29:149-153 (2011), each of which is incorporated by reference in its entirety. [00484] In some embodiments, the TALE is a dTALE (or designerTALE), see Zhang et al., Nature Biotechnology 29:149-153 (2011), incorporated herein by reference. [00485] In some embodiments, the TALE monomer comprises an RVD of HN or NH that preferentially binds to guanine, and the TALEs have high binding specificity for guanine containing target nucleic acid sequences. In come embodiments, polypeptide monomers having RVDs RN, NN, NK, SN, NH, KN, HN, NQ, HH, RG, KH, RH and SS preferentially bind to guanine. In some embodiments, polypeptide monomers having RVDs RN, NK, NQ, HH, KH, RH, SS and SN preferentially bind to guanine. In some embodiments, polypeptide monomers having RVDs HH, KH, NH, NK, NQ, RH, RN and SS preferentially bind to guanine. In some embodiments, the RVDs that have high binding specificity for guanine are RN, NH RH and KH. In some embodiments, polypeptide monomers having an RVD of NV preferentially bind to adenine and guanine as do monomers having the RVD HN. Monomers having an RVD of NC preferentially bind to adenine, guanine and cytosine, and monomers having an RVD of S (or S*), bind to adenine, guanine, cytosine and thymine with comparable affinity. In more embodiments, monomers having RVDs of H*, HA, KA, N*, NA, NC, NS, RA, and S* bind to adenine, guanine, cytosine and thymine with comparable affinity. Such polypeptide monomers allow for the generation of degenerative TALEs able to bind to a repertoire of related, but not identical, target nucleic acid sequences. [00486] In certain embodiments, the TALE polypeptide has a nucleic acid binding domain containing polypeptide monomers arranged in a predetermined N-terminus to C-terminus order such that each polypeptide monomer binds to a nucleotide of a predetermined target nucleic acid sequence, and where at least one of the polypeptide monomers has an RVD of HN or NH and preferentially binds to guanine, an RVD of NV and preferentially binds to adenine and guanine, an RVD of NC and preferentially binds to adenine, guanine and cytosine or an RVD of S and binds to adenine, guanine, cytosine and thymine. [00487] In some embodiments, each polypeptide monomer of the nucleic acid binding domain that binds to adenine has an RVD of NI, NN, NV, NC or S. [00488] In certain embodiments, each polypeptide monomer of the nucleic acid binding domain that binds to guanine has an RVD of HN, NH, NN, NV, NC or S. [00489] In certain embodiments, each polypeptide monomer of the nucleic acid binding domain that binds to cytosine has an RVD of HD, NC or S. [00490] In some embodiments, each polypeptide monomer that binds to thymine has an RVD of NG or S. [00491] In some embodiments, each polypeptide monomer of the nucleic acid binding domain that binds to adenine has an RVD of NI. [00492] In certain embodiments, each polypeptide monomer of the nucleic acid binding domain that binds to guanine has an RVD of HN or NH. [00493] In certain embodiments, each polypeptide monomer of the nucleic acid binding domain that binds to cytosine has an RVD of HD. [00494] In some embodiments, each polypeptide monomer that binds to thymine has an RVD of NG. [00495] In certain embodiments, the RVDs that have a specificity for adenine are NI, RI, KI, HI, and SI. [00496] In certain embodiments, the RVDs that have a specificity for adenine are HN, SI and RI, most preferably the RVD for adenine specificity is SI. [00497] In certain embodiments, the RVDs that have a specificity for thymine are NG, HG, RG and KG. [00498] In certain embodiments, the RVDs that have a specificity for thymine are KG, HG and RG, most preferably the RVD for thymine specificity is KG or RG. [00499] In certain embodiments, the RVDs that have a specificity for cytosine are HD, ND, KD, RD, HH, YG and SD. [00500] In certain embodiments, the RVDs that have a specificity for cytosine are SD and RD. [00501] FIG.4B of WO 2012/067428 provides representative RVDs and the nucleotides they target, the entire content of which is hereby incorporated herein by reference. [00502] In certain embodiments, the variant TALE monomers may comprise any of the RVDs that exhibit specificity for a nucleotide as depicted in FIG.4A of WO2012/067428. All such TALE monomers allow for the generation of degenerative TALEs able to bind to a repertoire of related, but not identical, target nucleic acid sequences. [00503] In certain embodiments, the RVD SH may have a specificity for G, the RVD IS may have a specificity for A, and the RVD IG may have a specificity for T. [00504] In certain embodiments, the RVD NT may bind to G and A. In certain embodiments, the RVD NP may bind to A, T and C. In certain embodiments, at least one selected RVD may be NI, HD, NG, NN, KN, RN, NH, NQ, SS, SN, NK, KH, RH, HH, KI, HI, RI, SI, KG, HG, RG, SD, ND, KD, RD, YG, HN, NV, NS, HA, S*, N*, KA, H*, RA, NA or NC. [00505] The predetermined N-terminal to C-terminal order of the one or more polypeptide monomers of the nucleic acid or DNA binding domain determines the corresponding predetermined target nucleic acid sequence to which the TALE or polypeptides of the invention may bind. [00506] As used herein the monomers and at least one or more half monomers are “specifically ordered to target” the genomic locus or gene of interest. In plant genomes, the natural TALE-binding sites always begin with a thymine (T), which may be specified by a cryptic signal within the non-repetitive N-terminus of the TALE polypeptide; in some cases this region may be referred to as repeat 0. In animal genomes, TALE binding sites do not necessarily have to begin with a thymine (T) and polypeptides of the invention may target DNA sequences that begin with T, A, G or C. The tandem repeat of TALE monomers always ends with a half- length repeat or a stretch of sequence that may share identity with only the first 20 amino acids of a repetitive full length TALE monomer and this half repeat may be referred to as a half- monomer (FIG.8 of WO 2012/067428). Therefore, it follows that the length of the nucleic acid or DNA being targeted is equal to the number of full monomers plus two (see FIG.44 of WO 2012/067428). [00507] In certain embodiments, nucleic acid binding domains are engineered to contain 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, or more polypeptide monomers arranged in a N-terminal to C-terminal direction to bind to a predetermined 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25 nucleotide length nucleic acid sequence. [00508] In certain embodiments, nucleic acid binding domains are engineered to contain 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26 or more full length polypeptide monomers that are specifically ordered or arranged to target nucleic acid sequences of length 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27 and 28 nucleotides, respectively. In certain embodiments, the polypeptide monomers are contiguous. In some embodiments, half-monomers may be used in the place of one or more monomers, particularly if they are present at the C-terminus of the TALE. [00509] Polypeptide monomers are generally 33, 34 or 35 amino acids in length. With the exception of the RVD, the amino acid sequences of polypeptide monomers are highly conserved or as described herein, the amino acids in a polypeptide monomer, with the exception of the RVD, exhibit patterns that effect TALE activity, the identification of which may be used in preferred embodiments of the invention. [00510] In certain embodiments, when the DNA binding domain may comprise (Xl-11- X12X13-X14-33 or 34 or 35)z, wherein Xl-11 is a chain of 11 contiguous amino acids, wherein X12X13 is a repeat variable diresidue (RVD), wherein X14-33 or 34 or 35 is a chain of 21, 22 or 23 contiguous amino acids, wherein z is at least 5 to 26, then the preferred combinations of amino acids are LTLD or LTLA or LTQV at Xl-4, or EQHG or RDHG at positions X30-33 or X31-34 or X32-35. Furthermore, other amino acid combinations of interest in the monomers are LTPD at Xl-4 and NQALE at XI 6-20 and DHG at X32-34 when the monomer is 34 amino acids in length. When the monomer is 33 or 35 amino acids long, then the corresponding shift occurs in the positions of the contiguous amino acids NQALE and DHG. In certain embodiments, NQALE is at X15-19 or X17-21 and DHG is at X31-33 or X33-35. [00511] In certain embodiments, amino acid combinations of interest in the monomers, are LTPD at Xl-4 and KRALE at X16-20 and AHG at X32-34 or LTPE at Xl-4 and KRALE at XI 6- 20 and DHG at X32-34 when the monomer is 34 amino acids in length. When the monomer is 33 or 35 amino acids long, the corresponding shift occurs in the positions of the contiguous amino acids KRALE, AHG and DHG. In certain embodiments, the positions of the contiguous amino acids may be (LTPD at Xl-4 and KRALE at X15-19 and AHG at X31-33) or (LTPE at Xl-4 and KRALE at X15-19 and DHG at X31-33) or (LTPD at Xl-4 and KRALE at X17-21 and AHG at X33-35) or (LTPE at Xl-4 and KRALE at X17-21 and DHG at X33-35). [00512] In certain embodiments, contiguous amino acids [NGKQALE] are present at positions X14-20 or X13-19 or X15-21. These representative positions put forward various embodiments of the invention and provide guidance to identify additional amino acids of interest or combinations of amino acids of interest in all the TALE monomers (see FIGs.24A-24F, and 25 of WO 2012/067428). [00513] Exemplary amino acid sequences of conserved portions of polypeptide monomers are provided below. The position of the RVD in each sequence is represented by XX or by X* (wherein (*) indicates that the RVD is a single amino acid and residue 13 (X13) is absent). LTPAQVVAIASXXGGKQALETVQRLLPVLCQDHG LTPAQVVAIASX*GGKQALETVQRLLPVLCQDHG LTPDQVVAIANXXGGKQALATVQRLLPVLCQDHG LTPDQVVAIANXXGGKQALETLQRLLPVLCQDHG LTPDQVVAIANXXGGKQALETVQRLLPVLCQDHG LTPDQVVAIASXXGGKQALATVQRLLPVLCQDHG LTPDQVVAIASXXGGKQALETVQRLLPVLCQDHG LTPDQVVAIASXXGGKQALETVQRVLPVLCQDHG LTPEQVVAIASXXGGKQALETVQRLLPVLCQAHG LTPYQVVAIASXXGSKQALETVQRLLPVLCQDHG LTREQVVAIASXXGGKQALETVQRLLPVLCQDHG LSTAQVVAIASXXGGKQALEGIGEQLLKLRTAPYG LSTAQVVAVASXXGGKPALEAVRAQLLALRAAPYG [00514] A further listing of TALE monomers excluding the RVDs which may be denoted in a sequence (X1-11-X14-34 or Xl-11-X14-35), wherein X is any amino acid and the subscript is the amino acid position is provided in FIG.24A-F of WO 2012/067428, which is incorporated herein by reference. [00515] In certain embodiments, TALE polypeptide binding efficiency is increased by including amino acid sequences from the “capping regions” that are directly N-terminal or C- terminal of the DNA binding region of naturally occurring TALEs into the engineered TALEs at positions N-terminal or C-terminal of the engineered TALE DNA binding region. Thus, in certain embodiments, the TALE polypeptides described herein further comprise an N-terminal capping region and/or a C-terminal capping region. [00516] An exemplary amino acid sequence of a N-terminal capping region is: MDPIRSRTPSPARELLSGPQPDGVQPTADRGVSPPAGGPLDGLPARRTMSRTRLPSPPAPSPAF SADSFSDLLRQFDPSLFNTSLFDSLPPFGAHHTEAATGEWDEVQSGLRAADAPPPTMRVAVTAA RPPRAKPAPRRRAAQPSDASPAAQVDLRTLGYSQQQQEKIKPKVRSTVAQHHEALVGHGFTHAH IVALSQHPAALGTVAVKYQDMIAALPEATHEAIVGVGKQWSGARALEALLTVAGELRGPPLQLD TGQLLKIAKRGGVTAVEAVHAWRNALTGAPLN [00517] An exemplary amino acid sequence of a C-terminal capping region is: RPALESIVAQLSRPDPALAALTNDHLVALACLGGRPALDAVKKGLPHAPALIKRTNRRIPERTS HRVADHAQVVRVLGFFQCHSHPAQAFDDAMTQFGMSRHGLLQLFRRVGVTELEARSGTLPPASQ RWDRILQASGMKRAKPSPTSTQTPDQASLHAFADSLERDLDAPSPMHEGDQTRAS [00518] As used herein the predetermined “N-terminus” to “C terminus” orientation of the N-terminal capping region, the DNA binding domain comprising the repeat TALE monomers and the C-terminal capping region provide structural basis for the organization of different domains in the d-TALEs or polypeptides of the invention. [00519] The entire N-terminal and/or C-terminal capping regions are not necessary to enhance the binding activity of the DNA binding region. Therefore, in certain embodiments, fragments of the N-terminal and/or C-terminal capping regions are included in the TALE polypeptides described herein. [00520] In certain embodiments, the TALE (including TALEs) polypeptides described herein contain a N-terminal capping region fragment that included at least 10, 20, 30, 40, 50, 54, 60, 70, 80, 87, 90, 94, 100, 102, 110, 117, 120, 130, 140, 147, 150, 160, 170, 180, 190, 200, 210, 220, 230, 240, 250, 260 or 270 amino acids of an N-terminal capping region. In certain embodiments, the N-terminal capping region fragment amino acids are of the C-terminus (the DNA-binding region proximal end) of an N-terminal capping region. N-terminal capping region fragments that include the C-terminal 240 amino acids enhance binding activity equal to the full length capping region, while fragments that include the C-terminal 147 amino acids retain greater than 80% of the efficacy of the full length capping region, and fragments that include the C-terminal 117 amino acids retain greater than 50% of the activity of the full-length capping region. [00521] In some embodiments, the TALE polypeptides described herein contain a C- terminal capping region fragment that included at least 6, 10, 20, 30, 37, 40, 50, 60, 68, 70, 80, 90, 100, 110, 120, 127, 130, 140, 150, 155, 160, 170, 180 amino acids of a C-terminal capping region. In certain embodiments, the C-terminal capping region fragment amino acids are of the N-terminus (the DNA-binding region proximal end) of a C-terminal capping region. In certain embodiments, C-terminal capping region fragments that include the C-terminal 68 amino acids enhance binding activity equal to the full length capping region, while fragments that include the C-terminal 20 amino acids retain greater than 50% of the efficacy of the full length capping region. [00522] In certain embodiments, the capping regions of the TALE polypeptides described herein do not need to have identical sequences to the capping region sequences provided herein. Thus, in some embodiments, the capping region of the TALE polypeptides described herein have sequences that are at least 50%, 60%, 70%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% identical or share identity to the capping region amino acid sequences provided herein. Sequence identity is related to sequence homology. Homology comparisons may be conducted by eye, or more usually, with the aid of readily available sequence comparison programs. These commercially available computer programs may calculate percent (%) homology between two or more sequences and may also calculate the sequence identity shared by two or more amino acid or nucleic acid sequences. In some preferred embodiments, the capping region of the TALE polypeptides described herein have sequences that are at least 95% identical or share identity to the capping region amino acid sequences provided herein. [00523] Sequence homologies may be generated by any of a number of computer programs known in the art, which include but are not limited to BLAST or FASTA. Suitable computer program for carrying out alignments like the GCG Wisconsin Bestfit package may also be used. Once the software has produced an optimal alignment, it is possible to calculate % homology, preferably % sequence identity. The software typically does this as part of the sequence comparison and generates a numerical result. % homology may be calculated over contiguous sequences, i.e., one sequence is aligned with the other sequence and each amino acid or nucleotide in one sequence is directly compared with the corresponding amino acid or nucleotide in the other sequence, one residue at a time. This is called an “ungapped” alignment. Typically, such ungapped alignments are performed only over a relatively short number of residues. [00524] Additional sequences for the conserved portions of polypeptide monomers and for N-terminal and C-terminal capping regions are included in the sequences with the following gene accession numbers: AAW59491.1, AAQ79773.2, YP_450163.1, YP_001912778.1, ZP_02242672.1, AAW59493.1, AAY54170.1, ZP_02245314.1, ZP_02243372.1, AAT46123.1, AAW59492.1, YP_451030.1, YP_001915105.1, ZP_02242534.1, AAW77510.1, ACD11364.1, ZP_02245056.1, ZP_02245055.1, ZP_02242539.1, ZP_02241531.1, ZP_02243779.1, AAN01357.1, ZP_02245177.1, ZP_02243366.1, ZP_02241530.1, AAS58130.3, ZP_02242537.1, YP_200918.1, YP_200770.1, YP_451187.1, YP_451156.1, AAS58127.2, YP_451027.1, UR_451025.1, AAA92974.1, UR_001913755.1, ABB70183.1, UR_451893.1, UR_450167.1, ABY60855.1, UR_200767.1, ZR_02245186.1, ZR_02242931.1, ZR_02242535.1, AAU54169.1, UR_450165.1, UR_001913452.1, AAS58129.3, ACM44927.1, ZR_02244836.1, AAT46125.1, UR_450161.1, ZR_02242546.1, AAT46122.1, UR_451897.1, AAF98343.1, UR_001913484.1, AAY54166.1, UR_001915093.1, UR_001913457.1, ZR_02242538.1, UR_200766.1, UR_453043.1, UR_001915089.1, UR_001912981.1, ZR_02242929.1, UR_001911730.1, UR_201654.1, UR_199877.1, ABB70129.1, UR_451696.1, UR_199876.1, AAS75145.1, AAT46124.1, UR_200914.1, UR 001915101.1, ZR_02242540.1, AAG02079.2, UR_451895.1, YP 451189.1, UR_200915.1, AAS46027.1, UR_001913759.1, UR_001912987.1, AAS58128.2, AAS46026.1, UR_201653.1, UR_202894.1, UR_001913480.1, ZR_02242666.1, R_001912775.1, ZR_02242662.1, AAS46025.1, AAC43587.1, BAA37119.1, NPJ544725.1, AB077779.1, BAA37120.1, ACZ62652.1, BAF46271.1, ACZ62653.1, NPJ544793.1, ABO77780.1, ZR_02243740.1, ZR_02242930.1, AAB69865.1, AAY54168.1, ZR_02245191.1, UR_001915097.1, ZR_02241539.1, UR_451158.1, BAA37121.1, UR_001913182.1, UR_200903.1, ZR_02242528.1, ZR_06705357.1, ZR_06706392.1, ADI48328.1, ZR_06731493.1, ADI48327.1, AB077782.1, ZR 06731656.1, NR_942641.1, AAY43360.1, ZR_06730254.1, ACN39605.1, UR_451894.1, UR_201652.1, UR_001965982.1, BAF46269.1, NPJ544708.1, ACN82432.1, AB077781.1, P14727.2, BAF46272.1, AAY43359.1, BAF46270.1, NR_644743.1, ABG37631.1, AAB00675.1, YP 199878.1, ZR_02242536.1, CAA48680.1, ADM80412.1, AAA27592.1, ABG37632.1, ABP97430.1, ZR_06733167.1, AAY43358.1, 2KQ5_A, BAD42396.1, ABO27075.1, UR_002253357.1, UR_002252977.1, ABO27074.1, ABO27067.1, ABO27072.1, ABO27068.1, UR_003750492.1, ABO27073.1, NR_519936.1, ABO27071.1, AB027070.1, and ABO27069.1, each of which is hereby incorporated by reference. [00525] In some embodiments, the TALEs described herein also include a nuclear localization signal and/or cellular uptake signal. Such signals are known in the art and may target a TALE to the nucleus and/or intracellular compartment of a cell. Such cellular uptake signals include, but are not limited to, the minimal Tat protein transduction domain which spans residues 47-57 of the human immunodeficiency virus Tat protein: YGRKKRRQRRR. [00526] In some embodiments, the TALEs described herein include a nucleic acid or DNA binding domain that is a non-TALE nucleic acid or a non-TALE DNA binding domain. [00527] As used herein the term “non-TALE DNA binding domain” refers to a DNA binding domain that has a nucleic acid sequence corresponding to a nucleic acid sequence which is not substantially homologous to a nucleic acid that encodes for a TALE protein or fragment thereof, e.g., a nucleic acid sequence which is different from a nucleic acid that encodes for a TALE protein and which is derived from the same or a different organism. [00528] In certain embodiments, the TALEs described herein include a nucleic acid or DNA binding domain that is linked to a non-TALE polypeptide. [00529] A “non-TALE polypeptide” refers to a polypeptide having an amino acid sequence corresponding to a protein which is not substantially homologous to a TALE protein or fragment thereof, e.g., a protein which is different from a TALE protein and which is derived from the same or a different organism. In this context, the term “linked” is intended include any manner by which the nucleic acid binding domain and the non-TALE polypeptide could be connected to each other, including, for example, through peptide bonds by being part of the same polypeptide chain or through other covalent interactions, such as a chemical linker. The non- TALE polypeptide may be linked, for example to the N-terminus and/or C-terminus of the nucleic acid binding domain, may be linked to a C-terminal or N-terminal cap region, or may be connected to the nucleic acid binding domain indirectly. [00530] In certain embodiments, the TALEs or polypeptides of the invention comprise chimeric DNA binding domains. Chimeric DNA binding domains may be generated by fusing a full TALE (including the N- and C- terminal capping regions) with another TALE or non-TALE DNA binding domain such as zinc finger (ZF), helix-loop-helix, or catalytically-inactivated DNA endonucleases (e.g., EcoRI, meganucleases, etc.), or parts of TALE may be fused to other DNA binding domains. The chimeric domain may have novel DNA binding specificity that combines the specificity of both domains. [00531] In certain embodiments, the TALE polypeptides of the invention include a nucleic acid binding domain linked to the one or more effector domains. In certain embodiments, the effector domain is a nickase or nuclease. [00532] In certain embodiments, the sequence-specific nuclease is a zinc finger nuclease (ZFN), such as an artificial zinc-finger nuclease having arrays of zinc-finger (ZF) modules to target new DNA-binding sites in a target sequence (e.g., target sequence or target site in the genome). Each zinc finger module in a ZF array targets three DNA bases. A customized array of individual zinc finger domains is assembled into a ZF protein (ZFP). The resulting ZFP can be linked to a functional domain such as a nuclease. [00533] ZF nucleases (ZFN) may be used as alternative programmable nucleases for use in retron-based editing in place of RNA-guide nucleases. ZFN proteins have been extensively described in the art, for example, in Carroll et al.,“Genome Engineering with Zinc-Finger Nucleases,” Genetics, Aug 2011, Vol.188: 773-782; Durai et al.,“Zinc finger nucleases: custom- designed molecular scissors for genome engineering of plant and mammalian cells,” Nucleic Acids Res, 2005, Vol.33: 5978-90; and Gaj et al., “ZFN, TALEN, and CRISPR/Cas-based methods for genome engineering,” Trends Biotechnol.2013, Vol.31: 397-405, each of which are incorporated herein by reference in their entireties. [00534] In certain embodiments, the ZF-linked nuclease is a catalytic domain of the Type IIS restriction enzyme FokI (see Kim et al., PNAS U.S.A.91:883-887, 1994; Kim et al., PNAS U.S.A.93:1156-1160, 1996, both incorporated herein by reference). [00535] In certain embodiments, the ZFN comprises paired ZFN heterodimers, resulting in increased cleavage specificity and/or decreased off-target activity. In this embodiment, each ZFN in the heterodimer targets different nucleotide sequences separated by a short spacer (see Doyon et al., Nat. Methods 8:74-79, 2011, incorporated herein by reference). [00536] In certain embodiments, the ZFN comprises a polynucleotide-binding domain (comprising multiple sequence-specific ZF modules) and a polynucleotide cleavage nickase domain. [00537] In certain embodiments, the ZFs are engineered using libraries of two finger modules. [00538] In certain embodiments, strings of two-finger units are used in ZFNs to improve DNA binding specificity from polyzinc finger peptides (see PNAS USA 98: 1437-1441, incorporated herein by reference). [00539] In certain embodiments, the ZFN has more than 3 fingers. In certain embodiments, the ZFN has 4, 5, or 6 fingers. In certain embodiments, the ZF modules in the ZFN are separated by one or more linkers to improve specificity. [00540] In certain embodiments, the ZF of the ZFN includes substitutions in the dimer interface of the cleavage domain that prevent homodimerization between ZFs, but allow heterodimers to form. [00541] In certain embodiments, the ZF of the ZFN has a design that retains activity while suppressing homodimerization. [00542] In certain embodiments, the ZFN is any one of the ZF nucleases in Table 1 of Carroll et al., Genetics 188(4):773-782, 2011, incorporated herein by reference. [00543] General principles and guidance for generating ZF, ZF arrays, and ZFN can be found in the art, such as the modular design (where the different modules can be rearranged and assembled into new combinations for new targets) of the ZF or ZF arrays in the ZFN as taught in Carroll et al., Nat. Protoc.1: 1329-1341, 2006 (incorporated herein by reference); the new three- finger sets for engineered ZFs generated by using partially randomized libraries; profiling the DNA-binding specificities of engineered Cys2His2 zinc finger domains using a rapid cell-based method (see Nucleic Acids Res.35: e81, incorporated by reference). ZFs for certain DNA triplets that work well in neighbor combination are described in Sander et al., 2011. Selection-free zinc- finger-nuclease engineering by context-dependent assembly (CoDA) is taught in Nat. Methods 8: 67-69). ToolGen describes the individual fingers in their collection that are best behaved in modular assembly (Kim et al., 2011). Preassembled zinc-finger arrays for rapid construction of ZFNs are taught in Nat. Methods 8:7. [00544] Additional, non-limiting ZFs and AFNz that can be adapted for use in the instant invention include those described in WO2010/065123, WO2000/041566, WO2003/080809, WO2015/143046, WO2016/183298, WO2013/044008, WO2015/031619, WO2017/136049, WO2016/014794, WO2017/091512, WO1995/009233, WO2000/023464, WO2000/042219, WO2002/026960, WO2001/083793; US9428756, US9145565, US8846578, US8524874, US6777185, US6599692, US7235354, US6503717, US7491531, US7943553, US7262054, US8680021, US7705139, US7273923, US6780590, US6785613, US7788044, US7177766, US6453242, US6794136, US7358085, US8383766, US7030215, US7013219, US7361635, US7939327, US8772453, US9163245, US7045304, US8313925, US9260726, US6689558, US8466267, US7253273, US7947873, US9388426, US8153399, US8569253, US8524221, US7951925, US9115409, US8772008, US9121072, US9624498, US6979539, US9491934, US6933113, US9567609, US7070934, US9624509, US8735153, US9567573, US6919204, US2002-0081614, US2004-0203064, US2006-0166263, US2006-0292621, US2003-0134318, US2006-0294617, US2007-0287189, US2007-0065931, US2003-0105593, US2003-0108880, US2009-0305402, US2008-0209587, US2013-0123484, US2004-0091991, US2009-0305977, US2008-0233641, US2014-0287500, US2011-0287512, US2009-0258363, US2013-0244332, US2007-0134796, US2010-0256221, US2005-0267061, US2012-0204282, US2012-0252122, US2010-0311124, US2016-0215298, US2008-0031109, US2014-0017214, US2015-0267205, US2004-0235002, US2004-0204345, US2015-0064789, US2006-0063231, US2011-0265198, US2017-0218349, alll incorporated herein by reference. [00545] Polynucleotides and vectors capable of expressing one or more of the ZFNs are also provided herein, which can be part of the vector system of the invention. The polynucleotides and vectors can be expressed in a cell, such as a eukaryotic cell, a mammalian cell, or a human cell. Suitable vectors, cells and expression systems are described in greater detail elsewhere herein, and can be suitable for use with the TALEs, the meganucleases, and the CRISPR-Cas nucleases. [00546] In certain embodiments, the sequence-specific nuclease is a meganuclease. [00547] Meganucleases are a class of sequence-specific endonucleases that recognize large DNA target sites (>12 bp). These proteins can cleave a unique chromosomal sequence without affecting overall genome integrity. Meganucleases create site specific DNA DSBs, and, in the presence of donor DNA, such as one present in the heterologous nucleic acid encompassed by or encoded by the engineered retron of the invention, promotes the integration of the donor DNA at the cleavage site through homologous recombination (HR). [00548] In certain embodiments, the meganuclease is a homing endonuclease, which is a widespread class of proteins found in eukaryotes, bacteria and archaea. In certain embodiments, the meganuclease is of the LAGLIDADG family of homing endonucleases. [00549] In certain embodiments, the meganuclease is I-Scel, I-Cre-I, I-Dmol, or an engineered or a naturally occurring variant thereof. The hallmark of these proteins is a well conserved LAGLIDADG peptide motif, termed (do)decapeptide, found in one or two copies. Homing endonucleases with only one such motif, such as I-Crel or I-Ceul, function as homodimers. In contrast, larger proteins bearing two (do)decapeptide motifs, such as I-Scel, Pl- Scel and I-Dmol are single chain proteins. [00550] Additional homing nucleases are found at the website of homingendonuclease.net, which provides a database listing basic properties of known LAGLIDADG homing endonucleases. See also Taylor et al., Nucleic Acids Research 40 (Wl): W110-W116, 2012 (all incorporated herein by reference). [00551] In certain embodiments, specificity (or polynucleotide recognition) of the meganuclease is modified by altering the amino acids within the meganuclease, and/or by fusing other effector domains with the meganuclease. [00552] In certain embodiments, the meganuclease is a megaTAL, which includes a DNA binding domain from a TALE. [00553] In certain embodiments, the meganuclease is engineered to have nickase activity. [00554] Additional suitable natural and engineered meganucleases and megaTALs are described in WO2006/097853, WO2004/067736, WO2012/030747, WO2007/123636, WO2010/001189, WO2018/071565, WO2007/049095, WO2009/068937, WO2005/105989, WO2008/102198, WO2007/057781, WO2019/126558, WO2010/046786, US2010-0151556, US2014-0121115, US2011-0207199, US2012-0301456, US2013-0189759, US2011-0158974, US2010-0144012, US2014-0112904, US2013-0196320, US2010-0203031, US2010-0167357, US2012-0272348, US2012-0258537, US2011-0072527, US2013-0183282, US2014-0178942, US2012-0260356, US2013-0236946, US2010-0325745, US2011-0041194, US2014-0004608, US2011-0263028, US2011-0225664, US2013-0145487, US2013-0045539, US2012-0171191, US2015-0315557, US2014-0017731, US2011-0091441, US2014-0038239, US2010-0229252, US2009-0222937, US2010-0146651, US2013-0059387, US2011-0179507, US2013-0326644, US2006-0078552, US2004-0002092, US2012-0052582, US2009-0162937, US2010-0086533, US2009-0220476, US8802437, US7842489, US8715992, US8426177, US8476072, US9365864, US9540623, US9273296, US9290748, US8163514, US8148098, US8143016, US8143015, US8133697, US8129134, US8124369, US8119361, US7897372, US9683257, US10287626, US10273524, US10000746, US10006052, US7919605, US9018364, US10407672, US8211685, US9365864, US7476500, all incorporated herein by reference. [00555] In certain embodiments, the sequence-specific nuclease is TnpB, which is a programmable RNA-guided DNA endonuclease. It is believed that TnpB is a functional progenitor of the CRISPR-Cas nucleases. [00556] Transposons are mobile genetic elements that contain only the genes required for their transposition and its regulation. These elements encode the tnpA transposase, which is essential for mobilization, and often carry an accessory tnpB gene, which is dispensable for transposition. TnpB has been shown to be a nuclease that is guided by an RNA, derived from the right-end element of a transposon, to cleave DNA next to the 5′-TTGAT transposon-associated motif, and TnpB can be reprogrammed to cleave DNA target sites in human cells. [00557] In certain embodiments, tnpB is from D. radiodurans ISDra2 of the IS200/IS605 family. [00558] In certain embodiments, tnpB is from transposon PsiTn554. [00559] In certain embodiments, the sequence-specific nuclease is a TnpB-like protein, such as Fanzor1 or Fanzor2. These proteins are widespread in diverse eukaryotic transposable elements (TEs), and in large double-stranded DNA (dsDNA) viruses infecting eukaryotes. Fanzor and TnpB proteins share the same conserved amino acid motif in their C-terminal half regions: D-X(125, 275)-[TS]-[TS]-X-X-[C4 zinc finger]-X(5,50)-RD, but are highly variable in their N-terminal regions. Fanzor1 proteins are frequently captured by DNA transposons from different superfamilies including Helitron, Mariner, IS4-like, Sola and MuDr. In contrast, Fanzor2 proteins appear only in some IS607-type elements. [00560] In certain embodiments, the sequence-specific nuclease is IscB. [00561] ISC (insertion sequences Cas9-like) is a novel group of bacterial and archaeal DNA transposons that encode Cas9 homologs. The ISC transposon-encoded two nuclease domain-containing proteins are the likely ancestors of the CRISPR-associated Cas9. The homology region includes the arginine-rich helix and the HNH nuclease domain that is inserted into the RuvC-like nuclease domain. ISC genes, however, are not linked to Cas genes or CRISPR. They represent a distinct group of nonautonomous transposons, with many diverse families of full-length ISC transposons. Their terminal sequences (particularly 3′ termini) are similar to those of IS605 superfamily transposons that are mobilized by the Y1 tyrosine transposase encoded by the TnpA gene, and often also encode the TnpB protein containing the RuvC-like endonuclease domain. The terminal regions of the ISC and IS605 transposons contain palindromic structures that are likely recognized by the Y1 transposase. The transposons from these two groups are inserted either exactly in the middle or upstream of specific 4-bp target sites, without target site duplication. [00562] In certain embodiments, the sequence-specific nuclease is a restriction endonuclease (RE), such as an RE with stringent / long recognition sequence of at least 8 nts. [00563] In certain embodiments, the RE is a rare-cutter RE with seven and eight base pair recognition sequences. Exemplary rare-cutter RE enzyme include NotI, which cuts after the first GC of a 5ʹ-GCGGCCGC-3ʹ sequence. [00564] In certain embodiments, the components of the system – e.g., the retron encoded ncRNA or msDNA in complex with the RT, the sequence-specific nuclease, and the DNA-repair modulating biomolecule, may form multiple complexes in a so-called split complex configuration. The multiple complexes may be brought together to form a functional complex. [00565] For example, in some embodiments, a first component in the system may be a split protein or domain. A fragment of the split protein or domain may associate with a second component of the system, while another fragment of the split protein or domain may associate with a third component of the system. The two fragments of the split protein or domain may be brought together (e.g., along with the other components of the system) to form a functional complex. [00566] In certain embodiments, the split protein or domain is the sequence-specific nuclease, e.g., a CRISPR/Cas effector enzyme (e.g., Cas protein such as Cas9 or Cas12), a ZFN, a TALEN, a meganuclease, TnpB, IscB, or a restriction endonuclease (RE). [00567] In certain embodiments, the split protein or domain is the reverse transcriptase domain. [00568] In certain embodiments, the split protein or domain is the DNA-repair modulating biomolecule. [00569] For example a first fragment of the sequence-specific nuclease may associate with the reverse transcriptase domain and a second fragment of the sequence-specific nuclease may associate with the DNA-repair modulating biomolecule. The two fragments of the split protein or domain may be brought together (e.g., along with the reverse transcriptase domain and the DNA- repair modulating biomolecule) to form a functional complex. The associations between the parts of the split protein or domain may be through adaptor proteins or linkers described herein (e.g., those used for associating Cas proteins with function domains). [00570] In certain embodiments, the split protein or domain is split in the sense that the two parts of the split protein or domain substantially comprise a functioning split protein or domain. Ideally, the split should always be so that the catalytic domain(s) are unaffected. That split protein or domain may function as a sequence-specific nuclease or it may be a dead-Cas which is essentially an RNA-binding protein with very little or no catalytic activity, due to typically mutation(s) in its catalytic domains. [00571] Each fragment of the split protein or domain may be fused to a dimerization partner. For example, rapamycin sensitive dimerization domains enables a chemically inducible split protein or domain for temporal control of the split protein or domain’s activity. The split protein or domain can thus be rendered chemically inducible by being split into two fragments and that rapamycin-sensitive dimerization domains may be used for controlled reassembly of the split protein or domain. The two parts of the split protein or domain can be thought of as the N’ terminal part and the C’ terminal part of the split protein or domain. The fusion is typically at the split point of the split protein or domain. In other words, the C’ terminal of the N’ terminal part of the split protein or domain is fused to one of the dimer halves, whilst the N’ terminal of the C’ terminal part is fused to the other dimer half. [00572] The split protein or domain does not have to be split in the sense that the break is newly created. The split point is typically designed in silico and cloned into the constructs. Together, the two parts of the split protein or domain, the N’ terminal and C’ terminal parts, form a full split protein or domain, comprising preferably at least 70% or more of the wildtype amino acids (or nucleotides encoding them), at least 80% or more, at least 90% or more, at least 95% or more, and at least 99% or more of the wildtype amino acids (or nucleotides encoding them). When the two parts are brought together, the desired split protein or domain function is restored or reconstituted. The dimer may be a homodimer or a heterodimer. [00573] In certain embodiments, the protein components of the system – e.g., the RT, the sequence-specific nuclease (a CRISPR/Cas effector enzyme, a ZFN, a TALEN, a meganuclease, TnpB, IscB, or a restriction endonuclease (RE)), and the DNA-repair modulating biomolecule, may further comprise one or more additional functional domains. [00574] In certain embodiments, the functional domain comprises a nuclear localization signal (NLS). In certain embodiments, one or more C-terminal or N-terminal NLSs are attached. In certain embodiments, a C-terminal NLS is attached for expression and nuclear targeting in eukaryotic cells, e.g., human cells. In certain embodiments, the NLS(s) may be at a location that is not at the C-terminus or N-terminus, for example, the NLS(s) may be between two polypeptides. [00575] Non-limiting examples of NLSs include an NLS sequence derived from: the NLS of the SV40 virus large T-antigen; the NLS from nucleoplasmin (e.g., the nucleoplasmin bipartite NLS); the c-myc NLS; the hRNPAl M9 NLS; the NLS of the IBB domain from importin-alpha; the NLS of the myoma T protein; the NLS of human p53; the NLS of mouse c- abl IV; the NLS of the influenza virus NS1; the NLS of the Hepatitis virus delta antigen; the NLS of the mouse Mxl protein; the NLS of the human poly(ADP-ribose) polymerase; and the NLS of the steroid hormone receptors (human) glucocorticoid. Exemplary NLS sequences include those described in paragraph [00106] of Feng Zhang et al. (WO2016/106236), all incorporated herein by reference. [00576] In certain embodiments, the functional domain comprises at least two NLS domains. The one or more NLS domain(s) may be positioned at or near or in proximity to a terminus of a polypeptide and, if two or more NLSs, each of the two may be positioned at or near or in proximity to a terminus of the polypeptide. [00577] In any of the fusion proteins, the fusion between the two domains (such as the RT and Cas enzyme, or the Cas and the DNA-repair modulating biomolecule) may be linked through a linker. [00578] A “linker” as used herein includes a peptide which joins two proteins or domains to form a fusion protein. Generally, such molecules have no specific biological activity other than to join or to preserve some minimum distance or other spatial relationship between the proteins / domains. However, in certain embodiments, the linker may be selected to influence some property of the linker and/or the fusion protein such as the folding, net charge, or hydrophobicity of the linker. Suitable linkers for use in the present disclosure are well-known to those of skill in the art and include, but are not limited to, straight or branched-chain carbon linkers, heterocyclic carbon linkers, or peptide linkers. However, as used herein, the linker may also be a covalent bond (carbon-carbon bond or carbon-heteroatom bond). [00579] In particular embodiments, the linker is used to separate the sequence-specific nuclease (a CRISPR/Cas effector enzyme, a ZFN, a TALEN, a meganuclease, TnpB, IscB, or a restriction endonuclease (RE)) and the RT and/or the DNA-repair modulating biomolecule by a distance sufficient to ensure that each protein domain retains its required functional property. [00580] Preferred peptide linker sequences adopt a flexible extended conformation and do not exhibit a propensity for developing an ordered secondary structure. In certain embodiments, the linker can be a chemical moiety which can be monomeric, dimeric, multimeric or polymeric. In certain embodiments, the linker comprises amino acids. Typical amino acids in flexible linkers include Gly, Asn and Ser. Accordingly, in particular embodiments, the linker comprises a combination of one or more of Gly, Asn and Ser amino acids. Other near neutral amino acids, such as Thr and Ala, also may be used in the linker sequence. [00581] In certain embodiments, the linker comprises a GlySer-rich sequence, such as a GnS linker (n = 1, 2, 3, 4, or 5, such as GS or G4S), or repeats thereof (such as 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20 repeats, optionally with an overall length of about 4-30 residues, 4-20 residues, or 4-10 residues). [00582] In certain embodiments, the linker comprises a G4S linker with 3, 6, 9, or 12 repeats. [00583] In certain embodiments, the linker is one disclosed in Maratea et al., Gene 40: 39- 46, 1985; Murphy et al., PNAS USA 83: 8258-62, 1986; US4,935,233; or US4,751,180, all incorporated by reference. [00584] In certain embodiments, the linker comprises a GlySer linker such as GGS, GGGS, GSG, GGGGS, optionally with repeats of 3 (such as (GGS)3, (GGGGS)3), 4, 5, 6, 7, 8, 9, 10, 11, or 12 or more, to provide suitable lengths. [00585] In certain embodiments, the linker comprises (GGGGS)3-15, such as (GGGGS)3-11, e.g., GGGGS with 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, or 11 repeats. [00586] In certain embodiments, the linker comprises LEPGEKPYKCPECGKSFSQSGALTRHQR THTR. [00587] In yet an additional embodiment, the linker is an XTEN linker. [00588] In certain embodiments, N-and/or C-terminal NLSs also function as linker (e.g., PKKKRKVEASSPKKRKVEAS). [00589] The gRNA and the various nucleases and fusions thereof can be provided in the form of a protein, optionally where the nuclease is complexed with a gRNA, or provided by a nucleic acid encoding the RNA-guided nuclease, such as an RNA (e.g., messenger RNA) or DNA (expression vector). In some embodiments, the RNA-guided nuclease and the gRNA are both provided by vectors. Both can be expressed by a single vector or separately on different vectors. The vectors encoding the RNA-guided nuclease and gRNA may be included in the vector system comprising the engineered retron msr gene, msd gene and ret gene sequences. [00590] Codon usage may be optimized to improve production of the engineered retron e.g., retron reverse transcriptase, ncRNA and/or RNA-guided nuclease in a particular cell or organism. For example, a nucleic acid encoding an ncRNA, RNA-guided nuclease or reverse transcriptase can be modified to substitute codons having a higher frequency of usage in a the particular cell such as a eukaryotic cell (e.g., yeast cell, a human cell, a non-human cell, a mammalian cell, a rodent cell, a mouse cell, a rat cell), or any other host cell of interest, as compared to the naturally occurring polynucleotide sequence. When a nucleic acid encoding the reverse transcriptase or ncRNA is introduced into cells, the protein can be transiently, conditionally, or constitutively expressed in the cell. F. RT-PN Fusion Proteins [00591] The recombinant retron-based editing system described herein contemplates fusion proteins comprising a programmable nuclease (PN) and a RT, optionally joined by a linker. The application contemplates any suitable programmable nuclease and RT (e.g., retron RTs of Table A) to be combined in a single fusion protein. In one embodiment, the RT is joined to the N-terminus of the PN. In another embodiment, the RT is joined to the C-terminus of the PN. The Examples of PNs and RTs are each defined herein. [00592] In various embodiments, the fusion proteins may comprise any suitable structural configuration. For example, the fusion protein may comprise from the N-terminus to the C- terminus direction, a PN fused to a RT. In other embodiments, the fusion protein may comprise from the N-terminus to the C-terminus direction, a RT fused to a NP. The fused domain may optionally be joined by a linker, e.g., an amino acid sequence. G. Nuclear Localization Signals [00593] In various embodiments, any of the polypeptide components of the retron-based editing systems may be engineered with one or more nuclear locatlization signals, which help promote translocation of a protein into the cell nucleus. The polypeptides of the retron-based editing system may comprise any known NLS sequence, including any of those described in Cokol et al.,“Finding nuclear localization signals,” EMBO Rep., 2000, 1(5): 411-415 and Freitas et al.,“Mechanisms and Signals for the Nuclear Import of Proteins,” Current Genomics, 2009, 10(8): 550-7, each of which are incorporated herein by reference. [00594] In various embodiments, the polypeptides disclosed herein may include one or more, preferably, at least two nuclear localization signals. The NLSs may be any known NLS sequence in the art. The NLSs may also be any future-discovered NLSs for nuclear localization. The NLSs also may be any naturally-occurring NLS, or any non-naturally occurring NLS (e.g., an NLS with one or more desired mutations). The term“nuclear localization sequence” or“NLS” refers to an amino acid sequence that promotes import of a protein into the cell nucleus, for example, by nuclear transport. Nuclear localization sequences are known in the art and would be apparent to the skilled artisan. For example, NLS sequences are described in Plank et al., International PCT application PCT/EP2000/011690, filed November 23, 2000, published as WO/2001/038547 on May 31, 2001, the contents of which are incorporated herein by reference. In some embodiments, an NLS comprises the amino acid sequence PKKKRKV. [00595] A representative nuclear localization signal is a peptide sequence that directs the protein to the nucleus of the cell in which the sequence is expressed. A nuclear localization signal is predominantly basic, can be positioned almost anywhere in a protein's amino acid sequence, generally comprises a short sequence of four amino acids (Autieri & Agrawal, (1998) J. Biol. Chem.273: 14731-37, incorporated herein by reference) to eight amino acids, and is typically rich in lysine and arginine residues (Magin et al., (2000) Virology 274: 11-16, incorporated herein by reference). Nuclear localization signals often comprise proline residues. A variety of nuclear localization signals have been identified and have been used to effect transport of biological molecules from the cytoplasm to the nucleus of a cell. See, e.g., Tinland et al., (1992) Proc. Natl. Acad. Sci. U.S.A.89:7442-46; Moede et al., (1999) FEBS Lett.461:229-34, which is incorporated by reference. Translocation is currently thought to involve nuclear pore proteins. [00596] Most NLSs can be classified in three general groups: (i) a monopartite NLS exemplified by the SV40 large T antigen NLS (PKKKRKV); (ii) a bipartite motif consisting of two basic domains separated by a variable number of spacer amino acids and exemplified by the Xenopus nucleoplasmin NLS (KRXXXXXXXXXXKKKL); and (iii) noncanonical sequences such as M9 of the hnRNP Al protein, the influenza virus nucleoprotein NLS, and the yeast Gal4 protein NLS (Dingwall and Laskey 1991). Nuclear localization signals appear at various points in the amino acid sequences of proteins. NLS’s have been identified at the N-terminus, the C- terminus and in the central region of proteins. Thus, the disclosure provides polypeptides that may be modified with one or more NLSs at the C-terminus, the N-terminus, as well as at in internal region of a polypeptide (including a fusion protein). [00597] The present disclosure contemplates any suitable means by which to modify a polypeptide to include one or more NLSs. In one aspect, a polypeptide (e.g., a programmable nuclease) may be engineered to express with a translationally fused NLS at its N-terminus or its C-terminus (or both), i.e., to form a polypeptide-NLS fusion construct. In addition, the NLSs may include various amino acid linkers or spacer regions encoded between a polypeptide and the N-terminally, C-terminally, or internally-attached NLS amino acid sequence, e.g, and in the central region of proteins. [00598] Thus, the present disclosure also provides for nucleotide constructs, vectors, and host cells for expressing fusion proteins that comprise a polypeptide and one or more NLSs. H. DNA-repair Modulating Biomolecules [00599] In certain embodiments, the engineered retron described herein (e.g., an engineered nucleic acid construct or engineered nucleic acid-enzyme construct described herein) further comprises or encodes a DNA-repair modulating biomolecule, which may further enhance the efficiency of integration of a transgene on the heterologous nucleic acid by homology dependent repair (HDR). [00600] In certain embodiments, the DNA-repair modulating biomolecule comprises a Nonhomologous end joining (NHEJ) inhibitor. [00601] In certain embodiments, the DNA-repair modulating biomolecule comprises a homologous directed repair (HDR) promoter. [00602] In certain embodiments, the DNA-repair modulating biomolecule comprises a NHEJ inhibitor and an HDR promoter. [00603] In certain embodiments, the DNA-repair modulating biomolecule enhances or improves more precise genome editing and/or the efficiency of homologous recombination, compared to the otherwise identical embodiment without the DNA-repair modulating biomolecule. [00604] HDR promoters and/or NHEJ inhibitors can, in some embodiments, comprise one or more small molecules. Systems bearing recombination enhancers such as small molecules that activate HDR and suppress NHEJ locally at the genomic site of the DNA damage can be tailored in their placement on the engineered systems to further enhance their efficiency. In general, the small molecule recombination enhancers can be synthesized to bear linkers and a functional group, such as maleimide for reacting with a thiol group on a Cys residue of a protein, for chemical conjugation to the engineered systems. Use of commercially available functionalized PEG linkers (alkyne, azide, cyclooctyne etc.) can also be employed for conjugation, and orthogonal conjugation chemistries can be utilized for the multivalent display. [00605] Conjugation sites can be readily identified where modifications do not affect the potency of the recombination enhancers selected. [00606] In certain embodiments, multivalent display of one or more DNA-repair modulating biomolecule can be affected, including multiple moieties of NHEJ inhibitors, HDR promoters, or a combination thereof. See, for example, “Genomic targeting of epigenetic probes using a chemically tailored Cas9 system” by Liszczak et al., Proc Natl Acad Sci U.S.A.114: 681- 686, 2017 (incorporated herein by reference). In certain embodiments, multivalent display of small molecule compounds can be achieved through sortase loop proteins used as a scaffold for their display. [00607] In some embodiments, the DNA-repair modulating biomolecule may comprise an HDR promoter. The HDR promoter may comprise small molecules, such as RSI or analogs thereof. In certain embodiments, the HDR promoter stimulates RAD51 activity or RAD52 motif protein 1 (RDMl) activity. In certain embodiments, the HDR promoter comprises Nocodazole, which can result in higher HDR selection. [00608] In certain embodiments, the HDR promoter may be administered prior to the delivery of the engineered retron described herein. [00609] In certain embodiments, the HDR promoter locally enhances HDR without NHEJ inhibition. For example, RAD5l is a protein involved in strand exchange and the search for homology regions during HDR repair. In certain embodiments, the HDR promoter is phenylbenzamide RSI, identified as a small-molecule RAD51-stimulator (see WO2019/135816 at [0200]-[0204], specifically incorporated herein by reference). [00610] In certain embodiments, the DNA-repair modulating biomolecule comprises C- terminal binding protein interacting protein (CtIP) or a functional fragment or homolog thereof. CtIP is a key protein in early steps of homologous recombination. According to this embodiment, the CtIP or the functional fragment or homolog thereof can be linked (e.g., fused) to the RT or the sequence-specific nuclease (e.g., a CRISPR/Cas effector enzyme, a ZFN, a TALEN, a meganuclease, TnpB, IscB, or a restriction endonuclease (RE)), and stimulates transgene integration by HDR. [00611] In certain embodiments, the CtIP fragment is a minimal N-terminal fragment of the wild-type CtIP, such as the N-terminal fragment comprising residues 1-296 of the full-length CtIP (the HE for HDR enhancer), as described in Charpentier et al. (Nature Comm., DOI: 10.1038/s41467-018-03475-7, incorporated herein by reference), shown to be sufficient to stimulate HDR. The activity of the fragment depends on CDK phosphorylation sites (e.g., S233, T245, and S276) and the multimerization domain essential for CtIP activity in homologous recombination. Thus alternative fragments comprising the CDK phosphorylation sites and the multimerization domain essential for CtIP activity are also within the scope of the invention. [00612] In certain embodiments, the DNA-repair modulating biomolecule comprises a dominant negative 53BP1. [00613] In certain embodiments, the DNA-repair modulating biomolecule comprises a cell cycle-specific degradation tag, such as the degradation domain of the (human) Geminin, and the (murine) CyclinB2. [00614] In certain embodiments, the DNA-repair modulating biomolecule comprises CyclinB2, a member of the B-type cyclins that associate with p34cdc2, and an essential component of the cell cycle regulatory machinery. CRISPR-mediated knock-in efficiency may be increased by promoting the relative increase in Cas9 activity in G2 phase of the cell cycle, when HDR is more active. In certain embodiments, the degradation domains of the (human) Geminin and (murine) CyclinB2 can be used as either N- or C-terminal fusion (e.g., fusions with a Cas, such as Cas9, or the retron RT) to serve as the DNA-repair modulating biomolecule. These domains are known to determine a cell-cycle specific profile of chimeric proteins, namely an increase in their relative concentration in S and G2 compared to G1, high-jacking the conventional CyclinB2 and Geminin degradation pathways. This produces active Geminin-Cas9 and CyclinB2-Cas9 chimeric proteins, which are degraded in a cell-cycle-dependent manner. Such chimeras shift the repair of the DSBs to the HDR repair pathway compared to the commonly used Cas9. [00615] While not wishing to be bound by particular theory, it is believed that the application of such cell cycle-specific degradation tags permits / promotes more efficient / secure gene editing. [00616] In certain embodiments, the DNA-repair modulating biomolecule comprises a Rad family member protein, such as Rad50, Rad51, Rad52, etc., which functions to promote foreign DNA integration into a host chromosome. Specifically, Rad52 is an important homologous recombinant protein, and its complex with Rad51 plays a key role in HDR, mainly involved in the regulation of foreign DNA in eukaryotes. Key steps in the process of HR include repair mediated by Rad51 and strand exchange. Co-expression of Rad52 as a DNA-repair modulating biomolecule significantly enhances the likelihood of HDR by, e.g., three-fold. [00617] In certain embodiments, the DNA-repair modulating biomolecule comprises a RAD52 protein as, e.g., either an N- or a C-terminal fusion. [00618] In certain embodiments, the DNA-repair modulating biomolecule comprises a RAD52 motif protein 1 (RDMl) that functions similarly as RAD52. RDM1 has been shown to be able to repair DSBs caused by DNA replication, prevent G2 or M cell cycle arrest, and improve HDR selection. [00619] In certain embodiments, the DNA-repair modulating biomolecule comprises a dominant negative version of the tumor suppressor p53-binding protein 1 (53BP1). The wild- type protein 53BP1 is a key regulator of the choice between NHEJ and HDR – it is a pro-NHEJ factor which limits HDR by blocking DNA end resection, and also by inhibiting BRCA1 recruitment to DSB sites. It has been shown that global inhibition of 53BP1 by a ubiquitin variant significantly improves Cas9-mediated HDR frequency in non-hematopoietic and hematopoietic cells with single-strand oligonucleotide delivery or double-strand donor in AAV. [00620] In certain embodiments, the dominant negative (DN) version of the 53BP1 comprises the minimal focus forming region, but lacks domains outside this region, e.g., towards the N-terminus and tandem C-terminal BRCT repeats that recruit key effectors involved in NHEJ, such as RIFl-PTIP and EXPAND, respectively. The 53BP1 adapter protein is recruited to specific histone marks at sites of DSBs via this minimal focus forming region, which comprises several conserved domains including an oligomerization domain (OD), a glycine-arginine rich (GAR) motif, a Tudor domain, and an adjacent ubiquitin-dependent recruitment (UDR) motif. The Tudor domain mediates interactions with histone H4 dimethylated at K2023. [00621] In certain embodiments, a dominant negative version of 53BP1 (DN1S) suppresses the accumulation of endogenous 53BP1 and downstream NHEJ proteins at sites of DNA damage, while upregulating the recruitment of the BRCA1 HDR protein. Such a DN version of the 53BP1 can be used as the DNA-repair modulating biomolecule, either as an N- or a C-terminal fusion (such as a Cas9 fusion, to locally inhibit NHEJ at the Cas9-target site defined by its gRNA, while promoting an increase in HDR, and does not globally affect NHEJ, thereby improving cell viability). [00622] In certain embodiments, the DNA-repair modulating biomolecule comprises an NHEJ inhibitor, such as an inhibitor of DNA ligase IV, a KU inhibitor (e.g., KU70 or KU80), a DNA-PKc inhibitor, or an artemis inhibitor. [00623] In certain embodiments, the NHEJ inhibitor inhibits the NHEJ pathway, enhances HDR, or modulates both. In certain embodiments, the NHEJ inhibitor is a small molecule inhibitor. [00624] In certain embodiments, the small molecule inhibitor of the NHEJ pathway comprises an SCR7 analog, for example, PK66, PK76, PK409. [00625] In certain embodiments, the NHEJ inhibitor comprises a KU inhibitor, for example, KU5788, and KU0060648. [00626] In certain embodiments, a small molecule NHEJ inhibitor is linked to a polyglycine tripeptide through PEG for sortase-mediated ligation, as described in WO2019/135816, Guimaraes et al., Nat Protoc 8:1787-99, 2013; Theile et al., Nat Protoc 8:1800-7, 2013; and Schmohl et al., Curr Opin Chem Biol 22:122-8, 2014 (all incorporated herein by reference). The same means can also be used for attaching small molecule HDR enhancers to protein. [00627] An exemplary method for conjugating a small molecule DNA-repair modulating biomolecule without loss of activity is described in WO2019135816, where SCR-7 conjugation of a poly-glycine peptide with the para-carboxylic moiety at ring 4 retained activity of the inhibitor, with rings 1, 2 and 3 of the molecule having involvement in the target-engagement, providing a simple and effective strategy to ligate a small molecule NHEJ inhibitor to the system described herein (e.g., to the sequence-specific nuclease including Cas enzymes, or to the RT) to precisely enhance HDR pathway near a nucleic acid target site. [00628] In certain embodiments, a nucleic acid targeting moiety conjugates based on small molecule inhibitor of DNA-dependent protein kinase (DNA-PK) or heterodimeric Ku (KU70/KU80) can be utilized. KU-0060648 is one potent KU-inhibitors, which can also be functionalized with poly-glycine and used for recombination enhancement. [00629] In certain embodiments, the DNA-repair modulating biomolecule comprises the Tumor Suppressor p53. p53 plays a direct role in DNA repair, including HR regulation, where it affects the extension of new DNA, thereby affecting HDR selection. In vivo, p53 binds to the nuclear matrix and is a rate-limiting factor in repairing DNA structure. p53 regulates DNA repair processes in almost all eukaryotes via transactivation-dependent and -independent pathways, but only the transactivation-independent function of p53 is involved in HR regulation. Wild-type p53 protein can link double stranded breaks to form intact DNA, as well as also playing a role in inhibiting NHEJ. p53 interacts with HR-related proteins, including Rad51, where it controls HR through direct interaction with Rad51. I. Vectors for Expression of Engineered Retrons [00630] Delivery of an engineered retron to a cell can generally be accomplished with or without vectors. Delivery of the ncRNA encoded by the engineered retron generally does not require a vector used to produce the ncRNA from the engineered retron. For example, the ncRNA can be packaged directly into a delivery vehicle such as a lipid nanoparticle and delivered into a host cell, as described in other sections. [00631] The engineered retrons (or vectors containing them) may be introduced into any type of cell, including any cell from a prokaryotic, eukaryotic, or archaeon organism, including bacteria, archaea, fungi, protists, plants (e.g., monocotyledonous and dicotyledonous plants); and animals (e.g., vertebrates and invertebrates). Examples of animals that may be transfected with an engineered retron include, without limitation, vertebrates such as fish, birds, mammals (e.g., human and non-human primates, farm animals, pets, and laboratory animals), reptiles, and amphibians. [00632] The engineered retrons can be introduced into a single cell or a population of cells. Cells from tissues, organs, and biopsies, as well as recombinant cells, genetically modified cells, cells from cell lines cultured in vitro, and artificial cells (e.g., nanoparticles, liposomes, polymersomes, or microcapsules encapsulating nucleic acids) may all be transfected with the engineered retrons. [00633] The engineered retrons can be introduced into cellular fragments, cell components, or organelles (e.g., mitochondria in animal and plant cells, plastids (e.g., chloroplasts) in plant cells and algae). [00634] Cells may be cultured or expanded after transfection with the engineered retron. [00635] Methods of introducing nucleic acids into a host cell are well known in the art. Commonly used methods include chemically induced transformation, typically using divalent cations (e.g., CaCl2), dextran-mediated transfection, polybrene mediated transfection, lipofectamine and LT-1 mediated transfection, electroporation, protoplast fusion, encapsulation of nucleic acids in liposomes, and direct microinjection of the nucleic acids comprising engineered retrons into nuclei. See, e.g., Sambrook et al. (2001) Molecular Cloning, a laboratory manual, 3rd edition, Cold Spring Harbor Laboratories, New York, Davis et al. (1995) Basic Methods in Molecular Biology, 2nd edition, McGraw-Hill, and Chu et al. (1981) Gene 13:197; herein incorporated by reference in their entireties. [00636] Methods for genetic transformation of plant cells are known in the art and include those set forth in US2022/0145296, and U.S. Pat. Nos.8,575,425; 7,692,068; 8,802,934; 7,541,517; each of which is herein incorporated by reference in its entirety. See, also, Rakoczy- Trojanowska, M. (2002) Cell Mol Biol Lett.7:849-858; Jones et al. (2005) Plant Methods 1:5; Rivera et al. (2012) Physics of Life Reviews 9:308-345; Bartlett et al. (2008) Plant Methods 4:1- 12; Bates, G. W. (1999) Methods in Molecular Biology 111:359-366; Binns and Thomashow (1988) Annual Reviews in Microbiology 42:575-606; Christou, P. (1992) The Plant Journal 2:275-281; Christou, P. (1995) Euphytica 85:13-27; Tzfira et al. (2004) TRENDS in Genetics 20:375-383; Yao et al. (2006) Journal of Experimental Botany 57:3737-3746; Zupan and Zambryski (1995) Plant Physiology 107:1041-1047; and Jones et al. (2005) Plant Methods 1:5. [00637] The plant cells that have been transformed may be grown into a transgenic organism, such as a plant, in accordance with conventional methods. See, for example, McCormick et al. (1986) Plant Cell Reports 5:81-84. [00638] Plant material that may be transformed with the engineered retrons described herein includes plant cells, plant protoplasts, plant cell tissue cultures from which plants can be regenerated, plant calli, plant clumps, and plant cells that are intact in plants or parts of plants such as embryos, pollen, ovules, seeds, leaves, flowers, branches, fruit, kernels, ears, cobs, husks, stalks, roots, root tips, anthers, and the like. Progeny, variants, and mutants of the regenerated plants are also included within the scope of the disclosure, provided that these parts comprise the genetic modification introduced by the engineered retron. Further provided is a processed plant product or byproduct that retains the genetic modification introduced by the engineered retron. [00639] The engineered retrons described herein may be used to produce transgenic plants with desired phenotypes, including but not limited to, increased disease resistance (e.g., increased viral, bacterial of fungal resistance), increased insect resistance, increased drought resistance, increased yield, and altered fruit ripening characteristics, sugar and oil composition, and color. [00640] In some embodiments, the retron msr gene, msd gene, and/or ret gene are expressed in vitro from a vector, such as in an in vitro transcription system. The resulting ncRNA or msDNA can be isolated before being packaged and/or formulated for direct delivery into a host cell. For example, the isolated ncRNA or msDNA can be packaged/formulated in a delivery vehicle such as lipid nanoparticles as described in other sections. [00641] In some embodiments, the retron msr gene, msd gene, and/or ret gene are expressed in vivo from a vector within a cell. The retron msr gene, msd gene, and/or ret gene can be introduced into a cell with a single vector or in multiple separate vectors to produce msDNA in a host subject. [00642] In other embodiments, the retron msr gene, msd gene, and/or ret gene, and any other components of the retron-based genome editing systems described herein (e.g., guide RNA in trans, programmable nuclease (e.g., in trans)) may be expressed in vivo from RNA delivered to the cell. The retron msr gene, msd gene, and/or ret gene can be introduced into a cell with a single vector or in multiple separate vectors to produce msDNA in a host subject. [00643] Vectors and/or nucleic acid molecules encoding the recombinant retron-based genome editing system or components thereof can include control elements operably linked to the retron sequences, which allow for the production of msDNA either in vitro, or in vivo in the subject species. For example, the retron msr gene, msd gene, and/or ret gene can be operably linked to a promoter to allow expression of the retron reverse transcriptase and/or the msDNA product. In some embodiments, heterologous sequences encoding desired products of interest (e.g., polynucleotide encoding polypeptide or regulatory RNA, donor polynucleotide for gene editing, or protospacer DNA for molecular recording) may be inserted in the msr gene and/or msd gene. [00644] Any eukaryotic, archaeal, or prokaryotic cell, capable of being transfected with a vector or retron delivery system comprising the engineered retron sequences, may be used to produce the msDNA in vivo. The ability of constructs to produce the msDNA along with other retron-encoded products can be empirically determined. For example the transfected cell can be assayed either through phenotypic changes that occur due to the introduced sequences or by direct DNA sequencing. [00645] In some embodiments, the engineered retron is produced by a vector system comprising one or more vectors. In the vector system, the msr gene, the msd gene, and/or the ret gene may be provided by the same vector (i.e., cis arrangement of all such retron elements), wherein the vector comprises a promoter operably linked to the msr gene and/or the msd gene. In some embodiments, the promoter is further operably linked to the ret gene. In other embodiments, the vector further comprises a second promoter operably linked to the ret gene. Alternatively, the ret gene may be provided by a second vector that does not include the msr gene and/or the msd gene (i.e., trans arrangement of msr-msd and ret). In yet other embodiments, the msr gene, the msd gene, and the ret gene are each provided by different vectors (i.e., trans arrangement of all retron elements). [00646] Numerous vectors are available for use in the vector or vector system, including but not limited to, linear polynucleotides, polynucleotides