CA2626262A1 - Rationally-designed meganucleases with altered sequence specificity and dna-binding affinity - Google Patents

Rationally-designed meganucleases with altered sequence specificity and dna-binding affinity Download PDF

Info

Publication number
CA2626262A1
CA2626262A1 CA002626262A CA2626262A CA2626262A1 CA 2626262 A1 CA2626262 A1 CA 2626262A1 CA 002626262 A CA002626262 A CA 002626262A CA 2626262 A CA2626262 A CA 2626262A CA 2626262 A1 CA2626262 A1 CA 2626262A1
Authority
CA
Canada
Prior art keywords
group
meganuclease
sense strand
modification
sequence
Prior art date
Legal status (The legal status 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 status listed.)
Granted
Application number
CA002626262A
Other languages
French (fr)
Other versions
CA2626262C (en
Inventor
Homme W. Hellinga
James Jefferson Smith
Derek Jantz
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Duke University
Original Assignee
Individual
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
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=37963286&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=CA2626262(A1) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Application filed by Individual filed Critical Individual
Priority to CA2891996A priority Critical patent/CA2891996A1/en
Publication of CA2626262A1 publication Critical patent/CA2626262A1/en
Application granted granted Critical
Publication of CA2626262C publication Critical patent/CA2626262C/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

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
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/43Enzymes; Proenzymes; Derivatives thereof
    • A61K38/46Hydrolases (3)
    • A61K38/465Hydrolases (3) acting on ester bonds (3.1), e.g. lipases, ribonucleases
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • 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
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/82Vectors or expression systems specially adapted for eukaryotic hosts for plant cells, e.g. plant artificial chromosomes (PACs)
    • C12N15/8201Methods for introducing genetic material into plant cells, e.g. DNA, RNA, stable or transient incorporation, tissue culture methods adapted for transformation
    • C12N15/8213Targeted insertion of genes into the plant genome by homologous recombination
    • 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
    • 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
    • C12N15/905Stable introduction of foreign DNA into chromosome using homologous recombination in yeast
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy

Abstract

Rationally-designed LAGLIDADG meganucleases and methods of making such meganucleases are provided. In addition, methods are provided for using the meganucleases to generate recombinant cells and organisms having a desired DNA
sequence inserted into a limited number of loci within the genome, as well as methods of gene therapy, for treatment of pathogenic infections, and for in vitro applications in diagnostics and research.

Description

RATIONALLY-DESIGNED MEGANUCLEASES
WITH ALTERED SEQUENCE SPECIFICITY AND DNA-BINDING AFFINITY
RELATED APPLICATION
[0001] This application claims benefit of priority to U.S. Provisional Patent Application No. 60/727,512, filed October 18, 2005, the entire disclosure of which is hereby incorporated by reference.

GOVERNMENT SUPPORT
[0002] The invention was supported in part by grants 2R01-GM-0498712, 5F32-GM072322 and 5 DPI OD000122 from the National Institute of General Medical Sciences of National Institutes of Health of the United States of America.
Therefore, the U.S. govermnent may have certain rights in the invention.

FIELD OF THE INVENTION
[0003) The invention relates to the field of molecular biology and recoinbinant nucleic acid technology. In particular, the invention relates to rationally-designed, non-naturally-occurring meganucleases with altered DNA recognition sequence specificity and/or altered affinity. The invention also relates to methods of producing such meganucleases, and methods of producing recombinant nucleic acids and organisms using such meganucleases.

BACKGROUND OF THE INVENTION
[0004] Genoine engineering requires the ability to insert, delete, substitute and otherwise manipulate specific genetic sequences within a genome, and has numerous therapeutic and biotechnological applications. The development of effective means for genoine modification remains a major goal in gene therapy, agrotechnology, and synthetic biology (Porteus et al. (2005), Nat. Biotechnol. 23: 967-73; Tzfira et al. (2005), Trends Biotechnol. 23: 567-9; McDaniel et al. (2005), Curr. Opin. Biotechnol.
16: 476-83). A coinmon method for inserting or modifying a DNA sequence involves introducing a transgenic DNA sequence flanked by sequences homologous to the genomic target and selecting or screening for a successful homologous recoinbination event.
Recoinbination with the transgenic DNA occurs rarely but can be stimulated by a double-stranded break in the genomic DNA at the target site. Numerous methods have been employed to create DNA double-stranded breaks, including irradiation and chemical treatments.
Although these methods efficiently stimulate recombination, the double-stranded breaks are randomly dispersed in the genome, which can be highly mutagenic and toxic. At present, the inability to target gene modifications to unique sites within a chromosoinal background is a major iinpediment to successful genome engineering.
[0005] One approach to achieving this goal is stimulating homologous recombination at a double-stranded break in a target locus using a nuclease with specificity for a sequence that is sufficiently large to be present at only a single site within the genome (see, e.g., Porteus et al. (2005), Nat. Biotechnol. 23: 967-73).
The effectiveness of this strategy has been demonstrated in a variety of organisms using chimeric fusions between an engineered zinc finger DNA-binding domain and the non-specific nuclease domain of the Fokl restriction enzyme (Porteus (2006), Mol 77hef 13:
438-46; Wright et al. (2005), Plant J. 44: 693-705; Ui-nov et al. (2005), Nature 435: 646-51). Although these artificial zinc finger nucleases stimulate site-specific recombination, they retain residual non-specific cleavage activity resulting from under-regulation of the nuclease domain and frequently cleave at unintended sites (Smith et al.
(2000), Nucleic Acids Res. 28: 3361-9). Such unintended cleavage can cause mutations and toxicity in the treated organism (Porteus et al. (2005), Nat. Biotechnol. 23: 967-73).
[0006] A group of naturally-occurring nucleases which recognize 15-40 base-pair cleavage sites commonly found in the genomes of plants and fungi may provide a less toxic genome engineering alternative. Such "meganucleases" or "homing endonucleases"
are frequently associated with parasitic DNA elements, such as group 1 self-splicing introns and inteins. They naturally promote homologous recombination or gene insertion at specific locations in the host genome by producing a double-stranded break in the chromosome, which recruits the cellular DNA-repair machinery (Stoddard (2006), Q.
Rev. Bioplays. 38: 49-95). Meganucleases are commonly grouped into four families: the LAGLIDADG family, the GIY-YIG family, the His-Cys box family and the HNH
family.
These families are characterized by structural motifs, which affect catalytic activity and recognition sequence. For instance, ineinbers of the LAGLIDADG family are characterized by having either one or two copies of the conserved LAGLIDADG
motif (see Chevalier et al. (2001), Nucleic Acids Res. 29(18): 3757-3774). The LAGLIDADG
meganucleases with a single copy of the LAGLIDADG motif fonn homodimers, whereas members with two copies of the LAGLIDADG motif are found as monomers.
Similarly, the GIY-YIG family meinbers have a GIY-YIG module, which is 70-100 residues long and includes four or five conserved sequence motifs with four invariant residues, two of which are required for activity (see Van Roey et al. (2002), Nature Struct.
Biol. 9: 806-811). The His-Cys box meganucleases are characterized by a highly conserved series of histidines and cysteines over a region encoinpassing several hundred amino acid residues (see Chevalier et al. (2001), Nucleic Acids Res. 29(18): 3757-3774). In the case of the NHN family, the members are defined by motifs containing two pairs of conserved histidines surrounded by asparagine residues (see Chevalier et al. (2001), Nucleic Acids Res. 29(18): 3757-3774). The four families of meganucleases are widely separated from one another with respect to conserved structural elements and, consequently, DNA
recognition sequence specificity and catalytic activity.
100071 Natural meganucleases, primarily from the LAGLIDADG family, have been used to effectively promote site-specific genome inodification in plants, yeast, Drosophila, mammalian cells and mice, but this approach has been limited to the modification of either homologous genes that conserve the meganuclease recognition sequence (Monnat et al. (1999),Biochem. Biophys. Res. Commun. 255: 88-93) or to pre-engineered genomes into which a recognition sequence has been introduced (Rouet et al.
(1994), Mol. Cell. Biol. 14: 8096-106; Chilton et al. (2003), Plant Physiol.
133: 956-65;
Puchta et al. (1996), Proc. Natl. Acad. Sci. USA 93: 5055-60; Rong et al.
(2002), Genes Dev. 16: 1568-81; Gouble et al. (2006), J. Gene Med. 8(5):616-622).
[0008] Systematic implementation of nuclease-stimulated gene modification requires the use of engineered enzyines with customized specificities to target DNA
breaks to existing sites in a genome and, therefore, there has been great interest in adapting meganucleases to promote gene modifications at medically or biotechnologically relevant sites (Porteus et al. (2005), Nat. Biotechnol. 23:
967-73;

Sussman et al. (2004), J. Mol. Biol. 342: 31-41; Epinat et al. (2003), Nucleic Acids Res.
31: 2952-62).
[0009] The meganuclease I-Crel from Chlamydomonas reinhardtii is a member of the LAGLIDADG family which recognizes and cuts a 22 base-pair recognition sequence in the chloroplast chroinosome, and which presents an attractive target for meganuclease redesign. The wild-type enzyme is a homodimer in which each monomer makes direct contacts with 9 base pairs in the full-length recognition sequence. Genetic selection techniques have been used to identify mutations in I-CreI that alter base preference at a single position in this recognition sequence (Sussman et al. (2004), J. Mol.
Biol. 342: 31-41; Chames et al. (2005), NucleicAcids Res. 33: e178; Seligman et al. (2002), Nucleic Acids Res. 30: 3870-9) or, inore recently, at three positions in the recognition sequence (Arnould et al. (2006), J. Mol. Biol. 355: 443-58). The I-CreI protein-DNA
interface contains nine amino acids that contact the DNA bases directly and at least an additional five positions that can form potential contacts in modified interfaces. The size of this interface imposes a combinatorial complexity that is unlikely to be sampled adequately in sequence libraries constructed to select for enzymes with drastically altered cleavage sites.
[00101 There remains a need for nucleases that will facilitate precise modification of a genome. In addition, there remains a need for techniques for generating nucleases with pre-detennined, rationally-designed recognition sequences that will allow manipulation of genetic sequences at specific genetic loci and for techniques utilizing such nucleases to genetically engineer organisms with precise sequence modifications.
SUMMARY OF THE INVENTION

[0011] The present invention is based, in part, upon the identification and characterization of specific amino acid residues in the LAGLIDADG family of meganucleases that make contacts with DNA bases and the DNA backbone when the meganucleases associate with a double-stranded DNA recognition sequence, and thereby affect the specificity and activity of the enzyines. This discovery has been used, as described in detail below, to identify amino acid substitutions which can alter the recognition sequence specificity and/or DNA-binding affinity of the meganucleases, and to rationally design and develop meganucleases that can recognize a desired DNA
sequence that naturally-occurring meganucleases do not recognize. The invention also provides methods that use such ineganucleases to produce recombinant nucleic acids and organisms by utilizing the meganucleases to cause recoinbination of a desired genetic sequence at a limited number of loci within the genome of the organism, for gene therapy, for treatment of pathogenic infections, and for in vitro applications in diagnostics and research.
[0012] Thus, in some embodiments, the invention provides recoinbinant ineganucleases having altered specificity for at least one recognition sequence half-site relative to a wild-type I-Crel meganuclease, in which the meganuclease includes a polypeptide having at least 85% sequence similarity to residues 2-153 of the wild-type I-Crel meganuclease of SEQ ID NO: 1, but in which the recombinant meganuclease has specificity for a recognition sequence half-site which differs by at least one base pair from a half-site within an I-Crel meganuclease recognition sequence selected from SEQ ID
NO: 2, SEQ ID NO: 3, SEQ ID NO: 4 and SEQ ID NO: 5, and in which the recombinant meganuclease includes at least one modification listed in Table 1 which is not an excluded modification found in the prior art.
[0013] In other embodiments, the invention provides recombinant meganucleases having altered specificity for at least one recognition sequence half-site relative to a wild-type I-Msol meganuclease, in which the meganuclease includes a polypeptide having at least 85% sequence similarity to residues 6-160 of the I-Msol meganuclease of SEQ ID
NO: 6, but in which the recombinant meganuclease has specificity for a recognition ' sequence half-site which differs by at least one base pair from a half-site within an I-Msol meganuclease recognition sequence selected from SEQ ID NO: 7 and SEQ ID NO: 8, and in which the recombinant meganuclease includes at least one modification listed in Table 2 which is not an excluded modification found in the prior art.
[0014] In other einbodiinents, the invention provides recoinbinant meganucleases having altered specificity for a recognition sequence relative to a wild-type I-SceI
meganuclease, in which the meganuclease includes a polypeptide having at least 85%
sequence similarity to residues 3-186 of the I-Scel meganuclease of SEQ ID NO:
9, but in which the recombinant meganuclease has specificity for a recognition sequence which differs by at least one base pair from an I-SceI ineganuclease recognition sequence of SEQ ID NO: 10 and SEQ ID NO: 11, and in which the recombinant meganuclease includes at least one modification listed in Table 3 which is not an excluded modification found in the prior art.
[0015] In other einbodiments, the invention provides recoinbinant meganucleases having altered specificity for at least one recognition sequence half-site relative to a wild-type I-CeuI meganuclease, in which the meganuclease includes a polypeptide having at least 85% sequence siinilarity to residues 5-211 of the I-Ceul meganuclease of SEQ ID
NO: 12, but in which the recoinbinant meganuclease has specificity for a recognition sequence half-site which differs by at least one base pair from a half-site within an I-CeuI
meganuclease recognition sequence selected from SEQ ID NO: 13 and SEQ ID NO:
14, and in which the recombinant meganuclease includes at least one modification listed in Table 4 which is not an excluded modification found in the prior art.
[0016] The meganucleases of the invention can include one, two, three or more of the modifications which have been disclosed herein in order to affect the sequence specificity of the recombinant meganucleases at one, two, three or more positions within the recognition sequence. The meganucleases can include only the novel modifications disclosed herein, or can include the novel modifications disclosed herein in combination with modifications found in the prior art. Specifically excluded, however, are recombinant meganucleases comprising only the modifications of the prior art.
[0017] In another aspect, the invention provides for recombinant meganucleases with altered binding affinity for double-stranded DNA which is not sequence-specific.
This is accoinplished by modifications of the meganuclease residues which make contacts with the backbone of the double-stranded DNA recognition sequence. The modifications can increase or decrease the binding affinity and, consequently, can increase or decrease the overall activity of the enzyine. Moreover, increases/decreases in binding and activity have been found to causes decreases/increases in sequence specificity. Thus, the invention provides a means for altering sequence specificity generally by altering DNA-binding affinity.
[0018] Thus, in some embodiments, the invention provides for recoinbinant meganucleases having altered binding affinity for double-stranded DNA relative to a wild-type I-Crel meganuclease, in which the meganuclease includes a polypeptide having at least 85% sequence similarity to residues 2-153 of the I-Crel meganuclease of SEQ ID
NO: 1, and in which the DNA-binding affinity has been either (1) increased by at least one modification corresponding to a substitution selected from (a) substitution of E80, D137, 181, Ll 12, P29, V64 or Y66 with H, N, Q, S, T, K or R, or (b) substitution of T46, T140 or T143 with K or R; or, conversely, (2) decreased by at least one modification corresponding to a substitution selected from (a) substitution of K34, K48, R5 1, K82, K116 or K139 with H, N, Q, S, T, D or E, or (b) substitution of 18 1, L112, P29, V64, Y66, T46, T140 or T143 with D or E.
[0019] In other embodiments, the invention provides for recombinant meganucleases having altered binding affinity for double-stranded DNA relative to a wild-type I-Msol meganuclease, in which the meganuclease includes a polypeptide having at least 85% sequence similarity to residues 6-160 of the I-Msol meganuclease of SEQ ID NO: 6, and in which the DNA-binding affinity has been either (1) increased by at least one modification corresponding to a substitution selected from(a) substitution of E147,185, G86 or Y118 with H, N, Q, S, T, K or R, or (b) substitution of Q41, N70, S87, T88, H89, Q122, Q139, S150 orN152 with K or R; or, conversely, (2) decreased by at least one modification corresponding to a substitution selected from (a) substitution of K36, R51, K123, K143 or R144 with H, N, Q, S, T, D or E, or (b) substitution of 185, G86, Y118, Q41, N70, S87, T88, H89, Q122, Q139, S150 or N152 with D or E.
[0020] In other embodiments, the invention provides for recombinant meganucleases having altered binding affinity for double-stranded DNA relative to a wild-type I-SceI meganuclease, in which the meganuclease includes a polypeptide having at least 85% sequence similarity to residues 3-186 of the I-SceI meganuclease of SEQ ID
NO: 9, and in which the DNA-binding affinity has been either (1) increased by at least one modification corresponding to a substitution selected from (a) substitution of D201, L19, L80, L92, Y151, Y188,1191, Y199 or Y222 with H, N, Q, S, T, K or R, or (b) substitution ofN15, N17, S81, H84, N94, N120, T156, N157, S159, N163, Q165, S166, N194 or S202 with K or R; or, conversely, (2) decreased by at least one modification corresponding to a substitution selected from (a) substitution of K20, K23, K63, K122, K148, K153, K190, K193, K195 or K223 with H, N, Q, S, T, D or E, or (b) substitution
-7-of L19, L80, L92, Y151, Y188, I191, Y199, Y222, N15, N17, S81, H84, N94, N120, T156, N157, S159, N163, Q165, S166, N194 or S202 with D or E.
[0021] In other einbodiments, the invention provides for recombinant meganucleases having altered binding affinity for double-stranded DNA relative to a wild-type I-CeuI meganuclease, in which the meganuclease includes a polypeptide having at least 85% sequence similarity to residues 5-211 of the I-Ceul meganuclease of SEQ ID
NO: 12, and in which the DNA-binding affinity has been either (1) increased by at least one modification corresponding to a substitution selected from (a) substitution of D25 or D 128 with H, N, Q, S, T, K or R, or (b) substitution of S68, N70, H94, S 117, N 120, N 129 or H172 with K or R; or, conversely, (2) decreased by at least one modification corresponding to a substitution selected from (a) substitution of K2 1, K28, K3 1, R112, R114 or R130 with H, N, Q, S, T, D or E, or (b) substitution of S68, N70, H94, S117, N 120, N 129 or H 172 with D or E.
[0022] The meganucleases of the invention can include one, two, three or more of the modifications of backbone contact residues which have been disclosed herein in order to affect DNA-binding affinity. In addition, these modifications affecting DNA-binding affinity can be combined with one or more of the novel modifications of the base contact residues described above which alter the sequence specificity of the recombinant meganucleases at specific positions within the recognition sequence, or with the prior art modifications described above, or with a combination of the novel modifications and prior art modifications. In particular, by combining backbone contact modifications and base contact modifications, recoinbinant meganucleases can be rationally-designed with desired specificity and activity. For example, increases in DNA-binding affinity can be designed which may offset losses in affinity resulting from designed changes to base contact residues, or decreases in affinity can be designed which may also decrease sequence specificity and broaden the set of recognition sequences for an enzyme.
[0023] In another aspect, the invention provides for rationally-designed meganuclease monomers with altered affinity for homo- or heterodimer formation. The affinity for dimer forination can be measured with the saine monomer (i.e., homodimer formation) or with a different monomer (i.e., heterodimer formation) such as a reference wild-type meganuclease. These recombinant ineganucleases have modifications to the
-8-
9 PCT/US2006/040919 amino acid residues which are present at the protein-protein interface between monomers in a meganuclease dimer. The modifications can be used to promote heterodimer formation and create meganucleases with non-palindromic recognition sequences.
[0024] Thus, in some einbodiinents, the invention provides recoinbinant meganuclease monomers having altered affinity for dimer forination with a reference meganuclease monomer, in which the recombinant monomer includes a polypeptide having at least 85% sequence similarity to residues 2-153 of the I-CreI
meganuclease of SEQ ID NO: 1, but in which affinity for dimer formation has been altered by at least one modification corresponding to a substitution selected from (a) substitution of K7, K57 or K96 with D or E, or (b) substitution of ES or E61 with K or R. Based upon such recoinbinant monomers, the invention also provides recombinant meganuclease heterodimers including (1) a first polypeptide having at least 85% sequence similarity to residues 2-153 of the I-CreI meganuclease of SEQ ID NO: 1, but in which affinity for dimer formation has been altered by at least one modification corresponding to a substitution selected from (a) substitution of K7, K57 or K96 with D or E, and (2) a second polypeptide having at least 85% sequence similarity to residues 2-153 of the I-CreI meganuclease of SEQ ID NO: 1, but i which affinity for dimer formation has been altered by at least one modification corresponding to a substitution selected from (b) substitution of E8 or E61 with K or R.
[0025] In other embodiments, the invention provides recombinant meganuclease monomers having altered affinity for dimer formation with a reference meganuclease monomer, in which the recombinant monomer includes a polypeptide having at least 85%
sequence similarity to residues 6-160 of the I-Msol meganuclease of SEQ ID NO:
6, but in which affinity for dimer formation has been altered by at least one modification corresponding to a substitution selected from (a) substitution of R302 with D
or E, or (b) substitution of D20, E11 or Q64 with K or R. Based upon such recoinbinant monomers, the invention also provides recoinbinant ineganuclease heterodimers including (1) a first polypeptide having at least 85% sequence similarity to residues 6-160 of the I-Msol meganuclease of SEQ ID NO: 6, but in which affinity for dimer formation has been altered by at least one modification corresponding to a substitution selected from (a) substitution of R302 with D or E, and (2) a second polypeptide having at least 85%

sequence similarity to residues 6-160 of the I-MsoI meganuclease of SEQ ID NO:
6, but in which affinity for diiner forination has been altered by at least one modification corresponding to a substitution selected from (b) substitution of D20, E11 or Q64 with K
or R.
[0026] In other einbodiinents, the invention provides recombinant meganuclease monomers having altered affinity for dimer formation with a reference meganuclease monomer, in which the recoinbinant monomer includes a polypeptide having at least 85%
sequence similarity to residues 5-211 of the I-CeuI meganuclease of SEQ ID NO:
12, but in which affinity for diiner forination has been altered by at least one modification corresponding to a substitution selected from (a) substitution of R93 with D
or E, or (b) substitution of E152 with K or R. Based upon such recombinant monomers, the invention also provides recombinant meganuclease heterodimers including (1) a first polypeptide having at least 85% sequence similarity to residues 5-211 of the I-Ceul meganuclease of SEQ ID NO: 12, but in which affinity for dimer formation has been altered by at least one modification corresponding to a substitution selected from (a) substitution of R93 with D
or E, and (2) a second polypeptide having at least 85% sequence similarity to residues 5-211 of the I-Ceul meganuclease of SEQ ID NO: 12, but in which affinity for dimer formation has been altered by at least one modification corresponding to a substitution selected from (b) substitution of E152 with K or R.
[0027] The recoinbinant meganuclease monomers or heterodimers with altered affinity for dimer formation can also include one, two, three or more of the modifications of base contact residues described above; one, two, three or more of the modifications of baclcbone contact residues described above; or combinations of both. Thus, for example, the base contacts of a monomer can be modified to alter sequence specificity, the backbone contacts of a monomer can be modified to alter DNA-binding affinity, and the protein-protein interface can be modified to affect diiner formation. Such a recombinant monomer can be combined with a similarly modified monomer to produce a rationally-designed meganuclease heterodimer with desired sequence specificity and activity.
100281 In another aspect, the invention provides for various methods of use for the rationally-designed meganucleases described and enabled herein. These methods include producing genetically-modified cells and organisms, treating diseases by gene therapy,
-10-treating pathogen infections, and using the recoinbinant meganucleases for in vitro applications for diagnostics and research.
[0029] Thus, in one aspect, the invention provides methods for producing a genetically-modified eukaryotic cell including an exogenous sequence of interest inserted in a chromosome, by transfecting the cell with (i) a first nucleic acid sequence encoding a meganuclease of the invention, and (ii) a second nucleic acid sequence including said sequence of interest, wherein the meganuclease produces a cleavage site in the chromosome and the sequence of interest is inserted into the chromosome at the cleavage site either by homologous recoinbination or non-homologous end-joining.
[0030] Alternatively, in another aspect, the invention provides methods for producing a genetically-modified eukaryotic cell including an exogenous sequence of interest inserted in a chromosome, by introducing a meganuclease protein of the invention into the cell, and transfecting the cell with a nucleic acid including the sequence of interest, wherein the meganuclease produces a cleavage site in the chromosome and the sequence of interest is inserted into the chromosome at the cleavage site either by homologous recombination or non-hoinologous end-joining.
[0031] In another aspect, the invention provides methods for producing a genetically-modified eukaryotic cell by disrupting a target sequence in a chromosome, by transfecting the cell with a nucleic acid encoding a meganuclease of the invention, wherein the meganuclease produces a cleavage site in the chromosome and the target sequence is disrupted by non-homologous end-joining at the cleavage site.
100321 In another aspect, the invention provides methods of producing a genetically-modified organism by producing a genetically-modified eukaryotic cell according to the methods described above, and growing the genetically-modified eukaryotic cell to produce the genetically-modified organism. In these embodiments, the eukaryotic cell can be selected from a gamete, a zygote, a blastocyst cell, an embryonic stem cell, and a protoplast cell.
[0033] In another aspect, the invention provides methods for treating a disease by gene therapy in a eukaryote, by transfecting at least one cell of the eukaryote with one or more nucleic acids including (i) a first nucleic acid sequence encoding a meganuclease of the invention, and (ii) a second nucleic acid sequence including a sequence of interest,
-11-wherein the meganuclease produces a cleavage site in the chromosome and the sequence of interest is inserted into the chromosome by homologous recombination or non-homologous end joining, and insertion of the sequence of interest provides gene therapy for the disease.
[0034] Alternatively, in another aspect, the invention provides methods for treating a disease by gene therapy in a eukaryote, by introducing a meganuclease protein of the invention into at least one cell of the eukaiyote, and transfecting the cell with a nucleic acid including a sequence of interest, wherein the meganuclease produces a cleavage site in the chromosome and the sequence of interest is inserted into the chromosome at the cleavage site by homologous recombination or non-homologous end-joining, and insertion of the sequence of interest provides gene therapy for the disease.
[0035] In another aspect, the invention provides methods for treating a disease by gene therapy in a eukaryote by disrupting a target sequence in a chromosome of the eukaryotic, by transfecting at least one cell of the eukaryote with a nucleic acid encoding a meganuclease of the invention, wherein the meganuclease produces a cleavage site in the chromosome and the target sequence is disrupted by non-homologous end-joining at the cleavage site, wherein disruption of the target sequence provides the gene therapy for the disease.
[0036] In another aspect, the invention provides methods for treating a viral or prokaryotic pathogen infection in a eukaryotic host by disrupting a target sequence in a genome of the pathogen, by transfecting at least one infected cell of the host with a nucleic acid encoding a meganuclease of the invention, wherein the meganuclease produces a cleavage site in the genome and the target sequence is disrupted by either (1) non-homologous end-joining at the cleavage site or (2) by homologous recombination with a second nucleic acid, and wherein disruption of the target sequence provides treatment for the infection.
[0037] More generally, in another aspect, the invention provides methods for rationally-designing recombinant meganucleases having altered specificity for at least one base position of a recognition sequence, by (1) deterinining at least a portion of a three-diinensional structure of a reference ineganuclease-DNA complex; (2) identifying amino acid residues forining a base contact surface at the base position; (3) determining a -
-12-distance between a(3-carbon of at least a first residue of the contact surface and at least a first base at the base position; and (4) identifying an amino acid substitution to promote the desired change by either (a) for a first residue which is < 6 A from the first base, selecting a substitution froin Group 1 and/or Group 2 which is a member of an appropriate one of Group G, Group C, Group T or Group A; or (b) for a first residue which is > 6 A from said first base, selecting a substitution from Group 2 and/or Group 3 which is a member of an appropriate one of Group G, Group C, Group T or Group A, where each of the Groups is defined herein. This method may be repeated for additional contact residues for the same base, and for contact residues for the other base at the same position, as well as for additional positions.
[0038] In addition, in another general aspect, the invention provides methods for rationally-designing a recoinbinant meganuclease having increased DNA-binding affinity, by (1) determining at least a portion of a three-dimensional structure of a reference ineganuclease-DNA complex; (2) identifying amino acid contact residues forming a backbone contact surface; and (3) identifying an amino acid substitution to increase the DNA-binding affinity by (a) for a contact residue having a negatively-charged or hydrophobic side chain, selecting a substitution having an uncharged/polar or positively-charged side chain; or (b) for a contact residue having an uncharged/polar side chain, selecting a substitution having a positively-charged side chain. Conversely, the invention also provides methods for rationally-designing a recombinant meganuclease having decreased DNA-binding affinity, by (1) determining at least a portion of a three-dimensional structure of a reference meganuclease-DNA complex; (2) identifying amino acid contact residues forining a backbone contact surface; (3) identifying an amino acid substitution to decrease the DNA-binding affinity by (a) for a contact residue having a positively-charged side chain, selecting a substitution having an uncharged/polar or negatively-charged side chain; or (b) for a contact residue having an hydrophobic or uncharged/polar side chain, selecting a substitution having a negatively-charged side chain.
[0039] These and other aspects and einbodiments of the invention will be apparent to one of ordinary slcill in the art based upon the following detailed description of the invention.
-13-BRIEF DESCRIPTION OF THE FIGURES

[0040] Figure 1(A) illustrates the interactions between the I-Crel hoinodiiner and its naturally-occurring double-stranded recognition sequence, based upon ciystallographic data. This scheinatic representation depicts the recognition sequence (SEQ ID
NO: 2 and SEQ ID NO: 3), shown as unwound for illustration purposes only, bound by the homodimer, shown as two ovals. The bases of each DNA half-site are numbered -1 through -9, and the amino acid residues of I-CreI which foi-in the recognition surface are indicated by one-letter amino acid designations and numbers indicating residue position.
Solid black lines: hydrogen bonds to DNA bases. Dashed lines: amino acid positions that form additional contacts in enzyme designs but do not contact the DNA in the wild-type coinplex. Arrows: residues that interact with the DNA backbone and influence cleavage activity.
[0041] Figure 1(B) illustrates the wild-type contacts between the A-T base pair at position -4 of the cleavage half-site on the right side of Figure 1(A).
Specifically, the residue Q26 is shown to interact with the A base. Residue 177 is in proximity to the base pair but not specifically interacting.
[0042] Figure 1(C) illustrates the interactions between a rationally-designed variant of the I-CreI meganuclease in which residue 177 has been modified to E77. As a result of this change, a G-C base pair is preferred at position -4. The interaction between Q26 and the G base is mediated by a water molecule, as has been observed crystallographically for the cleavage half-site on the left side of Figure 1(A).
[0043] Figure 1(D) illustrates the interactions between a rationally-designed variant of the I-Crel meganuclease in which residue Q26 has been modified to E26 and residue 177 has been modified to R77. As a result of this change, a C-G base pair is preferred at position -4.
[0044] Figure 1(E) illustrates the interactions between a rationally-designed variant of the I-CreI meganuclease in which residue Q26 has been modified to A26 and residue 177 has been modified to Q77. As a result of this change, a T-A base pair is preferred at position -4.
-14-100451 Figure 2(A) shows a comparison of one recognition sequence for each of the wild type I-Cre1 meganuclease (WT) and 11 rationally-designed meganuclease heterodimers of the invention. Bases that are conserved relative to the WT
recognition sequence are shaded. The 9 bp half-sites are bolded. WT: wild-type (SEQ ID NO:
4);
CF: AF508 allele of the human CFTR gene responsible for most cases of cystic fibrosis (SEQ ID NO: 25); MYD: the human DM kinase gene associated with myotonic dystrophy (SEQ ID NO: 27); CCR: the human CCR5 gene (a major HIV co-receptor) (SEQ ID NO: 26); ACH: the human FGFR3 gene correlated with achondroplasia (SEQ
ID NO: 23); TAT: the HIV-1 TAT/REV gene (SEQ ID NO: 15); HSV: the HSV-1 UL36 gene (SEQ ID NO: 28); LAM: the bacteriophage k p05 gene (SEQ ID NO: 22); POX:
the Variola (smallpox) viius gp009 gene (SEQ ID NO: 30); URA: the Saccharomyces cerevisiae URA3 gene (SEQ ID NO: 36); GLA: the Arabidopsis thali.ana GL2 gene (SEQ ID NO: 32); BRP: the Arabidopsis thaliaiia BP-1 gene (SEQ ID NO: 33).
100461 Figure 2(B) illustrates the results of incubation of each of wild-type I-CreI
(WT) and 11 rationally-designed meganuclease heterodimers with plasmids harboring the recognition sites for all 12 enzymes for 6 hours at 37 C. Percent cleavage is indicated in each box.
[0047] Figure 3 illustrates cleavage patterns of wild-type and rationally-designed I-CreI homodimers. (A) wild type I-CreI. (B) I-Crel K116D. (C-L) rationally-designed meganucleases of the invention. Enzymes were incubated with a set of plasmids harboring palindromes of the intended cleavage half-site the 27 corresponding single-base pair variations. Bar graphs show fractional cleavage (F) in 4 hours at 37 C.
Black bars:
expected cleavage patterns based on Table 1. Gray bars: DNA sites that deviate from expected cleavage patterns. White circles indicate bases in the intended recognition site.
Also shown are cleavage time-courses over two hours. The open circle time-course plots in C and L correspond to cleavage by the CCR1 and BRP2 enzyines lacking the mutation. The cleavage sites correspond to the 5' (left column) and 3' (right column) half-sites for the heterodiineric enzymes described in Fig. 2(A).
-15-DETAILED DESCRIPTION OF THE INVENTION

1.1 Introduction [0048] The present invention is based, in part, upon the identification and characterization of specific amino acids in the LAGLIDADG family of meganucleases that make specific contacts with DNA bases and non-specific contacts with the DNA
baclcbone when the ineganucleases associate with a double-stranded DNA
recognition sequence, and which thereby affect the recognition sequence specificity and DNA-binding affinity of the enzymes. This discovery has been used, as described in detail below, to identify amino acid substitutions in the meganucleases that can alter the specificity and/or affinity of the enzymes, and to rationally design and develop meganucleases that can recognize a desired DNA sequence that naturally-occurring meganucleases do not recognize, and/or that have increased or decreased specificity and/or affinity relative to the naturally-occurring meganucleases.
Furthermore, because DNA-binding affinity affects enzyine activity as well as sequence-specificity, the invention provides rationally-designed meganucleases with altered activity relative to naturally-occurring meganucleases. In addition, the invention provides rationally-designed meganucleases in which residues at the interface between the monomers associated to form a dimer have been modified in order to promote heterodimer formation. Finally, the invention provides uses for the rationally-designed meganucleases in the production of recombinant cells and organisms, as well as in gene therapy, anti-pathogen, anti-cancer, and in vitro applications, as disclosed herein.
[0049] As a general matter, the invention provides methods for generating rationally-designed LAGLIDADG meganucleases containing altered amino acid residues at sites within the meganuclease that are responsible for (1) sequence-specific binding to individual bases in the double-stranded DNA recognition sequence, or (2) non-sequence-specific binding to the phosphodiester baclcbone of a double-stranded DNA
molecule.
Because enzyine activity is correlated to DNA-binding affinity, however, altering the amino acids involved in binding to the DNA recognition sequence can alter not only the specificity of the meganuclease through specific base pair interactions, but also the activity of the meganuclease by increasing or decreasing overall binding affinity for the
-16-double-stranded DNA. Similarly, altering the amino acids involved in binding to the DNA backbone can alter not only the activity of the enzyme, but also the degree of specificity or degeneracy of binding to the recognition sequence by increasing or decreasing overall binding affinity for the double-stranded DNA.
[0050] As described in detail below, the methods of rationally-designing meganucleases include the identification of the amino acids responsible for DNA
recognition/binding, and the application of a series of rules for selecting appropriate amino acid changes. With respect to meganuclease sequence specificity, the rules include both steric considerations relating to the distances in a meganuclease-DNA
complex between the amino acid side chains of the meganuclease and the bases in the sense and anti-sense strands of the DNA, and considerations relating to the non-covalent chemical interactions between functional groups of the amino acid side chains and the desired DNA base at the relevant position.

