US20130227715A1 - Use of endonucleases for inserting transgenes into safe harbor loci - Google Patents

Use of endonucleases for inserting transgenes into safe harbor loci Download PDF

Info

Publication number
US20130227715A1
US20130227715A1 US13/702,066 US201113702066A US2013227715A1 US 20130227715 A1 US20130227715 A1 US 20130227715A1 US 201113702066 A US201113702066 A US 201113702066A US 2013227715 A1 US2013227715 A1 US 2013227715A1
Authority
US
United States
Prior art keywords
locus
seq
target sequence
protein
endonuclease
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.)
Abandoned
Application number
US13/702,066
Inventor
Olivier Danos
Aymeric Duclert
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.)
Cellectis SA
Original Assignee
Cellectis SA
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Cellectis SA filed Critical Cellectis SA
Priority to US13/702,066 priority Critical patent/US20130227715A1/en
Assigned to CELLECTIS reassignment CELLECTIS ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: DANOS, OLIVIER, DUCLERT, AYMERIC
Assigned to CELLECTIS reassignment CELLECTIS ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: DANOS, OLIVIER, DUCLERT, AYMERIC
Publication of US20130227715A1 publication Critical patent/US20130227715A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • 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
    • C12N15/87Introduction of foreign genetic material using processes not otherwise provided for, e.g. co-transformation
    • C12N15/90Stable introduction of foreign DNA into chromosome
    • 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)
    • 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