associated with ionic or amphiphilic compounds, plasmids, and viruses. [00647] Examples of viral vectors include, but are not limited to, adenoviral vectors, adeno-associated virus (AAV) vectors, retroviral vectors, lentiviral vectors, and the like. An expression construct can be replicated in a living cell, or it can be made synthetically. [00648] In some embodiments, the nucleic acid comprising an engineered retron sequence is under transcriptional control of a promoter. In some embodiments, the promoter is competent for initiating transcription of an operably linked coding sequence by a RNA polymerase I, II, or III. [00649] Exemplary promoters for mammalian cell expression include the SV40 early promoter, a CMV promoter such as the CMV immediate early promoter (see, U. S. Patent Nos. 5,168,062 and 5,385,839, incorporated herein by reference in their entireties), the mouse mammary tumor virus LTR promoter, the adenovirus major late promoter (Ad MLP), and the herpes simplex virus promoter, among others. Other nonviral promoters, such as a promoter derived from the murine metallothionein gene, will also find use for mammalian expression. [00650] Exemplary promoters for plant cell expression include the CaMV 35S promoter (Odell et al., 1985, Nature 313:810-812); the rice actin promoter (McElroy et al., 1990, Plant Cell 2:163-171); the ubiquitin promoter (Christensen et al., 1989, Plant Mol. Biol.12:619-632; and Christensen et al., 1992, Plant Mol. Biol.18:675-689); the pEMU promoter (Last et al., 1991, Theor. Appl. Genet.81:581-588); and the MAS promoter (Velten et al., 1984, EMBO J. 3:2723-2730). [00651] In additional embodiments, the retron-based vectors may also comprise tissue- specific promoters to start expression only after it is delivered into a specific tissue. Non-limiting exemplary tissue-specific promoters include B29 promoter, CD14 promoter, CD43 promoter, CD45 promoter, CD68 promoter, desmin promoter, elastase- 1 promoter, endoglin promoter, fibronectin promoter, Flt-1 promoter, GFAP promoter, GPIIb promoter, ICAM- 2 promoter, INF-b promoter, Mb promoter, Nphsl promoter, OG-2 promoter, SP-B promoter, SYN1 promoter, and WASP promoter. [00652] These and other promoters can be obtained from or incorporated into commercially available plasmids, using techniques well known in the art. See, e.g., Sambrook et al., supra. [00653] In some embodiments, one or more enhancer elements is/are used in association with the promoter to increase expression levels of the constructs. Examples include the SV40 early gene enhancer, as described in Dijkema et al., EMBOJ (1985) 4:761, the enhancer/promoter derived from the long terminal repeat (LTR) of the Rous Sarcoma Virus, as described in Gorman et al., Proc. Natl. Acad. Sci. USA (1982b) 79:6777, and elements derived from human CMV, as described in Boshart et al., Cell (1985) 41:521, such as elements included in the CMV intron A sequence. All such sequences are incorporated herein by reference. [00654] In one embodiment, an expression vector for expressing an engineered retron, including the msr gene, msd gene, and/or ret gene comprises a promoter operably linked to a polynucleotide encoding the msr gene, msd gene, and/or ret gene. [00655] In some embodiments, the vector or vector system also comprises a transcription terminator/polyadenylation signal. Examples of such sequences include, but are not limited to, those derived from SV40, as described in Sambrook et al., supra, as well as a bovine growth hormone terminator sequence (see, e.g., U.S. Patent No.5,122,458). [00656] Additionally, 5ʹ- UTR sequences can be placed adjacent to the coding sequence to further enhance the expression. Such sequences may include UTRs comprising an internal ribosome entry site (IRES). Inclusion of an IRES permits the translation of one or more open reading frames from a vector. The IRES element attracts a eukaryotic ribosomal translation initiation complex and promotes translation initiation. See, e.g., Kaufman et al., Nuc. Acids Res. (1991) 19:4485-4490; Gurtu et al., Biochem. Biophys. Res. Comm. (1996) 229:295-298: Rees et al., BioTechniques (1996) 20:102-110; Kobayashi et al., BioTechniques (1996) 21:399-402; and Mosser et al., BioTechniques (199722 ISO- 161)c . A multitude of IRES sequences are known and include sequences derived from a wide variety of viruses, such as from leader sequences of picomaviruses such as the encephalomyocarditis virus (EMCV) UTR (Jang et al.. Virol. (1989) 63:1651-1660). the polio leader sequence, the hepatitis A virus leader, the hepatitis C virus IRES, human rhinovirus type 2 IRES (Dobrikova et al., Proc. Natl. Acad. Sci. (2003) 100(251:15125-151301)). an IRES element from the foot and mouth disease virus (Ramesh et al., Nucl. Acid Res. (1996) 24:2697-2700), a giardiavirus IRES (Garlapati et al., J Biol. Chem. (2004) 279(51):3389-33971) and the like. A variety of nonviral IRES sequences will also find use herein, including, but not limited to IRES sequences from yeast, as well as the human angiotensin II type 1 receptor IRES (Martin et al., Mol. Cell Endocrinol. (2003) 212:51-61), fibroblast growth factor IRESs (FGF-1 IRES and FGF-2 IRES, Martineau et al. (2004) Mol. Cell. Biol.24( 17): 7622-7635), vascular endothelial growth factor IRES (Baranick et al. (2008) Proc. Natl. Acad Sci. U.S.A.105(12):4733-4738, Stein et al. (1998) Mol. Cell. Biol.18(6):3112- 3119, Bert et al. (2006) RNA 12(6): 1074-1083), and insulin-like growth factor 2 IRES (Pedersen et al. (2002) Biochem. J.363(Pt l):37-44). [00657] These elements are commercially available in plasmids sold, e.g., by Clontech (Mountain View, CA), Invivogen (San Diego, CA), Addgene (Cambridge, MA) and GeneCopoeia (Rockville, MD). See also IRESite: The database of experimentally verified IRES structures (iresite.org). An IRES sequence may be included in a vector, for example, to express multiple bacteriophage recombination proteins for recombineering or an RNA-guided nuclease (e.g., Cas9) for HDR in combination with a retron reverse transcriptase from an expression cassette. [00658] In some embodiments, a polynucleotide encoding a viral self-cleaving 2A peptide, such as a T2A peptide, can be used to allow production of multiple protein products (e.g., Cas9, bacteriophage recombination proteins, retron reverse transcriptase) from a single vector or a single transcription unit under one promoter. One or more 2A linker peptides can be inserted between the coding sequences in the multicistronic construct. The 2A peptide, which is self- cleaving, allows co-expressed proteins from the multicistronic construct to be produced at equimolar levels.2A peptides from various viruses may be used, including, but not limited to 2A peptides derived from the foot-and-mouth disease virus, equine rhinitis A virus, Jhosea asigna virus and porcine teschovirus-1. See, e.g., Kim et al. (2011) PLoS One 6(4): el8556, Trichas et al. (2008) BMC Biol.6:40, Provost et al. (2007) Genesis 45(10): 625-629, Furler et al. (2001) Gene Ther.8(11):864-873; herein incorporated by reference in their entireties. [00659] In some embodiments, the expression construct comprises a plasmid suitable for transforming a bacterial host. Numerous bacterial expression vectors are known to those of skill in the art, and the selection of an appropriate vector is a matter of choice. Bacterial expression vectors include, but are not limited to, pACYC177, pASK75, pBAD, pBADM, pBAT, pCal, pET, pETM, pGAT, pGEX, pHAT, pKK223, pMal, pProEx, pQE, and pZA31 Bacterial plasmids may contain antibiotic selection markers (e.g., ampicillin, kanamycin, erythromycin, carbenicillin, streptomycin, or tetracycline resistance), a lacZ gene (b-galactosidase produces blue pigment from x-gal substrate), fluorescent markers (e.g., GFP. mCherry), or other markers for selection of transformed bacteria. See, e.g., Sambrook et al., supra. [00660] In other embodiments, the expression construct comprises a plasmid suitable for transforming a yeast cell. Yeast expression plasmids typically contain a yeast-specific origin of replication (ORI) and nutritional selection markers (e.g., HIS3, URA3, LYS2, LEU2, TRP1, METIS, ura4+, leul+, ade6+), antibiotic selection markers (e.g., kanamycin resistance), fluorescent markers (e.g., mCherry), or other markers for selection of transformed yeast cells. The yeast plasmid may further contain components to allow shuttling between a bacterial host (e.g., E coif) and yeast cells. A number of different types of yeast plasmids are available including yeast integrating plasmids (Yip), which lack an ORI and are integrated into host chromosomes by homologous recombination; yeast replicating plasmids (YRp), which contain an autonomously replicating sequence (ARS) and can replicate independently; yeast centromere plasmids (YCp), which are low copy vectors containing a part of an ARS and part of a centromere sequence (CEN); and yeast episomal plasmids (YEp), which are high copy number plasmids comprising a fragment from a 2 micron circle (a natural yeast plasmid) that allows for 50 or more copies to be stably propagated per cell. [00661] In other embodiments, the expression construct does not comprise a plasmid suitable for transforming a yeast cell. [00662] In other embodiments, the expression construct comprises a virus or engineered construct derived from a viral genome. A number of viral based systems have been developed for gene transfer into mammalian cells. These include adenoviruses, retroviruses (g-retroviruses and lentiviruses), poxviruses, adeno-associated viruses, baculoviruses, and herpes simplex viruses (see e.g., Wamock et al. (2011) Methods Mol. Biol.737:1-25; Walther et al. (2000) Drugs 60(2):249-271; and Lundstrom (2003) Trends Biotechnol.21(3): 117-122; herein incorporated by reference in their entireties). The ability of certain viruses to enter cells via receptor-mediated endocytosis, to integrate into host cell genomes and express viral genes stably and efficiently have made them attractive candidates for the transfer of foreign genes into mammalian cells. [00663] For example, retroviruses provide a convenient platform for gene delivery systems. Selected sequences can be inserted into a vector and packaged in retroviral particles using techniques known in the art. The recombinant virus can then be isolated and delivered to cells of the subject either in vivo or ex vivo. A number of retroviral systems have been described (U.S. Pat. No.5,219,740; Miller and Rosman (1989) BioTechniques 7:980-990; Miller, A. D. (1990) Human Gene Therapy 1:5-14; Scarpa et al. (1991) Virology 180:849-852; Bums et al. (1993) Proc. Natl. Acad. Sci. USA 90:8033-8037; Boris-Lawrie and Temin (1993) Cur. Opin. Genet. Develop.3:102-109; and Ferry et al. (2011) Curr. Pharm. Des.17(24): 2516-2527). Lentiviruses are a class of retroviruses that are particularly useful for delivering polynucleotides to mammalian cells because they are able to infect both dividing and nondividing cells (see e.g., Lois et al. (2002) Science 295:868-872; Durand et al. (2011) Viruses 3(2): 132-159; herein incorporated by reference). [00664] A number of adenoviral vectors have also been described. Unlike retroviruses which integrate into the host genome, adenoviruses persist extrachromosomally thus minimizing the risks associated with insertional mutagenesis. [00665] Additionally, various adeno-associated vims (AAV) vector systems have been developed for gene delivery. AAV vectors can be readily constructed using techniques well known in the art. See, e.g., U.S. Pat. Nos.5,173,414 and 5,139,941; International Publication Nos. WO 92/01070 (published 23 January 1992) and WO 93/03769 (published 4 March 1993); Lebkowski et al., Molec. Cell. Biol. (1988) 8:3988-3996; Vincent et al., Vaccines 90 (1990) (Cold Spring Harbor LaboratoryPress); Carter, B. J. Current Opinion in Biotechnology (1992) 3:533-539; Muzyczka, N. Current Topics in Microbiol and Immunol. (1992) 158:97-129; Kotin, R. M. Human Gene Therapy (1994) 5:793-801; Shelling and Smith, Gene Therapy (1994) 1:165- 169; and Zhou et al., J. Exp. Med. (1994) 179:1867-1875. [00666] Another vector system useful for delivering nucleic acids encoding the engineered retrons is the enterically administered recombinant poxvirus vaccines described by Small, Jr., P. A., et al. (U.S. Pat. No.5,676,950, issued Oct.14, 1997, herein incorporated by reference). [00667] Other viral vectors include those derived from the pox family of viruses, including vaccinia virus and avian poxvirus. By way of example, vaccinia virus recombinants expressing a nucleic acid molecule of interest (e.g., engineered retron) can be constructed as follows. The DNA encoding the particular nucleic acid sequence is first inserted into an appropriate vector so that it is adjacent to a vaccinia promoter and flanking vaccinia DNA sequences, such as the sequence encoding thymidine kinase (TK). This vector is then used to transfect cells which are simultaneously infected with vaccinia. Homologous recombination serves to insert the vaccinia promoter plus the gene encoding the sequences of interest into the viral genome. The resulting TK-recombinant can be selected by culturing the cells in the presence of 5- bromodeoxyuridine and picking viral plaques resistant thereto. [00668] In some embodiments, avipoxviruses, such as the fowlpox and canarypox viruses, can also be used to deliver the nucleic acid molecules of interest. The use of an avipox vector is particularly desirable in human and other mammalian species since members of the avipox genus can only productively replicate in susceptible avian species and therefore are not infective in mammalian cells. Methods for producing recombinant avipoxviruses are known in the art and employ genetic recombination, as described above with respect to the production of vaccinia viruses. See, e.g., WO 91/12882; WO 89/03429; and WO 92/03545. [00669] Molecular conjugate vectors, such as the adenovirus chimeric vectors described in Michael et al., J. Biol. Chem. (1993) 268:6866-6869 and Wagner et al., Proc. Natl. Acad. Sci. USA (1992) 89:6099-6103, can also be used for gene delivery. [00670] Members of the alphavirus genus, such as, but not limited to, vectors derived from the Sindbis virus (SIN), Semliki Forest virus (SFV), and Venezuelan Equine Encephalitis virus (VEE), will also find use as viral vectors for delivering the polynucleotides of the present invention. For a description of Sindbis-virus derived vectors useful for the practice of the instant methods, see, Dubensky et al. (1996) J. Virol.70:508-519; and International Publication Nos. WO 95/07995, WO 96/17072; as well as, Dubensky, Jr., T. W., et al., U.S. Pat. No.5,843,723, issued Dec.1, 1998, and Dubensky, Jr., T. W., U.S. Patent No.5,789,245, issued Aug.4, 1998, both herein incorporated by reference. Particularly preferred are chimeric alphavirus vectors comprised of sequences derived from Sindbis virus and Venezuelan equine encephalitis virus. See, e.g., Perri et al. (2003) J. Virol.77: 10394-10403 and International Publication Nos. WO 02/099035, WO 02/080982, WO 01/81609, and WO 00/61772; herein incorporated by reference in their entireties. [00671] A vaccinia-based infection/transfection system can be conveniently used to provide for inducible, transient expression of the nucleic acids of interest (e.g., engineered retron) in a host cell. In this system, cells are first infected in vitro with a vaccinia virus recombinant that encodes the bacteriophage T7 RNA polymerase. This polymerase displays exquisite specificity in that it only transcribes templates bearing T7 promoters. Following infection, cells are transfected with the nucleic acid of interest, driven by a T7 promoter. The polymerase expressed in the cytoplasm from the vaccinia virus recombinant transcribes the transfected DNA into RNA. The method provides for high level, transient, cytoplasmic production of large quantities of RNA. See, e.g., Elroy-Stein and Moss, Proc. Natl. Acad. Sci. USA (1990) 87:6743- 6747; Fuerst et al., Proc. Natl. Acad. Sci. USA (1986) 83:8122-8126. [00672] In other approaches to infection with vaccinia or avipox virus recombinants, or to the delivery of nucleic acids using other viral vectors, an amplification system can be used that will lead to high level expression following introduction into host cells. Specifically, a T7 RNA polymerase promoter preceding the coding region for T7 RNA polymerase can be engineered. Translation of RNA derived from this template will generate T7 RNA polymerase which in turn will transcribe more templates. Concomitantly, there will be a cDNA whose expression is under the control of the T7 promoter. Thus, some of the T7 RNA polymerase generated from translation of the amplification template RNA will lead to transcription of the desired gene. Because some T7 RNA polymerase is required to initiate the amplification, T7 RNA polymerase can be introduced into cells along with the template(s) to prime the transcription reaction. The polymerase can be introduced as a protein or on a plasmid encoding the RNA polymerase. For a further discussion of T7 systems and their use for transforming cells, see, e.g., International Publication No. WO 94/26911; Studier and Moffatt, J. Mol. Biol. (1986) 189:113-130; Deng and Wolff, Gene (1994) 143:245-249; Gao et al., Biochem. Biophys. Res. Commun. (1994) 200:1201-1206; Gao and Huang, Nuc. Acids Res. (1993) 21:2867-2872; Chen et al., Nuc. Acids Res. (1994) 22:2114-2120; and U.S. Pat. No.5,135,855. [00673] Insect cell expression systems, such as baculovirus systems, can also be used and are known to those of skill in the art and described in, e.g., Baculovirus and Insect Cell Expression Protocols (Methods in Molecular Biology, D.W. Murhammer ed., Humana Press, 2nd edition, 2007) and L. King The Baculovirus Expression System: A laboratory guide (Springer, 1992). Materials and methods for baculovirus/insect cell expression systems are commercially available in kit form from, inter alia, Thermo Fisher Scientific (Waltham, MA) and Clontech (Mountain View, CA). [00674] Plant expression systems can also be used for transforming plant cells. Generally, such systems use virus-based vectors to transfect plant cells with heterologous genes. For a description of such systems see, e.g., Porta et al., Mol. Biotech. (1996) 5:209-221; and Hackland et al., Arch. Virol. (1994) 139:1-22. [00675] To obtain expression of the engineered retron or the ncRNA encoded thereby, the expression construct or the ncRNA must be delivered into a cell. This delivery may be accomplished in vitro, as in laboratory procedures for transforming cells lines, or in vivo or ex vivo, as in the treatment of certain disease states. One mechanism for delivery is via viral infection where the expression construct is encapsulated in an infectious viral particle. [00676] Several non-viral methods for the transfer of expression constructs into cultured cells also are contemplated. These include the use of calcium phosphate precipitation, DEAE- dextran, electroporation, direct microinjection, DNA-loaded liposomes, lipofectamine-DNA complexes, cell sonication, gene bombardment using high velocity microprojectiles, and receptor-mediated transfection (see, e.g., Graham and Van Der Eb (1973) Virology 52:456-467; Chen and Okayama (1987) Mol. Cell Biol.7:2745-2752; Rippe et al. (1990) Mol. Cell Biol. 10:689-695; Gopal (1985) Mol. Cell Biol.5:1188-1190; Tur-Kaspa et al. (1986) Mol. Cell. Biol. 6:716-718; Potter et al. (1984) Proc. Natl. Acad. Sci. USA 81:7161-7165); Harland and Weintraub (1985) J. Cell Biol.101:1094-1099); Nicolau & Sene (1982) Biochim. Biophys. Acta 721:185-190; Fraley et al. (1979) Proc. Natl. Acad. Sci. USA 76:3348-3352; Fechheimer et al. (1987) Proc Natl. Acad. Sci. USA 84:8463-8467; Yang et al. (1990) Proc. Natl. Acad. Sci. USA 87:9568-9572; Wu and Wu (1987) J. Biol. Chem.262:4429-4432; Wu and Wu (1988) Biochemistry 27:887-892; herein incorporated by reference). Some of these techniques may be successfully adapted for in vivo or ex vivo use. [00677] Once the expression construct has been delivered into the cell the nucleic acid comprising the engineered retron sequence may be positioned and expressed at different sites. In some embodiments, the nucleic acid comprising the engineered retron sequence may be stably integrated into the genome of the cell. This integration may be in the cognate location and orientation via homologous recombination (gene replacement) or it may be integrated in a random, non-specific location (gene augmentation). In yet further embodiments, the nucleic acid may be stably maintained in the cell as a separate, episomal segment of DNA. Such nucleic acid segments or episomes encode sequences sufficient to permit maintenance and replication independent of or in synchronization with the host cell cycle. How the expression construct is delivered to a cell and where in the cell the nucleic acid remains is dependent on the type of expression construct employed. [00678] In some embodiments, the expression construct may simply consist of naked recombinant DNA or plasmids comprising the engineered retron. Transfer of the construct may be performed by any of the methods mentioned above which physically or chemically permeabilize the cell membrane. This is particularly applicable for transfer in vitro but it may be applied to in vivo use as well. Dubensky et al. (Proc. Natl. Acad. Sci. USA (1984) 81:7529-7533) successfully injected polyomavirus DNA in the form of calcium phosphate precipitates into liver and spleen of adult and newborn mice demonstrating active viral replication and acute infection. Benvenisty & Neshif (Proc. Natl. Acad. Sci. USA (1986) 83:9551-9555) also demonstrated that direct intraperitoneal injection of calcium phosphate- precipitated plasmids results in expression of the transfected genes. It is envisioned that DNA encoding an engineered retron of interest may also be transferred in a similar manner in vivo and express retron products. [00679] In still another embodiment, a naked DNA expression construct may be transferred into cells by particle bombardment. This method depends on the ability to accelerate DNA-coated microprojectiles to a high velocity allowing them to pierce cell membranes and enter cells without killing them (Klein et al. (1987) Nature 327:70-73). Several devices for accelerating small particles have been developed. One such device relies on a high voltage discharge to generate an electrical current, which in turn provides the motive force (Yang et al. (1990) Proc. Natl. Acad. Sci. USA 87:9568-9572). The microprojectiles may consist of biologically inert substances, such as tungsten or gold beads. [00680] In a further embodiment, the expression construct may be delivered using liposomes. Liposomes are vesicular structures characterized by a phospholipid bilayer membrane and an inner aqueous medium. Multilamellar liposomes have multiple lipid layers separated by aqueous medium. They form spontaneously when phospholipids are suspended in an excess of aqueous solution. The lipid components undergo self-rearrangement before the formation of closed structures and entrap water and dissolved solutes between the lipid bilayers (Ghosh & Bachhawat (1991) Liver Diseases, Targeted Diagnosis and Therapy Using Specific Receptors and Ligands, Wu et al. (Eds.), Marcel Dekker, NY, 87-104). Also contemplated is the use of lipofectamine-DNA complexes. [00681] In some embodiments, the liposome may be complexed with a hemagglutinating virus (HVJ). This has been shown to facilitate fusion with the cell membrane and promote cell entry of liposome-encapsulated DNA (Kaneda et al. (1989) Science 243:375-378). In other embodiments, the liposome may be complexed or employed in conjunction with nuclear non- histone chromosomal proteins (HMG-I) (Kato et al. (1991) J. Biol. Chem.266(6):3361-3364). [00682] In yet further embodiments, the liposome may be complexed or employed in conjunction with both HVJ and HMG-I. Where a bacterial promoter is employed in the DNA construct, it also will be desirable to include within the liposome an appropriate bacterial polymerase. [00683] Other expression constructs which can be employed to deliver a nucleic acid into cells are receptor-mediated delivery vehicles. These take advantage of the selective uptake of macromolecules by receptor-mediated endocytosis in almost all eukaryotic cells. Because of the cell type-specific distribution of various receptors, the delivery can be highly specific (Wu and Wu (1993) Adv. Drug Delivery Rev.12:159- 167). Receptor-mediated gene targeting vehicles generally consist of two components: a cell receptor-specific ligand and a DNA-binding agent. Several ligands have been used for receptor-mediated gene transfer. The most extensively characterized ligands are asialoorosomucoid (ASOR) and transferrin (see, e.g., Wu and Wu (1987), supra; Wagner et al. (1990) Proc. Natl. Acad. Sci. USA 87(9):3410- 3414). A synthetic neoglycoprotein, which recognizes the same receptor as ASOR, has been used as a gene delivery vehicle (Ferkol et al. (1993) FASEB J.7:1081-1091; Perales et al. (1994) Proc. Natl. Acad. Sci. USA 91(9):4086-4090), and epidermal growth factor (EGF) has also been used to deliver genes to squamous carcinoma cells (Myers, EPO 0273085). [00684] In other embodiments, the delivery vehicle may comprise a ligand and a liposome. For example, Nicolau et al. (Methods Enzymol. (1987) 149:157-176) employed lactosy 1-ceramide, a galactose-terminal asialoganglioside, incorporated into liposomes and observed an increase in the uptake of the insulin gene by hepatocytes. Thus, it is feasible that a nucleic acid encoding a particular gene also may be specifically delivered into a cell by any number of receptor-ligand systems with or without liposomes. Also, antibodies to surface antigens on cells can similarly be used as targeting moieties. [00685] In some embodiments, the promoters that may be used in the retron delivery systems described herein may be constitutive, inducible, or tissue-specific. In some embodiments, the promoters may be a constitutive promoters. Non-limiting exemplary constitutive promoters include cytomegalovirus immediate early promoter (CMV), simian virus (SV40) promoter, adenovirus major late (MLP) promoter, Rous sarcoma virus (RSV) promoter, mouse mammary tumor virus (MMTV) promoter, phosphoglycerate kinase (PGK) promoter, elongation factor-alpha (EFla) promoter, ubiquitin promoters, actin promoters, tubulin promoters, immunoglobulin promoters, a functional fragment thereof, or a combination of any of the foregoing. In some embodiments, the promoter may be a CMV promoter. In some embodiments, the promoter may be a truncated CMV promoter. In other embodiments, the promoter may be an EFla promoter. In some embodiments, the promoter may be an inducible promoter. Non-limiting exemplary inducible promoters include those inducible by heat shock, light, chemicals, peptides, metals, steroids, antibiotics, or alcohol. In some embodiments, the inducible promoter may be one that has a low basal (non-induced) expression level, such as, e.g., the Tet-On® promoter (Clontech). In some embodiments, the promoter may be a tissue-specific promoter. In some embodiments, the tissue-specific promoter is exclusively or predominantly expressed in liver tissue. Non-limiting exemplary tissue-specific promoters include B29 promoter, CD14 promoter, CD43 promoter, CD45 promoter, CD68 promoter, desmin promoter, elastase- 1 promoter, endoglin promoter, fibronectin promoter, Flt-1 promoter, GFAP promoter, GPIIb promoter, ICAM- 2 promoter, INF-b promoter, Mb promoter, Nphsl promoter, OG-2 promoter, SP-B promoter, SYN1 promoter, and WASP promoter. J. Delivery Systems and Methods of Delivery [00686] The engineered retrons can be delivered by any known delivery system such as those described above. Non-limiting examples of delivery vehicles include lipid particles (e.g. Lipid nanoparticles (LNPs)), non-lipid nanoparticles, exosomes, liposomes, micelles, viral particles, Stable nucleic-acid-lipid particles (SNALPs), lipoplexes/polyplexes, DNA nanoclews, Gold nanoparticles, iTOP, Streptolysin O (SLO), multifunctional envelope-type nanodevice (MEND), lipid-coated mesoporous silica particles, inorganic nanoparticles, and polymeric delivery technology (e.g., polymer-based particles). All-RNA delivery system and RNA ratios [00687] In various embodiments, the retron editing systems disclosed herein may be delivered in an “all-RNA” format. As used herein, the term “all-RNA” format refers to the fact that each of the components of a retron editing system (e.g., the retron RT, the programmable nuclease, the sgRNA, and the ncRNA) are delivered and/or administered as RNA. In some embodiments, the RNA components may be delivered to cells and/or tissues by direct means, such as electroporation or transfection. In other embodiments, the RNA components may be delivered to cells and/or tissues by way of a delivery vehicle, such as an LNP or liposome. [00688] In various embodiments, the retron editing systems described herein may comprise a coding RNA (e.g., linear or circular mRNA) that encodes a retron reverse transcriptase (e.g., any RT from Table X or Table A), a coding RNA (e.g., linear or circular mRNA) that encodes a programmable nuclease, a retron ncRNA (e.g., a ncRNA from Table B), and a guide RNA. [00689] In some embodiments, RT and nuclease components may be encoded on the same coding RNA molecule. The proteins may also be expressed from separate coding RNA molecules. In still other embodiments, the RT and the nuclease components can be fused together as a singular fusion polypeptide having an RT domain and a nuclease domain optionally joined by a linker. [00690] In addition, in some embodiments, the ncRNA and the guide RNA may be fused together as a single RNA molecule. For example, the guide RNA may be located at the 5’ end of the ncRNA. In other embodiments, the guide RNA may be located at the 3’ end of the ncRNA. In some embodiments, the ncRNA may comprise a guide RNA at both the 3’ and the 5’ ends of the ncRNA. [00691] In still other embodiments, the ncRNA and the guide RNA may be separate molecules, i.e., delivered separately. [00692] In still other embodiments, the retron editing system may include both a ncRNA- guide RNA fusion and an additional guide RNA provided as a separate molecule. [00693] In various embodiments, the different RNA components of the all-RNA retron editing system can be combined and administered (e.g., directly or within a delivery vehicle) in different ratios. In some embodiments, the ratios of such RNA components or species can be expressed as molar ratios. [00694] For example, the molar ratio of RT coding RNA to nuclease coding RNA can be about 1:1, about 1:1.5, about 1:2, about 1:2.5, about 1:3, about 1:4, about 1:5, about 1:6, about 1:7, about 1:8, about 1:9, about 1:10, about 1:12, about 1:15, about 1:20. Useful ranges include from 1:1 to 1:2, from 1:1.5 to 1:4, from 1:2 to 1:4, from 1:2 to 1:8, from 1:2 to 1:10, from 1:3 to 1:9, from 1:3 to 1:12, from 1:3 to 1:15, from 1:4 to 1:8, from 1:4 to 1:12, from 1:4 to 1:20, from 1:5 to 1:10, from 1:5 to 1:15, from 1:5 to 1:20, from 1:10 to 1:20, or from 1:10 to 1:40. [00695] In another example, the molar ratio of nuclease coding RNA to RT coding RNA can be about 1:1, about 1:1.5, about 1:2, about 1:2.5, about 1:3, about 1:4, about 1:5, about 1:6, about 1:7, about 1:8, about 1:9, about 1:10, about 1:12, about 1:15, about 1:20. Useful ranges include from 1:1 to 1:2, from 1:1.5 to 1:4, from 1:2 to 1:4, from 1:2 to 1:8, from 1:2 to 1:10, from 1:3 to 1:9, from 1:3 to 1:12, from 1:3 to 1:15, from 1:4 to 1:8, from 1:4 to 1:12, from 1:4 to 1:20, from 1:5 to 1:10, from 1:5 to 1:15, from 1:5 to 1:20, from 1:10 to 1:20, or from 1:10 to 1:40. [00696] In still another example, the molar ratio of ncRNA or ncRNA-guide RNA fusion to separate guide RNA can be about 1:1, about 1:1.5, about 1:2, about 1:2.5, about 1:3, about 1:4, about 1:5, about 1:6, about 1:7, about 1:8, about 1:9, about 1:10, about 1:12, about 1:15, about 1:20. Useful ranges include from 1:1 to 1:2, from 1:1.5 to 1:4, from 1:2 to 1:4, from 1:2 to 1:8, from 1:2 to 1:10, from 1:3 to 1:9, from 1:3 to 1:12, from 1:3 to 1:15, from 1:4 to 1:8, from 1:4 to 1:12, from 1:4 to 1:20, from 1:5 to 1:10, from 1:5 to 1:15, from 1:5 to 1:20, from 1:10 to 1:20, or from 1:10 to 1:40. [00697] In still another example, the molar ratio of separate guide RNA to ncRNA or ncRNA-guide RNA fusion can be about 1:1, about 1:1.5, about 1:2, about 1:2.5, about 1:3, about 1:4, about 1:5, about 1:6, about 1:7, about 1:8, about 1:9, about 1:10, about 1:12, about 1:15, about 1:20. Useful ranges include from 1:1 to 1:2, from 1:1.5 to 1:4, from 1:2 to 1:4, from 1:2 to 1:8, from 1:2 to 1:10, from 1:3 to 1:9, from 1:3 to 1:12, from 1:3 to 1:15, from 1:4 to 1:8, from 1:4 to 1:12, from 1:4 to 1:20, from 1:5 to 1:10, from 1:5 to 1:15, from 1:5 to 1:20, from 1:10 to 1:20, or from 1:10 to 1:40. [00698] In still another example, the molar ratio of ncRNA to separate guide RNA can be about 1:1, about 1:1.5, about 1:2, about 1:2.5, about 1:3, about 1:4, about 1:5, about 1:6, about 1:7, about 1:8, about 1:9, about 1:10, about 1:12, about 1:15, about 1:20. Useful ranges include from 1:1 to 1:2, from 1:1.5 to 1:4, from 1:2 to 1:4, from 1:2 to 1:8, from 1:2 to 1:10, from 1:3 to 1:9, from 1:3 to 1:12, from 1:3 to 1:15, from 1:4 to 1:8, from 1:4 to 1:12, from 1:4 to 1:20, from 1:5 to 1:10, from 1:5 to 1:15, from 1:5 to 1:20, from 1:10 to 1:20, or from 1:10 to 1:40. [00699] In still another example, the molar ratio of separate guide RNA to ncRNA can be about 1:1, about 1:1.5, about 1:2, about 1:2.5, about 1:3, about 1:4, about 1:5, about 1:6, about 1:7, about 1:8, about 1:9, about 1:10, about 1:12, about 1:15, about 1:20. Useful ranges include from 1:1 to 1:2, from 1:1.5 to 1:4, from 1:2 to 1:4, from 1:2 to 1:8, from 1:2 to 1:10, from 1:3 to 1:9, from 1:3 to 1:12, from 1:3 to 1:15, from 1:4 to 1:8, from 1:4 to 1:12, from 1:4 to 1:20, from 1:5 to 1:10, from 1:5 to 1:15, from 1:5 to 1:20, from 1:10 to 1:20, or from 1:10 to 1:40. [00700] In still another example, the molar ratio of a coding RNA (e.g., encoding RT and/or nuclease) to ncRNA or ncRNA-guide RNA fusion, as the case may be, can be about 1:1, about 1:1.5, about 1:2, about 1:2.5, about 1:3, about 1:4, about 1:5, about 1:6, about 1:7, about 1:8, about 1:9, about 1:10, about 1:12, about 1:15, about 1:20. Useful ranges include from 1:1 to 1:2, from 1:1.5 to 1:4, from 1:2 to 1:4, from 1:2 to 1:8, from 1:2 to 1:10, from 1:3 to 1:9, from 1:3 to 1:12, from 1:3 to 1:15, from 1:4 to 1:8, from 1:4 to 1:12, from 1:4 to 1:20, from 1:5 to 1:10, from 1:5 to 1:15, from 1:5 to 1:20, from 1:10 to 1:20, or from 1:10 to 1:40. [00701] In still another example, the molar ratio of a coding RNA encoding a retron RT to ncRNA or ncRNA-guide RNA fusion, as the case may be, can be about 1:1, about 1:1.5, about 1:2, about 1:2.5, about 1:3, about 1:4, about 1:5, about 1:6, about 1:7, about 1:8, about 1:9, about 1:10, about 1:12, about 1:15, about 1:20. Useful ranges include from 1:1 to 1:2, from 1:1.5 to 1:4, from 1:2 to 1:4, from 1:2 to 1:8, from 1:2 to 1:10, from 1:3 to 1:9, from 1:3 to 1:12, from 1:3 to 1:15, from 1:4 to 1:8, from 1:4 to 1:12, from 1:4 to 1:20, from 1:5 to 1:10, from 1:5 to 1:15, from 1:5 to 1:20, from 1:10 to 1:20, or from 1:10 to 1:40. [00702] In still another example, the molar ratio of a coding RNA encoding a programmable nuclease to ncRNA or ncRNA-guide RNA fusion, as the case may be, can be about 1:1, about 1:1.5, about 1:2, about 1:2.5, about 1:3, about 1:4, about 1:5, about 1:6, about 1:7, about 1:8, about 1:9, about 1:10, about 1:12, about 1:15, about 1:20. Useful ranges include from 1:1 to 1:2, from 1:1.5 to 1:4, from 1:2 to 1:4, from 1:2 to 1:8, from 1:2 to 1:10, from 1:3 to 1:9, from 1:3 to 1:12, from 1:3 to 1:15, from 1:4 to 1:8, from 1:4 to 1:12, from 1:4 to 1:20, from 1:5 to 1:10, from 1:5 to 1:15, from 1:5 to 1:20, from 1:10 to 1:20, or from 1:10 to 1:40. [00703] In still another example, the molar ratio of a coding RNA encoding a retron RT or a nuclease to a separate guide RNA can be about 1:1, about 1:1.5, about 1:2, about 1:2.5, about 1:3, about 1:4, about 1:5, about 1:6, about 1:7, about 1:8, about 1:9, about 1:10, about 1:12, about 1:15, about 1:20. Useful ranges include from 1:1 to 1:2, from 1:1.5 to 1:4, from 1:2 to 1:4, from 1:2 to 1:8, from 1:2 to 1:10, from 1:3 to 1:9, from 1:3 to 1:12, from 1:3 to 1:15, from 1:4 to 1:8, from 1:4 to 1:12, from 1:4 to 1:20, from 1:5 to 1:10, from 1:5 to 1:15, from 1:5 to 1:20, from 1:10 to 1:20, or from 1:10 to 1:40. [00704] In still another example, the molar ratio of a separate guide RNA to a coding RNA encoding a retron RT or a nuclease can be about 1:1, about 1:1.5, about 1:2, about 1:2.5, about 1:3, about 1:4, about 1:5, about 1:6, about 1:7, about 1:8, about 1:9, about 1:10, about 1:12, about 1:15, about 1:20. Useful ranges include from 1:1 to 1:2, from 1:1.5 to 1:4, from 1:2 to 1:4, from 1:2 to 1:8, from 1:2 to 1:10, from 1:3 to 1:9, from 1:3 to 1:12, from 1:3 to 1:15, from 1:4 to 1:8, from 1:4 to 1:12, from 1:4 to 1:20, from 1:5 to 1:10, from 1:5 to 1:15, from 1:5 to 1:20, from 1:10 to 1:20, or from 1:10 to 1:40. [00705] In certain embodiments, the amount of ncRNA-sgRNA relative to RT mRNA is augmented. In certain embodiments the RT mRNA: ncRNA-sgRNA ratio is about 1:1.5, about 1:2, about 1:2.5, about 1:3, about 1:4, about 1:5, about 1:6, about 1:7, about 1:8, about 1:9, about 1:10, about 1:12, about 1:15, about 1:20. Useful ranges include from 1:1 to 1:2, from 1:1.5 to 1:4, from 1:2 to 1:4, from 1:2 to 1:8, from 1:2 to 1:10, from 1:3 to 1:9, from 1:3 to 1:12, from 1:3 to 1:15, from 1:4 to 1:8, from 1:4 to 1:12, from 1:4 to 1:20, from 1:5 to 1:10, from 1:5 to 1:15, from 1:5 to 1:20, from 1:10 to 1:20, or from 1:10 to 1:40. In certain embodiments, an RT-Cas9 (or Cas9-RT) fusion is encoded by an mRNA. In certain embodiments, the RT-Cas9 mRNA: ncRNA-sgRNA ratio is about 1:1.5, about 1:2, about 1:2.5, about 1:3, about 1:4, about 1:5, about 1:6, about 1:7, about 1:8, about 1:9, about 1:10, about 1:12, about 1:15, about 1:20. Useful ranges include from 1:1 to 1:2, from 1:1.5 to 1:4, from 1:2 to 1:4, from 1:2 to 1:8, from 1:2 to 1:10, from 1:3 to 1:9, from 1:3 to 1:12, from 1:3 to 1:15, from 1:4 to 1:8, from 1:4 to 1:12, from 1:4 to 1:20, from 1:5 to 1:10, from 1:5 to 1:15, from 1:5 to 1:20, from 1:10 to 1:20, or from 1:10 to 1:40. In certain embodiments, multiple genetic loci are targeted hence the ncRNA-sgRNA includes a mixture of ncRNA-sgRNA species and the same ratios and ranges are applicable. Format of ncRNA and guide RNA [00706] In certain embodiments, the ncRNA and the guide RNA can be delivered as a single molecule, i.e., with the guide RNA fused to the 5’ and/or 3’ end of the ncRNA. The ncRNA may have guide RNAs located at both ends in some embodiments. [00707] In other embodiments, the guide RNA and ncRNA may be provided and/or delivered as separate components. As shown in Example 4, separation of the guide RNA from the ncRNA can result in increased editing efficiency. [00708] In still other embodiments, a ncRNA-gRNA fusion may be co-delivered with a separate guide RNA. Modified ncRNAs [00709] In still other embodiments, the ncRNAs disclosed herein may be modified by introducing additional RNA motifs into the ncRNAs, e.g., at the 5ʹ and 3ʹ termini of the ncRNAs, or even at positions therein between (e.g., in the msr or msd regions) to improve transcriptional production and/or stability and/or function (e.g., RT-DNA production). Such structures may include, but are not limited to RNA hairpins, RNA step-loops, RNA quadruplexes, cap structures, and poly(A) tails, or ribozyme functions and the like. Also, ncRNAs could also be modified to include one or more nuclear localization sequences. [00710] Additional RNA motifs could also improve RT processivity of the ncRNA or enhance ncRNA activity by enhancing RT binding. Addition of dimerization motifs - such as kissing loops or a GNRA tetraloop/tetraloop receptor pair - at the 5ʹ and 3ʹ termini of the ncRNA could also result in effective circularization of the ncRNA, improving stability. Additionally, it is envisioned that addition of these motifs could enable the physical separation of ncRNA components, e.g., separation of the msr and msd regions. Short 5ʹ extensions or 3’ extensions to the ncRNA that form a small toehold hairpin at either or both ends of the ncRNA could also compete favorably against the annealing of intracomplementary regions along the length of the ncRNA. Finally, kissing loops could also be used to recruit other RNAs or proteins to the genomic site and enable swapping of RT activity from one RNA to the other. [00711] ncRNAs could be further improved via directed evolution, in an analogous fashion to how protein function can be improved. Directed evolution could enhance ncRNA recognition by RT and/or reduce off-site targeting and/or indels and/or improve precise editing efficiency. [00712] The present disclosure contemplates any such ways to further improve the stability and/or functionality of the ncRNAs disclosed here. [00713] In some embodiments, the RNAs (including the guide RNAs and the ncRNAs) used in the compositions of the disclosure have undergone a chemical or biological modification to render them more stable. Exemplary modifications to an RNA include the depletion of a base (e.g., by deletion or by the substitution of one nucleotide for another) or modification of a base, for example, the chemical modification of a base. The phrase "chemical modifications" as used herein, includes modifications which introduce chemistries which differ from those seen in naturally occurring RNA, for example, covalent modifications such as the introduction of modified nucleotides, (e.g., nucleotide analogs, or the inclusion of pendant groups which are not naturally found in such mRNA molecules). [00714] Other suitable polynucleotide modifications that may be incorporated into the RNAs used in the compositions of the disclosure include, but are not limited to, 4'- thio-modified bases: 4'-thio-adenosine, 4'-thio-guanosine, 4'-thio-cytidine, 4'-thio-uridine, 4'- thio-5-methyl- cytidine, 4'-thio-pseudouridine, and 4'-thio-2-thiouridine, pyridin-4-one ribonucleoside, 5-aza- uridine, 2-thio-5-aza-uridine, 2-thiouridine, 4-thio-pseudouridine, 2- thio-pseudouridine, 5- hydroxyuridine, 3-methyluridine, 5-carboxymethyl-uridine, 1- carboxymethyl-pseudouridine, 5- propynyl-uridine, 1-propynyl-pseudouridine, 5- taurinomethyluridine, 1-taurinomethyl- pseudouridine, 5-taurinomethyl-2-thio-uridine, 1- taurinomethyl-4-thio-uridine, 5-methyl- uridine, 1-methyl-pseudouridine, 4-thio-1-methyl- pseudouridine, 2-thio-1-methyl- pseudouridine, 1-methyl-1-deaza-pseudouridine, 2-thio-1- methyl-1-deaza-pseudouridine, dihydrouridine, dihydropseudouridine, 2-thio-dihydrouridine, 2-thio-dihydropseudouridine, 2- methoxyuridine, 2-methoxy-4-thio-uridine, 4-methoxy- pseudouridine, 4-methoxy-2-thio- pseudouridine, 5-aza-cytidine, pseudoisocytidine, 3-methyl- cytidine, N4-acetylcytidine, 5- formylcytidine, N4-methylcytidine, 5-hydroxymethylcytidine, 1-methyl-pseudoisocytidine, pyrrolo-cytidine, pyrrolo-pseudoisocytidine, 2-thio-cytidine, 2- thio-5-methyl-cytidine, 4-thio- pseudoisocytidine, 4-thio-1-methyl-pseudoisocytidine, 4-thio- 1-methyl-1-deaza- pseudoisocytidine, 1-methyl-1-deaza-pseudoisocytidine, zebularine, 5-aza- zebularine, 5-methyl- zebularine, 5-aza-2-thio-zebularine, 2-thio-zebularine, 2-methoxy- cytidine, 2-methoxy-5- methyl-cytidine, 4-methoxy-pseudoisocytidine, 4-methoxy-1-methyl- pseudoisocytidine, 2- aminopurine, 2,6-diaminopurine, 7-deaza-adenine, 7-deaza-8-aza- adenine, 7-deaza-2- aminopurine, 7-deaza-8-aza-2-aminopurine, 7-deaza-2,6-diaminopurine, 7-deaza-8-aza-2,6- diaminopurine, 1-methyladenosine, N6-methyladenosine, N6- isopentenyladenosine, N6-(cis- hydroxyisopentenyl)adenosine, 2-methylthio-N6-(cis- hydroxyisopentenyl)adenosine, N6- glycinylcarbamoyladenosine, N6- threonylcarbamoyladenosine, 2-methylthio-N6-threonyl carbamoyladenosine, N6,N6- dimethyladenosine, 7-methyladenine, 2-methylthio-adenine, and 2- methoxy-adenine, inosine, 1-methyl-inosine, wyosine, wybutosine, 7-deaza-guanosine, 7-deaza- 8-aza-guanosine, 6-thio- guanosine, 6-thio-7-deaza-guanosine, 6-thio-7-deaza-8-aza-guanosine, 7-methyl-guanosine, 6-thio-7-methyl-guanosine, 7-methylinosine, 6-methoxy-guanosine, 1- methylguanosine, N2- methylguanosine, N2,N2-dimethylguanosine, 8-oxo-guanosine, 7-methyl- 8-oxo-guanosine, 1-methyl-6-thio-guanosine, N2-methyl-6-thio-guanosine, and N2,N2-dimethyl- 6-thio- guanosine, and combinations thereof. The term modification also includes, for example, the incorporation of non-nucleotide linkages or modified nucleotides into the mRNA sequences of the present invention (e.g., modifications to one or both of the 3' and 5' ends of an mRNA molecule encoding a functional protein or enzyme). Such modifications include the addition of bases to an mRNA sequence (e.g., the inclusion of a poly A tail or a longer poly A tail), the alteration of the 3' UTR or the 5' UTR, complexing the mRNA with an agent (e.g., a protein or a complementary nucleic acid molecule), and inclusion of elements which change the structure of an RNA molecule (e.g., which form secondary structures). [00715] In some embodiments, RNAs (e.g., ncRNAs) include a 5' cap structure. A 5' cap is typically added as follows: first, an RNA terminal phosphatase removes one of the terminal phosphate groups from the 5' nucleotide, leaving two terminal phosphates; guanosine triphosphate (GTP) is then added to the terminal phosphates via a guanylyl transferase, producing a 5'5'5 triphosphate linkage; and the 7-nitrogen of guanine is then methylated by a methyltransferase. Examples of cap structures include, but are not limited to, m7G(5')ppp (5'(A,G(5')ppp(5')A and G(5')ppp(5')G. Naturally occurring cap structures comprise a 7- methyl guanosine that is linked via a triphosphate bridge to the 5'-end of the first transcribed nucleotide, resulting in a dinucleotide cap of m7G(5')ppp(5')N, where N is any nucleoside. In vivo, the cap is added enzymatically. The cap is added in the nucleus and is catalyzed by the enzyme guanylyl transferase. The addition of the cap to the 5' terminal end of RNA occurs immediately after initiation of transcription. The terminal nucleoside is typically a guanosine, and is in the reverse orientation to all the other nucleotides, i.e., G(5')ppp(5')GpNpNp. [00716] Additional cap analogs include, but are not limited to, a chemical structures selected from the group consisting of m7GpppG, m7GpppA, m7GpppC; unmethylated cap analogs (e.g., GpppG); dimethylated cap analog (e.g., m2,7GpppG), trimethylated cap analog (e.g., m2,2,7GpppG), dimethylated symmetrical cap analogs (e.g., m7Gpppm7G), or anti reverse cap analogs (e.g., ARCA; m7,2'OmeGpppG, m72'dGpppG, m7,3'OmeGpppG, m7,3'dGpppG and their tetraphosphate derivatives) (see, e.g., Jemielity, J. et al., "Novel 'anti-reverse' cap analogs with superior translational properties", RNA, 9: 1108-1122 (2003)). [00717] Typically, the presence of a "tail" serves to protect the RNA (e.g., ncRNA) from exonuclease degradation. A poly A or poly U tail is thought to stabilize natural messengers and synthetic sense RNA. Therefore, in certain embodiments a long poly A or poly U tail can be added to an RNA molecule thus rendering the RNA more stable. Poly A or poly U tails can be added using a variety of art-recognized techniques. For example, long poly A tails can be added to synthetic or in vitro transcribed RNA using poly A polymerase (Yokoe, et al. Nature Biotechnology.1996; 14: 1252-1256). A transcription vector can also encode long poly A tails. In addition, poly A tails can be added by transcription directly from PCR products. Poly A may also be ligated to the 3' end of a sense RNA with RNA ligase (see, e.g., Molecular Cloning A Laboratory Manual, 2nd Ed., ed. by Sambrook, Fritsch and Maniatis (Cold Spring Harbor Laboratory Press: 1991 edition)). [00718] Typically, the length of a poly A or poly U tail can be at least about 10, 50, 100, 200, 300, 400 at least 500 nucleotides. In some embodiments, a poly-A tail on the 3' terminus of mRNA typically includes about 10 to 300 adenosine nucleotides (e.g., about 10 to 200 adenosine nucleotides, about 10 to 150 adenosine nucleotides, about 10 to 100 adenosine nucleotides, about 20 to 70 adenosine nucleotides, or about 20 to 60 adenosine nucleotides). In some embodiments, mRNAs include a 3' poly(C) tail structure. A suitable poly-C tail on the 3' terminus of mRNA typically include about 10 to 200 cytosine nucleotides (e.g., about 10 to 150 cytosine nucleotides, about 10 to 100 cytosine nucleotides, about 20 to 70 cytosine nucleotides, about 20 to 60 cytosine nucleotides, or about 10 to 40 cytosine nucleotides). The poly-C tail may be added to the poly-A or poly U tail or may substitute the poly-A or poly U tail. [00719] RNAs according to the present disclosure (e.g., ncRNAs) may be synthesized according to any of a variety of known methods. For example, RNAs according to the present invention may be synthesized via in vitro transcription (IVT). Briefly, IVT is typically performed with a linear or circular DNA template containing a promoter, a pool of ribonucleotide triphosphates, a buffer system that may include DTT and magnesium ions, and an appropriate RNA polymerase (e.g., T3, T7 or SP6 RNA polymerase), DNAse I, pyrophosphatase, and/or RNAse inhibitor. The exact conditions will vary according to the specific application. An improved method of IVT of a ncRNA is disclosed in Example 5 herein. [00720] In a particular embodiment (as exemplified in Example 6 herein), the ncRNAs can comprise an MS2 modification, as specific RNA hairpin structure recognized in nature by a certain MS2-binding protein. This domain can help to stabilize the ncRNA and improve the editing efficiency. The disclosure contemplates other similar modifications. A review of other such MS2-like domains are described in the art, for example, in Johansson et al.,“RNA recognition by the MS2 phage coat protein,” Sem Virol., 1997, Vol.8(3): 176-185; Delebecque et al.,“Organization of intracellular reactions with rationally designed RNA assemblies,” Science, 2011, Vol.333: 470-474; Mali et al.,“Cas9 transcriptional activators for target specificity screening and paired nickases for cooperative genome engineering,” Nat. Biotechnol., 2013, Vol.31: 833- 838; and Zalatan et al.,“Engineering complex synthetic transcriptional programs with CRISPR RNA scaffolds,” Cell, 2015, Vol.160: 339-350, each of which are incorporated herein by reference in their entireties. Other systems include the PP7 hairpin, which specifically recruits the PCP protein, and the “com” hairpin, which specifically recruits the Com protein. See Zalatan et al. The nucleotide sequence of the MS2 hairpin (or equivalently referred to as the“MS2 aptamer”) is: GCCAACATGAGGATCACCCATGTCTGCAGGGCC. Lipid nanoparticles [00721] In some embodiments, the lipid delivery sytem includes lipid nanoparticles (LNP). In some embodiments the LNP are small solid or semi-solid particles possessing an exterior lipid layer with a hydrophilic exterior surface that is exposed to the non-LNP environment, an interior space which may aqueous (vesicle like) or non-aqueous (micelle like), and at least one hydrophobic inter-membrane space. LNP membranes may be lamellar or non- lamellar and may be comprised of 1, 2, 3, 4, 5 or more layers. In some embodiments, LNPs may comprise a nucleic acid (e.g. engineered retron) into their interior space, into the inter membrane space, onto their exterior surface, or any combination thereof. [00722] In some embodiments, an LNP of the present disclosure comprises an ionizable lipid, a structural lipid, a PEGylated lipid (aka PEG lipid), and a phospholipid. In alternative embodiments, an LNP comprises an ionizable lipid, a structural lipid, a PEGylated lipid (aka PEG lipid), and a zwitterionic amino acid lipid. In some embodiments, an LNP further comprises a 5th lipid, besides any of the aforementioned lipid components. In some embodiments, the LNP encapsulates one or more elements of the active agent of the present disclosure. In some embodiments, an LNP further comprises a targeting moiety covalently or non-covalently bound to the outer surface of the LNP. In some embodiments, the targeting moiety is a targeting moiety that binds to, or otherwise facilitates uptake by, cells of a particular organ system. [00723] In some embodiments, an LNP has a diameter of at least about 20nm, 30 nm, 40nm, 50nm, 60nm, 70nm, 80nm, or 90nm. In some embodiments, an LNP has a diameter of less than about 100nm, 110nm, 120nm, 130nm, 140nm, 150nm, or 160nm. In some embodiments, an LNP has a diameter of less than about 100nm. In some embodiments, an LNP has a diameter of less than about 90nm. In some embodiments, an LNP has a diameter of less than about 80nm. In some embodiments, an LNP has a diameter of about 60-100nm. In some embodiments, an LNP has a diameter of about 75-80nm. [00724] In some embodiments, the lipid nanoparticle compositions of the present disclosure are described according to the respective molar ratios of the component lipids in the formulation. As a non-limiting example, the mol-% of the ionizable lipid may be from about 10 mol-% to about 80 mol-%. As a non-limiting example, the mol-% of the ionizable lipid may be from about 20 mol-% to about 70 mol-%. As a non-limiting example, the mol-% of the ionizable lipid may be from about 30 mol-% to about 60 mol-%. As a non-limiting example, the mol-% of the ionizable lipid may be from about 35 mol-% to about 55 mol-%. As a non-limiting example, the mol-% of the ionizable lipid may be from about 40 mol-% to about 50 mol-%. [00725] In some embodiments, the mol-% of the phospholipid may be from about 1 mol- % to about 50 mol-%. In some embodiments, the mol-% of the phospholipid may be from about 2 mol-% to about 45 mol-%. In some embodiments, the mol-% of the phospholipid may be from about 3 mol-% to about 40 mol-%. In some embodiments, the mol-% of the phospholipid may be from about 4 mol-% to about 35 mol-%. In some embodiments, the mol-% of the phospholipid may be from about 5 mol-% to about 30 mol-%. In some embodiments, the mol-% of the phospholipid may be from about 10 mol-% to about 20 mol-%. In some embodiments, the mol- % of the phospholipid may be from about 5 mol-% to about 20 mol-%. [00726] In some embodiments, the mol-% of the structural lipid may be from about 10 mol-% to about 80 mol-%. In some embodiments, the mol-% of the structural lipid may be from about 20 mol-% to about 70 mol-%. In some embodiments, the mol-% of the structural lipid may be from about 30 mol-% to about 60 mol-%. In some embodiments, the mol-% of the structural lipid may be from about 35 mol-% to about 55 mol-%. In some embodiments, the mol-% of the structural lipid may be from about 40 mol-% to about 50 mol-%. [00727] In some embodiments, the mol-% of the PEG lipid may be from about 0.1 mol-% to about 10 mol-%. In some embodiments, the mol-% of the PEG lipid may be from about 0.2 mol-% to about 5 mol-%. In some embodiments, the mol-% of the PEG lipid may be from about 0.5 mol-% to about 3 mol-%. In some embodiments, the mol-% of the PEG lipid may be from about 1 mol-% to about 2 mol-%. In some embodiments, the mol-% of the PEG lipid may be about 1.5 mol-%. i. Ionizable lipids [00728] In some embodiments, an LNP disclosed herein comprises an ionizable lipid. In some embodiments, an LNP comprises two or more ionizable lipids. [00729] In some embodiments, an ionizable lipid has a dimethylamine or an ethanolamine head. In some embodiments, an ionizable lipid has an alkyl tail. In some embodiments, a tail has one or more ester linkages, which may enhance biodegradability. In some embodiments, a tail is branched, such as with 3 or more branches. In some embodiments, a branched tail may enhance endosomal escape. In some embodiments, an ionizable lipid has a pKa between 6 and 7, which may be measured, for example, by TNS assay. [00730] In some embodiments, an ionizable lipid has a structure of any of the formulas disclosed below, and all formulas disclosed in a reference publication and patent application publication cited below. In some embodiments, an ionizable lipid comprises a head group of any structure or formula disclosed below. In some embodiments, an ionizable lipid comprises a bridging moiety of any structure or formula disclosed below. In some embodiments, an ionizable lipid comprises any tail group, or combination of tail groups disclosed below. The present disclosure contemplates all permutations and combinations of head group, bridging moiety and tail group, or tail groups, disclosed herein. [00731] In some embodiments, a head, tail, or structure of an ionizable lipid is described in US patent application US20170210697A1. [00732] In some embodiments, a compound has a structure according to formula 1:
Figure imgf000240_0001
wherein: R1 is selected from the group consisting of C5-30 alkyl, C5-20 alkenyl, - R*YR", YR", and - R"M'R'; R2 and R3 are independently selected from the group consisting of H, C1-14 alkyl, C2-14 alkenyl, —R*YR", —YR", and —R*OR", or R2 and R3, together with the atom to which they are attached, form a heterocycle or carbocycle; R4 is selected from the group consisting of a C3-6 carbocycle, —(CH2)nQ, —(CH2)nCHQR, — CHQR, CO(R)2, and unsubstituted C1-6 alkyl, where Q is selected from a carbocycle, heterocycle, —OR, —O(CH2)nN(R)2, —C(O)OR, —OC(O)R, —CX3, —CX2H, -CXH2, -CN, N(R)2, –C(O)N(R)2, - N(R)C(O)R, -N(R)S(O)2R, -N(R)C(O)N(R)2, -N(R)C(S)N(R)2, -N(R)R8, -O(CH2)nOR, -N(R)C(=NR9)N(R)2 -N(R)C(=CHR9)N(R)2, -OC(O)N(R)2, -N(R)C(O)OR, - N(OR)C(O)R, -N(OR)S(O)2R, -N(OR)C(O)OR, -N(OR)C(O)N(R)2, -N(OR)C(S)N(R)2 - N(OR)C(—NR)N(R) - N(OR)C(=CHR9)N(R)2, -C(=NR9)N(R)2, —C(=NR9)R, -C(O)N(R)OR, and —C(R)N(R)2, C(O)OR, and each n is independently selected from 1, 2, 3, 4, and 5 or a head group disclosed in Table 1; each R5 is independently selected from the group consisting of C1-3 alkyl, C2-3 alkenyl, and H; each R6 is independently selected from the group consisting of C1-3 alkyl, C2-3 alkenyl, and H; M and M’ are independently selected from —C(O)O-, -OC(O)—, -C(O)N(R’)-, -N(R')C(O)-, - C(O)—, —C(S)—, —C(S)S-, —SC(S)—, —CH(OH)—, —P(O)(OR’)O-, —S(O)—, —S-S-, an aryl group, and a heteroaryl group; R7 is selected from the group consisting of C1-3alkyl, C2-3 alkenyl, and H; R8 is selected from the group consisting of C3-6 carbocycle and heterocycle; R9 is selected from the group consisting of H. CN, NO2, C1-6 alkyl, -OR, —S(O)2R, — S(O)2N(R)2, C2-6 alkenyl, C3-6 carbocycle and heterocycle; each R is independently selected from the group consisting of C1-3 alkyl, C2-3 alkenyl, and H; each R’ is independently selected from the group consisting of C1-18 alkyl, C2-18 alkenyl, — R*YR", —YR", and H; each R” is independently selected from the group consisting of C3-14 alkyl, C3-14 alkenyl, and H; each R* is independently selected from the group consisting of C1-12 alkyl and C2-12 alkenyl: each Y is independently a C3-6 carbocycle; each X is independently selected from the group consisting of F, Cl, Br, and I; each Q is is -OH, -NHC(S)N(R)2, -NHC(O)N(R)2, -N(R)C(O)R, -N(R)S(O)2R, -N(R)R8, - NHC(=NR9)N(R)2, -NHC(=CHR9)N(R)2, -OC(O)N(R)2, -N(R)C(O)OR, heteroaryl or heterocycloalkyl; and m is selected from 5, 6, 7, 8, 9, 10, 11, 12, and 13: and wherein when R4 is —(CH2)nQ, —(CH2)nCHQR, —CHQR, or —CQ(R)2, then (i) Q is not —N(R), when n is 1, 2, 3, 4 or 5, or (ii) Q is not 5, 6, or 7-membered heterocycloalkyl when n is 1 or 2. [00733] In some embodiments, R4 is in Table 1. [00734] In some embodiments, R4 in formula 1 is selected from head groups 1-47. Table 1 - Ionizable lipid head groups
Figure imgf000242_0001
Figure imgf000243_0001
Figure imgf000244_0001
Figure imgf000245_0001
Figure imgf000246_0001
Figure imgf000247_0001
[00735] In some embodiments, a subset of the compounds of formula 1 are also described by formula 1b:
Figure imgf000248_0001
wherein l is selected from 1, 2, 3, 4, and 5; M1 is a bond or M'; R4 is unsubstituted C1-3 alkyl, or -(CH2)nQ, in which n is 2, 3, or 4, and Q is -OH, -NHC(S)N(R)2, -NHC(O)N(R)2, -N(R)C(O)R, -N(R)S(O)2R, -N(R)R8, -NHC(=NR9)N(R)2, -NHC(=CHR9)N(R)2, -OC(O)N(R)2, - N(R)C(O)OR, heteroaryl or heterocycloalkyl; M and M' are independently selected from - C(O)O-, -OC(O)-, -C(O)N(R')-, -P(O)(OR')O-, -S-S-, an aryl group, and a heteroaryl group; and R2 and R3 are independently selected from the group consisting of H, C1-14 alkyl, and C2-14 alkenyl. In some embodiments, a head, tail, or structure of an ionizable lipid is described in international patent application PCT/US2018/058555. [00736] In some embodiments, an ionizable lipid has a structure according to formula 2:
Figure imgf000248_0002
wherein: one of L1 or L2 is -O(C=O)-, -(C=O)O-, -C(=O)-, -O-, -S(O)x-, -S-S-, -C(=O)S-, SC(=O)-, - NRaC(=O)-, -C(=O)NRa-, NRaC(=O)NRa-, -OC(=O)NRa- or -NRaC(=O)O-, and the other of L1 or L2 is -O(C=O)-, -(C=O)O-, -C(=O)-, -O-, -S(O)x-, -S-S-, -C(=O)S-, SC(=O)-, -NRaC(=O)- , -C(=O)NRa-, ,NRaC(=O)NRa-, -OC(=O)NRa- or -NRaC(=O)O- or a direct bond; Ra is H or C1-C12 alkyl; R1a and R1b are, at each occurrence, independently either (a) H or C1-C12 alkyl, or (b) R1a is H or C1-C12 alkyl, and R1b together with the carbon atom to which it is bound is taken together with an adjacent R1b and the carbon atom to which it is bound to form a carbon-carbon double bond; R2a and R2b are, at each occurrence, independently either (a) H or C1-C12 alkyl, or (b) R2a is H or C1-C12 alkyl, and R2b together with the carbon atom to which it is bound is taken together with an adjacent R2b and the carbon atom to which it is bound to form a carbon-carbon double bond; R3a and R3b are, at each occurrence, independently either (a) H or C1-C12 alkyl, or (b) R3a is H or C1-C12 alkyl, and R3b together with the carbon atom to which it is bound is taken together with an adjacent R3b and the carbon atom to which it is bound to form a carbon-carbon double bond; R4a and R4b are, at each occurrence, independently either (a) H or C1-C12 alkyl, or (b) R4a is H or C1-C12 alkyl, and R4b together with the carbon atom to which it is bound is taken together with an adjacent R4b and the carbon atom to which it is bound to form a carbon-carbon double bond; R5 and R6 are each independently methyl or cycloalkyl; R7 is, at each occurrence, independently H or C1-C12 alkyl; R8 and R9 are each independently unsubstituted C1-C12 alkyl; or R8 and R9, together with the nitrogen atom to which they are attached, form a 5, 6 or 7-membered heterocyclic ring comprising one nitrogen atom; a and d are each independently an integer from 0 to 24; b and c are each independently an integer from 1 to 24; e is 1 or 2; and x is 0, 1 or 2. [00737] In some embodiments, an ionizable lipid has a structure according to formula 3:
Figure imgf000250_0001
wherein: one of L1 or L2 is -O(C=O)-, -(C=O)O-, -C(=O)-, -O-, -S(O)x-, -S-S-, -C(=O)S-, SC(=O)-, - NRaC(=O)-, -C(=O)NRa-, NRaC(=O)NRa-, -OC(=O)NRa- or -NRaC(=O)O-, and the other of L1 or L2 is -O(C=O)-, -(C=O)O-, -C(=O)-, -O-, -S(O)x-, -S-S-, -C(=O)S-, SC(=O)-, -NRaC(=O)- , -C(=O)NRa-, NRaC(=O)NRa-, -OC(=O)NRa- or -NRaC(=O)O- or a direct bond; G1 is C1-C2 alkylene, -(C=O)-, -O(C=O)-, -SC(=O)-, -NRaC(=O)- or a direct bond: G2 is -C(=O)-, -(C=O)O-, -C(=O)S-, -C(=O)NRa- or a direct bond; G3 is C1-C6 alkylene; Ra is H or C1-C12 alkyl; R1a and R1b are, at each occurrence, independently either: (a) H or C1-C12 alkyl; or (b) R1a is H or C1-C12 alkyl, and R1b together with the carbon atom to which it is bound is taken together with an adjacent R1b and the carbon atom to which it is bound to form a carbon-carbon double bond; R2a and R2b are, at each occurrence, independently either: (a) H or C1-C12 alkyl; or (b) R2a is H or C1-C12 alkyl, and R2b together with the carbon atom to which it is bound is taken together with an adjacent R2b and the carbon atom to which it is bound to form a carbon-carbon double bond; R3a and R3b are, at each occurrence, independently either (a): H or C1-C12 alkyl; or (b) R3a is H or C1-C12 alkyl, and R3b together with the carbon atom to which it is bound is taken together with an adjacent R3b and the carbon atom to which it is bound to form a carbon-carbon double bond; R4a and R4b are, at each occurrence, independently either: (a) H or C1-C12 alkyl; or (b) R4a is H or C1-C12 alkyl, and R4b together with the carbon atom to which it is bound is taken together with an adjacent R4b and the carbon atom to which it is bound to form a carbon-carbon double bond; R5 and R6 are each independently H or methyl; R7 is C4-C20 alkyl; R8 and R9 are each independently C1-C12 alkyl; or R8 and R9, together with the nitrogen atom to which they are attached, form a 5, 6 or 7-membered heterocyclic ring; a, b, c and d are each independently an integer from 1 to 24; and x is 0, 1 or 2. In some embodiments, an ionizable lipid has a structure according to formula 4:
Figure imgf000251_0001
wherein: one of L1 or L2 is -O(C=O)-, -(C=O)O-, -C(=O)-, -O-, -S(O)x-, -S-S-, -C(=O)S-, SC(=O)-, - NRaC(=O)-, -C(=O)NRa-, NRaC(=O)NRa-, -OC(=O)NRa- or -NRaC(=O)O-, and the other of L1 or L2 is -O(C=O)-, -(C=O)O-, -C(=O)-, -O-, -S(O)x-, -S-S-, -C(=O)S-, SC(=O)-, -NRaC(=O)- , -C(=O)NRa-, ,NRaC(=O)NRa-, -OC(=O)NRa- or -NRaC(=O)O- or a direct bond; G1 and G2 are each independently unsubstituted C1-C12 alkylene or C1-C12 alkenylene; G3 is C1-C24 alkylene, C1-C24 alkenylene, C3-C8 cycloalkylene, C3-C8 cycloalkenylene; Ra is H or C1-C12 alkyl; R1 and R2 are each independently C6-C24 alkyl or C6-C24 alkenyl; R3 is H, OR5, CN, -C(=O)OR4, -OC(=O)R4 or -NR5C(=O)R4; R4 is C1-C12 alkyl; R5 is H or C1-C6 alkyl; and x is 0, 1 or 2. [0001] In some embodiments, an ionizable lipid has a structure according to formula 5:
Figure imgf000252_0001
wherein: one of G1 or G2 is, at each occurrence, -O(C=O)-, -(C=O)O-, -C(=O)-, -O-, -S(O)y, -S-S-, - C(=O)S-, SC(=O)-, -N(Ra)C(=O)-, -C(=O)N(Ra)-, -N(Ra)C(=O)N(Ra)-, -OC(=O)N(Ra)- or - N(Ra)C(=O)0-, and the other of G1 or G2 is, at each occurrence, -O(C=O)-, -(C=O)O-, -C(=O)-, -O-, -S(O)y, -S-S-, -C(=O)S-, -SC(=O)-, -N(Ra)C(=O)-, -C(=O)N(Ra)-, -N(Ra)C(=O)N(Ra)-, - OC(=O)N(Ra)- or -N(Ra)C(=O)O- or a direct bond; L is, at each occurrence, ~O(C=O)-, wherein ~ represents a covalent bond to X; X is CRa; Z is alkyl, cycloalkyl or a monovalent moiety comprising at least one polar functional group when n is 1; or Z is alkylene, cycloalkylene or a polyvalent moiety comprising at least one polar functional group when n is greater than 1; Ra is, at each occurrence, independently H, C1-C12 alkyl, C1-C12 hydroxylalkyl, C1-C12 aminoalkyl, C1-C12 alkylaminylalkyl, C1-C12 alkoxyalkyl, C1-C12 alkoxycarbonyl, C1-C12 alkylcarbonyloxy, C1-C12 alkylcarbonyloxyalkyl or C1-C12 alkylcarbonyl; R is, at each occurrence, independently either: (a) H or C1-C12 alkyl; or (b) R together with the carbon atom to which it is bound is taken together with an adjacent R and the carbon atom to which it is bound to form a carbon-carbon double bond; R1 and R2 have, at each occurrence, the following structure, respectively:
Figure imgf000253_0001
a1 and a2 are, at each occurrence, independently an integer from 3 to 12; b1 and b2 are, at each occurrence, independently 0 or 1; c1 and c2 are, at each occurrence, independently an integer from 5 to 10; d1 and d2 are, at each occurrence, independently an integer from 5 to 10; y is, at each occurrence, independently an integer from 0 to 2; and n is an integer from 1 to 6, wherein each alkyl, alkylene, hydroxylalkyl, aminoalkyl, alkylaminylalkyl, alkoxyalkyl, alkoxycarbonyl, alkylcarbonyloxy, alkylcarbonyloxyalkyl and alkylcarbonyl is optionally substituted with one or more substituent. [0002] In some embodiments, an ionizable lipid has a structure according to formula 6:
Figure imgf000253_0002
wherein: one of G1 or G2 is, at each occurrence, -O(C=O)-, -(C=O)O-, -C(=O)-, -O-, -S(O)y, -S-S-, - C(=O)S-, SC(=O)-, -N(Ra)C(=O)-, -C(=O)N(Ra)-, -N(Ra)C(=O)N(Ra)-, -OC(=O)N(Ra)- or - N(Ra)C(=O)O-, and the other of G1 or G2 is, at each occurrence, -O(C=O)-, -(C=O)O-, -C(=O)-, -O-, -S(O)y-, -S-S-, -C(=O)S-, -SC(=O)-, -N(Ra)C(=O)-, -C(=O)N(Ra)-, -N(Ra)C(=O)N(Ra)-, - OC(=O)N(Ra)- or -N(Ra)C(=O)O- or a direct bond; L is, at each occurrence, ~O(C=O)-, wherein ~ represents a covalent bond to X; X is CRa; Z is alkyl, cycloalkyl or a monovalent moiety comprising at least one polar functional group when n is 1; or Z is alkylene, cycloalkylene or a polyvalent moiety comprising at least one polar functional group when n is greater than 1; Ra is, at each occurrence, independently H, C1-C12 alkyl, C1-C12 hydroxylalkyl, C1-C12 aminoalkyl, C1-C12 alkylaminylalkyl, C1-C12 alkoxyalkyl, C1-C12 alkoxycarbonyl, C1-C12 alkylcarbonyloxy, C1-C12 alkylcarbonyloxyalkyl or C1-C12 alkylcarbonyl; R is, at each occurrence, independently either: (a) H or C1-C12 alkyl; or (b) R together with the carbon atom to which it is bound is taken together with an adjacent R and the carbon atom to which it is bound to form a carbon-carbon double bond; R1 and R2 have, at each occurrence, the following structure, respectively:
Figure imgf000254_0001