[0051] Finally, a majority of natural meganucleases that bind DNA as homodimers recognize pseudo- or coinpletely palindromic recognition sequences.
Because lengthy palindromes are expected to be rare, the likelihood of encountering a palindromic sequence at a genomic site of interest is exceedingly low.
Consequently, if these enzymes are to be redesigned to recognize genomic sites of interest, it is necessary to design two enzylne monomers recognizing different half-sites that can heterodimerize to cleave the non-palindromic hybrid recognition sequence. Therefore, in some aspects, the invention provides rationally-designed meganucleases in which monomers differing by at least one amino acid position are dimerized to form heterodimers. In some cases, both monomers are rationally-designed to form a heterodimer which recognizes a non-palindromic recognition sequence. A mixture of two different monomers can result in up to three active forms of meganuclease dimer: the two homodimers and the heterodimer.
In addition or alternatively, in some cases, amino acid residues are altered at the interfaces at which monomers can interact to form dimers, in order to increase or decrease the likelihood of forination of homodimers or heterodimers.
[0052] Thus, in one aspect, the invention provide methods for rationally designing LAGLIDADG meganucleases containing amino acid changes that alter the specificity and/or activity of the enzymes. In another aspect, the invention provides the rationally-
-17-designed meganucleases resulting from these methods. In another aspect, the invention provides methods that use such rationally-designed meganucleases to produce recoinbinant nucleic acids and organisms in which a desired DNA sequence or genetic locus within the genome of an organism is modified by the insertion, deletion, substitution or other manipulation of DNA sequences. In another aspect, the invention provides methods for reducing the survival of pathogens or cancer cells using rationally-designed meganucleases which have pathogen-specific or cancer-specific recognition sequences.

1.2 References and Definitions [0053] The patent and scientific literature referred to herein establishes knowledge that is available to those of skill in the art. The issued U.S.
patents, allowed applications, published foreign applications, and references, including GenBank database sequences, that are cited herein are hereby incorporated by reference to the same extent as if each was specifically and individually indicated to be incorporated by reference.
[0054] As used herein, the terin "meganuclease" refers to an endonuclease that binds double-stranded DNA at a recognition sequence that is greater than 12 base pairs.
Naturally-occurring meganucleases can be monomeric (e.g., I-Scel) or dimeric (e.g., I-CreI). The terin meganuclease, as used herein, can be used to refer to monomeric meganucleases, dimeric meganucleases, or to the monomers which associate to form a dimeric meganuclease. The term "homing endonuclease" is synonymous with the term "meganuclease."
[0055] As used herein, the term "LAGLIDADG meganuclease" refers either to meganucleases including a single LAGLIDADG motif, which are naturally diineric, or to meganucleases including two LAGLIDADG motifs, which are naturally monomeric.
The term "mono-LAGLIDADG meganuclease" is used herein to refer to meganucleases including a single LAGLIDADG motif, and the tenn "di-LAGLIDADG meganuclease"
is used herein to refer to meganucleases including two LAGLIDADG motifs, when it is necessary to distinguish between the two. Each of the two structural domains of a di-LAGLIDADG ineganuclease which includes a LAGLIDADG motif can be referred to as a LAGLIDADG subunit.
-18-[0056] As used herein, the term "rationally-designed" means non-naturally occurring and/or genetically engineered. The rationally-designed meganucleases of the invention differ from wild-type or naturally-occurring meganucleases in their amino acid sequence or primary structure, and may also differ in their secondary, tertiary or quatemary structure. In addition, the rationally-designed meganucleases of the invention also differ from wild-type or naturally-occurring meganucleases in recognition sequence-specificity and/or activity.
[0057] As used herein, with respect to a protein, the terin "recombinant"
means having an altered amino acid sequence as a result of the application of genetic engineering techniques to nucleic acids which encode the protein, and cells or organisms which express the protein. With respect to a nucleic acid, the term "recombinant" means having an altered nucleic acid sequence as a result of the application of genetic engineering techniques. Genetic engineering techniques include, but are not limited to, PCR and DNA cloning technologies; transfection, transformation and other gene transfer technologies; homologous recombination; site-directed mutagenesis; and gene fusion. In accordance with this definition, a protein having an ainino acid sequence identical to a naturally-occurring protein, but produced by cloning and expression in a heterologous host, is not considered recombinant.
[0058] As used herein with respect to recombinant proteins, the term "modification" means any insertion, deletion or substitution of an amino acid residue in the recombinant sequence relative to a reference sequence (e.g., a wild-type).
[0059] As used herein, the term "genetically-modified" refers to a cell or organism in which, or in an ancestor of which, a genoinic DNA sequence has been deliberately modified by recombinant technology. As used herein, the term "genetically-modified" encompasses the term "transgenic."
[0060] As used herein, the term "wild-type" refers to any naturally-occurring form of a meganuclease. The tenn "wild-type" is not intended to mean the most common allelic variant of the enzyme in nature but, rather, any allelic variant found in nature.
Wild-type meganucleases are distinguished from recombinant or non-naturally-occurring meganucleases.
-19-[0061] As used herein, the term "recognition sequence half-site" or simply "half site" means a nucleic acid sequence in a double-stranded DNA molecule which is recognized by a monomer of a mono-LAGLIDADG meganuclease or by one LAGLIDADG subunit of a di-LAGLIDADG meganuclease.
[0062] As used herein, the tenn "recognition sequence" refers to a pair of half-sites which is bound and cleaved by either a mono-LAGLIDADG meganuclease dimer or a di-LAGLIDADG meganuclease monomer. The two half-sites may or may not be separated by base pairs that are not specifically recognized by the enzyme. In the cases of I-Crel, I-Msol and I-CeuI, the recognition sequence half-site of each monomer spans 9 base pairs, and the two half-sites are separated by four base pairs which are not recognized specifically but which constitute the actual cleavage site (which has a 4 base pair overhang). Thus, the combined recognition sequences of the I-CreI, I-MsoI
and I-Ceul meganuclease dimers normally span 22 base pairs, including two 9 base pair half-sites flanking a 4 base pair cleavage site. The base pairs of each half-site are designated -9 through -1, with the -9 position being most distal from the cleavage site and the -1 position being adjacent to the 4 central base pairs, which are designated NI-N4. The strand of each half-site which is oriented 5' to 3' in the direction from -9 to -1 (i.e., towards the cleavage site), is designated the "sense" strand and the opposite strand is designated the "antisense strand", although neither strand may encode protein.
Thus, the "sense" strand of one half-site is the antisense strand of the other half-site. See, for example, Figure 1(A). In the case of the I-SceI meganuclease, which is a di-LAGLIDADG meganuclease monomer, the recognition sequence is an approximately bp non-palindromic sequence, and there are no central base pairs which are not specifically recognized. By convention, one of the two strands is referred to as the "sense" strand and the other the "antisense" strand, although neitlier strand may encode protein.

[0063] As used herein, the term "specificity" means the ability of a meganuclease to recognize and cleave double-stranded DNA molecules only at a particular sequence of base pairs referred to as the recognition sequence, or only at a particular set of recognition sequences. The set of recognition sequences will share certain conserved positions or sequence motifs, but may be degenerate at one or more positions. A highly-specific
-20-meganuclease is capable of cleaving only one or a very few recognition sequences.
Specificity can be determined in a cleavage assay as described in Example 1.
As used herein, a meganuclease has "altered" specificity if it binds to and cleaves a recognition sequence which is not bound to and cleaved by a reference meganuclease (e.g., a wild-type) or if the rate of cleavage of a recognition sequence is increased or decreased by a statistically significant (p < 0.05) ainount relative to a reference meganuclease.
[0064] As used herein, the term "degeneracy" ineans the opposite of "specificity."
A highly-degenerate meganuclease is capable of cleaving a large nuinber of divergent recognition sequences. A meganuclease can have sequence degeneracy at a single position within a half-site or at multiple, even all, positions within a half-site. Such sequence degeneracy can result from (i) the inability of any amino acid in the DNA-binding domain of a meganuclease to make a specific contact with any base at one or more positions in the recognition sequence, (ii) the ability of one or more amino acids in the DNA-binding domain of a meganuclease to make specific contacts with more than one base at one or more positions in the recognition sequence, and/or (iii) sufficient non-specific DNA binding affinity for activity. A"completely" degenerate position can be occupied by any of the four bases and can be designated with an "N" in a half-site. A
"partially" degenerate position can be occupied by two or three of the four bases (e.g., either purine (Pu), either pyrimidine (Py), or not G).
[0065] As used herein with respect to meganucleases, the term "DNA-binding affinity" or "binding affinity" means the tendency of a meganuclease to non-covalently associate with a reference DNA molecule (e.g., a recognition sequence or an arbitrary sequence). Binding affinity is measured by a dissociation constant, KD (e.g., the KD of I-CreI for the WT recognition sequence is approximately 0.1 nM). As used herein, a meganuclease has "altered" binding affinity if the KD of the recombinant meganuclease for a reference recognition sequence is increased or decreased by a statistically significant (p < 0.05) amount relative to a reference meganuclease.
[0066] As used herein with respect to meganuclease monomers, the term "affinity for dimer forination" means the tendency of a meganuclease monomer to non-covalently associate with a reference meganuclease monomer. The affinity for dimer formation can be measured with the same monomer (i.e., homodiiner formation) or with a different
-21-monomer (i.e., heterodimer forination) such as a reference wild-type meganuclease.
Binding affinity is measured by a dissociation constant, KD. As used herein, a meganuclease has "altered" affinity for dimer formation if the KD of the recombinant meganuclease monomer for a reference meganuclease monomer is increased or decreased by a statistically significant (p < 0.05) amount relative to a reference meganuclease monomer.
[0067] As used herein, the term "palindromic" refers to a recognition sequence consisting of inverted repeats of identical half-sites. In this case, however, the palindromic sequence need not be palindromic with respect to the four central base pairs, which are not contacted by the enzyme. In the case of dimeric meganucleases, palindromic DNA sequences are recognized by homodimers in which the two monomers make contacts with identical half-sites.
[0068] As used herein, the term "pseudo-palindromic" refers to a recognition sequence consisting of inverted repeats of non-identical or imperfectly palindromic half-sites. In this case, the pseudo-palindromic sequence not only need not be palindromic with respect to the four central base pairs, but also can deviate from a palindromic sequence between the two half-sites. Pseudo-palindromic DNA sequences are typical of the natural DNA sites recognized by wild-type homodimeric meganucleases in which two identical enzyme monomers make contacts with different half-sites.
[0069] As used herein, the term "non-palindromic" refers to a recognition sequence composed of two unrelated half-sites of a meganuclease. In this case, the non-palindromic sequence need not be palindromic with respect to either the four central base pairs or the two monomer half-sites. Non-palindromic DNA sequences are recognized by either di-LAGLIDADG meganucleases, highly degenerate mono-LAGLIDADG
meganucleases (e.g., I-CeuI) or by heterodimers of mono-LAGLIDADG meganuclease monomers that recognize non-identical half-sites.
[0070] As used herein, the terin "activity" refers to the rate at which a meganuclease of the invention cleaves a particular recognition sequence. Such activity is a measurable enzyinatic reaction, involving the hydrolysis of phosphodiester bonds of double-stranded DNA. The activity of a meganuclease acting on a particular DNA
substrate is affected by the affinity or avidity of the meganuclease for that particular DNA
-22-substrate which is, in turn, affected by both sequence-specific and non-sequence-specific interactions with the DNA.
[0071] As used herein, the terin "homologous recombination" refers to the natural, cellular process in which a double-stranded DNA-break is repaired using a homologous DNA sequence as the repair teinplate (see, e.g. Cahill et al.
(2006), Front.
Biosci. 11:1958-1976). The homologous DNA sequence may be an endogenous chroinosoinal sequence or an exogenous nucleic acid that was delivered to the cell. Thus, in some embodiinents, a rationally-designed meganuclease is used to cleave a recognition sequence within a target sequence and an exogenous nucleic acid with homology to or substantial sequence similarity with the target sequence is delivered into the cell and used as a teinplate for repair by homologous recoinbination. The DNA sequence of the exogenous nucleic acid, which may differ significantly from the target sequence, is thereby incorporated into the chroinosomal sequence. The process of homologous recombination occurs primarily in eukaryotic organisms. The term "homology" is used herein as equivalent to "sequence similarity" and is not intended to require identity by descent or phylogenetic relatedness.
[0072] As used herein, the term "non-homologous end-joining" refers to the natural, cellular process in which a double-stranded DNA-break is repaired by the direct joining of two non-homologous DNA segments (see, e.g. Cahill et al. (2006), Front.
Biosci. 11:1958-1976). DNA repair by non-homologous end-joining is error-prone and frequently results in the unteinplated addition or deletion of DNA sequences at the site of repair. Thus, in certain einbodiments, a rationally-designed meganuclease can be used to produce a double-stranded break at a meganuclease recognition sequence within a target sequence to disrupt a gene (e.g., by introducing base insertions, base deletions, or fraineshift mutations) by non-homologous end-joining. In other embodiments, an exogenous nucleic acid lacking homology to or substantial sequence similarity with the target sequence may be captured at the site of a meganuclease-stiinulated double-stranded DNA break by non-homologous end-joining (see, e.g. Salomon, et al. (1998), EMBO J.
17:6086-6095). The process of non-homologous end-joining occurs in both eukaryotes and prokaryotes such as bacteria.
- 23 -[0073] As used herein, the tenn "sequence of interest" means any nucleic acid sequence, whether it codes for a protein, RNA, or regulatory element (e.g., an enhancer, silencer, or promoter sequence), that can be inserted into a genome or used to replace a genomic DNA sequence using a meganuclease protein. Sequences of interest can have heterologous DNA sequences that allow for tagging a protein or RNA that is expressed from the sequence of interest. For instance, a protein can be tagged with tags including, but not limited to, an epitope (e.g., c-myc, FLAG) or other ligand (e.g., poly-His).
Furthermore, a sequence of interest can encode a fusion protein, according to techniques lcnown in the art (see, e.g., Ausubel et al., Current Protocols in Molecular Biology, Wiley 1999). In some embodiments, the sequence of interest is flanked by a DNA
sequence that is recognized by the recoinbinant meganuclease for cleavage. Thus, the flanking sequences are cleaved allowing for proper insertion of the sequence of interest into genomic recognition sequences cleaved by the recombinant meganuclease. In some embodiments, the entire sequence of interest is homologous to or has substantial sequence similarity with the a target sequence in the genome such that homologous recombination effectively replaces the target sequence with the sequence of interest. In other embodiments, the sequence of interest is flanked by DNA sequences with homology to or substantial sequence similarity with the target sequence such that homologous recombination inserts the sequence of interest within the genome at the locus of the target sequence. In some embodiments, the sequence of interest is substantially identical to the target sequence except for mutations or other modifications in the meganuclease recognition sequence such that the meganuclease can not cleave the target sequence after it has been modified by the sequence of interest.
[00741 As used herein with respect to both amino acid sequences and nucleic acid sequences, the terms "percentage similarity" and "sequence similarity" refer to a measure of the degree of similarity of two sequences based upon an alignment of the sequences which maximizes similarity between aligned amino acid residues or nucleotides, and which is a function of the number of identical or similar residues or nucleotides, the nuinber of total residues or nucleotides, and the presence and length of gaps in the sequence alignment. A variety of algorithms and computer programs are available for deterinining sequence similarity using standard parameters. As used herein, sequence
-24-similarity is measured using the BLASTp program for amino acid sequences and the BLASTn program for nucleic acid sequences, both of which are available through the National Center for Bioteclulology Information (www.ncbi.nlm.nih.gov/), and are described in, for exainple, Altschul et al. (1990), J. Mol. Biol. 215:403 -410; Gish and States (1993), Nature Genet. 3:266-272; Madden et al. (1996), Meth. Enzymol.
266:131-141; Altschul et al. (1997), Nucleic Acids Res. 25:33 89-3402); Zhang et al.
(2000), J.
Connput. Biol. 7(1-2):203-14.. As used herein, percent similarity of two amino acid sequences is the score based upon the following parameters for the BLASTp algorithm:
word size = 3; gap opening penalty = -11; gap extension penalty =-1; and scoring matrix = BLOSUM62. As used herein, percent similarity of two nucleic acid sequences is the score based upon the following parameters for the BLASTn algorithm: word size = 11;
gap opening penalty =-5; gap extension penalty = -2; match reward = 1; and mismatch penalty = -3.
[0075] As used herein with respect to modifications of two proteins or amino acid sequences, the tenn "corresponding to" is used to indicate that a specified modification in the first protein is a substitution of the same amino acid residue as in the modification in the second protein, and that the amino acid position of the modification in the first proteins corresponds to or aligns with the amino acid position of the modification in the second protein when the two proteins are subjected to standard sequence alignments (e.g., using the BLASTp program). Thus, the modification of residue "X" to amino acid "A" in the first protein will correspond to the modification of residue "Y" to amino acid "A" in the second protein if residues X and Y correspond to each other in a sequence alignment, and despite the fact that X and Y may be different numbers.
[00761 As used herein, the recitation of a numerical range for a variable is intended to convey that the invention may be practiced with the variable equal to any of the values within that range. Thus, for a variable which is inherently discrete, the variable can be equal to any integer value within the numerical range, including the end-points of the range. Similarly, for a variable which is inherently continuous, the var-iable can be equal to any real value within the numerical range, including the end-points of the range.
As an example, and without limitation, a variable which is described as having values between 0 and 2 can take the values 0, 1 or 2 if the variable is inherently discrete, and can
-25-take the values 0.0, 0.1, 0.01, 0.001, or any other real values > 0 and _ 2 if the variable is inherently continuous.
[0077] As used herein, unless specifically indicated otherwise, the word "or"
is used in the inclusive sense of "and/or" and not the exclusive sense of "either/or."

2.1 Rationally-Designed Meganucleases with Altered Sequence-Sbecificity [0078] In one aspect of the invention, methods for rationally designing recombinant LAGLIDADG family meganucleases are provided. In this aspect, recombinant meganucleases are rationally-designed by first predicting amino acid substitutions that can alter base preference at each position in the half-site. These substitutions can be experimentally validated individually or in coinbinations to produce meganucleases with the desired cleavage specificity.
[0079] In accordance with the invention, amino acid substitutions that can cause a desired change in base preference are predicted by determining the amino acid side chains of a reference meganuclease (e.g., a wild-type meganuclease, or a rion-naturally-occurring reference meganuclease) that are able to participate in making contacts with the nucleic acid bases of the meganuclease's DNA recognition sequence and the DNA
phosphodiester backbone, and the spatial and chemical nature of those contacts. These amino acids include but are not limited to side chains involved in contacting the reference DNA half-site. Generally, this detennination requires having knowledge of the structure of the coinplex between the ineganuclease and its double-stranded DNA recognition sequence, or knowledge of the structure of a highly similar complex (e.g., between the same meganuclease and an alternative DNA recognition sequence, or between an allelic or phylogenetic variant of the meganuclease and its DNA recognition sequence).
[0080] Three-dimensional structures, as described by atomic coordinates data, of a polypeptide or complex of two or more polypeptides can be obtained in several ways.
For example, protein structure detenninations can be made using techniques including, but not limited to, X-ray crystallography, NMR, and mass spectrometry. Another approach is to analyze databases of existing structural co-ordinates for the meganuclease of interest or a related meganuclease. Such structural data is often available from
-26-databases in the forin of three-diinensional coordinates. Often this data is accessible through online databases (e.g., the RCSB Protein Data Bank at www.rcsb.org/pdb).
[0081] Structural infonnation can be obtained experimentally by analyzing the diffraction patterns of, for example, X-rays or electrons, created by regular two- or three-dimensional arrays (e.g., crystals) of proteins or protein complexes.
Computational methods are used to transforin the diffraction data into three-dimensional atomic co-ordinates in space. For example, the field of X-ray crystallography has been used to generate three-dimensional stiuctural information on many protein-DNA
complexes, including meganucleases (see, e.g., Chevalier et al. (2001), Nucleic Acids Res. 29(18):
3757-3774).
[0082] Nuclear Magnetic Resonance (NMR) also has been used to determine inter-atomic distances of molecules in solution. Multi-dimensional NMR methods combined with coinputational methods have succeeded in determining the atomic co-ordinates of polypeptides of increasing size (see, e.g., Tzakos et al. (2006), Annu. Rev.
Biop/zys. Bionzol. Struct. 35:19-42.).
[0083] Alternatively, computational modeling can be used by applying algorithms based on the lcnown primary structures and, when available, secondary, tertiary and/or quaternary stiuctures of the protein/DNA, as well as the known physiochemical nature of the ainino acid side chains, nucleic acid bases, and bond interactions. Such methods can optionally include iterative approaches, or experimentally-derived constraints. An exainple of such computational software is the CNS program described in Adams et al.
(1999), Acta Crystallogr. D. Biol. Cnystallogr. 55 (Pt 1): 181-90. A variety of other coinputational programs have been developed that predict the spatial arrangement of amino acids in a protein structure and predict the interaction of the ainino acid side chains of the protein with various target molecules (see, e.g., U.S. Pat. No.
6,988,041).
[0084] Thus, in some einbodiments of the invention, computational models are used to identify specific amino acid residues that specifically interact with DNA nucleic acid bases and/or facilitate non-specific phosphodiester backbone interactions. For instance, computer models of the totality of the potential meganuclease-DNA
interaction can be produced using a suitable software prograin, including, but not limited to, MOLSCRIPTTM 2.0 (Avatar Software AB, Stockholm, Sweden), the graphical display
-27-program O(Jones et. al. (1991), Acta Crystallography, A47: 110), the graphical display program GRASPTM (Nicholls et al. (1991), PROTEINS, Structure, Function and Genetics 11(4): 281ff), or the graphical display program INSIGHTTM (TSI, Inc., Shoreview, MN).
Coinputer hardware suitable for producing, viewing and manipulating three-dimensional sti-uctural representations of protein-DNA complexes are coinmercially available and well known in the art (e.g., Silicon Graphics Workstation, Silicon Graphics, Inc., Mountainview, CA).
[0085] Specifically, interactions between a meganuclease and its double-stranded DNA recognition sequences can be resolved using methods known in the art. For exainple, a representation, or model, of the three dimensional stiucture of a multi-component complex structure, for which a ciystal has been produced, can be determined using techniques which include molecular replacement or SIR/MIR
(single/multiple isomorphous replacement) (see, e.g., Brunger (1997), Meth. Enzym. 276: 558-580;
Navaza and Saludjian (1997), Meth. Enzynz. 276: 581-594; Tong and Rossmann (1997), Meth. Enzym. 276: 594-611; and Bentley (1997), Meth. Enzyni. 276: 611-619) and can be performed using a software program, such as AMoRe/Mosflin (Navaza (1994), Acta Cjyst. A50: 157-163; CCP4 (1994), Acta Cryst. D50: 760-763) or XPLOR (see, Brunger et al. (1992), X-PLOR Version 3 1 A System for X-ray Crystallography and NMR, Yale University Press, New Haven, CT).
[0086] The deteimination of protein structure and potential meganuclease-DNA
interaction allows for rational choices concerning the amino acids that can be changed to affect enzyine activity and specificity. Decisions are based on several factors regarding amino acid side chain interactions with a particular base or DNA
phosphodiester backbone. Chemical interactions used to determine appropriate amino acid substitutions include, but are not limited to, van der Waals forces, steric hindrance, ionic bonding, hydrogen bonding, and hydrophobic interactions. Ainino acid substitutions can be selected which either favor or disfavor specific interactions of the meganuclease with a particular base in a potential recognition sequence half-site in order to increase or decrease specificity for that sequence and, to some degree, overall binding affinity and activity. In addition, amino acid substitutions can be selected which either increase or decrease binding affinity for the phosphodiester backbone of double-stranded DNA in
-28-order to increase or decrease overall activity and, to some degree, to decrease or increase specificity.
[0087] Thus, in specific einbodiments, a three-diinensional structure of a meganuclease-DNA complex is determined and a "contact surface" is defined for each base-pair in a DNA recognition sequence half-site. In some embodiments, the contact surface comprises those ainino acids in the enzyine with (3-carbons less than 9.0 A from a major groove hydrogen-bond donor or acceptor on either base in the pair, and with side chains oriented toward the DNA, irrespective of whether the residues make base contacts in the wild-type ineganuclease-DNA complex. In other embodiments, residues can be excluded if the residues do not make contact in the wild-type meganuclease-DNA
complex, or residues can be included or excluded at the discretion of the designer to alter the nuinber or identity of the residues considered. In one example, as described below, for base positions -2, -7, -8, and -9 of the wild-type I-CreI half-site, the contact surfaces were limited to the amino acid positions that actually interact in the wild-type enzyine-DNA complex. For positions -1, -3, -4, -5, and -6, however, the contact surfaces were defined to contain additional amino acid positions that are not involved in wild-type contacts but which could potentially contact a base if substituted with a different amino acid.
100881 It should be noted that, although a recognition sequence half-site is typically represented with respect to only one strand of DNA, meganucleases bind in the major groove of double-stranded DNA, and make contact with nucleic acid bases on both strands. In addition, the designations of "sense" and "antisense" strands are completely arbitraiy with respect to meganuclease binding and recognition. Sequence specificity at a position can be achieved either through interactions with one member of a base pair, or by a combination of interactions with both members of a base-pair. Thus, for exainple, in order to favor the presence of an A/T base pair at position X, where the A
base is on the "sense" strand and the T base is on the "antisense" strand, residues are selected which are sufficiently close to contact the sense strand at position X and which favor the presence of an A, and/or residues are selected which are sufficiently close to contact the antisense strand at position X and which favor the presence of a T. In accordance with the
-29-invention, a residue is considered sufficiently close if the (3-carbon of the residue is within 9A of the closest atom of the relevant base.
[0089] Thus, for example, an amino acid with a(3-carbon within 9A of the DNA
sense strand but greater than 9A from the antisense strand is considered for potential interactions with only the sense strand. Similarly, an amino acid with a(3-carbon within 9A of the DNA antisense strand but greater than 9A from the sense strand is considered for potential interactions with only the antisense strand. Amino acids with P-carbons that are within 9A of both DNA strands are considered for potential interactions with either strand.
[0090] For each contact surface, potential amino acid substitutions are selected based on their predicted ability to interact favorably with one or more of the four DNA
bases. The selection process is based upon two primary criteria: (i) the size of the amino acid side chains, which will affect their steric interactions with different nucleic acid bases, and (ii) the chemical nature of the amino acid side chains, which will affect their electrostatic and bonding interactions with the different nucleic acid bases.
[0091] With respect to the size of side chains, amino acids with shorter and/or smaller side chains can be selected if an amino acid 0-carbon in a contact surface is <6 A
from a base, and amino acids with longer and/or larger side chains can be selected if an amino acid (3-carbon in a contact surface is >6 A from a base. Amino acids with side chains that are interinediate in size can be selected if an amino acid (3-carbon in a contact surface is 5-8 A from a base.
[0092] The amino acids with relatively shorter and smaller side chains can be assigned to Group 1, including glycine (G), alanine (A), serine (S), threonine (T), cysteine (C), valine (V), leucine (L), isoleucine (I), aspartate (D), asparagine (N) and proline (P).
Proline, however, is expected to be used less frequently because of its relative inflexibility. In addition, glycine is expected to be used less frequently because it introduces unwanted flexibility in the peptide baclcbone and its very small size reduces the likelihood of effective contacts when it replaces a larger residue. On the other hand, glycine can be used in some instances for promoting a degenerate position. The amino acids with side chains of relatively intermediate length and size can be assigned to Group 2, including lysine (K), methionine (M), arginine (R), glutamate (E) and glutamine (Q).
-30-The amino acids with relatively longer and/or larger side chains can be assigned to Group 3, including lysine (K), methionine (M), arginine (R), histidine (H), phenylalanine (F), tyrosine (Y), and tryptophan (W). Tryptophan, however, is expected to be used less frequently because of its relative inflexibility. In addition, the side chain flexibility of lysine, arginine, and methionine allow these amino acids to make base contacts from long or intennediate distances, warranting their inclusion in both Groups 2 and 3.
These groups are also shown in tabular form below:

Group 1 Group 2 Group 3 glycine (G) glutamine (Q) arginine (R) alanine (A) glutamate (E) histidine (H) serine (S) lysine (K) phenylalanine (F) threonine (T) methionine (M) tyrosine (Y) cysteine (C) arginine (R) tryptophan (W) valine (V) lysine (K) leucine (L) methionine (M) isoleucine (I) aspartate (D) asparagine (N) proline (P) [0093] With respect to the chemical nature of the side chains, the different amino acids are evaluated for their potential interactions with the different nucleic acid bases (e.g., van der Waals forces, ionic bonding, hydrogen bonding, and hydrophobic interactions) and residues are selected which either favor or disfavor specific interactions of the meganuclease with a particular base at a particular position in the double-stranded DNA recognition sequence half-site. In some instances, it may be desired to create a half-site with one or more complete or partial degenerate positions. In such cases, one
-31-may choose residues which favor the presence of two or more bases, or residues which disfavor one or more bases. For example, partial degenerate base recognition can be achieved by sterically hindering a pyrimidine at a sense or antisense position.
[0094] Recognition of guanine (G) bases is achieved using amino acids with basic side chains that form hydrogen bonds to N7 and 06 of the base. Cytosine (C) specificity is confeiTed by negatively-charged side chains which interact unfavorably with the major groove electronegative groups present on all bases except C. Thymine (T) recognition is rationally-designed using hydrophobic and van der Waals interactions between hydrophobic side chains and the major groove methyl group on the base.
Finally, adenine (A) bases are recognized using the carboxamide side chains Asn and Gln or the hydroxyl side chain of Tyr through a pair of hydrogen bonds to N7 and N6 of the base.
Lastly, His can be used to confer specificity for a purine base (A or G) by donating a hydrogen bond to N7. These straightforward rules for DNA recognition can be applied to predict contact surfaces in which one or both of the bases at a particular base-pair position are recognized through a rationally-designed contact.
[0095] Thus, based on their binding interactions with the different nucleic acid bases, and the bases which they favor at a position with which they make contact, each amino acid residue can be assigned to one or more different groups corresponding to the different bases they favor (i.e., G, C, T or A). Thus, Group G includes arginine (R), lysine (K) and histidine (H); Group C includes aspartate (D) and glutamate (E); Group T
includes alanine (A), valine (V), leucine (L), isoleucine (I), cysteine (C), threonine (T), methionine (M) and phenylalanine (F); and Group A includes asparagine (N), glutamine (N), tyrosine (Y) and histidine (H). Note that histidine appears in both Group G and Group A; that serine (S) is not included in any group but may be used to favor a degenerate position; and that proline, glycine, and tryptophan are not included in any particular group because of predominant steric considerations. These groups are also shown in tabular forin below:
-32-Group G Group C Group T Group A
arginine (R) aspartate (D) alanine (A) asparagine (N) lysine (K) glutainate (E) valine (V) glutamine (Q) histidine (H) leucine (L) tyrosine (Y) isoleucine (I) histidine (H) cysteine (C) threonine (T) methionine (M) phenylalanine (F) [0096] Thus, in accordance with the invention, in order to effect a desired change in the recognition sequence half-site of a meganuclease at a given position X, (1) determine at least the relevant portion of the three-dimensional structure of the wild-type or reference ineganuclease-DNA complex and the amino acid residue side chains which define the contact surface at position X; (2) determine the distance between the 0-carbon of at least one residue comprising the contact surface and at least one base of the base pair at position X; and (3)(a) for a residue which is < 6 A from the base, select a residue from Group 1 and/or Group 2 which is a member of the appropriate one of Group G, Group C, Group T or Group A to promote the desired change, and/or (b) for a residue which is >
6 A from the base, select a residue from Group 2 and/or Group 3 which is a member of the appropriate one of Group G, Group C, Group T or Group A to promote the desired change. More than one such residue coinprising the contact surface can be selected for analysis and modification and, in some embodiments, each such residue is analyzed and multiple residues are modified. Similarly, the distance between the P-carbon of a residue included in the contact surface and each of the two bases of the base pair at position X
-33-can be determined and, if the residue is within 9A of both bases, then different substitutions can be made to affect the two bases of the pair (e.g., a residue froin Group I
to affect a proximal base on one strand, or a residue from Group 3 to affect a distal base on the other strand). Alternatively, a combination of residue substitutions capable of interacting with both bases in a pair can affect the specificity (e.g., a residue from the T
Group contacting the sense strand coinbined with a residue from the A Group contacting the antisense strand to select for T/A). Finally, multiple alternative modifications of the residues can be validated either empirically (e.g., by producing the recombinant ineganuclease and testing its sequence recognition) or coinputationally (e.g., by computer modeling of the meganuclease-DNA coinplex of the modified enzyme) to choose ainongst alteinatives.
[0097] Once one or inore desired amino acid modifications of the wild-type or reference meganuclease are selected, the rationally-designed meganuclease can be produced by recombinant methods and techniques well known in the art. In some embodiments, non-random or site-directed inutagenesis techniques are used to create specific sequence modifications. Non-limiting examples of non-random mutagenesis techniques include overlapping primer PCR (see, e.g., Wang et al. (2006), Nucleic Acids Res. 34(2): 517-527), site-directed mutagenesis (see, e.g., U.S. Pat. No.
7,041,814), cassette inutagenesis (see, e.g., U.S. Pat. No. 7,041,814), and the manufacturer's protocol for the Altered Sites II Mutagenesis Systems lcit commercially available from Promega Biosciences, Inc. (San Luis Obispo, CA).
[0098] The recognition and cleavage of a specific DNA sequence by a rationally-designed meganuclease can be assayed by any method known by one skilled in the art (see, e.g., U.S. Pat. Pub. No. 2006/0078552). In certain embodiments, the determination of ineganuclease cleavage is determined by in vitro cleavage assays. Such assays use in vitro cleavage of a polynucleotide substrate comprising the intended recognition sequence of the assayed meganuclease and, in certain embodiments, variations of the intended recognition sequence in which one or more bases in one or both half-sites have been changed to a different base. Typically, the polynucleotide substrate is a double-stranded DNA molecule comprising a target site which has been synthesized and cloned into a vector. The polynucleotide substrate can be linear or circular, and typically comprises
-34-only one recognition sequence. The meganuclease is incubated with the polynucleotide substrate under appropriate conditions, and the resulting polynucleotides are analyzed by known methods for identifying cleavage products (e.g., electrophoresis or chromatography). If there is a single recognition sequence in a linear, double-strand DNA substrate, the meganuclease activity is detected by the appearance of two bands (products) and the disappearance of the initial full-length substrate band. In one embodiment, meganuclease activity can be assayed as described in, for example, Wang et al. (1997), Nucleic Acid Res., 25: 3767-3776.
[0099] In other embodiments, the cleavage pattern of the meganuclease is determined using in vivo cleavage assays (see, e.g., U.S. Pat. Pub. No.
2006/0078552). In particular einbodiments, the in vivo test is a single-strand annealing recombination test (SSA). This kind of test is known to those of skill in the art (Rudin et al.
(1989), Genetics 122: 519-534; Fishman-Lobell et al. (1992), Science 258: 480-4).
[0100] As will be apparent to one of skill in the art, additional amino acid substitutions, insertions or deletions can be made to domains of the meganuclease enzyines other than those involved in DNA recognition and binding without complete loss of activity. Substitutions can be conservative substitutions of similar ainino acid residues at structurally or functionally constrained positions, or can be non-conservative substitutions at positions which are less structurally or functionally constrained. Such substitutions, insertions and deletions can be identified by one of ordinary skill in the art by routine experiinentation without undue effort. Thus, in some embodiments, the recombinant meganucleases of the invention include proteins having anywhere from 85%
to 99% sequence similarity (e.g., 85%, 87.5%, 90%, 92.5%, 95%, 97.5%, 99%) to a reference meganuclease sequence. With respect to each of the wild-type I-CreI, I-MsoI, I-Scel and I-CeuI proteins, the most N-terminal and C-tenninal sequences are not clearly visible in X-ray crystallography studies, suggesting that these positions are not structurally or functionally constrained. Therefore, these residues can be excluded from calculation of sequence similarity, and the following reference meganuclease sequences can be used: residues 2-153 of SEQ ID NO: 1 for I-Crel, residues 6-160 of SEQ
ID NO:
6 for I-Msol, residues 3-186 of SEQ ID NO: 9 for I-SceI, and residues 5-211 of SEQ ID
NO: 12 for I-CeuI.
-35-2.2 LAGLIDADG Family Meganucleases [0101] The LAGLIDADG meganuclease family is coinposed of more than 200 members from a diverse phylogenetic group of host organisms. All members of this family have one or two copies of a highly conseived LAGLIDADG motif along with other stilxctural motifs involved in cleavage of specific DNA sequences.
Enzymes that have a single copy of the LAGLIDADG motif (i.e., mono-LAGLIDADG meganucleases) function as dimers, whereas the enzymes that have two copies of this motif (i.e., di-LAGLIDADG meganucleases) function as monomers.
[0102] All LAGLIDADG family members recognize and cleave relatively long sequences (> 12bp), leaving four nucleotide 3' overhangs. These enzymes also share a number of structural motifs in addition to the LAGLIDADG motif, including a similar arrangement of anti-parallel (3-strands at the protein-DNA interface. Amino acids within these conserved structural motifs are responsible for interacting with the DNA
bases to confer recognition sequence specificity. . The overall structural similarity between some members of the family (e.g., I-Crel, I-MsoI, I-Scel and I-CeuI) has been elucidated by X-ray crystallography. Accordingly, the meinbers of this family can be modified at particular amino acids within such structural motifs to change the over-all activity or sequence-specificity of the enzymes, and corresponding modifications can reasonable be expected to have similar results in other family members. See, generally, Chevalier et al.
(2001), Nucleic Acid Res. 29(18): 3757-3774).

2.2.1 Meganucleases Derived from I-Crel [0103] In one aspect, the present invention relates to rationally-designed meganucleases which are based upon or derived from the I-CreI meganuclease of Chlarrrydomonas reinhardtii. The wild-type ainino acid sequence of the I-Crel meganuclease is shown in SEQ ID NO: 1, which corresponds to Genbank Accession #
P05725. Two recognition sequence half sites of the wild-type I-Crel meganuclease from crystal structure PDB # 1BP7 are shown below:
-36-Position -9-8-7-6-5-4-3-2-1 5' -G A A A C T G T C T C A C G A C G T T T T G-3' SEQ ID NO: 2 3' -C T T T G A C A G A G T G C T G C A A A A C-5' SEQ ID NO: 3 Position -1-2-3-4-5-6-7-8-9 Note that this natural recognition sequence is not perfectly palindromic, even outside the central four base pairs. The two recognition sequence half-sites are shown in bold on their respective sense strands.
[0104] Wild-type I-CreI also recognizes and cuts the following perfectly palindromic (except for the central NI -N4 bases) sequence:

Position -9-8-7-6-5-4-3-2-1 5' -C A A A C T G T C G T G A G A C A G T T T G-3' SEQ ID NO: 4 3' -G T T T G A C A G C A C T C T G T C A A A C-5' SEQ ID NO: 5 Position -1-2-3-4-5-6-7-8-9 [0105] The palindromic sequence of SEQ ID NO: 4 and SEQ ID NO: 5 is considered to be a better substrate for the wild-type I-CreI because the enzyme binds this site with higher affinity and cleaves it more efficiently than the natural DNA
sequence.
For the purposes of the following disclosure, and with particular regard to the experimental results presented herein, this palindromic sequence cleaved by wild-type I-CreI is referred to as "WT" (see, e.g., Figure 2(A)). The two recognition sequence half-sites are shown in bold on their respective sense strands.
[0106J Figure 1(A) depicts the interactions of a wild-type I-CreI meganuclease hoinodiiner with a double-stranded DNA recognition sequence, Figures 1(B) shows the specific interactions between amino acid residues of the enzyme and bases at the -4 position of one half-site for a wild-type enzyme and one wild-type recognition sequence, and Figures 1(C)-(E) show the specific interactions between amino acid residues of the enzyine and bases at the -4 position of one half-site for three rationally-designed meganucleases of the invention with altered specificity at position -4 of the half-site.
[0107] Thus, the base preference at any specified base position of the half-site can be rationally altered to each of the other three base pairs using the methods disclosed herein. First, the wild type recognition surface at the specified base position is determined (e.g., by analyzing ineganuclease-DNA coinplex co-crystal structures; or by
-37-computer modeling of the meganuclease-DNA coinplexes). Second, existing and potential contact residues are detennined based on the distances between the (3-carbons of the surrounding ainino acid positions and the nucleic acid bases on each DNA
strand at the specified base position. For exainple, and without limitation, as shown in Figure 1(A), the I-CreI wild type meganuclease-DNA contact residues at position -4 involve a glutamine at position 26 which hydrogen bonds to an A base on the antisense DNA
strand. Residue 77 was also identified as potentially being able to contact the -4 base on the DNA sense strand. The P-carbon of residue 26 is 5.9 A away from N7 of the A base on the antisense DNA strand, and the (3-carbon of residue 77 is 7.15 A away from the C5-inethyl of the T on the sense strand. According to the distance and base chemistry rules described herein, a C on the sense strand could hydrogen bond with a glutamic acid at position 77 and a G on the antisense strand could bond with glutamine at position 26 (mediated by a water molecule, as observed in the wild-type I-CreI crystal structure) (see Fig 1(C)); a G on the sense strand could hydrogen bond with an arginine at position 77 and a C on the antisense strand could hydrogen bond with a glutainic acid at position 26 (see Fig 1(D)); an A on the sense strand could hydrogen bond with a glutamine at position 77 and a T on the antisense strand could form hydrophobic contacts with an alanine at position 26 (see Fig. 1(E)). If the base specific contact is provided by position 77, then the wild-type contact, Q26, can be substituted (e.g., with a serine residue) to reduce or remove its influence on specificity. Alternatively, complementary mutations at positions 26 and 77 can be combined to specify a par-ticular base pair (e.g., A26 specifies a T on the antisense strand and Q77 specifies an A on the sense strand (Fig. 1(E)). These predicted residue substitutions have all been validated experimentally.
[0108] Thus, in accordance with the invention, a substantial number of amino acid modifications to the DNA recognition domain of the I-CreI meganuclease have been identified which, singly or in coinbination, result in recombinant meganucleases with specificities altered at individual bases within the DNA recognition sequence half-site, such that these rationally-designed meganucleases have half-sites different from the wild-type enzylne. The amino acid modifications of I-Crel and the resulting change in recognition sequence half-site specificity are shown in Table 1:
-38-H
(~ O o o (0 ~ ~
a ~ a ~ ~ U) :E
~

~ 00 , ro (9 Y
00 to cf) CY
to m 00 *
.~~ 00 L
Y
~
-+ y ~ o0 a a = Y
~ N m H o ~a U
U.
0 ~ ~ ~

r ti~ ti ~~ ~ Cfl CO
C~ U-> >- 0 2 2 d I CY :2 U-j u-YW D~
~ W W~ _~W ~W

i0 U~ ti~ d~ d~ V~ ~~~ dd N N ~ dN
~2G'2Y~ WO WL- WY w iOin0)io o~~~ q q o~ (o a~ J U} U Q ~ ~ Q>~ z U U N ~

O Ch d ~
a ~l-v ~ i~ izd ~zzc~

N
~
~ ~
~ CY) U ~
~
N
r'M
d.' Q Q Q =

O p O 0) M O) p0 00 e0 M C'7 M c'7 M N N N N
~ ~ J ~ LL U J> Q U
d~ ~
O
~
o ~ Y~~j tm1.T CY) ~Y
W~ W Y~ W 0 W

dU
Z~ uM..}

rn C?

Bold entries are wild-type contact residues and do not constitute "modifications" as used herein.
An asterisk indicates that the residue contacts the base on the antisense strand.
2.2.2 Meganucleases Derived from I-Msol [0109] In another aspect, the present invention relates to rationally-designed meganucleases which are based upon or derived from the I-Msol meganuclease of Monorn.astix sp. The wild-type amino acid sequence of the I-MsoI meganuclease is shown in SEQ ID NO: 6, which corresponds to Genbank Accession # AAL34387. Two recognition sequence half-sites of the wild-type I-MsoI meganuclease from crystal structure PDB # 1 M5X are shown below:

Position -9-8-7-6-5-4-3-2-1 5'-C A G A A C G T C G T G A G A C A G T T C C-3 ' SEQ ID NO : 7 3'-G T C T T G C A G C A C T C T G T C A A G G-5' SEQ ID NO: 8 Position -1-2-3-4-5-6-7-8-9 Note that the recognition sequence is not perfectly palindromic, even outside the central four base pairs. The two recognition sequence half-sites are shown in bold on their respective sense strands.
[0110] In accordance with the invention, a substantial number of amino acid modifications to the DNA recognition domain of the I-MsoI meganuclease have been identified which, singly or in coinbination, can result in recombinant meganucleases with specificities altered at individual bases within the DNA recognition sequence half-sites, such that these rationally-designed meganucleases have recognition sequences different from the wild-type enzyme. Ainino acid modifications of I-MsoI and the predicted change in recognition sequence half-site specificity are shown in Table 2:

Favored Sense-Strand Base Position A C G T
-'I K75* D77 K77 C77 A49* K49* E49* Q79*
C49* R75* E79*
K79* K75*
R79*
K79*

K81 D75 E47* C75 C47* R47* E81* V75 147* K47* 175 L47* K81* T75 R81* Q47*
Q81 *

C26* Y72 K72 Y72 L26* H26* Y26* H26*
V26* K26* F26*
A26* R26*
126*
-4 K28 K28* R83 K28 Q83 R28* K83 K83 E83 Q28*
-5 K28 K28* R45 Q28*
C28* R28* E28*
L28*
128*
-6 130* E43 R43 K43 V30* E85 K43 185 S30* K30* K85 V85 L30* R30* R85 L85 Q43 E30* Q30*
D30*

Bold entries are represent wild-type contact residues and do not constitute "modifications" as used herein.
An asterisk indicates that the residue contacts the base on the antisense strand.

2.2.3 Meganucleases Derived from I-Scel [0111] In another aspect, the present invention relates to rationally-designed meganucleases which are based upon or derived from the I-Scel meganuclease of Saccharoinyces cer-evisiae. The wild-type amino acid sequence of the I-Scel meganuclease is shown in SEQ ID NO: 9, which corresponds to Genbank Accession #
CAA09843. The recognition sequence of the wild-type I-Scel meganuclease from crystal structure PDB # 1R7M is shown below:

Sense 5'-T T A C C C T G T T A T C C C T A G-3' SEQ ID NO:
Antisense 3'-A A T G G G A C A A T A G G G A T C-5' SEQ ID NO:

Position 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 Note that the recognition sequence is non-palindromic and there are not four base pairs separating half-sites.
[0112] In accordance with the invention, a substantial number of amino acid modifications to the DNA recognition domain of the I-SceI meganuclease have been identified which, singly or in combination, can result in recombinant meganucleases with specificities altered at individual bases within the DNA recognition sequence, such that these rationally-designed meganucleases have recognition sequences different from the wild-type enzyine. The amino acid modifications of I-SceI and the predicted change in recognition sequence specificity are shown in Table 3:

Favored Sense-Strand Base Position A C G T
4 K50 R50* E50* K57 K50* R57 M57 E57 K57 Q50*
K48 R48* E48* Q48*
Q102 K48* K102 C102 6 K59 R59* K84 Q59*
K59* E59* Y46 7 C46* R46* K86 K68 L46* K46* R86 C86 V46* E86 E46* L86 Q46*
8 K61* E88 E61* K88 S61* R61* R88 Q61*
V61* H61* K88 H61*
A61*
L61*
9 T98* R98* E98* Q98*
C98* K98* D98*
V98*
L98*
V96* K96* D96* Q96*
C96* R96* E96*
A96*
11 C90* K90* E90* Q90 "
L90* R90*

13 0193"' K193* E193* Q193*
L193* R193* D193* C163 14 L192* E161 K147 K161 C192* R192* K161 Q192*
K192* R161 D 192*
E192*

17 N152* K152* N152* Q152*
S152* K150* S152* Q150*
C150* D152*
L150* D150*
V150* E150*
T150*
18 K155* R155* E155* H155*
C155* K155* Y155*
Bold entries are wild-type contact residues and do not constitute "modifications" as used herein.
An asterisk indicates that the residue contacts the base on the antisense strand.

2.2.4 Meganucleases Derived from I-CeuI
101131 In another aspect, the present invention relates to rationally-designed meganucleases which are based upon or derived from the I-CeuI meganuclease of Chlainydomorzas eugametos. The wild-type amino acid sequence of the I-CeuI
meganuclease is shown in SEQ ID NO: 12, which corresponds to Genbank Accession #
P32761. Two recognition sequence half sites of the wild-type I-Ceul meganuclease from crystal structure PDB # 2EX5 are shown below:

Position -9-8-7-6-5-4-3-2-1 5' -A T A A C G G T C C T A A G G T A G C G A A-3' SEQ ID NO:

3'-T A T T G C C A G G A T T C C A T C G C T T-5' SEQ ID NO:

Position -1-2-3-4-5-6-7-8-9 Note that the recognition sequence is non-palindromic, even outside the central four base pairs, despite the fact that I-Ceul is a homodimer, due to the natural degeneracy in the I-Ceul recognition interface (Spiegel et al. (2006), Structure 14:869-80). The two recognition sequence half-sites are shown in bold on their respective sense strands.
[0114] In accordance with the invention, a substantial number of amino acid modifications to the DNA recognition domain of the I-Ceul meganuclease have been identified which, singly or in combination, result in recombinant ineganucleases with specificities altered at individual bases within the DNA recognition sequence half-site, such that these rationally-designed meganucleases can have recognition sequences different from the wild-type enzyme. The amino acid modifications of I-CeuI
and the predicted change in recognition sequence specificity are shown in Table 4:

Favored Sense-Strand Base Position A C G T
-1 C92* K116* E116* Q116*
A92* R116* E92* Q92*
V92* D116*
K92*

C90* D117 R124 V117 L90* R174* K124 T117 V90* K124* E124* Q90*
K90* E90*
R90* D90*
K68*
-3 C70* K70* E70* Q70*
V70* E88*
T70*
L70*
K70*

K88* R88* E88* Q88*
L88* K88* D88*
C88* K72*
C72*
L72*
V72*
-5 C74* K74* E74* C128 L74* K128 L128 V74* R128 V128 T74* E128 T128 R84* R86 L86 K84* K86 E84*
-7 L76* R76* E76* H76*
C76* K76* R84 Q76*
K76* H76*

Bold entries are wild-type contact residues and do not constitute "modifications" as used herein.
An asterisk indicates that the residue contacts the base on the antisense strand.

2.2.5 Specifically-Excluded Recoinbinant Meganucleases [0115] The present invention is not intended to embrace certain recombinant meganucleases which have been described in the prior art, and which have been developed by alternative methods. These excluded meganucleases include those described by Arnould et al. (2006), J. Mol. Biol. 355: 443-58; Sussman et al.
(2004), J.
Mol. Bi.ol. 342: 31-41; Chames et al. (2005), Nucleic Acids Res. 33: e178;
Seliginan et al.
(2002), Nucleic Acids Res. 30: 3870-9; and Ashworth et al. (2006), Nature 441(7093):656-659; the entire disclosures of which are hereby incorporated by reference, including recombinant meganucleases based on I-CreI with single substitutions selected from C33, R33, A44, H33, K32, F33, R32, A28, A70, E33, V33, A26, and R66. Also excluded are recombinant meganucleases based on I-CreI with three substitutions selected from A68/N70/N75 and D44/D70/N75, or with four substitutions selected from K44/T68/G60/N75 and R44/A68/T70/N75. Lastly, specifically excluded is the recoinbinant meganuclease based on I-Msol with the pair of substitutions L28 and R83.
These substitutions or combinations of substitutions are referred to herein as the "excluded modifications."

2.2.6 Meganucleases with Multiple Changes in the Recognition Sequence Half-Site [0116] In another aspect, the present invention relates to rationally-designed meganucleases which are produced by coinbining two or more amino acid modifications as described in sections 2.2.1-2.2.4 above, in order to alter half-site preference at two or more positions in a DNA recognition sequence half-site. For exainple, without limitation, and as more fully described below, the enzyine DJI was derived from I-CreI by incorporating the modifications R30/E38 (which favor C at position -7), R40 (which favors G at position -6), R42 (which favors at G at position -5), and N32 (which favors coinplete degeneracy at position -9). The rationally-designed DJl meganuclease invariantly recognizes C_7 G_6 G_5 compared to the wild-type preference for A_7 A_6 C_5, and has increased tolerance for A at position -9.
[0117] The ability to combine residue substitutions that affect different base positions is due in part to the modular nature of the LAGLIDADG meganucleases.
A
majority of the base contacts in the LAGLIDADG recognition interfaces are made by individual ainino acid side chains, and the interface is relatively free of interconnectivity or hydrogen bonding networks between side chains that interact with adjacent bases. This generally allows manipulation of residues that interact with one base position without affecting side chain interactions at adjacent bases. The additive nature of the mutations listed in sections 2.2.1-2.2.4 above is also a direct result of the method used to identify these mutations. The method predicts side chain substitutions that interact directly with a single base. Interconnectivity or hydrogen bonding networlcs between side chains is generally avoided to maintain the independence of the substitutions within the recognition interface.
[0118] Certain combinations of side chain substitutions are coinpletely or partially incompatible with one another. When an incompatible pair or set of amino acids are incoiporated into a rationally-designed meganuclease, the resulting enzyme will have reduced or eliminated catalytic activity. Typically, these incompatibilities are due to steric interference between the side chains of the introduced amino acids and activity can be restored by identifying and removing this interference. Specifically, when two amino acids with large side chains (e.g., amino acids from group 2 or 3) are incorporated at ainino acid positions that are adjacent to one another in the meganuclease structure (e.g., positions 32 and 33, 28 and 40, 28 and 42, 42 and 77, or 68 and 77 in the case of meganucleases derived from I-CreI), it is likely that these two amino acids will interfere with one another and reduce enzyme activity. This interference be eliminated by substituting one or both incompatible amino acids to an amino acid with a smaller side chain (e.g., group 1 or group 2). For example, in rationally-designed meganucleases derived from I-CreI, K28 interferes with both R40 and R42. To maximize enzylne activity, R40 and R42 can be coinbined with a serine or aspartic acid at position 28.
[0119] Combinations of amino substitutions, identified as described herein, can be used to rationally alter the specificity of a wild-type meganuclease (or a previously modified meganuclease) from an original recognition sequence to a desired recognition sequence which may be present in a nucleic acid of interest (e.g., a genome).
Figure 2A, for exainple, shows the "sense" strand of the I-CreI meganuclease recognition sequence WT (SEQ ID NO: 4) as well as a number of other sequences for which a rationally-designed meganuclease would be useful. Conserved bases between the WT
recognition sequence and the desired recognition sequence are shaded. In accordance with the invention, recombinant meganucleases based on the I-CreI meganuclease can be rationally-designed for each of these desired recognition sequences, as well as any others, by suitable amino acid substitutions as described herein.

3. Rationally-Designed Meganucleases with Altered DNA-Binding Affinity [0120] As described above, the DNA-binding affinity of the recombinant meganucleases of the invention can be modulated by altering certain amino acids that forin the contact surface with the phosphodiester backbone of DNA. The contact surface comprises those amino acids in the enzyine with (3-carbons less than 9 A from the DNA
backbone, and with side chains oriented toward the DNA, irrespective of whether the residues make contacts with the DNA backbone in the wild-type meganuclease-DNA
coinplex. Because DNA-binding is a necessary precursor to enzyine activity, increases/decreases in DNA-binding affinity have been shown to cause increases/decreases, respectively, in enzyme activity. However, increases/decreases in DNA-binding affinity also have been shown to cause decreases/increases in the meganuclease sequence-specificity. Therefore, both activity and specificity can be modulated by modifying the phosphodiester backbone contacts.
[0121] Specifically, to increase enzyme activity/decrease enzyme specificity:
[0122] (i) Remove electrostatic repulsion between the enzyme and DNA
baclcbone. If an identified amino acid has a negatively-charged side chain (e.g., aspartic acid, glutamic acid) which would be expected to repulse the negatively-charged DNA

backbone, the repulsion can be eliminated by substituting an amino acid with an uncharged or positively-charged side chain, subject to effects of steric interference. An experimentally verified exainple is the mutation of glutainic acid 80 in I-CreI to glutamine.
[0123] (ii) Introduce electrostatic attraction interaction between the enzyme and the DNA backbone. At any of the positions of the contact surface, the introduction of an amino acid with a positively-charged side chain (e.g., lysine or arginine) is expected to increase binding affinity, subject to effects of steric interference.
[0124] (iii) Introduce a hydrogen-bond between the enzyine and the DNA
backbone. If an ainino acid of the contact surface does not make a hydrogen bond with the DNA backbone because it lacks an appropriate hydrogen-bonding functionality or has a side chain that is too short, too long, and/or too inflexible to interact with the DNA
backbone, a polar amino acid capable of donating a hydrogen bond (e.g., ser-ine, threonine, tyrosine, histidine, glutamine, asparagine, lysine, cysteine, or arginine) with the appropriate length and flexibility can be introduced, subject to effects of steric interference.
[0125] Specifically, to decrease enzyine activity/increase enzyme specificity:
[0126] (i) Introduce electrostatic repulsion between the enzyme and the DNA
baclcbone. At any of the positions of the contact surface, the introduction of an amino acid with a negatively-charged side chain (e.g.,, glutainic acid, aspartic acid) is expected to decrease binding affinity, subject to effects of steric interference.
[0127] (ii) Remove electrostatic attraction between the enzyme and DNA. If any amino acid of the contact surface has a positively-charged side chain (e.g., lysine or arginine) that interacts with the negatively-charged DNA backbone, this favorable interaction can be eliminated by substituting an amino acid with an uncharged or negatively-charged side chain, subject to effects of steric interference. An experimentally verified exainple is the mutation of lysine 116 in I-Crel to aspartic acid.
[0128] (iii) Remove a hydrogen-bond between the enzyine and the DNA
backbone. If any amino acid of the contact surface makes a hydrogen bond with the DNA backbone, it can be substituted to an ainino acid that would not be expected to make a similar hydrogen bond because its side chain is not appropriately functionalized or it lacks the necessary length/flexibility characteristics.
[0129] For exainple, in soine recombinant meganucleases based on I-CreI, the glutainic acid at position 80 in the I-CreI meganuclease is altered to either a lysine or a glutamine to increase activity. In another embodiment, the tyrosine at position 66 of I-Crel is changed to arginine or lysine, wliich increases the activity of the ineganuclease.
In yet another embodiment, enzyme activity is decreased by changing the lysine at position 34 of I-Crel to aspartic acid, changing the tyrosine at position 66 to aspartic acid, and/or changing the lysine at position 116 to aspartic acid.
[0130] The activities of the recombinant meganucleases can be modulated such that the recombinant enzyme has anywhere from no activity to very high activity with respect to a particular recognition sequence. For example, the DJ1 recombinant meganuclease when carrying glutamic acid mutation at position 26 loses activity completely. However, the combination of the glutainic acid substitution at position 26 and a glutainine substitution at position 80 creates a recombinant meganuclease with high specificity and activity toward a guanine at -4 within the recognition sequence half-site (see Figure 1(D)).
[0131] In accordance with the invention, amino acids at various positions in proximity to the phosphodiester DNA backbone can be changed to simultaneously affect both meganuclease activity and specificity. This "tuning" of the enzyme specificity and activity is accotnplished by increasing or decreasing the number of contacts made by amino acids with the phosphodiester backbone. A variety of contacts with the phosphodiester baclcbone can be facilitated by amino acid side chains. In some embodiments, ionic bonds, salt bridges, hydrogen bonds, and steric hindrance affect the association of amino acid side chains with the phosphodiester backbone. For example, for the I-CreI ineganuclease, alteration of the lysine at position 116 to an aspartic acid removes a salt bridge between nucleic acid base pairs at positions -8 and -9, reducing the rate of enzyme cleavage but increasing the specificity.
[0132] The residues forming the backbone contact surface of each of the wild-type I-CreI (SEQ ID NO: 1), I-Msol (SEQ ID NO: 6), I-Scel (SEQ ID NO: 9) and I-Ceul (SEQ ID NO: 12) meganucleases are identified in Table 5 below:

I-CreI I-MsoI I-SceI I-CeuI
P29, K34, T46, K36, Q41, R51, N15, N17, L19, K21, D25, K28, K48, R51, V64, N70, 185, G86, S87, K20, K23, K63, K31, S68, N70, Y66, E80, 181, K82, T88, H89, Y118, L80, S81, H84, L92, H94, R112, R114, Ll 12, K116, D137, Q122, K123, Q139, N94, N120, K122, S117, N120, D128, K139, T140, T143 K143, R144, E147, K148, Y151, K153, N129, R130, H172 S150, N152 T156, N157, S159, N163, Q165, S166, Y188, K190, 1191, K193, N194, K195, Y199, D201, S202, Y222, K223 [0133] To increase the affinity of an enzyme and thereby make it more active/less specific:
(1) Select an amino acid from Table 5 for the corresponding enzyme that is either negatively-charged (D or E), hydrophobic (A, C, F, G, I, L, M, P, V, W, Y), or uncharged/polar (H, N, Q, S, T).
(2) If the amino acid is negatively-charged or hydrophobic, mutate it to uncharged/polar (less effect) or positively-charged (K or R, more effect).
(3) If the amino acid is uncharged/polar, mutate it to positively-charged.

[0134] To decrease the affinity of an enzyme and thereby make it less active/more specific:
(1) Select an amino acid from Table 5 for the corresponding enzyme that is either positively-charged (K or R), hydrophobic (A, C, F, G, I, L, M, P, V, W, Y), or uncharged/polar (H, N, Q, S, T).