R' is, at each occurrence, independently H or C1-C12 alkyl; a1 and a2 are, at each occurrence, independently an integer from 3 to 12; b1 and b2 are, at each occurrence, independently 0 or 1; c1 and c2 are, at each occurrence, independently an integer from 2 to 12; d1 and d2 are, at each occurrence, independently an integer from 2 to 12; y is, at each occurrence, independently an integer from 0 to 2; and n is an integer from 1 to 6, wherein a1, a2, c1, c2, d1 and d2 are selected such that the sum of a1+c1+d1 is an integer from 18 to 30, and the sum of a2+c2+d2 is an integer from 18 to 30, and wherein each alkyl, alkylene, hydroxylalkyl, aminoalkyl, alkylaminylalkyl, alkoxyalkyl, alkoxycarbonyl, alkylcarbonyloxy, alkylcarbonyloxyalkyl and alkylcarbonyl is optionally substituted with one or more substituent. In certain embodiments of Formula (V), G1 and G2 are each independently -O(C=O)- or -(C=O)O-. [00738] In some embodiments, an ionizable lipid has a disulfide tail. [00739] In some embodiments, an ionizable lipid includes short peptides of 12-15 mer length as head groups. [00740] In some embodiments, the head of an ionizable lipid comprises the structure of Vitamin A, D, E, or K as described in the published Patent Application WO2019232095A1, which is incorporated by herein by reference in its entirety. [00741] In some embodiments, a lipid is described in international patent applications WO2021077067, or WO2019152557, each of which is incorporated herein by reference in its entirety. [00742] In some embodiments, an LNP described herein comprises a lipid, e.g., an ionizable lipid, disclosed in US 2019/0240354, which is incorporated herein by reference in its entirety. [00743] In some embodiments, the lipids disclosed in US 2019/0240354 are of Formula I:
Figure imgf000255_0001
or salts thereof, wherein: R1 and R2 are either the same or different and are independently hydrogen (H) or an optionally substituted C1-C6 alkyl, C2-C6 alkenyl, or C2-C6 alkynyl, or R1 and R2 may join to form an optionally substituted heterocyclic ring of 4 to 6 carbon atoms and 1 or 2 heteroatoms selected from the group consisting of nitrogen (N), oxygen (O), and mixtures thereof; R3 is either absent or is hydrogen (H) or a C1-C6 alkyl to provide a quaternary amine; R4 and R5 are either the same or different and are independently an optionally substituted C10-C24 alkyl, C10-C24 alkenyl, C10-C24 alkynyl, or C10-C24 acyl, wherein at least one of R4 and R5 comprises at least two sites of unsaturation; and n is 0, 1, 2, 3, or 4. [00744] In some embodiments, the lipids disclosed in US 2019/0240354 are of Formula II:
Figure imgf000256_0001
wherein R1 and R2 are either the same or different and are independently an optionally substituted C12-C24 alkyl, C12-C24 alkenyl, C12-C24 alkynyl, or C12-C24 acyl; R3 and R4 are either the same or different and are independently an optionally substituted C1-C6 alkyl, C2-C6 alkenyl, or C2- C6 alkynyl, or R3 and R4 may join to form an optionally substituted heterocyclic ring of 4 to 6 carbon atoms and 1 or 2 heteroatoms chosen from nitrogen and oxygen; R5 is either absent or is hydrogen (H) or a C1-C6 alkyl to provide a quaternary amine; m, n, and p are either the same or different and are independently either 0, 1, or 2, with the proviso that m, n, and p are not simultaneously 0; q is 0, 1, 2, 3, or 4; and Y and Z are either the same or different and are independently O, S, or NH. In some embodiments, q is 2. In some embodiments, the cationic lipid of Formula II is 2,2-dilinoleyl-4-(2- dimethylaminoethyl)-[1,3]-dioxolane, 2,2-dilinoleyl-4-(3-dimethylaminopropyl)-[1,3]-dioxolane, 2,2-dilinoleyl-4-(4-dimethylaminobutyl)-[1,3]-dioxolane, 2,2-dilinoleyl-5-dimethylaminomethyl- [1,3]-dioxane, 2,2-dilinoleyl-4-N-methylpepiazino-[1,3]-dioxolane, 2,2-dilinoleyl-4- dimethylaminomethyl-[1,3]-dioxolane, 2,2-dioleoyl-4-dimethylaminomethyl-[1,3]-dioxolane, 2,2-distearoyl-4-dimethylaminomethyl-[1,3]-dioxolane, 2,2-dilinoleyl-4-N-morpholino-[1,3]- dioxolane, 2,2-Dilinoleyl-4-trimethylamino-[1,3]-dioxolane chloride, 2,2-dilinoleyl-4,5- bis(dimethylaminomethyl)-[1,3]-dioxolane, 2,2-dilinoleyl-4-methylpiperzine-[1,3]-dioxolane, or mixtures thereof. In some embodiments, the cationic lipid of Formula II is 2,2-dilinoleyl-4-(2- dimethylaminoethyl)-[1,3]-dioxolane. [00745] In some embodiments, the lipids disclosed in US 2019/0240354 are of Formula III:
Figure imgf000256_0002
or salts thereof, wherein: R1 and R2 are either the same or different and are independently an optionally substituted C1-C6 alkyl, C2-C6 alkenyl, or C2-C6 alkynyl, or R1 and R2 may join to form an optionally substituted heterocyclic ring of 4 to 6 carbon atoms and 1 or 2 heteroatoms selected from the group consisting of nitrogen (N), oxygen (O), and mixtures thereof; R3 is either absent or is hydrogen (H) or a C1-C6 alkyl to provide a quaternary amine; R4 and R5 are either absent or present and when present are either the same or different and are independently an optionally substituted C1-C10 alkyl or C2-C10 alkenyl; and n is 0, 1, 2, 3, or 4. [00746] In some embodiments, the lipids disclosed in US 2019/0240354 are of Formula C: X-A-Y—Z1; (Formula C) or salts thereof, wherein: X is —N(H)R or —NR2; A is absent, C1 to C6 alkyl, C2 to C6 alkenyl, or C2 to C6 alkynyl, which C1 to C6 alkyl, C2 to C6 alkenyl, and C2 to C6 alkynyl is optionally substituted with one or more groups independently selected from oxo, halogen, heterocycle, —CN, —ORx, —NRxRy, —NRxC(═O)Ry, — NRxSO2Ry, —C(═O)Rx, —C(═O)ORx, —C(═O)NRxRy, —SOnRx, and —SOnNRxRy, wherein n is 0, 1, or 2, and Rx and Ry are each independently hydrogen, alkyl, or heterocycle, wherein each alkyl and heterocycle of Rx and Ry may be further substituted with one or more groups independently selected from oxo, halogen, —OH, —CN, alkyl, —ORx′, heterocycle, —NRx′Ry′, —NRx′C(═O)Ry′, —NRx′SO2Ry′, —C(═O)Rx′, —C(═O)ORx′, —C(═O)NRx′Ry′, —SOn′Rx′, and —SOn′NRx′Ry′, wherein n′ is 0, 1, or 2, and Rx′ and Ry′ are each independently hydrogen, alkyl, or heterocycle; Y is selected from the group consisting of absent, —C(═O)—, —O—, —OC(═O)—, — C(═O)O—, —N(Rb)C(═O)—, —C(═O)N(Rb)—, —N(Rb)C(═O)O—, and —OC(═O)N(Rb)—; Z1 is a C1 to C6 alkyl that is substituted with three or four Rx groups, wherein each Rx is independently selected from C6 to C11 alkyl, C6 to C11 alkenyl, and C6 to C11 alkynyl, which C6 to C11 alkyl, C6 to C11 alkenyl, and C6 to C11 alkynyl is optionally substituted with one or more groups independently selected from oxo, halogen, heterocycle, —CN, —ORx, —NRxRy, — NRxC(═O)Ry, —NRxSO2Ry, —C(═O)Rx, —C(═O)ORx, —C(═O)NRxRy, —SOnRx, and — SOnNRxRy, wherein n is 0, 1, or 2, and Rx and Ry are each independently hydrogen, alkyl, or heterocycle, wherein any alkyl and heterocycle of Rx and Ry may be further substituted with one or more groups independently selected from oxo, halogen, —OH, —CN, alkyl, —ORx′, heterocycle, —NRx′Ry′, —NRx′C(═O)Ry′, —NRx′SO2Ry′, —C(═O)Rx′, —C(═O)ORx′, — C(═O)NRx′Ry′, —SOn′Rx′, and —SOn′NRx′Ry′, wherein n′ is 0, 1, or 2, and Rx′ and Ry′ are each independently hydrogen, alkyl, or heterocycle; each R is independently alkyl, alkenyl, or alkynyl, that is optionally substituted with one or more groups independently selected from oxo, halogen, heterocycle, —CN, —ORx, —NRxRy, — NRxC(═O)Ry, —NRxSO2Ry, —C(═O)Rx, —C(═O)ORx, —C(═O)NRxRy, —SOnRx, and — SOnNRxRy, wherein n is 0, 1, or 2, and Rx and Ry are each independently hydrogen, alkyl, or heterocycle, wherein any alkyl and heterocycle of Rx and Ry may be further substituted with one or more groups independently selected from oxo, halogen, —OH, —CN, alkyl, —ORx′, heterocycle, —NRx′Ry′, —NRx′C(═O)Ry′, —NRx′SO2Ry′, —C(═O)Rx′, —C(═O)ORx′, — C(═O)NRx′Ry′, —SOn′Rx′, and —SOn′NRx′Ry′, wherein n′ is 0, 1, or 2, and Rx′ and Ry′ are each independently hydrogen, alkyl, or heterocycle; and each Rb is H or C1 to C6alkyl. [00747] In some embodiments, an LNP described herein comprises a lipid, e.g., an ionizable lipid, disclosed in US 2010/0130588, which is incorporated herein by reference in its entirety. [00748] In some embodiments, the lipids disclosed in US 2010/0130588 are of Formula I:
Figure imgf000258_0001
wherein R1 and R2 are independently selected and are H or C1-C3 alkyls, R3 and R4 are independently selected and are alkyl groups having from about 10 to about 20 carbon atoms, and at least one of R3 and R4 comprises at least two sites of unsaturation. In some embodiments, R3 and R4 are both the same, i.e., R3 and R4 are both linoleyl (C18), etc. In some embodiments, R3 and R4 are different, i.e., R3 is tetradectrienyl (C14) and R4 is linoleyl (C18). [00749] In some embodiments, the lipid of Formula I is 1,2-dilinoleyloxy-N,N- dimethylaminopropane (DLinDMA) or 1,2-dilinolenyloxy-N,N-dimethylaminopropane (DLenDMA). [00750] In some embodiments, the lipids disclosed in US 2010/0130588 are of Formula II:
Figure imgf000259_0001
wherein R1 and R2 are independently selected and are H or C1-C3 alkyls, R3 and R4 are independently selected and are alkyl groups having from about 10 to about 20 carbon atoms, and at least one of R3 and R4 comprises at least two sites of unsaturation. [00751] In some embodiments, an LNP described herein comprises a lipid, e.g., an ionizable lipid, disclosed in US 2021/0087135, which is incorporated herein by reference in its entirety. [00752] In some embodiments, the lipids disclosed in US 2021/0087135 are of Formula (A):
Figure imgf000259_0002
or its N-oxide, or a salt or isomer thereof, wherein R′a is R′branched or R′cyclic; wherein R′branched is:
Figure imgf000259_0003
R′cyclic is:
Figure imgf000259_0004
wherein:
Figure imgf000260_0001
denotes a point of attachment; wherein R is H, and R, R, and R are each independently selected from the group consisting of H, C2-12 alkyl, and C2-12 alkenyl, wherein at least one of R, R, and R is selected from the group consisting of C2-12 alkyl and C2-12 alkenyl; R2 and R3 are each C1-14 alkyl; R4 is selected from the group consisting of —(CH2)2OH, —(CH2)3OH, —(CH2)4OH, — (CH2)5OH and
Figure imgf000260_0002
wherein:
Figure imgf000260_0003
denotes a point of attachment; R10 is N(R)2; each R is independently selected from the group consisting of C1-6 alkyl, C2- 3 alkenyl, and H; and n2 is selected from the group consisting of 1, 2, 3, 4, 5, 6, 7, 8, 9, and 10; each R5 is independently selected from the group consisting of OH, C1-3 alkyl, C2-3 alkenyl, and H; each R6 is independently selected from the group consisting of OH, C1-3 alkyl, C2-3 alkenyl, and H; R7 is H; M and M′ are each independently selected from the group consisting of —C(O)O— and — OC(O)—; R′ is a C1-12 alkyl or C2-12 alkenyl; Ya is a C3-6 carbocycle; R*″a is selected from the group consisting of C1-15 alkyl and C2-15 alkenyl; 1 is selected from the group consisting of 1, 2, 3, 4, and 5; s is 2 or 3; and m is selected from the group consisting of 5, 6, 7, 8, 9, 10, 11, 12, and 13. [00753] In some embodiments, an LNP described herein comprises a lipid, e.g., an ionizable lipid, disclosed in US 2021/0128488, which is incorporated herein by reference in its entirety [00754] In some embodiments, the lipids disclosed in US 2021/0128488 are of structure (I):
Figure imgf000261_0001
or a pharmaceutically acceptable salt, prodrug or stereoisomer thereof, wherein: L1 is —O(C═O)R′, —(C═O)OR1, —C(═O)R1, —OR1, —S(O)xR1, —S—SR1, —C(′O)SR′, — SC(═O)R′, —NRaC(═O)R1, —C(═O)NRbRc, —NRaC(═O)NRbRc, —OC(═O)NRbRc or — NRaC(═O)OR1; L2 is —O(C═O)R2, —(C═O)OR2, —C(═O)R2, —OR2, —S(O)xR2, —S—SR2, —C(═O)SR2, — SC(═O)R2, —NRdC(═O)R2, —C(═O)NReRf, —NRdC(═O)NReRf, —OC(═O)NReRf; — NRdC(═O)OR2 or a direct bond to R2; G1 and G2 are each independently C2-C12 alkylene or C2-C12 alkenylene; G3 is C1-C24 alkylene, C2-C24 alkenylene, C3-C8 cycloalkylene or C3-C8 cycloalkenylene; Ra, Rb, Rd and Re are each independently H or C1-C12 alkyl or C1-C12 alkenyl; Rc and Rf are each independently C1-C12 alkyl or C2-C12 alkenyl; R1 and R2 are each independently branched C6-C24 alkyl or branched C6-C24 alkenyl; R3 is —N(R4)R5; R4 is C1-C12 alkyl; R5 is substituted C1-C12 alkyl; and x is 0, 1 or 2, and wherein each alkyl, alkenyl, alkylene, alkenylene, cycloalkylene, cycloalkenylene, aryl and aralkyl is independently substituted or unsubstituted unless otherwise specified. [00755] In some embodiments, an LNP described herein comprises a lipid, e.g., an ionizable lipid, disclosed in US 2020/0121809, which is incorporated herein by reference in its entirety. [00756] In some embodiments the lipids disclosed in US 2020/0121809 have a structure of Formula II:
Figure imgf000262_0001
or a pharmaceutically acceptable salt, tautomer, prodrug or stereoisomer thereof, wherein: one of L1 or L2 is —O(C═O)—, —(C═O)O—, —C(═O)—, —O—, —S(O)x—, —S—S—, — C(═O)S—, SC(═O)—, —NRaC(═O)—, —C(═O)NRa—, NRaC(═O)NRa—, —OC(═O)NRa— or —NRaC(═O)O—, and the other of L1 or L2 is —O(C═O)—, —(C═O)O—, —C(═O)—, — O—, —S(O)x—, —S—S—, —C(═O)S—, SC(═O)—, —NRaC(═O)—, —C(═O)NRa—, NRaC(═O)NRa—, —OC(═O)NRa— or —NRaC(═O)O— or a direct bond; G1 is C1-C2 alkylene, —(C═O)—, —O(C═O)—, —SC(═O)—, —NRaC(═O)— or a direct bond; G2 is —C(═O)—, —(C═O)O—, —C(═O)S—, —C(═O)NRa— or a direct bond; G3 is C1-C6 alkylene; Ra is H or C1-C12 alkyl; R1a and R1b are, at each occurrence, independently either: (a) H or C1-C12 alkyl; or (b) R1a is H or C1-C12 alkyl, and R1b together with the carbon atom to which it is bound is taken together with an adjacent R1b and the carbon atom to which it is bound to form a carbon-carbon double bond; R2a and R2b are, at each occurrence, independently either: (a) H or C1-C12 alkyl; or (b) R2a is H or C1-C12 alkyl, and R2b together with the carbon atom to which it is bound is taken together with an adjacent R2b and the carbon atom to which it is bound to form a carbon-carbon double bond; R3a and R3b are, at each occurrence, independently either (a): H or C1-C12 alkyl; or (b) R3a is H or C1-C12 alkyl, and R3b together with the carbon atom to which it is bound is taken together with an adjacent R3b and the carbon atom to which it is bound to form a carbon-carbon double bond; R4a and R4b are, at each occurrence, independently either: (a) H or C1-C12 alkyl; or (b) R4a is H or C1-C12 alkyl, and R4b together with the carbon atom to which it is bound is taken together with an adjacent R4b and the carbon atom to which it is bound to form a carbon-carbon double bond; R5 and R6 are each independently H or methyl; R7 is C4-C20 alkyl; R8 and R9 are each independently C1-C12 alkyl; or R8 and R9, together with the nitrogen atom to which they are attached, form a 5, 6 or 7-membered heterocyclic ring; a, b, c and d are each independently an integer from 1 to 24; and x is 0, 1 or 2. [00757] In some embodiments, the lipids disclosed in US 2020/0121809 have a structure of Formula III:
Figure imgf000263_0001
or a pharmaceutically acceptable salt, prodrug or stereoisomer thereof, wherein: one of L1 or L2 is —O(C═O)—, —(C═O)O—, —C(═O)—, —O—, —S(O)x—, —S—S—, — C(═O)S—, SC(═O)—, —NRaC(═O)—, —C(═O)NRa—, NRaC(═O)NRa—, —OC(═O)NRa— or —NRaC(═O)O—, and the other of L1 or L2 is —O(C═O)—, —(C═O)O—, —C(═O)—, — O—, —S(O)x—, —S—S—, —C(═O)S—, SC(═O)—, —NRaC(═O)—, —C(═O)NRa—, NRaC(═O)NRa—, —OC(═O)NRa— or —NRaC(═O)O— or a direct bond; G1 and G2 are each independently unsubstituted C1-C12 alkylene or C1-C12 alkenylene; G3 is C1-C24 alkylene, C1-C24 alkenylene, C3-C8 cycloalkylene, C3-C8 cycloalkenylene; Ra is H or C1-C12 alkyl; R1 and R2 are each independently C6-C24 alkyl or C6-C24 alkenyl; R3 is H, ORS, CN, —C(═O)OR4, —OC(═O)R4 or —NR5C(═O)R4; R4 is C1-C12 alkyl; R5 is H or C1-C6 alkyl; and x is 0, 1 or 2. [00758] In some embodiments, the lipids disclosed in US 2020/0121809 have a structure of Formula (IV):
Figure imgf000264_0001
or a pharmaceutically acceptable salt, prodrug or stereoisomer thereof, wherein: one of G1 or G2 is, at each occurrence, —O(C═O)—, —(C═O)O—, —C(═O)—, —O—, — S(O)y—, —S—S—, —C(═O)S—, SC(═O)—, —N(Ra)C(═O)—, —C(═O)N(Ra)—, — N(Ra)C(═O)N(Ra)—, —OC(═O)N(Ra)— or —N(Ra)C(═O)O—, and the other of G1 or G2 is, at each occurrence, —O(C═O)—, —(C═O)O—, —C(═O)—, —O—, —S(O)y—, —S—S—, — C(═O)S—, —SC(═O)—, —N(Ra)C(═O)—, —C(═O)N(Ra)—, —N(Ra)C(═O)N(Ra)—, — OC(═O)N(Ra)— or —N(Ra)C(═O)O— or a direct bond; L is, at each occurrence, —O(C═O)—, wherein - represents a covalent bond to X; X is CRa; Z is alkyl, cycloalkyl or a monovalent moiety comprising at least one polar functional group when n is 1; or Z is alkylene, cycloalkylene or a polyvalent moiety comprising at least one polar functional group when n is greater than 1; Ra is, at each occurrence, independently H, C1-C12 alkyl, C1-C12 hydroxylalkyl, C1- C12 aminoalkyl, C1-C12 alkylaminylalkyl, C1-C12 alkoxyalkyl, C1-C12 alkoxycarbonyl, C1- C12 alkylcarbonyloxy, C1-C12 alkylcarbonyloxyalkyl or C1-C12 alkylcarbonyl; R is, at each occurrence, independently either: (a) H or C1-C12 alkyl; or (b) R together with the carbon atom to which it is bound is taken together with an adjacent R and the carbon atom to which it is bound to form a carbon-carbon double bond; R1 and R2 have, at each occurrence, the following structure, respectively:
Figure imgf000265_0001
a1 and a2 are, at each occurrence, independently an integer from 3 to 12; b1 and b2 are, at each occurrence, independently 0 or 1; c1 and c2 are, at each occurrence, independently an integer from 5 to 10; d1 and d2 are, at each occurrence, independently an integer from 5 to 10; y is, at each occurrence, independently an integer from 0 to 2; and n is an integer from 1 to 6, wherein each alkyl, alkylene, hydroxylalkyl, aminoalkyl, alkylaminylalkyl, alkoxyalkyl, alkoxycarbonyl, alkylcarbonyloxy, alkylcarbonyloxyalkyl and alkylcarbonyl is optionally substituted with one or more substituent. [00759] In some embodiments, an LNP described herein comprises a lipid, e.g., an ionizable lipid, disclosed in US 2013/0108685, which is incorporated herein by reference in its entirety. [00760] In some embodiments, the lipids disclosed in US 2013/0108685 are represented by the following formula (I):
Figure imgf000265_0002
wherein: R1 and R2 are, the same or different, each linear or branched alkyl, alkenyl or alkynyl having 12 to 24 carbon atoms, or R1 and R2 are combined together to form dialkylmethylene, dialkenylmethylene, dialkynylmethylene or alkylalkenylmethylene, X1 and X3 are hydrogen atoms, or are combined together to form a single bond or alkylene, X3 is absent or represents alkyl having 1 to 6 carbon atoms, or alkenyl having 3 to 6 carbon atoms, when X3 is absent, Y is absent, a and b are 0, L3 is a single bond, R3 is alkyl having 1 to 6 carbon atoms, alkenyl having 3 to 6 carbon atoms, pyrrolidin-3-yl, piperidin-3-yl, piperidin-4-yl, or alkyl having 1 to 6 carbon atoms or alkenyl having 3 to 6 carbon atoms substituted with 1 to 3 substituent(s), which is(are), the same or different, amino, monoalkylamino, dialkylamino, trialkylammonio, hydroxy, alkoxy, carbamoyl, monoalkylcarbamoyl, dialkylcarbamoyl, pyrrolidinyl, piperidyl or morpholinyl, and L1 and L2 are —O—, Y is absent, a and b are, the same or different, 0 to 3, and are not 0 at the same time, L3 is a single bond, R3 is alkyl having 1 to 6 carbon atoms, alkenyl having 3 to 6 carbon atoms, pyrrolidin-3-yl, piperidin-3-yl, piperidin-4-yl, or alkyl having 1 to 6 carbon atoms or alkenyl having 3 to 6 carbon atoms substituted with 1 to 3 substituent(s), which is(are), the same or different, amino, monoalkylamino, dialkylamino, trialkylammonio, hydroxy, alkoxy, carbamoyl, monoalkylcarbamoyl, dialkylcarbamoyl, pyrrolidinyl, piperidyl or morpholinyl, L1 and L2 are, the same or different, —O—, —CO—O— or —O—CO—, Y is absent, a and b are, the same or different, 0 to 3, L3 is a single bond, R3 is a hydrogen atom, and L1 and L2 are, the same or different, —O—, —CO—O— or —O—CO—, or Y is absent, a and b are, the same or different, 0 to 3, L3 is —CO— or —CO—O—, R3 is pyrrolidin-2-yl, pyrrolidin-3-yl, piperidin-2-yl, piperidin-3-yl, piperidin-4-yl, morpholin-2-yl, morpholin-3-yl, or alkyl having 1 to 6 carbon atoms or alkenyl having 3 to 6 carbon atoms substituted with 1 to 3 substituent(s), which is(are), the same or different, amino, monoalkylamino, dialkylamino, trialkylammonio, hydroxy, alkoxy, carbamoyl, monoalkylcarbamoyl, dialkylcarbamoyl, pyrrolidinyl, piperidyl or morpholinyl, wherein at least one of the substituents is amino, monoalkylamino, dialkylamino, trialkylammonio, pyrrolidinyl, piperidyl or morpholinyl, and L1 and L2 are, the same or different, —O—, —CO—O— or —O— CO—, and when X3 is alkyl having 1 to 6 carbon atoms or alkenyl having 3 to 6 carbon atoms, Y is a pharmaceutically acceptable anion, a and b are, the same or different, 0 to 3, L3 is a single bond, R3 is alkyl having 1 to 6 carbon atoms, alkenyl having 3 to 6 carbon atoms, pyrrolidin-2-yl, pyrrolidin-3-yl, piperidin-2-yl, piperidin-3-yl, piperidin-4-yl, morpholin-2-yl, morpholin-3-yl, or alkyl having 1 to 6 carbon atoms or alkenyl having 3 to 6 carbon atoms substituted with 1 to 3 substituent(s), which is(are), the same or different, amino, monoalkylamino, dialkylamino, trialkylammonio, hydroxy, alkoxy, carbamoyl, monoalkylcarbamoyl, dialkylcarbamoyl, pyrrolidinyl, piperidyl or morpholinyl, L1 and L2 are, the same or different, —O—, —CO—O— or —O—CO—). [00761] In some embodiments, an LNP described herein comprises a lipid, e.g., an ionizable lipid, disclosed in US 2013/0195920, which is incorporated herein by reference in its entirety. [00762] In some embodiments, the lipids disclosed in US 2013/0195920 are of formula (I), which has a branched alkyl at the alpha position adjacent to the biodegradable group (between the biodegradable group and the teriary carbon):
Figure imgf000267_0001
or a salt thereof (e.g., a pharmaceutically acceptable salt thereof), wherein R′ is absent, hydrogen, or alkyl (e.g., C1-C4 alkyl); with respect to R1 and R2, (i) R1 and R2 are each, independently, optionally substituted alkyl, alkenyl, alkynyl, cycloalkylalkyl, heterocycle, or R10; (ii) R1 and R2, together with the nitrogen atom to which they are attached, form an optionally substituted heterocylic ring; or (iii) one of R1 and R2 is optionally substituted alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkylalkyl, or heterocycle, and the other forms a 4-10 member heterocyclic ring or heteroaryl (e.g., a 6- member ring) with (a) the adjacent nitrogen atom and (b) the (R)a group adjacent to the nitrogen atom; each occurrence of R is, independently, —(CR3R4)—; each occurrence of R3 and R4 are, independently H, halogen, OH, alkyl, alkoxy, —NH2, R10, alkylamino, or dialkylamino (In some embodiments, each occurrence of R3 and R4 are, independently H or C1-C4 alkyl); each occurrence of R10 is independently selected from PEG and polymers based on poly(oxazoline), poly(ethylene oxide), poly(vinyl alcohol), poly(glycerol), poly(N- vinylpyrrolidone), poly[N-(2-hydroxypropyl)methacrylamide] and poly(amino acid)s, wherein (i) the PEG or polymer is linear or branched, (ii) the PEG or polymer is polymerized by n subunits, (iii) n is a number-averaged degree of polymerization between 10 and 200 units, and (iv) wherein the compound of formula has at most two R10 groups (preferably at most one R10 group); the dashed line to Q is absent or a bond; when the dashed line to Q is absent then Q is absent or is —O—, —NH—, —S—, —C(O)—, — C(O)O —, —OC(O)—, —C(O)N(R4)—, —N(R5)C(O)—, —S—S—, —OC(O)O—, —O— N═C(R5)—, —C(R5)═N—O—, —OC(O)N(R5)—, —N(R5)C(O)N(R5)—, —N(R5)C(O)O—, — C(O)S—, —C(S)O— or —C(R5)═N—O—C(O)—; or when the dashed line to Q is a bond then (i) b is 0 and (ii) Q and the tertiary carbon adjacent to it (C*) form a substituted or unsubstituted, mono- or bi-cyclic heterocyclic group having from 5 to 10 ring atoms (e.g., the heteroatoms in the heterocyclic group are selected from O and S, preferably O); each occurrence of R5 is, independently, H or alkyl (e.g. C1-C4 alkyl); X and Y are each, independently, alkylene or alkenylene (e.g., C4 to C20 alkylene or C4 to C20 alkenylene); M1 and M2 are each, independently, a biodegradable group (e.g., —OC(O)—, —C(O)O—, — SC(O)—, —C(O)S—, —OC(S)—, —C(S)O, —S—S—, C(R5)═N—, —N═C(R5)—, — C(R5)═N—O—, —O—N═C(R5)—, —C(O)(NR5)—, —N(R5)C(O)—, —C(S)(NR5)—, — N(R5)C(O)—, —N(R5)C(O)N(R5)—, —OC(O)O—, —OSi(R5)2O—, —C(O)(CR3R4)C(O)O—, —OC(O)(CR3R4)C(O)—, or
Figure imgf000268_0001
wherein R11 is a C2-C8 alkyl or alkenyl; each occurrence of Rz is, independently, C1-C8 alkyl (e.g., methyl, ethyl, isopropyl, n-butyl, n- pentyl, or n-hexyl); a is 1, 2, 3, 4, 5 or 6; b is 0, 1, 2, or 3; and Z1 and Z2 are each, independently, C8-C14 alkyl or C8-C14 alkenyl, wherein the alkenyl group may optionally be substituted with one or two fluorine atoms at the alpha position to a double bond which is between the double bond and the terminus of Z1 or Z2. [00763] In some embodiments, the lipids disclosed in US 2013/0195920 are of formula (II), which has a branched alkyl at the alpha position adjacent to the biodegradable group (between the biodegradable group and the terminus of the tail, i.e., Z1 or Z2):
Figure imgf000269_0001
or a salt thereof (e.g., a pharmaceutically acceptable salt thereof), wherein R′ is absent, hydrogen, or alkyl (e.g., C1-C4 alkyl); with respect to R1 and R2, (i) R1 and R2 are each, independently, optionally substituted alkyl, alkenyl, alkynyl, cycloalkylalkyl, heterocycle, or R10; (ii) R1 and R2, together with the nitrogen atom to which they are attached, form an optionally substituted heterocylic ring; or (iii) one of R1 and R2 is optionally substituted alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkylalkyl, or heterocycle, and the other forms a 4-10 membered heterocyclic ring or heteroaryl (e.g., a 6- member ring) with (a) the adjacent nitrogen atom and (b) the (R)a group adjacent to the nitrogen atom; each occurrence of R is, independently, —(CR3R4)—; each occurrence of R3 and R4 are, independently H, halogen, OH, alkyl, alkoxy, —NH2, R10, alkylamino, or dialkylamino (In some embodiments, each occurrence of R3 and R4 are, independently H or C1-C4 alkyl); each occurrence of R10 is independently selected from PEG and polymers based on poly(oxazoline), poly(ethylene oxide), poly(vinyl alcohol), poly(glycerol), poly(N- vinylpyrrolidone), poly[N-(2-hydroxypropyl)methacrylamide] and poly(amino acid)s, wherein (i) the PEG or polymer is linear or branched, (ii) the PEG or polymer is polymerized by n subunits, (iii) n is a number-averaged degree of polymerization between 10 and 200 units, and (iv) wherein the compound of formula has at most two R10 groups (preferably at most one R10 group); the dashed line to Q is absent or a bond; when the dashed line to Q is absent then Q is absent or is —O—, —NH—, —S—, —C(O)—, — C(O)O —, —OC(O)—, —C(O)N(R4)—, —N(R5)C(O)—, —S—S—, —OC(O)O—, —O— N═C(R5)—, —C(R5)═N—O—, —OC(O)N(R5)—, —N(R5)C(O)N(R5)—, —N(R5)C(O)O—, — C(O)S—, —C(S)O— or —C(R5)═N—O—C(O)—; or when the dashed line to Q is a bond then (i) b is 0 and (ii) Q and the tertiary carbon adjacent to it (C*) form a substituted or unsubstituted, mono- or bi-cyclic heterocyclic group having from 5 to 10 ring atoms (e.g., the heteroatoms in the heterocyclic group are selected from O and S, preferably O); each occurrence of R5 is, independently, H or alkyl; X and Y are each, independently, alkylene (e.g., C6-C8 alkylene) or alkenylene, wherein the alkylene or alkenylene group is optionally substituted with one or two fluorine atoms at the alpha position to the M1 or M2 group M1 and M2 are each, independently, a biodegradable group (e.g., —OC(O)—, —C(O)O—, — SC(O)—, —C(O)S—, —OC(S)—, —C(S)O, —S—S—, C(R5)═N—, —N═C(R5)—, — C(R5)═N—O—, —O—N═C(R5)—, —C(O)(NR5)—, —N(R5)C(O)—, —C(S)(NR5)—, — N(R5)C(O)—, —N(R5)C(O)N(R5)—, —OC(O)O—, —OSi(R5)2O—, —C(O)(CR3R4)C(O)O—, —OC(O)(CR3R4)C(O)—, or
Figure imgf000270_0001
wherein R11 is a C2-C8 alkyl or alkenyl; each occurrence of Rz is, independently, C1-C8 alkyl (e.g., methyl, ethyl, isopropyl); a is 1, 2, 3, 4, 5 or 6; b is 0, 1, 2, or 3; and Z1 and Z2 are each, independently, C8-C14 alkyl or C8-C14 alkenyl, wherein (i) the alkenyl group may optionally be substituted with one or two fluorine atoms at the alpha position to a double bond which is between the double bond and the terminus of Z1 or Z2; and (ii) the terminus of at least one of Z1 and Z2 is separated from the group M1 or M2 by at least 8 carbon atoms. [00764] In some embodiments, the lipids disclosed in US 2013/0195920 are of formula (III), which has a branching point at a position that is 2-6 carbon atoms (i.e., at the beta (β), gamma (γ), delta (δ), epsilon (ε) or zeta position (ζ) adjacent to the biodegradable group (between the biodegradable group and the terminus of the tail, i.e., Z1 or Z2):
Figure imgf000271_0001
or a salt thereof (e.g., a pharmaceutically acceptable salt thereof), wherein R′, R1, R2, R, R3, R4, R10, Q, R5, M1, M2, Rz, a, and b are defined as in formula (I); L1 and L2 are each, independently, C1-C5 alkylene or C2-C5 alkenylene; X and Y are each, independently, alkylene (e.g., C4 to C20 alkylene or C6-C8 alkylene) or alkenylene (e.g., C4 to C20 alkenylene); and Z1 and Z2 are each, independently, C8-C14 alkyl or C8-C14 alkenyl, wherein the alkenyl group may optionally be substituted with one or two fluorine atoms at the alpha position to a double bond which is between the double bond and the terminus of Z1 or Z2. and with the proviso that the terminus of at least one of Z1 and Z2 is separated from the group M1 or M2 by at least 8 carbon atoms. [00765] In some embodiments, the cationic lipid disclosed in US 2013/0195920 is a compound of formula (IV), which has a branching point at a position that is 2-6 carbon atoms (i.e., at beta (β), gamma (γ), delta (δ), epsilon (ε) or zeta position (ζ) adjacent to the biodegradable group (between the biodegradable group and the terminus of the tail, i.e., Z1 or Z2):
Figure imgf000272_0001
or a salt thereof (e.g., a pharmaceutically acceptable salt thereof), wherein R′, R1, R2, R, R3, R4, R10, Q, R5, M2, Rz, a, and b are defined as in formula (I); L1 and L2 and are each, independently, C1-C5 alkylene or C2-C5 alkenylene; X and Y are each, independently, alkylene or alkenylene (e.g., C12-C20 alkylene or C12- C20 alkenylene); and each occurrence of Z is independently C1-C4 alkyl (preferably, methyl). [00766] For example, in some embodiments, -L1-C(Z)3 is —CH2C(CH3)3. In some embodiments, -L1-C(Z)3 is —CH2CH2C(CH3)3. [00767] In some embodiments, the lipids disclosed in US 2013/0195920 are of formula (V), which has an alkoxy or thioalkoxy (i.e., —S-alkyl) group substitution on at least one tail:
Figure imgf000272_0002
or a salt thereof (e.g., a pharmaceutically acceptable salt thereof), wherein R′, R1, R2, R, R3, R4, R10, Q, R5, M1, M2, a, and b are defined as in formula (I); X and Y are each, independently, alkylene (e.g., C6-C8 alkylene) or alkenylene, wherein the alkylene or alkenylene group is optionally substituted with one or two fluorine atoms at the alpha position to the M1 or M2 group; Z1 and Z2 are each, independently, C8-C14 alkyl or C8-C14 alkenyl, wherein (i) the C8-C14 alkyl or C8-C14 alkenyl of at least one of Z1 and Z2 is substituted by one or more alkoxy (e.g., a C1- C4 alkoxy such as —OCH3) or thioalkoxy (e.g., a C1-C4 thioalkoxy such as —SCH3) groups, and (ii) the alkenyl group may optionally be substituted with one or two fluorine atoms at the alpha position to a double bond which is between the double bond and the terminus of Z1 or Z2. [00768] In some embodiments, the lipids disclosed in US 2013/0195920 are of formula (VIA), which has one or more fluoro substituents on at least one tail at a position that is either alpha to a double bond or alpha to a biodegradable group:
Figure imgf000273_0002
or a salt thereof (e.g., a pharmaceutically acceptable salt thereof), wherein R1, R2, R, a, and b are as defined with respect to formula (I); Q is absent or is —O—, —NH—, —S—, —C(O)—, —C(O)O—, —OC(O)—, —C(O)N(R4)—, —N(R5)C(O)—, —S—S—, —OC(O)O—, —O—N═C(R5)—, —C(R5)═N—O—, — OC(O)N(R5)—, —N(R5)C(O)N(R5)—, —N(R5)C(O)O —, —C(O)S—, —C(S)O— or — C(R5)═N—O—C(O)—; R′ is absent, hydrogen, or alkyl (e.g., C1-C4 alkyl); and each of R9 and R10 are independently C12-C24 alkyl (e.g., C12-C20 alkyl), C12-C24 alkenyl (e.g., C12- C20 alkenyl), or C12-C24 alkoxy (e.g., C12-C20 alkoxy) (a) having one or more biodegradable groups and (b) optionally substituted with one or more fluorine atoms at a position which is (i) alpha to a biodegradable group and between the biodegradable group and the tertiary carbon atom marked with an asterisk (*), or (ii) alpha to a carbon-carbon double bond and between the double bond and the terminus of the R9 or R10 group; each biodegradable group independently interrupts the C12-C24 alkyl, alkenyl, or alkoxy group or is substituted at the terminus of the C12- C24 alkyl, alkenyl, or alkoxy group, wherein (i) at least one of R9 and R10 contains a fluoro group; (ii) the compound does not contain the following moiety:
Figure imgf000273_0001
wherein - - - - is an optional bond; and (iii) the terminus of R9 and R10 is separated from the tertiary carbon atom marked with an asterisk (*) by a chain of 8 or more atoms (e.g., 12 or 14 or more atoms). [00769] In some embodiments, the terminus of R9 and R10 is separated from the tertiary carbon atom marked with an asterisk (*) by a chain of 18-22 carbon atoms (e.g., 18-20 carbon atoms). [00770] In some embodiments, the lipids disclosed in US 2013/0195920 are of formula (VIB), which has one or more fluoro substituents on at least one tail at a position that is either alpha to a double bond or alpha to a biodegradable group:
Figure imgf000274_0001
or a salt thereof (e.g., a pharmaceutically acceptable salt thereof), wherein R′, R1, R2, R, R3, R4, R10, Q, R5, M1, M2, a, and b are defined as in formula (I); X and Y are each, independently, alkylene (e.g., C6-C8 alkylene) or alkenylene, wherein the alkylene or alkenylene group is optionally substituted with one or two fluorine atoms at the alpha position to the M1 or M2 group; and Z1 and Z2 are each, independently, C8-C14 alkyl or C8-C14 alkenyl, wherein said C8-C14 alkenyl is optionally substituted by one or more fluorine atoms at a position that is alpha to a double bond, wherein at least one of X, Y, Z1, and Z2 contains a fluorine atom. [00771] In some embodiments, the lipids disclosed in US 2013/0195920 are of formula (VII), which has an acetal group as a biodegradable group in at least one tail:
Figure imgf000274_0002
or a salt thereof (e.g., a pharmaceutically acceptable salt thereof), wherein R′, R1, R2, R, R3, R4, R10, Q, R5, a, and b are defined as in formula (I); X and Y are each, independently, alkylene (e.g., C6-C8 alkylene) or alkenylene, wherein the alkylene or alkenylene group is optionally substituted with one or two fluorine atoms at the alpha position to the M1 or M2 group M1 and M2 are each, independently, a biodegradable group (e.g., —OC(O)—, —C(O)O—, — SC(O)—, —C(O)S—, —OC(S)—, —C(S)O, —S—S—, C(R5)═N—, —N═C(R5)—, — C(R5)═N—O—, —O—N═C(R5)—, —C(O)(NR5)—, —N(R5)C(O)—, —C(S)(NR5)—, — N(R5)C(O)—, —N(R5)C(O)N(R5)—, —OC(O)O—, —OSi(R5)2O—, —C(O)(CR3R4)C(O)O—, —OC(O)(CR3R4)C(O)—, or
Figure imgf000275_0002
wherein R11 is a C4-C10 alkyl or C4-C10 alkenyl; with the proviso that at least one of M1 and M2 is
Figure imgf000275_0001
Z1 and Z2 are each, independently, C4-C14 alkyl or C4-C14 alkenyl, wherein the alkenyl group may optionally be substituted with one or two fluorine atoms at the alpha position to a double bond which is between the double bond and the terminus of Z1 or Z2. [00772] In some embodiments, an LNP described herein comprises a lipid, e.g., an ionizable lipid, disclosed in US 2015/0005363, which is incorporated herein by reference in its entirety. [00773] In some embodiments, an LNP described herein comprises a lipid, e.g., an ionizable lipid, disclosed in US 2014/0308304, which is incorporated herein by reference in its entirety. [00774] In some embodiments, the lipid disclosed in US 2014/0308304 is a compound of formula (I):
Figure imgf000276_0001
or a salt thereof (e.g., a pharmaceutically acceptable salt thereof), wherein Xaa is a D- or L-amino acid residue having the formula —NRN—CR1R2—(C═O)—, or a peptide of amino acid residues having the formula —{NRN—CR1R2—(C═O)}n—, wherein n is 2 to 20; R1 is independently, for each occurrence, a non-hydrogen, substituted or unsubstituted side chain of an amino acid; R2 and RN are independently, for each occurrence, hydrogen, an organic group consisting of carbon, oxygen, nitrogen, sulfur, and hydrogen atoms, or any combination of the foregoing, and having from 1 to 20 carbon atoms, C(1-5)alkyl, cycloalkyl, cycloalkylalkyl, C(3-5)alkenyl, C(3- 5)alkynyl, C(1-5)alkanoyl, C(1-5)alkanoyloxy, C(1-5)alkoxy, C(1-5)alkoxy-C(1-5)alkyl, C(1-5)alkoxy-C(1- 5)alkoxy, C(1-5)alkyl-amino-C(1-5)alkyl-, C(1-5)dialkyl-amino-C(1-5)alkyl-, nitro-C(1-5)alkyl, cyano- C(1-5)alkyl, aryl-C(1-5)alkyl, 4-biphenyl-C(1-5)alkyl, carboxyl, or hydroxyl; Z is NH, O, S, —CH2S—, —CH2S(O)—, or an organic linker consisting of 1-40 atoms selected from hydrogen, carbon, oxygen, nitrogen, and sulfur atoms (preferably, Z is NH or O); Rx and Ry are, independently, (i) a lipophilic tail derived from a lipid (which can be naturally- occurring or synthetic), phospholipid, glycolipid, triacylglycerol, glycerophospholipid, sphingolipid, ceramide, sphingomyelin, cerebroside, or ganglioside, wherein the tail optionally includes a steroid; (ii) an amino acid terminal group selected from hydrogen, hydroxyl, amino, and an organic protecting group; or (iii) a substituted or unsubstituted C(3-22)alkyl, C(6- 12)cycloalkyl, C(6-12)cycloalkyl-C(3-22)alkyl, C(3-22)alkenyl, C(3-22)alkynyl, C(3-22)alkoxy, or C(6-12)- alkoxy-C(3-22)alkyl; one of Rx and Ry is a lipophilic tail as defined above and the other is an amino acid terminal group, or both Rx and Ry are lipophilic tails; at least one of Rx and Ry is interrupted by one or more biodegradable groups (e.g., —OC(O)—, —C(O)O—, —SC(O)—, —C(O)S—, —OC(S)—, —C(S)O—, —S—S—, —C(R5)═N—, — N═C(R5)—, —C(R5)═N—O—, —O—N═C(R5)—, —C(O)(NR5)—, —N(R5)C(O)—, — C(S)(NR5)—, —N(R5)C(O)—, —N(R5)C(O)N(R5)—, —OC(O)O—, —OSi(R5)2O—, — C(O)(CR3R4)C(O)O—, —OC(O)(CR3R4)C(O)— or
Figure imgf000277_0001
, (wherein R11 is a C2-C8 alkyl or alkenyl), in which each occurrence of R5 is, independently, H or alkyl; and each occurrence of R3 and R4 are, independently H, halogen, OH, alkyl, alkoxy, — NH2, alkylamino, or dialkylamino; or R3 and R4, together with the carbon atom to which they are directly attached, form a cycloalkyl group (in some embodiments, each occurrence of R3 and R4 are, independently H or C1-C4 alkyl)); and Rx and Ry each, independently, optionally have one or more carbon-carbon double bonds. In some embodiments, the lipid disclosed in US 2014/0308304 is a compound of formula (IA):
Figure imgf000277_0002
or a salt thereof (e.g., a pharmaceutically acceptable salt thereof), wherein Z and Xaa are as defined with respect to formula (I) (the variables which are used in the definition of Xaa, namely RN, R1 and R2, are also as defined in formula (I)); each occurrence of R is, independently, —(CR3R4)—; each occurrence of R3 and R4 are, independently H, halogen, OH, alkyl, alkoxy, —NH2, alkylamino, or dialkylamino (in some embodiments, each occurrence of R3 and R4 are, independently H or C1-C4 alkyl); or R3 and R4, together with the carbon atom to which they are directly attached, form a cycloalkyl group, wherein no more than three R groups in each chain between the —Z-Xaa- C(O)— and Z2 moieties are cycloalkyl (e.g., cyclopropyl); Q1 and Q2 are each, independently, absent, —O—, —S—, —OC(O)—, —C(O)O—, —SC(O)—, —C(O)S—, —OC(S)—, —C(S)O—, —S—S—, —C(O)(NR5)—, —N(R5)C(O)—, — C(S)(NR5)—, —N(R5)C(O)—, —N(R5)C(O)N(R5)—, or —OC(O)O—; Q3 and Q4 are each, independently, H, —(CR3R4)—, cycloalkyl, heterocyclyl, heterocyclylalkyl, aryl, heteroaryl, or a cholesterol moiety; each occurrence of A1, A2, A3 and A4 is, independently, —(CR5R5—CR5═CR5)—; M1 and M2 are each, independently, a biodegradable group (e.g., —OC(O)—, —C(O)O—, — SC(O)—, —C(O)S—, —OC(S)—, —C(S)O—, —S—S—, —C(R5)═N—, —N═C(R5)—, — C(R5)═N—O—, —O—N═C(R5)—, —C(O)(NR5)—, —N(R5)C(O)—, —C(S)(NR5)—, — N(R5)C(O)—, —N(R5)C(O)N(R5)—, —OC(O)O—, —OSi(R5)2O—, —C(O)(CR3R4)C(O)O—, —OC(O)(CR3R4)C(O)—, or
Figure imgf000278_0001
(wherein R11 is a C2-C8 alkyl or alkenyl)); each occurrence of R5 is, independently, H or alkyl (e.g., C1-C4 alkyl); Z2 is absent, alkylene or —O—P(O)(OH)—O—; each ------ attached to Z2 is an optional bond, such that when Z2 is absent, Q3 and Q4 are not directly covalently bound together; c, d, e, f, i, j, m, n, q and r are each, independently, 0, 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10; g and h are each, independently, 0, 1 or 2; k and l are each, independently, 0 or 1, wherein at least one of k and l is 1; o and p are each, independently, 0, 1 or 2; and Q3 and Q4 are each, independently, separated from the —Z-Xaa-C(O)— moiety by a chain of 8 or more atoms (e.g., 12 or 14 or more atoms). In some embodiments the lipids disclosed in US 2014/0308304 are of the formula (IC):
Figure imgf000279_0001
or a salt thereof (e.g., a pharmaceutically acceptable salt thereof), wherein Z and Xaa are as defined with respect to formula (I) (the variables which are used in the definition of Xaa, namely RN, R1 and R2, are also as defined in formula (I)); each of R9 and R10 are, independently, alkylene or alkenylene; each of R11 and R12 are, independently, alkyl or alkenyl, optionally terminated by COOR13 wherein each R13 is independently unsubstituted alkyl (e.g., C1-C4 alkyl such as methyl or ethyl), substituted alkyl (such as benzyl), or cycloalkyl; M1 and M2 are each, independently, a biodegradable group (e.g., —OC(O)—, —C(O)O—, — SC(O)—, —C(O)S—, —OC(S)—, —C(S)O—, —S—S—, —C(R5)═N—, —N═C(R5)—, — C(R5)═N—O—, —O—N═C(R5)—, —C(O)(NR5)—, —N(R5)C(O)—, —C(S)(NR5)—, — N(R5)C(O)—, —N(R5)C(O)N(R5)—, —OC(O)O—, —OSi(R5)2O—, —C(O)(CR3R4)C(O)O—, —OC(O)(CR3R4)C(O)—, or
Figure imgf000279_0002
wherein R11 is a C2-C8 alkyl or alkenyl, in which each occurrence of R5 is, independently, H or alkyl; and each occurrence of R3 and R4 are, independently H, halogen, OH, alkyl, alkoxy, — NH2, alkylamino, or dialkylamino; or R3 and R4, together with the carbon atom to which they are directly attached, form a cycloalkyl group (in some embodiments, each occurrence of R3 and R4 are, independently H or C1-C4 alkyl)); R9, M1, and R11 are together at least 8 carbon atoms in length (e.g., 12 or 14 carbon atoms or longer); and R10, M2, and R12 are together at least 8 carbon atoms in length (e.g., 12 or 14 carbon atoms or longer). [00775] In some embodiments, the lipid disclosed in US 2014/0308304 is a compound of the formula II:
Figure imgf000280_0002
or a salt thereof (e.g., a pharmaceutically acceptable salt thereof), wherein: s is 1, 2, 3 or 4; and R7 is selected from lysyl, ornithyl, 2,3-diaminobutyryl, histidyl and an acyl moiety of the formula:
Figure imgf000280_0001
t is 1, 2 or 3; the NH3 + moiety in the acyl moiety in R7 is optionally absent; each occurrence of Y is independently a pharmaceutically acceptable anion (e.g., halide, such as chloride); R5 and R6 are each, independently a lipophilic tail derived from a naturally-occurring or synthetic lipid, phospholipid, glycolipid, triacylglycerol, glycerophospholipid, sphingolipid, ceramide, sphingomyelin, cerebroside, or ganglioside, wherein the tail may contain a steroid; or a substituted or unsubstituted C(3-22)alkyl, C(6-12)cycloalkyl, C(6-12)cycloalkyl-C(3-22)alkyl, C(3- 22)alkenyl, C(3-22)alkynyl, C(3-22)alkoxy, or C(6-12)alkoxy-C(3-22)alkyl; at least one of R5 and R6 is interrupted by one or more biodegradable groups (e.g., —SC(O)—, —C(O)S—, —OC(S)—, —C(S)O—, —S—S—, —C(O)(NRa)—, —N(Ra)C(O)—, — C(S)(NRa)—, —N(Ra)C(O)—, —N(Ra)C(O)N(Ra)—, or —OC(O)O—); each occurrence of Ra is, independently, H or alkyl; and R5 and R6 each, independently, optionally contain one or more carbon-carbon double bonds. [00776] In some embodiments, the lipids disclosed in US 2014/0308304 are of the formula (IIA):
Figure imgf000281_0001
or a salt thereof (e.g., a pharmaceutically acceptable salt thereof), wherein: R7 and s are as defined with respect to formula (II); each occurrence of R is, independently, —(CR3R4)—; each occurrence of R3 and R4 are, independently H, halogen, OH, alkyl, alkoxy, —NH2, alkylamino, or dialkylamino (in some embodiments, each occurrence of R3 and R4 are, independently H or C1-C4 alkyl); or R3 and R4, together with the carbon atom to which they are directly attached, form a cycloalkyl group, wherein no more than three R groups in each chain attached to the nitrogen N* are cycloalkyl (e.g., cyclopropyl); Q1 and Q2 are each, independently, absent, —O—, —S—, —OC(O)—, —C(O)O—, —SC(O)—, —C(O)S—, —OC(S)—, —C(S)O—, —S—S—, —C(O)(NR5)—, —N(R5)C(O)—, — C(S)(NR5)—, —N(R5)C(O)—, —N(R5)C(O)N(R5)—, or —OC(O)O—; Q3 and Q4 are each, independently, H, —(CR3R4)—, aryl, cycloalkyl, heterocyclyl, heterocyclylalkyl, heteroaryl, or a cholesterol moiety; each occurrence of A1, A2, A3 and A4 is, independently, —(CR5R5—CR5═CR5)—; M1 and M2 are each, independently, a biodegradable group (e.g., —OC(O)—, —C(O)O—, — SC(O)—, —C(O)S—, —OC(S)—, —C(S)O—, —S—S—, —C(R5)═N—, —N═C(R5)—, — C(R5)═N—O—, —O—N═C(R5)—, —C(O)(NR5)—, —N(R5)C(O)—, —C(S)(NR5)—, — N(R5)C(O)—, —N(R5)C(O)N(R5)—, —OC(O)O—, —OSi(R5)2O—, —C(O)(CR3R4)C(O)O—, —OC(O)(CR3R4)C(O)—, or
Figure imgf000281_0002
wherein R11 is a C2-C8 alkyl or alkenyl; each occurrence of R5 is, independently, H or alkyl; Z is absent, alkylene or —O—P(O)(OH)—O—; each ------ attached to Z is an optional bond, such that when Z is absent, Q3 and Q4 are not directly covalently bound together; c, d, e, f, i, j, m, n, q and r are each, independently, 0, 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10; g and h are each, independently, 0, 1 or 2; k and l are each, independently, 0 or 1, where at least one of k and l is 1; and o and p are each, independently, 0, 1 or 2. [00777] In some embodiments the lipid disclosed in US 2014/0308304 are of the formula (IIC):
Figure imgf000282_0001
or a salt thereof (e.g., a pharmaceutically acceptable salt thereof), wherein: R7 and s are as defined with respect to formula (II); each of R9 and R10 are independently alkyl (e.g., C12-C24 alkyl) or alkenyl (e.g., C12-C24 alkenyl); each of R11 and R12 are independently alkyl or alkenyl, optionally terminated by COOR13 where each R13 is independently alkyl (e.g., C1-C4 alkyl such as methyl or ethyl); M1 and M2 are each, independently, a biodegradable group (e.g., —OC(O)—, —C(O)O—, — SC(O)—, —C(O)S—, —OC(S)—, —C(S)O—, —S—S—, —C(R5)═N—, —N═C(R5)—, — C(R5)═N—O—, —O—N═C(R5)—, —C(O)(NR5)—, —N(R5)C(O)—, —C(S)(NR5)—, — N(R5)C(O)—, —N(R5)C(O)N(R5)—, —OC(O)O—, —OSi(R5)2O—, —C(O)(CR3R4)C(O)O—, —OC(O)(CR3R4)C(O)—, or
Figure imgf000282_0002
wherein R11 is a C2-C8 alkyl or alkenyl; in which each occurrence of R5 is, independently, H or alkyl; and each occurrence of R3 and R4 are, independently H, halogen, OH, alkyl, alkoxy, —NH2, alkylamino, or dialkylamino; or R3 and R4, together with the carbon atom to which they are directly attached, form a cycloalkyl group (in some embodiments, each occurrence of R3 and R4 are, independently, H or C1- C4 alkyl)); R9, M1, and R11 are together at least 8 carbons atoms in length (e.g., 12 or 14 carbon atoms or longer); and R10, M2, and R12 are together at least 8 carbons atoms in length (e.g., 12 or 14 carbon atoms or longer). [00778] In some embodiments, the lipid disclosed in US 2014/0308304 is a compound of the formula (4):
Figure imgf000283_0001
wherein: X is N or P; R1, R2, R, a, b, M1, and M2 are as defined with respect to formula (I); Q is absent or is —O—, —NH—, —S—, —C(O)O—, —OC(O)—, —C(O)N(R4)—, — N(R5)C(O)—, —S—S—, —OC(O)O—, —O—N═C(R5)—, —C(R5)═N—O—, — OC(O)N(R5)—, —N(R5)C(O)N(R5)—, —N(R5)C(O)O—, —C(O)S—, —C(S)O— or — C(R5)═N—O—C(O)—; R′ is absent, hydrogen, or alkyl (e.g., C1-C4 alkyl); each of R9 and R10 are independently alkylene, or alkenylene; and each of R11 and R12 are independently alkyl or alkenyl, optionally terminated by COOR13 where each R13 is independently alkyl (e.g., C1-C4 alkyl such as methyl or ethyl); R9, M1, and R11 are together at least 8 carbons atoms in length (e.g., 12 or 14 carbon atoms or longer); and R10, M2, and R12 are together at least 8 carbons atoms in length (e.g., 12 or 14 carbon atoms or longer). [00779] In some embodiments, the lipid disclosed in US 2014/0308304 is a compound of the formula (5)
Figure imgf000284_0001
wherein: X is N or P; R1, R2, R, a, and b are as defined with respect to formula (I); Q is absent or is —O—, —NH—, —S—, —C(O)O—, —OC(O)—, —C(O)N(R4)—, — N(R5)C(O)—, —S—S—, —OC(O)O—, —O—N═C(R5)—, —C(R5)═N—O—, — OC(O)N(R5)—, —N(R5)C(O)N(R5)—, —N(R5)C(O)O—, —C(O)S—, —C(S)O— or — C(R5)═N—O—C(O)—;R′ is absent, hydrogen, or alkyl (e.g., C1-C4 alkyl); each of R9 and R10 are independently C12-C24 alkyl or alkenyl substituted at its terminus with a biodegradable group, such as —COOR13 where each R13 is independently alkyl (preferably C1- C4 alkyl such as methyl or ethyl). [00780] In some embodiments the lipids disclosed in US 2014/0308304 are of Formula A:
Figure imgf000284_0002
or a pharmaceutically acceptable salt or stereoisomer thereof, wherein: n is 0-6 (e.g., n is 0, 1 or 2); R1 and R2 are independently selected from H, (C1-C6)alkyl, heterocyclyl, and a polyamine, wherein said alkyl, heterocyclyl and polyamine are optionally substituted with one or more sub stituents selected from R′, or R1 and R2 can be taken together with the nitrogen to which they are attached to form a monocyclic heterocycle with 3-7 (e.g., 4-7) members optionally containing, in addition to the nitrogen, one or two additional heteroatoms selected from N, O and S, said monocyclic heterocycle is optionally substituted with one or more substituents selected from R′; R3 is selected from H and (C1-C6)alkyl, wherein said alkyl is optionally substituted with one or more substituents selected from R′, or R3 can be taken together with R1 to form a monocyclic heterocycle with 3-7 (e.g., 4-7) members optionally containing, in addition to the nitrogen, one or two additional heteroatoms selected from N, O and S, said monocyclic heterocycle is optionally substituted with one or more substituents selected from R′; each occurrence of R4, R3′ and R4′ is independently selected from H, (C1-C6)alkyl and O-alkyl, said alkyl is optionally substituted with one or more substituents selected from R′; or R3′ and R4′ when directly bound to the same carbon atom form an oxo (═O) group, cyclopropyl or cyclobutyl; or R3 and R4 form an oxo (═O) group; R5 is selected from H and (C1-C6)alkyl; or R5 can be taken together with R1 to form a monocyclic heterocycle with 4-7 members optionally containing, in addition to the nitrogen, one or two additional heteroatoms selected from N, O and S, said monocyclic heterocycle is optionally substituted with one or more substituents selected from R′; each occurrence of R′ is independently selected from halogen, R″, OR″, SR″, CN, CO2R″ and CON(R″)2; each occurrence of R″ is selected from H and (C1-C6)alkyl, wherein said alkyl is optionally substituted with one or more substituents selected from halogen and OH; L1 is a C4-C22 alkyl or C4-C22 alkenyl, said alkyl or alkenyl is optionally interrupted by or terminated with one or more biodegradable groups; and said alkyl or alkenyl is optionally substituted with one or more sub stituents selected from R′; and L2 is a C4-C22 alkyl or C4-C22 alkenyl, said alkyl or alkenyl is optionally interrupted by or terminated with one or more biodegradable groups; and said alkyl or alkenyl is optionally substituted with one or more sub stituents selected from R′; with the proviso that the CR3′R4′ group when present adjacent to the nitrogen atom in formula A is not a ketone (—C(O)—). [00781] In some embodiments the lipids disclosed in US 2014/0308304 are of formula B:
Figure imgf000286_0001
or a pharmaceutically acceptable salt or stereoisomer thereof, wherein: n is 0, 1, 2, 3, 4, or 5; R6 and R7 are each independently (i) C1-C4 linear or branched alkyl (e.g., methyl or ethyl) optionally substituted with 1-4 R′, or (ii) C3-C8 cycloalkyl (e.g., C3-C6 cycloalkyl); or R6 and R7 together with the nitrogen atom adjacent to them form a 3-6 membered ring; L1 is a C4-C22 alkyl or C4-C22 alkenyl, said alkyl or alkenyl optionally interrupted by or terminated with one or more biodegradable groups; and said alkyl or alkenyl is optionally substituted with 1-5 sub stituents selected from R′; and L2 is a C4-C22 alkyl or C4-C22 alkenyl, said alkyl or alkenyl optionally interrupted by or terminated with one or more biodegradable groups; and said alkyl or alkenyl is optionally substituted with 1-5 sub stituents selected from R′; each occurrence of R′ is independently selected from halogen, R″, OR″, SR″, CN, CO2R″ and CON(R″)2; and each occurrence of R″ is independently selected from H and (C1-C6)alkyl, wherein said alkyl is optionally substituted with one or more substituents selected from halogen and OH. [00782] In some embodiments, lipids disclosed in US 2014/0308304 are of formula C:
Figure imgf000286_0002
or a pharmaceutically acceptable salt or stereoisomer thereof, wherein: n is 0, 1, 2, 3, 4, or 5; L1 is a C4-C22 alkyl or C4-C22 alkenyl, said alkyl or alkenyl optionally has one or more biodegradable groups; each biodegradable group independently interrupts the alkyl or alkenyl group or is substituted at the terminus of the alkyl or alkenyl group, and said alkyl or alkenyl is optionally substituted with 1-5 sub stituents selected from R′; and L2 is a C4-C22 alkyl or C4-C22 alkenyl, said alkyl or alkenyl optionally interrupted by or terminated with one or more biodegradable groups; and said alkyl or alkenyl is optionally substituted with 1-5 sub stituents selected from R′; each occurrence of R′ is independently selected from halogen, R″, OR″, SR″, CN, CO2R″ and CON(R″)2; and each occurrence of R″ is independently selected from H and (C1-C6)alkyl, wherein said alkyl is optionally substituted with one or more substituents selected from halogen and OH. [00783] In some embodiments, the lipid disclosed in US 2014/0308304 are of formula D:
Figure imgf000287_0001
or a pharmaceutically acceptable salt or stereoisomer thereof, wherein m is 0, 1, 2, or 3; n is 0, 1, 2, 3, 4, or 5; R6 and R7 are each independently (i) C1-C4 linear or branched alkyl (e.g., methyl or ethyl) optionally substituted with 1-4 R′, or (ii) C3-C8 cycloalkyl (e.g., C3-C6 cycloalkyl); or R6 and R7 together with the nitrogen atom adjacent to them form a 3-6 membered ring; L1 is a C4-C22 alkyl or C4-C22 alkenyl, said alkyl or alkenyl optionally interrupted by or terminated with one or more biodegradable groups; and said alkyl or alkenyl is optionally substituted with 1-5 substituents selected from R′; and L2 is a C4-C22 alkyl or C4-C22 alkenyl, said alkyl or alkenyl optionally interrupted by or terminated with one or more biodegradable groups; and said alkyl or alkenyl is optionally substituted with 1-5 substituents selected from R′; each occurrence of R′ is independently selected from halogen, R″, OR″, SR″, CN, CO2R″ and CON(R″)2; and each occurrence of R″ is independently selected from H and (C1-C6)alkyl, wherein said alkyl is optionally substituted with one or more substituents selected from halogen and OH. [00784] In some embodiments lipid disclosed in US 2014/0308304 are of formula E:
Figure imgf000288_0001
or a pharmaceutically acceptable salt or stereoisomer thereof, wherein n is 0, 1, 2, 3, 4, or 5; the group “amino acid” is an amino acid residue; L1 is a C4-C22 alkyl or C4-C22 alkenyl, said alkyl or alkenyl optionally interrupted by or terminated with one or more biodegradable groups, and said alkyl or alkenyl is optionally substituted with 1-5 sub stituents selected from R′; and L2 is a C4-C22 alkyl or C4-C22 alkenyl, said alkyl or alkenyl optionally interrupted by or terminated with one or more biodegradable groups, and said alkyl or alkenyl is optionally substituted with 1-5 sub stituents selected from R′; each occurrence of R′ is independently selected from halogen, R″, OR″, SR″, CN, CO2R″ and CON(R″)2; and each occurrence of R″ is independently selected from H and (C1-C6)alkyl, wherein said alkyl is optionally substituted with one or more substituents selected from halogen and OH. [00785] The amino acid residue in formula E may have the formula —C(O)—C(R9)(NH2), where R9 is an amino acid side chain. [00786] In some embodiments, the lipid disclosed in US 2014/0308304 are of formula F:
Figure imgf000288_0002
or a pharmaceutically acceptable salt or stereoisomer thereof, wherein: R6 and R7 are independently (i) C1-C4 linear or branched alkyl (e.g., methyl or ethyl) optionally substituted with 1-4 R′, or (ii) C3-C8 cycloalkyl (e.g., C3-C6 cycloalkyl); or R6 and R7 together with the nitrogen atom adjacent to them form a 3-6 membered ring; L1 is a C4-C22 alkyl or C4-C22 alkenyl optionally interrupted by or terminated with one or more biodegradable groups, and said alkyl or alkenyl is optionally substituted with 1-5 substituents selected from R′; and L2 is a C4-C22 alkyl or C4-C22 alkenyl optionally interrupted by or terminated with one or more biodegradable groups, and said alkyl or alkenyl is optionally substituted with 1-5 substituents selected from R′; each occurrence of R′ is independently selected from halogen, R″, OR″, SR″, CN, CO2R″ and CON(R″)2; each occurrence of R″ is independently selected from H and (C1-C6)alkyl, wherein said alkyl is optionally substituted with one or more substituents selected from halogen and OH. [00787] In some embodiments, the lipid disclosed in US 2014/0308304 are of formula G:
Figure imgf000289_0001
or a pharmaceutically acceptable salt or stereoisomer thereof, wherein: n is 0, 1, 2, 3, 4, or 5; q is 1, 2, 3, or 4 R6 and R7 are independently (i) C1-C4 linear or branched alkyl (e.g., methyl or ethyl) optionally substituted with 1-4 R′, or (ii) C3-C8 cycloalkyl (e.g., C3-C6 cycloalkyl); L1 is a C4-C22 alkyl or C4-C22 alkenyl optionally interrupted by or terminated with one or more biodegradable groups, and said alkyl or alkenyl is optionally substituted with 1-5 substituents selected from R′; and L2 is a C4-C22 alkyl or C4-C22 alkenyl optionally interrupted by or terminated with one or more biodegradable groups, and said alkyl or alkenyl is optionally substituted with 1-5 substituents selected from R′; each occurrence of R′ is independently selected from halogen, R″, OR″, SR″, CN, CO2R″ and CON(R″)2; each occurrence of R″ is independently selected from H and (C1-C6)alkyl, wherein said alkyl is optionally substituted with one or more substituents selected from halogen and OH. [00788] In some embodiments, an LNP described herein comprises a lipid, e.g., an ionizable lipid, disclosed in US 2013/0053572, which is incorporated herein by reference in its entirety. [00789] In some embodiments, the lipids disclosed in US 2013/0053572 are of Formula A:
Figure imgf000290_0001
wherein: n is 0, 1 or 2; R1 and R2 are independently selected from H, (C1-C6)alkyl, heterocyclyl, and a polyamine, wherein said alkyl, heterocyclyl and polyamine are optionally substituted with one or more substituents selected from R′, or R1, and R2 can be taken together with the nitrogen to which they are attached to form a monocyclic heterocycle with 4-7 members optionally containing, in addition to the nitrogen, one or two additional heteroatoms selected from N, O and S, said monocyclic heterocycle is optionally substituted with one or more substituents selected from R′; R3 is selected from H and (C1-C6)alkyl, wherein said alkyl is optionally substituted with one or more substituents selected from R′, or R3 can be taken together with R1 to form a monocyclic heterocycle with 4-7 members optionally containing, in addition to the nitrogen, one or two additional heteroatoms selected from N, O and S, said monocyclic heterocycle is optionally substituted with one or more substituents selected from R′; R4 is selected from H, (C1-C6)alkyl and O-alkyl, said alkyl is optionally substituted with one or more substituents selected from R′; R5 is selected from H and (C1-C6)alkyl; or R5 can be taken together with R1 to form a monocyclic heterocycle with 4-7 members optionally containing, in addition to the nitrogen, one or two additional heteroatoms selected from N, O and S, said monocyclic heterocycle is optionally substituted with one or more substituents selected from R′; R′ is independently selected from halogen, R″, OR″, CN, CO2R″ and CON(R″)2; R″ is selected from H and (C1-C6)alkyl, wherein said alkyl is optionally substituted with one or more substituents selected from halogen and OH; L1 is a C4-C22 alkenyl, said alkenyl is optionally substituted with one or more substituents selected from R′; and L2 is a C4-C22 alkenyl, said alkenyl is optionally substituted with one or more substituents selected from R′; or any pharmaceutically acceptable salt or stereoisomer thereof. [00790] In some embodiments, an LNP described herein comprises a lipid, e.g., an ionizable lipid, disclosed in US Application publication US2017/0119904, which is incorporated by reference herein, in its entirety. [00791] In some embodiments, an LNP described herein comprises a lipid, e.g., an ionizable lipid, disclosed in PCT Application publication WO2021/204179, which is incorporated by reference herein, in its entirety. [00792] In some embodiments, an LNP described herein comprises a lipid, e.g., an ionizable lipid, disclosed in PCT Application PCT/US2022/031383, which is incorporated by reference herein, in its entirety. [00793] In some embodiments, an LNP described herein comprises an ionizable lipid of Table 2: Table 2 – Exemplary Ionizable Lipids
Figure imgf000291_0001
Figure imgf000292_0001
Figure imgf000293_0001
ii. Structural lipids
[00794] In some embodiments, an LNP comprises a structural lipid. Structural lipids can be selected from the group consisting of, but are not limited to, cholesterol, fecosterol, fucosterol, beta sitosterol, sitosterol, ergosterol, campesterol, stigmasterol, brassicasterol, tomatidine, cholic acid, sitostanol, litocholic acid, tomatine, ursolic acid, alpha-tocopherol, and mixtures thereof. In some embodiments, the structural lipid is cholesterol. In some embodiments, the structural lipid includes cholesterol and a corticosteroid (such as prednisolone, dexamethasone, prednisone, and hydrocortisone), or any combinations thereof. In some embodiments, a structural lipid is described in international patent application WO2019152557A1, which is incorporated herein by reference in its entirety.
[00795] In some embodiments, a structural lipid is a cholesterol analog. Using a cholesterol analog may enhance endosomal escape as described in Patel et al., Naturally- occuring cholesterol analogues in lipid nanoparticles induce polymorphic shape and enhance intracellular delivery of mRNA, Nature Communications (2020), which is incorporated herein by reference.
[00796] In some embodiments, a structural lipid is a phytosterol. Using a phytosterol may enhance endosomal escape as described in Herrera et al., Illuminating endosomal escape of polymorphic lipid nanoparticles that boost mRNA delivery, Biomaterials Science (2020), which is incorporated herein by reference. [00797] In some embodiments, a structural lipid contains plant sterol mimetics for enhanced endosomal release. iii. PEGylated lipids [00798] A PEGylated lipid is a lipid modified with polyethylene glycol. In some embodiments, the LNP comprises a compound of Formula I or a pharmaceutically acceptable salt thereof, as described herein above. In some embodiments, the LNP comprises a compound of Formula II or a pharmaceutically acceptable salt thereof, as described herein above. [00799] In some embodiments, an LNP comprises an additional PEGylated lipid or PEG- modified lipid. A PEGylated lipid may be selected from the non-limiting group consisting of PEG-modified phosphatidylethanolamines, PEG-modified phosphatidic acids, PEG-modified ceramides, PEG-modified dialkylamines, PEG-modified diacylglycerols, PEG-modified dialkylglycerols, and mixtures thereof. For example, a PEG lipid may be PEG-c-DOMG, PEG- DMG, PEG-DLPE, PEG-DMPE, PEG-DPPC, or a PEG-DSPE lipid. [00800] In some embodiments, the LNP comprises a PEGylated lipid disclosed in one of US 2019/0240354; US 2010/0130588; US 2021/0087135; WO 2021/204179; US 2021/0128488; US 2020/0121809; US 2017/0119904; US 2013/0108685; US 2013/0195920; US 2015/0005363; US 2014/0308304; US 2013/0053572; WO 2019/232095A1; WO 2021/077067; WO 2019/152557; US 2015/0203446; US 2017/0210697; US 2014/0200257; or WO 2019/089828A1, each of which is incorporated by reference herein in their entirety. v. Phospholipids [00801] In some embodiments, an LNP of the present disclosure comprises a phospholipid. Phospholipids useful in the compositions and methods may be selected from the non-limiting group consisting of 1,2-distearoyl-sn-glycero-3-phosphocholine (DSPC), 1,2- dioleoyl-sn-glycero-3-phosphoethanolamine (DOPE), 1,2-dilinoleoyl-sn-glycero-3- phosphocholine (DLPC), 1,2-dimyristoyl-sn-glycero-phosphocholine (DMPC), 1.2-dioleoyl-sn- glycero-3-phosphocholine (DOPC), 1,2-dipalmitoyl-sn-glycero-3-phosphocholine (DPPC), 1,2- diundecanoyl-sn-glycero-phosphocholine (DUPC), 1-palmitoyl-2-oleoyl-sn-glycero-3- phosphocho line (POPC), 1,2-di-O-octadecenyl-sn-glycero-3-phosphocholine (18:0 Diether PC), 1-oleoyl-2-cholesterylhemisuc cinoyl-sn-glycero-3-phosphocholine (OChemsPC), 1-hexadecyl- sn-glycero-3-phosphocholine (C16 Lyso PC), 1,2-dilinolenoyl-sn-glycero-3-phosphocholine, 1,2-diarachidonoyl-sn-glycero-3-phosphocholine, 1,2-didocosahexaenoyl-sn-glycero-3- phosphocholine, 1,2-diphytanoylsn-glycero-3-phosphoethanolamine (ME 16.0 PE), 1,2- distearoyl-sn-glycero-3-phosphoethanolamine, 1,2-dilinoleoyl-sn-glycero-3- phosphoethanolamine, 1,2-dilinolenoyl-sn-glycero-3-phosphoethanolamine, 1,2-diarachidonoyl- sn-glycero-3-phosphoethanolamine, 1,2-didocosahexaenoyl-sn-glycero-3-phosphoethanolamine, 1,2-dioleoyl-sn-glycero-3-phospho-rac-(1-glycerol) sodium salt (DOPG), and sphingomyelin. In some embodiments, an LNP includes DSPC. In certain embodiments, an LNP includes DOPE. In some embodiments, an LNP includes both DSPC and DOPE. [00802] In some embodiments, a phospholipid tail may be modified in order to promote endosomal escape as described in U.S.2021/0121411, which is incorporated herein by reference. [00803] In some embodiments, the LNP comprises a phospholipid disclosed in one of US 2019/0240354; US 2010/0130588; US 2021/0087135; WO 2021/204179; US 2021/0128488; US 2020/0121809; US 2017/0119904; US 2013/0108685; US 2013/0195920; US 2015/0005363; US 2014/0308304; US 2013/0053572; WO 2019/232095A1; WO 2021/077067; WO 2019/152557; US 2017/0210697; or WO 2019/089828A1, each of which is incorporated by reference herein in their entirety. [00804] In some embodiments, phospholipids disclosed in US 2020/0121809 have the following structure:
Figure imgf000295_0001
wherein R1 and R2 are each independently a branched or straight, saturated or unsaturated carbon chain (e.g., alkyl, alkenyl, alkynyl). vi. Targeting moieties [00805] In some embodiments, the lipid nanoparticle further comprises a targeting moiety. The targeting moiety may be an antibody or a fragment thereof. The targeting moiety may be capable of binding to a target antigen. [00806] In some embodiments, the pharmaceutical composition comprises a targeting moiety that is operably connected to a lipid nanoparticle. In some embodiments, the targeting moiety is capable of binding to a target antigen. In some embodiments, the target antigen is expressed in a target organ. [00807] In some embodiments, the target antigen is expressed more in the target organ than it is in the liver. [00808] In some embodiments, the targeting moiety is an antibody as described in WO2016189532A1, which is incorporated herein by reference. For example, in some embodiments, the targeted particles are conjugated to a specific anti-CD38 monoclonal antibody (mAb), which allows specific delivery of the siRNAs encapsulated within the particles at a greater percentage to B-cell lymphocytes malignancies (such as MCL) than to other subtypes of leukocytes. [00809] In some embodiments, the lipid nanoparticles may be targeted when conjugated/attached/associated with a targeting moiety such as an antibody. vii. Zwitterionic amino lipids [00810] In some embodiments, an LNP comprises a zwitterionic lipid. In some embodiments, an LNP comprising a zwitterionic lipid does not comprise a phospholipid. [00811] Zwitterionic amino lipids have been shown to be able to self-assemble into LNPs without phospholipids to load, stabilize, and release mRNAs intracellular as described in U.S. Patent Application 20210121411, which is incorporated herein by reference in its entirety. Zwitterionic, ionizable cationic and permanently cationic helper lipids enable tissue-selective mRNA delivery and CRISPR-Cas9 gene editing in spleen, liver and lungs as described in Liu et al., Membrane-destablizing ionizable phospholipids for organ-selective mRNA delivery and CRISPR-Cas gene editing, Nat Mater. (2021), which is incorporated herein by reference in its entirety. [00812] The zwitterionic lipids may have head groups containing a cationic amine and an anionic carboxylate as described in Walsh et al., Synthesis, Characterization and Evaluation of Ionizable Lysine-Based Lipids for siRNA Delivery, Bioconjug Chem. (2013), which is incorporated herein by reference in its entirety. Ionizable lysine-based lipids containing a lysine head group linked to a long-chain dialkylamine through an amide linkage at the lysine α-amine may reduce immunogenicity as described in Walsh et al., Synthesis, Characterization and Evaluation of Ionizable Lysine-Based Lipids for siRNA Delivery, Bioconjug Chem. (2013). viii. Additional Lipid Components [00813] In some embodiments, the LNP compositions of the present disclosure further comprise one or more additional lipid components capable of influencing the tropism of the LNP. In some embodiments, the LNP further comprises at least one lipid selected from DDAB, EPC, 14PA, 18BMP, DODAP, DOTAP, and C12-200 (see Cheng, et al. Nat Nanotechnol.2020 April; 15(4): 313–320.; Dillard, et al. PNAS 2021 Vol.118 No.52.). ix. LNP pharmaceutical compositions [00814] In some embodiments, a nanoparticle includes an ionizable lipid, a phospholipid, a PEG lipid, and a structural lipid. In certain embodiments, the lipid component of the nanoparticle composition includes about 30 mol % to about 60 mol % ionizable lipid, about 0 mol % to about 30 mol % phospholipid, about 18.5 mol % to about 48.5 mol % structural lipid, and about 0 mol% to about 10 mol% of PEG lipid, provided that the total mol % does not exceed 100%. In some embodiments, the lipid component of the nanoparticle composition includes about 35 mol % to about 55 mol % ionizable lipid, about 5 mol % to about 25 mol % phospholipid, about 30 mol % to about 40 mol % structural lipid, and about 0 mol % to about 10 mol % of PEG lipid. In a particular embodiment, the lipid component includes about 50 mol % ionizable lipid, about 10 mol % phospholipid, about 38.5 mol % structural lipid, and about 1.5 mol% of PEG lipid. In another particular embodiment, the lipid component includes about 40 mol % ionizable lipid, about 20 mol % phospholipid, about 38.5 mol % structural lipid, and about 1.5 mol % of PEG lipid. In another particular embodiment, the lipid component includes about 48.5 mol % ionizable lipid, about 10 mol % phospholipid, about 40 mol % structural lipid, and about 1.5 mol % of PEG lipid. In another particular embodiment, the lipid component includes about 48.5 mol % ionizable lipid, about 10 mol % phospholipid, about 39 mol % structural lipid, and about 2.5 mol % of PEG lipid. In some embodiments, the phospholipid may be DOPE or DSPC. In other embodiments, the PEG lipid may be PEG-DMG and/or the structural lipid may be cholesterol. The amount of active agent in a nanoparticle composition may depend on the size, composition, desired target and/or application, or other properties of the nanoparticle composition as well as on the properties of the active agent. For example, the amount of active agent useful in a nanoparticle composition may depend on the size, sequence, and other characteristics of the active agent. The relative amounts of active agent and other elements (e.g., lipids) in a nanoparticle composition may also vary. In some embodiments, the wt/wt ratio of the lipid component to an enzyme in a nanoparticle composition may be from about 5:1 to about 60:1, such as 5:1, 6:1, 7:1, 8:1, 9:1, 10:1, 11:1, 12:1, 13:1, 14:1, 15:1, 16:1, 17:1, 18:1, 19:1, 20:1, 25:1, 30:1, 35:1, 40:1, 45:1, 50:1, and 60:1. The amount of a enzyme in a nanoparticle composition may, for example, be measured using absorption spectroscopy (e.g., ultraviolet-visible spectroscopy). [00815] In some embodiments, a nanoparticle composition comprising an active agent of the present disclosure is formulated to provide a specific E:P ratio. The E:P ratio of the composition refers to the molar ratio of nitrogen atoms in one or more lipids to the number of phosphate groups in an RNA active agent. In general, a lower E:P ratio is preferred. The one or more enzymes, lipids, and amounts thereof may be selected to provide an E:P ratio from about 2:1 to about 30:1, such as 2:1, 3:1, 4:1, 5:1, 6:1, 7:1, 8:1, 9:1, 10:1, 12:1, 14:1, 16:1, 18:1, 20:1, 22:1, 24:1, 26:1, 28:1, or 30:1. In certain embodiments, the E:P ratio may be from about 2:1 to about 8:1. In other embodiments, the E:P ratio is from about 5:1 to about 8:1. For example, the E:P ratio may be about 5.0:1, about 5.5:1, about 5.67:1, about 6.0:1, about 6.5:1, or about 7.0:1. [00816] The characteristics of a nanoparticle composition may depend on the components thereof. For example, a nanoparticle composition including cholesterol as a structural lipid may have different characteristics than a nanoparticle composition that includes a different structural lipid. Similarly, the characteristics of a nanoparticle composition may depend on the absolute or relative amounts of its components. For instance, a nanoparticle composition including a higher molar fraction of a phospholipid may have different characteristics than a nanoparticle composi tion including a lower molar fraction of a phospholipid. Characteristics may also vary depending on the method and conditions of preparation of the nanoparticle composition. Nanoparticle compositions may be characterized by a variety of methods. For example, microscopy (e.g., transmission electron microscopy or scanning electron microscopy) may be used to examine the morphology and size distribution of a nanoparticle composition. Dynamic light scattering or potentiometry (e.g., potentiometric titrations) may be used to measure Zeta potentials. Dynamic light scattering may also be utilized to determine particle sizes. Instruments such as the Zetasizer Nano ZS (Malvern Instruments Ltd, Malvern, Worcestershire, UK) may also be used to measure multiple characteristics of a nanoparticle composition, Such as particle size, polydispersity index, and Zeta potential. [00817] The mean size of a nanoparticle composition may be between 10s of nm and 100s of nm, e.g., measured by dynamic light scattering (DLS). For example, the mean size may be from about 40 nm to about 150 nm, such as about 40 nm, 45 nm, 50 nm, 55 nm, 60 nm, 65 nm, 70 nm, 75 nm, 80 nm, 85 nm, 90 nm, 95 nm, 100 nm, 105 nm, 110 nm, 115nm, 120 nm, 125 nm, 130 nm, 135 nm, 140 nm, 145 nm, or 150 nm. In some embodiments, the mean size of a nanoparticle composition may be from about 50 nm to about 100 nm, from about 50 nm to about 90 nm, from about 50 nm to about 80 nm, from about 50 nm to about 70 nm, from about 50 nm to about 60 nm, from about 60 nm to about 100 nm, from about 60 nm to about 90 nm, from about 60 nm to about 80 nm, from about 60 nm to about 70 nm, from about 70 nm to about 100 nm, from about 70 nm to about 90 nm, from about 70 nm to about 80 nm, from about 80 nm to about 100 nm, from about 80 nm to about 90 nm, or from about 90 nm to about 100 nm. In certain embodiments, the mean size of a nanoparticle composition may be from about 70 nm to about 100 nm. In a particular embodiment, the mean size may be about 80 nm. In other embodiments, the mean size may be about 100 nm. [00818] A nanoparticle composition may be relatively homogenous. A polydispersity index may be used to indicate the homogeneity of a nanoparticle composition, e.g., the particle size distribution of the nanoparticle compositions. A small (e.g., less than 0.3) polydispersity index generally indicates a narrow particle size distribution. A nanoparticle composition may have a polydispersity index from about 0 to about 0.25, such as 0.01, 0.02, 0.03, 0.04, 0.05, 0.06, 0.07, 0.08, 0.09, 0.10, 0.11, 0.12, 0.13, 0.14, 0.15, 0.16, 0.17, 0.18, 0.19, 0.20, 0.21, 0.22, 0.23, 0.24, or 0.25. [00819] The Zeta potential of a nanoparticle composition may be used to indicate the electrokinetic potential of the composition. For example, the Zeta potential may describe the surface charge of a nanoparticle composition. Nanoparticle compositions with relatively low charges, positive or negative, are generally desirable, as more highly charged species may interact undesirably with cells, tissues, and other elements in the body. In some embodiments, the Zeta potential of a nanoparticle composition may be from about -10 mV to about +20 mV, from about -10 mV to about +15 mV, from about -10 mV to about +10 mV, from about -10 mV to about +5 mV, from about -10 mV to about 0 mV, from about -10 mV to about -5 mV, from about -5 mV to about +20 mV, from about -5 mV to about +15 mV, from about -5 mV to about +10 mV, from about -5 mV to about +5 mV, from about -5 mV to about 0 mV, from about 0 mV, to about +20 mV, from about 0 mV to about +15 mV, from about 0 mV to about +10 mV, from about 0 mV to about +5 mV, from about +5 mV to about +20 mV, from about +5 mV, to about +15 mV, or from about +5 mV to about +10 mV. [00820] The efficiency of encapsulation of a payload describes the amount of payload that is encapsulated or otherwise associated with a nanoparticle composition after preparation, relative to the initial amount provided. The encapsulation efficiency is desirably high (e.g., close to 100%). The encapsulation efficiency may be measured, for example, by comparing the amount of payload in a solution containing the nanoparticle composition before and after breaking up the nanoparticle composition with one or more organic solvents or detergents. Fluorescence may be used to measure the amount of free payload in a solution. For the nanoparticle compositions described herein, the encapsulation efficiency of a therapeutic and/or prophylactic may be at least 50%, for example 50%, 55%, 60%.65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100%. In some embodiments, the encapsulation efficiency may be at least 80%. In certain embodiments, the encapsulation efficiency may be at least 90%. [00821] Lipids and their method of preparation are disclosed in, e.g., U.S. Patent Nos. 8,569,256, 5,965,542 and U.S. Patent Publication Nos.2016/0199485, 2016/0009637, 2015/0273068, 2015/0265708, 2015/0203446, 2015/0005363, 2014/0308304, 2014/0200257, 2013/086373, 2013/0338210, 2013/0323269, 2013/0245107, 2013/0195920, 2013/0123338, 2013/0022649, 2013/0017223, 2012/0295832, 2012/0183581, 2012/0172411, 2012/0027803, 2012/0058188, 2011/0311583, 2011/0311582, 2011/0262527, 2011/0216622, 2011/0117125, 2011/0091525, 2011/0076335, 2011/0060032, 2010/0130588, 2007/0042031, 2006/0240093, 2006/0083780, 2006/0008910, 2005/0175682, 2005/017054, 2005/0118253, 2005/0064595, 2004/0142025, 2007/0042031, 1999/009076 and PCT Pub. Nos. WO 99/39741, WO 2017/117528, WO 2017/004143, WO 2017/075531, WO 2015/199952, WO 2014/008334, WO 2013/086373, WO 2013/086322, WO 2013/016058, WO 2013/086373, WO2011/141705, and WO 2001/07548 and Semple et. al, Nature Biotechnology, 2010, 28, 172-176, the full disclosures of which are herein incorporated by reference in their entirety for all purposes. [00822] A nanoparticle composition may include any substance useful in pharmaceutical compositions. For example, the nanoparticle composition may include one or more pharmaceutically acceptable excipients or accessory ingredients such as, but not limited to, one or more solvents, dispersion media, diluents, dispersion aids, suspension aids, granulating aids, disintegrants, fillers, glidants, liquid vehicles, binders, surface active agents, isotonic agents, thickening or emulsifying agents, buffering agents, lubricating agents, oils, preservatives, and other species. Excipients such as waxes, butters, coloring agents, coating agents, flavorings, and perfuming agents may also be included. Pharmaceutically acceptable excipients are well known in the art (see for example Remington’s The Science and Practice of Pharmacy, 21st Edition, A. R. Gennaro: Lippincott, Williams & Wilkins, Baltimore, Md., 2006).Other different lipids or liposomal formulations including nanoparticles and methods of administration include, but are not limited to, U.S. Patent Publication 20030203865, 20020150626, 20030032615, and 20040048787, which are specifically incorporated by reference to the extent they disclose formulations and other related aspects of administration and delivery of nucleic acids. Methods used for forming particles are also disclosed in U.S. Pat. Nos.5,844,107, 5,877,302, 6,008,336, 6,077,835, 5,972,901, 6,200,801, and 5,972,900, which are incorporated by reference for those aspects. [00823] In some embodiments, the LNP encapsulates the engineered retron, e.g., an engineered nucleic acid construct, ncRNA, vector system, RNA molecule, and/or engineered nucleic acid-enzyme construct as described herein. [00824] In some embodiments, the lipid nanoparticle comprises: one or more ionizable lipids; one or more structural lipids; one or more PEGylated lipids; and one or more phospholipids. In some embodiments, the one or more ionizable lipids is selected from the group consisting of those disclosed in Table X. [00825] In some embodiments, the one or more structural lipids are selected from the group consisting of cholesterol, fecosterol, beta sitosterol, sitosterol, ergosterol, campesterol, stigmasterol, brassicasterol, tomatidine, tomatine, ursolic acid, alpha-tocopherol, prednisolone, dexamethasone, prednisone, and hydrocortisone. In some embodiments, the one or more PEGylated lipids are selected from the group consisting of PEG-c-DOMG, PEG-DMG, PEG- DLPE, PEG-DMPE, PEG-DPPC, and PEG-DSPE. [00826] In some embodiments, the one or more phospholipids are selected from the group consisting of 1,2-distearoyl-sn-glycero-3-phosphocholine (DSPC), 1,2-dioleoyl-sn-glycero-3- phosphoethanolamine (DOPE), 1,2-dilinoleoyl-sn-glycero-3-phosphocholine (DLPC), 1,2- dimyristoyl-sn-glycero-phosphocholine (DMPC), 1.2-dioleoyl-sn-glycero-3-phosphocholine (DOPC), 1,2-dipalmitoyl-sn-glycero-3-phosphocholine (DPPC), 1,2-diundecanoyl-sn-glycero- phosphocholine (DUPC), 1-palmitoyl-2-oleoyl-sn-glycero-3-phosphocho line (POPC), 1,2-di-O- octadecenyl-sn-glycero-3-phosphocholine (18:0 Diether PC), 1-oleoyl-2-cholesterylhemisuc cinoyl-sn-glycero-3-phosphocholine (OChemsPC), 1-hexadecyl-sn-glycero-3-phosphocholine (C16 Lyso PC), 1,2-dilinolenoyl-sn-glycero-3-phosphocholine, 1,2-diarachidonoyl-sn-glycero-3- phosphocholine, 1,2-didocosahexaenoyl-sn-glycero-3-phosphocholine, 1,2-diphytanoylsn- glycero-3-phosphoethanolamine (ME 16.0 PE), 1,2-distearoyl-sn-glycero-3- phosphoethanolamine, 1,2-dilinoleoyl-sn-glycero-3-phosphoethanolamine, 1,2-dilinolenoyl-sn- glycero-3-phosphoethanolamine, 1,2-diarachidonoyl-sn-glycero-3-phosphoethanolamine, 1,2- didocosahexaenoyl-sn-glycero-3-phosphoethanolamine, 1,2-dioleoyl-sn-glycero-3-phospho-rac- (1-glycerol) sodium salt (DOPG), and sphingomyelin. [00827] In some embodiments, the lipid nanoparticle comprises about 48.5 mol % ionizable lipid, about 10 mol % phospholipid, about 40 mol % structural lipid, and about 1.5 mol% of PEG lipid. [00828] In some embodiments, the lipid nanoparticle comprises about 48.5 mol % ionizable lipid, about 10 mol % phospholipid, about 39 mol % structural lipid, and about 2.5 mol% of PEG lipid. In some embodiments, the LNP further comprises a targeting moiety operably connected to the LNP. In some embodimens, the LNP further comprises one or more additional components selected from the group consisting of DDAB, EPC, 14PA, 18BMP, DODAP, DOTAP, and C12-200. [00829] In some embodiments, the engineered retron can be used for gene transfer, which may be performed under ex vivo or in vivo conditions. Ex vivo gene therapy refers to the isolation of cells from a subject, the delivery of a nucleic acid into cells in vitro, and the return of the modified cells back into the subject. This may involve the collection of a biological sample comprising cells from the subject. For example, blood can be obtained by venipuncture, and solid tissue samples can be obtained by surgical techniques according to methods well known in the art. [00830] Usually, but not always, the subject who receives the cells (e.g., the recipient) is also the subject from whom the cells are harvested or obtained, which provides the advantage that the donated cells are autologous. However, cells can be obtained from another subject (e.g., a donor), a culture of cells from a donor, or from established cell culture lines. Accordingly, in some embodiments the cells are allogeneic to the recipient. Cells may be obtained from the same or a different species than the subject to be treated, but preferably are of the same species, and more preferably of the same immunological profile as the subject. Such cells can be obtained, for example, from a biological sample comprising cells from a close relative or matched donor, then transfected with nucleic acids (e.g., comprising an engineered retron), and administered to a subject in need of genome modification, for example, for treatment of a disease or condition. [00831] In other embodiments, the engineered retron can be introduced in vivo (e.g., used in gene therapy) by physically delivering the engineered retron to a subject. Examples of physically introducing the engineered retron includes via injections, electroporation and transfection (e.g., calcium-mediated or liposome tranfection, or the like). K. Kits [00832] Also provided are kits comprising engineered retrons (e.g., engineered nucleic acid constructs, or engineered nucleic acid-enzyme constructs) as described herein. [00833] In some embodiments, the kit provides an engineered retron construct or a vector system comprising such a retron construct. In some embodiments, the engineered retron construct, included in the kit, comprises a heterologous sequence capable of providing a cell with a nucleic acid encoding a protein or regulatory RNA of interest, a cellular barcode, a donor polynucleotide suitable for use in gene editing, e.g., by homology directed repair (HDR) or recombination-mediated genetic engineering (recombineering), or a CRISPR protospacer DNA sequence for use in molecular recording. Other agents may also be included in the kit such as transfection agents, host cells, suitable media for culturing cells, buffers, and the like. [00834] In the context of a kit, agents can be provided in liquid or solid form in any convenient packaging (e.g., stick pack, dose pack, etc.). The agents of a kit can be present in the same or separate containers. The kits may contain any one or more of the components described herein in one or more containers. The components may be prepared sterilely, packaged in a syringe and shipped refrigerated. Alternatively it may be housed in a vial or other container for storage. A second container may have other components prepared sterilely. Alternatively the kits may include the active agents premixed and shipped in a vial, tube, or other container. [00835] The kits may have a variety of forms, such as a blister pouch, a shrink wrapped pouch, a vacuum sealable pouch, a sealable thermoformed tray, or a similar pouch or tray form, with the accessories loosely packed within the pouch, one or more tubes, containers, a box or a bag. The kits may be sterilized after the accessories are added, thereby allowing the individual accessories in the container to be otherwise unwrapped. The kits can be sterilized using any appropriate sterilization techniques, such as radiation sterilization, heat sterilization, or other sterilization methods known in the art. The kits may also include other components, depending on the specific application, for example, containers, cell media, salts, buffers, reagents, syringes, needles, a fabric, such as gauze, for applying or removing a disinfecting agent, disposable gloves, a support for the agents prior to administration, etc. Some aspects of this disclosure provide kits comprising a nucleic acid construct comprising a nucleotide sequence encoding the various components of the retron-based editing system described herein. [00836] In addition to the above components, the subject kits may further include (in some embodiments) instructions for practicing the subject methods. These instructions may be present in the subject kits in a variety of forms, one or more of which may be present in the kit. One form in which these instructions may be present is as printed information on a suitable medium or substrate, e.g., a piece or pieces of paper on which the information is printed, in the packaging of the kit, in a package insert, and the like. Yet another form of these instructions is a computer readable medium, e.g., diskette, compact disk (CD), flash drive, and the like, on which the information has been recorded. Yet another form of these instructions that may be present is a website address which may be used via the internet to access the information at a remote site. In some embodiments, information provided on the website is periodically updated to provide, for example, the most up-to-date information. The written instructions may be in a form prescribed by a governmental agency regulating the manufacture, use, or sale of pharmaceuticals or biological products, which can also reflect approval by the agency of manufacture, use or sale for animal administration. As used herein,“promoted” includes all methods of doing business including methods of education, hospital and other clinical instruction, scientific inquiry, drug discovery or development, academic research, pharmaceutical industry activity including pharmaceutical sales, and any advertising or other promotional activity including written, oral and electronic communication of any form, associated with the disclosure. [00837] Other aspects of this disclosure provide kits comprising one or more nucleic acid constructs (e.g., one or more mRNA or circular RNA molecules encoding the components of the retron-based genome editing system) In various embodiments, all nucleic acid constructs can be based on RNA molecules, i.e., and “all-RNA system.” For example, each of the components of the editing system could be expressed from a mRNA molecule, which would be delivered to a target cell by one more more delivery methods (e.g., LNP delivery). L. Cells [00838] One aspect of the disclosure provides an isolated host cell that includes one or more of the compositions described herein, including, but not limited to, engineered retrons and/or retron components, engineered ncRNAs, engineered msDNA, engineered RT, nucleic acid molecules encoding the engineered retrons and/or retron components, and vector or vector systems encoding the engineered retrons and/or retron components, and any combinations thereof. In some embodiments, the host cell is a prokaryotic cell, an archaeal cell, or a eukaryotic host cell. In some embodiments, the eukaryotic host cell is a mammalian cell, such as a human cell, a non-human cell, or a non-human mammalian cell. In some embodiments, the host cell is an artificial cell or genetically modified cell. In some embodiments, the host cell is in vitro, such as a tissue culture cell. In some embodiments, the host cell is within a living host organism. [00839] Cells that may contain any of the compositions described herein. The methods described herein are used to deliver recombinant retrons or components thereof into a eukaryotic cell (e.g., a mammalian cell, such as a human cell). In some embodiments, the cell is in vitro (e.g., cultured cell. In some embodiments, the cell is in vivo (e.g., in a subject such as a human subject). In some embodiments, the cell is ex vivo (e.g., isolated from a subject and may be administered back to the same or a different subject). [00840] The present disclosure contemplates the use of any suitable host cell. For example, the cell host can be a mammalian cell. Mammalian cells of the present disclosure include human cells, primate cells (e.g., vero cells), rat cells (e.g., GH3 cells, OC23 cells) or mouse cells (e.g., MC3T3 cells). There are a variety of human cell lines, including, without limitation, human embryonic kidney (HEK) cells, HeLa cells, cancer cells from the National Cancer Institute's 60 cancer cell lines (NCI60), DU145 (prostate cancer) cells, Lncap (prostate cancer) cells, MCF-7 (breast cancer) cells, MDA-MB-438 (breast cancer) cells, PC3 (prostate cancer) cells, T47D (breast cancer) cells, THP-1 (acute myeloid leukemia) cells, U87 (glioblastoma) cells, SHSY5Y human neuroblastoma cells (cloned from a myeloma) and Saos-2 (bone cancer) cells. In some embodiments, the cells can be human embryonic kidney (HEK) cells (e.g., HEK 293 or HEK 293T cells). In some embodiments, the cells can be stem cells (e.g., human stem cells) such as, for example, pluripotent stem cells (e.g., human pluripotent stem cells including human induced pluripotent stem cells (hiPSCs)). A stem cell refers to a cell with the ability to divide for indefinite periods in culture and to give rise to specialized cells. A pluripotent stem cell refers to a type of stem cell that is capable of differentiating into all tissues of an organism, but not alone capable of sustaining full organismal development. A human induced pluripotent stem cell refers to a somatic (e.g., mature or adult) cell that has been reprogrammed to an embryonic stem cell-like state by being forced to express genes and factors important for maintaining the defining properties of embryonic stem cells (see, e.g., Takahashi and Yamanaka, Cell 126 (4): 663–76, 2006, incorporated by reference herein). Human induced pluripotent stem cells express stem cell markers and are capable of generating cells characteristic of all three germ layers (ectoderm, endoderm, mesoderm). [00841] Some aspects of this disclosure provide cells comprising any of the compositions disclosed herein, including, but not limited to, engineered retrons and/or retron components, engineered ncRNAs, engineered msDNA, engineered RT, nucleic acid molecules encoding the engineered retrons and/or retron components, and vector or vector systems encoding the engineered retrons and/or retron components, and any combinations thereof. In some embodiments, a host cell is transiently or non-transiently transfected with one or more delivery systems described herein, including virus-based systems, virus-like particle systems, and non- virus-base delivery, including LNPs and liposomes. In some embodiments, a cell is transfected as it naturally occurs in a subject. In some embodiments, a cell that is transfected is taken from a subject, i.e., ex vivo transfection. In some embodiments, the cell is derived from cells taken from a subject, such as a cell line. A wide variety of cell lines for tissue culture are known in the art. Examples of cell lines include, but are not limited to, C8161, CCRF-CEM, MOLT, mIMCD- 3, NHDF, HeLa-S3, Huh1, Huh4, Huh7, HUVEC, HASMC, HEKn, HEKa, MiaPaCell, Panc1, PC- 3, TF1, CTLL-2, C1R, Rat6, CV1, RPTE, A10, T24, J82, A375, ARH-77, Calu1, SW480, SW620, SKOV3, SK-UT, CaCo2, P388D1, SEM-K2, WEHI-231, HB56, TIB55, Jurkat, J45.01, LRMB, Bcl-1, BC-3, IC21, DLD2, Raw264.7, NRK, NRK-52E, MRC5, MEF, Hep G2, HeLa B, HeLa T4, COS, COS-1, COS-6, COS-M6A, BS-C-1 monkey kidney epithelial, BALB/3T3 mouse embryo fibroblast, 3T3 Swiss, 3T3-L1, 132-d5 human fetal fibroblasts; 10.1 mouse fibroblasts, 293-T, 3T3, 721, 9L, A2780, A2780ADR, A2780cis, A 172, A20, A253, A431, A- 549, ALC, B16, B35, BCP-1 cells, BEAS-2B, bEnd.3, BHK-21, BR 293. BxPC3. C3H-10T1/2, C6/36, Cal-27, CHO, CHO-7, CHO-IR, CHO-K1, CHO-K2, CHO-T, CHO Dhfr -/-, COR-L23, COR-L23/CPR, COR-L23/5010, COR-L23/R23, COS-7, COV-434, CML T1, CMT, CT26, D17, DH82, DU145, DuCaP, EL4, EM2, EM3, EMT6/AR1, EMT6/AR10.0, FM3, H1299, H69, HB54, HB55, HCA2, HEK-293, HeLa, Hepa1c1c7, HL-60, HMEC, HT-29, Jurkat, JY cells, K562 cells, Ku812, KCL22, KG1, KYO1, LNCap, Ma-Mel 1-48, MC-38, MCF-7, MCF-10A, MDA-MB-231, MDA-MB-468, MDA-MB-435, MDCK II, MDCK 11, MOR/0.2R, MONO- MAC 6, MTD-1A, MyEnd, NCI- H69/CPR, NCI-H69/LX10, NCI-H69/LX20, NCI-H69/LX4, NIH-3T3, NALM-1, NW-145, OPCN/OPCT cell lines, Peer, PNT-1A/PNT 2, RenCa, RIN-5F, RMA/RMAS, Saos-2 cells, Sf-9, SkBr3, T2, T-47D, T84, THP1 cell line, U373, U87, U937, VCaP, Vero cells, WM39, WT-49, X63, YAC-1, YAR, and transgenic varieties thereof. [00842] Cell lines are available from a variety of sources known to those with skill in the art (see, e.g., the American Type Culture Collection (ATCC) (Manassus, Va.)). In some embodiments, a cell transfected with one or more retron delivery systems described herein is used to establish a new cell line comprising one or more nucleic acid molecules encoding the recombinant retron-based gene editing systems described herein, or encoding at last a component of said systems (e.g., a recombinant ncRNA or a recombinant retron RT). M. Pharmaceutical Compositions [00843] The engineered retron-based genome editing systems described herein, or one or more components thereof (e.g., engineered ncRNAs, engineered msDNA, engineered RT, nucleic acid molecules encoding the engineered retrons and/or retron components, guide RNAs, programmable nucleases) may be provided as pharmaceutical compositions. For example, one or more LNPs or other non-virus-based delivery system comprising one or more circular or linear RNA molecules encoding each of the components of the retron-based genome editing system may be formulated as a pharmaceutical composition for administering to a subject in need (e.g., a human in need of gene editing). [00844] Formulations can include, without limitation, saline, liposomes, lipid nanoparticles, polymers, peptides, proteins, cells transfected with viral vectors (e.g., for transfer or transplantation into a subject) and combinations thereof. [00845] Formulations of the pharmaceutical compositions described herein may be prepared by any method known or hereafter developed in the art of pharmacology. As used herein the term “pharmaceutical composition” refers to compositions comprising at least one active ingredient and optionally one or more pharmaceutically acceptable excipients. [00846] In general, such preparatory methods include the step of associating the active ingredient with an excipient and/or one or more other accessory ingredients. As used herein, the phrase “active ingredient” generally refers an engineered retron as described herein. [00847] A pharmaceutical composition in accordance with the present disclosure may be prepared, packaged, and/or sold in bulk, as a single unit dose, and/or as a plurality of single unit doses. As used herein, a “unit dose” refers to a discrete amount of the pharmaceutical composition comprising a predetermined amount of the active ingredient. The amount of the active ingredient is generally equal to the dosage of the active ingredient which would be administered to a subject and/or a convenient fraction of such a dosage such as, for example, one-half or one-third of such a dosage. [00848] Other aspects of the present disclosure relate to pharmaceutical compositions comprising any of the various components of the recombinant retron-based genome editing systems described herein, including, but not limited to, engineered retrons and/or retron components, engineered ncRNAs, engineered msDNA, engineered RT, nucleic acid molecules encoding the engineered retrons and/or retron components, programmable nucleases (e.g., RNA- guided nucleases), guide RNAs, and vector or vector systems encoding the engineered retrons and/or retron components, and any combinations thereof. The term“pharmaceutical composition”, as used herein, refers to a composition formulated for pharmaceutical use. In some embodiments, the pharmaceutical composition further comprises a pharmaceutically acceptable carrier. In some embodiments, the pharmaceutical composition comprises additional agents (e.g. for specific delivery, increasing half-life, or other therapeutic compounds). [00849] As used here, the term“pharmaceutically-acceptable carrier” means a pharmaceutically-acceptable material, composition or vehicle, such as a liquid or solid filler, diluent, excipient, manufacturing aid (e.g., lubricant, talc magnesium, calcium or zinc stearate, or steric acid), or solvent encapsulating material, involved in carrying or transporting the compound from one site (e.g., the delivery site) of the body, to another site (e.g., organ, tissue or portion of the body). A pharmaceutically acceptable carrier is“acceptable” in the sense of being compatible with the other ingredients of the formulation and not injurious to the tissue of the subject (e.g., physiologically compatible, sterile, physiologic pH, etc.). [00850] Some examples of materials which can serve as pharmaceutically-acceptable carriers include: (1) sugars, such as lactose, glucose and sucrose; (2) starches, such as corn starch and potato starch; (3) cellulose, and its derivatives, such as sodium carboxymethyl cellulose, methylcellulose, ethyl cellulose, microcrystalline cellulose and cellulose acetate; (4) powdered tragacanth; (5) malt; (6) gelatin; (7) lubricating agents, such as magnesium stearate, sodium lauryl sulfate and talc; (8) excipients, such as cocoa butter and suppository waxes; (9) oils, such as peanut oil, cottonseed oil, safflower oil, sesame oil, olive oil, corn oil and soybean oil; (10) glycols, such as propylene glycol; (11) polyols, such as glycerin, sorbitol, mannitol and polyethylene glycol (PEG); (12) esters, such as ethyl oleate and ethyl laurate; (13) agar; (14) buffering agents, such as magnesium hydroxide and aluminum hydroxide; (15) alginic acid; (16) pyrogen-free water; (17) isotonic saline; (18) Ringer's solution; (19) ethyl alcohol; (20) pH buffered solutions; (21) polyesters, polycarbonates and/or polyanhydrides; (22) bulking agents, such as polypeptides and amino acids (23) serum component, such as serum albumin, HDL and LDL; (22) C2-C12 alcohols, such as ethanol; and (23) other non-toxic compatible substances employed in pharmaceutical formulations. Wetting agents, coloring agents, release agents, coating agents, sweetening agents, flavoring agents, perfuming agents, preservative and antioxidants can also be present in the formulation. The terms such as“excipient”,“carrier”,“pharmaceutically acceptable carrier” or the like are used interchangeably herein. [00851] In some embodiments, the pharmaceutical composition is formulated for delivery to a subject, e.g., for gene editing. Suitable routes of administrating the pharmaceutical composition described herein include, without limitation: topical, subcutaneous, transdermal, intradermal, intralesional, intraarticular, intraperitoneal, intravesical, transmucosal, gingival, intradental, intracochlear, transtympanic, intraorgan, epidural, intrathecal, intramuscular, intravenous, intravascular, intraosseus, periocular, intratumoral, intracerebral, and intracerebroventricular administration. [00852] In some embodiments, the pharmaceutical composition described herein is administered locally to a diseased site (e.g., tumor site). In some embodiments, the pharmaceutical composition described herein is administered to a subject by injection, by means of a catheter, by means of a suppository, or by means of an implant, the implant being of a porous, non-porous, or gelatinous material, including a membrane, such as a sialastic membrane, or a fiber. [00853] In other embodiments, the pharmaceutical composition described herein is delivered in a controlled release system. In one embodiment, a pump may be used (see, e.g., Langer, 1990, Science 249:1527-1533; Sefton, 1989, CRC Crit. Ref. Biomed. Eng.14:201; Buchwald et al., 1980, Surgery 88:507; Saudek et al., 1989, N. Engl. J. Med.321:574). In another embodiment, polymeric materials can be used. (See, e.g., Medical Applications of Controlled Release (Langer and Wise eds., CRC Press, Boca Raton, Fla., 1974); Controlled Drug Bioavailability, Drug Product Design and Performance (Smolen and Ball eds., Wiley, New York, 1984); Ranger and Peppas, 1983, Macromol. Sci. Rev. Macromol. Chem.23:61. See also Levy et al., 1985, Science 228:190; During et al., 1989, Ann. Neurol.25:351; Howard et al., 1989, J. Neurosurg.71:105). Other controlled release systems are discussed, for example, in Langer, supra. [00854] In some embodiments, the pharmaceutical composition is formulated in accordance with routine procedures as a composition adapted for intravenous or subcutaneous administration to a subject, e.g., a human. In some embodiments, pharmaceutical composition for administration by injection are solutions in sterile isotonic aqueous buffer. Where necessary, the pharmaceutical can also include a solubilizing agent and a local anesthetic such as lignocaine to ease pain at the site of the injection. Generally, the ingredients are supplied either separately or mixed together in unit dosage form, for example, as a dry lyophilized powder or water free concentrate in a hermetically sealed container such as an ampoule or sachette indicating the quantity of active agent. Where the pharmaceutical is to be administered by infusion, it can be dispensed with an infusion bottle containing sterile pharmaceutical grade water or saline. Where the pharmaceutical composition is administered by injection, an ampoule of sterile water for injection or saline can be provided so that the ingredients can be mixed prior to administration. [00855] A pharmaceutical composition for systemic administration may be a liquid, e.g., sterile saline, lactated Ringer’s or Hank’s solution. In addition, the pharmaceutical composition can be in solid forms and re-dissolved or suspended immediately prior to use. Lyophilized forms are also contemplated. [00856] The pharmaceutical composition can be contained within a lipid particle or vesicle, such as a liposome or microcrystal or LNP, which is also suitable for parenteral administration. The particles can be of any suitable structure, such as unilamellar or plurilamellar, so long as compositions are contained therein. Compounds can be entrapped in “stabilized plasmid- lipid particles” (SPLP) containing the fusogenic lipid dioleoylphosphatidylethanolamine (DOPE), low levels (5-10 mol%) of cationic lipid, and stabilized by a polyethyleneglycol (PEG) coating (Zhang Y. P. et al., Gene Ther.1999, 6:1438- 47). Positively charged lipids such as N-[1-(2,3-dioleoyloxi)propyl]-N,N,N-trimethyl- amoniummethylsulfate, or “DOTAP,” are particularly preferred for such particles and vesicles. The preparation of such lipid particles is well known. See, e.g., U.S. Patent Nos.4,880,635; 4,906,477; 4,911,928; 4,917,951; 4,920,016; and 4,921,757; each of which is incorporated herein by reference. [00857] Further, the pharmaceutical composition can be provided as a pharmaceutical kit comprising (a) a container containing a recombinant retron-based genome editing system or one or more components thereof in lyophilized form and (b) a second container containing a pharmaceutically acceptable diluent (e.g., sterile water) for injection. The pharmaceutically acceptable diluent can be used for reconstitution or dilution of the lyophilized system of the invention. Optionally associated with such container(s) can be a notice in the form prescribed by a governmental agency regulating the manufacture, use or sale of pharmaceuticals or biological products, which notice reflects approval by the agency of manufacture, use or sale for human administration. [00858] In another aspect, an article of manufacture containing materials useful for the treatment of the diseases described above is included. In some embodiments, the article of manufacture comprises a container and a label. Suitable containers include, for example, bottles, vials, syringes, and test tubes. The containers may be formed from a variety of materials such as glass or plastic. In some embodiments, the container holds a composition that is effective for treating a disease described herein and may have a sterile access port. For example, the container may be an intravenous solution bag or a vial having a stopper pierce- able by a hypodermic injection needle. The active agent in the composition is a compound of the invention. In some embodiments, the label on or associated with the container indicates that the composition is used for treating the disease of choice. The article of manufacture may further comprise a second container comprising a pharmaceutically-acceptable buffer, such as phosphate-buffered saline, Ringer's solution, or dextrose solution. It may further include other materials desirable from a commercial and user standpoint, including other buffers, diluents, filters, needles, syringes, and package inserts with instructions for use. N. Use and Methods of Use [00859] The engineered retron comprising the heterologous nucleic acid sequence can be used in a variety of applications, several non-limiting examples of which are described herein. In general, the engineered retron can be used in any suitable organism. In some embodiments, the organism is a eukaryote. [00860] In some embodiments, the organism is an animal. In some embodiments, the animal is a fish, an amphibian, a reptile, a mammal, or a bird. In some embodiments, the animal is a farm animal or agriculture animal. Non-limiting examples of farm and agriculture animals include horses, goats, sheep, swine, cattle, llamas, alpacas, and birds, e.g., chickens, turkeys, ducks, and geese. In some embodiments, the animal is a non-human primate, e.g., baboons, capuchin monkeys, chimpanzees, lemurs, macaques, marmosets, tamarins, spider monkeys, squirrel monkeys, and vervet monkeys. In some embodiments, the animal is a pet. Non-limiting examples of pets include dogs, cats horses, wolfs, rabbits, ferrets, gerbils, hamsters, chinchillas, fancy rats, guinea pigs, canaries, parakeets, and parrots. [00861] In some embodiments, the organism is a plant. Plants that may be transfected with an engineered retron include monocots and dicots. Particular examples include, but are not limited to, corn (maize), sorghum, wheat, sunflower, potato, cotton, rice, soybean, sugarbeet, sugarcane, tobacco, barley, and oilseed rape, Brassica sp., alfalfa, rye, millet, safflower, peanuts, sweet potato, cassava, coffee, coconut, pineapple, citrus trees, cocoa, tea, banana, avocado, fig, guava, mango, olive, papaya, cashew, macadamia, almond, oats, vegetables, ornamentals, and conifers. Vegetables include, but are not limited to, crucifers, peppers, tomatoes, lettuce, green beans, lima beans, peas, and members of the genus Cucumis such as cucumber, cantaloupe, and musk melon. Ornamentals include, but are not limited to, azalea, hydrangea, hibiscus, roses, tulips, daffodils, petunias, carnation, poinsettia, and chrysanthemum. [00862] In some embodiments, heterologous nucleic acid sequences can be added to the subject engineered retron to provide a cell with a heterologous nucleic acid encoding a protein or regulatory RNA of interest, a cellular barcode, a donor polynucleotide suitable for use in gene editing, e.g., by homology directed repair (HDR) or recombination-mediated genetic engineering (recombineering), or a CRISPR protospacer DNA sequence for use in molecular recording, as discussed further below. Such heterologous sequences may be inserted, for example, into the msr locus or the msd locus such that the heterologous sequence is transcribed by the retron reverse transcriptase as part of the msDNA product. [00863] In some embodiments, the engineered retrons described herein may be used for research tools, such as kits, functional genomics assays, and generating engineered cell lines and animal models for research and drug screening. The kit may comprise one or more reagents in addition to the engineered retron, such as a buffer, a control reagent, a control vector, a control RNA polynucleotide, a reagent for in vitro production of the polypeptide from DNA, and adaptors for sequencing. A buffer can be, for example, a stabilization buffer, a reconstituting buffer, a diluting buffer, a wash buffer, or a buffer for introducing a polypeptide and/or polynucleotide of the kit into a cell. In some instances, a kit can comprise one or more additional reagents specific for plants. One or more additional reagents for plants can include, for example, soil, nutrients, plants, seeds, spores, Agrobacterium, a T-DNA vector, and a pBINAR vector. Production of Protein or RNA [00864] In some embodiments, the single-stranded msDNA generated by the engineered retron of the invention can be used to produce a desired product of interest in cells. [00865] In some embodiments, the retron is engineered with a heterologous sequence encoding a polypeptide of interest to allow production of the polypeptide from the retron msDNA generated in a cell. The polypeptide of interest may be any type of protein/peptide including, without limitation, an enzyme, an extracellular matrix protein, a receptor, transporter, ion channel, or other membrane protein, a hormone, a neuropeptide, an antibody, or a cytoskeletal protein, a functional fragment thereof, or a biologically active domain of interest. In some embodiments, the protein is a therapeutic protein, therapeutic antibody for use in treatment of a disease, or a template to fix a mutation or mutated exon in the genome. [00866] Non-limiting examples of polypeptides of interest include: growth hormones, insulin-like growth factors (IGF-1), Fat-1, Phytase, xylanase, beta-glucanase, Lysozyme or lysostaphin, Histone deacetylase such as HDAC6, CD163, etc. [00867] In other embodiments, the retron is engineered with a heterologous sequence encoding an RNA of interest to allow production of the RNA from the retron in a cell. The RNA of interest may be any type of RNA including, without limitation, a RNA interference (RNAi) nucleic acid or regulatory RNA such as, but not limited to, a microRNA (miRNA), a small interfering RNA (siRNA), a short hairpin RNA (shRNA), a small nuclear RNA (snRNA), a long non-coding RNA (IncRNA), an antisense nucleic acid, and the like. Gene Editing [00868] In some embodiments, the retron is used for genome editing a desired site. A retron is engineered with a heterologous nucleic acid sequence encoding a donor polynucleotide suitable for use with nuclease genome editing system. The nuclease is designed to specifically target a location proximal to the desired edit (the nuclease should be designed such that it will not cut the target once the edit is properly installed). The nuclease (e.g., CAS or non-CAS) is linked to the retron, either by direct fusion to the RT or by fusion of the msDNA to the gRNA (only applicable for RNA-guided nucleases). A heterologous nucleic acid sequence is inserted into the retron msd. See for example FIG.3, which shows a marker representing the edit. [00869] In some embodiments, the heterologous nucleic acid sequence has 10-100 or more bp of homologous nucleic acid sequence to the genome on both sides of the desired edit. The desired edit (insertion, deletion, or mutation) is in between the homologous sequence. [00870] In some embodiments, donor polynucleotides comprise a sequence comprising an intended genome edit flanked by a pair of homology arms responsible for targeting the donor polynucleotide to the target locus to be edited in a cell. The donor polynucleotide typically comprises a 5ʹ homology arm that hybridizes to a 5ʹ genomic target sequence and a 3ʹ homology arm that hybridizes to a 3ʹ genomic target sequence. The homology arms are referred to herein as 5ʹ and 3ʹ (i.e., upstream and downstream) homology arms, which relate to the relative position of the homology arms to the nucleotide sequence comprising the intended edit within the donor polynucleotide. The 5ʹ and 3ʹ homology arms hybridize to regions within the target locus in the genomic DNA to be modified, which are referred to herein as the “5ʹ target sequence” and “3ʹ target sequence,” respectively. [00871] The homology arm must be sufficiently complementary for hybridization to the target sequence to mediate homologous recombination between the donor polynucleotide and genomic DNA at the target locus. For example, a homology arm may comprise a nucleotide sequence having at least about 80-100% sequence identity to the corresponding genomic target sequence, including any percent identity within this range, such as at least 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% sequence identity thereto, wherein the nucleotide sequence comprising the intended edit can be integrated into the genomic DNA by HDR at the genomic target locus recognized (i.e., having sufficient complementary for hybridization) by the 5ʹ and 3ʹ homology arms. [00872] In some embodiments, the corresponding homologous nucleotide sequences in the genomic target sequence (i.e., the “5ʹ target sequence” and “3ʹ target sequence”) flank a specific site for cleavage and/or a specific site for introducing the intended edit. The distance between the specific cleavage site and the homologous nucleotide sequences (e.g., each homology arm) can be several hundred nucleotides. In some embodiments, the distance between a homology arm and the cleavage site is 200 nucleotides or less (e.g., 0, 10, 20, 30, 50, 75, 100, 125, 150, 175, and 200 nucleotides). In most cases, a smaller distance may give rise to a higher gene targeting rate. In some embodiments, the donor polynucleotide is substantially identical to the target genomic sequence, across its entire length except for the sequence changes to be introduced to a portion of the genome that encompasses both the specific cleavage site and the portions of the genomic target sequence to be altered. [00873] A homology arm can be of any length, e.g.10 nucleotides or more, 15 nucleotides or more, 20 nucleotides or more, 50 nucleotides or more, 100 nucleotides or more, 250 nucleotides or more, 300 nucleotides or more, 350 nucleotides or more, 400 nucleotides or more, 450 nucleotides or more, 500 nucleotides or more, 1000 nucleotides (1 kb) or more, 5000 nucleotides (5 kb) or more, 10000 nucleotides (10 kb) or more, etc. In some instances, the 5ʹ and 3ʹ homology arms are substantially equal in length to one another. However, in some instances the 5ʹ and 3ʹ homology arms are not necessarily equal in length to one another. For example, one homology arm may be 30% shorter or less than the other homology arm, 20% shorter or less than the other homology arm, 10% shorter or less than the other homology arm, 5% shorter or less than the other homology arm, 2% shorter or less than the other homology arm, or only a few nucleotides less than the other homology arm. In other instances, the 5ʹ and 3ʹ homology arms are substantially different in length from one another, e.g. one may be 40% shorter or more, 50% shorter or more, sometimes 60% shorter or more, 70% shorter or more, 80% shorter or more, 90% shorter or more, or 95% shorter or more than the other homology arm. [00874] The donor polynucleotide may be used in combination with an RNA-guided nuclease, which is targeted to a particular genomic sequence (i.e., genomic target sequence to be modified) by a guide RNA. A target-specific guide RNA comprises a nucleotide sequence that is complementary to a genomic target sequence, and thereby mediates binding of the nuclease- gRNA complex by hybridization at the target site. For example, the gRNA can be designed with a sequence complementary to the sequence of a minor allele to target the nuclease-gRNA complex to the site of a mutation. The mutation may comprise an insertion, a deletion, or a substitution. For example, the mutation may include a single nucleotide variation, gene fusion, translocation, inversion, duplication, frameshift, missense, nonsense, or other mutation associated with a phenotype or disease of interest. The targeted minor allele may be a common genetic variant or a rare genetic variant. In some embodiments, the gRNA is designed to selectively bind to a minor allele with single base-pair discrimination, for example, to allow binding of the nuclease-gRNA complex to a single nucleotide polymorphism (SNP). In particular, the gRNA may be designed to target disease-relevant mutations of interest for the purpose of genome editing to remove the mutation from a gene. Alternatively, the gRNA can be designed with a sequence complementary to the sequence of a major or wild-type allele to target the nuclease-gRNA complex to the allele for the purpose of genome editing to introduces a mutation into a gene in the genomic DNA of the cell, such as an insertion, deletion, or substitution. Such genetically modified cells can be used, for example, to alter phenotype, confer new properties, or produce disease models for drug screening. [00875] In some embodiments, the RNA-guided nuclease used for genome modification is a clustered regularly interspersed short palindromic repeats (CRISPR) system Cas nuclease. Any RNA-guided Cas nuclease capable of catalyzing site- directed cleavage of DNA to allow integration of donor polynucleotides by the HDR mechanism can be used in genome editing, including CRISPR system Class 1, Type I, II, or III Cas nucleases; Class 2, Type II nuclease (such as Cas9); a Class 2, Type V nuclease (such as Cpfl), or a Class 2, Type VI nuclease (such as C2c2). Examples of Cas proteins include Casl, CaslB, Cas2, Cas3, Cas4, Cas5, Cas5e (CasD), Cas6, Cas6e, Cas6f, Cas7, Cas8al, Cas8a2, Cas8b, Cas8c, Cas9 (Csnl or Csxl2), CaslO, CaslOd, CasF, CasG, CasH, Csyl, Csy2, Csy3, Csel (CasA), Cse2 (CasB), Cse3 (CasE), Cse4 (CasC), Cscl, Csc2, Csa5, Csn2, Csm2, Csm3, Csm4, Csm5, Csm6, Cmrl, Cmr3, Cmr4, Cmr5, Cmr6, Csbl, Csb2, Csb3, Csxl7, Csxl4, CsxlO, Csxl6, CsaX, Csx3, Csxl, Csxl5, Csfl, Csf2, Csf3, Csf4, and Cul966, and homologs or modified versions thereof. [00876] In some embodiments, a Class 1, type II CRISPR system Cas9 endonuclease is used. Cas9 nucleases from any species, or biologically active fragments, variants, analogs, or derivatives thereof that retain Cas9 endonuclease activity (i.e., catalyze site-directed cleavage of DNA to generate double-strand breaks) may be used to perform genome modification as described herein. The Cas9 need not be physically derived from an organism but may be synthetically or recombinantly produced. Cas9 sequences from a number of bacterial species are well known in the art and listed in the National Center for Biotechnology Information (NCBI) database. See, for example, NCBI entries for Cas9 from: Streptococcus pyogenes (WP 002989955, WP_038434062, WP_011528583); Campylobacter jejuni (WP_022552435, YP 002344900), Campylobacter coli (WP 060786116); Campylobacter fetus (WP 059434633); Corynebacterium ulcerans (NC_015683, NC_017317); Corynebacterium diphtheria (NC_016782, NC_016786); Enterococcus faecalis (WP 033919308); Spiroplasma syrphidicola (NC 021284); Prevotella intermedia (NC 017861); Spiroplasma taiwanense (NC 021846); Streptococcus iniae (NC 021314); Belliella baltica (NC 018010); Psychroflexus torquisl (NC O 18721); Streptococcus thermophilus (YP 820832), Streptococcus mutans (WP 061046374, WP 024786433); Listeria innocua (NP 472073); Listeria monocytogenes (WP 061665472); Legionella pneumophila (WP 062726656); Staphylococcus aureus (WP_001573634); Francisella tularensis (WP_032729892, WP_014548420), Enterococcus faecalis (WP 033919308); Lactobacillus rhamnosus (WP 048482595, WP_032965177); and Neisseria meningitidis (WP_061704949, YP_002342100); all of which sequences (as entered by the date of filing of this application) are herein incorporated by reference in their entireties. Any of these sequences or a variant thereof comprising a sequence having at least about 70-100% sequence identity thereto, including any percent identity within this range, such as 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98, or 99% sequence identity thereto, can be used for genome editing, as described herein. See also Fonfara et al. (2014) Nucleic Acids Res.42(4):2577-90; Kapitonov et al. (2015) J. Bacterid.198(5): 797- 807, Shmakov et al. (2015) Mol. Cell.60(3):385- 397, and Chylinski et al. (2014) Nucleic Acids Res.42(10):6091-6105); for sequence comparisons and a discussion of genetic diversity and phylogenetic analysis of Cas9. [00877] The genomic target site will typically comprise a nucleotide sequence that is complementary to the gRNA and may further comprise a protospacer adjacent motif (PAM). In some embodiments, the target site comprises 20-30 base pairs in addition to a 3 or more base pair PAM. Typically, the first nucleotide of a PAM can be any nucleotide, while the two or more other nucleotides will depend on the specific Cas9 protein that is chosen. Exemplary PAM sequences are known to those of skill in the art and include, without limitation, NNG, NGN, NAG, and NGG, wherein N represents any nucleotide. In some embodiments, the allele targeted by a gRNA comprises a mutation that creates a PAM within the allele, wherein the PAM promotes binding of the Cas9-gRNA complex to the allele. [00878] In some embodiments, the gRNA is 5-50 nucleotides, 10-30 nucleotides, 15- 25 nucleotides, 18-22 nucleotides, or 19-21 nucleotides in length, or any length between the stated ranges, including, for example, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, or 35 nucleotides in length. The guide RNA may be a single guide RNA comprising crRNA and tracrRNA sequences in a single RNA molecule, or the guide RNA may comprise two RNA molecules with crRNA and tracrRNA sequences residing in separate RNA molecules. [00879] In another embodiment, the CRISPR nuclease from Prevotella and Francisella 1 (Cpfl, or Cas12a) is used. Cpfl is another class II CRISPR/Cas system RNA-guided nuclease with similarities to Cas9 and may be used analogously. Unlike Cas9, Cpfl does not require a tracrRNA and only depends on a crRNA in its guide RNA, which provides the advantage that shorter guide RNAs can be used with Cpfl for targeting than Cas9. Cpfl is capable of cleaving either DNA or RNA. The PAM sites recognized by Cpfl have the sequences 5ʹ-YTN-3ʹ (where “Y” is a pyrimidine and “N” is any nucleobase) or 5ʹ-TTN-3ʹ, in contrast to the G-rich PAM site recognized by Cas9. Cpfl cleavage of DNA produces double-stranded breaks with a sticky-ends having a 4 or 5 nucleotide overhang. For a discussion of Cpfl, see, e.g., Ledford et al. (2015) Nature.526 (7571):17-17, Zetsche et al. (2015) Cell.163 (3):759-771, Murovec et al. (2017) Plant Biotechnol. J.15(8):917-926, Zhang et al. (2017) Front. Plant Sci.8:177, Fernandes et al. (2016) Postepy Biochem.62(3):315-326; herein incorporated by reference. [00880] C2c1 (Cas12b) is another class II CRISPR/Cas system RNA-guided nuclease that may be used. C2cl, similarly to Cas9, depends on both a crRNA and tracrRNA for guidance to target sites. See, e.g., Shmakov et al. (2015) Mol Cell.60(3):385-397, Zhang et al. (2017) Front Plant Sci.8:177; herein incorporated by reference. [00881] In yet another embodiment, an engineered RNA-guided Fokl nuclease may be used. RNA-guided Fokl nucleases comprise fusions of inactive Cas9 (dCas9) and the Fokl endonuclease (FokI-dCas9), wherein the dCas9 portion confers guide RNA-dependent targeting on Fokl. For a description of engineered RNA-guided Fold nucleases, see, e.g., Havlicek et al. (2017) Mol. Ther.25(2):342-355, Pan et al. (2016) Sci Rep.6:35794, Tsai et al. (2014) Nat Biotechnol.32(6):569-576; herein incorporated by reference. [00882] In other embodiments, any other Cas enzymes and variants described in other sections of the application (all incorporated herein) can be used similarly. [00883] In some embodiments, the RNA-guided nuclease is provided in the form of a protein, optionally where the nuclease is complexed with a gRNA to form a ribonucleoprotein (RNP) complex. In some embodiments, the RNA-guided nuclease is provided by a nucleic acid encoding the RNA-guided nuclease, such as an RNA (e.g., messenger RNA) or DNA (expression vector). In some embodiments, the RNA-guided nuclease and the gRNA are both provided by vectors, such as the vectors and the vector system described in other parts of the application (all incorporated herein by reference). Both can be expressed by a single vector or separately on different vectors. The vectors encoding the RNA-guided nuclease and gRNA may be included in the vector system comprising the engineered retron msr gene, msd gene and ret gene sequences. In some embodiments, the RNA-guided nuclease is fused to the RT and/or the msDNA. [00884] The RNP complex may be administered to a subject or delivered into a cell by methods known in the art, such as those described in U.S. Pat. No.11,390,884, which is incorporated by reference herein in its entirety. In some embodiments, the endonuclease/gRNA ribonucleoprotein (RNP) complexes are delivered to cells by electroporation. Direct delivery of the RNP complex to a subject or cell eliminates the need for expression from nucleic acids (e.g., transfection of plasmids encoding Cas9 and gRNA). It also eliminates unwanted integration of DNA segments derived from nucleic acid delivery (e.g., transfection of plasmids encoding Cas9 and gRNA). An endonuclease/gRNA ribonucleoprotein (RNP) complex usually is formed prior to administration. [00885] Codon usage may be optimized to further improve production of an RNA-guided nuclease and/or reverse transcriptase (RT) in a particular cell or organism. For example, a nucleic acid encoding an RNA-guided nuclease or reverse transcriptase can be modified to substitute codons having a higher frequency of usage in a yeast cell, a bacterial cell, a human cell, a non-human cell, a mammalian cell, a rodent cell, a mouse cell, a rat cell, or any other host cell of interest, as compared to the naturally occurring polynucleotide sequence. When a nucleic acid encoding the RNA-guided nuclease or reverse transcriptase is introduced into cells, the protein can be transiently, conditionally, or constitutively expressed in the cell. [00886] In some embodiments, the engineered retron used for genome editing with nuclease genome editing systems can further include accessory or enhancer proteins for recombination. Examples of recombination enhancers can include nonhomologous end joining (NHEJ) inhibitors (e.g., inhibitor of DNA ligase IV, a KU inhibitor (e.g., KU70 or KU80), a DNA-PKc inhibitor, or an artemis inhibitor) and homologous directed repair (HDR) promoters, or both, that can enhance or improve more precise genome editing and/or the efficiency of homologous recombination. In some embodiments, the recombination accessory or enhancers can comprise C-terminal binding protein interacting protein (CtIP), cyclinB2, Rad family members (e.g. Rad50, Rad51, Rad52, etc). [00887] CtIP is a transcription factor containing C2H2 zinc fingers that are involved in early steps of homologous recombination. Mammalian CtIP and its orthologs in other eukaryotes promote the resection of DNA double-strand breaks and are essential for meiotic recombination. HDR may be enhanced by using Cas9 nuclease associated (e.g. fused) to an N-terminal domain of CtIP, an approach that forces CtIP to the cleavage site and increases transgene integration by HDR. In some embodiments, an N-terminal fragment of CtIP, called HE for HDR enhancer, may be sufficient for HDR stimulation and requires the CtIP multimerization domain and CDK phosphorylation sites to be active. HDR stimulation by the Cas9-HE fusion depends on the guide RNA used, and therefore the guide RNA will be designed accordingly. [00888] Using the gene editing system described herein, any target gene or sequence in a host cell can be edited or modified for a desired trait, including but not limited to: Myostatin (e.g., GDF8) to increase muscle growth; Pc POLLED to induce hairlessness; KISS1R to induce bore taint; Dead end protein (dnd) to induce sterility; Nano2 and DDX to induce sterility; CD163 to induce PRRSV resistance; RELA to induce ASFV resilience; CD18 to induce Mannheimia (Pasteurella) haemolytica resilience; NRAMPl to induce tuberculosis resilience; Negative regulators of muscle mass (e.g., Myostatin) to increase muscle mass. Recombineering [00889] Recombineering (recombination-mediated genetic engineering) can be used in modifying chromosomal as well as episomal replicons in cells, for example, to create gene replacements, gene knockouts, deletions, insertions, inversions, or point mutations. Recombineering can also be used to modify a plasmid or bacterial artificial chromosome (BAC), for example, to clone a gene or insert markers or tags. [00890] The engineered retrons described herein can be used in recombineering applications to provide linear single-stranded or double-stranded DNA for recombination. Homologous recombination may be mediated by bacteriophage proteins such as RecE/RecT from Rac prophage or Redobd from bacteriophage lambda. The linear DNA should have sufficient homology at the 5ʹ and 3ʹ ends to a target DNA molecule present in a cell (e.g., plasmid, BAC, or chromosome) to allow recombination. [00891] The linear double-stranded or single-stranded DNA molecule used in recombineering (i.e. donor polynucleotide) comprises a sequence having the intended edit to be inserted flanked by two homology arms that target the linear DNA molecule to a target site for homologous recombination. Homology arms for recombineering typically range in length from 13-300 nucleotides, or 20 to 200 nucleotides, including any length within this range such as 13, 14, 15, 16, 17, 18, 19, 20, 22, 24, 26, 28, 30, 32, 34, 36, 38, 40, 42, 44, 46, 48, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, 100, 105, 110, 115, 120, 125, 130, 135, 140, 145, 150, 155, 160, 165, 170, 175, 180, 185, 190, 195, or 200 nucleotides in length. In some embodiments, a homology arm is at least 15, at least 20, at least 30, at least 40, or at least 50 or more nucleotides in length. Homology arms ranging from 40-50 nucleotides in length generally have sufficient targeting efficiency for recombination; however, longer homology arms ranging from 150 to 200 bases or more may further improve targeting efficiency. In some embodiments, the 5ʹ homology arm and the 3ʹ homology arm differ in length. For example, the linear DNA may have about 50 bases at the 5ʹ end and about 20 bases at the 3ʹ end with homology to the region to be targeted. [00892] The bacteriophage homologous recombination proteins can be provided to a cell as proteins or by one or more vectors encoding the recombination proteins, such as the vector or vector system. In some embodiments, one or more vectors encoding the bacteriophage recombination proteins are included in the vector system comprising the engineered retron msr gene, msd gene, and/or ret gene sequences. Additionally, a number of bacterial strains containing prophage recombination systems are available for recombineering, including, without limitation, DY380, containing a defective l prophage with recombination proteins exo, bet, and gam; EL250, derived from DY380, which in addition to the recombination genes found in DY380, also contains a tightly controlled arabinose-inducible flpe gene (flpe mediates recombination between two identical frt sites); EL350, also derived from DY380, which in addition to the recombination genes found in DY380, also contains a tightly controlled arabinose-inducible ere gene (ere mediates recombination between two identical loxP sites; SW102, derived from DY380, which is designed for BAC recombineering using a galK positive/negative selection; SW105, derived from EL250, which can also be used for galK positive/negative selection, but like EL250, contain an ara-inducible Flpe gene; and SW106, derived from EL350, which can be used for galK positive/negative selection, but like EL350, contains an ara-inducible Cre gene. Recombineering can be carried out by transfecting bacterial cells of such strains with an engineered retron comprising a heterologous sequence encoding a linear DNA suitable for recombineering. For a discussion of recombineering systems and protocols, see, e.g., Sharan et al. (2009) Nat Protoc.4(2): 206-223, Zhang et al. (1998) Nature Genetics 20: 123-128, Muyrers et al. (1999) Nucleic Acids Res.27: 1555-1557, Yu et al. (2000) Proc. Natl. Acad. Sci U.S.A.97 (11):5978-5983; herein incorporated by reference. Molecular Recording [00893] In some embodiments, the heterologous sequence in the engineered retron construct comprises a synthetic CRISPR protospacer DNA sequence to allow molecular recording. The endogenous CRISPR Cas1-Cas2 system is normally utilized by bacteria and archaea to keep track of foreign DNA sequences originating from viral infections by storing short sequences (i.e., protospacers) that confer sequence-specific resistance to invading viral nucleic acids within genome-based arrays. These arrays not only preserve the spacer sequences but also record the order in which the sequences are acquired, generating a temporal record of acquisition events. [00894] This system can be adapted to record arbitrary DNA sequences into a genomic CRISPR array in the form of “synthetic protospacers” that are introduced into cells using engineered retrons. Engineered retrons carrying the protospacer sequences can be used for integration of synthetic CRISPR protospacer sequences at a specific genomic locus by utilizing the CRISPR system Cas1-Cas2 complex. Molecular recording can be used to keep track of certain biological events by producing a stable genetic memory tracking code. See, e.g., Shipman et al. (2016) Science 353(6298): aafl 175 and International Patent Application Publication No. WO/2018/191525; herein incorporated by reference in their entireties. [00895] In some embodiments, the CRISPR-Cas system is harnessed to record specific and arbitrary DNA sequences into a bacterial genome. The DNA sequences can be produced by an engineered retron within the cell. For example, the engineered retron can be used to produce the protospacers within the cell, which are inserted into a CRISPR array within the cell. The cell may be modified to include one or more engineered returns (or vector systems encoding them) that can produce one or more synthetic protospacers in the cell, wherein the synthetic protospacers are added to the CRISPR array. A record of defined sequences, recorded over many days, and in multiple modalities can be generated. [00896] In some embodiments, the engineered retron comprises an msd protospacer nucleic acid region or an msr protospacer nucleic acid region. In the case of a msr protospacer nucleic acid region, the protospacer sequence is first incorporated into the msr RNA, which is reverse transcribed into protospacer DNA. Double stranded protospacer DNA is produced when two complementary protospacer DNA sequences having complementary sequences hybridize, or when a double-stranded structure (such as a hairpin) is formed in a single stranded protospacer DNA (e.g., a single msDNA can form an appropriate hairpin structure to provide the double stranded DNA protospacer). [00897] In