(2) If the amino acid is positively-charged, mutate it to uncharged/polar (less effect) or negatively-charged (inore effect).
(3) If the amino acid is hydrophobic or uncharged/polar, inutate it to negatively-charged.
4. Heterodimeric Meganucleases [0135] In another aspect, the invention provides meganucleases which are heterodimers forined by the association of two monomers, one of which may be a wild-type and one or both of which may be a non-naturally-occurring or recombinant form.
For exainple, wild-type I-CreI meganuclease is norinally a homodimer composed of two monomers that each bind to one half-site in the pseudo-palindromic recognition sequence.
A heterodimeric recombinant meganuclease can be produced by combining two meganucleases that recognize different half-sites, for example by co-expressing the two meganucleases in a cell or by mixing two meganucleases in solution. The formation of heterodimers can be favored over the formation of homodimers by altering ainino acids on each of the two monomers that affect their association into dimers. In particular embodiments, certain amino acids at the interface of the two monomers are altered from negatively-charged ainino acids (D or E) to positively- charged amino acids (K
or R) on a first monomer and from positively- charged amino acids to negatively-charged amino acids on a second monomer (Table 6). For example, in the case of meganucleases derived from I-CreI, lysines at positions 7 and 57 are mutated to glutamic acids in the first monomer and glutamic acids at positions 8 and 61 are mutated to lysines in the second monomer. The result of this process is a pair of monomers in which the first monomer has an excess of positively-charged residues at the dimer interface and the second monomer has an excess of negatively-charged residues at the dimer interface.
The first and second monomer will, therefore, associate preferentially over their identical monomer pairs due to the electrostatic interactions between the altered amino acids at the interface.

I-CreI: First Monomer I-Crel: Second Monomer Substitutions Substitutions K7 to E7 or D7 E8 to K8 or R8 K57 to E57 or D57 E61 to K61 or R61 K96 to E96 or D96 I-Msol: First Monomer I-Msol: Second Monomer Substitutions Substitutions R302 to E302 or D302 D20 to K60 or R60 E11 to K11 or R11 Q64 to K64 or R64 I-CeuI: First Monomer I-CeuI: Second Monomer Substitutions Substitutions R93 to E93 or D93 E152 to K152 or R152 101361 Alternatively, or in addition, certain ainino acids at the interface of the two monomers can be altered to sterically hinder homodimer formation.
Specifically, amino acids in the dimer interface of one monomer are substituted with larger or bulkier residues that will sterically prevent the homodimer. An7ino acids in the dimer interface of the second monomer optionally can be substituted with smaller residues to compensate for the bulkier residues in the first monomer and reinove any clashes in the heterodimer, or can be unmodified.
[0137] In another alternative or additional einbodiment, an ionic bridge or hydrogen bond can be buried in the hydrophobic core of a heterodimeric interface.
Specifically, a hydrophobic residue on one monomer at the core of the interface can be substituted with a positively charged residue. In addition, a hydrophobic residue on the second monomer, that interacts in the wild type homodimer with the hydrophobic residue substituted in the first monomer, can be substituted with a negatively charged residue.
Thus, the two substituted residues can forin an ionic bridge or hydrogen bond.
At the same time, the electrostatic repulsion of an unsatisfied charge buried in a hydrophobic interface should disfavor homodimer formation.

[0138] Finally, as noted above, each monomer of the heterodimer can have different amino acids substituted in the DNA recognition region such that each has a different DNA half-site and the combined diineric DNA recognition sequence is non-palindromic.

5. Methods of Producing Recombinant Cells and Or ang isins 101391 Aspects of the present invention further provide methods for producing recombinant, transgenic or otherwise genetically-modified cells and organisms using rationally-designed meganucleases. Thus, in certain einbodiments, recombinant meganucleases are developed to specifically cause a double-stranded break at a single site or at relatively few sites in the genomic DNA of a cell or an organism to allow for precise insertion(s) of a sequence of interest by homologous recombination. In other embodiments, recombinant meganucleases are developed to specifically cause a double-stranded break at a single site or at relatively few sites in the genomic DNA
of a cell or an organism to either (a) allow for rare insertion(s) of a sequence of interest by non-homologous end-joining or (b) allow for the disruption of the target sequence by non-homologous end-joining. As used herein with respect to homologous recoinbination or non-homologous end-joining of sequences of interest, the term "insertion"
means the ligation of a sequence of interest into a chromosome such that the sequence of interest is integrated into the chromosome. In the case of homologous recombination, an inserted sequence can replace an endogenous sequence, such that the original DNA is replaced by exogenous DNA of equal length, but with an altered nucleotide sequence.
Alternatively, an inserted sequence can include more or fewer bases than the sequence it replaces.
[0140] Therefore, in accordance with this aspect of the invention, the recombinant organisms include, but are not limited to, monocot plant species such as rice, wheat, corn (maize) and rye, and dicot species such as legumes (e.g., kidney beans, soybeans, lentils, peanuts, peas), alfalfa, clover, tobacco and Arabidopsis species. In addition, the recombinant organisms can include, but are not limited to, animals such as humans and non-human primates, horses, cows, goats, pigs, sheep, dogs, cats, guinea pigs, rats, inice, lizards, fish and insects such as Drosophila species. In other einbodunents, the organism is a fungus such as a Candida, Neurospora or Saccharomyces species.

[0141] In some einbodiinents, the methods of the invention involve the introduction of a sequence of interest into a cell such as a germ cell or stem cell that can become a mature recombinant organism or allow the resultant genetically-modified organism to give rise to progeny carrying the inserted sequence of interest in its genome.
[0142] Meganuclease proteins can be delivered into cells to cleave genomic DNA, which allows for homologous recombination or non-homologous end-joining at the cleavage site with a sequence of interest, by a variety of different mechanisms known in the art. For example, the recombinant meganuclease protein can introduced into a cell by techniques including, but not limited to, microinjection or liposome transfections (see, e.g., LipofectainineTM, Invitrogen Corp., Carlsbad, CA). The liposome formulation can be used to facilitate lipid bilayer fusion with a target cell, thereby allowing the contents of the liposome or proteins associated with its surface to be brought into the cell.
Alternatively, the enzyme can be fused to an appropriate uptake peptide such as that from the HIV TAT protein to direct cellular uptake (see, e.g., Hudecz et al.
(2005), Med. Res.
Rev. 25: 679-736).
[0143] Alternatively, gene sequences encoding the meganuclease protein are inserted into a vector and transfected into a eukaryotic cell using techniques known in the art (see, e.g., Ausubel et. al., Current Protocols in Molecular Biology, Wiley 1999). The sequence of interest can be introduced in the same vector, a different vector, or by other means known in the art.
[0144] Non-limiting examples of vectors for DNA transfection include virus vectors, plasmids, cosmids, and YAC vectors. Transfection of DNA sequences can be accomplished by a variety of methods known to those of skill in the art. For instance, liposomes and iinmunoliposoines are used to deliver DNA sequences to cells (see, e.g., Lasic et al. (1995), Science 267: 1275-76). In addition, viruses can be utilized to introduce vectors into cells (see, e.g., U.S. Pat. No. 7,037,492).
Alternatively, transfection strategies can be utilized such that the vectors are introduced as naked DNA
(see, e.g., Rui et al. (2002), Life Sci. 71(15): 1771-8).
[0145] General methods for delivering nucleic acids into cells include: (1) chemical methods (Graham et al. (1973), Virology 54(2):536-539; Zatloukal et al. (1992), Ann. N.Y. Acad. Sci., 660:136-153; (2) physical methods such as microinjection (Capecchi (1980), Cell 22(2):479-488, electroporation (Wong et al. (1982), Biochim.
Biophys. Res. Com.naun. 107(2):584-587; Fromm et al. (1985), Proc. Nat'l Acad.
Sci. USA
82(17):5824-5828; U.S. Pat. No. 5,384,253) and ballistic injection (Johnston et al. (1994), Methods Cell. Biol. 43(A): 353-365; Fynan et al. (1993), Proc. Nat'l Acad.
Sci. USA
90(24): 11478-11482); (3) viral vectors (Clapp (1993), Clin. Perinatol. 20(1):
155-168;
Lu et al. (1993), J. Exp. Med. 178(6):2089-2096; Eglitis et al. (1988), Avd.
Exp. Med.
Biol. 241:19-27; Eglitis et al. (1988), Biotechniques 6(7):608-614); and (4) receptor-mediated mechanisms (Curiel et al. (1991), Proc. Nat'l Acad. Sci. USA
88(19):8850-8854; Curiel et al. (1992), Hum. Gen. Tlier. 3(2):147-154; Wagner et al.
(1992), Proc.
Nat'lAcad. Sci. USA 89 (13):6099-6103).
101461 In certain embodiments, a genetically-modified plant is produced, which contains the sequence of interest inserted into the genome. In certain embodiments, the genetically-modified plant is produced by transfecting the plant cell with DNA
sequences corresponding to the recombinant meganuclease and the sequence of interest, which may or may not be flanked by the meganuclease recognition sequences and/or sequences substantially identical to the target sequence. In other embodiments, the genetically-modified plant is produced by transfecting the plant cell with DNA sequences corresponding to the recombinant meganuclease only, such that cleavage promotes non-homologous end-joining and disrupts the target sequence containing the recognition sequence. In such einbodiments, the meganuclease sequences are under the control of regulatory sequences that allow for expression of the meganuclease in the host plant cells.
These regulatory sequences include, but are not limited to, constitutive plant promoters such as the NOS promoter, chemically-inducible gene promoters such as the dexainethasone-inducible promoter (see, e.g., Gremillon et al. (2004), Plant J. 37:218-228), and plant tissue specific promoters such as the LGC1 promoter (see, e.g., Singh et al. (2003), FEBS Lett. 542:47-52).
[0147] Suitable methods for introducing DNA into plant cells include virtually any method by which DNA can be introduced into a cell, including but not limited to Agrobacterium infection, PEG-mediated transformation of protoplasts (Omirulleh et al.
(1993), Plant Molecular Biology, 21:415-428), desiccation/inhibition-mediated DNA

uptake, electroporation, agitation with silicon carbide fibers, ballistic injection or inicroprojectile bombardment, and the like.
[0148] In other einbodiments, a genetically-modified animal is produced using a recombinant meganuclease. As with plant cells, the nucleic acid sequences can be introduced into a germ cell or a cell that will eventually become a transgenic organism.
In some embodiments, the cell is a fertilized egg, and exogenous DNA molecules can be injected into the pro-nucleus of the fertilized egg. The micro-injected eggs are then transferred into the oviducts of pseudopregnant foster mothers and allowed to develop.
The recoinbinant meganuclease is expressed in the fertilized egg (e.g., under the control of a constitutive promoter, such as 3-phosphoglycerate kinase), and facilitates homologous recombination of the sequence of interest into one or a few discrete sites in the genome. Alternatively, the genetically-modified animals can be obtained by utilizing recombinant embryonic stem ("ES") cells for the generation of the transgenics, as described by Gossler et al. (1986), Proc. Natl. Acad. Sci. USA 83:9065 9069.
[01491 In certain embodiments, a recombinant mainmalian expression vector is capable of directing tissue-specific expression of the nucleic acid preferentially in a particular cell type. Tissue-specific regulatory elements are known in the art. Non-limiting examples of suitable tissue-specific promoters include the albumin promoter (liver-specific; Pinkert et al. (1987), Genes Dev. 1: 268-277), lymphoid-specific promoters (Calame and Eaton (1988), Adv. Imrnunol. 43: 235-275), in particular promoters of T cell receptors (Winoto and Baltimore (1989), EMBO.I. 8: 729-733) and iminunoglobulins (Banerji et al. (1983), Cell 33: 729-740; Queen and Baltimore (1983), Cell 33: 741-748), neuron-specific promoters (e.g., the neurofilament promoter; Byrne and Ruddle (1989), Proc. Natl. Acad. Sci. USA 86: 5473-5477), pancreas-specific promoters (Edlund et al. (1985), Science 230: 912-916), and maminary gland-specific promoters (e.g., milk whey promoter; U.S. Pat. No. 4,873,316 and European Pat.
Pub. EP
0 264 166). Developmentally-regulated promoters are also encompassed, e.g., the inurine hox promoters (Kessel and Gruss (1990), Scieyice 249: 374-379) and the a-fetoprotein promoter (Campes and Tilghinan (1989), Genes Dev. 3: 537-546).
[01501 In certain embodiments, a rationally-designed meganuclease may be tagged with a peptide epitope (e.g., an HA, FLAG, or Myc epitope) to inonitor expression levels or localization. In some embodiments, the meganuclease may be fused to a sub-cellular localization signal such as a nuclear-localization signal (e.g., the nuclear localization signal from SV40) or chloroplast or mitochondrial localization signals. In other embodiments, the meganuclease may be fused to a nuclear export signal to localize it to the cytoplasm. The meganuclease may also be fused to an unrelated protein or protein domain such as a protein that stimulates DNA-repair or homologous recombination (e.g., recA, RAD51, RAD52, RAD54, RAD57 or BRCA2).

6. Methods for Gene Theraby [0151] Aspects of the invention allow for the use of recombinant meganuclease for gene therapy. As used herein, "gene therapy" means therapeutic treatments that coinprise introducing into a patient a functional copy of at least one gene, or gene regulatory sequence such as a promoter, enhancer, or silencer to replace a gene or gene regulatory region that is defective in its structure and/or function. The term "gene therapy" can also refer to modifications made to a deleterious gene or regulatory element (e.g., oncogenes) that reduce or eliminate expression of the gene. Gene therapy can be performed to treat congenital conditions, conditions resulting from mutations or damage to specific genetic loci over the life of the patient, or conditions resulting from infectious organisms.
[0152] In some aspects of the invention, dysfunctional genes are replaced or disabled by the insertion of exogenous nucleic acid sequences into a region of the genome affecting gene expression. In certain embodiments, the recoinbinant meganuclease is targeted to a particular sequence in the region of the genome to be modified so as to alleviate the condition. The sequence can be a region within an exon, intron, promoter, or other regulatory region that is causing dysfunctional expression of the gene.
As used herein, the terin "dysfunctional expression" means aberrant expression of a gene product either by the cell producing too little of the gene product, too inuch of the gene product, or producing a gene product that has a different function such as lacking the necessary function or having more than the necessary function.
[0153] Exogenous nucleic acid sequences inserted into the modified region can be used to provide "repaired" sequences that normalize the gene. Gene repair can be accomplished by the introduction of proper gene sequences into the gene allowing for proper function to be reestablished. In these embodiments, the nucleic acid sequence to be inserted can be the entire coding sequence for a protein or, in certain embodiments, a fragment of the gene coinprising only the region to be repaired. In other embodiments the nucleic acid sequence to be inserted coinprises a promoter sequence or other regulatory elements such that inutations causing abnorinal expression or regulation are repaired. In other embodiments, the nucleic acid sequence to be inserted contains the appropriate translation stop codon lacking in a mutated gene. The nucleic acid sequence can also have sequences for stopping transcription in a recombinant gene lacking appropriate transcriptional stop signals.
[0154] Alternatively, the nucleic acid sequences can eliminate gene function altogether by disrupting the regulatory sequence of the gene or providing a silencer to eliminate gene function. In some embodiments, the exogenous nucleic acid sequence provides a translation stop codon to prevent expression of the gene product.
In other einbodiinents, the exogenous nucleic acid sequences provide transcription stop element to prevent expression of a full length RNA molecule. In still other einbodiinents, gene function is disi-upted directly by the meganuclease by introducing base insertions, base deletions, and/or fraineshift mutations through non-homologous end-joining.
[0155] In many instances, it is desirable to direct the proper genetic sequences to a target cell or population of cells that is the cause of the disease condition.
Such targeting of therapeutics prevents healthy cells from being targeted by the therapeutics. This increases the efficacy of the treatment, while decreasing the potentially adverse effects that the treatment could have on healthy cells.
[0156] Delivery of recombinant meganuclease genes and the sequence of interest to be inserted into the genome to the cells of interest can be accomplished by a variety of mechanisms. In some embodiments, the nucleic acids are delivered to the cells by way of viruses with particular viral genes inactivated to prevent reproduction of the virus. Thus, a virus can be altered so that it is capable only of delivery and maintenance within a target cell, but does not retain the ability to replicate within the target cell or tissue. One or more DNA sequences can be introduced to the altered viral genome, so as to produce a viral genome that acts like a vector, and may or may not be inserted into a host genome and subsequently expressed. More specifically, certain embodiments include employing a retroviral vector such as, but not limited to, the MFG or pLJ vectors. An MFG vector is a siinplified Moloney inurine leukemia virus vector (MoMLV) in which the DNA
sequences encoding the pol and env proteins have been deleted to render it replication defective. A pLJ retroviral vector is also a form of the MoMLV (see, e.g., Korman et al.
(1987), Proc. Nat'l Acad. Sci., 84:2150-2154). In other embodiments, a recombinant adenovirus or adeno-associated virus can be used as a delivery vector.
[0157] In other embodiments, the delivery of recoinbinant meganuclease protein and/or recoinbinant meganuclease gene sequences to a target cell is accoinplished by the use of liposomes. The production of liposomes containing nucleic acid and/or protein cargo is known in the art (see, e.g., Lasic et al. (1995), Science 267: 1275-76).
Iminunoliposomes incorporate antibodies against cell-associated antigens into liposomes, and can delivery DNA sequences for the meganuclease or the meganuclease itself to specific cell types (see, e.g., Lasic et al. (1995), Science 267: 1275-76;
Young et al.
(2005), J. Calif. Dent. Assoc. 33(12): 967-71; Pfeiffer et al. (2006), J.
Vasc. Surg.
43(5):1021-7). Methods for producing and using liposome formulations are well known in the art, (see, e.g., U.S. Pat. No. 6,316,024, U.S. Pat. No. 6,379,699, U.S.
Pat. No.
6,387,397, U.S. Pat. No. 6,511,676 and U.S. Pat. No. 6,593,308, and references cited therein). In some embodiments, liposomes are used to deliver the sequences of interest as well as the recombinant meganuclease protein or recombinant meganuclease gene sequences.

7. Methods for Treating Pathogen Infection.
[0158] Aspects of the invention also provide methods of treating infection by a pathogen. Pathogenic organisms include viruses such as, but not limited to, herpes simplex virus 1, herpes siinplex virus 2, human immunodeficiency virus 1, human iminunodeficiency virus 2, variola virus, polio virus, Epstein-Barr virus, and human papilloma virus and bacterial organisms such as, but not limited to, Bacillus anthracis, Haenzoph.ilzts species, Pneumococcus species, Staphylococcus aureus, Streptococcus species, methicillin-resistant Staphylococcus aureus, and Mycoplasma tuberculosis.

Pathogenic organisms also include fungal organisms such as, but not limited to, Candida, Blaston?yces, Cyyptococcus, and Histoplasina species.
[0159] In some einbodiments, a rationally-designed meganuclease can be targeted to a recognition sequence within the pathogen genoine, e.g., to a gene or regulatory element that is essential for growth, reproduction, or toxicity of the pathogen. In certain embodiments, the recognition sequence may be in a bacterial plasmid.
Meganuclease-inediated cleavage of a recognition sequence in a pathogen genome can stimulate mutation within a targeted, essential gene in the forin of an insertion, deletion or frameshift, by stimulating non-homologous end-joining. Alternatively, cleavage of a bacterial plasmid can result in loss of the plasmid along with any genes encoded on it, such as toxin genes (e.g., B. anthracis Lethal Factor gene) or antibiotic resistance genes.
As noted above, the meganuclease may be delivered to the infected patient, animal, or plant in either protein or nucleic acid form using techniques that are common in the art.
In certain einbodiments, the meganuclease gene may be incorporated into a bacteriophage genome for delivery to pathogenic bacteria.
[0160] Aspects of the invention also provide therapeutics for the treatment of certain forms of cancer. Because human viruses are often associated with tumor formation (e.g., Epstein-Barr Virus andnasopharyngeal carcinomas; Human Papilloma Virus and cervical cancer) inactivation of these viral pathogens may prevent cancer development or progression. Alternatively, double-stranded breaks targeted to the genomes of these tuinor-associated viruses using rationally-designed meganucleases may be used to trigger apoptosis through the DNA damage response pathway. In this manner, it may be possible to selectively induce apoptosis in tumor cells harboring the viral genome.

8. Methods for Genotyping and Pathogen Identification [0161] Aspects of the invention also provide tools for in vitro molecular biology research and development. It is common in the art to use site-specific endonucleases (e.g., restriction enzymes) for the isolation, cloning, and manipulation of nucleic acids such as plasmids, PCR products, BAC sequences, YAC sequences, viruses, and genomic sequences from eukaryotic and prokaryotic organisms (see, e.g., Ausubel et al., Current Protocols in Molecular Biology, Wiley 1999). Thus, in some embodiments, a rationally-designed meganuclease may be used to manipulate nucleic acid sequences in vitro. For exainple, rationally-designed meganucleases recognizing a pair of recognition sequences within the same DNA molecule can be used to isolate the intervening DNA
segment for subsequent manipulation such as ligation into a bacterial plasmid, BAC, or YAC.
[0162] In another aspect, this invention provides tools for the identification of pathogenic genes and organisms. In one embodiment, rationally-designed meganucleases can be used to cleave recognition sites corresponding to polymorphic genetic regions correlated to disease to distinguish disease-causing alleles from healthy alleles (e.g., a rationally-designed meganuclease which recognizes the AF-508 allele of the huinan CFTR gene, see example 4). In this einbodiinent, DNA sequences isolated from a human patient or other organism are digested with a rationally-designed meganuclease, possibly in conjunction with additional site-specific nucleases, and the resulting DNA
fragment pattern is analyzed by gel electrophoresis, capillary electrophoresis, mass spectrometry, or other methods known in the art. This fragmentation pattern and, specifically, the presence or absence of cleavage by the rationally-designed meganuclease, indicates the genotype of the organism by revealing whether or not the recognition sequence is present in the genome. In another embodiment, a rationally-designed ineganuclease is targeted to a polymorphic region in the genome of a pathogenic virus, fungus, or bacterium and used to identify the organism. In this embodiment, the rationally-designed meganuclease cleaves a recognition sequence that is unique to the pathogen (e.g., the spacer region between the 16S and 23S rRNA genes in a bacterium; see, e.g., van der Giessen et al.
(1994), Micy obiology 140:1103-1108) and can be used to distinguish the pathogen from other closely-related organisms following endonuclease digest of the genome and subsequent analysis of the fragmentation pattern by electrophoresis, mass spectrometry, or other methods known in the art.

9. Methods for the Production of Custom DNA-binding Domains.
[0163] In another aspect, the invention provides rationally-designed DNA-binding proteins that lack endonuclease cleavage activity. The catalytic activity of a rationally-designed meganuclease can be eliminated by mutating ainino acids involved in catalysis (e.g., the inutation of Q47 to E in I-CreI, see Chevalier et al. (2001), Biochem.istry.
43:14015-14026); the mutation of D44 or D145 to N in I-Scel; the mutation of E66 to Q
in I-CeuI; the inutation of D22 to N in I-Msol). The inactivated meganuclease can then be fused to an effector domain from another protein including, but not limited to, a transcription activator (e.g., the GAL4 transactivation domain or the VP 16 transactivation domain), a transcription repressor (e.g., the KRAB domain from the Kruppel protein), a DNA methylase domain (e.g., M.CviPI or M.Sssl), or a histone acetyltransferase domain (e.g., HDAC1 or HDAC2). Chimeric proteins consisting of an engineered DNA-binding domain, most notably an engineered zinc finger domain, and an effector domain are known in the art (see, e.g., Papworth et al. (2006), Gene 366:27-38).

EXAMPLES
[0164] This invention is further illustrated by the following examples, which should not be construed as limiting. Those skilled in the art will recognize, or be able to ascertain, using no more than routine experimentation, numerous equivalents to the specific substances and procedures described herein. Such equivalents are intended to be encoinpassed in the scope of the claims that follow the examples below.
Examples 1-4 below refer specifically to rationally-designed meganucleases based on I-CreI, but rationally-designed meganucleases based on I-Scel, I-Msol, I-Ceul, and other LAGLIDADG meganucleases can be similarly produced and used, as described herein.

Rational Design of Meganucleases Recognizing the HIV-1 TAT Gene 1. Meganuclease Design.
[0165] A pair of meganucleases were designed to recognize and cleave the DNA
site 5'-GAAGAGCTCATCAGAACAGTCA-3' (SEQ ID NO: 15) found in the HIV-1 TAT Gene. In accordance with Table 1, two meganucleases, TAT1 and TAT2, were designed to bind the half-sites 5'-GAAGAGCTC-3' (SEQ ID NO: 16) and 5'-TGACTGTTC-3' (SEQ ID NO: 17), respectively, using the following base contacts (non-WT contacts are in bold):

TAT 1:
Position -9 -8 -7 -6 -5 -4 -3 -2 -1 Base G A A G A G c T C
Contact Residues S32 Y33 N30/ R40 K28 S26/ K24/ Q44 R70 TAT2:
Position -9 -8 -7 -6 -5 -4 -3 -2 -1 Base T G A C T G T T C
Contact Residues C32 R33 N30/ R28/ M66 S26/ Y68 044 R70 [0166] The two enzymes were cloned, expressed in E. coli, and assayed for enzyme activity against the corresponding DNA recognition sequence as described below. In both cases, the rationally-designed meganucleases were found to be inactive.
A second generation of each was then produced in which E80 was mutated to Q to improve contacts with the DNA backbone. The second generation TAT2 enzyme was found to be active against its intended recognition sequence while the second generation TATI enzyme remained inactive. Visual inspection of the wild-type I-Crel co-crystal structure suggested that TATI was inactive due to a steric clash between R40 and K28.
To alleviate this clash, TAT1 variants were produced in which K28 was mutated to an amino acid with a smaller side chain (A, S, T, or C) while maintaining the Q80 mutation.
When these enzyines were produced in E. coli and assayed, the TAT1 variants with S28 and T28 were both found to be active against the intended recognition sequence while maintaining the desired base preference at position -7.

2. Construction of Recombinant Meganucleases.
101671 Mutations for the redesigned I-CreI enzyines were introduced using mutagenic primers in an overlapping PCR strategy. Recombinant DNA fragments of I-Crel generated in a primary PCR were joined in a secondary PCR to produce full-length recoinbinant nucleic acids. All recombinant I-CreI constructs were cloned into pET21 a vectors with a six histidine tag fused at the 3' end of the gene for purification (Novagen Corp., San Diego, CA). All nucleic acid sequences were confinned using Sanger Dideoxynucleotide sequencing (see Sanger et al. (1977), Proc. Natl. Acad. Sci.
USA.
74(12): 5463-7).

101681 Wild-type I-CreI and all engineered meganucleases were expressed and purified using the following method. The constructs cloned into a pET21 a vector were transforined into chemically competent BL21 (DE3) pLysS, and plated on standard 2xYT
plates containing 200 g/inl carbanicillin. Following overnight growth, transforined bacterial colonies were scraped fioin the plates and used to inoculate 50 ml of 2XYT
broth. Cells were grown at 37 C with shaking until they reached an optical density of 0.9 at a wavelength of 600 nin. The growth teinperature was then reduced from 37 C
to 22 C. Protein expression was induced by the addition of 1mM IPTG, and the cells were incubated with agitation for two and a half hours. Cells were then pelleted by centrifugation for 10 min. at 6000 xg. Pellets were resuspended in 1m1 binding buffer (20 mM Tris-HCL, pH 8.0, 500 mM NaC1, 10 mM imidazole) by vortexing. The cells were then disrupted with 12 pulses of sonication at 50% power and the cell debris was pelleted by centrifugation for 15 min. at 14,000 xg. Cell supernatants were diluted in 4 ml binding buffer and loaded onto a 200 l nickel-charged metal-chelating Sepharose column (Pharmacia).
[0169] The column was subsequently washed with 4 ml wash buffer (20 mM Tris-HCI, pH 8.0, 500 mM NaCl, 60 mM imidazole) and with 0.2 ml elution buffer (20 mM
Tris-HCI, pH 8.0, 500 mM NaCI, 400 mM imidazole). Meganuclease enzyines were eluted with an additional 0.6 ml of elution buffer and concentrated to 50-130 l using Vivospin disposable concentrators (ISC, Inc., Kaysville, UT). The enzyines were exchanged into SA buffer (25 mM Tris-HCL, pH 8.0, 100 mM NaCl, 5 mM MgC12, 51nM
EDTA) for assays and storage using Zeba spin desalting colunms (Pierce Biotechnology, Inc., Rockford, IL). The enzyine concentration was determined by absorbance at 280 nm using an extinction coefficient of 23,590 M"Icin"1. Purity and molecular weight of the enzyines was then confirined by MALDI-TOF mass spectrometry.
[0170] Heterodimeric enzymes were produced either by purifying the two proteins independently, and mixing them in vitro or by constructing an artificial operon for tandem expression of the two proteins in E. coli. In the former case, the purified meganucleases were mixed 1:1 in solution and pre-incubated at 42 C for 20 minutes prior to the addition of DNA substrate. In the latter case, the two genes were cloned sequentially into the pET-21 a expression vector using Ndel/EcoRl and EcoRl/HiradllZ
The first gene in the operon ends with two stop codons to prevent read-through errors during transcription. A 12-base pair nucleic acid spacer and a Shine-Dalgarno sequence from the pET21 vector separated the first and second genes in the artificial operon.

3. Cleavage Assays.
[0171] All enzyines purified as described above were assayed for activity by incubation with linear, double-stranded DNA substrates containing the meganuclease recognition sequence. Synthetic oligonucleotides corresponding to both sense and antisense strands of the recognition sequence were annealed and were cloned into the Smal site of the pUC19 plasmid by blunt-end ligation. The sequences of the cloned binding sites were confirmed by Sanger dideoxynucleotide sequencing. All plasmid substrates were linearized with XmnI, Scal or Bpnal concurrently with the meganuclease digest. The enzyine digests contained 5 l 0.05 M DNA substrate, 2.5 l 5 M
recombinant I-Crel meganuclease, 9.5 l SA buffer, and 0.5 l Xmnl, Scal, or Bp77zl.
Digests were incubated at either 37 C, or 42 C for certain meganuclease enzyines, for four hours. Digests were stopped by adding 0.3 mg/ml Proteinase K and 0.5%
SDS, and incubated for one hour at 37 C. Digests were analyzed on 1.5% agarose and visualized by ethidium bromide staining.
[0172] To evaluate meganuclease half-site preference, rationally-designed meganucleases were incubated with a set of DNA substrates corresponding to a perfect palindrome of the intended half-site as well as each of the 27 possible single-base-pair substitutions in the half-site. In this manner, it was possible to detennine how tolerant each enzyme is to deviations from its intended half-site.

4. Recognition Sequence-Specificity.
[0173] Purified recombinant TAT1 and TAT2 meganucleases recognized DNA
sequences that were distinct from the wild-type meganuclease recognition sequence (Fig.
2(B)). The wild-type I-CreI meganuclease cleaves the WT recognition sequence, but cuts neither the intended sequence for TAT1 nor the intended sequence for TAT2.
TAT1 and TAT2, likewise, cut their intended recognition sequences but not the wild-type sequence.
The meganucleases were then evaluated for half-site preference and overall specificity (Fig 3). Wild-type I-CreI was found to be highly tolerant of single-base-pair substitutions in its natural half-site. In contrast, TAT1 and TAT2 were found to be highly-specific and coinpletely intolerant of base substitutions at positions -1, -2, -3, -6, and -8 in the case of TAT1, and positions -1, -2, and -6 in the case of TAT2.

Rational Design of Meganucleases with Altered DNA-Binding Affinity 1. Meganucleases with increased affinity and increased activity.
[0174] The meganucleases CCRl and BRP2 were designed to cleave the half-sites 5'-AACCCTCTC-3' (SEQ ID NO: 18) and 5'-CTCCGGGTC-3' (SEQ ID NO: 19), respectively. These enzymes were produced in accordance with Table 1 as in Example 1:
CCR1:
Position -9 8 -7 -6 -5 -4 -3 -2 -1 Base A A C C C T C T C
Contact Residues N32 Y33 R30/ R28/ E42 G26 K24/ 444 R70 BRP2:
Position -9 -8 -7 -6 -5 -4 '3 -2 -1 Base C T C C G G G T C
Contact Residues S32 C33 R30/ R28/ R42 S26/ R68 Q44 R70 [0175] Both enzymes were expressed in E. coli, purified, and assayed as in Example 1. Both first generation enzyines were found to cleave their intended recognition sequences with rates that were considerably below that of wild-type I-CreI
with its natural recognition sequence. To alleviate this loss in activity, the DNA-binding affinity of CCRl and BRP2 was increased by mutating E80 to Q in both enzymes.
These second-generation versions of CCR1 and BRP2 were found to cleave their intended recognition sequences with substantially increased catalytic rates.