some embodiments, a single stranded DNA produced in vivo from a first engineered retron may be hybridized with a complementary single-stranded DNA produced in vivo from the same retron or a second engineered retron or may form a hairpin structure and then used as a protospacer sequence to be inserted into a CRISPR array as a spacer sequence. The engineered retron(s) should provide sufficient levels of the protospacer sequence within a cell for incorporation into the CRISPR array. The use of protospacers generated within the cell extends the in vivo molecular recording system from only capturing information known to a user, to capturing biological or environmental information that may be previously unknown to a user. For example, an msDNA protospacer sequence in an engineered retron construct may be driven by a promoter that is downstream of a sensor pathway for a biological phenomenon or environmental toxin. The capture and storage of the protospacer sequence in the CRISPR array records the event. If multiple msDNA protospacers are driven by different promoters, the activity of those promoters is recorded (along with anything that may be upstream of the promoters) as well as the relative order of promoter activity (based on the relative position of spacer sequences in the CRISPR array). At any point after the recording has taken place, the CRISPR array may be sequenced to determine whether a given biological or environmental event has taken place and the order of multiple events, given by the presence and relative position of msDNA-derived spacers in the CRISPR array. [00898] In some embodiments, the synthetic protospacer further comprises an AAG PAM sequence at its 5ʹ end. Protospacers including the 5ʹ AAG PAM are acquired by the CRISPR array with greater efficiency than those that do not include a PAM sequence. [00899] In some embodiments, Cas1 and Cas2 are provided by a vector that expresses the Cas1 and Cas2 at a level sufficient to allow the synthetic protospacer sequences produced by engineered retrons to be acquired by a CRISPR array in a cell. Such a vector system can be used to allow molecular recording in a cell that lacks endogenous Cas proteins. Therapeutic Applications [00900] Also provided herein are methods of diagnosing, prognosing, treating, and/or preventing a disease, state, or condition in or of a subject, using the engineered retron of the invention. [00901] Generally, the methods of diagnosing, prognosing, treating, and/or preventing a disease, state, or condition in or of a subject can include modifying a polynucleotide in a subject or cell thereof using a composition, system, or component thereof of the engineered retron as described herein, and/or include detecting a diseased or healthy polynucleotide in a subject or cell thereof using a composition, system, or component thereof of the engineered retron as described herein. [00902] In some embodiments, the method of treatment or prevention can include using a composition, system, or component of the engineered retron to modify a polynucleotide of an infectious organism (e.g. bacterial or virus) within a subject or cell thereof. [00903] In some embodiments, the method of treatment or prevention can include using a composition, system, or component of the engineered retron to modify a polynucleotide of an infectious organism or symbiotic organism within a subject. [00904] In some embodiments, the composition, system, and components of the engineered retron can be used to develop models of diseases, states, or conditions. [00905] In some embodiments, the composition, system, and components of the engineered retron can be used to detect a disease state or correction thereof, such as by a method of treatment or prevention described herein. [00906] In some embodiments, the composition, system, and components of the engineered retron can be used to screen and select cells that can be used, for example, as treatments or preventions described herein. [00907] In some embodiments, the composition, system, and components thereof can be used to develop biologically active agents that can be used to modify one or more biologic functions or activities in a subject or a cell thereof. [00908] In general, the method can include delivering a composition, system, and/or component of the engineered retron to a subject or cell thereof, or to an infectious or symbiotic organism by a suitable delivery technique and/or composition. Once administered, the components can operate as described elsewhere herein to elicit a nucleic acid modification event. In some embodiments, the nucleic acid modification event can occur at the genomic, epigenomic, and/or transcriptomic level. DNA and/or RNA cleavage, gene activation, and/or gene deactivation can occur. [00909] The composition, system, and components of the engineered retron as described elsewhere herein can be used to treat and/or prevent a disease, such as a genetic and/or epigenetic disease, in a subject; to treat and/or prevent genetic infectious diseases in a subject, such as bacterial infections, viral infections, fungal infections, parasite infections, and combinations thereof; to modify the composition or profile of a microbiome in a subject, which can in turn modify the health status of the subject; to modify cells ex vivo, which can then be administered to the subject whereby the modified cells can treat or prevent a disease or symptom thereof; or to treat mitochondrial diseases, where the mitochondrial disease etiology involves a mutation in the mitochondrial DNA. [00910] Also provided is a method of treating a subject, e.g., a subject in need thereof, comprising inducing gene editing by transforming the subject with the polynucleotide encoding one or more components of the composition, system, or complex or any of polynucleotides or vectors described herein of the engineered retron, and administering them to the subject. [00911] Also provided is a method of treating a subject, e.g., a subject in need thereof, comprising inducing transcriptional activation or repression of multiple target gene loci by transforming the subject with the polynucleotides or vectors described herein, wherein said polynucleotide or vector encodes or comprises one or more components of composition, system, complex or component of the engineered retron, and comprising multiple Cas effectors. [00912] Also provided is a method of treating a subject, e.g., a subject in need thereof, comprising inducing gene editing by transforming the subject with the Cas effector(s), and encoding and expressing in vivo the remaining portions of the composition, system, (e.g., RNA, guides), complex or component of the engineered retron. A suitable repair template may also be provided by the engineered retron as described herein elsewhere. [00913] Also provided is a method of treating a subject, e.g., a subject in need thereof, comprising inducing transcriptional activation or repression by transforming the subject with the systems or compositions herein. [00914] Also provided is a method of inducing one or more polynucleotide modifications in a eukaryotic or prokaryotic cell or component thereof (e.g. a mitochondria) of a subject, infectious organism, and/or organism of the microbiome of the subject. The modification can include the introduction, deletion, or substitution of one or more nucleotides at a target sequence of a polynucleotide of one or more cell(s). The modification can occur in vitro, ex vivo, in situ, or in vivo. [00915] In some embodiments, the method of treating or inhibiting a condition or a disease caused by one or more mutations in a genomic locus in a eukaryotic organism or a non- human organism can include manipulation of a target sequence within a coding, non-coding or regulatory element of said genomic locus in a target sequence in a subject or a non-human subject in need thereof comprising modifying the subject or a non -human subject by manipulation of the target sequence and wherein the condition or disease is susceptible to treatment or inhibition by manipulation of the target sequence including providing treatment comprising delivering a composition comprising the particle delivery system or the delivery system or the virus particle of any one of the above embodiment or the cell of any one of the above embodiment. [00916] Also provided herein is the use of the particle delivery system or the delivery system or the virus vector (in viral particle) of any one of the above embodiment or the cell of any one of the above embodiment in ex vivo or in vivo gene or genome editing; or for use in in vitro, ex vivo or in vivo gene therapy. [00917] Also provided herein are particle delivery systems, non-viral delivery systems, and/or the virus particle of any one of the above embodiments or the cell of any one of the above embodiments used in the manufacture of a medicament for in vitro, ex vivo or in vivo gene or genome editing or for use in in vitro, ex vivo or in vivo gene therapy or for use in a method of modifying an organism or a non-human organism by manipulation of a target sequence in a genomic locus associated with a disease or in a method of treating or inhibiting a condition or disease caused by one or more mutations in a genomic locus in a eukaryotic organism or a non- human organism. [00918] In some embodiments, target polynucleotide modification using the subject engineered retron and the associated composition, vectors, system and methods comprises addition, deletion, or substitution of 1-about 10k nucleotides at each target sequence of said polynucleotide of said cell(s). The modification can include the addition, deletion, or substitution of at least 1, 5, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 35, 40, 45, 50, 75, 100, 200, 250, 300, 500, 1000, 1500, 2000, 2500, 3000, 3500, 4000, 5000, 6000, 7000, 8000, 9000, 10,000 or more nucleotides at each target sequence. [00919] In some embodiments, formation of system or complex results in cleavage, nicking, and/or another modification of one or both strands in or near (e.g. within 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 20, 50, or more base pairs from) the target sequence. [00920] In some embodiments, a method of modifying a target polynucleotide in a cell to treat or prevent a disease can include allowing a composition, system, or component of the subject engineered retron to bind to the target polynucleotide, e.g., to effect cleavage, nicking, or other modification as the composition, system, is capable of said target polynucleotide, thereby modifying the target polynucleotide, wherein the composition, system, or component thereof, complex with a guide sequence, and hybridize said guide sequence to a target sequence within the target polynucleotide, wherein said guide sequence is optionally linked to a tracr mate sequence, which in turn can hybridize to a tracr sequence. In some embodiments, modification can include cleaving or nicking one or two strands at the location of the target sequence by one or more components of the composition, system, or component thereof. [00921] In some embodiments, the engineered retron and the associated compositions, systems, vectors, uses, and methods of use, can be used to treat diseases of the circulatory system. In some embodiments, the treatment can be carried out by using an AAV or a lentiviral vector to deliver the engineered retron, composition, system, and/or vector described herein to modify hematopoietic stem cells (HSCs) or iPSCs in vivo or ex vivo. In some embodiments, the treatment can be carried out by correcting HSCs or iPSCs as to the disease using a composition, system, herein or a component thereof, wherein the composition, system, optionally includes a suitable HDR repair template (e.g., a template in the msDNA of the engineered retron). [00922] In some embodiments, the treatment or prevention for treating a circulatory system or blood disease can include modifying a human cord blood cell. In some embodiments, the treatment or prevention for treating a circulatory system or blood disease can include modifying a granulocyte colony-stimulating factor-mobilized peripheral blood cell (mPB) with any modification described herein. In some embodiments, the human cord blood cell or mPB can be CD34+. In some embodiments, the cord blood cells or mPB cells modified are autologous. In some embodiments, the cord blood cells or mPB cells are allogenic. In addition to the modification of the disease genes, allogenic cells can be further modified using the composition, system, described herein to reduce the immunogenicity of the cells when delivered to the recipient. The modified cord blood cells or mPB cells can be optionally expanded in vitro. The modified cord blood cell(s) or mPB cells can be derived to a subject in need thereof using any suitable delivery technique. [00923] The composition and system may be engineered to target genetic locus or loci in HSCs. In some embodiments, the components of the systems can be codon-optimized for a eukaryotic cell and especially a mammalian cell, e.g., a human cell, for instance, HSC, or iPSC and sgRNA targeting a locus or loci in HSC, such as circulatory disease, can be prepared. These may be delivered via particles, such as the lipid nanoparticle delivery system described herein. The particles may be formed by the components of the systems herein being admixed. [00924] In some embodiments, after ex vivo modification the HSCs or iPCS can be expanded prior to administration to the subject. Expansion of HSCs can be via any suitable method such as that described by, Lee, “Improved ex vivo expansion of adult hematopoietic stem cells by overcoming CUL4-mediated degradation of HOXB4.” Blood.2013 May 16;121(20):4082-9. doi: 10.1182/blood-2012-09-455204. Epub 2013 Mar 21. [00925] In some embodiments, the HSCs or iPSCs modified are autologous. In some embodiments, the HSCs or iPSCs are allogenic. In addition to the modification of the disease genes, allogenic cells can be further modified using the composition, system, described herein to reduce the immunogenicity of the cells when delivered to the recipient. [00926] In some embodiments, the engineered retron and the associated compositions, systems, vectors, uses, and methods of use, can be used to treat neurological diseases. In some embodiments, the neurological diseases comprise diseases of the brain and CNS. [00927] Delivery options for the diseases in the brain include encapsulation of the systems in the form of either DNA or RNA into liposomes and conjugating to molecular Trojan horses for trans-blood brain barrier (BBB) delivery. Molecular Trojan horses have been shown to be effective for delivery of B-gal expression vectors into the brain of non-human primates. The same approach can be used to delivery vectors or vector systems of the invention. In other embodiments, an artificial virus can be generated for CNS and/or brain delivery. [00928] In some embodiments, the engineered retron and the associated compositions, systems, vectors, uses, and methods of use, can be used to treat hearing diseases or hearing loss in one or both ears. Deafness is often caused by lost or damaged hair cells that cannot relay signals to auditory neurons. In some embodiments, the composition, system, or modified cells can be delivered to one or both ears for treating or preventing hearing disease or loss by any suitable method or technique known in the art, such as US20120328580 (e.g., auricular administration), by intratympanic injection (e.g., into the middle ear), and/or injections into the outer, middle, and/or inner ear; administration in situ, via a catheter or pump (U.S. 2006/0030837) and Jacobsen (U.S. Pat. No.7,206,639). Also see US20120328580. Cells resulting from such methods can then be transplanted or implanted into a patient in need of such treatment. [00929] In some embodiments, the engineered retron and the associated compositions, systems, vectors, uses, and methods of use, can be used to treat diseases in non-dividing cells. Exemplary non-dividing cells include muscle cells or neurons. In such cells, homologous recombination (HR) is generally suppressed in the G1 cell-cycle phase, but can be turned back on using art-recognized methods, such as Orthwein et al. (Nature.2015 Dec 17; 528(7582): 422– 426). [00930] In some embodiments, the engineered retron and the associated compositions, systems, vectors, uses, and methods of use, can be used to treat diseases of the eye.
[00931] In some embodiments, the engineered retron and the associated compositions, systems, vectors, uses, and methods of use, can be used to treat muscle diseases and cardiovascular diseases.
[00932] In some embodiments, the engineered retron and the associated compositions, systems, vectors, uses, and methods of use, can be used to treat diseases of the liver and kidney.
[00933] In some embodiments, the engineered retron and the associated compositions, systems, vectors, uses, and methods of use, can be used to treat epithelial and lung diseases.
[00934] In some embodiments, the engineered retron and the associated compositions, systems, vectors, uses, and methods of use, can be used to treat diseases of the skin.
[00935] In some embodiments, the engineered retron and the associated compositions, systems, vectors, uses, and methods of use, can be used to treat cancer.
[00936] In some embodiments, the engineered retron and the associated compositions, systems, vectors, uses, and methods of use, can be used in adoptive cell therapy.
[00937] In some embodiments, the engineered retron and the associated compositions, systems, vectors, uses, and methods of use, can be used to treat infectious diseases.
[00938] In some embodiments, the engineered retron and the associated compositions, systems, vectors, uses, and methods of use, can be used to treat mitochondrial diseases.
[00939] All publications, published patent documents, and patent applications cited herein are hereby incorporated by reference to the same extent as though each individual publication, published patent document, or patent application was specifically and individually indicated as being incorporated by reference.
O. Sequences
[00940] The instant Specification discloses and claims recombinant retrons and components thereof (e.g., recombinant ncRNAs and recombinant retron RTs), as well as genome modification systems comprising said recombinant retrons and/or recombinant components thereof. Without limitation, such systems include recombinant retron-based genome editing systems, recombineering systems, and cell-recording systems. The Specification also discloses and claims the various components and aspects making up the systems, as well as their uses and applications, including without limitation: (a) recombinant nucleic acid molecules encoding recombinant retrons and retron-based genome modification systems, (b) vector systems (including viral and non-viral) comprising one or more components of said retron-based genome modification systems, including all-DNA vector systems, all-RNA vector systems, and DNA/RNA vector systems, (c) delivery systems for delivering said vector systems and/or components of the retron-based genome modification systems (e.g., lipid particles, lipid nanoparticles, and other delivery vehicle formats), (d) formulations comprising any of the aforementioned components for delivery to cells and/or tissues, including in vitro, in vivo, and ex vivo delivery, (e) cells modified by the recombinant retron-based genome modification systems and methods described herein, and (f) methods of modifying cells by conducting genome editing, recombineering, or cell recording using the herein disclosed retron-based genome modification systems, (g) methods of making the recombinant retrons, retron-based genome modification systems, vectors, and formulations described herein, and (h) pharmaceutical compositions and kits for modifying cells under in vitro, in vivo, and ex vivo conditions. [00941] The recombinant retrons and retron-based genome modification systems described herein can be made, in various embodiments, by introducing one or more modifications into a known retron, e.g., a retron that is publicly known (Group I retrons) or a novel retron that is found in nature but which has not previously been recognized or described (Group II retrons). Examples of such retron sequences (and in some cases sequences of their components) are disclosed herein and summarized below: Table X: Previously known retron reverse transcriptases [00942] Table X provides non-limiting examples of 1928 retron reverse transcriptases (AA sequences associated with each NCBI Accession No. assigned to sequence identifiers SEQ ID NO: 19413-21341) that may be modified in accordance with the herein methods to obtain a recombinant retron reverse transcriptase for use in the compositions, systems, and methods described herein. These retron sequences were reported in Mestre et al., “Systematic Prediction of Genes Functionally Associated with Bacterial Retrons and Classification of The Encoded Tripartite Systems,” Nucleic Acids Research, Volume 48, Issue 22, 16 December 2020, Pages 12632-12647, the contents of which are incorporated herein by reference. In some embodiments, the Table X retrons are meant to be excluded from the scope of the claimed subject matter.
Figure imgf000333_0001
Figure imgf000334_0001
Figure imgf000335_0001
Figure imgf000336_0001
Figure imgf000337_0001
Figure imgf000338_0001
Figure imgf000339_0001
Figure imgf000340_0001
Figure imgf000341_0001
Figure imgf000342_0001
Figure imgf000343_0001
Figure imgf000344_0001
Figure imgf000345_0001
Figure imgf000346_0001
Figure imgf000347_0001
Figure imgf000348_0001
Figure imgf000349_0001
Figure imgf000350_0001
Figure imgf000351_0001
Figure imgf000352_0001
Figure imgf000353_0001
Figure imgf000354_0001
Figure imgf000355_0001
Figure imgf000356_0001
Figure imgf000357_0001
Figure imgf000358_0001
Figure imgf000359_0001
Figure imgf000360_0001
Figure imgf000361_0001
Figure imgf000362_0001
Figure imgf000363_0001
Figure imgf000364_0001
Figure imgf000365_0001
Figure imgf000366_0001
Figure imgf000367_0001
Figure imgf000368_0001
Figure imgf000369_0001
Figure imgf000370_0001
Figure imgf000371_0001
Figure imgf000372_0001
Figure imgf000373_0001
Figure imgf000374_0001
Figure imgf000375_0001
Figure imgf000376_0001
Thus, in various aspects, the present disclosure describes making recombinant retrons and retron- based genome editing sy tables summarize the amino acid and nucleotide sequences disclosed herein, each being ascribed an individual sequence identifier (i.e., SEQ ID NO). In some instances, the sequences are disclosed in the body of the Specification. In other instances, the sequences are disclosed in the Sequence Listing, which forms a part of the instant Specification.
[00943] The following is a sequence key to summarize and identify the enclosed sequences. The sequence key accompanies the visual summary provided in FIGs. 28 and 29.
Table A and Sub-Tables A1-A45: Novel Retron Reverse Transcriptase Sequences
[00944] Table A provides non-limiting examples of retron reverse transcriptases that may be modified in accordance with the herein methods to obtain a recombinant retron reverse transcriptase or use in the compositions, systems, and methods described herein.
[00945] In particular, Table A provides sequence identifiers corresponding to the novel retron RTs identified as a result of the computational discovery work described in the Examples. The table provides sequence identifiers corresponding to the contents of the Sequence Listing included as part of this Specification. The table includes both RT amino acid sequences and RT nucleic acid sequences. The table is organized into forty-five sub-tables each of which represents those sequences forming a single phylogenetic clade of related retron RTs as determined by the computational work described in the Examples.
Figure imgf000377_0001
Figure imgf000378_0001
Table B and Sub-Tables B1-B45: Exemplary ncRNA Sequences
[00946] Table B provides non-limiting examples of retron ncRNA sequences that may be modified in accordance with the herein methods to obtain a recombinant retron ncRNA sequences for use in the compositions, systems, and methods described herein.
[00947] In particular, Table B provides sequence identifiers corresponding to the novel retron RTs identified as a result of the computational discovery work described in the Examples. The table provides sequence identifiers corresponding to the contents of the Sequence Listing included as part of this Specification. The table is organized into forty-five sub-tables each of which represents those sequences forming a single phylogenetic clade of related retron ncRNAs as determined by the computational work described in the Examples.
[00948] In various embodiments and in the claims, the disclosure provides recombinant retron-based genome editing systems which comprise combining in a cell through various delivery strategies a retron RT together with an ncDNA. In various aspects, the retron RTs and the ncDNA constituting the recombinant retron-based genome editing system can be based on pairing together such components that are naturally found associated to one another in nature, i.e., sourced from the same bacterial species. These are referred to as the “cognate” pairings of retron RT and retron ncRNA. A cognate pairing of a ncRNA and an RT (amino acid or nucleotide sequence) can easily be identified from Sequence Listing by identifying those ncRNA sequences from Table B and those RT sequences from Table A have the same genomic accession number that is indicated for each sequence in the Sequence Listing. In various other aspects, the retron RT component and the ncRNA component can be from different bacterial species, i.e., are not found together in nature as cognate pairs. In still other embodiments, the retron RT component and the ncRNA component can both be from the same phylogenetic functional type (e.g., Type I-A, Type I-B1, Type I-B2, Type IC, etc.). For example, a recombinant retron-based genome editing system may be comprised of a retron RT from Type I-A (i.e., SEQ ID Nos: 3980-4178 for AA and SEQ ID Nos: 11231-11429 for NT– see Table A) and a retron ncRNA also from Type I-A (i.e., SEQ ID Nos: 16886-17078 – see Table B).
Figure imgf000379_0001
Figure imgf000380_0001
Table C: Consensus RT amino acid sequence [00949] Table C provides a consensus amino acid sequence for each type of RT identified in Table A.
Figure imgf000380_0002
Figure imgf000381_0001
Figure imgf000382_0001
EXEMPLARY EMBODIMENTS Exemplary embodiment Group A [00950] The following paragraphs describe exemplary and non-limiting embodiments of the present disclosure. [00951] Paragraph 1. An engineered retron, comprising: a) an msr locus (that encodes the msr RNA portion of an msDNA); b) an msd locus encoding the msd RNA portion of the msDNA; c) a retron reverse transcriptase (RT), wherein said msd RNA is capable of being reverse transcribed to form the msDNA by the retron reverse transcriptase (RT); d) a heterologous nucleic acid inserted at or within the msd locus, upstream of the msr locus, upstream or downstream of the msd locus; and, e) a sequence modification (e.g., insertion, deletion, and/or substitution of one or more nucleotide(s)) in the msr locus and/or the msd locus that: i) modulates (e.g., enhances) reverse transcription, processivity, acuracy/fidelity, and/or production of the msDNA (e.g., in the mammalian cell); ii) modulates (e.g., reduces) immunogenicity of ncRNA encoded by the engineered retron (e.g., the msr locus and/or the msd locus) in a host (e.g., a host comprising the mammalian cell); and/or iii) comprises a nucleotide sequence that modulates (e.g., inhibits, either permanently or transiently) a function of the msDNA; and/or iv) modulates (e.g., improves) efficiency of targeted genome editing / engineering. [00952] Paragraph 2. The engineered retron of paragraph 1, wherein said engineered retron is engineered based on and/or to resemble a secondary structure of a wild-type or consensus retron encoding a wild-type or consensus retron ncRNA encompassed by: 1) any one of the sequences and/or structures as depicted in SEQ ID NOs: 1-21 and/or FIGs. 2-22; or 2) a variant of 1), having: A) up to 1, 2, or 3 (e.g., up to 1) nucleotide changes per 10 red lettered-nucleotides; B) up to 4, 5, or 6 (e.g., up to 1 or 2) nucleotide changes per 10 black lettered- nucleotides; and/or C) up to 7, 8, or 9 (e.g., up to 3 or 4) nucleotide changes per 10 grey lettered- nucleotides; and/or optionally further comprising: a) 7, 8, 9, or 10 (e.g., 9 or 10) nucleotides present per 10 red-circled nucleotides; b) 6, 7, 8, 9, or 10 (e.g., 8, 9 or 10) nucleotides present per 10 black-circled nucleotides; c) 4, 5, 6, 7, 8, 9, or 10 (e.g., 6, 7, 8, 9 or 10) nucleotides present per 10 grey-circled nucleotides; and/or d) 2, 3, 4, 5, 6, 7, 8, 9, or 10 (e.g., 4, 5, 6, 7, 8, 9, or 10) nucleotides present per 10 white-circled nucleotides. [00953] Paragraph 3. The engineered retron of any one of paragraphs 1-2, wherein said engineered retron is engineered by introducing said sequence modification into a wild-type retron encoding a wild-type ncRNA, or into a synthetic/artificial retron encoding the consensus retron ncRNA. [00954] Paragraph 4. The engineered retron of any one of paragraphs 1-3, wherein the sequence modification comprises, in the encoded retron ncRNA, one or more of: (i) a modified (e.g., mutated, reduced, or eliminated) bulge in a1, a2 or both a1 and a2; (ii) an extension or shortening of a1, a2 or both a1 and a2; (iii) an extension or shortening of a spacer sequence between hairpin loops (e.g., S1, S2, S3, S4, S5 and/or S6); (iv) a modified (e.g., mutated or eliminated) bulge in hairpin loops (e.g., L1, L2, L3, L4, L5 and/or L6 (e.g., by removing unpaired bases in the bulge, or by replacing unpaired bases with an equivalent number of base pairs)); (v) a modified (e.g., extended or shortened) length of hairpin loops (e.g., L1, L2, L3, L4, L5 and/or L6); (vi) an alternative L1 and/or L2 having complement, reverse, or reverse complement sequences; (vii) a modified (e.g., increased) number of unpaired bases at the tip of hairpin loops (e.g., L1, L2, L3, L4, L5 and/or L6); (viii) a modified (e.g., increased or decreased) GC content in hairpin loops (e.g., L1, L2, L3, L4, L5 and/or L6); (ix) an insertion of the heterologous nucleic acid in spacer sequences between hairpin loops (e.g., S1, S2, S3, S4, S5 and/or S6), or at the tip of hairpin loops (e.g., L1, L2, L3, L4, L5 and/or L6); (x) a deletion of hairpin loops (e.g., L1, L2, L3, L4, L5 and/or L6); (xi) an addition of a new loop in spacer sequences between hairpin loops (e.g., S1, S2, S3, S4, S5 and/or S6); (xii) circularization of the ncRNA with the 5’ end and the 3’ end of the ncRNA being connected either directly, or via a spacer sequence; (xiii) a repositioned branching guanosine capable of initiating reverse transcription priming; (xiv) a staggered end sequence that reduces immunogenicity of the retron ncRNA, created by, e.g., adding or removing the 5’ a1 nucleotides and/or the 3’ a2 nucleotides; and/or, (xv) an antisense sequence complementary to a CRISPR/Cas guide RNA (gRNA) sequence encoded by the heterologous nucleic acid, wherein the antisense sequence hybridizes to and inhibits said gRNA in the encoded retron ncRNA, and wherein said antisense sequence is removed upon reverse transcription of the msDNA. [00955] Paragraph 5. The engineered retron of any one of paragraphs 1-4, comprising: (i) a heterologous nucleic acid (such as the coding sequence for an RNA aptamer or a ribozyme) inserted into the msr locus or the msd locus (such as in an S region (e.g., S1, S2, S3, S4, S5 and/or S6), or the tip of an L region (e.g., L1, L2, L3, L4, L5 and/or L6)), or upstream or downstream of either the msr locus or the msd locus; or, (ii) a first heterologous nucleic acid inserted into the msd locus, and a second heterologous nucleic acid inserted either upstream of the msr locus or downstream of the msd locus, wherein the second heterologous nucleic acid encodes a CRISPR/Cas guide RNA (gRNA). [00956] Paragraph 6. The engineered retron of any one of paragraphs 1-5, wherein said retron reverse transcriptase (RT) is a cognate RT, a retron RT from a species within the same clade of the cognate RT, or a retron RT not within the same clade of the cognate RT (such as an unrelated RT or an engineered RT). [00957] Paragraph 7. The engineered retron of any one of paragraphs 1-6, wherein said heterologous nucleic acid encodes a protein or peptide of interest, or wherein said heterologous nucleic acid comprises or encodes a donor / template sequence (e.g., a donor that corrects / repairs / removes a mutation at the target genome site, such as a mutated exon in a disease gene; a functional DNA element (such as a promoter, an enhancer, a protein binding sequence, a methylation site, a homology region for assiting gene editing, etc.); or a coding sequence for a functional RNA element (ncRNAs, etc)). [00958] Paragraph 8. The engineered retron paragraph 7, wherein said protein or peptide of interest comprises a therapeutic protein (such as a wt protein defective in a disease cell, or a therapeutic antibody or antigen-binding fragment thereof) useful in treating a disease. [00959] Paragraph 9. The engineered retron of paragraph 7, wherein said donor / template sequence corrects/repairs/removes a mutation at the target genome site (such as a mutated exon in a disease gene). [00960] Paragraph 10. The engineered retron of any one of paragraphs 1-9, further comprising or encoding a sequence-specific nuclease (such as a CRISPR/Cas effector enzyme, a ZFN, a TALEN, a meganuclease, TnpB, IscB, or a restriction endonuclease (RE)), and/or a DNA-repair modulating biomolecule. [00961] Paragraph 11. The engineered retron of paragraph 10, wherein the sequence- specific nuclease is fused to the RT, optionally via a flexible linker (e.g., a flexible linker comprising Gly and Ser rich sequences such as G4S repeats or GS repeats) or by a generally disordered protein sequence (such as unstructured hydrophilic, biodegradable protein polymer, e.g., an XTEN peptide polymer). [00962] Paragraph 12. The engineered retron of paragraph 10 or 11, wherein the nuclease is a CRISPR/Cas effector enzyme that forms a complex with a guide RNA (gRNA) recognizing a target sequence, wherein the gRNA is linked to the ncRNA and/or the msDNA, either directly or through a linker / spacer polynucleotide. [00963] Paragraph 13. The engineered retron of paragraph 10, wherein the DNA-repair modulating biomolecule is a regulatory protein that modulates (e.g., enhances) HDR, and the regulatory protein is fused to the RT or to the sequence-specific nuclease, optionally via a flexible linker (e.g., the flexible linker comprising Gly and Ser rich sequences such as G4S repeats or GS repeats) or by a generally disordered protein sequence (such as unstructured hydrophilic, biodegradable protein polymer, e.g., an XTEN peptide polymer). [00964] Paragraph 14. A vector system comprising a vector comprising the engineered retron of any one of paragraphs 1-13. [00965] Paragraph 15. The vector system of paragraph 14, wherein the msr locus, the msd locus, and the RT gene are provided by the same vector. [00966] Paragraph 16. The vector system of paragraph 14 or 15, wherein the same vector comprises a promoter operably linked to the msr locus and/or the msd locus. [00967] Paragraph 17. The vector system of any one of paragraphs 14-16, wherein the promoter is further operably linked to the RT gene. [00968] Paragraph 18. The vector system of paragraph 14, wherein the msr locus, the msd locus, and the RT gene are provided by at least two different vectors. [00969] Paragraph 19. The vector system of paragraph 14 or 18, wherein said RT gene is provided in trans with respect to the msr gene and/or the msd gene. [00970] Paragraph 20. The vector system of any one of paragraphs 14-19, wherein the one or more vectors comprise a viral vector and/or a nonviral vector. [00971] Paragraph 21. The vector system of paragraph 20, wherein the nonviral vector comprises a plasmid. [00972] Paragraph 22. The vector system of any one of paragraphs 14-21, comprising a vector encoding a sequence-specific nuclease. [00973] Paragraph 23. The vector system of paragraph 22, wherein the sequence-specific nuclease comprises an RNA-guided sequence-specific nuclease (e.g., a CRISPR/Cas effector enzyme, an engineered RNA-guided FokI-nuclease (e.g., dCas-FokI), an RNA-guided DNA endonuclease, TnpB, IscB, or a transposon-associated nuclease), or a non-RNA-guided sequence-specific nuclease (e.g., a meganuclease, a zinc finger nuclease (ZFN), a TALE nuclease (TALEN), or a restriction endonuclease (RE)). [00974] Paragraph 24. The vector system of paragraph 23, wherein the Cas effector enzyme is a Class 1, Type I, II, or III Cas; a Class 2, Type II Cas (e.g., Cas9); or a Class 2, Type V Cas (e.g., Cpfl). [00975] Paragraph 25. The vector system of paragraph 23, wherein: 1) the RNA-guided sequence-specific nuclease comprises the CRISPR/Cas effector enzyme, the engineered RNA-guided FokI-nuclease (e.g., dCas-FokI), the RNA-guided DNA endonuclease, TnpB, IscB, IsrB, or the transposon-associated nuclease; or, 2) non-RNA-guided sequence-specific nuclease comprises the meganuclease, the zinc finger nuclease (ZFN), the TALE nuclease (TALEN), or the restriction endonuclease (RE). [00976] Paragraph 26. The vector system of any one of paragraphs 14-25, further comprising a vector encoding a homologous recombination enhancer protein. [00977] Paragraph 27. An isolated host cell comprising the engineered retron of any one of paragraphs 1-13, or the vector system of any of paragraphs 14-26. [00978] Paragraph 28. The host cell of paragraph 27, wherein the host cell is a prokaryotic, archeon, or eukaryotic host cell. [00979] Paragraph 29. The host cell of paragraph 28, wherein the eukaryotic host cell is a mammalian host cell. [00980] Paragraph 30. The host cell of paragraph 28, wherein the eukaryotic host cell is a non-human host cell. [00981] Paragraph 31. The host cell of paragraph 29, wherein the mammalian host cell is a human host cell. [00982] Paragraph 32. The host cell of any of paragraphs 28-31, wherein the host cell is an artificial cell or genetically modified cell. [00983] Paragraph 33. A pharmaceutical composition comprising the engineered retron of any one of paragraphs 1-13. [00984] Paragraph 34. A delivery vehicle comprising the engineered retron of any one of paragraphs 1-13 or ncRNA encoded by the engineered retron of any one of paragraphs 1-13, the vector of any one of paragraphs 14-26, the host cell of any one of paragraphs 27-32, or the pharmaceutical composition of paragraph 33. [00985] Paragraph 35. The delivery vehicle of paragraph 34, which is a lipid nanoparticle. [00986] Paragraph 36. The delivery vehicle of paragraph 35, wherein the lipid nanoparticle comprises at least one cationic lipid selected from the group consists of: a lipid in Table (I), a lipid having a structure of Formula (I), a lipid having a structure of Formula (II), a lipid having a structure of Formula (III), a lipid having a structure of Formula (IV), a lipid having a structure of Formula (V), a lipid having a structure of Formula (VI), and combinations thereof. [00987] Paragraph 37. A kit comprising the engineered retron of any one of paragraphs 1- 13 or ncRNA encoded by the engineered retron of any one of paragraphs 1-13, the vector or vector system of any one of paragraphs 14-26, the host cell of any one of paragraphs 27-32, the pharmaceutical composition of paragraph 33, or the delivery vehicle of any one of paragraphs 34-36, and instructions for genetically modifying a cell with said engineered retron, ncRNA, vector / vector system, host cell, pharmaceutical composition, or delivery vehicle. [00988] Paragraph 38. A method of modifying a target DNA sequence in a host (e.g., mammalian) cell, the method comprising introducing into the mammalian cell the engineered retron of any one of paragraphs 1-13 or ncRNA encoded by the engineered retron of any one of paragraphs 1-13, the vector or vector system of any one of paragraphs 14-26, the pharmaceutical composition of paragraph 33, or the delivery vehicle of any one of paragraphs 34-36, to allow the production of the msDNA in the host (e.g., mammalian) cell, wherein the encoded heterologous nucleic acid in the msDNA is integrated into the genome of the host (e.g., mammalian) cell at the target DNA sequence. [00989] Paragraph 39. Use of the engineered retron of any one of paragraphs 1-13 or ncRNA encoded by the engineered retron of any one of paragraphs 1-13, the vector or vector system of any one of paragraphs 14-26, the pharmaceutical composition of paragraph 33, or the delivery vehicle of any one of paragraphs 34-36, to effect modification of a target DNA sequence in a host (e.g., mammalian) cell. Exemplary embodiment Group B [00990] The following paragraphs further describe exemplary and non-limiting embodiments of the present disclosure. [00991] Paragraph 1. An engineered nucleic acid construct comprising: a) a first polynucleotide encoding a non-coding RNA (ncRNA), said first polynucleotide comprising: 1) an msr locus encoding the msr RNA portion of a multi-copy single-stranded DNA (msDNA); and 2) an msd locus encoding the msd RNA portion of the msDNA; and b) one or more heterologous nucleic acids inserted at or within a location selected from: the msd locus, upstream of the msr locus, upstream of the msd locus, and downstream of the msd locus, wherein the ncRNA comprises: (I) an ncRNA listed in Table B, or an ncRNA having at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%%, at least 99.1%, at least 99.2%, at least 99.3%, at least 99.4%, at least 99.5%, at least 99.6%, at least 99.7%, at least 99.8% or at least 99.9% sequence identity with an ncRNA listed in Table B; and/or (II) an ncRNA having a conserved structure of any one of the ncRNA structures of FIGs.2- 27; and wherein the ncRNA optionally excludes any ncRNA associated in nature with any one of the retron reverse transcriptases of Table X. [00992] Paragraph 2. The engineered nucleic acid construct of paragraph 1, further comprising a second polynucleotide encoding a reverse transcriptase (RT), or a portion thereof, wherein the encoded RT or portion thereof is capable of synthesizing a DNA copy of at least a portion of the msd locus encoding the msDNA. [00993] Paragraph 3. The engineered nucleic acid construct of paragraph 2, wherein the second polynucleotide comprises: III) a polynucleotide listed in Table A, or a polynucleotide having at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%%, at least 99.1%, at least 99.2%, at least 99.3%, at least 99.4%, at least 99.5%, at least 99.6%, at least 99.7%, at least 99.8% or at least 99.9% sequence identity to a polynucleotide listed in Table A; and/or IV) encodes a consensus amino acid sequence of Table C, or encodes an amino acid sequence having at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%%, at least 99.1%, at least 99.2%, at least 99.3%, at least 99.4%, at least 99.5%, at least 99.6%, at least 99.7%, at least 99.8% or at least 99.9% sequence identity to an amino acid sequence listed in Table C; and/or wherein the second polynucleotide encodes: V) a polypeptide listed in Table A, or a polypeptide having at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%%, at least 99.1%, at least 99.2%, at least 99.3%, at least 99.4%, at least 99.5%, at least 99.6%, at least 99.7%, at least 99.8% or at least 99.9% sequence identity to a polypeptide listed in Table A; and/or VI) a polypeptide comprising a polypeptide consensus sequence listed in Table C, or a polypeptide having at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%%, at least 99.1%, at least 99.2%, at least 99.3%, at least 99.4%, at least 99.5%, at least 99.6%, at least 99.7%, at least 99.8% or at least 99.9% sequence identity to an amino acid sequence listed in Table C; and/or wherein the second polynucleotide optionally does not encode an amino acid sequence listed in Table X. [00994] Paragraph 4. An engineered nucleic acid construct comprising: a) a first polynucleotide encoding a non-coding RNA (ncRNA), said first polynucleotide comprising: 1) an msr locus encoding the msr RNA portion of a multi-copy single-stranded DNA (msDNA); and 2) an msd locus encoding the msd RNA portion of the msDNA; b) one or more heterologous nucleic acids inserted at or within a location selected from: the msd locus, upstream of the msr locus, upstream of the msd locus, and downstream of the msd locus; and c) a second polynucleotide encoding a reverse transcriptase (RT), or a portion thereof, wherein the encoded RT or portion thereof is capable of synthesizing a DNA copy of at least a portion of the msd locus encoding the msDNA, and, wherein the non-coding RNA (ncRNA) of the first polynucleotide optionally has a conserved structure of any one of the ncRNA structures of FIGs.2-27; wherein the second polynucleotide comprises: I) a polynucleotide listed in Table A, or a polynucleotide having at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%%, at least 99.1%, at least 99.2%, at least 99.3%, at least 99.4%, at least 99.5%, at least 99.6%, at least 99.7%, at least 99.8% or at least 99.9% sequence identity to a polynucleotide listed in Table A; and/or wherein the second polynucleotide encodes: II) a polypeptide listed in Table A, or a polypeptide having at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%%, at least 99.1%, at least 99.2%, at least 99.3%, at least 99.4%, at least 99.5%, at least 99.6%, at least 99.7%, at least 99.8% or at least 99.9% sequence identity to a polypeptide listed in Table A; and/or IV) a polypeptide comprising a polypeptide consensus sequence listed in Table C, or a polypeptide having at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%%, at least 99.1%, at least 99.2%, at least 99.3%, at least 99.4%, at least 99.5%, at least 99.6%, at least 99.7%, at least 99.8% or at least 99.9% sequence identity to a polypeptide listed in Table C; and wherein the second polynucleotide optionally does not encode an amino acid sequence of Table X. [00995] Paragraph 4a. An engineered nucleic acid construct, comprising: 1) an msr locus (that encodes the msr RNA portion of an msDNA); 2) an msd locus encoding the msd RNA portion of the msDNA; 3) a sequence encoding a retron reverse transcriptase (RT), wherein said msd RNA is capable of being reverse transcribed to form the msDNA by the retron reverse transcriptase (RT); and, 4) a heterologous nucleic acid inserted at or within the msd locus, upstream of the msr locus, upstream or downstream of the msd locus; wherein the engineered nucleic acid construct optionally has (a) a secondary structure of a wild- type ncRNA of any one of FIGs.2-27 or b) a variant of a), having: i) up to 1, 2, or 3 (e.g., up to 1) nucleotide changes per 10 red lettered-nucleotides; ii) up to 4, 5, or 6 (e.g., up to 1 or 2) nucleotide changes per 10 black lettered- nucleotides; and/or iii) up to 7, 8, or 9 (e.g., up to 3 or 4) nucleotide changes per 10 grey lettered- nucleotides. [00996] Paragraph 5. The engineered nucleic acid construct of any one of paragraphs 1 to 4a, comprising one or more sequence modifications (e.g., an insertion, deletion, and/or substitution of one or more nucleotide(s)) in the msr locus and/or the msd locus that: a) modulates (e.g., enhances) reverse transcription, processivity, accuracy/fidelity, and/or production of the msDNA (e.g., in the mammalian cell); b) modulates (e.g., reduces) immunogenicity of ncRNA encoded by the engineered retron (e.g., the msr locus and/or the msd locus) in a host (e.g., a host comprising the mammalian cell); c) modulates (e.g., inhibits, either permanently or transiently) a function of the msDNA; and/or d) modulates (e.g., improves) efficiency of targeted genome editing / engineering. [00997] Paragraph 6. The engineered nucleic acid construct of any one of paragraphs 1 to 4, wherein said engineered nucleic acid construct has a secondary structure of a wild-type retron encoding a wild-type retron ncRNA encompassed by: a) any one of the structures as depicted in FIGs.2-27, or b) a variant of a), having: i) up to 1, 2, or 3 (e.g., up to 1) nucleotide changes per 10 red lettered-nucleotides; ii) up to 4, 5, or 6 (e.g., up to 1 or 2) nucleotide changes per 10 black lettered- nucleotides; and/or iii) up to 7, 8, or 9 (e.g., up to 3 or 4) nucleotide changes per 10 grey lettered- nucleotides; and/or optionally further comprising: i) 7, 8, 9, or 10 (e.g., 9 or 10) nucleotides present per 10 red-circled nucleotides; ii) 6, 7, 8, 9, or 10 (e.g., 8, 9 or 10) nucleotides present per 10 black-circled nucleotides; iii) 4, 5, 6, 7, 8, 9, or 10 (e.g., 6, 7, 8, 9 or 10) nucleotides present per 10 grey-circled nucleotides; and/or iv) 2, 3, 4, 5, 6, 7, 8, 9, or 10 (e.g., 4, 5, 6, 7, 8, 9, or 10) nucleotides present per 10 white-circled nucleotides. [00998] Paragraph 7. The engineered nucleic acid construct of any one of paragraphs 1- 6, wherein the nucleic acid construct is engineered by introducing the one or more sequence modifications into a wild-type retron encoding a wild-type ncRNA listed in Table B. [00999] Paragraph 8. The engineered nucleic acid construct of any one of paragraphs 1- 7, wherein the one or more sequence modifications in the ncRNA comprises one or more of: (i) a modified (e.g., mutated, reduced, or eliminated) bulge in a1, a2, or both a1 and a2; (ii) an extension or shortening of a1, a2, or both a1 and a2; (iii) an extension or shortening of a spacer sequence between hairpin loops (e.g., S1, S2, S3, and/or S4); (iv) an additional or modified (e.g., mutated or eliminated) bulge in hairpin loops (e.g., L2 and/or L3 (e.g., by removing unpaired bases in the bulge, or by replacing unpaired bases with an equivalent number of base pairs)); (v) a modified (e.g., extended or shortened) length of hairpin loops (e.g., L1, L2, L3, and/or L4); (vi) an alternative L1 and/or L2 having complement, reverse, or reverse complement sequences; (vii) a modified (e.g., increased) number of unpaired bases at the tip of hairpin loops (e.g., L1, L2, L3, and/or L4); (viii) a modified (e.g., increased or decreased) GC content in hairpin loops (e.g., L1, L2, L3, and/or L4); (ix) an insertion of the heterologous nucleic acid in spacer sequences between hairpin loops (e.g., S1, S2, S3 and/or S4), or at the tip of hairpin loops (e.g., L1, L2, L3, and/or L4); (x) a deletion of one or more hairpin loops (e.g., L1, L2, L3 and/or L4); (xi) an addition of a new loop in a spacer sequence between hairpin loops (e.g., S1, S2, S3, and/or S4); (xii) circularization of the ncRNA with the 5ʹ end and the 3ʹ end of the ncRNA being connected either directly, or via a spacer sequence; (xiii) a repositioned branching guanosine capable of initiating reverse transcription priming; (xiv) a staggered end sequence that reduces immunogenicity of the retron ncRNA, created by, e.g., adding or removing the 5ʹ a1 nucleotides and/or the 3ʹ a2 nucleotides; and/or, (xv) an antisense sequence complementary to a CRISPR/Cas guide RNA (gRNA) sequence encoded by the heterologous nucleic acid, wherein the antisense sequence hybridizes to and inhibits said gRNA in the encoded retron ncRNA, and wherein said antisense sequence is removed upon reverse transcription of the msDNA. [001000] Paragraph 9. The engineered nucleic acid construct of any one of paragraphs 1- 8, wherein the one or more heterologous nucleic acid sequences comprise: a) a heterologous nucleic acid (such as the coding sequence for an RNA aptamer or a ribozyme) inserted into the msr locus or the msd locus (such as in an S region (e.g., S1, S2, S3 and/or S4), or the tip of an L region (e.g., L1, L2, L3 and/or L4), or upstream or downstream of either the msr locus or the msd locus; or b) a first heterologous nucleic acid inserted into the msd locus, and a second heterologous nucleic acid inserted either upstream of the msr locus or downstream of the msd locus, wherein the second heterologous nucleic acid encodes a guide RNA. [001001] Paragraph 10. The engineered nucleic acid construct of any one of paragraphs 1- 9, wherein said heterologous nucleic acid encodes: (a) a protein or peptide of interest, or wherein said heterologous nucleic acid comprises; (b) a DNA donor template sequence; (c) a functional DNA element selected from a promoter, an enhancer, a protein binding sequence, a methylation site, a homology region for assisting gene editing, and the like; or (d) a coding sequence for a functional RNA element selected from a guide RNA and a ncRNA. [001002] Paragraph 11. The engineered nucleic acid construct of paragraph 10, wherein said protein or peptide of interest comprises a therapeutic protein useful in treating a disease. [001003] Paragraph 12. The engineered nucleic acid construct of paragraph 10, wherein said DNA donor template sequence corrects/repairs/removes a mutation at the target genome site. [001004] Paragraph 13. The engineered nucleic acid construct of any one of paragraphs 1- 12, further comprising or encoding a sequence-specific nuclease (such as a CRISPR/Cas effector enzyme, a ZFN, a TALEN, a meganuclease, TnpB, IscB, or a restriction endonuclease (RE)), and/or a DNA-repair modulating biomolecule. [001005] Paragraph 13b. The engineered nucleic acid construct of paragraphs 1-13 wherein the engineered nucleic acid is an all-RNA component system. [001006] Paragraph 13c.The engineered nucleic acid construct of paragraphs 1-13 wherein the engineered nucleic acid is an all-DNA molecule system. [001007] Paragraph 14. The engineered nucleic acid construct of paragraph 13, wherein the sequence-specific nuclease is fused to the RT, optionally via a flexible linker (e.g., a flexible linker comprising Gly and Ser rich sequences such as G4S repeats or GS repeats) or by a generally disordered protein sequence (such as unstructured hydrophilic, biodegradable protein polymer, e.g., an XTEN peptide polymer). [001008] Paragraph 15. The engineered nucleic acid construct of paragraph 13 or 14, wherein the nuclease is a CRISPR/Cas effector enzyme that forms a complex with a guide RNA (gRNA) recognizing a target sequence, wherein the gRNA is linked to the ncRNA and/or the msDNA, either directly or through a linker / spacer polynucleotide. [001009] Paragraph 16. The engineered nucleic acid construct of paragraph 13, wherein the DNA-repair modulating biomolecule is a regulatory protein that modulates (e.g., enhances) HDR, and the regulatory protein is optionally fused to the RT or to the sequence-specific nuclease, optionally via a flexible linker (e.g., the flexible linker comprising Gly and Ser rich sequences such as G4S repeats or GS repeats) or by a generally disordered protein sequence (such as unstructured hydrophilic, biodegradable protein polymer, e.g., an XTEN peptide polymer). [001010] Paragraph 17. A vector system comprising one or more vectors comprising the engineered nucleic acid construct of any one of paragraphs 1-16, wherein the vector system is optionally all-RNA. [001011] Paragraph 18. The vector system of paragraph 17, wherein the msr locus, the msd locus, and the polynucleotide encoding the RT are comprised within the same vector. [001012] Paragraph 19. The vector system of paragraph 17 or 18, wherein the same vector further comprises a promoter operably linked to the msr locus and/or the msd locus. [001013] Paragraph 20. The vector system of paragraph 19, wherein the promoter is further operably linked to the polynucleotide encoding the RT. [001014] Paragraph 21. A vector system comprising one or more vectors, comprising the engineered nucleic acid construct of paragraph 1 or 2, wherein the vector system further comprises a second polynucleotide encoding a reverse transcriptase (RT), or a portion thereof, wherein the encoded RT is capable of synthesizing a DNA copy of at least a portion of the msd locus encoding the msDNA, and wherein the msr locus, the msd locus, and the second polynucleotide encoding the RT are provided by at least two different vectors. [001015] Paragraph 22. The vector system of paragraph 21, wherein: a) the second polynucleotide comprises: i) a polynucleotide listed in Table A, or a polynucleotide having at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%%, at least 99.1%, at least 99.2%, at least 99.3%, at least 99.4%, at least 99.5%, at least 99.6%, at least 99.7%, at least 99.8% or at least 99.9% sequence identity to a polynucleotide listed in Table A; and/or b) the second polynucleotide encodes: i) a polypeptide listed in Table A, or a polypeptide having at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%%, at least 99.1%, at least 99.2%, at least 99.3%, at least 99.4%, at least 99.5%, at least 99.6%, at least 99.7%, at least 99.8% or at least 99.9% sequence identity to a polypeptide listed in Table A; and/or ii) a polypeptide listed in Table C, or a polypeptide having at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%%, at least 99.1%, at least 99.2%, at least 99.3%, at least 99.4%, at least 99.5%, at least 99.6%, at least 99.7%, at least 99.8% or at least 99.9% sequence identity to a polypeptide listed in Table C; and wherein the second polynucleotide optionally does not encode a polypeptide listed in Table X. [001016] Paragraph 23. The vector system of paragraph 21 or 22, wherein the polynucleotide encoding the RT is provided in trans with respect to the msr gene and/or the msd gene. [001017] Paragraph 24. The vector system of any one of paragraphs 17-23, wherein the one or more vectors comprise a viral vector. [001018] Paragraph 25. The vector system of paragraph 24, wherein the viral vector is a retroviral vector, a lentiviral vector, an adenoviral vector, an adeno-associated viral vector, a vaccinia viral vector, a poxviral vector, or a herpes simplex viral vector. [001019] Paragraph 26. The vector system of any one of paragraphs 17-23, wherein the one or more vectors comprise a non-viral vector. [001020] Paragraph 27. The vector system of paragraph 26, wherein the non-viral vector comprises a plasmid. [001021] Paragraph 28. The vector system of paragraph 26, wherein the non-viral vector comprises a liposome, a lipid nanoparticle (LNP), a cationic polymer, a vesicle, or a gold nanoparticle. [001022] Paragraph 29. The vector system of any one of paragraphs 17-28, comprising a vector encoding a sequence-specific nuclease. [001023] Paragraph 30. The vector system of paragraph 29, wherein the sequence-specific nuclease comprises an RNA-guided sequence-specific nuclease (e.g., a CRISPR/Cas effector enzyme, an engineered RNA-guided FokI-nuclease (e.g., dCas-FokI), an RNA-guided DNA endonuclease, TnpB, IscB, or a transposon-associated nuclease), or a non-RNA-guided sequence-specific nuclease (e.g., a meganuclease, a zinc finger nuclease (ZFN), a TALE nuclease (TALEN), or a restriction endonuclease (RE)). [001024] Paragraph 31. The vector system of paragraph 30, wherein the Cas effector enzyme is a Class 1, Type I, II, or III Cas; a Class 2, Type II Cas (e.g., Cas9); or a Class 2, Type V Cas (e.g., Cpfl). [001025] Paragraph 32. The vector system of paragraph 30, wherein: 1) the RNA-guided sequence-specific nuclease comprises the CRISPR/Cas effector enzyme, the engineered RNA-guided FokI-nuclease (e.g., dCas-FokI), the RNA-guided DNA endonuclease, TnpB, IscB, IsrB, or the transposon-associated nuclease; or, 2) non-RNA-guided sequence-specific nuclease comprises the meganuclease, the zinc finger nuclease (ZFN), the TALE nuclease (TALEN), or the restriction endonuclease (RE). [001026] Paragraph 33. The vector system of any one of paragraphs 17-32, further comprising a vector encoding a homologous recombination enhancer protein. [001027] Paragraph 34. An RNA molecule encoded by the engineered nucleic acid construct of any one of paragraphs 1-16. [001028] Paragraph 35. A recombinant retron-based genome editing system comprising: a) a non-coding RNA (ncRNA) comprising: i) an msr locus encoding the msr RNA portion of a multi-copy single-stranded DNA (msDNA); and ii) an msd locus encoding the msd RNA portion of the msDNA; b) a heterologous nucleic acid inserted at or within a location selected from: the msd locus, upstream of the msr locus, upstream of the msd locus, and downstream of the msd locus; and c) a sequence encoding a reverse transcriptase (RT), or a domain thereof comprising: i) a polypeptide listed in Table A, or a polypeptide having at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%%, at least 99.1%, at least 99.2%, at least 99.3%, at least 99.4%, at least 99.5%, at least 99.6%, at least 99.7%, at least 99.8% or at least 99.9% sequence identity to a polypeptide listed in Table A; and/or ii) a polypeptide listed in Table C, or a polypeptide having at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%%, at least 99.1%, at least 99.2%, at least 99.3%, at least 99.4%, at least 99.5%, at least 99.6%, at least 99.7%, at least 99.8% or at least 99.9% sequence identity to a polypeptide listed in Table C; and wherein, the RT optionally does not comprise a polypeptide listed in Table X. [001029] Paragraph 36. A recombinant retron-based genome editing system comprising: a) a non-coding RNA (ncRNA) comprising: i) an msr locus encoding the msr RNA portion of a multi-copy single-stranded DNA (msDNA); and ii) an msd locus encoding the msd RNA portion of the msDNA; wherein the ncRNA comprises: i) an ncRNA listed in Table B, or an ncRNA having at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%%, at least 99.1%, at least 99.2%, at least 99.3%, at least 99.4%, at least 99.5%, at least 99.6%, at least 99.7%, at least 99.8% or at least 99.9% sequence identity to an ncRNA listed in Table B; and/or ii) an ncRNA having a conserved structure of any one of the ncRNA structures of FIGs.2-27; and wherein the ncRNA optionally excludes any ncRNA associated in nature with any one of the retron reverse transcriptases of Table X; b) a heterologous nucleic acid inserted at or within a location selected from: the msd locus; upstream of the msr locus; upstream of the msd locus; and downstream of the msd locus; and c) a reverse transcriptase (RT), or a portion thereof, wherein the RT is capable of synthesizing a DNA copy of at least a portion of the msd locus encoding the msDNA. [001030] Paragraph 37. A recombinant retron-based genome editing system comprising: a) a non-coding RNA (ncRNA) comprising: i) an msr locus encoding the msr RNA portion of a multi-copy single-stranded DNA (msDNA); and ii) an msd locus encoding the msd RNA portion of the msDNA; wherein, the ncRNA comprises: i) an ncRNA listed in Table B, or an ncRNA having at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%%, at least 99.1%, at least 99.2%, at least 99.3%, at least 99.4%, at least 99.5%, at least 99.6%, at least 99.7%, at least 99.8% or at least 99.9% sequence identity to an ncRNA listed in Table B; and/or ii) an ncRNA having a conserved structure of any one of the ncRNA structures of FIGs.2-27; and wherein the ncRNA optionally excludes any ncRNA associated in nature with any one of the retron reverse transcriptases of Table X; b) a heterologous nucleic acid inserted at or within a location selected from: the msd locus, upstream of the msr locus, upstream of the msd locus, and downstream of the msd locus; and c) a reverse transcriptase (RT) or a domain thereof: wherein the RT comprises: i) an RT listed in Table A, or an RT having at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%%, at least 99.1%, at least 99.2%, at least 99.3%, at least 99.4%, at least 99.5%, at least 99.6%, at least 99.7%, at least 99.8% or at least 99.9% sequence identity to an RT listed in Table A; and/or ii) a consensus sequence listed in Table C, or a polypeptide having at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%%, at least 99.1%, at least 99.2%, at least 99.3%, at least 99.4%, at least 99.5%, at least 99.6%, at least 99.7%, at least 99.8% or at least 99.9% sequence identity to an amino acid sequence listed in Table C; and wherein the RT does not optionally comprise an RT listed in Table X. [001031] Paragraph 38. An isolated host cell comprising the engineered nucleic acid construct of any one of paragraphs 1-16, the vector system of any of paragraphs 17-33, the RNA molecule of paragraph 34, or the recombinant retron-based genome editing system of any one of paragraphs 35-37. [001032] Paragraph 39. The isolated host cell of paragraph 38, wherein the host cell is a prokaryotic, archeon, or eukaryotic host cell. [001033] Paragraph 40. The isolated host cell of paragraph 38, wherein the eukaryotic host cell is a mammalian host cell. [001034] Paragraph 41. The isolated host cell of paragraph 39, wherein the eukaryotic host cell is a non-human host cell. [001035] Paragraph 42. The isolated host cell of paragraph 40, wherein the mammalian host cell is a human host cell. [001036] Paragraph 43. The isolated host cell of any one of paragraphs 38-42, wherein the host cell is an artificial cell or genetically modified cell. [001037] Paragraph 44. A pharmaceutical composition comprising: a) the engineered nucleic acid construct of any one of paragraphs 1-16, ncRNA encoded by the engineered nucleic acid construct of any one of paragraphs 1-16, the vector system of any one of paragraphs 17-33, the RNA molecule of paragraph 34, the recombinant retron-based genome editing system of any one of paragraphs 35-37, and/or the isolated host cell of any one of paragraphs 38-43; and b) a pharmaceutically acceptable carrier. [001038] Paragraph 45. A pharmaceutical composition comprising: a) a lipid nanoparticle (LNP); and b) the engineered nucleic acid construct of any one of paragraphs 1-16, ncRNA encoded by the engineered nucleic acid construct of any one of paragraphs 1-16, the vector system of any one of paragraphs 17-33, the RNA molecule of paragraph 34, and/or the recombinant retron- based genome editing system of any one of paragraphs 35-37. [001039] Paragraph 46. The pharmaceutical composition of paragraph 45, wherein the LNP encapsulates the engineered nucleic acid construct, ncRNA, vector system, RNA molecule, and/or engineered nucleic acid-enzyme construct. [001040] Paragraph 47. The pharmaceutical composition of paragraph 45 or 46, wherein the lipid nanoparticle comprises: a) one or more ionizable lipids; b) one or more structural lipids; c) one or more PEGylated lipids; and d) one or more phospholipids. [001041] Paragraph 48. The pharmaceutical composition of paragraph 47, wherein the one or more ionizable lipids is selected from the group consisting of those disclosed in Table 2. [001042] Paragraph 49. The pharmaceutical composition of paragraph 47 or 48, wherein the one or more structural lipids are selected from the group consisting of cholesterol, fecosterol, beta sitosterol, sitosterol, ergosterol, campesterol, stigmasterol, brassicasterol, tomatidine, tomatine, ursolic acid, alpha-tocopherol, prednisolone, dexamethasone, prednisone, and hydrocortisone. [001043] Paragraph 50. The pharmaceutical composition of any one of paragraphs 47-49, wherein the one or more PEGylated lipids are selected from the group consisting of PEG-c- DOMG, PEG-DMG, PEG-DLPE, PEG-DMPE, PEG-DPPC, and PEG-DSPE. [001044] Paragraph 51. The pharmaceutical composition of any one of paragraphs 47-50, wherein the one or more phospholipids are selected from the group consisting of 1,2-distearoyl- sn-glycero-3-phosphocholine (DSPC), 1,2-dioleoyl-sn-glycero-3-phosphoethanolamine (DOPE), 1,2-dilinoleoyl-sn-glycero-3-phosphocholine (DLPC), 1,2-dimyristoyl-sn-glycero- phosphocholine (DMPC), 1.2-dioleoyl-sn-glycero-3-phosphocholine (DOPC), 1,2-dipalmitoyl- sn-glycero-3-phosphocholine (DPPC), 1,2-diundecanoyl-sn-glycero-phosphocholine (DUPC), 1- palmitoyl-2-oleoyl-sn-glycero-3-phosphocho line (POPC), 1,2-di-O-octadecenyl-sn-glycero-3- phosphocholine (18:0 Diether PC), 1-oleoyl-2-cholesterylhemisuc cinoyl-sn-glycero-3- phosphocholine (OChemsPC), 1-hexadecyl-sn-glycero-3-phosphocholine (C16 Lyso PC), 1,2- dilinolenoyl-sn-glycero-3-phosphocholine, 1,2-diarachidonoyl-sn-glycero-3-phosphocholine, 1,2-didocosahexaenoyl-sn-glycero-3-phosphocholine, 1,2-diphytanoylsn-glycero-3- phosphoethanolamine (ME 16.0 PE), 1,2-distearoyl-sn-glycero-3-phosphoethanolamine, 1,2- dilinoleoyl-sn-glycero-3-phosphoethanolamine, 1,2-dilinolenoyl-sn-glycero-3- phosphoethanolamine, 1,2-diarachidonoyl-sn-glycero-3-phosphoethanolamine, 1,2- didocosahexaenoyl-sn-glycero-3-phosphoethanolamine, 1,2-dioleoyl-sn-glycero-3-phospho-rac- (1-glycerol) sodium salt (DOPG), and sphingomyelin. [001045] Paragraph 52. The pharmaceutical composition of any one of paragraphs 47-51, wherein the lipid nanoparticle comprises about 48.5 mol % ionizable lipid, about 10 mol % phospholipid, about 40 mol % structural lipid, and about 1.5 mol% of PEG lipid. [001046] Paragraph 53. The pharmaceutical composition of any one of paragraphs 47-52, wherein the lipid nanoparticle comprises about 48.5 mol % ionizable lipid, about 10 mol % phospholipid, about 39 mol % structural lipid, and about 2.5 mol% of PEG lipid. [001047] Paragraph 54. The pharmaceutical composition of any one of paragraphs 47-53, wherein the LNP further comprises a targeting moiety operably connected to the LNP. [001048] Paragraph 55. The pharmaceutical composition of any one of paragraphs 47-54, wherein the LNP further comprises one or more additional components selected from the group consisting of DDAB, EPC, 14PA, 18BMP, DODAP, DOTAP, and C12-200. [001049] Paragraph 56. The pharmaceutical composition of paragraph 45, wherein the lipid nanoparticle comprises at least one cationic lipid selected from the group consisting of: a lipid in Table 2, a lipid having a structure of Formula (I), a lipid having a structure of Formula (II), a lipid having a structure of Formula (III), a lipid having a structure of Formula (IV), a lipid having a structure of Formula (V), a lipid having a structure of Formula (VI), and combinations thereof. [001050] Paragraph 57. A kit comprising the engineered nucleic acid construct of any one of paragraphs 1-16, ncRNA encoded by the engineered nucleic acid construct of any one of paragraphs 1-16, the vector system of any one of paragraphs 17-33, the RNA molecule of paragraph 34, the recombinant retron-based genome editing system of any one of paragraphs 35- 37, the host cell of any one of paragraphs 38-43, or the pharmaceutical composition of any one of paragraphs 44-56, and instructions for genetically modifying a cell with said engineered nucleic acid construct, ncRNA, vector system, host cell, or pharmaceutical composition. [001051] Paragraph 58. A method of modifying a target DNA sequence in a host (e.g., mammalian) cell, the method comprising introducing into the mammalian cell the engineered nucleic acid construct of any one of paragraphs 1-16, ncRNA encoded by the engineered nucleic acid construct of any one of paragraphs 1-16, the vector system of any one of paragraphs 17-33, the RNA molecule of paragraph 34, the recombinant retron-based genome editing system of any one of paragraphs 35-37, or the pharmaceutical composition of any one of paragraphs 44-56, to allow production of the msDNA in the host (e.g., mammalian) cell, wherein the heterologous nucleic acid in the msDNA modifies the genome of the host. [001052] Paragraph 59. The method of paragraph 58, wherein the modifying comprises introducing an insertion, deletion and/or substitution into the target DNA sequence. [001053] Paragraph 60. A method of treating a disease or condition in a subject in need thereof, the method comprising administering a therapeutically effective amount of the engineered nucleic acid construct of any one of paragraphs 1-16, ncRNA encoded by the engineered nucleic acid construct of any one of paragraphs 1-16, the vector system of any one of paragraphs 17-33, the RNA molecule of paragraph 34, the recombinant retron-based genome editing system of any one of paragraphs 35-37, the host cell of any one of paragraphs 38-43, or the pharmaceutical composition of any one of paragraphs 44-56 to the subject, thereby treating the disease or condition in the subject. [001054] Paragraph 61. A method of treating a disease or condition in a subject in need thereof, the method comprising administering a therapeutically effective amount of the host cell of any one of paragraphs 38-43 to the subject, thereby treating the disease or condition in the subject. [001055] Paragraph 62. The method of paragraph 61, wherein the host cell is autologous to the subject. [001056] Paragraph 63. The method of paragraph 61, wherein the host cell is allogeneic to the subject. Exemplary embodiment Group C [001057] The following paragraphs also describe exemplary and non-limiting embodiments of the present disclosure. [001058] Paragraph 1. A gene editing system comprising one or more delivery vehicles, wherein: the delivery vehicle(s) comprise RNA cargo, said RNA cargo comprises (a) at least one mRNA molecule encoding (i) a nucleic acid programmable nuclease and (ii) a retron reverse transcriptase, (b) an engineered retron ncRNA, and (c) guide RNA for the programmable nuclease, each delivery vehicle contains (a)(i) and/or (a)(ii) and/or (b) and/or (c), whereby one delivery vehicle or more than one delivery vehicle delivers (a)(i), (a)(ii), (b), and (c). [001059] Paragraph 2. The gene editing system of paragraph 1, wherein (a)(i) and (a)(ii) comprise a single mRNA molecule encoding the nucleic acid programmable nuclease and the retron reverse transcriptase. [001060] Paragraph 3. The gene editing system of paragraph 2, wherein (a)(i) and (a)(ii) are encoded and expressed as a fusion protein. [001061] Paragraph 4. The gene editing system of paragraph 3, wherein the fusion protein comprises the C-terminal end of the nucleic acid programmable nuclease fused to the N-terminal end of the retron reverse transcriptase (nuclease:RT fusion).