2. Meganucleases with decreased DNA-bindin ag ffinity and decreased activity but increased specificity.
[0176] Wild-type I-CreI was found to be highly-tolerant of substitutions to its half-site (Fig. 3(A)). In an effort to make the enzyme more specific, the lysine at position 116 of the enzyme, which norinally makes a salt-bridge with a phosphate in the DNA
backbone, was mutated to aspartic acid to reduce DNA-binding affinity. This rationally-designed enzyme was found to cleave the wild-type recognition sequence with substantially reduced activity but the recombinant enzyme was considerably more specific than wild-type. The half-site preference of the K116D variant was evaluated as in Exainple 1 and the enzyine was found to be entirely intolerant of deviation from its natural half-site at positions -1, -2, and -3, and displayed at least partial base preference at the remaining 6 positions in the half-site (Fig. 3(B)).

Rationally-Designed Meganuclease uclease Heterodimers 1. Cleavage of non-palindromic DNA sites by meganuclease heterodimers formed in solution.
[0177] Two meganucleases, LAM1 and LAM2, were designed to cleave the half-sites 5'-TGCGGTGTC-3' (SEQ ID NO: 20) and 5'-CAGGCTGTC-3' (SEQ ID NO: 21), respectively. The heterodimer of these two enzymes was expected to recognize the DNA
sequence 5'-TGCGGTGTCCGGCGACAGCCTG-3' (SEQ ID NO: 22) found in the bacteriophage X p05 gene.

LAM 1:
Position -9 -8 -7 -6 -5 -4 -3 -2 -1 Base T G C G T G T c Contact Residues C32 R33 R30/ D28/ R42 Q26 R68 044 R70 LAM2:
Position -9 -8 -7 -6 -5 -4 -3 -2 -1 Base c A G G C T G T C
Contact Residues S32 Y33 E30/ R40 K28/ 026 R68 Q44 R70 [0178] LAM1 and LAM 2 were cloned, expressed in E. coli, and purified individually as described in Example 1. The two enzymes were then mixed 1:1 and incubated at 42 C for 20 minutes to allow them to exchange subunits and re-equilibrate.
The resulting enzyine solution, expected to be a mixture of LAM1 homodimer, homodimer, and LAM1/LAM2 heterodimer, was incubated with three different recognition sequences corresponding to the perfect palindrome of the LAM 1 half-site, the perfect palindrome of the LAM2 half-site, and the non-palindromic hybrid site found in the bacteriophage k genome. The purified LAM1 enzyme alone cuts the LAM1 palindromic site, but neither the LAM2 palindromic site, nor the LAM1/LAM2 hybrid site. Likewise, the purified LAM2 enzyme alone cuts the LAM2 palindromic site but neither the LAM1 palindromic site nor the LAM1/LAM2 hybrid site. The 1:1 mixture of LAMI and LAM2, however, cleaves all three DNA sites. Cleavage of the LAM1/LAM2 hybrid site indicates that two distinct redesigned meganucleases can be mixed in solution to forin a heterodimeric enzyine capable of cleaving a non-palindromic DNA
site.

2. Cleavage of non palindroinic DNA sites by meganuclease heterodimers formed by co-expression.
[0179] Genes encoding the LAM1 and LAM2 enzyines described above were arranged into an operon for simultaneous expression in E. coli as described in Example 1.
The co-expressed enzymes were purified as in Example 1 and the enzyme mixture incubated with the three potential recognition sequences described above. The co-expressed enzyine mixture was found to cleave all three sites, including the LAM1/LAM2 hybrid site, indicating that two distinct rationally-designed meganucleases can be co-expressed to form a heterodimeric enzyme capable of cleaving a non-palindromic DNA site.

3. Preferential cleavage of non-palindromic DNA sites by meganuclease heterodimers with modified protein-protein interfaces.

[0180] For applications requiring the cleavage of non-palindromic DNA sites, it is desirable to promote the fonnation of enzyme heterodiiners while minimizing the foimation of homodimers that recognize and cleave different (palindromic) DNA
sites.
To this end, variants of the LAM1 enzyme were produced in which lysines at positions 7, 57, and 96 were changed to glutamic acids. This enzyme was then co-expressed and purified as in above with a variant of LAM2 in which glutamic acids at positions 8 and 61 were changed to lysine. In this case, formation of the LAM1 homodimer was expected to be reduced due to electrostatic repulsion between E7, E57, and E96 in one monomer and E8 and E61 in the other monomer. Likewise, formation of the LAM2 homodimer was expected to be reduced due to electrostatic repulsion between K7, K57, and K96 on one monomer and K8 and K61 on the other monomer. Conversely, the LAM 1/LAM2 heterodimer was expected to be favored due to electrostatic attraction between E7, E57, and E96 in LAM1 and K8 and K61 in LAM2. When the two meganucleases with modified interfaces were co-expressed and assayed as described above, the hybrid site was found to be cleaved preferentially over the two palindromic sites, indicating that substitutions in the meganuclease protein-protein interface can drive the preferential fonnation of heterodimers.

Additional Meganuclease Heterodimers Which Cleave Ph siologic DNA Sequences 1. Meganuclease heterodimers which cleave DNA sequences relevant to gene therapy.
[0181] A rationally-designed meganuclease heterodimer (ACH1/ACH2) can be produced that cleaves the sequence 5'-CTGGGAGTCTCAGGACAGCCTG-3' (SEQ ID
NO: 23) in the huinan FGFR3 gene, inutations in which cause achondroplasia.
For exainple, a meganuclease was designed based on the I-Crel ineganuclease, as described above, with the following contact residues and recognition sequence half-sites:

ACH 1:

Position -9 -8 -7 -6 -5 -4 -3 -2 -1 Base C T G G G A G T C
Contact Residues D32 C33 E30/ R40/ R42 A26/ R68 044 R70 ACH2:
Position -9 -8 -7 -6 -5 -4 -3 -2 -1 Base c A G G C T G T C
Contact Residues D32 Y33 E30/ R40 K28/ 026 R68 Q44 R70 [0182] A rationally-designed meganuclease heterodimer (HGH1/HGH2) can be produced that cleaves the sequence 5'-CCAGGTGTCTCTGGACTCCTCC-3' (SEQ ID
NO: 24) in the promoter of the Human Growth Hormone gene. For example, a meganuclease was designed based on the I-CreI meganuclease, as described above, with the following contact residues and recognition sequence half-sites:

HGH 1:

Position -9 -8 -7 -6 -5 -4 -3 -2 -1 Base C C A G G T T c Contact Residues D32 C33 N301 R40/ R42 ~26 R68 044 R70 HGH2:

Position -9 -8 -7 -6 -5 -4 -3 -2 -1 Base G G A G G A G T C
Contact Residues K32 R33 N30/ R40/ R42 A26 R68 044 R70 [0183] A rationally-designed meganuclease heterodimer (CF1/CF2) can be produced that cleaves the sequence 5'-GAAAATATCATTGGTGTTTCCT-3' (SEQ ID
NO: 25) in the AF508 allele of the human CFTR gene. For example, a meganuclease was designed based on the I-CreI meganuclease, as described above, with the following contact residues and recognition sequence half-sites:

CF1:
Position -9 -8 -7 -6 -5 -4 -3 -2 -1 Base G A A A A T A T c Contact Residues S32 Y33 N30/ ,040 K28 026 H68/ Q44 R70 CF2:
Position -9 -8 -7 -6 -5 -4 -3 -2 -1 Base A G G A A A C A C
Contact Residues N32 R33 E30/ ~40 K28 A26 Y68/ T44 R70 [0184] A rationally-designed meganuclease heterodimer (CCR1/CCR2) can be produced that cleaves the sequence 5'-AACCCTCTCCAGTGAGATGCCT-3' (SEQ ID
NO: 26) in the human CCR5 gene (an HIV co-receptor). For example, a meganuclease was designed based on the I-CreI meganuclease, as described above, with the following contact residues and recognition sequence half-sites:

CCR1:

Position -9 -8 -7 -5 -4 -3 -2 -1 Base A A C C C T C T C
Contact Residues N32 Y33 R30/ E40/ E42 026 Y68/ 044 R70 CCR2:
Position -9 -8 -7 -6 -5 -4 -3 -2 -1 Base A G G C A T c T c Contact Residues N32 R33 E30/ E40 K28 026 Y68/ 044 R70 [0185] A rationally-designed meganuclease heterodimer (MYD1/MYD2) can be produced that cleaves the sequence 5'-GACCTCGTCCTCCGACTCGCTG-3' (SEQ ID
NO: 27) in the 3' untranslated region of the huinan DM kinase gene. For example, a meganuclease was designed based on the I-CreI meganuclease, as described above, with the following contact residues and recognition sequence half-sites:

MYD 1:
Position -9 -8 -7 -6 -5 -4 -3 -2 Base G A C C T C G T C
Contact Residues S32 Y33 R30/ E40/ K66 216/ R68 044 R70 MYD 1:
Position -9 -8 -7 -6 -5 -4 -3 -2 -1 Base C A G c G A G T c Contact Residues S32 Y33 E30/ E40/ R42 A26 R68 Q44 R70 2. Meganuclease heterodimers which cleave DNA sequences in pathogen genomes.
[0186] A rationally-designed meganuclease heterodimer (HSV1/HSV2) can be produced that cleaves the sequence 5'-CTCGATGTCGGACGACACGGCA-3' (SEQ ID
NO: 28) in the UL36 gene of Herpes Siinplex Virus-1 and Herpes Simplex Virus-2. For example, a meganuclease was designed based on the I-CreI meganuclease, as described above, with the following contact residues and recognition sequence half-sites:

HSV1:
Position -9 -8 -7 -6 -5 -4 -3 -2 -1 Base C T C G A T G T C
Contact Residues S32 C33 R30/ R40/ ,042/ 026 R68 044 R70 HSV2:
Position -9 -8 -7 -6 -5 -4 -3 -2 -1 Base T G C C G T G T C
Contact Residues C32 R33 R30/ E40/ R42 026 R68 44 R70 [01871 A rationally-designed meganuclease heterodimer (ANT1/ANT2) can be produced that cleaves the sequence 5'-ACAAGTGTCTATGGACAGTTTA-3' (SEQ ID
NO: 29) in the Bacillus anthracis genome. For example, a meganuclease was designed based on the I-CreI meganuclease, as described above, with the following contact residues and recognition sequence half-sites:
ANT 1:
Position -9 -8 -7 -6 -5 -4 -3 -2 -1 Base A C A A G T G T C
Contact Residues N32 C33 N30/ 40/ R42 026 R68 044 R70 ANT2:
Position -9 -8 -7 -6 -5 -4 -3 -2 -1 Base T A A A C T G T C
Contact Residues C32 Y33 N30/ 40 E42 026 R68 044 R70 [0188] A rationally-designed meganuclease heterodimer (POX1/POX2) can be produced that cleaves the sequence 5'-AAAACTGTCAAATGACATCGCA-3' (SEQ ID
NO: 30) in the Variola (smallpox) virus gp009 gene. For example, a meganuclease was designed based on the I-CreI meganuclease, as described above, with the following contact residues and recognition sequence half-sites:
POX1:
Position -9 -8 -7 -6 -5 -4 -3 -2 -1 Base A A A A C T G T C
Contact Residues N32 C33 N30/ QO K28 ~26 R68 044 R70 POX2:
Position -9 -8 -7 -6 -5 -4 -3 -2 -1 Base T G C G A T G T C
Contact Residues C32 R33 R30/ R40 C28/ ~26 R68 ~44 R70 E38 ~L42 [0189] A rationally-designed meganuclease homodimer (EBB1/EBB1) can be produced that cleaves the pseudo-palindromic sequence 5'-CGGGGTCTCGTGCGAGGCCTCC-3' (SEQ ID NO: 31) in the Epstein-BaiT Virus BALF2 gene. For example, a meganuclease was designed based on the I-CreI
meganuclease, as described above, with the following contact residues and recognition sequence half-sites:

EBB 1:
Position -9 -8 -7 -6 -5 -4 -3 -2 -1 Base C G G G G T C T C
Contact Residues S32 R33 D30/ R40/ R42 026 Y68/ 044 R70 EBB 1:

Position -9 -8 -7 -6 -5 -4 -3 -2 -1 Base G G A G G C C T C
Contact Residues S32 R33 D30/ R40/ R42 (26 Y68/ 044 R70 3. Meganuclease heterodiiners which cleave DNA sequences in plant genoines.
[0190) A rationally-designed meganuclease heterodimer (GLA1/GLA2) can be produced that cleaves the sequence 5'-CACTAACTCGTATGAGTCGGTG-3' (SEQ ID
NO: 32) in the Arabidopsis thalianna GL2 gene. For example, a meganuclease was designed based on the I-CreI meganuclease, as described above, with the following contact residues and recognition sequence half-sites:

GLA 1:

Position -9 -8 -7 -6 -5 -4 -3 -2 -1 Base c A c T A A c T C
Contact Residues S32 Y33 R30/ S40/ K28 A26/ Y68/ 44 R70 GLA2:
Position -9 -8 -7 -6 -5 -4 -3 -2 -1 Base C A C C G A C T C
Contact Residues S32 Y33 R30/ E40/ R42 A26 Y68/ 44 R70 [01911 A rationally-designed meganuclease heterodimer (BRP1/BRP2) can be produced that cleaves the sequence 5'-TGCCTCCTCTAGAGACCCGGAG-3' (SEQ ID
NO: 33) in the Arabi.dopsis thalianna BPI gene. For example, a meganuclease was designed based on the I-Crel meganuclease, as described above, with the following contact residues and recognition sequence half-sites:

BRP 1:

Position -9 -8 -7 -6 -5 -4 -3 -2 -1 Base T G C C T C C T C
Contact Residues C32 R33 R30/ R28/ K66 026/ Y68/ 044 R7Q

BRP2:
Position -9 -8 -7 -6 -5 -4 -3 -2 -1 Base C T C C G G G T c Contact Residues S32 C33 R30/ E40/ R42 S26 R68 044, R70 [0192] A rationally-designed meganuclease heterodimer (MGCI/MGC2) can be produced that cleaves the sequence 5'-TAAAATCTCTAAGGTCTGTGCA-3' (SEQ ID
NO: 34) in the Nicotiasza tabacum Magnesium Chelatase gene. For example, a meganuclease was designed based on the I-CreI meganuclease, as described above, with the following contact residues and recognition sequence half-sites:

MGCI:
Position -9 -8 -7 -6 -5 -4 -3 -2 =1 Base T A A A A T c T C
Contact Residues C32 Y33 N30/ 040/ K28 Q26 Y68/ 044 R70 MGC2:
Position -9 -8 -7 -6 -5 -4 -3 -2 -1 Base T G C A C A G A C
Contact Residues S32 R33 R30/ 040 K28 A26 R68 T44 R70 [0193] A rationally-designed meganuclease heterodiiner (CYP/HGH2) can be produced that cleaves the sequence 5'-CAAGAATTCAAGCGAGCATTAA-3' (SEQ ID
NO: 35) in the Nicotiana tabacz-cin CYP82E4 gene. For exainple, a rneganuclease was designed based on the I-CreI meganuclease, as described above, with the following contact residues and recognition sequence half-sites:
CYP:
Position -9 -8 -7 -6 -5 -4 -3 -2 -1 Base C A A G A A T T C
Contact Residues D32 Y33 N30/ R40/ K28 ,077/ Y68 ,044 R70 HGH2:
Position -9 -8 -7 -6 -5 -4 -3 -2 -1 Base T T A A T G C T C
Contact Residues S32 C33 N30/ 040 K66 R77/ Y68 044 R70 4. Meganuclease heterodiiners which cleave DNA sequences in yeast genomes.
[0194] A rationally-designed meganuclease heterodimer (UR.A1/URA2) can be produced that cleaves the sequence 5'-TTAGATGACAAGGGAGACGCAT-3' (SEQ ID
NO: 36) in the Saccharoinyces cerevisiae URA3 gene. For example, a meganuclease was designed based on the I-Crel meganuclease, as described above, with the following contact residues and recognition sequence half-sites:
URA1:
Position -9 -8 -7 -6 -5 -4 -3 -2 -1 Base T T A G A T G A C
Contact Residues S32 C33 N30/ R40 K28 026 R68 T44 R70 URA2:

Position -9 -8 -7 -6 -5 -4 -3 -2 -1 Base A T G C G T C T C
Contact Residues N32 C33 E30/ E40/ R42 Q26 Y68/ 044 R70 5. Recognition Sequence SpecificitY.
[0195] The rationally-designed meganucleases outlined above in this Example were cloned, expressed in E. coli, and purified as in Example 1. Each purified meganuclease was then mixed 1:1 with its corresponding heterodimerization partner (e.g., ACH1 with ACH2, HGHl with HGH2, etc.) and incubated with a linearized DNA
substrate containing the intended non-palindroinic DNA recognition sequence for each meganuclease heterodiiner. As shown in Figure 3, each rationally-designed meganuclease heterodimer cleaves its intended DNA site.

Claims (48)

1. A recombinant meganuclease having altered specificity for at least one recognition sequence half-site relative to a wild-type I-CreI meganuclease, comprising:
a polypeptide having at least 85% sequence similarity to residues 2-153 of the I-CreI meganuclease of SEQ ID NO: 1; and having specificity for a recognition sequence half-site which differs by at least one base pair from a half-site within an I-CreI meganuclease recognition sequence selected from the group consisting of SEQ ID NO: 2, SEQ ID NO: 3, SEQ ID NO: 4 and SEQ
ID
NO: 5;
wherein said recombinant meganuclease comprises at least one modification of Table 1 which is not an excluded modification.
2. A recombinant meganuclease having altered specificity for at least one recognition sequence half-site relative to a wild-type I-MsoI meganuclease, comprising:
a polypeptide having at least 85% sequence similarity to residues 6-160 of the I-MsoI meganuclease of SEQ ID NO: 6; and having specificity for a recognition sequence half-site which differs by at least one base pair from a half-site within an I-MsoI meganuclease recognition sequence selected from the group consisting of SEQ ID NO: 7 and SEQ ID NO: 8;
wherein said recombinant meganuclease comprises at least one modification of Table 2 which is not an excluded modification.
3. A recombinant meganuclease having altered specificity for a recognition sequence relative to a wild-type I-SceI meganuclease, comprising:
a polypeptide having at least 85% sequence similarity to residues 3-186 of the I-SceI meganuclease of SEQ ID NO: 9; and having specificity for a recognition sequence which differs by at least one base pair from an I-SceI meganuclease recognition sequence of SEQ ID NO: 10 and SEQ
ID
NO: 11;

wherein said recombinant meganuclease comprises at least one modification of Table 3 which is not an excluded modification.
4. A recombinant meganuclease having altered specificity for at least one recognition sequence half-site relative to a wild-type I-CeuI meganuclease, coinprising:
a polypeptide having at least 85% sequence similarity to residues 5-211 of the I-CeuI meganuclease of SEQ ID NO: 12; and having specificity for a recognition sequence half-site which differs by at least one base pair from a half-site within an I-CeuI meganuclease recognition sequence selected from the group consisting of SEQ ID NO: 13 and SEQ ID NO: 14;
wherein said recombinant meganuclease comprises at least one modification of Table 4 which is not an excluded modification.
5. A recombinant meganuclease having altered specificity for at least one recognition sequence half-site relative to a wild-type I-CreI meganuclease, comprising:
a polypeptide having at least 85% sequence similarity to residues 2-153 of the I-CreI meganuclease of SEQ ID NO: 1; and having specificity for a recognition sequence half-site which differs by at least one base pair from a half-site within an I-CreI meganuclease recognition sequence selected from the group consisting of SEQ ID NO: 2, SEQ ID NO: 3, SEQ ID NO: 4 and SEQ
ID
NO: 5;
wherein:
(1) specificity at position -1 has been altered:
(a) to a T on a sense strand by a modification selected from the group consisting of Q70, C70, L70, Y75, Q75, H75, H139, Q46 and H46;
(b) to an A on a sense strand by a modification selected from the group consisting of Y75, L75, C75, Y139, C46 and A46;
(c) to a G on a sense strand by a modification selected from the group consisting of K70, E70, E75, E46 and D46;
(d) to a C on a sense strand by a modification selected from the group consisting of H75, R75, H46, K46 and R46; or (e) to any base on a sense strand by a modification selected from the group consisting of G70, A70, S70 and G46; and/or (2) specificity at position -2 has been altered:
(a) to an A on a sense strand by a modification selected from the group consisting of Q70, T44, A44, V44, I44, L44, and N44;
(b) to a C on a sense strand by a modification selected from the group consisting of E70, D70, K44 and R44;
(c) to a G on a sense strand by a modification selected from the group consisting of H70, D44 and E44; or (d) to an A or T on a sense strand by a modification comprising C44;
and/or (3) specificity at position -3 has been altered:
(a) to an A on a sense strand by a modification selected from the group consisting of Q68 and C24;
(b) to a C on a sense strand by a modification selected from the group consisting of E68, F68, K24 and R24;
(c) to a T on a sense strand by a modification selected from the group consisting of M68, C68, L68 and F68;
(d) to an A or C on a sense strand by a modification comprising H68;
(e) to a C or T on a sense strand by a modification comprising Y68; or (f) to a G or T on a sense strand by a modification comprising K68; and/or (4) specificity at position -4 has been altered:
(a) to a C on a sense strand by a modification selected from the group consisting of E77 and K26;
(b) to a G on a sense strand by a modification selected from the group consisting of E26 and R77;
(c) to a C or T on a sense strand by a modification coinprising S77; or (d) to a any base on a sense strand by a modification comprising S26;
and/or (5) specificity at position -5 has been altered:
(a) to a C on a sense strand by a modification coinprising E42;

(b) to a G on a sense strand by a modification comprising R42;
(c) to an A or G on a sense strand by a modification selected from the group consisting of C28 and Q42; or (d) to any base on a sense strand by a modification of selected from the group consisting of M66 and K66; and/or (6) specificity at position -6 has been altered:
(a) to a T on a sense strand by a modification selected from the group consisting of C40, I40, V40, C79, I79, V79, and Q28;
(b) to a C on a sense strand by a modification selected from the group consisting of E40 and R28; or (c) to a G on a sense strand by a modification comprising R40; and/or (7) specificity at position -7 has been altered:
(a) to a C on a sense strand by a modification selected from the group consisting of E38, K30 and R30;
(b) to a G on a sense strand by a modification selected from the group consisting of K38, R38 and E30;
(c) to a T on a sense strand by a modification selected from the group consisting of I38 and L38; or (d) to an A or G on a sense strand by a modification comprising C38; or (e) to any base on a sense strand by a modification selected from the group consisting of H38, N38 and Q30; and/or (8) specificity at position -8 has been altered:
(a) to a T on a sense strand by a modification selected from the group consisting of L33, V33, I33, F33 and C33;
(b) to a C on a sense strand by a modification selected from the group consisting of E33 and D33;
(c) to a G on a sense strand by a modification consisting of K33;
(d) to an A or C on a sense strand by a modification comprising R32; or (e) to an A or G on a sense strand by a modification comprising R33;
and/or (9) specificity at position -9 has been altered:

(a) to a C on a sense strand by a modification comprising E32;
(b) to a G on a sense strand by a modification selected from the group consisting of R32 and K32;
(c) to a T on a sense strand by a modification selected from the group consisting of L32, V32, A32 and C32;
(d) to a C or T on a sense strand by a modification selected from the group consisting of D32 and I32; or (e) to any base on a sense strand by a modification selected from the group consisting of S32, N32, H32, Q32 and T32.
6. A recombinant meganuclease having altered specificity for at least one recognition sequence half-site relative to a wild-type I-MsoI meganuclease, comprising:
a polypeptide having at least 85% sequence similarity to residues 6-160 of the I-MsoI meganuclease of SEQ ID NO: 6; and having specificity for a recognition sequence half-site which differs by at least one base pair from a half-site within an I-MsoI meganuclease recognition sequence selected from the group consisting of SEQ ID NO: 7 and SEQ ID NO: 8;
wherein:
(1) specificity at position -1 has been altered:
(a) to an A on a sense strand by a modification selected from the group consisting of K75, Q77, A49, C49 and K79;
(b) to a T on a sense strand by a modification selected from the group consisting of C77, L77 and Q79; or (c) to a G on a sense strand by a modification selected from the group consisting of K77, R77, E49 and E79; and/or (2) specificity at position -2 has been altered:
(a) to an A on a sense strand by a modification selected from the group consisting of Q75, K81, C47, I 47 and L47;
(b) to a C on a sense strand by a modification selected from the group consisting of E75, D75, R47, K47, K81 and R81; or (c) to a G on a sense strand by a modification selected from the group consisting of K75, E47 and E81; and/or (3) specificity at position -3 has been altered:
(a) to an A on a sense strand by a modification selected from the group consisting of Q72, C26, L26, V26, A26 and 126;
(b) to a C on a sense strand by a modification selected from the group consisting of E72, Y72, H26, K26 and R26; or (c) to a T on a sense strand by a modification selected from the group consisting of K72, Y72 and H26; and/or (4) specificity at position -4 has been altered:
(a) to a T on a sense strand by a modification selected from the group consisting of K28, K83 and Q28;
(b) to a G on a sense strand by a modification selected from the group consisting of R83 and K83; or (c) to an A on a sense strand by a modification selected from the group consisting of K28 and Q83; and/or (5) specificity at position -5 has been altered:
(a) to a G on a sense strand by a modification selected from the group consisting of R45 and E28;
(b) to a T on a sense strand by a modification comprising Q28; or (c) to a C on a sense strand by a modification comprising R28; and/or (6) specificity at position -6 has been altered:
(a) to a T on a sense strand by a modification selected from the group consisting of K43, V85, L85 and Q30;
(b) to a C on a sense strand by a modification selected from the group consisting of E43, E85, K30 and R30; or (c) to a G on a sense strand by a modification selected from the group consisting of R43, K43, K85, R85, E30 and D30; and/or (7) specificity at position -7 has been altered:
(a) to a C on a sense strand by a modification selected from the group consisting of E32 and E41;
(b) to a G on a sense strand by a modification selected from the group consisting of R32, R41 and K41;
(c) to a T on a sense strand by a modification selected from the group consisting of K32, M41, L41 and I41; and/or (8) specificity at position -8 has been altered:
(a) to a T on a sense strand by a modification selected from the group consisting of K32 and K35;

(b) to a C on a sense strand by a modification comprising E32; or (c) to a G on a sense strand by a modification consisting of K32, K35 and R35; and/or (9) specificity at position -9 has been altered:
(a) to an A on a sense strand by a modification selected from the group consisting of N34 and H34;
(b) to a T on a sense strand by a modification selected from the group consisting of S34, C34, V34, T34 and A34; or (c) to a G on a sense strand by a modification selected from the group consisting of K34, R34 and H34.
7. A recombinant meganuclease having altered specificity for a recognition sequence relative to a wild-type I-SceI meganuclease, comprising:
a polypeptide having at least 85% sequence similarity to residues 3-186 of the I-SceI meganuclease of SEQ ID NO: 9; and having specificity for a recognition sequence which differs by at least one base pair from an I-SceI meganuclease recognition sequence of SEQ ID NO: 10 and SEQ
ID
NO: 11;
wherein:
(1) specificity at position 4 has been altered:
(a) to an A on a sense strand by a modification comprising K50;
(b) to a T on a sense strand by a modification selected from the group consisting of K57, M57 and Q50; or (c) to a G on a sense strand by a modification selected from the group consisting of E50, R57 and K57; and/or (2) specificity at position 5 has been altered:
(a) to an A on a sense strand by a modification selected from the group consisting of K48, Q102;
(b) to a G on a sense strand by a modification selected from the group consisting of E48, K102 and R102; or (c) to a T on a sense strand by a modification selected from the group consisting of Q48, C102, L102 and V102; and/or (3) specificity at position 6 has been altered:
(a) to an A on a sense strand by a modification comprising K59;
(b) to a C on a sense strand by a modification selected from the group consisting of R59 and K59; or (b) to a G on a sense strand by a modification selected from the group consisting of K84 and E59; and/or (4) specificity at position 7 has been altered:
(a) to a C on a sense strand by a modification selected from the group consisting of R46, K46 and E86;
(b) to a G on a sense strand by a modification selected from the group consisting of K86, R86 and E46; or (c) to an A on a sense strand by a modification selected from the group consisting of C46, L46 and V46; and/or (5) specificity at position 8 has been altered:
(a) to a C on a sense strand by a modification selected from the group consisting of E88, R61 and H61;
(b) to a T on a sense strand by a modification selected from the group consisting of K88, Q61 and H61; or (c) to an A on a sense strand by a modification selected from the group consisting of K61, S61, V61, A61 and L61; and/or (6) specificity at position 9 has been altered:
(a) to an A on a sense strand by a modification selected from the group consisting of C98, V98 and L98;
(b) to a C on a sense strand by a modification selected from the group consisting of R98 and K98; or (c) to a G on a sense strand by a modification selected from the group consisting of E98 and D98; and/or (7) specificity at position 10 has been altered:
(a) to a C on a sense strand by a modification selected from the group consisting of K96 and R96;
(b) to a G on a sense strand by a modification selected from the group consisting of D96 and E96; or (c) to an A on a sense strand by a modification selected from the group consisting of C96 and A96; and/or (8) specificity at position 11 has been altered:
(a) to a T on a sense strand by a modification comprising Q90;
(b) to a C on a sense strand by a modification selected from the group consisting of K90 and R90; or (c) to a G on a sense strand by a modification comprising E90; and/or (9) specificity at position 12 has been altered:
(a) to an A on a sense strand by a modification comprising Q193;
(b) to a C on a sense strand by a modification selected from the group consisting of E165, E193 and D193; or (c) to a G on a sense strand by a modification selected from the group consisting of K165 and R165; and/or (10) specificity at position 13 has been altered:
(a) to a T on a sense strand by a modification selected from the group consisting of Q193, C163 and L163;
(b) to a G on a sense strand by a modification selected from the group consisting of E193, D193, K163 and R192; or (c) to an A on a sense strand by a modification selected from the group consisting of C193 and L193; and/or (11) specificity at position 14 has been altered:
(a) to a T on a sense strand by a modification selected from the group consisting of K161 and Q192;

(b) to an A on a sense strand by a modification selected from the group consisting of L192 and C192;