[001062] Paragraph 5. The gene editing system of paragraph 3, wherein the fusion protein comprises the N-terminal end of the nucleic acid programmable nuclease fused to the C-terminal end of the retron reverse transcriptase (RTmuclease fusion).
[001063] Paragraph 6. The gene editing system of paragraph 1, wherein (a)(i) and (a)(ii) comprise a first mRNA molecule encoding the nucleic acid programmable nuclease and a second mRNA molecule encoding the retron reverse transcriptase.
[001064] Paragraph 7. The gene editing system of paragraph 1, wherein (c) is separate from (a)(i), (a)(ii) and (b) or is provided in trans.
[001065] Paragraph 8. The gene editing system of paragraph 1, wherein (b) the engineered retron ncRNA, and (c) the guide RNA are fused or are provided in cis.
[001066] Paragraph 9. The gene editing system of paragraph 8, wherein the guide RNA is fused to the 5’ end of the retron ncRNA.
[001067] Paragraph 10. The gene editing system of paragraph 8, wherein the guide RNA is fused to the 3 ’ end of the retron ncRNA.
[001068] Paragraph 11. The gene editing system of paragraph 8, wherein the engineered ncRNA comprises a first guide RNA fused to the 5’ end of the retron ncRNA, and a second guide RNA fused to the 3’ end of the retron ncRNA, and the first and second guide RNAs target different sequences.
[001069] Paragraph 12. The gene editing system of paragraph 1, wherein the one or more delivery vehicles comprise a liposome or a lipid nanoparticle (LNP).
[001070] Paragraph 13. The gene editing system of paragraph 1, wherein (a) the at least one mRNA molecule encoding (i) the nucleic acid programmable nuclease and (ii) the retron reverse transcriptase, and (b) the engineered retron ncRNA are in the same delivery vehicle. [001071] Paragraph 14. The gene editing system of paragraph 1, wherein the (a) the at least one mRNA molecule encoding (i) the nucleic acid programmable nuclease and (ii) the retron reverse transcriptase, and (b) the engineered retron ncRNA are in separate delivery vehicles. [001072] Paragraph 15. The gene editing system of paragraph 1, wherein the nucleic acid programmable nuclease and the retron reverse transcriptase are encoded on separate mRNA molecules and those separate mRNA molecules of (a)(i) and (a)(ii) are contained in the same delivery vehicle. [001073] Paragraph 16. The gene editing system of paragraph 1, wherein the nucleic acid programmable nuclease and the retron reverse transcriptase are encoded on separate mRNA molecules and those separate mRNA molecules of (a)(i) and (a)(ii) are contained in different delivery vehicles. [001074] Paragraph 17. The gene editing system of paragraph 1, wherein the engineered retron ncRNA includes a sequence of interest encoding a donor polynucleotide comprising an intended edit to be integrated at a target sequence in a cell, and wherein the donor polynucleotide is flanked by a 5' homology arm that hybridizes to a sequence 5’ to the target sequence and a 3' homology arm that hybridizes to a sequence 3' to the target sequence. [001075] Paragraph 18. The gene editing system of paragraph 1, wherein the nucleic acid programmable nuclease comprises a Cas9 nuclease, a TnpB nuclease, or a Cas12a nuclease. [001076] Paragraph 19. The gene editing system of paragraph 18, wherein the nucleic acid programmable nuclease comprises a Cas9 nuclease. [001077] Paragraph 20. The gene editing system of paragraph 1, wherein the retron reverse transcriptase has an amino acid sequence of Table A, or an amino acid sequence having at least 90%, 95%, 99%, or 100% sequence identity with any amino acid sequence of Table A. [001078] Paragraph 21. The gene editing system of paragraph 1, wherein the engineered retron ncRNA comprises: A) a pre-msr sequence having a first complementary region of the retron ncRNA; B) an msr sequence including an msr stem-loop structure; C) an msd sequence including an msd stem-loop structure and a sequence of interest, wherein said msd sequence templates a single strand DNA product (RT-DNA) in the presence of the retron reverse transcriptase; and D) a post-msd sequence having a second complementary region, wherein the first and second complementary regions form an a1/a2 duplex region of the retron ncRNA, wherein the msr stem-loop structure, the msd stem-loop structure, or the a1/a2 duplex comprise a modification which result in increased editing efficiency in the presence of a nucleic acid programmable nuclease that associates with the one or more guide RNAs, and wherein optionally one or more of the guide RNAs of (c) are coupled to the pre-msr sequence, the post-msd sequence, or both the pre-msr sequence and the post-msd sequence. [001079] Paragraph 22. The gene editing system of paragraph 21, wherein the sequence of interest encodes a donor polynucleotide comprising an intended edit to be integrated at a target sequence of a cell, wherein the donor polynucleotide is flanked by a 5' homology arm that hybridizes to a sequence 5’ to the target sequence and a 3' homology arm that hybridizes to a sequence 3' to the target sequence. [001080] Paragraph 23. The gene editing system of paragraph 21, wherein the ncRNA has a nucleotide sequence of any sequences of Table B, or a nucleotide sequence having at least 90%, 95%, 99%, or 100% sequence identity with any sequence of Table B. [001081] Paragraph 24. An isolated cell comprising the gene editing system of paragraph 1. [001082] Paragraph 25. The isolated cell of paragraph 24, wherein the isolated cell is a mammalian cell. [001083] Paragraph 26. The isolated cell of paragraph 25, wherein the mammalian cell is a human cell. [001084] Paragraph 27. A composition comprising: a) the gene editing system of paragraph 1; and b) a pharmaceutically or veterinarily acceptable carrier. [0100] Paragraph 28. The composition of paragraph 27, wherein the delivery vehicle is a lipid nanoparticle comprising: a) one or more ionizable lipids; b) one or more structural lipids; c) one or more PEGylated lipids; and d) one or more phospholipids. [0101] Paragraph 29. The composition of paragraph 28, wherein the one or more ionizable lipids comprises an ionizable lipid set forth in Table 2. [0102] Paragraph 30. A method of genetically modifying a cell comprising: contacting the gene editing system of paragraph 17 with the cell, thereby delivering the RNA cargo to the cell, wherein: the nucleic acid programmable nuclease forms a complex with the guide RNA, wherein said guide RNA directs the complex to the target sequence, the nucleic acid programmable nuclease creates a double-stranded break in in the target sequence, the retron reverse transcriptase and engineered retron ncRNA create RT DNA that comprises the donor polynucleotide, and the donor polynucleotide becomes integrated at the target sequence. EXAMPLES Example 1: Retron Engineering [001085] This example demonstrates that an engineered (or recombinant) retron as described herein can be engineered based on existing sequence information in a sequence database. [001086] Specifically, retron-like reverse transcriptases (RTs) are first identified from various genomic or metagenomic sequence databases. The identified ncRNA regions of the retrons are then predicted in silico, and/or are determined empirically by, for example, reconstituting the putative retron system in a living cell, to assay for msDNA production. [001087] Once a particular wild-type retron is identified and confirmed, one or more sequence elements of the retron are modified based on the methods described herein, and/or the associated RT is modified or engineered to enhance the overall activity and/or processivity of the retron. For example, the wild-type retron can be engineered by one or more of the following methods: (a) addition of a heterologous nucleic acid sequence of interest (e.g., a nucleotide sequence encoding an HDR donor template) to the various portions and structures of the msd locus; (b) performing any/all of the structural modifications described herein; (c) optionally linking the engineered retron ncRNA to one or more CRISPR gRNA, e.g., a gRNA linked to the 3ʹ end of a retron ncRNA, or a gRNA linked to the 5ʹ end of a retron ncRNA, or a pair of gRNAs linked one to the 3ʹ end and one to the 5ʹ end of a retron ncRNA. [001088] The engineered retron or its encoded ncRNA are optionally linked to a sequence- specific nuclease such as CRISPR/Cas enzyme and/or gRNA, ZFN, TALEN, TnpBs, or IscBs, and the like [001089] For example, the RT is fused to the CRISPR/Cas enzyme (such as Cas9 or Cpf1) as a N- or C-terminal fusion, optionally further fused to a nuclear localization signal (NLS) at the N- and/or C-terminal of the fusion. The ncRNA or the resulting msDNA after reverse transcription is also linked to a guide RNA (gRNA) at the 5ʹ or 3ʹ end, and can be used with the cognate CRISPR/Cas enzyme in the method of the invention. [001090] In another example, the RT is linked to a DNA-repair modulating biomolecule, such as a HDR promoter and/or an NHEJ peptide inhibitor, as described herein above. Example 2: Genome Engineering [001091] This example demonstrates that the engineered retron can be used to introduce a heterologous nucleic acid sequence (e.g., a targeting DNA donor or template) into a host cell genome (e.g., a human cell). [001092] First, a targeting DNA is introduced into the msd portion of the engineered retron, as shown in FIG.3 as a “marker.” The heterologous nucleic acid sequence is designed such that it is flanked by 10-100 or more base pairs of homologous sequence substantially identical/homologous to the genomic sequence at the target site. Thus, the desired edit on the “marker” is in between the homologous sequence arms, and includes an insertion, a deletion, and/or other mutations. [001093] A sequence-specific nuclease, such as a Cas9 nuclease, forms a complex with a guide RNA that specifically targets a location at or near the desired editing site on the genomic sequence. The nuclease is designed such that it does not cut the target once the edit is properly installed. In this experiment, the Cas9 nuclease is linked to the retron reverse transcriptase (RT) as a fusion protein. The Cas9 gRNA is also linked to the ncRNA or msDNA produced by the engineered retron. [001094] The engineered retron encoding the sequence-specific nuclease (and the fused RT) as well as the ncRNA (and the linked gRNA) are then introduced into a host cell, such as a human cell. [001095] The engineered retron is introduced as part of a plasmid for transfection into the cell, or as part of a viral vector (such as an AAV vector) for infecting the cell. [001096] Alternatively, the transcribed ncRNA (and the linked gRNA) can be formulated in vitro in, for example, the lipid nanoparticle or delivery system of the invention, for direct delivery into the host cell. The sequence-specific nuclease (and the fused RT) can either be separately delivered into the host cell (using transfection of plasmid or infection by AAV, etc.), or delivered with the ncRNA in lipid nanoparticles. For example, the coding sequence (e.g., mRNA) of the Cas9-RT fusion is formulated with the ncRNA in the same lipid nanoparticle, or separately formulated as lipid nanoparticles to be delivered together, either simultaneously or one after another. [001097] Once present inside the cell, the Cas9-RT fusion is translated by the host cell translation machinery, while the ncRNA is transcribed from the engineered retron (if the ncRNA is not directly delivered into the cell). The RT portion of the fusion proceeds to reverse transcribe the ncRNA and convert it to msDNA, which includes the heterologous nucleic acid sequence as the cargo / donor / template. Meanwhile, double-stranded breaks (DSBs) are generated at the target site by the CRISPR/Cas9 nuclease. The cargo/donor/template sequence is then integrated into the host genome at the target site, via host cell DNA repair (e.g., HDR) after the Cas9 nuclease forms the DSB. [001098] The cells of interest are then assayed for correct installation of the edit on the heterologous nucleic acid sequence, either through phenotypic changes that occur due to the edit, or by direct DNA sequencing of the target site, or both. Example 3: Computational discovery of novel retrons and phylogenetic analysis to increase retron diversity for genome editing applications [001099] Reverse transcriptases (RTs), also called RNA-directed DNA polymerases, are enzymes capable of synthesizing DNA using RNA as a template. Although they are present in the three domains of life and viruses; prokaryotic RTs have been traditionally less explored in comparison with their eukaryotic counterparts. Prokaryotic RTs can be divided into 6 main groups: (1) Group II introns, (2) CRISPR-associated RTs, (3) Diversity Generating Retroelements, (4) Retrons, (5) Abi (Abortive infection) RTs and (6) Unknown Groups of RTs (UG). In the last five years, a burst of research has increased the knowledge about prokaryotic RTs that led to the discovery of novel putative systems with potential antiphage properties, including retrons. In this Example, a systematic search of public databases was performed with the aim of increasing the number and diversity of known retrons for possible use in genome editing applications. As a result, new types of retrons have been identified, and the increase in data has allowed the identification of new associated ncRNAs. [001100] As a first step in this work, a set of known retron RTs was manually curated, trimmed, and aligned to create suitable input, which was then used to train an HMM model for identifying new retron RTs. Next, this model was applied to existing databases of protein sequences (e.g., nr database from NCBI) to identify potential candidate retron RTs. As a next step, the identified candidates were then grouped by sequence identity and individual representative sequences from each group were chosen. These RT domains of these representative candidates were then aligned, and a phylogenetic tree was built (data not shown). Using information about other known classes of RTs, the full phylogeny was separated into bona fide retron RT candidates and RTs that could belong to other classes (such as group II introns, DGRs, CRISPR-Cas RTs, etc). A new alignment and phylogenetic tree were then constructed from these verified candidates, as shown in FIG.28. For all sequences in the new alignment, a matrix of the protein neighborhood was built, indicating which proteins were present in the vicinity of the candidate retron RTs. From there, types and subtypes of retrons were defined based on the RT phylogeny and identity of associated effector proteins. [001101] To predict ncRNAs proximal to the candidate retron RTs, genomic regions proximal to the candidate retron RTs were extracted, aligned, and analyzed for structural covariance using an iterative convergence model. [001102] ncRNA classification methodology: [001103] To identify what type of ncRNA a sequence may belong to, sequences can be checked against the covariance model of existing ncRNA types. The Infernal suite (http://eddylab.org/infernal/) provides tools to do this. Briefly, covariance models can be built from structural alignment of known ncRNAs for each type, then collated into a CM database. From there, new sequences can be searched against the databases to verify whether the sequence fits with any of the represented families. Example 4: Retron-mediated precise genome editing: unique ability to produce donor DNA template known as msDNA (multicopy single-stranded DNA) inside cells beyond current limit of insertion size using an all-RNA system and in the presence of guide RNA provided in trans [001104] Current genome editing methods can efficiently disrupt target genes by inserting and deleting genetic information using programmable nucleases. However, gene disruption that does not discriminate mutant from wild type allele impairs not only mutant allele in pathogenesis but also wild type allele in normal physiology of the organism. In Transthyretin (TTR) induced amyloidosis, for example, mutated TTR alleles cause the disease but non-pathogenic wild type allele plays a role in neuro protection and injury response (1). As a single nucleotide exchange is a dominant variant in TTR (2), the vast majority of pathogenic alleles in human differ from their nonpathogenic counterparts by small modifications that require much more precise editing technologies to correct. Homology-directed repair (HDR) stimulated by nuclease-mediated DNA breaks has been widely used to install precise editing. However, HDR relies on the delivery of exogenous donor DNA, that can trigger strong immune response and has been shown inefficient to deliver universally high abundance in recipients (3). Current therapeutical ex-vivo and in vivo delivery of donor DNA relies on AAV (adeno-associated virus) transduction. However, AAV manufacturing is complicated and expensive. Additionally, AAV has raised safety concerns in 30% of 255 complete or ongoing clinical trials surveyed by meta-analysis (4). RNA delivery in contrast, successfully demonstrated its safety, easier manufacturing and efficacy in two SARS- Covid2 vaccines. Based on this amenability of RNA as a cargo, we searched the system, where donor DNA for precise editing could be generated from delivered RNA within cells. [001105] Retrons are defined by their unique ability to produce an unusual satellite DNA known as msDNA (multicopy single-stranded DNA) from RNA inside the cell (5). These bacterial elements are involved in phage defense (6) and consist of a non-coding RNA and a reverse transcriptase (RT) that can reverse transcribe non-coding RNA (ncRNA) into msDNA. Their tightly defined sites of reverse transcription enable to insert donor DNA sequences in non- coding RNA substrate. Moreover, their compact size is amenable to deliver along with programmable nuclease in all RNA format. These features place retron an attractive tool for precise genome editing in therapeutic application. [001106] Independent approach to generate new DNA sequences by reverse transcription in the cell is prime editing and its virus originating RT can achieve up to 44 bp insertion (7). Retron in natural sequence can produce up to 163 nucleotide single strand DNA (8), suggesting that longer insertion in retron mediated editing is feasible. In line with this, newly characterized retron Aco1 could insert 56 bp with 6 bp deletion in EMX1 locus of HEK293T cells in S.pyogenes Cas9 nuclease system. Evidence of retron from different clades in precise editing [001107] As a first step, over 7,000 retron/retron-like RTs in bacterial genomes were identified and analyzed. Phylogenetic analysis of the over 7,000 retrons produced the phylogenetic tree of FIG.28 Non-coding RNA gene was searched in the genomic neighborhood of the retron/retron-like RT sequences for conserved RNA secondary structures resembling characterized msr-msd transcripts: self-hybridizing inverted repeat at the ends, hairpin structures in msr and msd, such as those represented by FIGs.2-27. Using covariance models and consensus structure detection, ncRNAs were identified with a high degree of confidence in certain RTs. To evaluate gene editing applicability of retron across diverse phylogeny, Eco1, Eco3, Eco5, Aco1, RTX3-2042, 6083 v1 and 6943 were selected for further analysis (FIG.30). Eco1, Eco3 and Eco5 were previoulsy experimentally validated retrons to show msDNA production (9). Aco1 was recently annotated in the literature but has not been experimentally validated as producing msDNA (10). RTX3-2042, 6083 v1 and 6943 are novel retrons. [001108] A gene editing assay was first performed in plasmid DNA format. Retron elements are assembled in a plasmid, where RT is transcribed under CAG promoter and ncRNA fused with single guide RNA (sgRNA) of Cas9 nuclease under U6 or H1 RNA polymerase III promoter (see FIGs.31A and 31B). Desired sequence to insert at EMX1 targeting site flanked with homology arm at both sides is inserted within msD stem loop of ncRNA of each retron. As described in FIGs.32 and 33, the plasmid was transfected to Cas9 expressing human embryonic kidney 293T (HEK293T) cells via lipofection. Three days after transfection, EMX1 target genomic locus was amplified by PCR and sequences were analyzed by next generation sequencing (NGS). The precise edit assayed is characterized as a 10 bp insertion and a 6 bp substitution and its percentage is calculated by CRISPResso 2 analysis among other editing outcomes. A representative editing outcome is shown in FIG.35. Eco1 (FIG.36), Aco1 (FIG. 37), RTX3-2042 (FIG.38), RTX3-6083 v1 and 6943 (FIG.39A) showed precise editing efficiency of 0.3%, 0.1%, 0.25%, 0.06% and 0.05% (left graph in FIGs.36, 37, 38, and 39, respectively). Undesired editing outcome defined as indels that incorporated random nucleotides or deleted sequences near Cas9 cutting site amounted to 50%, 3%, 5%, 2% and 4% (right graph in FIGs.36, 37, 38, and 39A, respectively). Follow up experiments using the same assay indicated that RTX3_6083v1 and RTX3_6943 generated 3-4 fold more precise edits than Eco1 while indels generated from these two retrons were 2-3 fold lower (FIGs.39B and 39C). RTX3_2042 showed precise editing at similar frequencies to Eco1 but had more variability than other samples (FIGs.39B and 39C). These data demonstrated that recently reported (Aco1) and three novel retrons (RTX3-2042, RTX6083 v1, RTX3-6943) enable precise gene editing in human cells, in addition to previously characterized Eco1 (11). [001109] Follow up experiments using the same assay indicated that RTX3-6342S and RTX3-6342L and RTX-1262 generated precise edits 10-20 fold more frequently than Eco1 and approximately 5-10 fold more frequent than RTX3-2042, 6083v1, 6943, and 0637 (see FIG.39E – left). Furthermore, indels generated using RTX3-6342S, 6342L, 1262, 0637, and 6083v1 were two-fold less frequent compared to Eco1 (FIG.39E – right). Indels generated using RTX3-2042 were decreased by 80% compared to Eco1 (FIG.39E – right). Additionally, these retrons all generated precise edits more frequently than another literature validated retron, Eco3, albeit with similar or higher frequencies of indels (FIG.39E). A small group of retrons were also modified with deletions in the msd stem to assess if precise editing could be improved. We noted that precise editing was increased after shortening the msd stem in six retrons while precise editing was lowered or unchanged in the remaining three retrons (FIG.57 and FIG.58). [001110] These data demonstrated that recently reported (Aco1) and six novel retrons (RTX3-0637, RTX3-1262, RTX3-2042, RTX6083 v1, RTX3-6342, RTX3-6943) enable precise gene editing in human cells, in addition to previously characterized Eco1 and Eco3 in gene editing. Furthermore, modifying the msd stem length can alter precise editing but the effects can be retron specific. Evidence of retron-mediated gene editing in all-RNA system [0003] To establish RNA-based editing system, gene editing assay with two RNA components (RT mRNA and ncRNA fused to sgRNA of Cas9 nuclease) was performed first, in HEK293T cells constitutively expressing Cas9 nuclease. With RT and ncRNA being separated in RNA format, the condition for higher precise editing and lower indels could be easier identified than when they are together in the same plasmid by optimizing the ratio between RT enzyme and its substrate. RT mRNA and ncRNA fused to sgRNA of Cas9 nuclease were produced by in vitro transcription. Following experimental scheme in FIG.40. Eco1, Eco3 and Eco5 mediated gene editing activities were compared at two different ratios of RT mRNA and ncRNA-sgRNA fusion in Cas9 expressing HEK293T cells. More amount of ncRNA than RT mRNA was transfected to maximize msDNA production for precise gene editing. Results showed precise edits up to 0.4% for Eco3 and as low as 10% indels for Eco3 (FIG.41). Precise editing frequency in RNA based assay is comparable to that in plasmid-based assay. [001111] Given that Eco3 produced highest editing, RNA load and RT mRNA: ncRNA- sgRNA fusion ratio was further optimized in Eco3 retron system. To 0.2 or 0.5 µg of RT mRNA, ncRNA-sgRNA fusion was added at ratios of RT mRNA to ncRNA, 1:2, 1:3, 1:4, 1:5, 1:8, 1:10, respectively.0.5 µg of RT mRNA gave rise to more precise editing than 0.2 ug at any ratio and increasing ncRNA correlated with higher precise editing (FIG.42, left). Indels was as low as 6% (FIG.42, right). With two RNA components, precise editing was achieved up to 1% of cell population. [001112] Next, Cas9 mRNA was added to RT mRNA and ncRNA-sgRNA fusion, making all RNA editing system in HEK293T cells (FIG.43). At the optimized RNA load and ratio of RT mRNA and ncRNA-sgRNA fusion, the amount of Cas9 mRNA was titrated with Eco3 retron (FIG.44). Precise editing was observed up to 0.1% of cell population with 0.2 µg of Cas9 mRNA and adding more it did not increase editing efficiency. While the precise editing frequency is an order of magnitude lower than two RNA system, the editing occurred by specific action of Cas9 nuclease and retron, since the absence of either abrogated the precise editing. [001113] The delivery of all RNA system to cells was also made via lipofection using MessengerMAX reagent (FIG.45). This approach is more resembling in vivo delivery of therapeutic RNAs loaded in lipid nanoparticle (LNP), in terms of formation of RNA/lipid complexes and cell uptake mechanism. When applied to Aco1 retron system that consists of Aco1 RT mRNA, Aco1 ncRNA-sgRNA fusion, and Cas9 mRNA, precise editing of 56 bp insertion with 6 bp deletion was observed in 0.1% of cell population and the frequency of indels was about 1.5% (FIG.46). Aco1 retron was recently annotated but has not been experimentally validated yet (10). The precise gene editing mediated by Aco1 strongly suggest that Aco1 retron could produce msDNA inside cells. Moreover, the length of insertion in precise editing exceeds those made by other reverse transcriptase mediated editing (7). Elevated retron-mediated gene editing in all-RNA system with sgRNA spike-in [001114] In all experiments above, retron ncRNA fused with sgRNA of Cas9 nuclease was used. This fusion RNA serves as a template of RT and at same time sgRNA portion could complex with Cas9 nuclease. Two enzymes acting on a single RNA piece could create steric effects that affect the activity of either enzyme. In line with this hypothesis, frequency of indels that tells overall Cas9 nuclease activity is only about 1.5 % with 400ng of ncRNA-sgRNA fusion (FIG.46, right). [001115] To test systematically the hypothesis, Cas9 nuclease activity by frequency of indels was compared between equimolar amounts of ncRNA-sgRNA fusion and separated sgRNA. When electroporated with 200 ng of Cas9 mRNA, 1 µg (7.5 pmol) of ncRNA-sgRNA fusion shows 20-fold lower activity than 0.266 µg (7.5 pmol) of separated sgRNA alone (FIG. 47) The result suggests that ncRNA-sgRNA fusion might inhibit either formation of complex with Cas9 protein or the activity of Cas9-sgRNA complex or both. In addition, chemically modified sgRNA shows 6-fold higher activity than unmodified sgRNA. These results indicate that affected Cas9 cleavage activity by ncRNA-sgRNA fusion could limit precise editing and addition of separated sgRNA could compensate for it. Modification of separated sgRNA is expected to further enhance precise editing. [001116] FIG.48 summarizes the assay of sgRNA spike-in in all RNA system and Fig. R. shows the results using Eco3 retron. At 0.2 µg of Cas9 mRNA and 0.5 µg of RT mRNA, the amount of sgRNA spike-in is titrated at 50, 100, and 200 ng (Fig. R, left). The titration was performed at two different ratios of RT mRNA: ncRNA-sgRNA fusion=1:6 or 1:8. With addition of 50 ng of sgRNA spike-in to 1:6 ratio, the precise editing was achieved 40-fold and gradually increased with more sgRNA, reaching up to 50-fold increase at 200ng.1:8 ratio of RT mRNA:ncRNA-sgRNA fusion responded similarly to sgRNA spike-in and obtained slightly higher activity than 1:6, up to 13% of precise editing. Higher precise editing with sgRNA spike- in came along with higher indels on the right graph. [001117] The effect of sgRNA spike-in in precise editing was confirmed using orthogonal delivery method, lipofection (FIG.50). Although overall amount of RNA to transfect was five times lower than in electroporation, 3.4% of precise editing was obtained without sgRNA spike- in, which is 7.5-fold higher than that achieved by electroporation (FIG.49 and FIG.51, left graph). With addition of sgRNA as low as 2 ng, precise editing further increased up to 3.5-fold, reaching 12% efficiency (FIG.51, left). Increasing the amount of sgRNA up to 10 ng did not further change the efficiency. The precise editing observed is dependent on retron since the absence of retron components (RT mRNA and ncRNA-sgRNA fusion) completely abrogated the editing. [001118] These data support that physical fusion of ncRNA and sgRNA of Cas9 nuclease could be a limiting factor for Cas9 activity and consequently precise editing and either spike-in of sgRNA demonstrated here or separation of ncRNA and sgRNA would be an improved strategy for efficient precise editing. Separation of ncRNA and sgRNA [001119] The impact of separating sgRNA from ncRNA on editing was tested in Eco3 retron system comparing the editing efficiency between ncRNA-sgRNA fusion and separated ncRNA and sgRNA side by side (FIG.52). Increasing amount of sgRNA was added to 300 ng of ncRNA that is equimolar amount of 400 ng of ncRNA-sgRNA fusion. With no sgRNA added, no precise editing was observed as expected. With 10 ng of sgRNA, precise editing peaked at 2.23% compared to 1.78% obtained with ncRNA-sgRNA fusion (FIG.52, left). The increasing amount of sgRNA beyond 10 ng did not further improve precise editing. Frequency of indels (FIG.52, right) showed a similar trend as precise editing. [001120] These data demonstrates that separation of ncRNA and sgRNA could achieve comparable or even higher precise editing than ncRNA-sgRNA fusion. Materials and Methods: [001121] Mammalian cell culture HEK293T (ATCC CRF-3216) or 293T-Cas9 (Genecopoeia SL502) cells were cultured in Dulbecco’s modified Eagle’s medium (DMEM) plus GlutaMAX (Thermo Fisher Scientitic), supplemented with 10% (v/v) fetal bovine serum (Gibco). Cells were maintained at 37ºC with 5% CO2. [001122] Genomic DNA extraction After incubation, the media was removed from the cells and genomic DNA was extracted by the addition of prepGEM reagent (Thomas Scientific: PUN0050) directly into each well of the tissue culture plate. Lysed cells were transferred to a 96 well PCR plate and incubated at 72ºC for 10 minutes, followed by 95ºC enzyme inactivation step for 2 minutes. [001123] High-throughput DNA sequencing of genomic DNA samples Human EMX1 gene locus was amplified from genomic DNA samples and sequenced on an Illumina NextSeq. Briefly, amplification primers containing Illumina forward and reverse adapters were used for a first round of PCR (PCR1) amplifying EMX1 targeting site.25 µl PCR1 reactions were performed with 0.3 µM of each forward and reverse primer, 1 µl of genomic DNA extract and 12.5 µl of KAPA HIFI HOTSTART PCR master mix. PCR reactions were carried out as follows: 95 ºC for 1 minute, then 25 cycles of [98ºC for 20 seconds, 65ºC for 15 seconds, and 72ºC for 15 seconds], followed by a final 72ºC extension for 2 minutes. PCR reactions were purified using Ampure XP beads (Beckman Coulter) and eluted in 20 µl H20. Unique Illumina barcoding primer pairs were added to each sample in a secondary PCR reaction (PCR2). Specifically, 25 µl PCR2 reactions were performed with 5 µl of IDT for Illumina UDI primers (Illumina), 1 µl of purified PCR1 reaction, and 12.5 µl of KAPA HIFI HOTSTART PCR master mix. PCR reactions were carried out as follows: 95 ºC for 3 minutes, then 10 cycles of [95ºC for 30 seconds, 55ºC for 30 seconds, and 72ºC for 30 seconds], followed by a final 72ºC extension for 5 minutes. PCR2 reactions were purified by SequalPrep Normalization plate kit (Thermo Fisher Scintific) and pooled. Size and purity were evaluated by Tape station D1000 assay (Agilent). DNA concentration was measured by fluorometric quantification (Qubit, ThermoFisher Scientitfic) and libraries were sequenced with 30% PhiX sequencing control on an Illumina NextSeq 2000 instrument using P1 or P2300 cycle kit. Sequencing reads were demultiplexed and alignment of amplicon sequences to a reference sequence was performed using CRISPresso2. CRISPresso2 was run in HDR mode using the desired allele as the expected allele and precise editing yield was calculated as the number of HDR aligned reads divided by total aligned reads. [001124] Junction qPCR for GFP integration qPCR was used to determine presence of GFP at the EMX1 locus post editing. Primers were designed to flank the 5’ and 3’ junction of GFP insertion at EMX1 locus. Briefly, 25 µl qPCR reactions were performed with 12.5 µl of KAPA HiFi HotStart ReadyMix (Roche), 0.3 µM of each forward and reverse primer, 2 µl of gDNA, 1X of SYBR Green (Thermo Fisher Scientific: S7563), and 1X of Rox Reference Dye (Thermo Fisher Scientific:54881). qPCR reactions were carried out as follows: 50 ◦C for 2 minutes, 95◦C for 3 minutes, then 40 cycles of [98◦C for 30 seconds, 67◦C for 30 seconds, and 72◦C for 30 seconds], 72◦C extension for 2 minutes, followed by a melt curve analysis with 95 ◦C for 15 seconds, 60 ◦C for 1 minute and 95 ◦C for 1 second. Delta Ct was obtained by subtracting edited samples’ Ct value from negative/unedited samples’ Ct value to relatively quantify GFP insertion at EMX1 locus. Additionally, qPCR samples were run on Tapestation 4200 (Agilent) with High-Sensitivity D1000 Reagents (Agilent) to confirm band size at expectation. [001125] Plasmid based gene editing Plasmids encoding retrons were synthesized by Twist Bioscience. For transfections, 25,000293T-Cas9 cells (Genecopoeia: SL502) were transfected using Lipofectamine 3000 (Thermofisher: L3000001) according to the manufacturer’s protocol. Transfected cells were incubated at 37ºC for 72 hrs before lysis in prepGEM reagent (Thomas Scientific: PUN0050) according to the manufacturer’s protocol. A 1 uL aliquot of crude lysate was used as template for amplification of the EMX1 CRISPR target site by PCR. Illumina adaptors and UDIs were added in a second round of PCR before loading onto an Illumina NextSeq. The resulting FASTQ files were used as input for CRISPResso2 which quantified precise editing and indels at the EMX1 locus. [001126] In vitro transcription Plasmid templates were synthesized by Twist Bioscience. T7 promoter was inserted upstream of RT or ncRNA sequences. Plasmid was linearized by Sbf1- HF enzyme (NEB, R3642S) or EcoR V (NEB, R0195L) for 16 hour at 37ºC and the complete linearization was checked by agarose gel electrophoresis. Linearized template was purified by GeneRT PCR purificiation kit (K0701, Thermo Fisher Scientific) and quantified by Nanodrop. mRNA was synthesized for 16 hour at 37ºC using HiScribe T7 mRNA kit with CleanCap Reagent AG (NEB, E2080S) and ncRNA using HiScribe T7 High Yield RNA synthesis kit (NEB, 2040S) following manufacturer’s protocol. After DNAse1 treatment, RNA was purified using Qiagen RNeasy midi kit (Qiagen, 75144). RNA concentration was measured by Nanodrop (Thermo Fisher) and purity and size were evaluated by RNA ScreenTape analysis on Tape station (Agilent). RT mRNA was further poly (A) tailed by E.Coli Poly(A) polymerase (NEB, M0276) and purified by Qiagen RNeasy midi kit. Tail length was estimated by RNA ScreenTape analysis by comparing before and after poly (A) adenylation. [001127] RNA Transfection by Neon Electroporation system HEK293T (ATCC CRF- 3216) or 293T-Cas9 (Genecopoeia SL502) cells were harvested and resuspended in R buffer. 50,000 cells with 1 µl of RNA mixture were electroporated by 10 µl Neon tip at 1150 voltage, 20 pulses. Electroporated cells were gently transferred to 96 well plate (VWR) containing culture media and cultured for 72 hours before genomic DNA extraction. [001128] RNA Transfection by Lipofectamine MessengerMAX 20,000 HEK293T (ATCC CRF-3216) cells were seeded in 96 well plates.16-24 hours post-seeding, cells were transfected with 0.3 µl of Lipofectamine MessengerMax (Thermo Fisher Scientific) with appropriate amount of RNA mixture. Cells were cultured for 72 hours before genomic DNA extraction. Example 5: improved ncRNAs generated by transcription from template linearized plasmid having blunt ends show higher precise editing efficiency [001129] Previously made in vitro transcription experiments to produce ncRNA used a double-stranded DNA template containing a 3’ overhang (on same strand as T7 promoter sequence). A new template with a blunt end was designed and tested. See FIG.52. As shown in FIG.53, four RNAs (cas9 mRNA, Eco3 RT mRNA, gRNA targeting EMX1 locus and ncRNA) were transfected into 293T cells using MessengerMAX lipofection reagent. Cells were harvested after 3 days, genomic DNA isolated, the EMX1 locus amplified, the Illumina libraries produced and sequenced on NextSeq. The percent of precise edits improves more than 5-fold for ncRNA produced using blunt end template as compared to the precise editing by ncRNA produced using overhang template. Example 6: improved ncRNAs due to improved in vitro template optimization and ncRNA end protection with MS2 hairpin [001130] Additional elements can be added to the ncRNA by encoding them on the plasmid template. FIG.53 shows a modified Eco3 ncRNA that comprises an MS2 stem loop hairpin structure added to its 3’ end. As shown in FIG.54, the precise editing of ncRNA comprising the 3’ MS2 structure resulted in nearly 15-fold increase in precise editing compared to an ncRNA generated from a overhang template, and nearly 3-fold increase in precise editing compared to an ncRNA generated from a blunt end template. [001131] FIG.53 schematic describes the modification made to improve RNA quality. First approach was about how to linearize plasmid DNA template for in vitro transcription (FIG.53, A). Two identical plasmids encoding Eco3 ncRNA were made except different restriction enzyme sites at 3’ of ncRNA: One generates 3’ overhang and the other generates blunt end. Templates with 3’ overhangs can cause the RNA polymerase to extend on the opposite strand, resulting in longer transcripts containing the sequence of the complementary strand (13). Second consideration was to add a MS2 hairpin at 3’ of ncRNA (FIG.53, B). Retron ncRNA contains mutually reverse complementary sequences at 5’ and 3 ends that forms a stem loop structure. However, these sequences could also mediate intermolecular interactions, creating higher-order structures. The addition of MS2 hairpin at one end could mitigate such intermolecular interactions. MS2 sequence is a bacteriophage-derived RNA aptamer that does not interact with endogenous proteins in mammalian cells. [001132] Testing these new RNAs in gene editing assay is shown in FIG.54. Four components of all RNA system (RT mRNA, Cas9 mRNA, ncRNA, sgRNA) were delivered to HEK293T cells by Lipofectamine MessengerMAX. All RNA was transfected at a fixed amount as shown on the graphs. Using RNA with the template containing a 3’ overhang produced 1.35% precise edits. Using the template containing a blunt end increased precise editing to 5.94%. Adding an MS2 stem loop to the 3’ end of the ncRNA further increased precise editing to 12.39%. On the right graph, frequency of indels is shown for respective conditions.Altogether, precise editing increased 9 fold by optimizing in vitro transcription template and 3’ end protection by MS2 hairpin. Example 7: modified ncRNAs with caps and/or tails [001133] ncRNA performance can be modified by adding a cap structure to the 5’ end and/or adding a poly(A) tail at the 3’ end, as depicted in FIG.55. As shown in FIG.56, using ncRNA with either or both protection by cap and tail lowered indels as compared uncapped/untailed ncRNAs. In this experiment, a 4 component all-RNA system (RT mRNA + Cas9 mRNA + ncRNA-sgRNA + sgRNA) was delivered to HEK293T cells by Lipofectamine MessengerMAX. All RNA was transfected at a fixed amount RT mRNA 100 ng, ncRNA- sgRNA 400ng, Cas9 mRNA 100 ng, and sgRNA 5 ng. ncRNA-gRNA fusion was either capped (+cap –tail) or poly-A tailed (-cap +tail) or both capped and poly-A tailed (+cap +tail). Using RNA without end protection (-cap –tail) produced ~4.5 % precise edits and the editing was dependent on retron since the absence of RT abrogated precise editing. Using RNA with either or both protection by cap and tail produced lower precise editing (left graph) but lowered indels (right graph) than without cap and tail. Example 8: Precise editing with Cas9 Nickase [0103] Wild Type (WT) Cas9 nuclease generates double strand break (DSB) that can potentially increase genomic instability by generating larger deletion or chromosome translocation. Moreover, DSB induces p53-mediated DNA damage response pathway that often compromises the physiology of edited cells. Single point mutations in either of Cas9 nuclease domains can generate nickases that cut only one strand of targeting site. D10A variant cuts target strand using intact HNH nuclease domain and H840A variant cuts non-target strand using intact RuvC domain. Nicks can initiate homology-directed repair (HDR) and cause less mutagenic end joining than DSBs. These features draw a considerable interest in using nickases in retron- mediated precise editing. [0104] FIG.73 shows the results testing Cas9 nickases activity in 4 component all-RNA system (RT mRNA + Cas9 mRNA + ncRNA + sgRNA) delivered to HEK293T cells by Lipofectamine MessengerMAX. D10A and H840A nickases were used in parallel with WT Cas9 and they were tested either with separated ncRNA or with ncRNA-sgRNA fusion. All RNA was transfected at a fixed amount RT mRNA 100 ng, ncRNA/ncRNA-sgRNA 400ng, Cas9 variant mRNA 100 ng, and sgRNA 5 ng. Precise editing by Cas9 WT (Wild Type) was ~9% when either ncRNA or ncRNA-sgRNA was used with additional sgRNA. With Cas9 H480A mutant that nicks non-target strand, activity was barely seen above background in any condition. With Cas9 D10A mutant that nicks target strand, precise editing was observed ~0.1% frequency in both separated and fused ncRNA and sgRNA format. Indels are shown at the right graph at respective conditions of left graph. As expected, indels are extremely low for either nickases with or without additional sgRNA and only WT Cas9 generated significant indels. [0105] FIG.74 shows negative controls that lack one of components (Cas9 variant or ncRNA/ncRNA-sgRNA or RT) in all RNA system. They were tested with or without additional sgRNA. All controls in top graph do not show precise editing activity beyond background level, supporting that precise editing achieved by Cas9 WT or Cas9 nickase is dependent on each component of all RNA system. Example 9: dual sgRNAs achieve higher precise editing than single sgRNA [0106] It was reasoned that use of multiple sgRNAs at the proximity of targeting site could generate more free DNA ends upon cleavage and recruit more repair machinery, that would provide higher chance of precise editing. Two pairs of sgRNA were found to test this hypothesis, sgRNA1 at immediately left side of insertion in combination with either sgRNA2 or sgRNA3 at the right side of insertion on EMX1 gene (FIG.68 schematic at right). Results of testing dual sgRNAs vs single sgRNA was shown in Fig.37. RTX2042 retron in all RNA system was delivered to HEK293T cells by lipofection. At given amount of RT mRNA, Cas9 mRNA and ncRNA-sgRNA, additional sgRNA was added. The same ncRNA-sgRNA with template encoded a 25 bp insertion on the Cas9 sgRNA’s non-targeting strand was used for all conditions. With one additional sgRNA (#1), precise editing was 0.27%, which increased up to 5 -fold to either 1.33% (#2) or 1.13% (#3) in the presence of a second sgRNA. On the bottom graph, frequency of indels is shown for respective conditions. [0107] These data demonstrates that dual sgRNA at the vicinity of targeting site could enhance the precise editing mediated by retron. Example 10. Editing with larger insert size [0108] Most of experiments so far tested 10 bp insertion at 3’ of EMX1 gene by retron- mediated precise editing. The feasibility of longer insertion was examined by designing new ncRNAs with gradually increased insert length: 25, 50, 75 and 100bp for Eco3, Aco1, RTX- 2042, and RTX-6983 retrons. Sequences to insert are randomly chosen that they do not share any homology with human genome and GC content is 50% for all inserts to generate no bias. At given amount of RT mRNA (100 ng) and Cas9 mRNA (100 ng), 400 ng of ncRNA-sgRNA fusion with different template lengths ranging from 10 to 100 bp were added and delivered to HEK 293T cells via lipofection. Overall, the efficiency is comparable cross the size of insert for all retrons. For 100 bp insertions, precise editing was achieved at 1.04% for Eco3, 1.52% for Aco1,1.37% for RTX2042 and 0.05% frequency for RTX6943 retron (FIG.59A). On the bottom row of graphs, frequency of indels for respective conditions shows a very similar trend to precise editing (FIG.59B). [0109] When sgRNA spike-in was added to above conditions (FIG.60A and 60B), the overall activity increased and activity was not compromised with longer inserts. For 100 bp insertions, precise editing increased 1.8~8 fold from without sgRNA spike-in, 1.82% for Eco3, 4.50% for Aco1,4.40% for RTX-2042 and 0.4% for RTX-6943. At the bottom panel, frequency of indels also increased for respective conditions. [0110] In combination with Cas9 nuclease, retron can insert 100 bp precisely at target site of the genome up to 4.5% efficiency. Example 11. Inserting a whole gene by Retron-mediated precise editing [0111] Correcting large mutation hot spot or insert a moderate size of gene precisely into the genome broadens the appilicability of Retron-mediated gene editing. To interrogate this, a GFP gene with its own promoter and poly-A signal (1267 bp, FIG.61A) was integrated in ncRNA of retron and gene editing assay was performed with three retrons, Eco3, Aco1 and RTX6943. As a readout of insertion, 5’ and 3’ junctions between EMX1 host and insert sequences were amplified by qPCR. ΔCt (Ct of without RT sample - Ct of with RT sample) value was used for relative quantification of integration. The bigger value of ΔCt indicates the higher frequency of insert in the sample. With all three retrons, amplicons of both 5’ and 3’ junctions are detectable significantly above background level (FIG.61B). Amplicon of 3’ junction is more abundant than that of 5’ junction probably because insertion at 3’ is facilitated by enriched 5’ end of msDNA from reverse transcription. FIG.61D shows PCR products from Aco1 retron analyzed by Agilent Tapestation High Sensitivity D1000 screentape. In the presence of RT (+RT sample, left on the gel), the expected size of amplicons were detected at both 5’ and 3’ junctions of edited alleles as indicated by arrows. Edited alleles were not detected in the absence of RT (-RT sample, right on the gel). Triangle indicates the expected amplicon for unedited alleles, which is present in both - RT and +RT samples. In addition, FIG.61C shows qPCR data depicting GFP insertion at 5’ or 3’ junctions at EMX1 locus by two retrons. ncRNA-sgRNA engineered to contain GFP gene insert was transfected to HEK293T cells, together with RT mRNA of each retron, Cas9 mRNA, with/without additional sgRNA. Genomic DNA was analyzed by indicated primer pairs in the table to detect GFP gene insertion at 5’ or 3’ junctions on the edited allele. ΔCt was obtained by subtracting junction Ct value from EMX1 Ct value for relative quantification of GFP insertion at EMX1 locus. With all two retrons, amplicons of both 5’ and 3’ junctions are detectable significantly above background level. Amplicon of 3’ junction is slightly more abundant than that of 5’ junction probably because insertion at 3’ is facilitated by more enriched 5’ end of msDNA from reverse transcription. Thus, two different retrons have been shown to insert a whole gene by retron editing. [0112] In all, these results demonstrated the feasibility of inserting an entire gene by retron- mediated precise editing. Example 12. Order of sgRNA and ncRNA in fused RNA [0113] Next, the order of sgRNA and ncRNA in fusion was addressed. The order might play an important role in ensuring the activity of two enzymes (Cas9 and RT of retron) on one fused substrate. A. ncRNA-sgRNA and B. sgRNA-ncRNA order were compared for precise editing with Eco3, Aco1, and RTX-2042 retron (FIG.62A). For Eco3 and RTX-2042, sgRNA-ncRNA order allowed significantly higher activity for both precise editing (6- and 3- fold, respectively) and indels (3- and 2-fold respectively) than ncRNA-sgRNA order. The same order provided higher indels for Aco1 (2 fold), suggesting that sgRNA at 5’ of the fusion permits higher Cas9 activity. However, precise editing with Aco1 was comparable between two orders. [0114] When sgRNA spike-in was added to above conditions, the overall activity slightly increased (FIG.62B). The addition of sgRNA spike-in compensated inferior Cas9 activity with ncRNA-sgRNA (to that with sgRNA-ncRNA), so that precise editing and indels became comparable to the inverse order for all retrons. [0115] The precise editing was dependent on the presence of RT and Cas9 (FIG.63). When any one of these components is removed, precise editing is undetectable or at background levels with either order of ncRNA and sgRNA fusion (FIG.63 top graphs). This was consistent with all three retrons. On the bottom graphs, frequency of indels is shown for respective conditions (FIG. 63 bottom graphs). Example 13. Separation of ncRNA and sgRNA [0116] The addition of sgRNA spike-in to either order of ncRNA and sgRNA fusion increased precise editing and indels (FIGs.62A, 62B and FIG.63). These data suggest that physical fusion of ncRNA and sgRNA of Cas9 nuclease could be a limiting factor for Cas9 activity and consequently precise editing thus separation of ncRNA and sgRNA could be a better strategy for precise editing. [0117] The impact of separating sgRNA from ncRNA on editing was tested in Eco3, Aco1,RTX-2042 and RTX-6943 retron system comparing the editing efficiency between A. ncRNA-sgRNA or B. sgRNA-ncRNA fusion and C. separated ncRNA and sgRNA with or without additional sgRNA (FIG.64). With Eco3, the separation significantly outperformed (~5 fold) any order of fusion of ncRNA and sgRNA in precise editing. With no sgRNA added to separated ncRNA, no precise editing was observed, as expected. On the other hand, separated version of Aco1 and RTX-2042 ncRNAs achieved a comparable level of precise editing to fused RNA. Separated ncRNA of RTX-6943 showed lower activity than fused version but the overall activity of this retron is a magnitude lower than others. [0118] These data demonstrates that separation of ncRNA and sgRNA could achieve comparable or even higher precise editing than ncRNA and sgRNA fusion depending on the type of retron. Example 14. Reton mediated precise editing occurs at another genomic locus, AAVS1 [0119] All previous gene editing assays were done at EMX1 gene locus. With Eco3, Aco1, RTX2042 and RTX6943 retrons, AAVS1 locus was targeted for precise editing. AAVS1 locus, in intron 1 of PPP1R12C (protein phosphatase 1 regulatory subunit 12C) gene, is known as a “safe harbor”, because its disruption does not have adverse effects on the cell, and robust transcription of locus is permissive for stable expression of an exogenously inserted gene. Within msD stem loop of ncRNA from each retron, 25bp insertion donor sequence was incorporated and to the 3’ end of engineered ncRNA, sgRNA targeting AAVS1 was fused. [0120] FIG.65 shows the result of testing four retrons to edit AAVS1 locus. For all four retrons, precise editing was enhanced in the presence of additional sgRNA and ranged from 0.73% to 2.05%. On the right, frequency of indels at respective conditions shares a similar trend of precise editing. Removing either RT, ncRNA-sgRNA or Cas9 mRNA abrogated precise editing showing Retron and Cas9 specificity of editing (FIG.66), while indels are robust in the bottom graph. Example 15. Testing template strand bias for precise editing [0121] Cas9 uses different endonuclease domain to make cleavage on either sgRNA targeting or non-targeting strand. Differential release timing of each nuclease domains of Cas9 from DNA substrates after cutting might influence how single strand donor interact with broken DNA and repair pathway (12). [0122] Results of testing donor templates on the opposite strand relative to Cas9 sgRNA targeting strand are shown in FIG.67. Eco3 and Aco1 retrons in all RNA system were delivered to HEK293T cells by lipofection. The given amount of RT mRNA, Cas9 mRNA,additional sgRNA targeting EMX1 locus, and appropriate ncRNA-sgRNA described underneath the graph were added. All donor templates tested contained 25 bp insert. All ncRNAs used in previous experiments encoded the donor template of the strand targeted by the Cas9 sgRNA and denoted by the letter T. ncRNA that contains reverse complementary sequence of Cas9 targeting strand is denoted as NT. For Eco3, in the presence of additional sgRNA, precise editing was 1.67% when template was on the targeting strand and 0.85% when the template was on the non-targeting strand. For Aco1, in the presence of additional sgRNA, precise editing was 2.96% when template was on the targeting strand and 3.40% when the template was on the non-targeting strand. On the bottom row of graphs, frequency of indels is shown for respective conditions. [0123] At least in EMX1 locus, retron ncRNA encoding either Cas9 targeting or non- targeting strand produced a similar level of precise editing. Group A Sequence - sequences used in the Examples (SUMMARY)
Figure imgf000430_0001
Figure imgf000431_0001
Group A Sequence - sequences used in the Examples EMX1 s.p Cas9 gRNA GAGTCCGAGCAGAAGAAGAA (SEQ ID NO: 19109) Eco1 ncRNA + HDR_Template TGATAAGATTCCGTATGCGCACCCTTAGCGAGAGGTTTATCATTAAGGTCAACCTCT GGATGTTGTTTCGGCATCCTGCATTGAATCTGAGTTACTGTCTGTTTTCCTACAAACG GCAGAAGCTGGAGGAGGAAGGGCCTGAGTCCGAGCAGAAGAAAAAGTTCTCC CATCACATCAACCGGTGGCGCATTGCCACGAAGCAGGCCAATGGGGAGGACAT CGATGTCAAGGAAACCCGTTTCTTCTGACGTAAGGGTGCGCATACGGAATCTTATC A (SEQ ID NO: 19110) Aco1 ncRNA + HDR Template CCGTAGTGGGAGCCTCAGGCGAGGGTGTGTATCATGCCCGTTCTGCCAAGACCCACC AAAGAAGGGCACCGTGGAGGACAAACGGCAGAAGCTGGAGGAGGAAGGGCCTG AGTCCGAGCAGAAGAAACAGAAGTGCAAAGTTCTCCCATCACATCAACCGGTG GCGCATTGCCACGAAGCAGGCCAATGGGGAGGACATCGATGTCACCTCCACGG TGCAATGCGAAAGCAACTTGAGGCTTTGCTTAGTATGAGGCTCCCACTACGG (SEQ ID NO: 19111) RTX3_2042 RT MKDDQYSQWKKYYESRGILPEIQDKLLNYAKIHIDNNTPVIFNFEHLTLLLGREKNYLSS VVNSPDSHYRKFKIKKRSGGEREITAPYLSLLEMQYWIYRNILINVKIHYAAHGFAQDKS IITNSRNHLGQKHLLKMDLKDFFPSIKLNRIIYIFKSLGYPNIIAFYLASICSYKGHLPQGSP TSPILSNIVSITLDNRLVKFARKMKLRYSRYADDLTFSGDKIPTNYIKYITDIINDEGFEVN DTKTKLYLKAGKRIVTGISVIGNDPKLPREYKRKLKQELHYIFTYGIGSHMAKKKIKKIN YLYRIIGKVNFWLNIEPDNEYARNAKAKLLLLIDN* (SEQ ID NO: 19112) RTX3_2042 ncRNA + HDR_Template GTTAAGGTGGTTATATTCTAGTATTTATGAAGTGTAGTCGCTTCGATCGTTAAGGCTG ATTTTAACCTCTGCATAATAATATCGGTAGATATTATTATGCACGCTCCCTTTAGCAG AGCTAAGAATCGCTCACTCAGGCACAAGCTTTGAGGACAAACGGCAGAAGCTGGA GGAGGAAGGGCCTGAGTCCGAGCAGAAGAATCCTAAGAGAAAAGTTCTCCCAT CACATCAACCGGTGGCGCATTGCCACGAAGCAGGCCAATGGGGAGGACATCGA TGTCACCTCAAAGCTTGTGCCTGAGTGAGAGCTAAAGAAAAGAAAAGTAGAATAAG CCACCTTAAC (SEQ ID NO: 19113) RTX3_6083v1 RT MSNPQPTRAEIFERIKQSSKQEVILEEMQRLGFWPRSEGQPEVAADLIQREGELQRELAE LNKKLAVKRNPERALREMRKQRMKDARDKREVTKRAQAQQRYDKALLWHEKRASHV AYLGPGVSASLHENSSATQEQGDKGKPKRARDRAVPDLQRLTLNGLPALISAAQLAES MGVSVAELRFLSFHREVARTNHYHSFTLPKKTGGERLISAPMPRLKRAQYWVLDNVLA KMPAHDAAHGFLAGRSIISNAKPHAGQDVVINLDVKDFFPSIAFGRIKGVFRQLGYGESI ATVFALLCSENRAQAWQVDGERLFVGGKARERVLPQGAPTSPMLTNLLCRRMDRRLL GLAKQLGFVYTRYADDLTFSASGEPARDNVGKLLSRVRWILRDEGFTPHPDKERVMRK GRRQEVTGLVVNSDTPSVSRETRRRLRAALHRASQPDAASKPAHWQGHTAQPSQLLGL ATFVHQIDPKQGKTLLADAQQLMRSPIDRANDAAKSASRADAAQQSFRVLAAAGKPPV LADGKNWWQPAPPATPVLEKTDQQRREERQATRRQQAAAAAPPPSSTRRNERPQQAA HEQQGDAQPQNEAPPRFDPDQYAPPPRNVMTYWAQIAISFFLGSILHNRLITIFAMVAVI ALYYMRRQRWDVFMGILVVATLLGYLVRGMG* (SEQ ID NO: 19114) RTX3_6083v1 ncRNA + HDR Template GCTCCGGAGCAATGAGCAGGCTCTTGCAATCCGGGCGGTGTTTCGCCGCCCTTGTGA ACTGCCGTTTCATGCACCACGGGCGCCGTTTTCACTGTGCCACCCCAGCCACGGTAG TGCTACAAACGGCAGAAGCTGGAGGAGGAAGGGCCTGAGTCCGAGCAGAAGAA CTACAAGGTTAAAGTTCTCCCATCACATCAACCGGTGGCGCATTGCCACGAAG CAGGCCAATGGGGAGGACATCGATGTCAAGCACTACCGTGGCGGGGTGGCGTCG AGCGAACAGCTCCCGTCCCGTGAGCCCTACAGGCTCTTCGACGAGATGCACATTGCT CCGGAGC (SEQ ID NO: 19115) RTX3_6943 RT MEESTNYKLLVWGLSVIQPATPNEVLNYLTSTLNDNGLLPDVEKMIHYFELLDQLGYIH QVSKRNNLYSLTPRGNERLTPALKRLRDKIRLFMLDNCHSISKLGVLASTDTENMGGDS PSLQLRHNLKEVPHPSLSWAAGTLPSSPRQAWVRIYEQLNIGSMSSDEASTPTTARNAPL SFVGRLGFSLNYYSFNKIDEPLFNNDGVTAIASCIGISPGLITAMVKSPKRYYRTFNLRKK SGGFRSILAPRKFIKTIQYWLKDHVLNRLKIHSSCYSYRSGVSIKDNAINHVKKKFVASID ISDYFGSINKKMVKDCFYKNNIPDHIVNTISGIVTYNDVLPQGAPTSPIISNAILFEFDEEM TAHALTLDCIYTRYSDDISISSDYKENIAILINIAEANLLSAGFTLNRQKQRIASDNSRQVV TGILVNESIRPTRCYRKKIRSAFDHALKEQDGSQLTINKLRGYLNYLKSFETYGFKFNEK KYKETLDFLIALKQS* (SEQ ID NO: 19116) RTX3_6943 ncRNA + HDR Template GCGGAGTGCTGGCCTCAACTGATACAGAGAATATGGGCGGTGATTCGCCGTCTTTAC AGTTAAGGCACAATTTAAAAGAGGTTCCGCACCCAAGCCTGTCTTGGGCTGCACAA ACGGCAGAAGCTGGAGGAGGAAGGGCCTGAGTCCGAGCAGAAGAACGTATGG CCTAAAGTTCTCCCATCACATCAACCGGTGGCGCATTGCCACGAAGCAGGCCA ATGGGGAGGACATCGATGTCAGCAGCCCAAGACAGGCTTGGGTGCGGATCTACGA GCAATTAAATATTGGTTCGATGTCCAGTGATGAGGCCAGCACCCGC (SEQ ID NO: 19117) Eco1 RT > Coding sequence only. RNA was produced from a CRO and proprietary 5' cap, 5' UTR, 3' UTR and encoded polyA of around 120 bases were added. Full substitution with 1N- methyl-pseudouridine) AUGAAAUCUGCAGAGUAUCUGAAUACGUUCCGCCUUAGGAAUUUGGGCCUCCCCG UGAUGAACAAUCUCCACGAUAUGAGCAAGGCGACUCGAAUAUCCGUGGAAACGC UGAGACUGCUCAUCUAUACAGCAGACUUUCGGUACAGGAUCUACACGGUCGAAA AGAAGGGGCCUGAGAAACGCAUGCGAACAAUUUAUCAACCUAGCCGAGAGCUCA AGGCGUUGCAGGGCUGGGUUCUUCGAAACAUCCUUGACAAACUCUCAUCAUCACC CUUUAGUAUUGGGUUUGAAAAGCACCAAAGCAUCCUUAACAACGCGACGCCACAC AUAGGUGCCAAUUUCAUAUUGAACAUCGACUUGGAGGAUUUUUUUCCGAGCCUC ACAGCCAAUAAAGUGUUCGGUGUUUUUCACAGUCUUGGGUACAAUCGCCUUAUU AGUUCCGUUCUUACCAAGAUUUGUUGUUACAAGAAUCUCUUGCCCCAGGGAGCAC CCAGCAGUCCGAAAUUGGCGAAUUUGAUUUGUUCCAAGCUCGAUUAUCGAAUAC AAGGGUACGCGGGCAGCCGGGGACUCAUCUAUACCCGCUACGCAGACGAUCUUAC GCUGUCUGCCCAAUCAAUGAAGAAGGUCGUAAAGGCGCGGGAUUUCUUGUUUUC UAUCAUCCCGUCCGAGGGCUUGGUAAUUAAUUCCAAAAAGACUUGUAUCUCAGG ACCACGAUCUCAGCGAAAAGUGACAGGACUCGUCAUUUCUCAAGAAAAAGUCGG UAUAGGGAGAGAGAAGUAUAAGGAAAUCCGCGCGAAGAUCCACCACAUAUUCUG UGGCAAGAGCAGCGAGAUAGAACACGUCCGAGGCUGGUUGUCCUUCAUACUGAG CGUGGACUCAAAAAGCCACCGCCGGUUGAUCACCUAUAUUUCAAAACUGGAAAAG AAAUAUGGAAAGAACCCACUCAACAAAGCUAAAACACCACCAAAGAAGAAAAGA AAGGUCUGA (SEQ ID NO: 19118) Eco1 ncRNA ncRNA-sgRNA fusion (6 bp substition in EMX1 gene) GAAAUGAUAAGAUUCCGUAUGCGCACCCUUAGCGAGAGGUUUAUCAUUAAGGUC AACCUCUGGAUGUUGUUUCGGCAUCCUGCAUUGAAUCUGAGUUACUGUCUGUUU UCCUACAAACGGCAGAAGCUGGAGGAGGAAGGGCCUGAGUCCGAGCAGAAGAAA AAGUUCUCCCAUCACAUCAACCGGUGGCGCAUUGCCACGAAGCAGGCCAAUGGGG AGGACAUCGAUGUCAAGGAAACCCGUUUCUUCUGACGUAAGGGUGCGCAUACGG AAUCUUAUCAGAGUCCGAGCAGAAGAAGAAGUUUCAGAGCUAUGCUGGAAACAG CAUAGCAAGUUGAAAUAAGGCUAGUCCGUUAUCAACUUGAAAAAGUGGCACCGA GUCGGUGCUUUUUUUUUUUU (SEQ ID NO: 19119) Eco3 RT > Coding sequence only. RNA was produced from a CRO and proprietary 5' cap, 5' UTR, 3' UTR and encoded polyA of around 120 bases were added. Full substitution with 1N- methyl-pseudouridine AUGCGCAUUUACUCUCUGAUCGACAGCCAAACCUUAAUGACCAAAGGGUUCGCAU CCGAGGUCAUGAGGAGCCCAGAACCCCCUAAGAAGUGGGACAUUGCGAAGAAGA AGGGCGGAAUGCGUACGAUAUACCAUCCCUCUUCUAAGGUGAAGCUGAUACAGU ACUGGCUGAUGAACAACGUGUUCUCCAAAUUGCCGAUGCACAACGCCGCGUACGC UUUCGUGAAGAAUAGAUCUAUCAAGUCUAACGCACUGCUGCACGCAGAGAGUAA GAACAAAUACUACGUUAAGAUUGACCUGAAGGACUUCUUUCCAAGCAUCAAGUU CACAGACUUCGAAUAUGCCUUUACCCGGUACCGUGACAGAAUAGAGUUCACGACC GAGUACGACAAAGAACUGCUUCAGCUGAUUAAGACCAUUUGUUUCAUUUCUGAC UCUACACUGCCAAUAGGCUUCCCCACUUCCCCUCUUAUAGCCAAUUUCGUCGCCA GGGAGCUGGACGAGAAGCUCACUCAGAAGCUGAACGCUAUAGACAAGCUCAACGC UACGUACACUCGCUACGCAGACGACAUAAUCGUGAGCACGAACAUGAAGGGCGCC UCUAAGCUGAUCUUAGACUGCUUCAAGCGGACCAUGAAGGAAAUCGGACCCGAU UUCAAGAUCAAUAUCAAGAAGUUCAAAAUAUGCUCUGCCAGUGGCGGCUCAAUU GUCGUGACGGGCCUUAAGGUCUGUCAUGACUUCCACAUAACUCUGCACCGGUCUA UGAAGGACAAGAUCCGCCUGCACCUCUCUCUCCUGUCCAAAGGUAUUCUGAAGGA CGAGGACCACAACAAGCUGUCCGGGUACAUCGCCUACGCUAAGGACAUCGAUCCA CACUUCUACACCAAGCUCAAUAGGAAGUACUUCCAGGAGAUCAAGUGGAUACAA AACCUGCAUAAUAAGGUGGAGCCACCAAAGAAGAAAAGAAAGGUCUGA (SEQ ID NO: 19120) Eco3 ncRNA ncRNA-sgRNA fusion (10 bp insertion in EMX1 gene) GAAAUGAUAAGAUUCCGUAAGAGCCAAACCUAGCAUUUUAUGGGUUAAUAGCCC AUCGGGCCAUGAGUCAUGGUUUCGCCUAGUAUUUUAGCUAUGCCCGUCGUUCAG UUCGCUGAACAAACGGCAGAAGCUGGAGGAGGAAGGGCCUGAGUCCGAGCAGAA GAAUUACGUCUGCAAAGUUCUCCCAUCACAUCAACCGGUGGCGCAUUGCCACGAA GCAGGCCAAUGGGGAGGACAUCGAUGUCAUCAGCGAACUGAUCGACGUGCUCAA GUAGGUUUGGCUCUUACGGAAUCUUAUCAGAGUCCGAGCAGAAGAAGAAGUUUC AGAGCUAUGCUGGAAACAGCAUAGCAAGUUGAAAUAAGGCUAGUCCGUUAUCAA CUUGAAAAAGUGGCACCGAGUCGGUGCUUUUUUUUUUUU (SEQ ID NO: 19121) Eco5 RT > Coding sequence only. RNA was produced from a CRO and proprietary 5' cap, 5' UTR, 3' UTR and encoded polyA of around 120 bases were added. Full substitution with 1N- methyl-pseudouridine AUGGACGCUACCAGAACGACUCUCCUUGCAUUGGAUCUCUUCGGAUCUCCAGGUU GGUCCGCCGAUAAAGAAAUUCAGAGGCUUCAUGCGCUCAGUAAUCAUGCUGGAA GGCAUUACAGAAGGAUUAUAUUAAGUAAAAGGCACGGCGGACAGCGUCUUGUGC UUGCACCUGAUUACUUGUUAAAGACCGUUCAGCGCAACAUUUUGAAGAACGUUU UGAGUCAAUUUCCACUGUCACCAUUUGCUACAGCCUACAGACCGGGAUGCCCAAU CGUGUCUAACGCGCAGCCACACUGCCAACAGCCACAGAUCUUGAAACUCGAUAUA GAAAACUUCUUCGAUUCUAUUAGUUGGUUGCAGGUGUGGCGGGUGUUUCGCCAG GCCCAGUUGCCCCGAAAUGUCGUAACGAUGCUCACUUGGAUAUGUUGUUAUAAC GACGCACUUCCGCAGGGUGCCCCUACAUCCCCUGCAAUUUCCAAUCUCGUCAUGA GAAGGUUUGAUGAACGGAUUGGAGAAUGGUGUCAGGCUCGAGGGAUUACCUACA CUCGCUACUGCGAUGACAUGACGUUUAGUGGACACUUCAAUGCAAGGCAGGUCA AGAAUAAAGUCUGCGGUCUCUUAGCUGAGCUGGGCCUUUCCCUGAAUAAACGGA AAGGCUGCCUCAUAGCGGCUUGUAAGCGCCAGCAAGUCACCGGCAUUGUUGUGAA UCACAAGCCACAGCUUGCCCGAGAAGCCAGGCGUGCCCUGCGUCAGGAAGUGCAC CUGUGCCAGAAAUAUGGAGUUAUCUCUCAUCUCUCACAUAGAGGUGAACUGGAU CCUAGCGGAGAUCUGCACGCUCAGGCGACAGCGUAUCUCUAUGCACUCCAGGGGA GAAUUAACUGGCUUCUUCAAAUUAACCCUGAGGAUGAGGCGUUUCAACAGGCCC GGGAGUCCGUUAAGAGGAUGUUAGUUGCCUGGCCACCAAAGAAGAAAAGAAAGG UCUGA (SEQ ID NO: 19122) Eco5 ncRNA > ncRNA-sgRNA fusion (10 bp insertion in EMX1 gene) GAAAUGAUAAGAUUCCGUACGCCAGCAGUGGCAAUAGCGUUUCCGGCCUUUUGU GCCGGGAGGGUCGGCGAGUCGCUGACUUAACGCCAGUAGUAUGUCCAUAUACCCA AAGUCGCUUCAUUGUAACAAACGGCAGAAGCUGGAGGAGGAAGGGCCUGAGUCC GAGCAGAAGAAUCCUUCGAGCAAAGUUCUCCCAUCACAUCAACCGGUGGCGCAUU GCCACGAAGCAGGCCAAUGGGGAGGACAUCGAUGUCAUACAGUUACGCGCCUUCG GGAUGGUUUAAUGGUAUUGCCGCUGUUGGCGUACGGAAUCUUAUCAGAGUCCGA GCAGAAGAAGAAGUUUCAGAGCUAUGCUGGAAACAGCAUAGCAAGUUGAAAUAA GGCUAGUCCGUUAUCAACUUGAAAAAGUGGCACCGAGUCGGUGCUUUUUUUUUU UU (SEQ ID NO: 19123) EMX1 sgRNA modified > Contains chemical modifications from Synthego: 2'-F, 2'-O-methyl, phosphorothioates GAGUCCGAGCAGAAGAAGAAGUUUCAGAGCUAUGCUGGAAACAGCAUAGCAAGU UGAAAUAAGGCUAGUCCGUUAUCAACUUGAAAAAGUGGCACCGAGUCGGUGCUU UUU (SEQ ID NO: 19124) EMX1 sgRNA unmodified > No modifications GAGUCCGAGCAGAAGAAGAAGUUUCAGAGCUAUGCUGGAAACAGCAUAGCAAGU UGAAAUAAGGCUAGUCCGUUAUCAACUUGAAAAAGUGGCACCGAGUCGGUGCUU UUU (SEQ ID NO: 19125) Group B Sequence - Additional sequences used in the Examples (SUMMARY CHART)
Figure imgf000436_0001
Figure imgf000437_0001
Figure imgf000438_0001
Figure imgf000439_0001
Figure imgf000440_0001
Figure imgf000441_0001
Figure imgf000442_0001
Group B Sequences - Additional sequences used in the Examples
Figure imgf000443_0001
Figure imgf000444_0001
Figure imgf000445_0001
Figure imgf000446_0001
Figure imgf000447_0001
Figure imgf000448_0001
Figure imgf000449_0001
Figure imgf000450_0001
Figure imgf000451_0001
Figure imgf000452_0001
Figure imgf000453_0001
Figure imgf000454_0001
Figure imgf000455_0001
Figure imgf000456_0001
Figure imgf000457_0001
Figure imgf000458_0001
Figure imgf000459_0001
Figure imgf000460_0001
Figure imgf000461_0001
Figure imgf000462_0001
Figure imgf000463_0001
Figure imgf000464_0001
Figure imgf000465_0001
Figure imgf000466_0001
Figure imgf000467_0001
Figure imgf000468_0001
Figure imgf000469_0001
Figure imgf000470_0001
Primer sequence
Figure imgf000471_0001
References cited in Examples Are we creating a new phenotype? MF, Dohrn.2021, Neurological Research and Practice. Val50Met hereditary transthyretin amyloidosis:not just a medical problem but a psychosocial burden. J, Gonzalez-Moreno. s.l. : Orphanet Journal of Rare Diseases, 2021, Vol.16. synthetic DNA delivery systems. D, Luo.2000, Nature Biotechnology, pp.33-37. rAAV immunogenicity, toxicity, and durability in 255 clinical trials:A meta-analysis. W, Shen. 2022, Frontiers in Immunology. Reverse transcriptase associated with the biosynthesis of the branched RNA-linked msDNA in myxococcus xanthus. S, inouye. s.l. : Cell, 1989, Vol.56.709-717. Bacterial retrons funciton in anti-pahge defense. A, Millman. s.l. : Cell, 2020, Vol.183.1551- 1561. Search-and-replace genome editing without double-strand breaks or donor DNA. AV, anzalone.
2019, Nature, pp. 149-157.
Branched RNA covalently linked to the 5' end of a single-stdnaded DNA in Stigmatella aurantiaca. T, Furuichi. 1, s.l. : Cell, 1987, Vol. 48. 47-53.
Retrons and their applications in genome engineering. AJ, Simon. 21, s.l. : Nucleic Acids Research, 2019, Vol. 47.
Systematic prediction of genes functionally associated with bacterial retrons and classification of the encoded tripartite system. MR, Mestre. 22, s.l. : Nucleic Acids Research, 2020, Vol. 48.
Precise genome editing across kingdoms of life using retron-derived DNA. SC, lopez. 2, s.l. : Nature Chemical Biology, 2021, Vol. 18.