(c) to a G on a sense strand by a modification selected from the group consisting of K147, K161, R161, R197, D192 and E192; or (d) to a T on a sense strand by a modification selected from the group consisting of K161 and Q192; and/or (12) specificity at position 15 has been altered:
(a) to a T on a sense strand by a modification selected from the group consisting of C151, L151 and K151;
(b) to a G on a sense strand by a modification comprising K151; or (c) to a C on a sense strand by a modification comprising E151; and/or (13) specificity at position 17 has been altered:
(a) to a T on a sense strand by a modification selected from the group consisting of G152 and Q150;
(b) to a C on a sense strand by a modification selected from the group consisting of K152 and K150; or (c) to a G on a sense strand by a modification selected from the group consisting of N152, S152, D152, D150 and E150; and/or (14) specificity at position 18 has been altered:
(a) to a T on a sense strand by a modification selected from the group consisting of H155 and Y155;
(b) to a C on a sense strand by a modification selected from the group consisting of R155 and K155; or (c) to an A on a sense strand by a modification selected from the group consisting of K155 and C155.
8. A recombinant meganuclease having altered specificity for at least one recognition sequence half-site relative to a wild-type I-CeuI meganuclease, comprising:
a polypeptide having at least 85% sequence similarity to residues 5-211 of the I-CeuI meganuclease of SEQ ID NO: 12; and having specificity for a recognition sequence half-site which differs by at least one base pair from a half-site within an I-CeuI meganuclease recognition sequence selected from the group consisting of SEQ ID NO: 13 and SEQ ID NO: 14;
wherein:
(1) specificity at position -1 has been altered:
(a) to an A on a sense strand by a modification selected from the group consisting of C92, A92 and V92;
(b) to a T on a sense strand by a modification selected from the group consisting of Q116 and Q92; or (c) to a G on a sense strand by a modification selected from the group consisting of E116 and E92; and/or (2) specificity at position -2 has been altered:
(a) to an A on a sense strand by a modification selected from the group consisting of Q117, C90, L90 and V90;
(b) to a G on a sense strand by a modification selected from the group consisting of K117, R124, K124, E124, E90 and D90; or (c) to a C on a sense strand by a modification selected from the group consisting of E117, D117, R174, K124, K90, R90 and K68; and/or (3) specificity at position -3 has been altered:
(a) to an A on a sense strand by a modification selected from the group consisting of C70, V70, T70, L70 and K70;
(b) to a T on a sense strand by a modification comprising Q70;
(b) to a C on a sense strand by a modification consisting of K70; and/or (4) specificity at position -4 has been altered:
(a) to a C on a sense strand by a modification selected from the group consisting of E126, D126, R88, K88 and K72;
(b) to a T on a sense strand by a modification selected from the group consisting of K126, L126 and Q88; or (c) to an A on a sense strand by a modification selected from the group consisting of Q126, N126, K88, L88, C88, C72, L72 and V72; and/or (5) specificity at position -5 has been altered:
(a) to a G on a sense strand by a modification selected from the group consisting of E74, K128, R128 and E128;
(b) to a T on a sense strand by a modification selected from the group consisting of C128, L128, V128 and T128; or (c) to an A on a sense strand by a modification selected from the group consisting of C74, L74, V74 and T74; and/or (6) specificity at position -6 has been altered:
(a) to a T on a sense strand by a modification selected from the group consisting of K86, C86 and L86;
(b) to a C on a sense strand by a modification selected from the group consisting of D86, E86, R84 and K84; or (c) to a G on a sense strand by a modification selected from the group consisting of K128, R128, R86, K86 and E84; and/or (7) specificity at position -7 has been altered:
(a) to a C on a sense strand by a modification selected from the group consisting of R76, K76 and H76;
(b) to a G on a sense strand by a modification selected from the group consisting of E76 and R84; or (c) to a T on a sense strand by a modification consisting of H76 and Q76;
and/or (8) specificity at position -8 has been altered:
(a) to an A on a sense strand by a modification selected from the group consisting of Y79, R79 and Q76;
(b) to a C on a sense strand by a modification selected from the group consisting of D79, E79, D76 and E76; or (c) to a G on a sense strand by a modification selected from the group consisting of R79, K79, K76 and R76; and/or (9) specificity at position -9 has been altered:
(a) to a T on a sense strand by a modification selected from the group consisting of K78, V78, L78, C78 and T78;
(b) to a C on a sense strand by a modification selected from the group consisting of D78 and E78; or (c) to a G on a sense strand by a modification selected from the group consisting of R78, K78 and H78.
9. A recombinant meganuclease having altered binding affinity for double-stranded DNA relative to a wild-type I-CreI meganuclease, comprising:
a polypeptide having at least 85% sequence similarity to residues 2-153 of the I-CreI meganuclease of SEQ ID NO: 1;
wherein DNA-binding affinity has been increased by at least one modification corresponding to a substitution selected from the group consisting of:
(a) substitution of E80, D137, I81, L112, P29, V64 or Y66 with H, N, Q, S, T, K
or R; or (b) substitution of T46, T140 or T143 with K or R.
10. A recombinant meganuclease having altered binding affinity for double-stranded DNA relative to a wild-type I-CreI meganuclease, comprising:
a polypeptide having at least 85% sequence similarity to residues 2-153 of the I-CreI meganuclease of SEQ ID NO: 1;
wherein DNA-binding affinity has been decreased by at least one modification corresponding to a substitution selected from the group consisting of:
(a) substitution of K34, K48, R51, K82, K116 or K139 with H, N, Q, S, T, D or E;
or (b) substitution of I81, L112, P29, V64, Y66, T46, T140 or T143 with D or E.
11. A recombinant meganuclease having altered binding affinity for double-stranded DNA relative to a wild-type I-MsoI meganuclease, comprising:
a polypeptide having at least 85% sequence similarity to residues 6-160 of the I-MsoI meganuclease of SEQ ID NO: 6;
wherein DNA-binding affinity has been increased by at least one modification corresponding to a substitution selected from the group consisting of:
(a) substitution of E147,185, G86 or Y118 with H, N, Q, S, T, K or R; or (b) substitution of Q41, N70, S87, T88, H89, Q122, Q139, S150 or N152 with K

or R.
12. A recombinant meganuclease having altered binding affinity for double-stranded DNA relative to a wild-type I-MsoI meganuclease, coinprising:
a polypeptide having at least 85% sequence similarity to residues 6-160 of the I-MsoI meganuclease of SEQ ID NO: 6;
wherein DNA-binding affinity has been decreased by at least one modification corresponding to a substitution selected from the group consisting of:
(a) substitution of K36, R51, K123, K143 or R144 with H, N, Q, S, T, D or E;
or (b) substitution of 185, G86, Y118, Q41, N70, S87, T88, H89, Q122, Q139, S150 or N152 with D or E.
13. A recombinant meganuclease having altered binding affinity for double-stranded DNA relative to a wild-type I-SceI meganuclease, comprising:
a polypeptide having at least 85% sequence similarity to residues 3-186 of the I-SceI meganuclease of SEQ ID NO: 9;
wherein DNA-binding affinity has been increased by at least one modification corresponding to a substitution selected from the group consisting of:
(a) substitution of D201, L19, L80, L92, Y151, Y188,I191, Y199 or Y222 with H, N, Q, S, T, K or R; or (b) substitution of N15, N17, S81, H84, N94, N120, T156, N157, S159, N163, Q165, S166, N194 or S202 with K or R.
14. A recombinant meganuclease having altered binding affinity for double-stranded DNA relative to a wild-type I-SceI meganuclease, comprising:
a polypeptide having at least 85% sequence similarity to residues 3-186 of the I-SceI meganuclease of SEQ ID NO: 9;
wherein DNA-binding affinity has been decreased by at least one modification corresponding to a substitution selected from the group consisting of:
(a) substitution of K20, K23, K63, K122, K148, K153, K190, K193, K195 or K223 with H, N, Q, S, T, D or E; or (b) substitution of L19, L80, L92, Y151, Y188, I191, Y199, Y222, N15, N17, S81, H84, N94, N120, T156, N157, S159, N163, Q165, S166, N194 or S202 with D
or E.
15. A recombinant meganuclease having altered binding affinity for double-stranded DNA relative to a wild-type I-CeuI meganuclease, comprising:
a polypeptide having at least 85% sequence similarity to residues 5-211 of the I-CeuI meganuclease of SEQ ID NO: 12;
wherein DNA-binding affinity has been increased by at least one modification corresponding to a substitution selected from the group consisting of:
(a) substitution of D25 or D128 with H, N, Q, S, T, K or R; or (b) substitution of S68, N70, H94, S l 17, N120, N129 or H172 with K or R.
16. A recombinant meganuclease having altered binding affinity for double-stranded DNA relative to a wild-type I-CeuI meganuclease, comprising:
a polypeptide having at least 85% sequence similarity to residues 5-211 of the I-CeuI meganuclease of SEQ ID NO: 12;
wherein DNA-binding affinity has been decreased by at least one modification corresponding to a substitution selected from the group consisting of:
(a) substitution of K21, K28, K31, R112, R114 or R130 with H, N, Q, S, T, D or E; or (b) substitution of S68, N70, H94, S117, N120, N129 or H172 with D or E.
17. A recombinant meganuclease monomer having altered affinity for dimer formation with a reference meganuclease monomer, comprising:
a polypeptide having at least 85% sequence similarity to residues 2-153 of the I-CreI meganuclease of SEQ ID NO: 1;
wherein affinity for dimer formation has been altered by at least one modification corresponding to a substitution selected from the group consisting of:
(a) substitution of K7, K57 or K96 with D or E; or (b) substitution of E8 or E61 with K or R.
18. A recombinant meganuclease heterodimer comprising:
a first polypeptide having at least 85% sequence similarity to residues 2-153 of the I-CreI meganuclease of SEQ ID NO: 1;
wherein affinity for dimer formation has been altered by at least one modification corresponding to a substitution selected from the group consisting of:
(a) substitution of K7, K57 or K96 with D or E; and a second polypeptide having at least 85% sequence similarity to residues 2-153 of the I-CreI meganuclease of SEQ ID NO: 1;
wherein affinity for dimer formation has been altered by at least one modification corresponding to a substitution selected from the group consisting of:
(b) substitution of E8 or E61 with K or R.
19. A recombinant meganuclease monomer having altered affinity for dimer formation with a reference meganuclease monomer, comprising:
a polypeptide having at least 85% sequence similarity to residues 6-160 of the I-MsoI meganuclease of SEQ ID NO: 6;
wherein affinity for dimer formation has been altered by at least one modification corresponding to a substitution selected from the group consisting of:
(a) substitution of R302 with D or E; or (b) substitution of D20, E11 or Q64 with K or R.
20. A recombinant meganuclease heterodimer comprising:
a first polypeptide having at least 85% sequence similarity to residues 6-160 of the I-MsoI meganuclease of SEQ ID NO: 6;
wherein affinity for dimer formation has been altered by at least one modification corresponding to a substitution selected from the group consisting of:
(a) substitution of R302 with D or E; and a second polypeptide having at least 85% sequence similarity to residues 6-160 of the I-MsoI meganuclease of SEQ ID NO: 6;
wherein affinity for dimer formation has been altered by at least one modification corresponding to a substitution selected from the group consisting of:

(b) substitution of D20, E11 or Q64 with K or R.
21. A recombinant meganuclease monomer having altered affinity for dimer formation with a reference meganuclease monomer, comprising:
a polypeptide having at least 85% sequence similarity to residues 5-211 of the I-CeuI meganuclease of SEQ ID NO: 12;
wherein affinity for dimer formation has been altered by at least one modification corresponding to a substitution selected from the group consisting of:
(a) substitution of R93 with D or E; or (b) substitution of E152 with K or R.
22. A recombinant meganuclease heterodimer comprising:
a first polypeptide having at least 85% sequence similarity to residues 5-211 of the I-CeuI meganuclease of SEQ ID NO: 12;
wherein affinity for dimer formation has been altered by at least one modification corresponding to a substitution selected from the group consisting of:
(a) substitution of R93 with D or E; and a second polypeptide having at least 85% sequence similarity to residues 5-211 of the I-CeuI meganuclease of SEQ ID NO: 12;
wherein affinity for dimer formation has been altered by at least one modification corresponding to a substitution selected from the group consisting of:
(b) substitution of E152 with K or R.
23. A recombinant meganuclease monomer or heterodimer of claim 17 or claim 18, further comprising:
at least one modification selected from Table 1;
wherein said modification is not an excluded modification.
24. A recombinant meganuclease monomer or heterodimer of claim 19 or claim 20, further comprising:
at least one modification selected from Table 2;

wherein said modification is not an excluded modification.
25. A recombinant meganuclease monomer or heterodimer of claim 21 or claim 22, further comprising:
at least one modification selected from Table 4;
wherein said modification is not an excluded modification.
26. A method for producing a genetically-modified eukaryotic cell including an exogenous sequence of interest inserted in a chromosome of said eukaryotic cell, comprising:
transfecting a eukaryotic cell with one or more nucleic acids including (i) a first nucleic acid sequence encoding a meganuclease, and (ii) a second nucleic acid sequence including said sequence of interest;
wherein said meganuclease produces a cleavage site in said chromosome and said sequence of interest is inserted into said chromosome at said cleavage site;
and wherein said meganuclease is a recombinant meganuclease of any one of claims 1-25.
27. A method as in claim 26 wherein:
said second nucleic acid further comprises sequences homologous to sequences flanking said cleavage site and said sequence of interest is inserted at said cleavage site by homologous recombination.
28. A method as in claim 26 wherein:
said second nucleic acid lacks substantial homology to said cleavage site and said sequence of interest is inserted into said chromosome by non-homologous end-joining.
29. A method for producing a genetically-modified eukaryotic cell including an exogenous sequence of interest inserted in a chromosome of said eukaryotic cell, comprising:
introducing a meganuclease protein into a eukaryotic cell; and transfecting said eukaryotic cell with a nucleic acid including said sequence of interest;
wherein said meganuclease produces a cleavage site in said chromosome and said sequence of interest is inserted into said chromosome at said cleavage site;
and wherein said meganuclease is a recombinant meganuclease of any one of claims 1-25.
30. A method as in claim 29 wherein:
said nucleic acid further comprises sequences homologous to sequences flanking said cleavage site and said sequence of interest is inserted at said cleavage site by homologous recombination.
31. A method as in claim 29 wherein:
said nucleic acid lacks substantial homology to said cleavage site and said sequence of interest is inserted into said chromosome by non-homologous end-joining.
32. A method for producing a genetically-modified eukaryotic cell by disrupting a target sequence in a chromosome of said eukaryotic cell, comprising:
transfecting a eukaryotic cell with a nucleic acid encoding a meganuclease;
wherein said meganuclease produces a cleavage site in said chromosome and said target sequence is disrupted by non-homologous end-joining at said cleavage site; and wherein said meganuclease is a recombinant meganuclease of any one of claims 1-25.
33. A method of producing a genetically-modified organism comprising:
producing a genetically-modified eukaryotic cell according to the method of any one of claims 26-32; and growing said genetically-modified eukaryotic cell to produce said genetically-modified organism.
34. A method as in claim 33 wherein:
said eukaryotic cell is selected from the group consisting of a gamete, a zygote, a blastocyst cell, an embryonic stem cell, and a protoplast cell.
35. A method for treating a disease by gene therapy in a eukaryote, comprising:
transfecting at least one cell of said eukaryote with one or more nucleic acids including (i) a first nucleic acid sequence encoding a meganuclease, and (ii) a second nucleic acid sequence including a sequence of interest;
wherein said meganuclease produces a cleavage site in said chromosome and said sequence of interest is inserted into said chromosome at said cleavage site;
wherein said meganuclease is a recombinant meganuclease of any one of claims 1-25; and wherein insertion of said sequence of interest provides said gene therapy for said disease.
36. A method as in claim 35 wherein:
said second nucleic acid sequence further comprises sequences homologous to sequences flanking said cleavage site and said sequence of interest is inserted at said cleavage site by homologous recombination.
37. A method as in claim 35 wherein:
said second nucleic acid sequence lacks substantial homology to said cleavage site and said sequence of interest is inserted into said chromosome by non-homologous end-joining.
38. A method for treating a disease by gene therapy in a eukaryote, comprising:
introducing a meganuclease protein into at least one cell of said eukaryote;
and transfecting said eukaryotic cell with a nucleic acid including a sequence of interest;

wherein said meganuclease produces a cleavage site in said chromosome and said sequence of interest is inserted into said chromosome at said cleavage site;
wherein said meganuclease is a recombinant meganuclease of any one of claims 1-25; and wherein insertion of said sequence of interest provides said gene therapy for said disease.
39. A method as in claim 38 wherein:
said nucleic acid further comprises sequences homologous to sequences flanking said cleavage site and said sequence of interest is inserted at said cleavage site by homologous recombination.
40. A method as in claim 38 wherein:
said nucleic acid lacks substantial homology to said cleavage site and said sequence of interest is inserted into said chromosome by non-homologous end-joining.
41. A method for treating a disease by gene therapy in a eukaryote by disrupting a target sequence in a chromosome of said eukaryotic cell, comprising:
transfecting at least one cell of said eukaryote with a nucleic acid encoding a meganuclease;
wherein said meganuclease produces a cleavage site in said chromosome and said target sequence is disrupted by non-homologous end-joining at said cleavage site;
wherein said meganuclease is a recombinant meganuclease of any one of claims 1-25; and wherein disruption of said target sequence provides said gene therapy for said disease.
42. A method for treating a viral pathogen infection in a eukaryotic host by disrupting a target sequence in a genome of said viral pathogen, comprising:
transfecting at least one infected cell of said eukaryotic host with a nucleic acid encoding a meganuclease;

wherein said meganuclease produces a cleavage site in said viral genome and said target sequence is disrupted by non-homologous end-joining at said cleavage site;
wherein said meganuclease is a recombinant meganuclease of any one of claims 1-25; and wherein disruption of said target sequence provides treatment for said infection.
43. A method for treating a viral pathogen infection in a eukaryotic host by disrupting a target sequence in a genome of said viral pathogen, comprising:
transfecting at least one infected cell of said eukaryotic host with a first nucleic acid encoding a meganuclease and a second nucleic acid;
wherein said meganuclease produces a cleavage site in said viral genome and said target sequence is disrupted by homologous recombination of said viral genome and said second nucleic acid at said cleavage site;
wherein said meganuclease is a recombinant meganuclease of any one of claims 1-25;
wherein said second nucleic acid comprises sequences homologous to sequences flanking said cleavage site; and wherein disruption of said target sequence provides treatment for said infection.
44. A method for treating a prokaryotic pathogen infection in a eukaryotic host by disrupting a target sequence in a genome of said prokaryotic pathogen, comprising:
transfecting at least cell of said prokaryotic pathogen infecting said eukaryotic host with a nucleic acid encoding a meganuclease;
wherein said meganuclease produces a cleavage site in said prokaryotic genome and said target sequence is disrupted by non-homologous end-joining at said cleavage site;
wherein said meganuclease is a recombinant meganuclease of any one of claims 1-25; and wherein disruption of said target sequence provides treatment for said infection.
45. A method for treating a prokaryotic pathogen infection in a eukaryotic host by disrupting a target sequence in a genome of said prokaryotic pathogen, comprising:
transfecting at least cell of said prokaryotic pathogen infecting said eukaryotic host with a first nucleic acid encoding a meganuclease and a second nucleic acid;
wherein said meganuclease produces a cleavage site in said prokaryotic genome and said target sequence is disrupted by homologous recombination of said prokaryotic genome and said second nucleic acid at said cleavage site;
wherein said meganuclease is a recombinant meganuclease of any one of claims 1-25;
wherein said second nucleic acid comprises sequences homologous to sequences flanking said cleavage site; and wherein disruption of said target sequence provides treatment for said infection.
46. A method for rationally-designing a recombinant meganuclease having altered specificity for at least one base position of a recognition sequence, said altered specificity including at least one desired change, the method comprising:
(1) determining at least a portion of a three-dimensional structure of a reference meganuclease-DNA complex;
(2) identifying amino acid residues forming a base contact surface at said base position;
(3) determining a distance between a .beta.-carbon of at least a first residue comprising said contact surface and at least a first base at said base position;
(4) identifying an amino acid substitution to promote said desired change by (a) for a first residue which is < 6 .ANG. from said first base, selecting a substitution from Group 1 and/or Group 2 which is a member of an appropriate one of Group G, Group C, Group T or Group A to promote said desired change; or (b) for a first residue which is > 6 A from said first base, selecting a substitution from Group 2 and/or Group 3 which is a member of an appropriate one of Group G, Group C, Group T or Group A to promote said desired change.
47. A method for rationally-designing a recombinant meganuclease having increased DNA-binding affinity, coinprising:
(1) determining at least a portion of a three-dimensional structure of a reference meganuclease-DNA complex;
(2) identifying amino acid contact residues forming a backbone contact surface;
(3) identifying an amino acid substitution to increase said DNA-binding affinity by (a) for a contact residue having a negatively-charged or hydrophobic side chain, selecting a substitution having an uncharged/polar or positively-charged side chain;
or (b) for a contact residue having an uncharged/polar side chain, selecting a substitution having a positively-charged side chain.
48. A method for rationally-designing a recombinant meganuclease having decreased DNA-binding affinity, comprising:
(1) determining at least a portion of a three-dimensional structure of a reference meganuclease-DNA complex;
(2) identifying amino acid contact residues forming a backbone contact surface;
(3) identifying an amino acid substitution to decrease said DNA-binding affinity by (a) for a contact residue having a positively-charged side chain, selecting a substitution having an uncharged/polar or negatively-charged side chain; or (b) for a contact residue having an hydrophobic or uncharged/polar side chain, selecting a substitution having a negatively-charged side chain.
CA2626262A 2005-10-18 2006-10-18 Rationally-designed meganucleases with altered sequence specificity and dna-binding affinity Active CA2626262C (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CA2891996A CA2891996A1 (en) 2005-10-18 2006-10-18 Rationally-designed meganucleases with altered sequence specificity and dna-binding affinity

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US72751205P 2005-10-18 2005-10-18
US60/727,512 2005-10-18
PCT/US2006/040919 WO2007047859A2 (en) 2005-10-18 2006-10-18 Rationally-designed meganucleases with altered sequence specificity and dna-binding affinity

Related Child Applications (1)

Application Number Title Priority Date Filing Date
CA2891996A Division CA2891996A1 (en) 2005-10-18 2006-10-18 Rationally-designed meganucleases with altered sequence specificity and dna-binding affinity

Publications (2)

Publication Number Publication Date
CA2626262A1 true CA2626262A1 (en) 2007-04-26
CA2626262C CA2626262C (en) 2015-09-08

Family

ID=37963286

Family Applications (3)

Application Number Title Priority Date Filing Date
CA3055968A Pending CA3055968A1 (en) 2005-10-18 2006-10-18 Rationally-designed meganucleases with altered sequence specificity and dna-binding affinity
CA2626262A Active CA2626262C (en) 2005-10-18 2006-10-18 Rationally-designed meganucleases with altered sequence specificity and dna-binding affinity
CA2891996A Pending CA2891996A1 (en) 2005-10-18 2006-10-18 Rationally-designed meganucleases with altered sequence specificity and dna-binding affinity

Family Applications Before (1)

Application Number Title Priority Date Filing Date
CA3055968A Pending CA3055968A1 (en) 2005-10-18 2006-10-18 Rationally-designed meganucleases with altered sequence specificity and dna-binding affinity

Family Applications After (1)

Application Number Title Priority Date Filing Date
CA2891996A Pending CA2891996A1 (en) 2005-10-18 2006-10-18 Rationally-designed meganucleases with altered sequence specificity and dna-binding affinity

Country Status (9)

Country Link
US (19) US8021867B2 (en)
EP (13) EP2650366B1 (en)
JP (9) JP5937292B2 (en)
AT (1) ATE549401T1 (en)
AU (1) AU2006304668B2 (en)
CA (3) CA3055968A1 (en)
DK (9) DK2628794T3 (en)
ES (9) ES2533370T3 (en)
WO (1) WO2007047859A2 (en)

Families Citing this family (421)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2005520519A (en) 2002-03-15 2005-07-14 セレクティス Hybrid and single chain meganucleases and uses thereof
WO2009095742A1 (en) 2008-01-31 2009-08-06 Cellectis New i-crei derived single-chain meganuclease and uses thereof
EP1590453B1 (en) * 2003-01-28 2013-11-27 Cellectis Custom-made meganuclease and use thereof
EP1863909B2 (en) * 2005-03-15 2014-09-10 Cellectis I-crei meganuclease variants with modified specificity, method of preparation and uses thereof
WO2009134714A2 (en) * 2008-04-28 2009-11-05 Precision Biosciences, Inc. Fusion molecules of rationally-designed dna-binding proteins and effector domains
WO2007060495A1 (en) * 2005-10-25 2007-05-31 Cellectis I-crei homing endonuclease variants having novel cleavage specificity and use thereof
ES2378333T3 (en) * 2006-05-25 2012-04-11 Sangamo Biosciences, Inc. Methods and compositions for gene inactivation
WO2008093152A1 (en) * 2007-02-01 2008-08-07 Cellectis Obligate heterodimer meganucleases and uses thereof
JP5524627B2 (en) * 2007-02-19 2014-06-18 セレクティス LAGLIDADG homing endonuclease variant with novel substrate specificity and use thereof
CA2689345C (en) 2007-06-05 2017-07-11 Bayer Bioscience N.V. Methods and means for exact replacement of target dna in eukaryotic organisms
CN101677987A (en) * 2007-06-22 2010-03-24 赛多斯有限责任公司 solubilized formulation of docetaxel without tween 80
CN101849010A (en) 2007-06-29 2010-09-29 先锋高级育种国际公司 Methods for altering the genome of a monocot plant cell
WO2009013559A1 (en) * 2007-07-23 2009-01-29 Cellectis Meganuclease variants cleaving a dna target sequence from the human hemoglobin beta gene and uses thereof
WO2009019528A1 (en) * 2007-08-03 2009-02-12 Cellectis Meganuclease variants cleaving a dna target sequence from the human interleukin-2 receptor gamma chain gene and uses thereof
US11235026B2 (en) 2007-09-27 2022-02-01 Sangamo Therapeutics, Inc. Methods and compositions for modulating PD1
US8563314B2 (en) 2007-09-27 2013-10-22 Sangamo Biosciences, Inc. Methods and compositions for modulating PD1
AU2008305567B2 (en) * 2007-09-27 2014-04-10 Sangamo Therapeutics, Inc. Rapid in vivo identification of biologically active nucleases
ES2732735T3 (en) 2007-10-31 2019-11-25 Prec Biosciences Inc Single-chain meganucleases designed rationally with non-palindromic recognition sequences
US20110041194A1 (en) * 2007-11-28 2011-02-17 Cellectis I-msoi homing endonuclease variants having novel substrate specificity and use thereof
WO2009076292A2 (en) * 2007-12-07 2009-06-18 Precision Biosciences, Inc. Rationally-designed meganucleases with recognition sequences found in dnase hypersensitive regions of the human genome
MX341747B (en) 2008-02-29 2016-08-31 Monsanto Technology Llc Corn plant event mon87460 and compositions and methods for detection thereof.
WO2009114321A2 (en) 2008-03-11 2009-09-17 Precision Biosciencs, Inc. Rationally-designed meganucleases for maize genome engineering
US20100071083A1 (en) * 2008-03-12 2010-03-18 Smith James J Temperature-dependent meganuclease activity
AU2009238629C1 (en) 2008-04-14 2015-04-30 Sangamo Therapeutics, Inc. Linear donor constructs for targeted integration
ES2705756T3 (en) 2008-07-14 2019-03-26 Prec Biosciences Inc Recognition sequences for meganucleases derived from I-Crel and its uses
CA2734235C (en) * 2008-08-22 2019-03-26 Sangamo Biosciences, Inc. Methods and compositions for targeted single-stranded cleavage and targeted integration
EP2370575B1 (en) * 2008-12-17 2017-11-01 Dow AgroSciences LLC Targeted integration into the zp15 locus
EP2393506B1 (en) 2009-02-04 2018-03-28 Sangamo Therapeutics, Inc. Methods and compositions for treating neuropathies
US20110294217A1 (en) * 2009-02-12 2011-12-01 Fred Hutchinson Cancer Research Center Dna nicking enzyme from a homing endonuclease that stimulates site-specific gene conversion
JP5932632B2 (en) 2009-03-20 2016-06-15 サンガモ バイオサイエンシーズ, インコーポレイテッド Modification of CXCR4 using modified zinc finger protein
WO2010122367A2 (en) * 2009-04-21 2010-10-28 Cellectis Meganuclease variants cleaving the genomic insertion of a virus and uses thereof
US8772008B2 (en) 2009-05-18 2014-07-08 Sangamo Biosciences, Inc. Methods and compositions for increasing nuclease activity
WO2010136841A2 (en) * 2009-05-26 2010-12-02 Cellectis Meganuclease variants cleaving the genome of a non-genomically integrating virus and uses thereof
WO2010136981A2 (en) * 2009-05-26 2010-12-02 Cellectis Meganuclease variants cleaving the genome of a pathogenic non-integrating virus and uses thereof
WO2011002503A1 (en) * 2009-06-30 2011-01-06 Sangamo Biosciences, Inc. Rapid screening of biologically active nucleases and isolation of nuclease-modified cells
WO2011016840A2 (en) 2009-07-28 2011-02-10 Sangamo Biosciences, Inc. Methods and compositions for treating trinucleotide repeat disorders
WO2011017315A2 (en) * 2009-08-03 2011-02-10 Recombinetics, Inc. Methods and compositions for targeted gene modification
NZ598053A (en) * 2009-08-11 2014-01-31 Sangamo Biosciences Inc Organisms homozygous for targeted modification
ES2657067T3 (en) 2009-10-22 2018-03-01 Dow Agrosciences Llc Genetically modified zinc finger proteins that target plant genes involved in fatty acid biosynthesis
EP2319872A1 (en) 2009-11-04 2011-05-11 BASF Plant Science GmbH Amylopectin type starch with enhanced retrogradation stability
CA2782014C (en) * 2009-11-27 2021-08-31 Basf Plant Science Company Gmbh Optimized endonucleases and uses thereof
AU2010325564A1 (en) 2009-11-27 2012-07-12 Basf Plant Science Company Gmbh Chimeric endonucleases and uses thereof
CN102686726B (en) 2009-11-27 2015-12-16 巴斯夫植物科学有限公司 Chimeric endonuclease and uses thereof
CA2793596A1 (en) 2009-12-30 2011-07-07 Pioneer Hi-Bred International, Inc. Methods and compositions for targeted polynucleotide modification
US20110203012A1 (en) * 2010-01-21 2011-08-18 Dotson Stanton B Methods and compositions for use of directed recombination in plant breeding
EP2660318A1 (en) 2010-02-09 2013-11-06 Sangamo BioSciences, Inc. Targeted genomic modification with partially single-stranded donor molecules
CA2791116A1 (en) * 2010-02-26 2011-09-01 Olivier Danos Use of endonucleases for inserting transgenes into safe harbor loci
US20130198897A1 (en) 2010-03-22 2013-08-01 Philip Morris Products S.A. Modifying enzyme activity in plants
EP2566972B1 (en) 2010-05-03 2020-01-15 Sangamo Therapeutics, Inc. Compositions for linking zinc finger modules
EP2571512B1 (en) 2010-05-17 2017-08-23 Sangamo BioSciences, Inc. Novel dna-binding proteins and uses thereof
SG185668A1 (en) * 2010-06-09 2012-12-28 Bayer Cropscience Nv Methods and means to modify a plant genome at a nucleotide sequence commonly used in plant genome engineering
US9593317B2 (en) * 2010-06-09 2017-03-14 Bayer Cropscience Nv Methods and means to modify a plant genome at a nucleotide sequence commonly used in plant genome engineering
EP2596011B1 (en) 2010-07-21 2018-10-03 Sangamo Therapeutics, Inc. Methods and compositions for modification of a hla locus
EP2601297B1 (en) * 2010-08-02 2020-09-16 Cellectis Method for targeted genomic events in algae
CA2811364C (en) 2010-09-27 2022-01-04 Sangamo Biosciences, Inc. Methods and compositions for inhibiting viral entry into cells
CN103354715A (en) 2011-01-14 2013-10-16 双刃基金会 Citrus trees with resistance to citrus canker
BR112013024337A2 (en) 2011-03-23 2017-09-26 Du Pont complex transgenic trace locus in a plant, plant or seed, method for producing in a plant a complex transgenic trace locus and expression construct
WO2012138901A1 (en) * 2011-04-05 2012-10-11 Cellectis Sa Method for enhancing rare-cutting endonuclease efficiency and uses thereof
WO2012151254A1 (en) 2011-05-02 2012-11-08 Board Of Regents Of The University Of Nebraska Plants with useful traits and related methods
EP2535416A1 (en) 2011-05-24 2012-12-19 BASF Plant Science Company GmbH Development of phytophthora resistant potato with increased yield
ES2713529T3 (en) 2011-06-01 2019-05-22 Prec Biosciences Inc Procedures and products for the production of modified mammalian cell lines with amplified transgenes
EP2718443B1 (en) 2011-06-06 2017-11-29 Bayer CropScience NV Methods and means to modify a plant genome at a preselected site
US9758796B2 (en) 2011-06-10 2017-09-12 Basf Plant Science Company Gmbh Nuclease fusion protein and uses thereof
US9574208B2 (en) 2011-06-21 2017-02-21 Ei Du Pont De Nemours And Company Methods and compositions for producing male sterile plants
WO2013009935A2 (en) 2011-07-12 2013-01-17 Two Blades Foundation Late blight resistance genes
EP2737063B1 (en) 2011-07-25 2016-06-01 Sangamo BioSciences, Inc. Methods and compositions for alteration of a cystic fibrosis transmembrane conductance regulator (cftr) gene
AU2013204310C1 (en) * 2011-08-22 2015-12-10 BASF Agricultural Solutions Seed US LLC Methods and means to modify a plant genome
MX348003B (en) 2011-08-22 2017-03-08 Bayer Cropscience Nv Methods and means to modify a plant genome.
WO2013044008A2 (en) 2011-09-21 2013-03-28 Sangamo Biosciences, Inc. Methods and compositions for regulation of transgene expression
JP6188703B2 (en) 2011-10-27 2017-08-30 サンガモ セラピューティクス, インコーポレイテッド Methods and compositions for modifying the HPRT locus
EP2612918A1 (en) 2012-01-06 2013-07-10 BASF Plant Science Company GmbH In planta recombination
WO2013109754A1 (en) 2012-01-17 2013-07-25 Pioneer Hi-Bred International, Inc. Plant transcription factors, promoters and uses thereof
BR112014018294B1 (en) 2012-01-26 2022-01-11 Norfolk Plant Sciences, Ltd METHOD FOR PRODUCING A PLANT, EXPRESSION CASSETTE, AND BACTERIAL CELL
KR102084539B1 (en) 2012-02-29 2020-03-04 상가모 테라퓨틱스, 인코포레이티드 Methods and compositions for treating huntington's disease
WO2013136274A1 (en) 2012-03-13 2013-09-19 University Of Guelph Myb55 promoter and use thereof
US20150218578A1 (en) 2012-03-13 2015-08-06 University Of Guelph Methods of increasing tolerance to heat stress and amino acid content of plants
EP2841581B2 (en) 2012-04-23 2023-03-08 BASF Agricultural Solutions Seed US LLC Targeted genome engineering in plants
MX369788B (en) 2012-05-02 2019-11-21 Dow Agrosciences Llc Targeted modification of malate dehydrogenase.
BR112014027468A2 (en) 2012-05-04 2017-06-27 Du Pont isolated or recombinant polynucleotide, recombinant DNA construction, cell, plant, plant explant, transgenic seed, isolated polypeptide, composition, methods of producing meganuclease, introducing disruption and integrating a polynucleotide.
EP2847338B1 (en) 2012-05-07 2018-09-19 Sangamo Therapeutics, Inc. Methods and compositions for nuclease-mediated targeted integration of transgenes
CA2878037C (en) 2012-07-11 2021-08-31 Sangamo Biosciences, Inc. Methods and compositions for delivery of biologics
HUE041553T2 (en) 2012-07-11 2019-05-28 Sangamo Therapeutics Inc Methods and compositions for the treatment of monogenic diseases
CA3171494A1 (en) 2012-08-29 2014-03-06 Sangamo Biosciences, Inc. Cells genetically modified within the bcl11a gene by a nuclease and various aspects related thereto
HUE050797T2 (en) 2012-10-10 2021-01-28 Sangamo Therapeutics Inc T cell modifying compounds and uses thereof
CN109554350B (en) 2012-11-27 2022-09-23 儿童医疗中心有限公司 Targeting BCL11A distal regulatory elements for fetal hemoglobin re-induction
AR093980A1 (en) 2012-12-13 2015-07-01 Dow Agrosciences Llc PRECISION GENES THAT HAVE A PARTICULAR LOCUS IN THE CORN AS THE OBJECTIVE
CA2901676C (en) * 2013-02-25 2023-08-22 Sangamo Biosciences, Inc. Methods and compositions for enhancing nuclease-mediated gene disruption
JP6392315B2 (en) 2013-03-14 2018-09-19 イミュソフト コーポレーション In vitro memory B cell differentiation method and transduction method using VSV-G pseudotype virus vector
BR112015025006A2 (en) 2013-04-02 2017-10-10 Bayer Cropscience Nv genomic engineering targeted on eukaryotes
KR102192599B1 (en) 2013-04-05 2020-12-18 다우 아그로사이언시즈 엘엘씨 Methods and compositions for integration of an exogenous sequence within the genome of plants
WO2015006437A1 (en) 2013-07-10 2015-01-15 Majzoub Joseph A Mrap2 knockouts
WO2015026887A1 (en) 2013-08-22 2015-02-26 E. I. Du Pont De Nemours And Company A soybean u6 polymerase iii promoter and methods of use
EP3039136B8 (en) 2013-08-28 2020-12-16 Sangamo Therapeutics, Inc. Compositions for linking dna-binding domains and cleavage domains
WO2015033343A1 (en) 2013-09-03 2015-03-12 Yissum Research Development Company Of The Hebrew University Of Jerusalem Ltd. Compositions and methods for expressing recombinant polypeptides
US10767188B2 (en) 2013-09-25 2020-09-08 Nutech Ventures Methods and compositions for obtaining useful plant traits
KR20150037550A (en) * 2013-09-30 2015-04-08 주식회사 엘지화학 Polarizing plate having locally depolarizied area and method for producing thereof
CA2926078C (en) 2013-10-17 2021-11-16 Sangamo Biosciences, Inc. Delivery methods and compositions for nuclease-mediated genome engineering in hematopoietic stem cells
EP3058072B1 (en) 2013-10-17 2021-05-19 Sangamo Therapeutics, Inc. Delivery methods and compositions for nuclease-mediated genome engineering
JP6560205B2 (en) 2013-11-04 2019-08-14 ダウ アグロサイエンシィズ エルエルシー Optimal soybean locus
BR102014027438B1 (en) 2013-11-04 2022-09-27 Dow Agrosciences Llc RECOMBINANT NUCLEIC ACID MOLECULE AND PRODUCTION METHOD OF A TRANSGENIC PLANT CELL
UA120503C2 (en) 2013-11-04 2019-12-26 Дау Агросайєнсиз Елелсі Optimal maize loci
JP2016537341A (en) 2013-11-11 2016-12-01 サンガモ バイオサイエンシーズ, インコーポレイテッド Methods and compositions for treating Huntington's disease
HRP20211706T1 (en) 2013-11-13 2022-02-04 The Children's Medical Center Corporation Nuclease-mediated regulation of gene expression
HUE051628T2 (en) 2013-12-09 2021-03-01 Sangamo Therapeutics Inc Methods and compositions for genome engineering
US20170166920A1 (en) 2014-01-30 2017-06-15 Two Blades Foundation Plants with enhanced resistance to phytophthora
HUE051232T2 (en) 2014-02-03 2021-03-01 Sangamo Therapeutics Inc Methods and compositions for treatment of a beta thalessemia
EP3110454B1 (en) 2014-02-24 2020-11-18 Sangamo Therapeutics, Inc. Methods and compositions for nuclease-mediated targeted integration
TW201538518A (en) 2014-02-28 2015-10-16 Dow Agrosciences Llc Root specific expression conferred by chimeric gene regulatory elements
JP2017512767A (en) 2014-03-12 2017-05-25 プレシジョン バイオサイエンシズ,インク. Deletion of dystrophin gene exons using a modified nuclease
CA2942762C (en) 2014-03-18 2023-10-17 Sangamo Biosciences, Inc. Methods and compositions for regulation of zinc finger protein expression
GB201407852D0 (en) 2014-05-02 2014-06-18 Iontas Ltd Preparation of libraries od protein variants expressed in eukaryotic cells and use for selecting binding molecules
WO2015171603A1 (en) 2014-05-06 2015-11-12 Two Blades Foundation Methods for producing plants with enhanced resistance to oomycete pathogens
AU2015255877B2 (en) 2014-05-08 2020-03-26 Chdi Foundation, Inc. Methods and compositions for treating huntington's disease
DE102014209264B8 (en) * 2014-05-15 2017-01-12 Olympus Winter & Ibe Gmbh High-frequency surgical device
EP3149170A1 (en) * 2014-05-30 2017-04-05 The Board of Trustees of the Leland Stanford Junior University Compositions and methods to treat latent viral infections
MX2017000422A (en) 2014-07-11 2017-05-01 Du Pont Compositions and methods for producing plants resistant to glyphosate herbicide.
US9616090B2 (en) 2014-07-30 2017-04-11 Sangamo Biosciences, Inc. Gene correction of SCID-related genes in hematopoietic stem and progenitor cells
SI3194570T1 (en) 2014-09-16 2022-01-31 Sangamo Therapeutics, Inc. Methods and compositions for nuclease-mediated genome engineering and correction in hematopoietic stem cells
IL287561B2 (en) 2014-10-01 2024-03-01 Massachusetts Gen Hospital Methods for increasing efficiency of nuclease-induced homology-directed repair
WO2016100333A1 (en) 2014-12-15 2016-06-23 Syngenta Participations Ag Pesticidal microrna carriers and use thereof
US9963710B2 (en) 2014-12-23 2018-05-08 Syngenta Participations Ag Methods and compositions for identifying and enriching for cells comprising site specific genomic modifications
WO2016118726A2 (en) 2015-01-21 2016-07-28 Sangamo Biosciences, Inc. Methods and compositions for identification of highly specific nucleases
WO2016157175A1 (en) 2015-03-27 2016-10-06 Yeda Research And Development Co. Ltd. Methods of treating motor neuron diseases
EP3277823B1 (en) 2015-04-03 2023-09-13 Dana-Farber Cancer Institute, Inc. Composition and methods of genome editing of b-cells
RU2017137599A (en) 2015-04-15 2019-05-15 ДАУ АГРОСАЙЕНСИЗ ЭлЭлСи VEGETABLE PROMOTER FOR THE TRANSGEN EXPRESSION
EP3283632A1 (en) 2015-04-15 2018-02-21 Dow Agrosciences LLC Plant promoter for transgene expression
US11491342B2 (en) 2015-07-01 2022-11-08 Btl Medical Solutions A.S. Magnetic stimulation methods and devices for therapeutic treatments
EP3288570A4 (en) 2015-04-29 2018-11-21 Fred Hutchinson Cancer Research Center Modified stem cells and uses thereof
EP3289076B1 (en) * 2015-05-01 2021-11-17 Precision Biosciences, Inc. Precise deletion of chromoscomal sequences in vivo
EP3294873B1 (en) 2015-05-08 2020-08-19 The Children's Medical Center Corporation Targeting bcl11a enhancer functional regions for fetal hemoglobin reinduction
WO2016182881A1 (en) 2015-05-09 2016-11-17 Two Blades Foundation Late blight resistance gene from solanum americanum and methods of use
WO2016181393A1 (en) 2015-05-11 2016-11-17 Yeda Research And Development Co. Ltd. Citrin inhibitors for the treatment of cancer
ES2962885T3 (en) 2015-05-15 2024-03-21 Massachusetts Gen Hospital Tumor necrosis factor receptor superfamily antagonist antibodies
WO2016187459A1 (en) 2015-05-20 2016-11-24 The Regents Of The University Of California Method for generating human dendritic cells for immunotherapy
ES2920477T3 (en) 2015-06-19 2022-08-04 Prec Biosciences Inc Self-limiting viral vectors encoding nucleases
EP3313420B1 (en) 2015-06-25 2024-03-13 The Children's Medical Center Corporation Methods and compositions relating to hematopoietic stem cell expansion, enrichment, and maintenance
CN108024544B (en) 2015-07-13 2022-04-29 桑格摩生物治疗股份有限公司 Delivery methods and compositions for nuclease-mediated genome engineering
CA3033617A1 (en) 2015-08-11 2017-02-16 Anie Philip Peptidic tgf-beta antagonists
EP3337908A4 (en) 2015-08-18 2019-01-23 The Broad Institute, Inc. Methods and compositions for altering function and structure of chromatin loops and/or domains
AU2016321244B2 (en) 2015-09-08 2020-09-24 Precision Biosciences, Inc. Treatment of retinitis pigmentosa using engineered meganucleases
TW201718862A (en) 2015-09-22 2017-06-01 Dow Agrosciences Llc Plant promoter and 3' UTR for transgene expression
TW201718861A (en) 2015-09-22 2017-06-01 道禮責任有限公司 Plant promoter and 3'UTR for transgene expression
EP4108255A1 (en) 2015-10-05 2022-12-28 Precision Biosciences, Inc. Genetically-modified cells comprising a modified human t cell receptor alpha constant region gene
WO2017062439A1 (en) * 2015-10-05 2017-04-13 Precision Biosciences, Inc. Engineered meganucleases with recognition sequences found in the human t cell receptor alpha constant region gene
WO2017062790A1 (en) 2015-10-09 2017-04-13 Two Blades Foundation Cold shock protein receptors and methods of use
AU2016340893B2 (en) 2015-10-22 2019-06-27 Corteva Agriscience Llc Plant promoter for transgene expression
TWI812584B (en) 2015-10-30 2023-08-21 美國加利福尼亞大學董事會 Methods of generating t-cells from stem cells and immunotherapeutic methods using the t-cells
WO2017079428A1 (en) 2015-11-04 2017-05-11 President And Fellows Of Harvard College Site specific germline modification
WO2017078935A1 (en) 2015-11-04 2017-05-11 Dow Agrosciences Llc Plant promoter for transgene expression
WO2017079724A1 (en) 2015-11-06 2017-05-11 The Jackson Laboratory Large genomic dna knock-in and uses thereof
AU2016355178B9 (en) 2015-11-19 2019-05-30 Massachusetts Institute Of Technology Lymphocyte antigen CD5-like (CD5L)-interleukin 12B (p40) heterodimers in immunity
JP7128741B2 (en) 2015-12-18 2022-08-31 サンガモ セラピューティクス, インコーポレイテッド Targeted disruption of T-cell receptors
IL297018A (en) 2015-12-18 2022-12-01 Sangamo Therapeutics Inc Targeted disruption of the mhc cell receptor
AU2016379393B2 (en) 2015-12-23 2023-01-05 Precision Biosciences, Inc. Engineered meganucleases with recognition sequences found in the human beta-2 microglobulin gene
US10828318B2 (en) 2016-01-06 2020-11-10 Yeda Research And Development Co. Ltd. Compositions and methods for treating malignant, autoimmune and inflammatory diseases
BR112018013663A2 (en) 2016-01-11 2019-01-22 Univ Leland Stanford Junior chimeric proteins and immunotherapy methods
WO2017123559A2 (en) 2016-01-11 2017-07-20 The Board Of Trustees Of The Leland Stanford Junior University Chimeric proteins and methods of regulating gene expression
WO2017125931A1 (en) 2016-01-21 2017-07-27 The State Of Israel, Ministry Of Agriculture & Rural Development, Agricultural Research Organization (Aro) (Volcani Center) Parthenocarpic plants and methods of producing same
WO2017130205A1 (en) 2016-01-31 2017-08-03 Hadasit Medical Research Services And Development Ltd. Autosomal-identical pluripotent stem cell populations having non-identical sex chromosomal composition and uses thereof
JP7012650B2 (en) 2016-02-02 2022-01-28 サンガモ セラピューティクス, インコーポレイテッド Composition for linking DNA binding domain and cleavage domain
WO2017138008A2 (en) 2016-02-14 2017-08-17 Yeda Research And Development Co. Ltd. Methods of modulating protein exocytosis and uses of same in therapy
CN109069870B (en) 2016-02-24 2022-04-29 洛克菲勒大学 Embryonic cell-based therapeutic candidate screening systems, models for huntington's disease and uses thereof
WO2017153982A1 (en) 2016-03-06 2017-09-14 Yeda Research And Development Co. Ltd. Method for modulating myelination
EP3429603B1 (en) 2016-03-15 2021-12-29 Children's Medical Center Corporation Methods and compositions relating to hematopoietic stem cell expansion
US9981980B2 (en) 2016-04-27 2018-05-29 University Of Puerto Rico 1,5-Disubstituted 1,2,3-triazoles are inhibitors of Rac/Cdc42 GTPases
CA3022801A1 (en) 2016-05-03 2017-11-09 Precision Biosciences, Inc. Engineered nucleases useful for treatment of hemophilia a
CN109415737A (en) 2016-05-25 2019-03-01 嘉吉公司 For generating the engineered nucleic acid enzyme of mutation in plant
WO2017202715A1 (en) 2016-05-26 2017-11-30 Nunhems B.V. Seedless fruit producing plants
CN109689894A (en) 2016-06-19 2019-04-26 耶路撒冷希伯来大学伊萨姆研发有限公司 Chemotherapy resistance is screened in human haploid cell
WO2017223129A1 (en) 2016-06-22 2017-12-28 North Carolina State University Method for increasing nitrogen-use efficiency and or nitrogen-utilisation efficiency in plants
WO2018015936A2 (en) 2016-07-21 2018-01-25 Maxcyte, Inc. Methods and compositions for modifying genomic dna
IL247368A0 (en) 2016-08-18 2016-11-30 Yeda Res & Dev Diagnostic and therapeutic uses of exosomes
IL300783A (en) 2016-08-24 2023-04-01 Sangamo Therapeutics Inc Engineered target-specific zinc finger nucleases
US20190225974A1 (en) 2016-09-23 2019-07-25 BASF Agricultural Solutions Seed US LLC Targeted genome optimization in plants
EA201990591A1 (en) 2016-09-23 2019-08-30 Фред Хатчинсон Кэнсер Рисерч Сентер TCR SPECIFIC TO HISTORIC COMPATIBILITY (H) HA-1 MINORAL ANTIGEN AND THEIR APPLICATIONS
EP3518657B1 (en) 2016-10-03 2022-07-13 Corteva Agriscience LLC Plant promoter for transgene expression
EP3519574B1 (en) 2016-10-03 2022-01-19 Corteva Agriscience LLC Plant promoter for transgene expression
JP6887492B6 (en) 2016-10-04 2021-07-14 プレシジョン バイオサイエンシズ,インク. Co-stimulation domain for use in genetically modified cells
KR102305215B1 (en) 2016-10-14 2021-09-28 프리시젼 바이오사이언시스 인코포레이티드 Engineered meganucleases specific for recognition sequences in the hepatitis B virus genome
WO2018073393A2 (en) 2016-10-19 2018-04-26 Cellectis Tal-effector nuclease (talen) -modified allogenic cells suitable for therapy
AU2017345430A1 (en) 2016-10-20 2019-05-02 Sangamo Therapeutics, Inc. Methods and compositions for the treatment of Fabry disease
EP3532106A4 (en) 2016-10-31 2020-06-24 Sangamo Therapeutics, Inc. Gene correction of scid-related genes in hematopoietic stem and progenitor cells
WO2018076335A1 (en) 2016-10-31 2018-05-03 Institute Of Genetics And Developmental Biology, Chinese Academy Of Sciences Compositions and methods for enhancing abiotic stress tolerance
CA3046199A1 (en) 2016-12-08 2018-06-14 Case Western Reserve University Methods and compositions for enhancing functional myelin production
US20200124615A1 (en) 2016-12-29 2020-04-23 Ukko Inc. Methods for identifying and de-epitoping allergenic polypeptides
EA201992131A1 (en) 2017-03-15 2020-02-06 Фред Хатчинсон Кэнсер Рисерч Сентер HIGH-AFFINE SPECIFIC TO MAGE-A1 TCR AND THEIR APPLICATION
TW201841916A (en) 2017-04-12 2018-12-01 美商麥珍塔治療學股份有限公司 Aryl hydrocarbon receptor antagonists and uses thereof
EP3610266A4 (en) 2017-04-12 2021-04-21 Massachusetts Eye and Ear Infirmary Tumor signature for metastasis, compositions of matter methods of use thereof
WO2018191520A1 (en) 2017-04-12 2018-10-18 The Broad Institute, Inc. Respiratory and sweat gland ionocytes
EP4269596A3 (en) * 2017-04-21 2024-02-14 Precision Biosciences, Inc. Engineered meganucleases specific for recognition sequences in the pcsk9 gene
EP3612232A1 (en) 2017-04-21 2020-02-26 The Broad Institute, Inc. Targeted delivery to beta cells
EP3615510B1 (en) 2017-04-28 2024-03-27 Acuitas Therapeutics, Inc. Novel carbonyl lipids and lipid nanoparticle formulations for delivery of nucleic acids
EP3615662A1 (en) 2017-04-28 2020-03-04 Precision Biosciences, Inc. Methods for reducing dna-induced cytotoxicity and enhancing gene editing in primary cells
AU2018261366A1 (en) 2017-05-03 2019-10-31 Sangamo Therapeutics, Inc. Methods and compositions for modification of a cystic fibrosis transmembrane conductance regulator (CTFR) gene
WO2018204777A2 (en) 2017-05-05 2018-11-08 The Broad Institute, Inc. Methods for identification and modification of lncrna associated with target genotypes and phenotypes
IL252151A0 (en) 2017-05-07 2017-07-31 Fainzilber Michael Methods of treating psychiatric stress disorders
CA3062698A1 (en) 2017-05-08 2018-11-15 Precision Biosciences, Inc. Nucleic acid molecules encoding an engineered antigen receptor and an inhibitory nucleic acid molecule and methods of use thereof
CA3064000A1 (en) 2017-05-24 2018-11-29 Effector Therapeutics, Inc. Methods and compositions for cellular immunotherapy
GB201708662D0 (en) 2017-05-31 2017-07-12 Tropic Biosciences Uk Ltd Compositions and methods for increasing shelf-life of banana
EP3630973A1 (en) 2017-05-31 2020-04-08 Tropic Biosciences UK Limited Methods of selecting cells comprising genome editing events
GB201708665D0 (en) 2017-05-31 2017-07-12 Tropic Biosciences Uk Ltd Compositions and methods for increasing extractability of solids from coffee beans
WO2018224861A1 (en) 2017-06-07 2018-12-13 International Rice Research Institute Increasing hybrid seed production through higher outcrossing rate in cytoplasmic male sterile gramineae plants and related materials and methods
EP3638218A4 (en) 2017-06-14 2021-06-09 The Broad Institute, Inc. Compositions and methods targeting complement component 3 for inhibiting tumor growth
US20200291413A1 (en) 2017-06-23 2020-09-17 University Of Kentucky Research Foundation Method
US20210093667A1 (en) 2017-06-26 2021-04-01 The Broad Institute, Inc. Crispr/cas-adenine deaminase based compositions, systems, and methods for targeted nucleic acid editing
US11053484B2 (en) 2017-06-30 2021-07-06 Precision Biosciences, Inc. Genetically-modified T cells comprising a modified intron in the T cell receptor alpha gene
IL253642A0 (en) 2017-07-24 2017-09-28 Seger Rony Combination therapy for the treatment of cancer
AR112465A1 (en) 2017-07-31 2019-10-30 Reynolds Tobacco Co R METHODS AND COMPOSITIONS FOR THE EDITION OF VIRAL BASED GENES IN PLANTS
US10430138B2 (en) * 2017-08-10 2019-10-01 Canon Kabushiki Kaisha Communication apparatus
WO2019038771A1 (en) 2017-08-23 2019-02-28 Technion Research & Development Foundation Limited Compositions and methods for improving alcohol tolerance in yeast
CN111051349A (en) 2017-09-06 2020-04-21 弗雷德哈钦森癌症研究中心 STREP-TAG specific chimeric receptor and application thereof
WO2019051135A1 (en) 2017-09-06 2019-03-14 Fred Hutchinson Cancer Research Center Methods for improving adoptive cell therapy
DK3684930T3 (en) 2017-09-19 2022-06-07 Tropic Biosciences Uk Ltd Modification of the specificity of non-coding plant RNA molecules for inactivation of gene expression
WO2019070856A1 (en) 2017-10-03 2019-04-11 Precision Biosciences, Inc. Modified epidermal growth factor receptor peptides for use in genetically-modified cells
US20210069241A1 (en) 2017-10-20 2021-03-11 Fred Hutchinson Cancer Research Center Compositions and methods of immunotherapy targeting tigit and/or cd112r or comprising cd226 overexpression
EP3704232A1 (en) 2017-10-31 2020-09-09 Magenta Therapeutics, Inc. Compositions and methods for the expansion of hematopoietic stem and progenitor cells
US20190343885A1 (en) 2017-10-31 2019-11-14 Magenta Therapeutics Inc. Compositions and methods for hematopoietic stem and progenitor cell transplant therapy
EP3704238B1 (en) 2017-11-01 2024-01-03 Precision Biosciences, Inc. Engineered nucleases that target human and canine factor viii genes as a treatment for hemophilia a
CN111565730A (en) 2017-11-09 2020-08-21 桑格摩生物治疗股份有限公司 Genetic modification of cytokine-induced SH2-containing protein (CISH) gene
WO2019094983A1 (en) 2017-11-13 2019-05-16 The Broad Institute, Inc. Methods and compositions for treating cancer by targeting the clec2d-klrb1 pathway
IL255664A0 (en) 2017-11-14 2017-12-31 Shachar Idit Hematopoietic stem cells with improved properties
WO2019108619A1 (en) 2017-11-28 2019-06-06 Two Blades Foundation Methods and compositions for enhancing the disease resistance of plants
WO2019109047A1 (en) 2017-12-01 2019-06-06 Fred Hutchinson Cancer Research Center Binding proteins specific for 5t4 and uses thereof
US11503757B2 (en) 2017-12-03 2022-11-22 Seedx Technologies Inc. Systems and methods for sorting of seeds
EP3707642A1 (en) 2017-12-03 2020-09-16 Seedx Technologies Inc. Systems and methods for sorting of seeds
WO2019106639A1 (en) 2017-12-03 2019-06-06 Seedx Technologies Inc. Systems and methods for sorting of seeds
CN111712262A (en) 2017-12-06 2020-09-25 美真达治疗公司 Dosing regimens for mobilizing hematopoietic stem and progenitor cells
CN111902411A (en) 2018-01-03 2020-11-06 美真达治疗公司 Compositions and methods for expanding hematopoietic stem and progenitor cells and treating inherited metabolic disorders
WO2019140278A1 (en) 2018-01-11 2019-07-18 Fred Hutchinson Cancer Research Center Immunotherapy targeting core binding factor antigens
BR112020014168A2 (en) 2018-01-12 2020-12-08 Basf Se PROTEIN, ISOLATED NUCLEIC ACID, RECOMBINANT GENE, VECTOR, HOSTING CELL, PLANT, PART OF PLANT OR WHEAT SEED, METHODS OF PRODUCTION, WHEAT PRODUCT, FLOUR, WHOLE MUSHROOM, STARCH, MUSHROOMS AND MUSHROOMS. AND / OR SELECTING A WHEAT PLANT
WO2019140351A1 (en) 2018-01-12 2019-07-18 Two Blades Foundation Stem rust resistance genes and methods of use
WO2019155465A1 (en) 2018-02-08 2019-08-15 Yeda Research And Development Co. Ltd. Methods of identifying and using agents for treating diseases associated with intestinal barrier dysfunction
JP2021514631A (en) 2018-02-26 2021-06-17 フレッド ハッチンソン キャンサー リサーチ センター Compositions and Methods for Cellular Immunotherapy
CN111936617A (en) 2018-03-16 2020-11-13 益缪索夫特公司 B cells genetically engineered to secrete follistatin and methods of using the same to treat follistatin-related diseases, conditions, disorders, and enhance muscle growth and strength
IT201800004253A1 (en) 2018-04-05 2019-10-05 Compositions and methods for the treatment of hereditary dominant catecholaminergic polymorphic ventricular tachycardia.
KR20240005139A (en) 2018-04-12 2024-01-11 프리시젼 바이오사이언시스 인코포레이티드 Optimized engineered nucleases having specificity for the human t cell receptor alpha constant region gene
TW201945388A (en) 2018-04-12 2019-12-01 美商精密生物科學公司 Optimized engineered meganucleases having specificity for a recognition sequence in the hepatitis B virus genome
US20210163937A1 (en) 2018-04-18 2021-06-03 B.R.A.I.N. Biotechnology Research And Information Network Ag Enrichment of genome-edited cells
PL3560330T3 (en) 2018-04-24 2022-09-19 KWS SAAT SE & Co. KGaA Plants with improved digestibility and marker haplotypes
WO2019210268A2 (en) 2018-04-27 2019-10-31 The Broad Institute, Inc. Sequencing-based proteomics
GB201807192D0 (en) 2018-05-01 2018-06-13 Tropic Biosciences Uk Ltd Compositions and methods for reducing caffeine content in coffee beans
US20210386829A1 (en) 2018-05-04 2021-12-16 The Broad Institute, Inc. Compositions and methods for modulating cgrp signaling to regulate innate lymphoid cell inflammatory responses
US11690921B2 (en) 2018-05-18 2023-07-04 Sangamo Therapeutics, Inc. Delivery of target specific nucleases
US20210371932A1 (en) 2018-06-01 2021-12-02 Massachusetts Institute Of Technology Methods and compositions for detecting and modulating microenvironment gene signatures from the csf of metastasis patients
EP3806619A1 (en) 2018-06-15 2021-04-21 Nunhems B.V. Seedless watermelon plants comprising modifications in an abc transporter gene
US11873322B2 (en) 2018-06-25 2024-01-16 Yeda Research And Development Co. Ltd. Systems and methods for increasing efficiency of genome editing
AU2019297677A1 (en) 2018-07-04 2021-02-25 Ukko Inc. Methods of de-epitoping wheat proteins and use of same for the treatment of celiac disease
WO2020018964A1 (en) 2018-07-20 2020-01-23 Fred Hutchinson Cancer Research Center Compositions and methods for controlled expression of antigen-specific receptors
GB201812603D0 (en) 2018-08-02 2018-09-19 British American Tobacco Investments Ltd Method
US20210317429A1 (en) 2018-08-20 2021-10-14 The Broad Institute, Inc. Methods and compositions for optochemical control of crispr-cas9
US20210340201A1 (en) 2018-08-22 2021-11-04 Fred Hutchinson Cancer Research Center Immunotherapy targeting kras or her2 antigens
EP3844266A1 (en) 2018-08-28 2021-07-07 Fred Hutchinson Cancer Research Center Methods and compositions for adoptive t cell therapy incorporating induced notch signaling
KR20210071976A (en) 2018-09-04 2021-06-16 마젠타 테라퓨틱스 인코포레이티드 Aryl hydrocarbon receptor antagonists and methods of use
AU2019344927A1 (en) 2018-09-18 2021-04-01 Sangamo Therapeutics, Inc. Programmed cell death 1 (PD1) specific nucleases
EP3852813A4 (en) 2018-09-18 2022-06-22 VNV Newco Inc. Arc-based capsids and uses thereof
WO2020068702A1 (en) 2018-09-24 2020-04-02 Fred Hutchinson Cancer Research Center Chimeric receptor proteins and uses thereof
US20220411783A1 (en) 2018-10-12 2022-12-29 The Broad Institute, Inc. Method for extracting nuclei or whole cells from formalin-fixed paraffin-embedded tissues
WO2020081730A2 (en) 2018-10-16 2020-04-23 Massachusetts Institute Of Technology Methods and compositions for modulating microenvironment
IL262658A (en) 2018-10-28 2020-04-30 Memorial Sloan Kettering Cancer Center Prevention of age related clonal hematopoiesis and diseases associated therewith
GB201817971D0 (en) 2018-11-02 2018-12-19 British American Tobacco Investments Ltd Method
BR112021008973A8 (en) 2018-11-09 2023-05-02 Juno Therapeutics Inc SPECIFIC T CELL RECEPTORS FOR MESOTHELIN AND THEIR USE IN IMMUNOTHERAPY
GB201818715D0 (en) 2018-11-16 2019-01-02 British American Tobacco Investments Ltd Method
AU2019406778A1 (en) 2018-12-17 2021-07-22 Massachusetts Institute Of Technology Crispr-associated transposase systems and methods of use thereof
WO2020132659A1 (en) 2018-12-21 2020-06-25 Precision Biosciences, Inc. Genetic modification of the hydroxyacid oxidase 1 gene for treatment of primary hyperoxaluria
CA3123890A1 (en) 2019-01-04 2020-07-09 Cargill Incorporated Engineered nucleases to generate mutations in plants
US11739156B2 (en) 2019-01-06 2023-08-29 The Broad Institute, Inc. Massachusetts Institute of Technology Methods and compositions for overcoming immunosuppression
KR20210138569A (en) 2019-01-11 2021-11-19 아퀴타스 테라퓨틱스 인크. Lipids for Lipid Nanoparticle Delivery of Active Agents
GB201900940D0 (en) 2019-01-23 2019-03-13 British American Tobacco Investments Ltd Method
CN112805026A (en) 2019-02-06 2021-05-14 桑格摩生物治疗股份有限公司 Methods for treating mucopolysaccharidosis type I
US20230053540A1 (en) 2019-02-19 2023-02-23 Massachusetts Institute Of Technology Treatment of liver injury
CN114026116A (en) 2019-02-20 2022-02-08 弗雷德哈钦森癌症研究中心 RAS neoantigen specific binding proteins and uses thereof
JP2022521010A (en) 2019-02-21 2022-04-04 イッスム・リサーチ・デベロプメント・カムパニー・オブ・ザ・ヘブリュー・ユニバシティー・オブ・エルサレム リミテッド Methods for reducing drug-induced nephrotoxicity
EP3934417A1 (en) 2019-03-05 2022-01-12 The State of Israel - Ministry of Agriculture & Rural Development, Agricultural Research Organization (ARO) (Volcani Center) Genome-edited birds
CA3132845A1 (en) 2019-03-11 2020-09-17 Fred Hutchinson Cancer Research Center High avidity wt1 t cell receptors and uses thereof
WO2020186101A1 (en) 2019-03-12 2020-09-17 The Broad Institute, Inc. Detection means, compositions and methods for modulating synovial sarcoma cells
WO2020186237A1 (en) 2019-03-13 2020-09-17 The Broad Institute, Inc. Microglial progenitors for regeneration of functional microglia in the central nervous system and therapeutics uses thereof
GB201903521D0 (en) 2019-03-14 2019-05-01 Tropic Biosciences Uk Ltd No title
GB201903519D0 (en) 2019-03-14 2019-05-01 Tropic Biosciences Uk Ltd Introducing silencing activity to dysfunctional rna molecules and modifying their specificity against a gene of interest
WO2020186235A1 (en) 2019-03-14 2020-09-17 The Broad Institute, Inc. Compositions and methods for modulating cgrp signaling to regulate intestinal innate lymphoid cells
GB201903520D0 (en) 2019-03-14 2019-05-01 Tropic Biosciences Uk Ltd Modifying the specificity of non-coding rna molecules for silencing genes in eukaryotic cells
US20220152148A1 (en) 2019-03-18 2022-05-19 The Broad Institute, Inc. Modulation of type 2 immunity by targeting clec-2 signaling
EP3942023A1 (en) 2019-03-18 2022-01-26 The Broad Institute, Inc. Compositions and methods for modulating metabolic regulators of t cell pathogenicity
EP3946384A1 (en) 2019-04-02 2022-02-09 Sangamo Therapeutics, Inc. Methods for the treatment of beta-thalassemia
KR20210148293A (en) 2019-04-03 2021-12-07 프리시젼 바이오사이언시스 인코포레이티드 Gene-modified immune cells containing microRNA-adapted shRNAs (shRNAmiRs)
US20220204994A1 (en) 2019-04-05 2022-06-30 Precision Biosciences, Inc. Methods of preparing populations of genetically-modified immune cells
KR20220005555A (en) 2019-05-07 2022-01-13 프리시젼 바이오사이언시스 인코포레이티드 Optimization of engineered meganucleases for recognition sequences
UY38693A (en) 2019-05-13 2020-12-31 Kws Saat Se & Co Kgaa DROUGHT TOLERANCE IN CORN
GB201906768D0 (en) 2019-05-14 2019-06-26 British American Tobacco Investments Ltd Method
WO2020232271A1 (en) 2019-05-14 2020-11-19 The Broad Institute, Inc. Compositions and methods for targeting multinucleated cells
AR118995A1 (en) 2019-05-25 2021-11-17 Kws Saat Se & Co Kgaa HAPLOID INDUCTION ENHANCER
WO2020243371A1 (en) 2019-05-28 2020-12-03 Massachusetts Institute Of Technology Methods and compositions for modulating immune responses
US20220243178A1 (en) 2019-05-31 2022-08-04 The Broad Institute, Inc. Methods for treating metabolic disorders by targeting adcy5
BR112021026248A2 (en) 2019-06-27 2022-03-03 Two Blades Found Isolated nucleic acid molecule encoding an engineered atrlp23 protein, engineered atrlp23 protein, expression cassette, vector, host cell, plant or plant cell, methods for making an engineered atrlp23 protein, for making a nucleic acid molecule encoding an atrlp23 protein engineered and to intensify the resistance of a plant
GB201909563D0 (en) 2019-07-03 2019-08-14 British American Tobacco Investments Ltd Method
GB201909562D0 (en) 2019-07-03 2019-08-14 British American Tobacco Investments Ltd Method
AU2020300101A1 (en) 2019-07-04 2022-02-17 Ukko Inc. De-epitoped alpha gliadin and use of same for the management of celiac disease and gluten sensitivity
WO2021011348A1 (en) 2019-07-12 2021-01-21 The Regents Of The University Of California Plants with enhanced resistance to bacterial pathogens
CN114127304A (en) 2019-07-15 2022-03-01 免疫医疗有限公司 Tri-moiety systems for protein dimerization and methods of use
IL268111A (en) 2019-07-16 2021-01-31 Fainzilber Michael Methods of treating pain
WO2021016608A1 (en) 2019-07-25 2021-01-28 Precision Biosciences, Inc. Compositions and methods for sequential stacking of nucleic acid sequences into a genomic locus
WO2021019536A1 (en) 2019-07-30 2021-02-04 The State Of Israel, Ministry Of Agriculture & Rural Development, Agricultural Research Organization (Aro) (Volcani Center) Methods of controlling cannabinoid synthesis in plants or cells and plants and cells produced thereby
US20220267732A1 (en) 2019-08-01 2022-08-25 Sana Biotechnology, Inc. Dux4 expressing cells and uses thereof
EP3772542A1 (en) 2019-08-07 2021-02-10 KWS SAAT SE & Co. KGaA Modifying genetic variation in crops by modulating the pachytene checkpoint protein 2
US20220282333A1 (en) 2019-08-13 2022-09-08 The General Hospital Corporation Methods for predicting outcomes of checkpoint inhibition and treatment thereof
CN114555790A (en) 2019-08-20 2022-05-27 弗雷德哈钦森癌症研究中心 WT-1 specific T cell immunotherapy
EP4017526A1 (en) 2019-08-20 2022-06-29 Precision BioSciences, Inc. Lymphodepletion dosing regimens for cellular immunotherapies
WO2021035170A1 (en) 2019-08-21 2021-02-25 Precision Biosciences, Inc. Compositions and methods for tcr reprogramming using fusion proteins
CA3148370A1 (en) 2019-08-23 2021-03-04 Sana Biotechnology, Inc. Cd24 expressing cells and uses thereof
WO2021041922A1 (en) 2019-08-30 2021-03-04 The Broad Institute, Inc. Crispr-associated mu transposase systems
US20230002459A1 (en) 2019-10-02 2023-01-05 Sangamo Therapeutics, Inc. Zinc Finger Protein Transcription Factors for Treatment of Prion Disease
EP4041754A1 (en) 2019-10-02 2022-08-17 Sangamo Therapeutics, Inc. Zinc finger protein transcription factors for repressing alpha-synuclein expression
US11793787B2 (en) 2019-10-07 2023-10-24 The Broad Institute, Inc. Methods and compositions for enhancing anti-tumor immunity by targeting steroidogenesis
AU2020366566A1 (en) 2019-10-17 2022-04-21 KWS SAAT SE & Co. KGaA Enhanced disease resistance of crops by downregulation of repressor genes
US20220411479A1 (en) 2019-10-30 2022-12-29 Precision Biosciences, Inc. Cd20 chimeric antigen receptors and methods of use for immunotherapy
US11844800B2 (en) 2019-10-30 2023-12-19 Massachusetts Institute Of Technology Methods and compositions for predicting and preventing relapse of acute lymphoblastic leukemia
IL270306A (en) 2019-10-30 2021-05-31 Yeda Res & Dev Prevention and treatment of pre-myeloid and myeloid malignancies
US20220401481A1 (en) 2019-11-01 2022-12-22 Magenta Therapeutics, Inc. Dosing regimens for the mobilization of hematopoietic stem and progenitor cells
AU2019474716A1 (en) 2019-11-19 2022-07-07 Protalix Ltd. Removal of constructs from transformed cells
EP4069285A1 (en) 2019-12-06 2022-10-12 Precision BioSciences, Inc. Methods for cancer immunotherapy, using lymphodepletion regimens and cd19, cd20 or bcma allogeneic car t cells
US20230031465A1 (en) 2019-12-06 2023-02-02 Precision Biosciences, Inc. Optimized engineered meganucleases having specificity for a recognition sequence in the hepatitis b virus genome
EP3835309A1 (en) 2019-12-13 2021-06-16 KWS SAAT SE & Co. KGaA Method for increasing cold or frost tolerance in a plant
IL271656A (en) 2019-12-22 2021-06-30 Yeda Res & Dev Systems and methods for identifying cells that have undergone genome editing
EP4096647A1 (en) 2020-01-30 2022-12-07 Yeda Research and Development Co. Ltd Treating acute liver disease with tlr-mik inhibitors
WO2021158915A1 (en) 2020-02-06 2021-08-12 Precision Biosciences, Inc. Recombinant adeno-associated virus compositions and methods for producing and using the same
TW202202155A (en) 2020-03-25 2022-01-16 美商薩那生物科技公司 Hypoimmunogenic neural cells for the treatment of neurological disorders and conditions
CN115697044A (en) 2020-03-31 2023-02-03 艾洛生物系统有限公司 Regulation of endogenous mogroside pathway genes in watermelon and other cucurbitaceae
US20230159945A1 (en) 2020-04-09 2023-05-25 R.J. Reynolds Tobacco Company Method
AU2021263754A1 (en) 2020-04-27 2022-12-01 Ensoma, Inc. Methods and compositions for transducing hematopoietic stem and progenitor cells in vivo
CA3177621A1 (en) 2020-05-06 2021-11-11 Alexandre Juillerat Methods to genetically modify cells for delivery of therapeutic proteins
EP4146812A1 (en) 2020-05-06 2023-03-15 Cellectis S.A. Methods for targeted insertion of exogenous sequences in cellular genomes
WO2021231259A1 (en) 2020-05-11 2021-11-18 Precision Biosciences, Inc. Self-limiting viral vectors encoding nucleases
US20230193230A1 (en) 2020-05-12 2023-06-22 Precision Biosciences, Inc. Treatment of retinitis pigmentosa using improved engineered meganucleases
IL298138A (en) 2020-05-12 2023-01-01 Univ Pennsylvania Compositions for drg-specific reduction of transgene expression
US20230190871A1 (en) 2020-05-20 2023-06-22 Sana Biotechnology, Inc. Methods and compositions for treatment of viral infections
AR122206A1 (en) 2020-05-29 2022-08-24 Kws Saat Se & Co Kgaa INDUCTION OF HAPLOIDS IN PLANTS
WO2022035793A1 (en) 2020-08-10 2022-02-17 Precision Biosciences, Inc. Antibodies and fragments specific for b-cell maturation antigen and uses thereof
AU2021325941A1 (en) 2020-08-13 2023-03-09 Sana Biotechnology, Inc. Methods of treating sensitized patients with hypoimmunogenic cells, and associated methods and compositions
WO2022038536A1 (en) 2020-08-18 2022-02-24 International Rice Research Institute Methods of increasing outcrossing rates in gramineae
AU2021329403A1 (en) 2020-08-21 2023-05-04 Precision Biosciences, Inc. Engineered meganucleases having specificity for a recognition sequence in the transthyretin gene
EP4204545A2 (en) 2020-08-25 2023-07-05 Kite Pharma, Inc. T cells with improved functionality
WO2022066973A1 (en) 2020-09-24 2022-03-31 Fred Hutchinson Cancer Research Center Immunotherapy targeting pbk or oip5 antigens
WO2022066965A2 (en) 2020-09-24 2022-03-31 Fred Hutchinson Cancer Research Center Immunotherapy targeting sox2 antigens
WO2022067122A1 (en) 2020-09-25 2022-03-31 Sangamo Therapeutics, Inc. Zinc finger fusion proteins for nucleobase editing
EP4225902A1 (en) 2020-10-05 2023-08-16 Protalix Ltd. Dicer-like knock-out plant cells
TW202222841A (en) 2020-10-06 2022-06-16 福瑞德哈金森腫瘤研究中心 Compositions and methods for treating mage-a1-expressing disease
US20230365995A1 (en) 2020-10-07 2023-11-16 Precision Biosciences, Inc. Lipid nanoparticle compositions
WO2022079719A1 (en) 2020-10-15 2022-04-21 Yeda Research And Development Co. Ltd. Method of treating myeloid malignancies
WO2022087527A1 (en) 2020-10-23 2022-04-28 Elo Life Systems, Inc. Methods for producing vanilla plants with improved flavor and agronomic production
US20230407333A1 (en) 2020-10-29 2023-12-21 The Trustees Of The University Of Pennsylvania Aav capsids and compositions containing same
EP4244342A1 (en) 2020-11-12 2023-09-20 Precision BioSciences, Inc. Engineered meganucleases having specificity for recognition sequences in the dystrophin gene
EP4251641A1 (en) 2020-11-26 2023-10-04 Ukko Inc. Modified high molecular weight glutenin subunit and uses thereof
WO2022132836A2 (en) 2020-12-14 2022-06-23 Fred Hutchinson Cancer Research Center Compositions and methods for cellular immunotherapy
IL279559A (en) 2020-12-17 2022-07-01 Yeda Res & Dev Controlling ubiquitination of mlkl for treatment of disease
JP2024501971A (en) 2020-12-31 2024-01-17 サナ バイオテクノロジー,インコーポレイテッド Methods and compositions for modulating CAR-T activity
WO2022150616A1 (en) 2021-01-08 2022-07-14 Precision Biosciences, Inc. Engineered meganucleases having specificity for a recognition sequence in the hydroxyacid oxidase 1 gene
WO2022165111A1 (en) 2021-01-28 2022-08-04 Precision Biosciences, Inc. Modulation of tgf beta signaling in genetically-modified eukaryotic cells
EP4308694A1 (en) 2021-03-16 2024-01-24 Magenta Therapeutics, Inc. Dosing regimens for hematopoietic stem cell mobilization for stem cell transplants in multiple myeloma patients
WO2022226303A1 (en) 2021-04-22 2022-10-27 Precision Biosciences, Inc. Engineered meganucleases that target human mitochondrial genomes
CA3173245A1 (en) 2021-04-22 2022-10-22 James Jefferson Smith Engineered meganucleases that target human mitochondrial genomes
WO2022226316A1 (en) 2021-04-22 2022-10-27 Precision Biosciences, Inc. Compositions and methods for generating male sterile plants
TW202309066A (en) 2021-04-27 2023-03-01 賓州大學委員會 Porcine-derived adeno-associated virus capsids and uses thereof
BR112023023486A2 (en) 2021-05-10 2024-01-30 Yissum Res Dev Co Of Hebrew Univ Jerusalem Ltd PHARMACEUTICAL COMPOSITION, AND, METHOD OF TREATMENT OF A DISEASE OR CONDITION IN WHICH A BENEFICIAL CLINICAL EFFECT IS OBTAINED BY THE REDUCTION IN NEURONAL NITRIC OXIDE SYNTHASE ACTIVITY
EP4336997A1 (en) 2021-05-11 2024-03-20 Two Blades Foundation Methods for preparing a library of plant disease resistance genes for functional testing for disease resistance
EP4342983A1 (en) 2021-05-18 2024-03-27 Cells & Genes Biotech (Shanghai) Co., Ltd Method for modifying cell
BR112023024434A2 (en) 2021-05-27 2024-02-20 Sana Biotechnology Inc HYPOIMMUNOGENIC CELLS COMPRISING GENETICALLY MODIFIED HLA-E OR HLA-G
WO2023275255A1 (en) 2021-07-02 2023-01-05 Tropic Biosciences UK Limited Delay or prevention of browning in banana fruit
IL310089A (en) 2021-07-14 2024-03-01 Sana Biotechnology Inc Altered expression of y chromosome-linked antigens in hypoimmunogenic cells
TW202317602A (en) 2021-07-15 2023-05-01 福瑞德哈金森腫瘤中心 Chimeric polypeptides
WO2023006933A1 (en) 2021-07-30 2023-02-02 KWS SAAT SE & Co. KGaA Plants with improved digestibility and marker haplotypes
IL310550A (en) 2021-08-04 2024-03-01 Univ Colorado Regents Lat activating chimeric antigen receptor t cells and methods of use thereof
CA3228570A1 (en) 2021-08-10 2023-02-16 Yona Geffen Engineered nk cells, methods of their production and uses thereof
AU2022325231A1 (en) 2021-08-11 2024-02-08 Sana Biotechnology, Inc. Genetically modified cells for allogeneic cell therapy to reduce complement-mediated inflammatory reactions
AU2022325955A1 (en) 2021-08-11 2024-02-08 Sana Biotechnology, Inc. Genetically modified cells for allogeneic cell therapy to reduce instant blood mediated inflammatory reactions
CA3227108A1 (en) 2021-08-11 2023-02-16 Xiaomeng HU Genetically modified primary cells for allogeneic cell therapy
AU2022326565A1 (en) 2021-08-11 2024-02-08 Sana Biotechnology, Inc. Genetically modified cells for allogeneic cell therapy
AU2022327174A1 (en) 2021-08-11 2024-02-15 Sana Biotechnology, Inc. Inducible systems for altering gene expression in hypoimmunogenic cells
GB202112865D0 (en) 2021-09-09 2021-10-27 Tropic Biosciences Uk Ltd Resistance to black sigatoka disease in banana
GB202112866D0 (en) 2021-09-09 2021-10-27 Tropic Biosciences Uk Ltd Resistance to fusarium wilt in a banana
CA3233676A1 (en) 2021-09-30 2023-04-06 The Sainsbury Laboratory Plant disease resistance genes against stem rust and methods of use
WO2023064872A1 (en) 2021-10-14 2023-04-20 Precision Biosciences, Inc. Combinations of anti-bcma car t cells and gamma secretase inhibitors
WO2023070002A2 (en) 2021-10-19 2023-04-27 Precision Biosciences, Inc. Gene editing methods for treating alpha-1 antitrypsin (aat) deficiency
WO2023070003A1 (en) 2021-10-19 2023-04-27 Precision Biosciences, Inc. Gene editing methods for treating alpha-1 antitrypsin (aat) deficiency
WO2023069790A1 (en) 2021-10-22 2023-04-27 Sana Biotechnology, Inc. Methods of engineering allogeneic t cells with a transgene in a tcr locus and associated compositions and methods
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
WO2023081767A1 (en) 2021-11-05 2023-05-11 Precision Biosciences, Inc. Methods for immunotherapy
US11753677B2 (en) 2021-11-10 2023-09-12 Encodia, Inc. Methods for barcoding macromolecules in individual cells
WO2023087019A2 (en) 2021-11-15 2023-05-19 The Trustees Of The University Of Pennsylvania Compositions for drg-specific reduction of transgene expression
WO2023091910A1 (en) 2021-11-16 2023-05-25 Precision Biosciences, Inc. Methods for cancer immunotherapy
WO2023093862A1 (en) 2021-11-26 2023-06-01 Epigenic Therapeutics Inc. Method of modulating pcsk9 and uses thereof
WO2023102550A2 (en) 2021-12-03 2023-06-08 The Broad Institute, Inc. Compositions and methods for efficient in vivo delivery
WO2023111130A1 (en) 2021-12-17 2023-06-22 Tropic Biosciences UK Limited Modified agrobacteria for editing plants
WO2023122722A1 (en) 2021-12-22 2023-06-29 Sangamo Therapeutics, Inc. Novel zinc finger fusion proteins for nucleobase editing
WO2023122337A1 (en) 2021-12-23 2023-06-29 Sana Biotechnology, Inc. Chimeric antigen receptor (car) t cells for treating autoimmune disease and associated methods
WO2023133525A1 (en) 2022-01-07 2023-07-13 Precision Biosciences, Inc. Optimized polynucleotides for protein expression
WO2023141602A2 (en) 2022-01-21 2023-07-27 Renagade Therapeutics Management Inc. Engineered retrons and methods of use
WO2023148478A1 (en) 2022-02-03 2023-08-10 Nicoventures Trading Limited Method of modulating alkaloid content in tobacco plants
WO2023148476A1 (en) 2022-02-03 2023-08-10 Nicoventures Trading Limited Method of modulating alkaloid content in tobacco plants
WO2023148475A1 (en) 2022-02-04 2023-08-10 Nicoventures Trading Limited Method of modulating alkaloid content in tobacco plants
WO2023154578A1 (en) 2022-02-14 2023-08-17 Sana Biotechnology, Inc. Methods of treating patients exhibiting a prior failed therapy with hypoimmunogenic cells
WO2023158836A1 (en) 2022-02-17 2023-08-24 Sana Biotechnology, Inc. Engineered cd47 proteins and uses thereof
WO2023161178A1 (en) 2022-02-22 2023-08-31 Ecole Polytechnique Federale De Lausanne (Epfl) Cgas super-enzymes for cancer immunotherapy
WO2023173123A1 (en) 2022-03-11 2023-09-14 Sana Biotechnology, Inc. Genetically modified cells and compositions and uses thereof
WO2023196818A1 (en) 2022-04-04 2023-10-12 The Regents Of The University Of California Genetic complementation compositions and methods
GB202205148D0 (en) 2022-04-07 2022-05-25 Nicoventures Trading Ltd Method
GB202205149D0 (en) 2022-04-07 2022-05-25 Nicoventures Trading Ltd Method
GB202205562D0 (en) 2022-04-14 2022-06-01 Nicoventures Trading Ltd Method
GB202205561D0 (en) 2022-04-14 2022-06-01 Nicoventures Trading Ltd Method
GB202206109D0 (en) 2022-04-27 2022-06-08 Nicoventures Trading Ltd Method
GB202206107D0 (en) 2022-04-27 2022-06-08 Nicoventures Trading Ltd Method
WO2023215725A1 (en) 2022-05-02 2023-11-09 Fred Hutchinson Cancer Center Compositions and methods for cellular immunotherapy
WO2023215498A2 (en) 2022-05-05 2023-11-09 Modernatx, Inc. Compositions and methods for cd28 antagonism
WO2023220040A1 (en) 2022-05-09 2023-11-16 Synteny Therapeutics, Inc. Erythroparvovirus with a modified capsid for gene therapy
WO2023220043A1 (en) 2022-05-09 2023-11-16 Synteny Therapeutics, Inc. Erythroparvovirus with a modified genome for gene therapy
WO2023220035A1 (en) 2022-05-09 2023-11-16 Synteny Therapeutics, Inc. Erythroparvovirus compositions and methods for gene therapy
EP4278891A1 (en) 2022-05-20 2023-11-22 KWS SAAT SE & Co. KGaA Clubroot resistance and markers in brassica
WO2024015966A2 (en) 2022-07-15 2024-01-18 The Trustees Of The University Of Pennsylvania Recombinant aav having aav clade d and clade e capsids and compositions containing same
WO2024040222A1 (en) 2022-08-19 2024-02-22 Generation Bio Co. Cleavable closed-ended dna (cedna) and methods of use thereof
WO2024044723A1 (en) 2022-08-25 2024-02-29 Renagade Therapeutics Management Inc. Engineered retrons and methods of use
WO2024042199A1 (en) 2022-08-26 2024-02-29 KWS SAAT SE & Co. KGaA Use of paired genes in hybrid breeding
WO2024056902A2 (en) 2022-09-16 2024-03-21 Christopher Shaw Compositions and methods for treating neurological diseases
GB202305021D0 (en) 2023-04-04 2023-05-17 Tropic Biosciences Uk Ltd Methods for generating breaks in a genome