Claims

WHAT IS CLAIMED: 1. A pharmaceutical composition for gene editing comprising a delivery vehicle comprising RNA cargo, said RNA cargo comprises (a) at least one mRNA molecule encoding (i) a nucleic acid programmable nuclease and (ii) a retron reverse transcriptase, (b) an engineered retron ncRNA, and (c) guide RNA for the programmable nuclease, wherein said RNA cargo is in an amount effective to edit a genome at a target site.
2. The pharmaceutical composition of claim 1, wherein the delivery vehicle comprises a liposome or a lipid nanoparticle (LNP).
3. The pharmaceutical composition of claim 2, wherein the delivery vehicle is a lipid nanoparticle comprising: a) one or more ionizable lipids; b) one or more structural lipids; c) one or more PEGylated lipids; and d) one or more phospholipids.
4. The pharmaceutical composition of claim 3, wherein the one or more ionizable lipids comprises an ionizable lipid set forth in Table 2.
5. The pharmaceutical composition of claim 1, wherein the at least one mRNA molecule encodes both nucleic acid programmable nuclease and the retron reverse transcriptase.
6. The pharmaceutical composition of claim 1, wherein the at least one mRNA molecule comprises a first mRNA molecule encoding the nucleic acid programmable nuclease and a second mRNA molecule encoding the retron reverse transcriptase.
7. A gene editing system comprising one or more delivery vehicles, wherein: the delivery vehicle(s) comprise RNA cargo, said RNA cargo comprises (a) at least one mRNA molecule encoding (i) a nucleic acid programmable nuclease and (ii) a retron reverse transcriptase, (b) an engineered retron ncRNA, and (c) guide RNA for the programmable nuclease, each delivery vehicle contains (a)(i) and/or (a)(ii) and/or (b) and/or (c), whereby one delivery vehicle or more than one delivery vehicle delivers (a)(i), (a)(ii), (b), and (c).
8. The gene editing system of claim 1, wherein (a)(i) and (a)(ii) comprise a single mRNA molecule encoding the nucleic acid programmable nuclease and the retron reverse transcriptase.
9. The gene editing system of claim 2, wherein (a)(i) and (a)(ii) are encoded and expressed as a fusion protein.
10. The gene editing system of claim 3, wherein the fusion protein comprises the C- terminal end of the nucleic acid programmable nuclease fused to the N-terminal end of the retron reverse transcriptase (nuclease:RT fusion).
11. The gene editing system of claim 3, wherein the fusion protein comprises the N- terminal end of the nucleic acid programmable nuclease fused to the C-terminal end of the retron reverse transcriptase (RT:nuclease fusion).
12. The gene editing system of claim 1, wherein (a)(i) and (a)(ii) comprise a first mRNA molecule encoding the nucleic acid programmable nuclease and a second mRNA molecule encoding the retron reverse transcriptase.
13. The gene editing system of claim 1, wherein (c) is separate from (a)(i), (a)(ii) and (b) or is provided in trans.
14. The gene editing system of claim 1, wherein (b) the engineered retron ncRNA, and (c) the guide RNA are fused or are provided in cis.
15. The gene editing system of claim 8, wherein the guide RNA is fused to the 5’ end of the retron ncRNA.
16. The gene editing system of claim 8, wherein the guide RNA is fused to the 3’ end of the retron ncRNA.
17. The gene editing system of claim 8, wherein the engineered ncRNA comprises a first guide RNA fused to the 5’ end of the retron ncRNA, and a second guide RNA fused to the 3’ end of the retron ncRNA, and the first and second guide RNAs target different sequences.
18. The gene editing system of claim 1, wherein the one or more delivery vehicles comprise a liposome or a lipid nanoparticle (LNP).
19. The gene editing system of claim 1, wherein (a) the at least one mRNA molecule encoding (i) the nucleic acid programmable nuclease and (ii) the retron reverse transcriptase, and (b) the engineered retron ncRNA are in the same delivery vehicle.
20. The gene editing system of claim 1, wherein the (a) the at least one mRNA molecule encoding (i) the nucleic acid programmable nuclease and (ii) the retron reverse transcriptase, and (b) the engineered retron ncRNA are in separate delivery vehicles.
21. The gene editing system of claim 1, wherein the nucleic acid programmable nuclease and the retron reverse transcriptase are encoded on separate mRNA molecules and those separate mRNA molecules of (a)(i) and (a)(ii) are contained in the same delivery vehicle.
22. The gene editing system of claim 1, wherein the nucleic acid programmable nuclease and the retron reverse transcriptase are encoded on separate mRNA molecules and those separate mRNA molecules of (a)(i) and (a)(ii) are contained in different delivery vehicles.
23. The gene editing system of claim 1, wherein the engineered retron ncRNA includes a heterologous sequence of interest encoding a donor polynucleotide comprising an intended edit to be integrated at a target sequence in a cell, and wherein the donor polynucleotide is flanked by a 5' homology arm that hybridizes to a sequence 5’ to the target sequence and a 3' homology arm that hybridizes to a sequence 3' to the target sequence.
24. The gene editing system of claim 1, wherein the nucleic acid programmable nuclease comprises a Cas9 nuclease, a TnpB nuclease, or a Cas12a nuclease.
25. The gene editing system of claim 18, wherein the nucleic acid programmable nuclease comprises a Cas9 nuclease is encoded by SEQ ID NO: 19132.
26. The gene editing system of claim 25, wherein the Cas9 is further modified to be a nickase.
27. The gene editing system of claim 1, wherein the retron reverse transcriptase has an amino acid sequence comprising any RT amino acid sequence of Table A.
28. The gene editing system of claim 1, wherein the retron reverse transcriptase has an amino acid sequence comprising SEQ ID NO: 19112 (RTX3_2042), SEQ ID NO: 19114 (RTX_6083v1), SEQ ID NO: 19116 (RTX_6943), or an amino acid sequence having at least 90%, 95%, 99%, or 100% sequence identity with SEQ ID Nos: 19112, 19114, or 19116.
29. The gene editing system of claim 1, wherein the engineered retron ncRNA comprises: A) a pre-msr sequence having a first complementary region of the retron ncRNA; B) an msr sequence including an msr stem-loop structure; C) an msd sequence including an msd stem-loop structure and a heterologous sequence of interest, wherein said msd sequence templates a single strand DNA product (RT-DNA) in the presence of the retron reverse transcriptase; and D) a post-msd sequence having a second complementary region, wherein the first and second complementary regions form an a1/a2 duplex region of the retron ncRNA, wherein the msr stem-loop structure, the msd stem-loop structure, or the a1/a2 duplex comprise a modification which result in increased editing efficiency in the presence of a nucleic acid programmable nuclease that associates with the one or more guide RNAs, and wherein optionally one or more of the guide RNAs of (c) are coupled to the pre-msr sequence, the post-msd sequence, or both the pre-msr sequence and the post-msd sequence.
30. The gene editing system of claim 23, wherein the heterologous sequence of interest encodes a donor polynucleotide comprising an intended edit to be integrated at a target sequence of a cell, wherein the donor polynucleotide is flanked by a 5' homology arm that hybridizes to a sequence 5’ to the target sequence and a 3' homology arm that hybridizes to a sequence 3' to the target sequence.
31. The gene editing system of claim 23, wherein the ncRNA has a nucleotide comprising SEQ ID NO: 19113 (RTX3_2042), SEQ ID NO: 19115 (RTX_6083v1), SEQ ID NO: 19117 (RTX_6943), or an amino acid sequence having at least 90%, 95%, 99%, or 100% sequence identity with SEQ ID Nos: 19113, 19115, or 19117.
32. The gene editing system of claim 1, wherein the ncRNA and the retron reverse transcriptase are derived from the same organism.
33. The gene editing system of claim 1, wherein the ncRNA and the retron reverse transcriptase are derived from different organisms.
34. An isolated cell comprising the gene editing system of claim 1.
35. The isolated cell of claim 28, wherein the isolated cell is a mammalian cell.
36. The isolated cell of claim 29, wherein the mammalian cell is a human cell.
37. A composition comprising: a) the gene editing system of claim 1; and b) a pharmaceutically or veterinarily acceptable carrier.
38. The composition of claim 31, wherein the delivery vehicle is a lipid nanoparticle comprising: a) one or more ionizable lipids; b) one or more structural lipids; c) one or more PEGylated lipids; and d) one or more phospholipids.
39. The composition of claim 32, wherein the one or more ionizable lipids comprises an ionizable lipid set forth in Table 2.
40. A method of genetically modifying a cell comprising: contacting the gene editing system of claim 17 with the cell, thereby delivering the RNA cargo to the cell, wherein: the nucleic acid programmable nuclease forms a complex with the guide RNA, wherein said guide RNA directs the complex to the target sequence, the nucleic acid programmable nuclease creates a double-stranded break in in the target sequence, the retron reverse transcriptase and engineered retron ncRNA create RT DNA that comprises the donor polynucleotide, and the donor polynucleotide becomes integrated at the target sequence, whereby editing the cell is genetically modified.
41. The method of claim 34, wherein the retron reverse transcriptase and the engineered ncRNA are derived from the same organism.
42. The method of claim 34, wherein the retron reverse transcriptase and the engineered ncRNA are derived from different organism.
43. The method of claim 34, wherein the retron reverse transcriptase and the engineered ncRNA are predicted to be from a single phylogenetic clade of any of FIGs.2-27.
44. The method of claim 34, wherein the retron reverse transcriptase and the engineered ncRNA are predicted to be from different phylogenetic clades of any of FIGs.2-27.
45. An engineered retron ncRNA comprising a nucleotide sequence of SEQ ID NO: 19113 (RTX3_2042), SEQ ID NO: 19115 (RTX_6083v1), SEQ ID NO: 19117 (RTX_6943), or an amino acid sequence having at least 90%, 95%, 99%, or 100% sequence identity with SEQ ID Nos: 19113, 19115, or 19117.
46. The engineered retron ncRNA of claim 39 is further modified to comprise one or more guide RNAs.
47. The engineered retron ncRNA of claim 40 comprising a first guide RNA coupled to the 5’ or 3’ end of the ncRNA.
48. The engineered retron ncRNA of claim 41 comprising a second guide RNA coupled to other end of the ncRNA.
49. An engineered retron ncRNA comprising a nucleotide sequence of any one of the ncRNA sequences listed in Table B, or a nucleotide sequence having least 90%, 95%, 99%, or 100% sequence identify with any of the ncRNA sequence listed in Table B, wherein the engineered retron ncRNA comprises a modified msd sequence having a heterologous sequence of interest wherein said modified msd sequence is reverse transcribed to form an RT DNA in the presence of a retron reverse transcriptase.
50. The engineered retron ncRNA of claim 43 is further modified to comprise one or more guide RNAs.
51. The engineered retron ncRNA of claim 44 comprising a first guide RNA coupled to the 5’ or 3’ end of the engineered retron ncRNA.
52. The engineered retron ncRNA of claim 45 comprising a second guide RNA coupled to other end of the engineered retron ncRNA.
53. An engineered retron ncRNA comprising: A) a pre-msr sequence having a first complementary region of the retron ncRNA; B) an msr sequence including an msr stem-loop structure; C) an msd sequence including an msd stem-loop structure; D) a post-msd sequence having a second complementary region, wherein the first and second complementary regions form an a1/a2 duplex region of the retron ncRNA, and wherein the engineered ncRNA comprises a sequence listed in Table B, or a nucleotide sequence having at least 90%, 95%, 99%, or 100% sequence identify with any of the ncRNA sequence listed in Table B; and wherein the msd sequence is further modified to comprise a heterologous sequence of interest inserted within the msd stem-loop structure, wherein said modified msd sequence templates a single strand DNA product (RT-DNA) comprising the heterologous sequence of interest in the presence of a retron reverse transcriptase.
54. The engineered retron ncRNA of claim 47, wherein the heterologous sequence of interest is a donor polynucleotide comprising an intended edit to be integrated at a target sequence of a cell, wherein the donor polynucleotide is flanked by a 5' homology arm that hybridizes to a sequence 5’ to the target sequence and a 3' homology arm that hybridizes to a sequence 3' to the target sequence.
55. The engineered retron ncRNA of claim 47, wherein the msr stem-loop structure, the msd stem-loop structure, or the a1/a2 duplex comprise a modification which result in increased editing efficiency and/or reduced indels in the presence of a nucleic acid programmable nuclease that associates with the one or more guide RNAs.
56. The engineered retron ncRNA of claim 49, wherein the modification comprises an MS2 hairpin coupled to either end of the a1/a2 duplex.
57. The engineered retron ncRNA of claim 49, wherein the modification comprises a 5’ cap coupled to the 5’ end of the engineered retron ncRNA.
58. The engineered retron ncRNA of claim 49, wherein the modification comprises a poly-A tail coupled to the 3’ end of the engineered retron ncRNA.
59. The engineered retron ncRNA of claim 49, wherein the modification comprises a 5’ cap coupled to the 5’ end of the engineered retron ncRNA and a poly-A tail coupled to the 3’ end of the engineered retron ncRNA.
60. The engineered retron ncRNA of claim 47, wherein the ncRNA has a conserved structure of any one of the ncRNA structures of FIGs.2-27.
61. The engineered retron ncRNA of claim 47 is further modified to comprise one or more guide RNAs.
62. The engineered retron ncRNA of claim 51 comprising a first guide RNA coupled to the 5’ or 3’ end of the engineered retron ncRNA.
63. The engineered retron ncRNA of claim 51 comprising a second guide RNA coupled to other end of the engineered retron ncRNA.
64. The engineered retron ncRNA of any of claims 51-57, wherein the engineered retron ncRNA is an in vitro transcription product of a template linearized plasmid having blunt ends.
65. A method of producing the engineered retron ncRNA of any of claims 51-57 comprising contacting a blunt-ended linearized plasmid encoding the engineered retron ncRNA with an RNA polymerase.
66. A gene editing system comprising one or more delivery vehicles, wherein: the delivery vehicle(s) comprise RNA cargo, said RNA cargo comprises (a) at least one mRNA molecule encoding (i) a nucleic acid programmable nuclease and (ii) a retron reverse transcriptase, (b) an engineered retron ncRNA of any one of claims 39-58, and (c) guide RNA for the programmable nuclease, each delivery vehicle contains (a)(i) and/or (a)(ii) and/or (b) and/or (c), whereby one delivery vehicle or more than one delivery vehicle delivers (a)(i), (a)(ii), (b), and (c).
67. An isolated cell comprising the gene editing system of claim 60.
68. The isolated cell of claim 61, wherein the isolated cell is a mammalian cell.
69. The isolated cell of claim 62, wherein the mammalian cell is a human cell.
70. A composition comprising: a) the gene editing system of claim 60; and b) a pharmaceutically or veterinarily acceptable carrier.
71. The composition of claim 64, wherein the delivery vehicle is a lipid nanoparticle comprising: a) one or more ionizable lipids; b) one or more structural lipids; c) one or more PEGylated lipids; and d) one or more phospholipids.
72. The composition of claim 65, wherein the one or more ionizable lipids comprises an ionizable lipid set forth in Table 2.
73. A method of genetically modifying a cell comprising: contacting the gene editing system of claim 60 with the cell, thereby delivering the RNA cargo to the cell, wherein: the nucleic acid programmable nuclease forms a complex with the guide RNA, wherein said guide RNA directs the complex to the target sequence, the nucleic acid programmable nuclease creates a double-stranded break in in the target sequence, the retron reverse transcriptase and engineered retron ncRNA create RT DNA that comprises the donor polynucleotide, and the donor polynucleotide becomes integrated at the target sequence, whereby editing the cell is genetically modified.
PCT/US2023/061038 2022-01-21 2023-01-20 Engineered retrons and methods of use WO2023141602A2 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
PCT/US2023/072872 WO2024044723A1 (en) 2022-08-25 2023-08-24 Engineered retrons and methods of use

Applications Claiming Priority (8)

Application Number Priority Date Filing Date Title
US202263301936P 2022-01-21 2022-01-21
US63/301,936 2022-01-21
US202263370880P 2022-08-09 2022-08-09
US63/370,880 2022-08-09
US202263373545P 2022-08-25 2022-08-25
US63/373,545 2022-08-25
US202263476900P 2022-12-22 2022-12-22
US63/476,900 2022-12-22

Publications (2)

Publication Number Publication Date
WO2023141602A2 true WO2023141602A2 (en) 2023-07-27
WO2023141602A3 WO2023141602A3 (en) 2023-11-02

Family

ID=85283659

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2023/061038 WO2023141602A2 (en) 2022-01-21 2023-01-20 Engineered retrons and methods of use

Country Status (1)

Country Link
WO (1) WO2023141602A2 (en)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2024044673A1 (en) * 2022-08-24 2024-02-29 The J. David Gladstone Institutes, A Testamentary Trust Established Under The Will Of J. David Gladstone Dual cut retron editors for genomic insertions and deletions
WO2024044723A1 (en) * 2022-08-25 2024-02-29 Renagade Therapeutics Management Inc. Engineered retrons and methods of use

Citations (221)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4751180A (en) 1985-03-28 1988-06-14 Chiron Corporation Expression using fused genes providing for protein product
WO1989003429A1 (en) 1987-08-28 1989-04-20 Health Research Inc. Recombinant avipox virus
US4935233A (en) 1985-12-02 1990-06-19 G. D. Searle And Company Covalently linked polypeptide cell modulators
WO1991012882A1 (en) 1990-02-22 1991-09-05 Medgenix Group S.A. Microspheres for the controlled release of water-soluble substances and process for preparing them
WO1992001070A1 (en) 1990-07-09 1992-01-23 The United States Of America, As Represented By The Secretary, U.S. Department Of Commerce High efficiency packaging of mutant adeno-associated virus using amber suppressions
WO1992003545A1 (en) 1990-08-15 1992-03-05 Virogenetics Corporation Flavivirus recombinant poxvirus vaccine
US5122458A (en) 1984-08-24 1992-06-16 The Upjohn Company Use of a bgh gdna polyadenylation signal in expression of non-bgh polypeptides in higher eukaryotic cells
US5135855A (en) 1986-09-03 1992-08-04 The United States Of America As Represented By The Department Of Health And Human Services Rapid, versatile and simple system for expressing genes in eukaryotic cells
US5139941A (en) 1985-10-31 1992-08-18 University Of Florida Research Foundation, Inc. AAV transduction vectors
US5168062A (en) 1985-01-30 1992-12-01 University Of Iowa Research Foundation Transfer vectors and microorganisms containing human cytomegalovirus immediate-early promoter-regulatory DNA sequence
US5173414A (en) 1990-10-30 1992-12-22 Applied Immune Sciences, Inc. Production of recombinant adeno-associated virus vectors
CA2075515A1 (en) 1991-08-30 1993-03-01 Shohei Miyata Method for synthesizing stable single-stranded cdna in eucaryotes by means of a bacterial retron, products and uses therefor
WO1993003769A1 (en) 1991-08-20 1993-03-04 THE UNITED STATES OF AMERICA, represented by THE SECRETARY, DEPARTEMENT OF HEALTH AND HUMAN SERVICES Adenovirus mediated transfer of genes to the gastrointestinal tract
US5219740A (en) 1987-02-13 1993-06-15 Fred Hutchinson Cancer Research Center Retroviral gene transfer into diploid fibroblasts for gene therapy
US5320958A (en) 1989-02-24 1994-06-14 University Of Medicine And Dentistry Of New Jersey Isolated bacterial reverse transcriptase
WO1994026911A1 (en) 1993-05-14 1994-11-24 Ohio University Edison Animal Biotechnology Institute A gene expression system utilizing an rna polymerase prebound to dna
WO1995007995A2 (en) 1993-09-13 1995-03-23 Applied Immune Sciences, Inc. Adeno-associated viral (aav) liposomes and methods related thereto
WO1995009233A1 (en) 1993-09-27 1995-04-06 The Johns-Hopkins University Functional domains in flavobacterium okeanokoites (foki) restriction endonuclease
US5405775A (en) 1989-02-24 1995-04-11 The University Of Medicine And Dentistry Of New Jersey Retrons coding for hybrid DNA/RNA molecules
WO1996017072A2 (en) 1994-11-30 1996-06-06 Chiron Viagene, Inc. Recombinant alphavirus vectors
WO1996039154A1 (en) 1995-06-06 1996-12-12 Isis Pharmaceuticals, Inc. Oligonucleotides having phosphorothioate linkages of high chiral purity
WO1997003211A1 (en) 1995-07-13 1997-01-30 Isis Pharmaceuticals, Inc. Antisense inhibition of hepatitis b virus replication
US5676950A (en) 1994-10-28 1997-10-14 University Of Florida Enterically administered recombinant poxvirus vaccines
US5780269A (en) 1989-02-24 1998-07-14 The University Of Medicine And Denistry Of New Jersey Hybrid molecules
US5789245A (en) 1993-09-15 1998-08-04 Chiron Corporation Alphavirus structural protein expression cassettes
US5844107A (en) 1994-03-23 1998-12-01 Case Western Reserve University Compacted nucleic acids and their delivery to cells
US5849563A (en) 1989-02-24 1998-12-15 The University Of Medecine And Dentistry Of New Jersey Eukaryotes expressing single stranded hybrid molecules
US5877302A (en) 1994-03-23 1999-03-02 Case Western Reserve University Compacted nucleic acids and their delivery to cells
WO1999039741A2 (en) 1998-02-03 1999-08-12 Inex Pharmaceuticals Corporation Systemic delivery of serum stable plasmid lipid particles for cancer therapy
US5965542A (en) 1997-03-18 1999-10-12 Inex Pharmaceuticals Corp. Use of temperature to control the size of cationic liposome/plasmid DNA complexes
US5972901A (en) 1994-03-23 1999-10-26 Case Western Reserve University Serpin enzyme complex receptor--mediated gene transfer
US6017737A (en) 1989-02-24 2000-01-25 The University Of Medicine And Denistry Of New Jersey E. coli msDNA synthesizing system, products and uses
WO2000023464A2 (en) 1998-10-16 2000-04-27 Novartis Ag Zinc finger binding domains for gnn
US6077835A (en) 1994-03-23 2000-06-20 Case Western Reserve University Delivery of compacted nucleic acid to cells
WO2000042219A1 (en) 1999-01-12 2000-07-20 Sangamo Biosciences, Inc. Selection of sites for targeting by zinc finger proteins and methods of designing zinc finger proteins to bind to preselected sites
WO2000041566A1 (en) 1999-01-12 2000-07-20 Sanagamo Biosciences, Inc. Regulation of endogenous gene expression in cells using zinc finger proteins
WO2000061772A2 (en) 1999-04-14 2000-10-19 Chiron Corporation Compositions and methods for generating an immune response utilizing alphavirus-based vector systems
WO2001007548A1 (en) 1999-07-26 2001-02-01 The Procter & Gamble Company Cationic charge boosting systems
WO2001038547A2 (en) 1999-11-24 2001-05-31 Mcs Micro Carrier Systems Gmbh Polypeptides comprising multimers of nuclear localization signals or of protein transduction domains and their use for transferring molecules into cells
WO2001081609A2 (en) 2000-03-22 2001-11-01 Chiron Corporation Compositions and methods for generating an immune response utilizing alphavirus-based vector systems
WO2001083793A2 (en) 2000-04-28 2001-11-08 Sangamo Biosciences, Inc. Targeted modification of chromatin structure
WO2002026960A2 (en) 2000-09-29 2002-04-04 Sangamo Biosciences, Inc. Modulation of gene expression using localization domains
US20020081614A1 (en) 1999-09-14 2002-06-27 Sangamo Biosciences, Inc. Functional genomics using zinc finger proteins
US20020150626A1 (en) 2000-10-16 2002-10-17 Kohane Daniel S. Lipid-protein-sugar particles for delivery of nucleic acids
WO2002080982A2 (en) 2001-01-12 2002-10-17 Chiron Corporation Nucleic acid mucosal immunization
WO2002099035A2 (en) 2001-05-31 2002-12-12 Chiron Corporation Chimeric alphavirus replicon particles
US6503717B2 (en) 1999-12-06 2003-01-07 Sangamo Biosciences, Inc. Methods of using randomized libraries of zinc finger proteins for the identification of gene function
US20030032615A1 (en) 1989-03-21 2003-02-13 Vical Incorporated Lipid-mediated polynucleotide administration to deliver a biologically active peptide and to induce a cellular immune response
US20030108880A1 (en) 2001-01-22 2003-06-12 Sangamo Biosciences Modified zinc finger binding proteins
WO2003080809A2 (en) 2002-03-21 2003-10-02 Sangamo Biosciences, Inc. Methods and compositions for using zinc finger endonucleases to enhance homologous recombination
US20030203865A1 (en) 2001-04-30 2003-10-30 Pierrot Harvie Lipid-comprising drug delivery complexes and methods for their production
US20040002092A1 (en) 2002-03-15 2004-01-01 Sylvain Arnould Hybrid and single chain meganucleases and use thereof
US6689558B2 (en) 2000-02-08 2004-02-10 Sangamo Biosciences, Inc. Cells for drug discovery
US20040048787A1 (en) 2000-05-31 2004-03-11 Copernicus Therapeutics, Inc. Lyophilizable and enhanced compacted nucleic acids
US20040091991A1 (en) 2002-08-29 2004-05-13 Sangamo Biosciences, Inc. Simultaneous modulation of multiple genes
US20040142025A1 (en) 2002-06-28 2004-07-22 Protiva Biotherapeutics Ltd. Liposomal apparatus and manufacturing methods
WO2004067736A2 (en) 2003-01-28 2004-08-12 Cellectis Custom-made meganuclease and use thereof
US6780590B2 (en) 1999-09-14 2004-08-24 Sangamo Biosciences, Inc. Gene identification
US6794136B1 (en) 2000-11-20 2004-09-21 Sangamo Biosciences, Inc. Iterative optimization in the design of binding proteins
US20040235002A1 (en) 2002-09-20 2004-11-25 Sangamo Biosciences, Inc. Multiplex screening assays
US20050017054A1 (en) 2003-07-23 2005-01-27 Tom Iverson Flyback transformer wire attach method to printed circuit board
US20050064595A1 (en) 2003-07-16 2005-03-24 Protiva Biotherapeutics, Inc. Lipid encapsulated interfering RNA
US20050118253A1 (en) 1998-02-03 2005-06-02 Protiva Biotherapeutics, Inc. Systemic delivery of serum stable plasmid lipid particles for cancer therapy
US6919204B2 (en) 2000-09-29 2005-07-19 Sangamo Biosciences, Inc. Modulation of gene expression using localization domains
US20050175682A1 (en) 2003-09-15 2005-08-11 Protiva Biotherapeutics, Inc. Polyethyleneglycol-modified lipid compounds and uses thereof
WO2005105989A1 (en) 2004-04-30 2005-11-10 Cellectis I-dmoi derivatives with enhanced activity at 37°c and use thereof.
US20050267061A1 (en) 2004-04-08 2005-12-01 Sangamo Biosciences, Inc. Methods and compositions for treating neuropathic and neurodegenerative conditions
US20060008910A1 (en) 2004-06-07 2006-01-12 Protiva Biotherapeuties, Inc. Lipid encapsulated interfering RNA
US7013219B2 (en) 1999-01-12 2006-03-14 Sangamo Biosciences, Inc. Regulation of endogenous gene expression in cells using zinc finger proteins
US20060063231A1 (en) 2004-09-16 2006-03-23 Sangamo Biosciences, Inc. Compositions and methods for protein production
US7030215B2 (en) 1999-03-24 2006-04-18 Sangamo Biosciences, Inc. Position dependent recognition of GNN nucleotide triplets by zinc fingers
US20060083780A1 (en) 2004-06-07 2006-04-20 Protiva Biotherapeutics, Inc. Cationic lipids and methods of use
US7070934B2 (en) 1999-01-12 2006-07-04 Sangamo Biosciences, Inc. Ligand-controlled regulation of endogenous gene expression
WO2006097853A1 (en) 2005-03-15 2006-09-21 Cellectis I-crei meganuclease variants with modified specificity, method of preparation and uses thereof
US20070042031A1 (en) 2005-07-27 2007-02-22 Protiva Biotherapeutics, Inc. Systems and methods for manufacturing liposomes
WO2007049095A1 (en) 2005-10-25 2007-05-03 Cellectis Laglidadg homing endonuclease variants having mutations in two functional subdomains and use thereof
US20070134796A1 (en) 2005-07-26 2007-06-14 Sangamo Biosciences, Inc. Targeted integration and expression of exogenous nucleic acid sequences
US7253273B2 (en) 2004-04-08 2007-08-07 Sangamo Biosciences, Inc. Treatment of neuropathic pain with zinc finger proteins
US7262054B2 (en) 2002-01-22 2007-08-28 Sangamo Biosciences, Inc. Zinc finger proteins for DNA binding and gene regulation in plants
WO2007123636A2 (en) 2006-03-27 2007-11-01 Children's Hospital & Regional Medical Center Compositions and methods comprising the use of cell surface displayed homing endonucleases
US20080031109A1 (en) 2005-07-15 2008-02-07 Yoshikazu Takashima Method and apparatus for processing information, method and apparatus for manufacturing information recording medium, information recording medium, and computer program
US7358085B2 (en) 2005-02-28 2008-04-15 Sangamo Biosciences, Inc. Anti-angiogenic methods and compositions
WO2008102198A1 (en) 2007-02-19 2008-08-28 Cellectis Laglidadg homing endonuclease variants having novel substrate specificity and use thereof
US7476500B1 (en) 2001-03-19 2009-01-13 President And Fellows Of Harvard College In vivo selection system for enzyme activity
US7541517B2 (en) 2003-12-22 2009-06-02 Pioneer Hi-Bred International, Inc. Bacillus thuringiensis CRY9 nucleic acids
WO2009068937A1 (en) 2007-11-28 2009-06-04 Cellectis I-msoi homing endonuclease variants having novel substrate specificity and use thereof
US20090222937A1 (en) 2006-02-13 2009-09-03 Cellectis Meganuclease variants cleaving a dna target sequence from a xeroderma pigmentosum gene and uses thereof
US20090305977A1 (en) 2006-02-09 2009-12-10 Sangamo Bioscience, Inc. Method for treating peripheral arterial disease with zinc finger proteins
WO2010001189A1 (en) 2008-07-03 2010-01-07 Cellectis The crystal structure of i-dmoi in complex with its dna target, improved chimeric meganucleases and uses thereof
US7692068B2 (en) 2003-10-14 2010-04-06 Athenix Corporation AXMI-010, a delta-endotoxin gene and methods for its use
WO2010046786A1 (en) 2008-10-23 2010-04-29 Cellectis Meganuclease recombination system
US20100130588A1 (en) 2008-04-15 2010-05-27 Protiva Biotherapeutics, Inc. Novel lipid formulations for nucleic acid delivery
WO2010065123A1 (en) 2008-12-04 2010-06-10 Sangamo Biosciences, Inc. Genome editing in rats using zinc-finger nucleases
US20100146651A1 (en) 2006-11-14 2010-06-10 Cellectis Meganuclease variants cleaving a dna target sequence from the hprt gene and uses thereof
US20100151556A1 (en) 2002-03-15 2010-06-17 Cellectis Hybrid and single chain meganucleases and use thereof
US20100167357A1 (en) 2007-02-01 2010-07-01 Cellectis Obligate heterodimer meganucleases and uses thereof
US20100203031A1 (en) 2007-06-06 2010-08-12 Cellectis Method for enhancing the cleavage activity of i-crei derived meganucleases
US20100229252A1 (en) 2007-07-23 2010-09-09 Cellectis Meganuclease variants cleaving a dna target sequence from the human hemoglobin beta gene and uses thereof
US20100311124A1 (en) 2008-10-29 2010-12-09 Sangamo Biosciences, Inc. Methods and compositions for inactivating glutamine synthetase gene expression
US20100325745A1 (en) 2007-06-06 2010-12-23 Cellectis Meganuclease variants cleaving a dna target sequence from the mouse rosa26 locus and uses thereof
US20110076335A1 (en) 2009-07-01 2011-03-31 Protiva Biotherapeutics, Inc. Novel lipid formulations for delivery of therapeutic agents to solid tumors
US7919605B1 (en) 2010-08-30 2011-04-05 Amyris, Inc. Nucleic acids, compositions and methods for the excision of target nucleic acids
US20110091441A1 (en) 2007-08-03 2011-04-21 Cellectis Meganuclease variants cleaving a dna target sequence from the human interleukin-2 receptor gamma chain gene and uses thereof
US20110117125A1 (en) 2008-01-02 2011-05-19 Tekmira Pharmaceuticals Corporation Compositions and methods for the delivery of nucleic acids
US7947873B2 (en) 2002-04-17 2011-05-24 Sangamo Biosciences, Inc. Compositions and methods for regulation of plant gamma-tocopherol methyltransferase
US7951925B2 (en) 2006-05-25 2011-05-31 Sangamo Biosciences, Inc. Methods and compositions for gene inactivation
US20110225664A1 (en) 2008-09-08 2011-09-15 Cellectis Meganuclease variants cleaving a dna target sequence from a glutamine synthetase gene and uses thereof
US20110265198A1 (en) 2010-04-26 2011-10-27 Sangamo Biosciences, Inc. Genome editing of a Rosa locus using nucleases
WO2011141705A1 (en) 2010-05-12 2011-11-17 Protiva Biotherapeutics, Inc. Novel cationic lipids and methods of use thereof
US20110287512A1 (en) 2010-05-03 2011-11-24 Sangamo Biosciences, Inc. Compositions for linking zinc finger modules
US20110311583A1 (en) 2008-11-10 2011-12-22 Alnylam Pharmaceuticals, Inc. Novel lipids and compositions for the delivery of therapeutics
US20120027803A1 (en) 2010-06-03 2012-02-02 Alnylam Pharmaceuticals, Inc. Biodegradable lipids for the delivery of active agents
US8119361B2 (en) 2005-10-18 2012-02-21 Duke University Methods of cleaving DNA with rationally-designed meganucleases
WO2012067428A2 (en) 2010-11-16 2012-05-24 주식회사 에피밸리 Group-iii nitride semiconductor light-emitting device
US20120172411A1 (en) 2010-09-17 2012-07-05 Protiva Biotherapeutics, Inc. Novel trialkyl cationic lipids and methods of use thereof
US20120171191A1 (en) 2009-05-26 2012-07-05 Cellectis Meganuclease variants cleaving the genome of a pathogenic non-integrating virus and uses thereof
US20120204282A1 (en) 2011-02-04 2012-08-09 Sangamo Biosciences, Inc. Methods and compositions for treating occular disorders
US20120252122A1 (en) 2011-01-05 2012-10-04 Sangamo Biosciences, Inc. Methods and compositions for increasing production of induced pluripotent stem cells (ipscs)
US20120260356A1 (en) 2009-04-21 2012-10-11 Cellectis Meganuclease variants cleaving at least one target in the genome of a retrovirus and uses thereof
US20120272348A1 (en) 2009-07-17 2012-10-25 Cellectis Viral vectors encoding a dna repair matrix and containing a virion-associated site specific meganuclease for gene targeting
US20120295832A1 (en) 2011-05-17 2012-11-22 Arrowhead Research Corporation Novel Lipids and Compositions for Intracellular Delivery of Biologically Active Compounds
US20120301456A1 (en) 2009-09-24 2012-11-29 Cellectis Meganuclease reagents of uses thereof for treating genetic diseases caused by frame shift/non sense mutations
US20130017223A1 (en) 2009-12-18 2013-01-17 The University Of British Columbia Methods and compositions for delivery of nucleic acids
US20130022649A1 (en) 2009-12-01 2013-01-24 Protiva Biotherapeutics, Inc. Snalp formulations containing antioxidants
WO2013016058A1 (en) 2011-07-22 2013-01-31 Merck Sharp & Dohme Corp. Novel bis-nitrogen containing cationic lipids for oligonucleotide delivery
US20130045539A1 (en) 2010-02-18 2013-02-21 Cellectis Meganuclease recombination system
US8383766B2 (en) 1999-03-24 2013-02-26 Sangamo Biosciences, Inc. Position dependent recognition of GNN nucleotide triplets by zinc fingers
US20130053572A1 (en) 2010-01-22 2013-02-28 Steven L. Colletti Novel Cationic Lipids for Oligonucleotide Delivery
WO2013044008A2 (en) 2011-09-21 2013-03-28 Sangamo Biosciences, Inc. Methods and compositions for regulation of transgene expression
US20130086373A1 (en) 2011-09-29 2013-04-04 Apple Inc. Customized content for electronic devices
US20130108685A1 (en) 2010-04-28 2013-05-02 Takeshi Kuboyama Cationic lipid
US20130145487A1 (en) 2010-05-12 2013-06-06 Cellectis Meganuclease variants cleaving a dna target sequence from the dystrophin gene and uses thereof
WO2013086373A1 (en) 2011-12-07 2013-06-13 Alnylam Pharmaceuticals, Inc. Lipids for the delivery of active agents
WO2013086322A1 (en) 2011-12-07 2013-06-13 Alnylam Pharmaceuticals, Inc. Branched alkyl and cycloalkyl terminated biodegradable lipids for the delivery of active agents
US20130183282A1 (en) 2010-05-12 2013-07-18 Cellectis Meganuclease variants cleaving a DNA target sequence from the rhodopsin gene and uses thereof
US20130189759A1 (en) 2010-07-07 2013-07-25 Cellectis Meganucleases variants cleaving a dna target sequence in the nanog gene and uses thereof
US20130196320A1 (en) 2010-06-15 2013-08-01 Cellectis Method for improving cleavage of dna by endonuclease sensitive to methylation
US20130195920A1 (en) 2011-12-07 2013-08-01 Alnylam Pharmaceuticals, Inc. Biodegradable lipids for the delivery of active agents
US20130245107A1 (en) 2011-12-16 2013-09-19 modeRNA Therapeutics Dlin-mc3-dma lipid nanoparticle delivery of modified polynucleotides
US8569256B2 (en) 2009-07-01 2013-10-29 Protiva Biotherapeutics, Inc. Cationic lipids and methods for the delivery of therapeutic agents
US8575425B2 (en) 2009-07-02 2013-11-05 Athenix Corporation AXMI-205 pesticidal gene and methods for its use
US20130323269A1 (en) 2010-07-30 2013-12-05 Muthiah Manoharan Methods and compositions for delivery of active agents
US20130338210A1 (en) 2009-12-07 2013-12-19 Alnylam Pharmaceuticals, Inc. Compositions for nucleic acid delivery
WO2014008334A1 (en) 2012-07-06 2014-01-09 Alnylam Pharmaceuticals, Inc. Stable non-aggregating nucleic acid lipid particle formulations
US20140017214A1 (en) 2012-07-11 2014-01-16 Sangamo Biosciences, Inc. Methods and compositions for delivery of biologics
US20140068797A1 (en) 2012-05-25 2014-03-06 University Of Vienna Methods and compositions for rna-directed target dna modification and for rna-directed modulation of transcription
US8735153B2 (en) 2001-09-24 2014-05-27 Sangamo Biosciences, Inc. Modulation of stem cells using zinc finger proteins
US20140170753A1 (en) 2012-12-12 2014-06-19 Massachusetts Institute Of Technology Crispr-cas systems and methods for altering expression of gene products
US20140179770A1 (en) 2012-12-12 2014-06-26 Massachusetts Institute Of Technology Delivery, engineering and optimization of systems, methods and compositions for sequence manipulation and therapeutic applications
US20140179006A1 (en) 2012-12-12 2014-06-26 Massachusetts Institute Of Technology Crispr-cas component systems, methods and compositions for sequence manipulation
US20140186958A1 (en) 2012-12-12 2014-07-03 Feng Zhang Engineering and optimization of systems, methods and compositions for sequence manipulation with functional domains
US20140186919A1 (en) 2012-12-12 2014-07-03 Feng Zhang Engineering and optimization of improved systems, methods and enzyme compositions for sequence manipulation
US20140186843A1 (en) 2012-12-12 2014-07-03 Massachusetts Institute Of Technology Methods, systems, and apparatus for identifying target sequences for cas enzymes or crispr-cas systems for target sequences and conveying results thereof
US8772008B2 (en) 2009-05-18 2014-07-08 Sangamo Biosciences, Inc. Methods and compositions for increasing nuclease activity
US20140200257A1 (en) 2011-01-11 2014-07-17 Alnylam Pharmaceuticals, Inc. Pegylated lipids and their use for drug delivery
US8802934B2 (en) 2010-08-19 2014-08-12 Pioneer Hi Bred International Inc Bacillus thuringiensis gene with lepidopteran activity
US20140234972A1 (en) 2012-12-12 2014-08-21 Massachusetts Institute Of Technology CRISPR-CAS Nickase Systems, Methods And Compositions For Sequence Manipulation in Eukaryotes
US20140242702A1 (en) 2013-02-25 2014-08-28 Sigma Aldrich Co. Llc Methods and compositions for enhancing nuclease-mediated gene disruption
US20140242664A1 (en) 2012-12-12 2014-08-28 The Broad Institute, Inc. Engineering of systems, methods and optimized guide compositions for sequence manipulation
US20140273233A1 (en) 2013-03-15 2014-09-18 Sigma-Aldrich Co., Llc Crispr-based genome modification and regulation
US20140273037A1 (en) 2013-03-15 2014-09-18 System Biosciences, Llc Compositions and methods directed to crispr/cas genomic engineering systems
US20140273235A1 (en) 2013-03-15 2014-09-18 Regents Of The University Of Minnesota ENGINEERING PLANT GENOMES USING CRISPR/Cas SYSTEMS
US20140287938A1 (en) 2013-03-15 2014-09-25 The Broad Institute, Inc. Recombinant virus and preparations thereof
US8846578B2 (en) 2008-04-16 2014-09-30 The General Hospital Corporation Zinc finger nuclease for the CFTR gene and methods of use thereof
US20140298547A1 (en) 2012-12-13 2014-10-02 Dow Agrosciences Llc Dna detection methods for site specific nuclease activity
US20140295556A1 (en) 2013-03-15 2014-10-02 The General Hospital Corporation Using RNA-guided FokI Nucleases (RFNs) to Increase Specificity for RNA-Guided Genome Editing
US20140304853A1 (en) 2013-04-05 2014-10-09 Sangamo Biosciences, Inc. Methods and compositions for integration of an exogenous sequence within the genome of plants
US20140309487A1 (en) 2013-04-16 2014-10-16 Regeneron Pharmaceuticals, Inc. Targeted modification of rat genome
US20140315985A1 (en) 2013-03-14 2014-10-23 Caribou Biosciences, Inc. Compositions and methods of nucleic acid-targeting nucleic acids
US20140335063A1 (en) 2013-05-10 2014-11-13 Sangamo Biosciences, Inc. Delivery methods and compositions for nuclease-mediated genome engineering
US20140342456A1 (en) 2012-12-17 2014-11-20 President And Fellows Of Harvard College RNA-Guided Human Genome Engineering
US20140349400A1 (en) 2013-03-15 2014-11-27 Massachusetts Institute Of Technology Programmable Modification of DNA
US20140349405A1 (en) 2013-05-22 2014-11-27 Wisconsin Alumni Research Foundation Rna-directed dna cleavage and gene editing by cas9 enzyme from neisseria meningitidis
US20140357530A1 (en) 2012-12-12 2014-12-04 The Broad Institute Inc. Functional genomics using crispr-cas systems, compositions, methods, knock out libraries and applications thereof
US20140356956A1 (en) 2013-06-04 2014-12-04 President And Fellows Of Harvard College RNA-Guided Transcriptional Regulation
US20140357523A1 (en) 2013-05-29 2014-12-04 Agilent Technologies, Inc. Method for fragmenting genomic dna using cas9
US20140364333A1 (en) 2013-03-15 2014-12-11 President And Fellows Of Harvard College Methods for Live Imaging of Cells
US20140377868A1 (en) 2013-03-15 2014-12-25 The General Hospital Corporation Rna-guided targeting of genetic and epigenomic regulatory proteins to specific genomic loci
US20150064789A1 (en) 2013-08-28 2015-03-05 Sangamo Biosciences, Inc. Compositions for linking dna-binding domains and cleavage domains
US20150203446A1 (en) 2011-09-27 2015-07-23 Takeda Pharmaceutical Company Limited Di-aliphatic substituted pegylated lipids
US9115409B2 (en) 2007-09-27 2015-08-25 Sangamo Biosciences, Inc. Rapid screening of biologically active nucleases and isolation of nuclease-modified cells
US20150267205A1 (en) 2014-03-18 2015-09-24 Sangamo Biosciences, Inc. Methods and compositions for regulation of zinc finger protein expression
US9145565B2 (en) 2002-01-23 2015-09-29 University Of Utah Research Foundation Targeted chromosomal mutagenesis using zinc finger nucleases
WO2015199952A1 (en) 2014-06-25 2015-12-30 Acuitas Therapeutics Inc. Novel lipids and lipid nanoparticle formulations for delivery of nucleic acids
WO2016014794A1 (en) 2014-07-25 2016-01-28 Sangamo Biosciences, Inc. Methods and compositions for modulating nuclease-mediated genome engineering in hematopoietic stem cells
US9290748B2 (en) 2010-02-26 2016-03-22 Cellectis Use of endonucleases for inserting transgenes into safe harbor loci
WO2016106236A1 (en) 2014-12-23 2016-06-30 The Broad Institute Inc. Rna-targeting system
US20160215298A1 (en) 2006-05-25 2016-07-28 Sangamo Biosciences, Inc. Methods and compositions for increased transgene expression
US9428756B2 (en) 2006-08-11 2016-08-30 Dow Agrosciences Llc Zinc finger nuclease-mediated homologous recombination
WO2016183298A2 (en) 2015-05-12 2016-11-17 Sangamo Biosciences, Inc. Nuclease-mediated regulation of gene expression
WO2016189532A1 (en) 2015-05-26 2016-12-01 Ramot At Tel-Aviv University Ltd. Targeted lipid particles for systemic delivery of nucleic acid molecules to leukocytes
WO2017004143A1 (en) 2015-06-29 2017-01-05 Acuitas Therapeutics Inc. Lipids and lipid nanoparticle formulations for delivery of nucleic acids
US9540623B2 (en) 2011-07-08 2017-01-10 Cellectis Method for increasing the efficiency of double-strand-break induced mutagenesis
WO2017070633A2 (en) 2015-10-23 2017-04-27 President And Fellows Of Harvard College Evolved cas9 proteins for gene editing
US20170119904A1 (en) 2015-10-28 2017-05-04 Acuitas Therapeutics, Inc. Novel lipids and lipid nanoparticle formulations for delivery of nucleic acids
WO2017091512A1 (en) 2015-11-23 2017-06-01 Sangamo Biosciences, Inc. Methods and compositions for engineering immunity
US9683257B2 (en) 2008-07-14 2017-06-20 Precision Biosciences, Inc. Recognition sequences for I-CreI-derived meganucleases and uses thereof
WO2017117528A1 (en) 2015-12-30 2017-07-06 Acuitas Therapeutics, Inc. Lipids and lipid nanoparticle formulations for delivery of nucleic acids
US20170210697A1 (en) 2015-09-17 2017-07-27 Modernatx, Inc. Compounds and compositions for intracellular delivery of therapeutic agents
US20170218349A1 (en) 2016-02-02 2017-08-03 Sangamo Biosciences, Inc. Compositions for linking dna-binding domains and cleavage domains
WO2018071565A1 (en) 2016-10-11 2018-04-19 Bluebird Bio, Inc. TCRa HOMING ENDONUCLEASE VARIANTS
US10000746B2 (en) 2013-05-31 2018-06-19 Cellectis LAGLIDADG homing endonuclease cleaving the T cell receptor alpha gene and uses thereof
US10006052B2 (en) 2013-05-31 2018-06-26 Cellectis Laglidadg homing endonuclease cleaving the C-C chemokine receptor type-5 (CCR5) gene and uses thereof
WO2019089828A1 (en) 2017-10-31 2019-05-09 Acuitas Therapeutics, Inc. Lamellar lipid nanoparticles
WO2019126558A1 (en) 2017-12-20 2019-06-27 Bluebird Bio, Inc. Ahr homing endonuclease variants, compositions, and methods of use
WO2019135816A2 (en) 2017-10-23 2019-07-11 The Broad Institute, Inc. Novel nucleic acid modifiers
WO2019152557A1 (en) 2018-01-30 2019-08-08 Modernatx, Inc. Compositions and methods for delivery of agents to immune cells
US20190240354A1 (en) 2016-06-30 2019-08-08 Arbutus Biopharma Corporation Compositions and methods for delivering messenger rna
WO2019232095A1 (en) 2018-05-30 2019-12-05 Translate Bio, Inc. Vitamin cationic lipids
US20200010835A1 (en) 2016-08-24 2020-01-09 President And Fellows Of Harvard College Incorporation of unnatural amino acids into proteins using base editing
WO2020191233A1 (en) 2019-03-19 2020-09-24 The Broad Institute, Inc. Methods and compositions for editing nucleotide sequences
US10858639B2 (en) 2013-09-06 2020-12-08 President And Fellows Of Harvard College CAS9 variants and uses thereof
US10947530B2 (en) 2016-08-03 2021-03-16 President And Fellows Of Harvard College Adenosine nucleobase editors and uses thereof
US20210087135A1 (en) 2019-09-19 2021-03-25 Modernatx, Inc. Branched tail lipid compounds and compositions for intracellular delivery of therapeutic agents
WO2021077067A1 (en) 2019-10-18 2021-04-22 The Trustees Of The University Of Pennsylvania Lipid nanoparticles and formulations thereof for car mrna delivery
US20210121411A1 (en) 2018-06-19 2021-04-29 The Board Of Regents Of The University Of Texas System Lipid nanoparticle compositions for delivery of mrna and long nucleic acids
US20210128488A1 (en) 2017-08-16 2021-05-06 Acuitas Therapeutics, Inc. Lipids for use in lipid nanoparticle formulations
US11071790B2 (en) 2014-10-29 2021-07-27 Massachusetts Eye And Ear Infirmary Method for efficient delivery of therapeutic molecules in vitro and in vivo
WO2021204179A1 (en) 2020-04-09 2021-10-14 Suzhou Abogen Biosciences Co., Ltd. Nucleic acid vaccines for coronavirus
US20220145296A1 (en) 2018-12-27 2022-05-12 LifeEDIT Therapeutics, Inc. Polypeptides useful for gene editing and methods of use
US20220204975A1 (en) 2019-04-12 2022-06-30 President And Fellows Of Harvard College System for genome editing

Family Cites Families (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2021178709A1 (en) * 2020-03-04 2021-09-10 Flagship Pioneering Innovations Vi, Llc Methods and compositions for modulating a genome
EP4114937A2 (en) * 2020-03-04 2023-01-11 Flagship Pioneering Innovations VI, LLC Methods and compositions for modulating a genome
WO2023081756A1 (en) * 2021-11-03 2023-05-11 The J. David Gladstone Institutes, A Testamentary Trust Established Under The Will Of J. David Gladstone Precise genome editing using retrons

Patent Citations (364)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5122458A (en) 1984-08-24 1992-06-16 The Upjohn Company Use of a bgh gdna polyadenylation signal in expression of non-bgh polypeptides in higher eukaryotic cells
US5168062A (en) 1985-01-30 1992-12-01 University Of Iowa Research Foundation Transfer vectors and microorganisms containing human cytomegalovirus immediate-early promoter-regulatory DNA sequence
US5385839A (en) 1985-01-30 1995-01-31 University Of Iowa Research Foundation Transfer vectors and microorganisms containing human cytomegalovirus immediate-early promoter regulatory DNA sequence
US4751180A (en) 1985-03-28 1988-06-14 Chiron Corporation Expression using fused genes providing for protein product
US5139941A (en) 1985-10-31 1992-08-18 University Of Florida Research Foundation, Inc. AAV transduction vectors
US4935233A (en) 1985-12-02 1990-06-19 G. D. Searle And Company Covalently linked polypeptide cell modulators
US5135855A (en) 1986-09-03 1992-08-04 The United States Of America As Represented By The Department Of Health And Human Services Rapid, versatile and simple system for expressing genes in eukaryotic cells
US5219740A (en) 1987-02-13 1993-06-15 Fred Hutchinson Cancer Research Center Retroviral gene transfer into diploid fibroblasts for gene therapy
WO1989003429A1 (en) 1987-08-28 1989-04-20 Health Research Inc. Recombinant avipox virus
US5849563A (en) 1989-02-24 1998-12-15 The University Of Medecine And Dentistry Of New Jersey Eukaryotes expressing single stranded hybrid molecules
US5405775A (en) 1989-02-24 1995-04-11 The University Of Medicine And Dentistry Of New Jersey Retrons coding for hybrid DNA/RNA molecules
US5780269A (en) 1989-02-24 1998-07-14 The University Of Medicine And Denistry Of New Jersey Hybrid molecules
US5436141A (en) 1989-02-24 1995-07-25 University Of Medicine And Dentistry Of New Jersey Method for synthesizing stable single-stranded CDNA in eukaryotes by means of a bacterial retron and products
US5320958A (en) 1989-02-24 1994-06-14 University Of Medicine And Dentistry Of New Jersey Isolated bacterial reverse transcriptase
US6017737A (en) 1989-02-24 2000-01-25 The University Of Medicine And Denistry Of New Jersey E. coli msDNA synthesizing system, products and uses
US20030032615A1 (en) 1989-03-21 2003-02-13 Vical Incorporated Lipid-mediated polynucleotide administration to deliver a biologically active peptide and to induce a cellular immune response
WO1991012882A1 (en) 1990-02-22 1991-09-05 Medgenix Group S.A. Microspheres for the controlled release of water-soluble substances and process for preparing them
WO1992001070A1 (en) 1990-07-09 1992-01-23 The United States Of America, As Represented By The Secretary, U.S. Department Of Commerce High efficiency packaging of mutant adeno-associated virus using amber suppressions
WO1992003545A1 (en) 1990-08-15 1992-03-05 Virogenetics Corporation Flavivirus recombinant poxvirus vaccine
US5173414A (en) 1990-10-30 1992-12-22 Applied Immune Sciences, Inc. Production of recombinant adeno-associated virus vectors
WO1993003769A1 (en) 1991-08-20 1993-03-04 THE UNITED STATES OF AMERICA, represented by THE SECRETARY, DEPARTEMENT OF HEALTH AND HUMAN SERVICES Adenovirus mediated transfer of genes to the gastrointestinal tract
CA2075515A1 (en) 1991-08-30 1993-03-01 Shohei Miyata Method for synthesizing stable single-stranded cdna in eucaryotes by means of a bacterial retron, products and uses therefor
WO1994026911A1 (en) 1993-05-14 1994-11-24 Ohio University Edison Animal Biotechnology Institute A gene expression system utilizing an rna polymerase prebound to dna
WO1995007995A2 (en) 1993-09-13 1995-03-23 Applied Immune Sciences, Inc. Adeno-associated viral (aav) liposomes and methods related thereto
US5843723A (en) 1993-09-15 1998-12-01 Chiron Corporation Alphavirus vector constructs
US5789245A (en) 1993-09-15 1998-08-04 Chiron Corporation Alphavirus structural protein expression cassettes
WO1995009233A1 (en) 1993-09-27 1995-04-06 The Johns-Hopkins University Functional domains in flavobacterium okeanokoites (foki) restriction endonuclease
US5877302A (en) 1994-03-23 1999-03-02 Case Western Reserve University Compacted nucleic acids and their delivery to cells
US5844107A (en) 1994-03-23 1998-12-01 Case Western Reserve University Compacted nucleic acids and their delivery to cells
US6200801B1 (en) 1994-03-23 2001-03-13 Case Western Reserve University Serpin enzyme complex receptor-mediated gene transfer
US5972901A (en) 1994-03-23 1999-10-26 Case Western Reserve University Serpin enzyme complex receptor--mediated gene transfer
US5972900A (en) 1994-03-23 1999-10-26 Case Western Reserve University Delivery of nucleic acid to cells
US6008336A (en) 1994-03-23 1999-12-28 Case Western Reserve University Compacted nucleic acids and their delivery to cells
US6077835A (en) 1994-03-23 2000-06-20 Case Western Reserve University Delivery of compacted nucleic acid to cells
US5676950A (en) 1994-10-28 1997-10-14 University Of Florida Enterically administered recombinant poxvirus vaccines
WO1996017072A2 (en) 1994-11-30 1996-06-06 Chiron Viagene, Inc. Recombinant alphavirus vectors
WO1996039154A1 (en) 1995-06-06 1996-12-12 Isis Pharmaceuticals, Inc. Oligonucleotides having phosphorothioate linkages of high chiral purity
WO1997003211A1 (en) 1995-07-13 1997-01-30 Isis Pharmaceuticals, Inc. Antisense inhibition of hepatitis b virus replication
US5965542A (en) 1997-03-18 1999-10-12 Inex Pharmaceuticals Corp. Use of temperature to control the size of cationic liposome/plasmid DNA complexes
WO1999039741A2 (en) 1998-02-03 1999-08-12 Inex Pharmaceuticals Corporation Systemic delivery of serum stable plasmid lipid particles for cancer therapy
US20050118253A1 (en) 1998-02-03 2005-06-02 Protiva Biotherapeutics, Inc. Systemic delivery of serum stable plasmid lipid particles for cancer therapy
WO2000023464A2 (en) 1998-10-16 2000-04-27 Novartis Ag Zinc finger binding domains for gnn
US7788044B2 (en) 1999-01-12 2010-08-31 Sangamo Biosciences, Inc. Selection of sites for targeting by zinc finger proteins and methods of designing zinc finger proteins to bind to preselected sites
US20030105593A1 (en) 1999-01-12 2003-06-05 Sangamo Biosciences, Inc. Selection of sites for targeting by zinc finger proteins and methods of designing zinc finger proteins to bind to preselected sites
US20040204345A1 (en) 1999-01-12 2004-10-14 Sangamo Biosciences, Inc. Modulation of endogenous gene expression in cells
US9491934B2 (en) 1999-01-12 2016-11-15 Sangamo Biosciences, Inc. Regulation of endogenous gene expression in cells using zinc finger proteins
WO2000041566A1 (en) 1999-01-12 2000-07-20 Sanagamo Biosciences, Inc. Regulation of endogenous gene expression in cells using zinc finger proteins
US6785613B2 (en) 1999-01-12 2004-08-31 Sangamo Biosciences, Inc. Selection of sites for targeting by zinc finger proteins and methods of designing zinc finger proteins to bind to preselected sites
US20070287189A1 (en) 1999-01-12 2007-12-13 Sangamo Biosciences, Inc. Selection of sites for targeting by zinc finger proteins and methods of designing zinc finger proteins to bind to preselected sites
US6453242B1 (en) 1999-01-12 2002-09-17 Sangamo Biosciences, Inc. Selection of sites for targeting by zinc finger proteins and methods of designing zinc finger proteins to bind to preselected sites
US6933113B2 (en) 1999-01-12 2005-08-23 Sangamo Biosciences, Inc. Modulation of endogenous gene expression in cells
US7177766B2 (en) 1999-01-12 2007-02-13 Sangamo Biosciences, Inc. Selection of sites for targeting by zinc finger proteins and methods of designing zinc finger proteins to bind to preselected sites
US6979539B2 (en) 1999-01-12 2005-12-27 Sangamo Biosciences, Inc. Regulation of endogenous gene expression in cells using zinc finger proteins
US7070934B2 (en) 1999-01-12 2006-07-04 Sangamo Biosciences, Inc. Ligand-controlled regulation of endogenous gene expression
WO2000042219A1 (en) 1999-01-12 2000-07-20 Sangamo Biosciences, Inc. Selection of sites for targeting by zinc finger proteins and methods of designing zinc finger proteins to bind to preselected sites
US7013219B2 (en) 1999-01-12 2006-03-14 Sangamo Biosciences, Inc. Regulation of endogenous gene expression in cells using zinc finger proteins
US7030215B2 (en) 1999-03-24 2006-04-18 Sangamo Biosciences, Inc. Position dependent recognition of GNN nucleotide triplets by zinc fingers
US8524874B2 (en) 1999-03-24 2013-09-03 Sangamo Biosciences Position dependent recognition of GNN nucleotide triplets by zinc fingers
US20130123484A1 (en) 1999-03-24 2013-05-16 Sangamo Biosciences, Inc. Position dependent recognition of gnn nucleotide triplets by zinc fingers
US8383766B2 (en) 1999-03-24 2013-02-26 Sangamo Biosciences, Inc. Position dependent recognition of GNN nucleotide triplets by zinc fingers
WO2000061772A2 (en) 1999-04-14 2000-10-19 Chiron Corporation Compositions and methods for generating an immune response utilizing alphavirus-based vector systems
WO2001007548A1 (en) 1999-07-26 2001-02-01 The Procter & Gamble Company Cationic charge boosting systems
US6777185B2 (en) 1999-09-14 2004-08-17 Sangamo Biosciences, Inc. Functional genomics using zinc finger proteins
US6780590B2 (en) 1999-09-14 2004-08-24 Sangamo Biosciences, Inc. Gene identification
US7235354B2 (en) 1999-09-14 2007-06-26 Sangamo Biosciences, Inc. Functional genomics using zinc finger proteins
US20020081614A1 (en) 1999-09-14 2002-06-27 Sangamo Biosciences, Inc. Functional genomics using zinc finger proteins
US20040203064A1 (en) 1999-09-14 2004-10-14 Sangamo Biosciences, Inc. Functional genomics using zinc finger proteins
US6599692B1 (en) 1999-09-14 2003-07-29 Sangamo Bioscience, Inc. Functional genomics using zinc finger proteins
WO2001038547A2 (en) 1999-11-24 2001-05-31 Mcs Micro Carrier Systems Gmbh Polypeptides comprising multimers of nuclear localization signals or of protein transduction domains and their use for transferring molecules into cells
US20060166263A1 (en) 1999-12-06 2006-07-27 Sangamo Biosciences, Inc. Randomized libraries of zinc finger proteins
US7491531B2 (en) 1999-12-06 2009-02-17 Sangamo Biosciences, Inc. Randomized libraries of zinc finger proteins
US6503717B2 (en) 1999-12-06 2003-01-07 Sangamo Biosciences, Inc. Methods of using randomized libraries of zinc finger proteins for the identification of gene function
US20060292621A1 (en) 1999-12-06 2006-12-28 Sangamo Biosciences, Inc. Randomized libraries of zinc finger proteins
US7943553B2 (en) 1999-12-06 2011-05-17 Sangamo Biosciences, Inc. Randomized libraries of zinc finger proteins
US20030134318A1 (en) 1999-12-06 2003-07-17 Sangamo Biosciences, Inc. Methods of using randomized libraries of zinc finger proteins for the identification of gene function
US6689558B2 (en) 2000-02-08 2004-02-10 Sangamo Biosciences, Inc. Cells for drug discovery
US7045304B2 (en) 2000-02-08 2006-05-16 Sangamo Biosciences, Inc. Cells for drug discovery
WO2001081609A2 (en) 2000-03-22 2001-11-01 Chiron Corporation Compositions and methods for generating an immune response utilizing alphavirus-based vector systems
WO2001083793A2 (en) 2000-04-28 2001-11-08 Sangamo Biosciences, Inc. Targeted modification of chromatin structure
US20040048787A1 (en) 2000-05-31 2004-03-11 Copernicus Therapeutics, Inc. Lyophilizable and enhanced compacted nucleic acids
US6919204B2 (en) 2000-09-29 2005-07-19 Sangamo Biosciences, Inc. Modulation of gene expression using localization domains
WO2002026960A2 (en) 2000-09-29 2002-04-04 Sangamo Biosciences, Inc. Modulation of gene expression using localization domains
US20020150626A1 (en) 2000-10-16 2002-10-17 Kohane Daniel S. Lipid-protein-sugar particles for delivery of nucleic acids
US6794136B1 (en) 2000-11-20 2004-09-21 Sangamo Biosciences, Inc. Iterative optimization in the design of binding proteins
WO2002080982A2 (en) 2001-01-12 2002-10-17 Chiron Corporation Nucleic acid mucosal immunization
US20070065931A1 (en) 2001-01-22 2007-03-22 Sangamo Biosciences, Inc. Zinc finger proteins for DNA binding and gene regulation in plants
US8680021B2 (en) 2001-01-22 2014-03-25 Sangamo Biosciences, Inc. Zinc finger proteins for DNA binding and gene regulation in plants
US7705139B2 (en) 2001-01-22 2010-04-27 Sangamo Biosciences, Inc. Zinc finger proteins for DNA binding and gene regulation in plants
US20060294617A1 (en) 2001-01-22 2006-12-28 Sangamo Biosciences, Inc. Zinc finger proteins for DNA binding and gene regulation in plants
US20030108880A1 (en) 2001-01-22 2003-06-12 Sangamo Biosciences Modified zinc finger binding proteins
US7273923B2 (en) 2001-01-22 2007-09-25 Sangamo Biosciences, Inc. Zinc finger proteins for DNA binding and gene regulation in plants
US7476500B1 (en) 2001-03-19 2009-01-13 President And Fellows Of Harvard College In vivo selection system for enzyme activity
US20030203865A1 (en) 2001-04-30 2003-10-30 Pierrot Harvie Lipid-comprising drug delivery complexes and methods for their production
WO2002099035A2 (en) 2001-05-31 2002-12-12 Chiron Corporation Chimeric alphavirus replicon particles
US8735153B2 (en) 2001-09-24 2014-05-27 Sangamo Biosciences, Inc. Modulation of stem cells using zinc finger proteins
US9624509B2 (en) 2001-09-24 2017-04-18 Sangamo Biosciences, Inc. Modulation of stem cells using zinc finger proteins
US7262054B2 (en) 2002-01-22 2007-08-28 Sangamo Biosciences, Inc. Zinc finger proteins for DNA binding and gene regulation in plants
US9145565B2 (en) 2002-01-23 2015-09-29 University Of Utah Research Foundation Targeted chromosomal mutagenesis using zinc finger nucleases
US20060078552A1 (en) 2002-03-15 2006-04-13 Sylvain Arnould Hybrid and single chain meganucleases and use thereof
US20040002092A1 (en) 2002-03-15 2004-01-01 Sylvain Arnould Hybrid and single chain meganucleases and use thereof
US20100151556A1 (en) 2002-03-15 2010-06-17 Cellectis Hybrid and single chain meganucleases and use thereof
WO2003080809A2 (en) 2002-03-21 2003-10-02 Sangamo Biosciences, Inc. Methods and compositions for using zinc finger endonucleases to enhance homologous recombination
US20080209587A1 (en) 2002-03-21 2008-08-28 Sangamo Biosciences, Inc. Methods and compositions for using zinc finger endonucleases to enhance homologous recombination
US20090305402A1 (en) 2002-03-21 2009-12-10 Sangamo Biosciences, Inc. Methods and compositions for using zinc finger endonucleases to enhance homologous recombination
US7947873B2 (en) 2002-04-17 2011-05-24 Sangamo Biosciences, Inc. Compositions and methods for regulation of plant gamma-tocopherol methyltransferase
US20110216622A1 (en) 2002-06-28 2011-09-08 Protiva Biotherapeutics, Inc. Liposomal apparatus and manufacturing method
US20040142025A1 (en) 2002-06-28 2004-07-22 Protiva Biotherapeutics Ltd. Liposomal apparatus and manufacturing methods
US7939327B2 (en) 2002-08-29 2011-05-10 Sangamo Biosciences, Inc. Simultaneous modulation of multiple genes
US20040091991A1 (en) 2002-08-29 2004-05-13 Sangamo Biosciences, Inc. Simultaneous modulation of multiple genes
US7361635B2 (en) 2002-08-29 2008-04-22 Sangamo Biosciences, Inc. Simultaneous modulation of multiple genes
US20080233641A1 (en) 2002-08-29 2008-09-25 Sangamo Biosciences, Inc. Simultaneous modulation of multiple genes
US20040235002A1 (en) 2002-09-20 2004-11-25 Sangamo Biosciences, Inc. Multiplex screening assays
WO2004067736A2 (en) 2003-01-28 2004-08-12 Cellectis Custom-made meganuclease and use thereof
US7842489B2 (en) 2003-01-28 2010-11-30 Cellectis Use of meganucleases for inducing homologous recombination ex vivo and in toto in vertebrate somatic tissues and application thereof
US20140121115A1 (en) 2003-01-28 2014-05-01 Cellectis Custom-made meganuclease and use thereof
US20100144012A1 (en) 2003-01-28 2010-06-10 Cellectis Use of meganucleases for inducing homologous recombination ex vivo and in toto in vertebrate somatic tissues and application thereof
US20060240093A1 (en) 2003-07-16 2006-10-26 Protiva Biotherapeutics, Inc. Lipid encapsulated interfering rna
US20120058188A1 (en) 2003-07-16 2012-03-08 Protiva Biotherapeutics, Inc. Lipid encapsulated interfering rna
US20050064595A1 (en) 2003-07-16 2005-03-24 Protiva Biotherapeutics, Inc. Lipid encapsulated interfering RNA
US20050017054A1 (en) 2003-07-23 2005-01-27 Tom Iverson Flyback transformer wire attach method to printed circuit board
US9260726B2 (en) 2003-08-08 2016-02-16 Sangamo Biosciences, Inc. Targeted integration and expression on exogenous nucleic acid sequences
US20130244332A1 (en) 2003-08-08 2013-09-19 Sangamo Biosciences, Inc. Taregted integration and expression of exogenous nucleic acid sequences
US20110091525A1 (en) 2003-09-15 2011-04-21 Protiva Biotherapeutics, Inc. Polyethyleneglycol-modified lipid compounds and uses thereof
US20050175682A1 (en) 2003-09-15 2005-08-11 Protiva Biotherapeutics, Inc. Polyethyleneglycol-modified lipid compounds and uses thereof
US7692068B2 (en) 2003-10-14 2010-04-06 Athenix Corporation AXMI-010, a delta-endotoxin gene and methods for its use
US7541517B2 (en) 2003-12-22 2009-06-02 Pioneer Hi-Bred International, Inc. Bacillus thuringiensis CRY9 nucleic acids
US7253273B2 (en) 2004-04-08 2007-08-07 Sangamo Biosciences, Inc. Treatment of neuropathic pain with zinc finger proteins
US20100256221A1 (en) 2004-04-08 2010-10-07 Sangamo Biosciences, Inc. Methods and compositions for treating neuropathic and neurodegenerative conditions
US8466267B2 (en) 2004-04-08 2013-06-18 Sangamo Biosciences, Inc. Nucleic acid encoding a zinc finger that recognizes the sodium channel Nav 1.8 (PN3) gene
US20050267061A1 (en) 2004-04-08 2005-12-01 Sangamo Biosciences, Inc. Methods and compositions for treating neuropathic and neurodegenerative conditions
WO2005105989A1 (en) 2004-04-30 2005-11-10 Cellectis I-dmoi derivatives with enhanced activity at 37°c and use thereof.
US8211685B2 (en) 2004-04-30 2012-07-03 Cellectis I-DmoI derivatives with enhanced activity at 37° C and use thereof
US20060008910A1 (en) 2004-06-07 2006-01-12 Protiva Biotherapeuties, Inc. Lipid encapsulated interfering RNA
US20110060032A1 (en) 2004-06-07 2011-03-10 Protiva Biotherapeutics, Inc. Lipid encapsulating interfering rna
US20060083780A1 (en) 2004-06-07 2006-04-20 Protiva Biotherapeutics, Inc. Cationic lipids and methods of use
US20110262527A1 (en) 2004-06-07 2011-10-27 Protiva Biotherapeutics, Inc. Cationic lipids and methods of use
US20060063231A1 (en) 2004-09-16 2006-03-23 Sangamo Biosciences, Inc. Compositions and methods for protein production
US7358085B2 (en) 2005-02-28 2008-04-15 Sangamo Biosciences, Inc. Anti-angiogenic methods and compositions
WO2006097853A1 (en) 2005-03-15 2006-09-21 Cellectis I-crei meganuclease variants with modified specificity, method of preparation and uses thereof
US20110158974A1 (en) 2005-03-15 2011-06-30 Cellectis Heterodimeric Meganucleases and Use Thereof
US7897372B2 (en) 2005-03-15 2011-03-01 Cellectis I-CreI meganuclease variants with modified specificity, method of preparation and uses thereof
US20120258537A1 (en) 2005-03-15 2012-10-11 Cellectis I-crei meganuclease variants with modified specificity, method of preparation and uses thereof
US20110072527A1 (en) 2005-03-15 2011-03-24 Cellectis I-crei meganuclease variants with modified specificity, method of preparation and uses thereof
US8715992B2 (en) 2005-03-15 2014-05-06 Cellectis I-CreI meganuclease variants with modified specificity, method of preparation and uses thereof
US20080031109A1 (en) 2005-07-15 2008-02-07 Yoshikazu Takashima Method and apparatus for processing information, method and apparatus for manufacturing information recording medium, information recording medium, and computer program
US20070134796A1 (en) 2005-07-26 2007-06-14 Sangamo Biosciences, Inc. Targeted integration and expression of exogenous nucleic acid sequences
US8313925B2 (en) 2005-07-26 2012-11-20 Sangamo Biosciences, Inc. Zinc finger proteins and method for inactivating a dhfr gene in a chinese hamster ovary cell
US20090258363A1 (en) 2005-07-26 2009-10-15 Sangamo Biosciences, Inc. Targeted integration and expression of exogenous nucleic acid sequences
US20070042031A1 (en) 2005-07-27 2007-02-22 Protiva Biotherapeutics, Inc. Systems and methods for manufacturing liposomes
US8119361B2 (en) 2005-10-18 2012-02-21 Duke University Methods of cleaving DNA with rationally-designed meganucleases
US8148098B2 (en) 2005-10-18 2012-04-03 Duke University Methods of cleaving DNA with rationally-designed meganucleases
US8143016B2 (en) 2005-10-18 2012-03-27 Duke University Methods of cleaving DNA with rationally-designed meganucleases
US8163514B2 (en) 2005-10-18 2012-04-24 Duke University Methods of cleaving DNA with rationally-designed meganucleases
US8143015B2 (en) 2005-10-18 2012-03-27 Duke University Methods of cleaving DNA with rationally-designed meganucleases
US8124369B2 (en) 2005-10-18 2012-02-28 Duke University Method of cleaving DNA with rationally-designed meganucleases
US8129134B2 (en) 2005-10-18 2012-03-06 Duke University Methods of cleaving DNA with rationally-designed meganucleases
US8133697B2 (en) 2005-10-18 2012-03-13 Duke University Methods of cleaving DNA with rationally-designed meganucleases
US20130326644A1 (en) 2005-10-25 2013-12-05 Cellectis, S.A. Laglidadg homing endonuclease variants having mutations in two functional subdomains and use thereof
US20110179507A1 (en) 2005-10-25 2011-07-21 Paques Frederic Laglidadg homing endonuclease variants having mutations in two functional subdomains and use thereof
US20090220476A1 (en) 2005-10-25 2009-09-03 Cellectis Laglidadg homing endonuclease variants having mutations in two functional subdomains and use thereof
WO2007049095A1 (en) 2005-10-25 2007-05-03 Cellectis Laglidadg homing endonuclease variants having mutations in two functional subdomains and use thereof
WO2007057781A2 (en) 2005-10-25 2007-05-24 Cellectis Laglidadg homing endonuclease variants having mutations in two functional subdomains and use thereof.
US20090305977A1 (en) 2006-02-09 2009-12-10 Sangamo Bioscience, Inc. Method for treating peripheral arterial disease with zinc finger proteins
US20090222937A1 (en) 2006-02-13 2009-09-03 Cellectis Meganuclease variants cleaving a dna target sequence from a xeroderma pigmentosum gene and uses thereof
WO2007123636A2 (en) 2006-03-27 2007-11-01 Children's Hospital & Regional Medical Center Compositions and methods comprising the use of cell surface displayed homing endonucleases
US20090162937A1 (en) 2006-03-27 2009-06-25 Children's Hospital & Regional Medical Center Compositions and methods comprising the use of cell surface displayed homing endonucleases
US10407672B2 (en) 2006-03-27 2019-09-10 Seattle Children's Hospital Compositions and methods comprising the use of cell surface displayed homing endonucleases
US8569253B2 (en) 2006-05-25 2013-10-29 Sangamo Biosciences, Inc. Methods and compositions for gene inactivation
US7951925B2 (en) 2006-05-25 2011-05-31 Sangamo Biosciences, Inc. Methods and compositions for gene inactivation
US20160215298A1 (en) 2006-05-25 2016-07-28 Sangamo Biosciences, Inc. Methods and compositions for increased transgene expression
US8524221B2 (en) 2006-05-25 2013-09-03 Sangamo Biosciences, Inc. Methods and compositions for gene inactivation
US9428756B2 (en) 2006-08-11 2016-08-30 Dow Agrosciences Llc Zinc finger nuclease-mediated homologous recombination
US20130059387A1 (en) 2006-11-14 2013-03-07 Cellectis Meganuclease variants cleaving a dna target sequence from the hprt gene and uses thereof
US20100146651A1 (en) 2006-11-14 2010-06-10 Cellectis Meganuclease variants cleaving a dna target sequence from the hprt gene and uses thereof
US20100167357A1 (en) 2007-02-01 2010-07-01 Cellectis Obligate heterodimer meganucleases and uses thereof
US20100086533A1 (en) 2007-02-19 2010-04-08 Cellectis Laglidadg homing endonuclease variants having novel substrate specificity and use thereof
WO2008102198A1 (en) 2007-02-19 2008-08-28 Cellectis Laglidadg homing endonuclease variants having novel substrate specificity and use thereof
US20140112904A9 (en) 2007-06-06 2014-04-24 Cellectis Method for enhancing the cleavage activity of i-crei derived meganucleases
US20100325745A1 (en) 2007-06-06 2010-12-23 Cellectis Meganuclease variants cleaving a dna target sequence from the mouse rosa26 locus and uses thereof
US20100203031A1 (en) 2007-06-06 2010-08-12 Cellectis Method for enhancing the cleavage activity of i-crei derived meganucleases
US20130236946A1 (en) 2007-06-06 2013-09-12 Cellectis Meganuclease variants cleaving a dna target sequence from the mouse rosa26 locus and uses thereof
US8426177B2 (en) 2007-06-06 2013-04-23 Cellectis Meganuclease variants cleaving a DNA target sequence from the mouse ROSA26 locus and uses thereof
US20100229252A1 (en) 2007-07-23 2010-09-09 Cellectis Meganuclease variants cleaving a dna target sequence from the human hemoglobin beta gene and uses thereof
US20140038239A1 (en) 2007-07-23 2014-02-06 Cellectis Meganuclease variants cleaving a dna target sequence from the human hemoglobin beta gene and uses thereof
US20110091441A1 (en) 2007-08-03 2011-04-21 Cellectis Meganuclease variants cleaving a dna target sequence from the human interleukin-2 receptor gamma chain gene and uses thereof
US20140017731A1 (en) 2007-08-03 2014-01-16 Cellectis Meganuclease variants cleaving a dna target sequence from the human interleukin-2 receptor gamma chain gene and uses thereof
US20110207199A1 (en) 2007-08-03 2011-08-25 Cellectis Novel method to generate meganucleases with altered characteristics
US9121072B2 (en) 2007-09-27 2015-09-01 Sangamo Biosciences, Inc. Rapid screening of biologically active nucleases and isolation of nuclease-modified cells
US9115409B2 (en) 2007-09-27 2015-08-25 Sangamo Biosciences, Inc. Rapid screening of biologically active nucleases and isolation of nuclease-modified cells
US20110041194A1 (en) 2007-11-28 2011-02-17 Cellectis I-msoi homing endonuclease variants having novel substrate specificity and use thereof
WO2009068937A1 (en) 2007-11-28 2009-06-04 Cellectis I-msoi homing endonuclease variants having novel substrate specificity and use thereof
US20110117125A1 (en) 2008-01-02 2011-05-19 Tekmira Pharmaceuticals Corporation Compositions and methods for the delivery of nucleic acids
US20120183581A1 (en) 2008-04-15 2012-07-19 Protiva Biotherapeutics, Inc Novel lipid formulations for nucleic acid delivery
US20100130588A1 (en) 2008-04-15 2010-05-27 Protiva Biotherapeutics, Inc. Novel lipid formulations for nucleic acid delivery
US8846578B2 (en) 2008-04-16 2014-09-30 The General Hospital Corporation Zinc finger nuclease for the CFTR gene and methods of use thereof
WO2010001189A1 (en) 2008-07-03 2010-01-07 Cellectis The crystal structure of i-dmoi in complex with its dna target, improved chimeric meganucleases and uses thereof
US10273524B2 (en) 2008-07-14 2019-04-30 Precision Biosciences, Inc. Recognition sequences for I-CreI-derived meganucleases and uses thereof
US10287626B2 (en) 2008-07-14 2019-05-14 Precision Biosciences, Inc. Recognition sequences for I-CreI-derived meganucleases and uses thereof
US9683257B2 (en) 2008-07-14 2017-06-20 Precision Biosciences, Inc. Recognition sequences for I-CreI-derived meganucleases and uses thereof
US9273296B2 (en) 2008-09-08 2016-03-01 Cellectis Meganuclease variants cleaving a DNA target sequence from a glutamine synthetase gene and uses thereof
US20110225664A1 (en) 2008-09-08 2011-09-15 Cellectis Meganuclease variants cleaving a dna target sequence from a glutamine synthetase gene and uses thereof
US9365864B2 (en) 2008-10-23 2016-06-14 Cellectis Meganuclease recombination system
WO2010046786A1 (en) 2008-10-23 2010-04-29 Cellectis Meganuclease recombination system
US20140004608A1 (en) 2008-10-23 2014-01-02 Cellectis Meganuclease recombination system
US20110263028A1 (en) 2008-10-23 2011-10-27 Cellectis Meganuclease recombination system
US8476072B2 (en) 2008-10-23 2013-07-02 Cellectis Meganuclease recombination system
US20100311124A1 (en) 2008-10-29 2010-12-09 Sangamo Biosciences, Inc. Methods and compositions for inactivating glutamine synthetase gene expression
US9388426B2 (en) 2008-10-29 2016-07-12 Sangamo Biosciences, Inc. Methods and compositions for inactivating glutamine synthetase gene expression
US8153399B2 (en) 2008-10-29 2012-04-10 Sangamo Biosciences, Inc. Methods and compositions for inactivating glutamine synthetase gene expression
US20150265708A1 (en) 2008-11-10 2015-09-24 Tekmira Pharmaceuticals Corporation Novel lipids and compositions for the delivery of therapeutics
US20110311582A1 (en) 2008-11-10 2011-12-22 Muthiah Manoharan Novel lipids and compositions for the delivery of therapeutics
US20160199485A1 (en) 2008-11-10 2016-07-14 Tekmira Pharmaceuticals Corporation Novel lipids and compositions for the delivery of therapeutics
US20110311583A1 (en) 2008-11-10 2011-12-22 Alnylam Pharmaceuticals, Inc. Novel lipids and compositions for the delivery of therapeutics
WO2010065123A1 (en) 2008-12-04 2010-06-10 Sangamo Biosciences, Inc. Genome editing in rats using zinc-finger nucleases
US20140178942A1 (en) 2009-04-21 2014-06-26 Cellectis Meganuclease variants cleaving at least one target in the genome of a retrovirus and uses thereof
US20120260356A1 (en) 2009-04-21 2012-10-11 Cellectis Meganuclease variants cleaving at least one target in the genome of a retrovirus and uses thereof
US8772008B2 (en) 2009-05-18 2014-07-08 Sangamo Biosciences, Inc. Methods and compositions for increasing nuclease activity
US20120171191A1 (en) 2009-05-26 2012-07-05 Cellectis Meganuclease variants cleaving the genome of a pathogenic non-integrating virus and uses thereof
US20150315557A1 (en) 2009-05-26 2015-11-05 Cellectis Meganuclease variants cleaving the genome of a pathogenic non-integrating virus and uses thereof
US20110076335A1 (en) 2009-07-01 2011-03-31 Protiva Biotherapeutics, Inc. Novel lipid formulations for delivery of therapeutic agents to solid tumors
US8569256B2 (en) 2009-07-01 2013-10-29 Protiva Biotherapeutics, Inc. Cationic lipids and methods for the delivery of therapeutic agents
US8575425B2 (en) 2009-07-02 2013-11-05 Athenix Corporation AXMI-205 pesticidal gene and methods for its use
US20120272348A1 (en) 2009-07-17 2012-10-25 Cellectis Viral vectors encoding a dna repair matrix and containing a virion-associated site specific meganuclease for gene targeting
US8802437B2 (en) 2009-09-24 2014-08-12 Cellectis Meganuclease reagents of uses thereof for treating genetic diseases caused by frame shift/non sense mutations
US20120301456A1 (en) 2009-09-24 2012-11-29 Cellectis Meganuclease reagents of uses thereof for treating genetic diseases caused by frame shift/non sense mutations
US20130022649A1 (en) 2009-12-01 2013-01-24 Protiva Biotherapeutics, Inc. Snalp formulations containing antioxidants
US20130338210A1 (en) 2009-12-07 2013-12-19 Alnylam Pharmaceuticals, Inc. Compositions for nucleic acid delivery
US20130017223A1 (en) 2009-12-18 2013-01-17 The University Of British Columbia Methods and compositions for delivery of nucleic acids
US20130053572A1 (en) 2010-01-22 2013-02-28 Steven L. Colletti Novel Cationic Lipids for Oligonucleotide Delivery
US20130045539A1 (en) 2010-02-18 2013-02-21 Cellectis Meganuclease recombination system
US9290748B2 (en) 2010-02-26 2016-03-22 Cellectis Use of endonucleases for inserting transgenes into safe harbor loci
US9567573B2 (en) 2010-04-26 2017-02-14 Sangamo Biosciences, Inc. Genome editing of a Rosa locus using nucleases
US20110265198A1 (en) 2010-04-26 2011-10-27 Sangamo Biosciences, Inc. Genome editing of a Rosa locus using nucleases
US20130108685A1 (en) 2010-04-28 2013-05-02 Takeshi Kuboyama Cationic lipid
US20140287500A1 (en) 2010-05-03 2014-09-25 Sangamo Biosciences, Inc. Compositions for linking zinc finger modules
US9163245B2 (en) 2010-05-03 2015-10-20 Sangamo Biosciences, Inc. Compositions for linking zinc finger modules
US8772453B2 (en) 2010-05-03 2014-07-08 Sangamo Biosciences, Inc. Compositions for linking zinc finger modules
US20110287512A1 (en) 2010-05-03 2011-11-24 Sangamo Biosciences, Inc. Compositions for linking zinc finger modules
US20130183282A1 (en) 2010-05-12 2013-07-18 Cellectis Meganuclease variants cleaving a DNA target sequence from the rhodopsin gene and uses thereof
US20130123338A1 (en) 2010-05-12 2013-05-16 Protiva Biotherapeutics, Inc. Novel cationic lipids and methods of use thereof
WO2011141705A1 (en) 2010-05-12 2011-11-17 Protiva Biotherapeutics, Inc. Novel cationic lipids and methods of use thereof
US20130145487A1 (en) 2010-05-12 2013-06-06 Cellectis Meganuclease variants cleaving a dna target sequence from the dystrophin gene and uses thereof
US20120027803A1 (en) 2010-06-03 2012-02-02 Alnylam Pharmaceuticals, Inc. Biodegradable lipids for the delivery of active agents
US20160009637A1 (en) 2010-06-03 2016-01-14 Alnylam Pharmaceuticals, Inc. Biodegradable lipids for the delivery of active agents
US20130196320A1 (en) 2010-06-15 2013-08-01 Cellectis Method for improving cleavage of dna by endonuclease sensitive to methylation
US20130189759A1 (en) 2010-07-07 2013-07-25 Cellectis Meganucleases variants cleaving a dna target sequence in the nanog gene and uses thereof
US20130323269A1 (en) 2010-07-30 2013-12-05 Muthiah Manoharan Methods and compositions for delivery of active agents
US8802934B2 (en) 2010-08-19 2014-08-12 Pioneer Hi Bred International Inc Bacillus thuringiensis gene with lepidopteran activity
US20120052582A1 (en) 2010-08-30 2012-03-01 Amyris, Inc. Nucleic acids, compositions and methods for the excision of target nucleic acids
US7919605B1 (en) 2010-08-30 2011-04-05 Amyris, Inc. Nucleic acids, compositions and methods for the excision of target nucleic acids
WO2012030747A1 (en) 2010-08-30 2012-03-08 Amyris, Inc. Nucleic acids, compositions and methods for the excision of target nucleic acids
US9018364B2 (en) 2010-08-30 2015-04-28 Amyris, Inc. Nucleic acids, compositions and methods for the excision of target nucleic acids
US20120172411A1 (en) 2010-09-17 2012-07-05 Protiva Biotherapeutics, Inc. Novel trialkyl cationic lipids and methods of use thereof
WO2012067428A2 (en) 2010-11-16 2012-05-24 주식회사 에피밸리 Group-iii nitride semiconductor light-emitting device
US20120252122A1 (en) 2011-01-05 2012-10-04 Sangamo Biosciences, Inc. Methods and compositions for increasing production of induced pluripotent stem cells (ipscs)
US20140200257A1 (en) 2011-01-11 2014-07-17 Alnylam Pharmaceuticals, Inc. Pegylated lipids and their use for drug delivery
US20120204282A1 (en) 2011-02-04 2012-08-09 Sangamo Biosciences, Inc. Methods and compositions for treating occular disorders
US20120295832A1 (en) 2011-05-17 2012-11-22 Arrowhead Research Corporation Novel Lipids and Compositions for Intracellular Delivery of Biologically Active Compounds
US9540623B2 (en) 2011-07-08 2017-01-10 Cellectis Method for increasing the efficiency of double-strand-break induced mutagenesis
WO2013016058A1 (en) 2011-07-22 2013-01-31 Merck Sharp & Dohme Corp. Novel bis-nitrogen containing cationic lipids for oligonucleotide delivery
WO2013044008A2 (en) 2011-09-21 2013-03-28 Sangamo Biosciences, Inc. Methods and compositions for regulation of transgene expression
US20150203446A1 (en) 2011-09-27 2015-07-23 Takeda Pharmaceutical Company Limited Di-aliphatic substituted pegylated lipids
US20130086373A1 (en) 2011-09-29 2013-04-04 Apple Inc. Customized content for electronic devices
WO2013086322A1 (en) 2011-12-07 2013-06-13 Alnylam Pharmaceuticals, Inc. Branched alkyl and cycloalkyl terminated biodegradable lipids for the delivery of active agents
US20140308304A1 (en) 2011-12-07 2014-10-16 Alnylam Pharmaceuticals, Inc. Lipids for the delivery of active agents
US20150273068A1 (en) 2011-12-07 2015-10-01 Alnylam Pharmaceuticals, Inc. Biodegradable lipids for the delivery of active agents
WO2013086373A1 (en) 2011-12-07 2013-06-13 Alnylam Pharmaceuticals, Inc. Lipids for the delivery of active agents
US20150005363A1 (en) 2011-12-07 2015-01-01 Alnylam Pharmaceuticals, Inc. Branched Alkyl And Cycloalkyl Terminated Biodegradable Lipids For The Delivery Of Active Agents
US20130195920A1 (en) 2011-12-07 2013-08-01 Alnylam Pharmaceuticals, Inc. Biodegradable lipids for the delivery of active agents
US20130245107A1 (en) 2011-12-16 2013-09-19 modeRNA Therapeutics Dlin-mc3-dma lipid nanoparticle delivery of modified polynucleotides
US20140068797A1 (en) 2012-05-25 2014-03-06 University Of Vienna Methods and compositions for rna-directed target dna modification and for rna-directed modulation of transcription
WO2014008334A1 (en) 2012-07-06 2014-01-09 Alnylam Pharmaceuticals, Inc. Stable non-aggregating nucleic acid lipid particle formulations
US20140017214A1 (en) 2012-07-11 2014-01-16 Sangamo Biosciences, Inc. Methods and compositions for delivery of biologics
US20140189896A1 (en) 2012-12-12 2014-07-03 Feng Zhang Crispr-cas component systems, methods and compositions for sequence manipulation
US20140357530A1 (en) 2012-12-12 2014-12-04 The Broad Institute Inc. Functional genomics using crispr-cas systems, compositions, methods, knock out libraries and applications thereof
US20140170753A1 (en) 2012-12-12 2014-06-19 Massachusetts Institute Of Technology Crispr-cas systems and methods for altering expression of gene products
US20140179770A1 (en) 2012-12-12 2014-06-26 Massachusetts Institute Of Technology Delivery, engineering and optimization of systems, methods and compositions for sequence manipulation and therapeutic applications
US20140179006A1 (en) 2012-12-12 2014-06-26 Massachusetts Institute Of Technology Crispr-cas component systems, methods and compositions for sequence manipulation
US20140273231A1 (en) 2012-12-12 2014-09-18 The Broad Institute, Inc. Crispr-cas component systems, methods and compositions for sequence manipulation
US20140186958A1 (en) 2012-12-12 2014-07-03 Feng Zhang Engineering and optimization of systems, methods and compositions for sequence manipulation with functional domains
US20140335620A1 (en) 2012-12-12 2014-11-13 The Broad Institute, Inc. Engineering and optimization of improved systems, methods and enzyme compositions for sequence manipulation
US20140273234A1 (en) 2012-12-12 2014-09-18 The Board Institute, Inc. Engineering and optimization of improved systems, methods and enzyme compositions for sequence manipulation
US20140248702A1 (en) 2012-12-12 2014-09-04 The Broad Institute, Inc. CRISPR-Cas Nickase Systems, Methods And Compositions For Sequence Manipulation in Eukaryotes
US20140242664A1 (en) 2012-12-12 2014-08-28 The Broad Institute, Inc. Engineering of systems, methods and optimized guide compositions for sequence manipulation
US20140310830A1 (en) 2012-12-12 2014-10-16 Feng Zhang CRISPR-Cas Nickase Systems, Methods And Compositions For Sequence Manipulation in Eukaryotes
US20140186919A1 (en) 2012-12-12 2014-07-03 Feng Zhang Engineering and optimization of improved systems, methods and enzyme compositions for sequence manipulation
US20140256046A1 (en) 2012-12-12 2014-09-11 Massachusetts Institute Of Technology Engineering and optimization of systems, methods and compositions for sequence manipulation with functional domains
US20140186843A1 (en) 2012-12-12 2014-07-03 Massachusetts Institute Of Technology Methods, systems, and apparatus for identifying target sequences for cas enzymes or crispr-cas systems for target sequences and conveying results thereof
US20140273232A1 (en) 2012-12-12 2014-09-18 The Broad Institute, Inc. Engineering of systems, methods and optimized guide compositions for sequence manipulation
US8795965B2 (en) 2012-12-12 2014-08-05 The Broad Institute, Inc. CRISPR-Cas component systems, methods and compositions for sequence manipulation
US20140242699A1 (en) 2012-12-12 2014-08-28 Massachusetts Institute Of Technology Delivery, engineering and optimization of systems, methods and compositions for sequence manipulation and therapeutic applications
US20140227787A1 (en) 2012-12-12 2014-08-14 The Broad Institute, Inc. Crispr-cas systems and methods for altering expression of gene products
US20140234972A1 (en) 2012-12-12 2014-08-21 Massachusetts Institute Of Technology CRISPR-CAS Nickase Systems, Methods And Compositions For Sequence Manipulation in Eukaryotes
US20140242700A1 (en) 2012-12-12 2014-08-28 Massachusetts Institute Of Technology Engineering and optimization of improved systems, methods and enzyme compositions for sequence manipulation
US20140298547A1 (en) 2012-12-13 2014-10-02 Dow Agrosciences Llc Dna detection methods for site specific nuclease activity
US20140356958A1 (en) 2012-12-17 2014-12-04 President And Fellows Of Harvard College RNA-Guided Human Genome Engineering
US20140342458A1 (en) 2012-12-17 2014-11-20 President And Fellows Of Harvard College RNA-Guided Human Genome Engineering
US20140342457A1 (en) 2012-12-17 2014-11-20 President And Fellows Of Harvard College RNA-Guided Human Genome Engineering
US20140342456A1 (en) 2012-12-17 2014-11-20 President And Fellows Of Harvard College RNA-Guided Human Genome Engineering
US20140242702A1 (en) 2013-02-25 2014-08-28 Sigma Aldrich Co. Llc Methods and compositions for enhancing nuclease-mediated gene disruption
US20140315985A1 (en) 2013-03-14 2014-10-23 Caribou Biosciences, Inc. Compositions and methods of nucleic acid-targeting nucleic acids
US20140349400A1 (en) 2013-03-15 2014-11-27 Massachusetts Institute Of Technology Programmable Modification of DNA
US20140364333A1 (en) 2013-03-15 2014-12-11 President And Fellows Of Harvard College Methods for Live Imaging of Cells
US20140273037A1 (en) 2013-03-15 2014-09-18 System Biosciences, Llc Compositions and methods directed to crispr/cas genomic engineering systems
US20140273226A1 (en) 2013-03-15 2014-09-18 System Biosciences, Llc Crispr/cas systems for genomic modification and gene modulation
US20140273235A1 (en) 2013-03-15 2014-09-18 Regents Of The University Of Minnesota ENGINEERING PLANT GENOMES USING CRISPR/Cas SYSTEMS
US20140273230A1 (en) 2013-03-15 2014-09-18 Sigma-Aldrich Co., Llc Crispr-based genome modification and regulation
US20140377868A1 (en) 2013-03-15 2014-12-25 The General Hospital Corporation Rna-guided targeting of genetic and epigenomic regulatory proteins to specific genomic loci
US20140273233A1 (en) 2013-03-15 2014-09-18 Sigma-Aldrich Co., Llc Crispr-based genome modification and regulation
US20140287938A1 (en) 2013-03-15 2014-09-25 The Broad Institute, Inc. Recombinant virus and preparations thereof
US20140295556A1 (en) 2013-03-15 2014-10-02 The General Hospital Corporation Using RNA-guided FokI Nucleases (RFNs) to Increase Specificity for RNA-Guided Genome Editing
US20140295557A1 (en) 2013-03-15 2014-10-02 The General Hospital Corporation Using Truncated Guide RNAs (tru-gRNAs) to Increase Specificity for RNA-Guided Genome Editing
US20140304853A1 (en) 2013-04-05 2014-10-09 Sangamo Biosciences, Inc. Methods and compositions for integration of an exogenous sequence within the genome of plants
US20140310828A1 (en) 2013-04-16 2014-10-16 Regeneron Pharmaceuticals, Inc. Targeted modification of rat genome
US20140309487A1 (en) 2013-04-16 2014-10-16 Regeneron Pharmaceuticals, Inc. Targeted modification of rat genome
US20140335063A1 (en) 2013-05-10 2014-11-13 Sangamo Biosciences, Inc. Delivery methods and compositions for nuclease-mediated genome engineering
US20140349405A1 (en) 2013-05-22 2014-11-27 Wisconsin Alumni Research Foundation Rna-directed dna cleavage and gene editing by cas9 enzyme from neisseria meningitidis
US20140356867A1 (en) 2013-05-29 2014-12-04 Agilent Technologies, Inc. Nucleic acid enrichment using cas9
US20140357523A1 (en) 2013-05-29 2014-12-04 Agilent Technologies, Inc. Method for fragmenting genomic dna using cas9
US10006052B2 (en) 2013-05-31 2018-06-26 Cellectis Laglidadg homing endonuclease cleaving the C-C chemokine receptor type-5 (CCR5) gene and uses thereof
US10000746B2 (en) 2013-05-31 2018-06-19 Cellectis LAGLIDADG homing endonuclease cleaving the T cell receptor alpha gene and uses thereof
US20140356956A1 (en) 2013-06-04 2014-12-04 President And Fellows Of Harvard College RNA-Guided Transcriptional Regulation
US20140356959A1 (en) 2013-06-04 2014-12-04 President And Fellows Of Harvard College RNA-Guided Transcriptional Regulation
US20150064789A1 (en) 2013-08-28 2015-03-05 Sangamo Biosciences, Inc. Compositions for linking dna-binding domains and cleavage domains
US9567609B2 (en) 2013-08-28 2017-02-14 Sangamo Biosciences, Inc. Compositions for linking DNA-binding domains and cleavage domains
WO2015031619A1 (en) 2013-08-28 2015-03-05 Sangamo Biosciences, Inc. Compositions for linking dna-binding domains and cleavage domains
US10858639B2 (en) 2013-09-06 2020-12-08 President And Fellows Of Harvard College CAS9 variants and uses thereof
US20150267205A1 (en) 2014-03-18 2015-09-24 Sangamo Biosciences, Inc. Methods and compositions for regulation of zinc finger protein expression
US9624498B2 (en) 2014-03-18 2017-04-18 Sangamo Biosciences, Inc. Methods and compositions for regulation of zinc finger protein expression
WO2015143046A2 (en) 2014-03-18 2015-09-24 Sangamo Biosciences, Inc. Methods and compositions for regulation of zinc finger protein expression
WO2015199952A1 (en) 2014-06-25 2015-12-30 Acuitas Therapeutics Inc. Novel lipids and lipid nanoparticle formulations for delivery of nucleic acids
WO2016014794A1 (en) 2014-07-25 2016-01-28 Sangamo Biosciences, Inc. Methods and compositions for modulating nuclease-mediated genome engineering in hematopoietic stem cells
US11071790B2 (en) 2014-10-29 2021-07-27 Massachusetts Eye And Ear Infirmary Method for efficient delivery of therapeutic molecules in vitro and in vivo
WO2016106236A1 (en) 2014-12-23 2016-06-30 The Broad Institute Inc. Rna-targeting system
WO2016183298A2 (en) 2015-05-12 2016-11-17 Sangamo Biosciences, Inc. Nuclease-mediated regulation of gene expression
WO2016189532A1 (en) 2015-05-26 2016-12-01 Ramot At Tel-Aviv University Ltd. Targeted lipid particles for systemic delivery of nucleic acid molecules to leukocytes
WO2017004143A1 (en) 2015-06-29 2017-01-05 Acuitas Therapeutics Inc. Lipids and lipid nanoparticle formulations for delivery of nucleic acids
US20170210697A1 (en) 2015-09-17 2017-07-27 Modernatx, Inc. Compounds and compositions for intracellular delivery of therapeutic agents
WO2017070633A2 (en) 2015-10-23 2017-04-27 President And Fellows Of Harvard College Evolved cas9 proteins for gene editing
US20200121809A1 (en) 2015-10-28 2020-04-23 Erikc A. HARWOOD Lipid nanoparticle formulations
US20170119904A1 (en) 2015-10-28 2017-05-04 Acuitas Therapeutics, Inc. Novel lipids and lipid nanoparticle formulations for delivery of nucleic acids
WO2017075531A1 (en) 2015-10-28 2017-05-04 Acuitas Therapeutics, Inc. Novel lipids and lipid nanoparticle formulations for delivery of nucleic acids
WO2017091512A1 (en) 2015-11-23 2017-06-01 Sangamo Biosciences, Inc. Methods and compositions for engineering immunity
WO2017117528A1 (en) 2015-12-30 2017-07-06 Acuitas Therapeutics, Inc. Lipids and lipid nanoparticle formulations for delivery of nucleic acids
WO2017136049A1 (en) 2016-02-02 2017-08-10 Sangamo Biosciences, Inc. Compositions for linking dna-binding domains and cleavage domains
US20170218349A1 (en) 2016-02-02 2017-08-03 Sangamo Biosciences, Inc. Compositions for linking dna-binding domains and cleavage domains
US20190240354A1 (en) 2016-06-30 2019-08-08 Arbutus Biopharma Corporation Compositions and methods for delivering messenger rna
US10947530B2 (en) 2016-08-03 2021-03-16 President And Fellows Of Harvard College Adenosine nucleobase editors and uses thereof
US20200010835A1 (en) 2016-08-24 2020-01-09 President And Fellows Of Harvard College Incorporation of unnatural amino acids into proteins using base editing
WO2018071565A1 (en) 2016-10-11 2018-04-19 Bluebird Bio, Inc. TCRa HOMING ENDONUCLEASE VARIANTS
US20210128488A1 (en) 2017-08-16 2021-05-06 Acuitas Therapeutics, Inc. Lipids for use in lipid nanoparticle formulations
WO2019135816A2 (en) 2017-10-23 2019-07-11 The Broad Institute, Inc. Novel nucleic acid modifiers
WO2019089828A1 (en) 2017-10-31 2019-05-09 Acuitas Therapeutics, Inc. Lamellar lipid nanoparticles
WO2019126558A1 (en) 2017-12-20 2019-06-27 Bluebird Bio, Inc. Ahr homing endonuclease variants, compositions, and methods of use
WO2019152557A1 (en) 2018-01-30 2019-08-08 Modernatx, Inc. Compositions and methods for delivery of agents to immune cells
WO2019232095A1 (en) 2018-05-30 2019-12-05 Translate Bio, Inc. Vitamin cationic lipids
US20210121411A1 (en) 2018-06-19 2021-04-29 The Board Of Regents Of The University Of Texas System Lipid nanoparticle compositions for delivery of mrna and long nucleic acids
US20220145296A1 (en) 2018-12-27 2022-05-12 LifeEDIT Therapeutics, Inc. Polypeptides useful for gene editing and methods of use
WO2020191233A1 (en) 2019-03-19 2020-09-24 The Broad Institute, Inc. Methods and compositions for editing nucleotide sequences
US11447770B1 (en) 2019-03-19 2022-09-20 The Broad Institute, Inc. Methods and compositions for prime editing nucleotide sequences
US20220204975A1 (en) 2019-04-12 2022-06-30 President And Fellows Of Harvard College System for genome editing
US20210087135A1 (en) 2019-09-19 2021-03-25 Modernatx, Inc. Branched tail lipid compounds and compositions for intracellular delivery of therapeutic agents
WO2021077067A1 (en) 2019-10-18 2021-04-22 The Trustees Of The University Of Pennsylvania Lipid nanoparticles and formulations thereof for car mrna delivery
WO2021204179A1 (en) 2020-04-09 2021-10-14 Suzhou Abogen Biosciences Co., Ltd. Nucleic acid vaccines for coronavirus

Non-Patent Citations (188)

* Cited by examiner, † Cited by third party
Title
"Methods in Molecular Biology", 2007, HUMANA PRESS, article "Baculovirus and Insect Cell Expression Protocols"
"Molecular Cloning A Laboratory Manual", 1991, COLD SPRING HARBOR LABORATORY PRESS
ALTSCHUL ET AL., J. MOL. BIOL., vol. 215, 1990, pages 403 - 410
ALTSCHUL, S. F. ET AL., J. MOLEC. BIOL., vol. 215, 1990, pages 403
AUER ET AL., GENOME RES., 31 October 2013 (2013-10-31)
AUTIERIAGRAWAL, J. BIOL. CHEM., vol. 273, 1998, pages 14731 - 37
BARANICK ET AL., PROC. NATL. ACAD SCI. U.S.A., vol. 105, no. 12, 2008, pages 4733 - 4738
BARTLETT ET AL., PLANT METHODS, vol. 4, 2008, pages 1 - 12
BATES, G. W., METHODS IN MOLECULAR BIOLOGY, vol. 111, 1999, pages 359 - 366
BENVENISTYNESHIF, PROC. NATL. ACAD. SCI. USA, vol. 83, 1986, pages 9551 - 9555
BERT ET AL., RNA, vol. 12, no. 6, 2006, pages 1074 - 1083
BINNSTHOMASHOW, ANNUAL REVIEWS IN MICROBIOLOGY, vol. 42, 1988, pages 575 - 606
BORIS-LAWRIETEMIN, CUR. OPIN. GENET. DEVELOP., vol. 3, 1993, pages 102 - 109
BOSHART ET AL., CELL, vol. 41, 1985, pages 521
BRINER ET AL., MOL CELL, vol. 56, no. 2, 23 October 2014 (2014-10-23), pages 333 - 9
BUMS, PROC. NATL. ACAD. SCI. USA, vol. 90, 1993, pages 8033 - 8037
BURSTEIN ET AL.: "New CRISPR-Cas systems from uncultivated microbes", CELL RES, 21 February 2017 (2017-02-21)
CABIOS, vol. 4, 1989, pages 11 - 17
CARILLO, H.LIPMAN, D., SIAM J APPLIED MATH., vol. 48, 1988, pages 1073
CARROLL ET AL., GENETICS, vol. 188, no. 4, 2011, pages 773 - 782
CARROLL ET AL., NAT. PROTOC., vol. 1, 2006, pages 1329 - 1341
CARROLL ET AL.: "Genome Engineering with Zinc-Finger Nucleases", GENETICS, vol. 188, August 2011 (2011-08-01), pages 773 - 782, XP055171682, DOI: 10.1534/genetics.111.131433
CARTER, B. J., CURRENT OPINION IN BIOTECHNOLOGY, vol. 3, 1992, pages 533 - 539
CHANG ET AL., PROC. NATL. ACAD SCI USA, vol. 84, 1987, pages 4959 - 4963
CHARPENTIER ET AL., NATURE COMM.
CHEN ET AL., NUC. ACIDS RES., vol. 22, 1994, pages 2114 - 2120
CHENG ET AL., CELL RES., vol. 23, no. 10, October 2013 (2013-10-01), pages 1163 - 71
CHENG ET AL., NAT NANOTECHNOL., vol. 15, no. 4, April 2020 (2020-04-01), pages 313 - 320
CHENOKAYAMA, MOL. CELL BIOL., vol. 7, 1987, pages 2745 - 2752
CHO ET AL., GENETICS, vol. 195, no. 3, November 2013 (2013-11-01), pages 1177 - 80
CHRISTENSEN ET AL., PLANT MOL. BIOL., vol. 12, 1989, pages 619 - 632
CHRISTENSEN ET AL., PLANT MOL. BIOL., vol. 18, 1992, pages 675 - 689
CHRISTOU, P., EUPHYTICA, vol. 85, 1995, pages 13 - 27
CHRISTOU, P., THE PLANT JOURNAL, vol. 2, 1992, pages 275 - 281
CHU ET AL., GENE, vol. 13, 1981, pages 197
CHYLINSKI ET AL., RNA BIOL., vol. 10, no. 5, May 2013 (2013-05-01), pages 726 - 37
COKOL ET AL.: "Finding nuclear localization signals", EMBO REP., vol. 1, no. 5, 2000, pages 411 - 415
CRUZ-BECERRAKADONAGA, ELIFE 2020, vol. 9, 2020, pages e55780
DELEBECQUE ET AL.: "Organization of intracellular reactions with rationally designed RNA assemblies", SCIENCE, vol. 333, 2011, pages 470 - 474
DENGWOLFF, GENE, vol. 143, 1994, pages 245 - 249
DEVEREUX, J. ET AL., NUCLEIC ACIDS RESEARCH, vol. 12, no. 1, 1984, pages 387
DICARLO ET AL., NUCLEIC ACIDS RES., vol. 41, no. 7, April 2013 (2013-04-01), pages 4336 - 43
DICKINSON ET AL., NAT METHODS., vol. 10, no. 10, October 2013 (2013-10-01), pages 1028 - 63
DIJKEMA ET AL., EMBOJ, vol. 4, 1985, pages 761
DILLARD ET AL., PNAS, vol. 118, no. 52, 2021
DOBRIKOVA ET AL., PROC. NATL. ACAD. SCI., vol. 100, no. 251, 2003, pages 15125 - 151301
DUBENSKY ET AL., J. VIROL., vol. 70, 1996, pages 508 - 519
DUBENSKY ET AL., PROC. NATL. ACAD. SCI. USA, vol. 81, 1984, pages 7529 - 7533
DURAI ET AL.: "Zinc finger nucleases: custom-designed molecular scissors for genome engineering of plant and mammalian cells", NUCLEIC ACIDS RES, vol. 33, 2005, pages 5978 - 90, XP002511419, DOI: 10.1093/NAR/GKI912
DURAND ET AL., VIRUSES, vol. 3, no. 2, 2011, pages 132 - 159
E. J. AIRD ET AL.: "Increasing Cas9-mediated homology-directed repair efficiency through covalent tethering of DNA repair template.", COMMUN. BIOL., vol. 1, 2018, pages 54
EBINA ET AL., SCI REP., vol. 3, 2013, pages 2510
ELROY-STEINMOSS, PROC. NATL. ACAD. SCI. USA, vol. 87, no. 9, 1990, pages 3410 - 3414
FERKOL ET AL., FASEB J., vol. 7, 1993, pages 1081 - 1091
FERRY ET AL., CURR. PHARM. DES., vol. 17, no. 24, 2011, pages 2516 - 2527
FRALEY ET AL., PROC. NATL. ACAD. SCI. USA, vol. 76, 1979, pages 3348 - 3352
FREITAS ET AL.: "Mechanisms and Signals for the Nuclear Import of Proteins", CURRENT GENOMICS, vol. 10, no. 8, 2009, pages 550 - 7, XP055502464
FRIETAS ET AL., CUN- GENOMICS, vol. 10, 2009, pages 550 - 7
FURLER ET AL., GENE THER., vol. 8, no. 11, 2001, pages 864 - 873
GAJ ET AL.: "ZFN, TALEN, and CRISPR/Cas-based methods for genome engineering", TRENDS BIOTECHNOL, vol. 31, 2013, pages 397 - 405
GAO ET AL., BIOCHEM. BIOPHYS. RES. COMMUN., vol. 200, 1994, pages 1201 - 1206
GAOHUANG, NUC. ACIDS RES., vol. 21, 1993, pages 2867 - 2872
GARLAPATI ET AL., J BIOL. CHEM., vol. 279, no. 51, 2004, pages 3389 - 33971
GOPAL, MOL. CELL BIOL., vol. 5, 1985, pages 1188 - 1190
GORMAN ET AL., PROC. NATL. ACAD. SCI. USA, vol. 79, 1982, pages 6777
GRAHAMVAN DER EB, VIROLOGY, vol. 52, 1973, pages 456 - 467
GUIMARAES ET AL., NAT PROTOC, vol. 8, 2013, pages 1800 - 99
GURTU ET AL., BIOCHEM. BIOPHYS. RES. COMM., vol. 229, 1996, pages 295 - 298
HACKLAND ET AL., ARCH. VIROL., vol. 139, 1994, pages 1 - 22
HALEMARHAM, THE HARPER COLLINS DICTIONARY OF BIOLOGY, 1991
HARLANDWEINTRAUB, J. CELL BIOL., vol. 101, 1985, pages 1094 - 1099
HAVLICEK, MOL. THER., vol. 25, no. 2, 2017, pages 342 - 355
HERRERA ET AL.: "Illuminating endosomal escape of polymorphic lipid nanoparticles that boost mRNA delivery", BIOMATERIALS SCIENCE, 2020
HU ET AL., CELL RES., vol. 23, no. ll, November 2013 (2013-11-01), pages 1322 - 5
J. CARLSON-STEVERMER ET AL.: "Assembly of CRISPR ribonucleoproteins with biotinylated oligonucleotides via an RNA aptamer for precise gene editing", NAT. COMMUN., vol. 8, 2017, pages 1711, XP055456055, DOI: 10.1038/s41467-017-01875-9
JANG ET AL., VIROL, vol. 63, 1989, pages 1651 - 1660
JEMIELITY, J. ET AL.: "Novel 'anti-reverse' cap analogs with superior translational properties", RNA, vol. 9, 2003, pages 1108 - 1122, XP002466761, DOI: 10.1261/rna.5430403
JIANG ET AL., NUCLEIC ACIDS RES., vol. 41, no. 20, 1 November 2013 (2013-11-01), pages el88
JINEK ET AL., ELIFE, vol. 2, 2013, pages e00471
JINEK ET AL., SCIENCE, vol. 337, no. 6096, 17 August 2012 (2012-08-17), pages 816 - 21
JOHANSSON ET AL.: "RNA recognition by the MS2 phage coat protein", SEM VIROL., vol. 8, no. 3, 1997, pages 176 - 185
JONES ET AL., PLANT METHODS, vol. 1, 2005, pages 5
K. LEE ET AL.: "Synthetically modified guide RNA and donor DNA are a versatile platform for CRISPR-Cas9 engineering", ELIFE, vol. 6, 2017, pages e25312, XP055456761, DOI: 10.7554/eLife.25312
KANEDA ET AL., SCIENCE, vol. 243, 1989, pages 375 - 378
KATO ET AL., J. BIOL. CHEM., vol. 266, no. 6, 1991, pages 3361 - 3364
KAUFMAN ET AL., NUC. ACIDS RES., vol. 19, 1991, pages 4485 - 4490
KIM ET AL., PLOS ONE, vol. 6, no. 4, 2011, pages el8556
KIM ET AL., PNAS U.S.A., vol. 91, 1994, pages 883 - 887
KIM ET AL., PNAS U.S.A., vol. 93, 1996, pages 1156 - 1160
KIM ET AL.: "Preassembled zinc-finger arrays for rapid construction of ZFNs are taught", NAT. METHODS, vol. 8, 2011, pages 7
KLEIN ET AL., NATURE, vol. 327, 1987, pages 70 - 73
KOBAYASHI ET AL., BIOTECHNIQUES, vol. 21, 1996, pages 399 - 402
KOTIN, R. M., HUMAN GENE THERAPY, vol. 5, 1994, pages 793 - 801
L. KING: "The Baculovirus Expression System: A laboratory guide", 1992, SPRINGER
LARSON ET AL., NAT PROTOC., vol. 8, no. ll, November 2013 (2013-11-01), pages 2281 - 308
LAST ET AL., THEOR. APPL. GENET., vol. 81, 1991, pages 581 - 588
LEBKOWSKI ET AL., MOLEC. CELL. BIOL., vol. 8, 1988, pages 3988 - 3996
LING ET AL.: "Improving the efficiency of precise genome editing with site-specific Cas9-oligonucleotide conjugates", SCIENCE ADVANCES, vol. 6, no. 15, 2020, pages 1 - 8
LISZCZAK ET AL.: "Genomic targeting of epigenetic probes using a chemically tailored Cas9 system", PROC NATL ACAD SCI U.S.A., vol. 114, 2017, pages 681 - 686, XP055534035, DOI: 10.1073/pnas.1615723114
LIU ET AL.: "Membrane-destablizing ionizable phospholipids for organ-selective mRNA delivery and CRISPR-Cas gene editing", NAT MATER., 2021
LOIS ET AL., SCIENCE, vol. 295, 2002, pages 868 - 872
LOPEZ ET AL.: "Precise genome editing across kingdoms of life using retron-derived DNA", NATURE CHEMICAL BIOLOGY, vol. 18, no. 2022, 12 December 2021 (2021-12-12), pages 199 - 206, XP037679572, DOI: 10.1038/s41589-021-00927-y
LUNDSTROM, TRENDS BIOTECHNOL., vol. 21, no. 3, 2003, pages 117 - 122
MA ET AL., BIOMED RES INT., vol. 2013, 2013, pages 270805
MAGIN ET AL., VIROLOGY, vol. 274, 2000, pages 11 - 16
MAKAROVA ET AL., NAT REV MICROBIOL., vol. 13, no. 11, November 2015 (2015-11-01), pages 722 - 36
MALI ET AL.: "Cas9 transcriptional activators for target specificity screening and paired nickases for cooperative genome engineering", NAT. BIOTECHNOL., vol. 31, 2013, pages 833 - 838, XP055693153, DOI: 10.1038/nbt.2675
MARATEA ET AL., GENE, vol. 40, 1985, pages 39 - 46
MARTIN ET AL., HELV. CHIM. ACTA, vol. 78, 1995, pages 486 - 504
MARTIN ET AL., MOL. CELL ENDOCRINOL., vol. 212, 2003, pages 51 - 61
MARTINEAU ET AL., MOL. CELL. BIOL., vol. 24, no. 17, 2004, pages 7622 - 7635
MATZ ET AL., NATURE BIOTECHNOL., vol. 17, 1999, pages 969 - 973
MCCORMICK ET AL., PLANT CELL REPORTS, vol. 5, 1986, pages 81 - 84
MCELROY ET AL., PLANT CELL, vol. 2, 1990, pages 163 - 171
MESTERE ET AL., UG/ABI: ''A HIGHLY DIVERSE FAMILY OF PROKARYOTIC REVERSE TRANSCRIPTASES ASSOCIATED WITH DEFENSE FUNCTIONS
MESTRE ET AL., NUCLEIC ACIDS RESEARCH, vol. 48, 16 December 2020 (2020-12-16), pages 12632 - 12647
MESTRE ET AL.: "Systematic Prediction of Genes Functionally Associated with Bacterial Retrons and Classification of The Encoded Tripartite Systems", NUCLEIC ACIDS RESEARCH, vol. 48, 16 December 2020 (2020-12-16), pages 12632 - 12647
MICHAEL ET AL., J. BIOL. CHEM., vol. 268, 1993, pages 6866 - 6869
MILLER, A. D., HUMAN GENE THERAPY, vol. 1, 1990, pages 5 - 14
MILLERROSMAN, BIOTECHNIQUES, vol. 7, 1989, pages 980 - 990
MOEDE ET AL., FEBS LETT, vol. 461, 1999, pages 229 - 34
MOSCOU ET AL., SCIENCE, vol. 326, 2009, pages 1509 - 1512
MOSSER ET AL., BIOTECHNIQUES, vol. 22, 1997
MURPHY ET AL., PNAS USA, vol. 83, 1986, pages 8258 - 1441
MUZYCZKA, N., CURRENT TOPICS IN MICROBIOL AND IMMUNOL., vol. 158, 1992, pages 97 - 129
N. SAVIC ET AL.: "Covalent linkage of the DNA repair template to the CRISPR-Cas9 nuclease enhances homology-directed repair", ELIFE, vol. 7, 2018, pages e33761, XP055642567, DOI: 10.7554/eLife.33761
NAKAYAMA ET AL., GENESIS, vol. 51, no. 12, December 2013 (2013-12-01), pages 835 - 43
NEHLS ET AL., SCIENCE, vol. 272, 1996, pages 886 - 889
NICOLAU ET AL., METHODS ENZYMOL., vol. 149, 1987, pages 157 - 176
NICOLAUSENE, BIOCHIM. BIOPHYS. ACTA, vol. 721, 1982, pages 185 - 190
NUCLEIC ACIDS RES., vol. 35, pages e81
ODELL ET AL., NATURE, vol. 313, 1985, pages 810 - 812
PAN ET AL., SCI REP., vol. 6, 2016, pages 35794
PATEL ET AL.: "Naturally-occuring cholesterol analogues in lipid nanoparticles induce polymorphic shape and enhance intracellular delivery of mRNA", NATURE COMMUNICATIONS, 2020
PATTANAYAK ET AL., NAT BIOTECHNOL., vol. 31, no. 9, September 2013 (2013-09-01), pages 839 - 43
PEDERSEN ET AL., BIOCHEM. J., vol. 363, 2002, pages 37 - 44
PERALES ET AL., PROC. NATL. ACAD. SCI. USA, vol. 91, no. 9, 1994, pages 4086 - 4090
PERRI ET AL., J. VIROL., vol. 77, 2003, pages 10394 - 10403
PORTA ET AL., MOL. BIOTECH., vol. 5, 1996, pages 209 - 221
PROC NATL. ACAD. SCI. USA, vol. 84, 1987, pages 8463 - 8467
PROVOST ET AL., GENESIS, vol. 45, no. 10, 2007, pages 625 - 629
QI ET AL., CELL, vol. 152, no. 5, 28 February 2013 (2013-02-28), pages 1173 - 83
RAKOCZY-TROJANOWSKA, M., CELL MOL BIOL LETT., vol. 7, 2002, pages 849 - 858
RAMESH ET AL., NUCL. ACID RES., vol. 24, 1996, pages 2697 - 2700
REMINGTONA. R. GENNARO: "The Science and Practice of Pharmacy", 2006, LIPPINCOTT, WILLIAMS & WILKINS
RIPPE ET AL., MOL. CELL BIOL., vol. 10, 1990, pages 689 - 695
RIVERA ET AL., PHYSICS OF LIFE REVIEWS, vol. 9, 2012, pages 308 - 345
SANDER ET AL.: "Selection-free zinc-finger-nuclease engineering by context-dependent assembly (CoDA) is taught", NAT. METHODS, vol. 8, 2011, pages 67 - 69, XP055752406, DOI: 10.1038/nmeth.1542
SCARPA ET AL., VIROLOGY, vol. 180, 1991, pages 849 - 852
SCHMOHL ET AL., CURR OPIN CHEM BIOL, vol. 22, 2014, pages 122 - 8
SEMPLE, NATURE BIOTECHNOLOGY, vol. 28, 2010, pages 172 - 176
SHANER ET AL., NAT. METHODS, vol. 2, 2005, pages 905 - 909
SHELLINGSMITH, GENE THERAPY, vol. 1, 1994, pages 165 - 169
SHMAKOV ET AL., MOL CELL., vol. 60, no. 3, 5 November 2015 (2015-11-05), pages 385 - 97
SMITHWATERMAN, ADV. APPL. MATH., vol. 2, 1981, pages 482 - 489
STEIN ET AL., MOL. CELL. BIOL., vol. 18, no. 6, 1998, pages 3112 - 3119
STUDIERMOFFATT, J. MOL. BIOL., vol. 189, 1986, pages 113 - 130
TAYLOR ET AL., NUCLEIC ACIDS RESEARCH, vol. 40, no. Wl, 2012, pages W 110 - W 116
TENCHOV ET AL.: "Lipid Nanoparticles - From Liposomes to mRNA Vaccine Delivery, a Landscape of Diversity and Advancement", ACS NANO, vol. 15, 2021, pages 16982 - 17015, XP093009618, DOI: 10.1021/acsnano.1c04996
TINLAND ET AL., PROC. NATL. ACAD. SCI. U.S.A., vol. 89, 1992, pages 7442 - 46
TRICHAS ET AL., BMC BIOL., vol. 6, 2008, pages 40
TSAI ET AL., NAT BIOTECHNOL., vol. 32, no. 6, 2014, pages 569 - 576
TUR-KASPA ET AL., MOL. CELL. BIOL., vol. 6, 1986, pages 716 - 718
TZFIRA ET AL., TRENDS IN GENETICS, vol. 20, 2004, pages 375 - 383
UPADHYAY ET AL., G3 (BETHESDA)., vol. 3, no. 12, 9 December 2013 (2013-12-09), pages 2233 - 8
VARSHNEY ET AL.: "DNA-guided genome editing using structure-guided endonucleases", GENOME BIOLOGY, vol. 17, no. 1, 2016, pages 187, XP055419357, DOI: 10.1186/s13059-016-1055-4
VELTEN ET AL., EMBO J., vol. 3, 1984, pages 2723 - 2730
VERMAGREENBURG, GENES DEV., vol. 30, no. 10, 2016, pages 1138 - 1154
WAGNER ET AL., PROC. NATL. ACAD. SCI. USA, vol. 89, 1992, pages 6099 - 6103
WALSH ET AL., PROC NATL ACAD SCI USA., vol. 110, no. 39, 24 September 2013 (2013-09-24), pages 15514 - 5
WALSH ET AL.: "Synthesis, Characterization and Evaluation of Ionizable Lysine-Based Lipids for siRNA Delivery", BIOCONJUG CHEM., 2013
WALTHER ET AL., DRUGS, vol. 60, no. 2, 2000, pages 249 - 271
WAMOCK ET AL., METHODS MOL. BIOL., vol. 737, 2011, pages 1 - 25
WANG ET AL., CELL, vol. 153, no. 4, 9 May 2013 (2013-05-09), pages 910 - 8
WUWU, ADV. DRUG DELIVERY REV., vol. 12, 1993, pages 159 - 167
WUWU, BIOCHEMISTRY, vol. 27, 1988, pages 887 - 892
WUWU, J. BIOL. CHEM., vol. 262, 1987, pages 4429 - 4432
XIE ET AL., MOL PLANT., 9 October 2013 (2013-10-09)
YAMANO ET AL.: "Crystal structure of Cpfl in complex with guide RNA and target DNA", CELL, vol. 165, 2016, pages 949 - 962
YANG ET AL., CELL, vol. 154, no. 6, 12 September 2013 (2013-09-12), pages 1370 - 9
YAO ET AL., JOURNAL OF EXPERIMENTAL BOTANY, vol. 57, 2006, pages 3737 - 3746
YOKOE ET AL., NATURE BIOTECHNOLOGY, vol. 14, 1996, pages 1252 - 1256
ZALATAN ET AL.: "Engineering complex synthetic transcriptional programs with CRISPR RNA scaffolds", CELL, vol. 160, 2015, pages 339 - 350, XP055278878, DOI: 10.1016/j.cell.2014.11.052
ZETSCHE ET AL., CELL, vol. 163, no. 3, 22 October 2015 (2015-10-22), pages 759 - 71
ZHANG ET AL., NATURE BIOTECHNOLOGY, vol. 29, 2011, pages 149 - 153
ZHANGMADDEN, GENOME RES., vol. 7, 1997, pages 649 - 656
ZHOU ET AL., J. EXP. MED., vol. 179, 1994, pages 1867 - 1875
ZUPANZAMBRYSKI, PLANT PHYSIOLOGY, vol. 107, 1995, pages 1041 - 1047

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2024044673A1 (en) * 2022-08-24 2024-02-29 The J. David Gladstone Institutes, A Testamentary Trust Established Under The Will Of J. David Gladstone Dual cut retron editors for genomic insertions and deletions
WO2024044723A1 (en) * 2022-08-25 2024-02-29 Renagade Therapeutics Management Inc. Engineered retrons and methods of use

Also Published As

Publication number Publication date
WO2023141602A3 (en) 2023-11-02

Similar Documents

Publication Publication Date Title
AU2021204395B2 (en) Delivery, use and therapeutic applications of the CRISPR-Cas systems and compositions for genome editing
AU2019283878B2 (en) Delivery, engineering and optimization of systems, methods and compositions for sequence manipulation and therapeutic applications
US11447774B2 (en) Nuclease systems for genetic engineering
US11920150B2 (en) Engineered muscle targeting compositions
AU2014281030B2 (en) Delivery, engineering and optimization of systems, methods and compositions for targeting and modeling diseases and disorders of post mitotic cells
CN105793425B (en) Delivery, use and therapeutic applications of CRISPR-CAS systems and compositions for targeting disorders and diseases using viral components
US11666641B2 (en) CRISPR nuclease
WO2023141602A2 (en) Engineered retrons and methods of use
US11946077B2 (en) OMNI-59, 61, 67, 76, 79, 80, 81, and 82 CRISPR nucleases
WO2023081756A1 (en) Precise genome editing using retrons
US11866728B2 (en) Engineered retrons and methods of use
US20230087228A1 (en) Novel type iv and type i crispr-cas systems and methods of use thereof
US20220380758A1 (en) Type i-b crispr-associated transposase systems
WO2024044723A1 (en) Engineered retrons and methods of use
US20230303989A1 (en) Omni-103 crispr nuclease
US20240131121A1 (en) Novel omni-50 crispr nuclease
WO2024020346A2 (en) Gene editing components, systems, and methods of use
CA3228373A1 (en) Novel omni 115, 124, 127, 144-149, 159, 218, 237, 248, 251-253 and 259 crispr nucleases
CA3206576A1 (en) Omni 103 crispr nuclease
IL307855A (en) NOVEL OMNI 117, 140, 150-158, 160-165, 167-177, 180-188, 191-198, 200, 201, 203, 205-209, 211-217, 219, 220, 222, 223, 226, 227, 229, 231-236, 238-245, 247, 250, 254, 256, 257, 260 and 262 CRISPR NUCLEASES