Family Cites Families (42)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DE122007000007I1 (en) 1986-04-09 2007-05-16 Genzyme Corp Genetically transformed animals secreting a desired protein in milk
US4873316A (en) 1987-06-23 1989-10-10 Biogen, Inc. Isolation of exogenous recombinant proteins from the milk of transgenic mammals
US6156304A (en) 1990-12-20 2000-12-05 University Of Pittsburgh Of The Commonwealth System Of Higher Education Gene transfer for studying and treating a connective tissue of a mammalian host
US5384253A (en) 1990-12-28 1995-01-24 Dekalb Genetics Corporation Genetic transformation of maize cells by electroporation of cells pretreated with pectin degrading enzymes
WO1993004169A1 (en) 1991-08-20 1993-03-04 Genpharm International, Inc. Gene targeting in animal cells using isogenic dna constructs
US5792640A (en) 1992-04-03 1998-08-11 The Johns Hopkins University General method to clone hybrid restriction endonucleases using lig gene
US5436150A (en) 1992-04-03 1995-07-25 The Johns Hopkins University Functional domains in flavobacterium okeanokoities (foki) restriction endonuclease
US5792632A (en) 1992-05-05 1998-08-11 Institut Pasteur Nucleotide sequence encoding the enzyme I-SceI and the uses thereof
US6395959B1 (en) 1992-05-05 2002-05-28 Institut Pasteur Nucleotide sequence encoding the enzyme I SceI and the use thereof
US5474896A (en) 1992-05-05 1995-12-12 Institut Pasteur Nucleotide sequence encoding the enzyme I-SceI and the uses thereof
US5997861A (en) 1994-10-31 1999-12-07 Burstein Laboratories, Inc. Antiviral supramolecules containing target-binding molecules and therapeutic molecules bound to spectrin
US5830729A (en) 1996-04-18 1998-11-03 Institut Pasteur I Sce I-induced gene replacement and gene conversion in embryonic stem cells
US6265196B1 (en) 1996-05-07 2001-07-24 Johns Hopkins University Methods for inactivating target DNA and for detecting conformational change in a nucleic acid
US6056973A (en) 1996-10-11 2000-05-02 Sequus Pharmaceuticals, Inc. Therapeutic liposome composition and method of preparation
US6060082A (en) 1997-04-18 2000-05-09 Massachusetts Institute Of Technology Polymerized liposomes targeted to M cells and useful for oral or mucosal drug delivery
US7041814B1 (en) 1998-02-18 2006-05-09 Genome Therapeutics Corporation Nucleic acid and amino acid sequences relating to Enterobacter cloacae for diagnostics and therapeutics
CA2360878A1 (en) 1999-02-03 2000-08-10 The Children's Medical Center Corporation Gene repair involving excision of targeting dna
JP2002535995A (en) 1999-02-03 2002-10-29 ザ チルドレンズ メディカル センター コーポレイション Gene repair involving induction of double-stranded DNA breaks at chromosomal target sites
US6794136B1 (en) 2000-11-20 2004-09-21 Sangamo Biosciences, Inc. Iterative optimization in the design of binding proteins
US6511676B1 (en) 1999-11-05 2003-01-28 Teni Boulikas Therapy for human cancers using cisplatin and other drugs or genes encapsulated into liposomes
US6593308B2 (en) 1999-12-03 2003-07-15 The Regents Of The University Of California Targeted drug delivery with a hyaluronan ligand
WO2001055443A1 (en) 2000-01-31 2001-08-02 Pharmacia & Upjohn Company Crystallization and structure determination of staphylococcus aureus nad synthetase
DE10131786A1 (en) 2001-07-04 2003-01-16 Sungene Gmbh & Co Kgaa Recombination systems and methods for removing nucleic acid sequences from the genome of eukaryotic organisms
US7314712B2 (en) 2001-07-27 2008-01-01 The United States Of America As Represented By The Secretary Of The Department Of Health And Human Services Systems for in vivo site-directed mutagenesis using oligonucleotides
WO2003087341A2 (en) 2002-01-23 2003-10-23 The University Of Utah Research Foundation Targeted chromosomal mutagenesis using zinc finger nucleases
JP2005520519A (en) 2002-03-15 2005-07-14 セレクティス Hybrid and single chain meganucleases and uses thereof
DE10226316B4 (en) * 2002-06-10 2004-10-28 Siemens Ag Method for setting up an additional service in a mobile radio network
EP2806025B1 (en) 2002-09-05 2019-04-03 California Institute of Technology Use of zinc finger nucleases to stimulate gene targeting
WO2004031346A2 (en) 2002-09-06 2004-04-15 Fred Hutchinson Cancer Research Center Methods and compositions concerning designed highly-specific nucleic acid binding proteins
US7285699B2 (en) 2002-10-07 2007-10-23 Stowers Institute For Medical Research Ends-out gene targeting method
US20030175968A1 (en) 2002-10-30 2003-09-18 Golic Kent G. Gene targeting method
EP1590453B1 (en) 2003-01-28 2013-11-27 Cellectis Custom-made meganuclease and use thereof
US7888121B2 (en) 2003-08-08 2011-02-15 Sangamo Biosciences, Inc. Methods and compositions for targeted cleavage and recombination
PL1689870T3 (en) 2003-11-18 2009-06-30 Bayer Cropscience Nv Improved targeted dna insertion in plants
EP1591521A1 (en) 2004-04-30 2005-11-02 Cellectis I-Dmo I derivatives with enhanced activity at 37 degrees C and use thereof
WO2007034262A1 (en) 2005-09-19 2007-03-29 Cellectis Heterodimeric meganucleases and use thereof
WO2006097784A1 (en) 2005-03-15 2006-09-21 Cellectis I-crei meganuclease variants with modified specificity, method of preparation and uses thereof
EP1863909B2 (en) * 2005-03-15 2014-09-10 Cellectis I-crei meganuclease variants with modified specificity, method of preparation and uses thereof
WO2007049095A1 (en) 2005-10-25 2007-05-03 Cellectis Laglidadg homing endonuclease variants having mutations in two functional subdomains and use thereof
WO2007060495A1 (en) 2005-10-25 2007-05-31 Cellectis I-crei homing endonuclease variants having novel cleavage specificity and use thereof
WO2007093836A1 (en) 2006-02-13 2007-08-23 Cellectis Meganuclease variants cleaving a dna target sequence from a xp gene and uses thereof
WO2008010009A1 (en) 2006-07-18 2008-01-24 Cellectis Meganuclease variants cleaving a dna target sequence from a rag gene and uses thereof

Also Published As

Publication number Publication date
ES2440801T3 (en) 2014-01-30
EP2341135A2 (en) 2011-07-06
EP1945762A2 (en) 2008-07-23
US8163514B2 (en) 2012-04-24
US8377674B2 (en) 2013-02-19
US8129134B2 (en) 2012-03-06
EP3246403A3 (en) 2018-02-14
EP2650366A3 (en) 2014-02-12
US20120021465A1 (en) 2012-01-26
US20120021521A1 (en) 2012-01-26
EP3795682A2 (en) 2021-03-24
DK2628794T3 (en) 2016-08-15
DK2484758T3 (en) 2014-01-06
JP2024001024A (en) 2024-01-09
US20130227720A1 (en) 2013-08-29
CA2891996A1 (en) 2007-04-26
EP2368981A2 (en) 2011-09-28
US8143016B2 (en) 2012-03-27
EP2357226A1 (en) 2011-08-17
WO2007047859A2 (en) 2007-04-26
US20070117128A1 (en) 2007-05-24
US8119361B2 (en) 2012-02-21
CA2626262C (en) 2015-09-08
EP2650365A1 (en) 2013-10-16
EP2628794A3 (en) 2013-12-04
DK2650366T3 (en) 2017-07-10
JP2018039808A (en) 2018-03-15
JP6148437B2 (en) 2017-06-14
EP2484758A1 (en) 2012-08-08
DK1945762T3 (en) 2013-07-29
US20120030783A1 (en) 2012-02-02
DK2360244T3 (en) 2015-04-07
JP2022070928A (en) 2022-05-13
US8133697B2 (en) 2012-03-13
AU2006304668B2 (en) 2013-03-07
US8021867B2 (en) 2011-09-20
JP6050611B2 (en) 2016-12-21
EP3246403B1 (en) 2020-08-26
EP2628794B1 (en) 2016-05-04
ES2533370T3 (en) 2015-04-09
AU2006304668A1 (en) 2007-04-26
DK3246403T3 (en) 2020-10-26
EP2327774A3 (en) 2011-09-28
EP2662442A1 (en) 2013-11-13
ES2602184T3 (en) 2017-02-20
US20120266266A1 (en) 2012-10-18
EP2341135A3 (en) 2011-10-12
EP2650366B1 (en) 2017-03-22
DK2650365T3 (en) 2016-12-05
EP2650366A2 (en) 2013-10-16
JP2017048179A (en) 2017-03-09
US20210284980A1 (en) 2021-09-16
US20120015421A1 (en) 2012-01-19
EP3246403A2 (en) 2017-11-22
EP3795682A3 (en) 2021-06-16
WO2007047859A3 (en) 2007-11-15
US20120021520A1 (en) 2012-01-26
JP2019077698A (en) 2019-05-23
US20170335299A1 (en) 2017-11-23
ES2539616T3 (en) 2015-07-02
EP2368981A3 (en) 2011-10-12
DK2662442T3 (en) 2015-07-06
EP2628794A2 (en) 2013-08-21
JP2013034473A (en) 2013-02-21
US20200109383A1 (en) 2020-04-09
EP2484758B1 (en) 2013-10-02
JP5937292B2 (en) 2016-06-22
EP2327774A2 (en) 2011-06-01
US20120040405A1 (en) 2012-02-16
US20120070877A1 (en) 2012-03-22
US8119381B2 (en) 2012-02-21
ATE549401T1 (en) 2012-03-15
ES2626025T3 (en) 2017-07-21
US8124369B2 (en) 2012-02-28
US20120028315A1 (en) 2012-02-02
DK2357226T3 (en) 2012-06-25
US20120015406A1 (en) 2012-01-19
US20120028314A1 (en) 2012-02-02
US8148098B2 (en) 2012-04-03
US20120040406A1 (en) 2012-02-16
EP2357226B1 (en) 2012-03-14
US8304222B1 (en) 2012-11-06
JP2009511085A (en) 2009-03-19
ES2384440T3 (en) 2012-07-04
ES2829549T3 (en) 2021-06-01
EP2360244B1 (en) 2014-12-24
ES2425022T3 (en) 2013-10-10
US20120264189A1 (en) 2012-10-18
US8143015B2 (en) 2012-03-27
EP2650365B1 (en) 2016-09-14
JP2021054830A (en) 2021-04-08
EP1945762B1 (en) 2013-05-01
US20150135345A1 (en) 2015-05-14
JP2012196214A (en) 2012-10-18
ES2582091T3 (en) 2016-09-09
CA3055968A1 (en) 2007-04-26
EP2360244A1 (en) 2011-08-24
EP2662442B1 (en) 2015-03-25

Similar Documents

Publication Publication Date Title
US20210284980A1 (en) Rationally-designed meganucleases with altered sequence specificity and dna-binding affinity
AU2008335324A1 (en) Rationally-designed meganucleases with recognition sequences found in DNase hypersensitive regions of the human genome

Legal Events

Date Code Title Description
EEER Examination request