Definitions

  • the present invention concerns the endonucleases capable of cleaving a target sequence located in a “safe harbor loci”, i.e. a loci allowing safe expression of a transgene.
  • the present invention further concerns the use of such endonucleases for inserting transgenes into a cell, tissue or organism.
  • Meganucleases also referred to as homing endonucleases, were the first endonucleases used to induce double-strand breaks and recombination in living cells (Rouet et al. PNAS 1994 91:6064-6068; Rouet et al. Mol Cell Biol. 1994 14:8096-8106; Choulika et al. Mol Cell Biol. 1995 15:1968-1973; Puchta et al. PNAS 1996 93:5055-5060).
  • their use has long been limited by their narrow specificity.
  • these processes rely on the identifications of locally engineered variants with a substrate specificity that differs from the substrate specificity of the wild-type meganuclease by only a few nucleotides. Up to four sets of mutations identified in such proteins can then be assembled in new proteins in order to generate new meganucleases with entirely redesigned binding interface.
  • the integrase from the Streptomyces phage PhiC31 was used early for targeted gene transfer in an endogenous locus.
  • This enzyme mediates recombination of the phage genome into the bacterial chromosome through a site-specific reaction between the phage attachment site (attP) and the bacterial attachment site (attB) (Kuhstoss et al. J Mol Biol 1991 222:897-908; Rausch et al. NAR 1991 19:5187-5189).
  • This can occur from plasmids carrying attB sites into native genomic sequences harboring partial identity with attP, called pseudo attP sites (attP′).
  • the PhiC31 integrase has been used to transfer several transgenes, including hFIX, in the human genome (Olivares et al. Nat Biotech 2002 20:1124-1128; Ginsburg et al. Adv Genet. 2005 54:179-187; Calos Curr Gene Ther 2006 6:633-645; Chalberg et al. J Mol Biol 2006 357:28-48; Aneja et al. J Gene Med 2007 9:967-975).
  • the drawback here is that the site where integration can occur cannot be chosen (Chalberg et al. J Mol Biol 2006 357:28-48), and one has to rely on pseudo attP sites within the human genome loci, for precise integration.
  • Site-specific recombinases such as the Cre recombinase from bacteriophage P1, or the Flp protein from Saccharomyces cerevisiae have been used to induce recombination between pre-engineered sequences containing their cognate sites.
  • the Cre recombinase recognizes and mediates recombination between two identical 34 bp sites known as loxP (Abremski et al. Cell 1983 32:1301-1311).
  • loxP Adbremski et al. Cell 1983 32:1301-131.
  • an engineered Cre recombinase variant has been used to excise an HIV type 1 provirus (Sarkar et al. Science 2007 316:1912-1915) from cells.
  • the recombinase was redesigned to target the proviral LTRs, and used to induce the excision of all intervening sequences.
  • Engineering attempts have also been made with the Flp recombinase, targeting the FRT (Flp Recombination Target) sequence (Buchholzt et al. Nat Biotech 1998 16:657-662), and variants recognizing non-native Flp recombination targets have been obtained (Voziyanov et al. J Mol Biol 2003 326:65-76).
  • FRT Fep Recombination Target
  • Transposons such as Piggy Back and Sleeping Beauty can provide efficient tools for insertion of sequences into vertebrate cells and have been proposed as an alternative to viral mediated gene delivery to achieve long-lasting expression (Izsvak et al. Mol ther 2004 9:147-156; Ivics et al. Curr Gene Ther 2006 6:593-607; Mates et al. Nat Genet. 2009 41:753-761).
  • Transposons are a natural means of gene delivery in which a DNA sequence present in a DNA molecule is inserted in another location, through the action of the transposase.
  • An engineered SB transposase, called SB100X was recently shown to increase the efficiency of the process (Mates et al. Nat Genet. 2009 41:753-761).
  • Transposition is random on a genomic level (for example, SB integrates into TA dinucleotides (Vigdal et al. J Mol Biol 2002 323:441-452), and should therefore not be considered as tools for targeted approaches.
  • SB integrates into TA dinucleotides
  • chromosomal transposition mediated by engineered transposases in human cells, by fusing the transposase catalytic domain to specific DNA binding domains (Ivics, et al. Mol Ther 2007 15:1137-1144), paving the way for the development of a new category of targeted tools.
  • Site-specific integration would be a promising alternative to random integration of viral vectors since it could alleviate the risks of insertional mutagenesis (Kolb et al. Trends Biotechnol. 2005 23:399-406; Porteus et al. Nat. Biotechnol. 2005 23:967-973; Paques et al. Curr Gen Ther. 2007 7:49-66).
  • insertional mutagenesis Kolb et al. Trends Biotechnol. 2005 23:399-406; Porteus et al. Nat. Biotechnol. 2005 23:967-973; Paques et al. Curr Gen Ther. 2007 7:49-66.
  • each tool has its intrinsic properties in terms of activity and specificity.
  • a variant endonuclease capable of cleaving a target sequence for use in inserting a transgene into the genome of an individual, wherein
  • locus is selected from the SH3 locus on human chromosome 6p25.1, the SH4 locus on human chromosome 7q31.2, the SH6 locus on human chromosome 21q21.1, the SH12 locus on human chromosome 13q34, the SH13 locus on human chromosome 3p12.2, the SH19 locus on human chromosome 22, the SH20 locus on human chromosome 12q21.2, the SH21 locus on human chromosome 3p24.1, the SH33 locus on human chromosome 6p12.2, the SH7 locus on human chromosome 2p16.1 and the SH8 locus on human chromosome 5.
  • a variant dimeric I-CreI protein comprising two monomers that comprise a sequence at least 80% identical to SEQ ID NO: 1 or SEQ ID NO: 42, wherein:
  • dimeric I-CreI protein according to embodiment 11, wherein said dimeric I-CreI protein is capable of cleaving a target sequence located within the SH3 locus on human chromosome 6p25.1.
  • the dimeric I-CreI protein according to embodiment 12, wherein said target sequence comprises the sequence of SEQ ID NO: 2.
  • polypeptide comprises:
  • polypeptide comprises:
  • polypeptide comprises:
  • dimeric I-CreI protein according to embodiment 11, wherein said dimeric I-CreI protein is capable of cleaving a target sequence located within the SH4 locus on human chromosome 7q31.2.
  • the dimeric I-CreI protein according to embodiment 18, wherein said target sequence comprises the sequence of SEQ ID NO: 3.
  • polypeptide comprises:
  • dimeric I-CreI protein according to embodiment 11, wherein said dimeric I-CreI protein is capable of cleaving a target sequence located within the SH6 locus on human chromosome 21q21.1.
  • polypeptide comprises:
  • polypeptide comprises:
  • a fusion protein comprising the monomers of the dimeric I-CreI protein according to any one of embodiments 11 to 26.
  • fusion protein according to embodiment 27, wherein said monomers are connected by a peptidic linker comprising a sequence of SEQ ID NO: 43.
  • fusion protein according to any one of embodiments 27 to 29, wherein said fusion protein comprises a sequence selected from the group consisting of SEQ ID Nos. 25-40 and 76-96.
  • An expression vector comprising the nucleic acid according to embodiment 31.
  • the expression vector according to embodiment 32 further comprising a targeting construct comprising a transgene and two sequences homologous to the genomic sequence flanking a target sequence recognized by the endonuclease as defined in one of embodiments 1-9 or by the protein as defined in any one of embodiments 11 to 30.
  • a pharmaceutical composition comprising the expression vector as defined in any one of embodiments 32 to 34 or the combination as defined in embodiment 36 and a pharmaceutically active carrier.
  • a method of treating an individual by gene therapy comprising administering an effective amount of the expression vector as defined in any one of embodiments 32 to 34 or of the combination as defined in embodiment 36 to an individual in need thereof.
  • a method for obtaining an endonuclease suitable for inserting a transgene into the genome of an individual comprising the step of:
  • embodiment 40 for making a non-human animal model of a hereditary disorder.
  • embodiment 40 for producing a recombinant protein.
  • a non-human transgenic animal comprising a nucleic acid according to embodiment 31, or an expression vector according to any one of embodiments 32-34, or a combination according to embodiment 36 in its genome.
  • the inventors have identified “safe harbors” loci within the genome allowing safe expression of a transgene through targeted insertion wherein (i) said loci are close to a retroviral insertion site identified in a cell from a patient treated by gene therapy, and (ii) said retroviral insertion are not associated with cancer or abnormal cell proliferation.
  • the safe harbor loci according to the invention may either be located within the intron of a gene, or within an intergenic region.
  • endonucleases could be engineered in such a way as to target said safe harbors for gene addition.
  • the inventors have engineered several I-CreI meganucleases that are capable of recognizing and cleaving target sequences located within different safe harbors loci, for instance the SH6, the SH3 locus, the SH4 locus, the SH12 locus, the SH13 locus, the SH19, the SH20 locus, the SH21 locus, the SH33 locus, the SH7 locus, the SH8 locus, the SH18 locus, the SH31 locus, the SH38 locus, the SH39 locus, the SH41 locus, the SH42 locus, the SH43 locus, the SH44 locus, the SH45 locus, the SH46 locus, the SH47 locus, the SH48 locus, the SH49 locus, the SH50 locus, the SH51 locus, the SH52 locus, the SH70 locus, the SH71 locus, the SH72 locus, the SH73 locus, the SH74 locus, the SH75 locus, the SH101 locus, the SH106 locus, the SH107 loci
  • meganucleases as well as other enymes like integrases, recombinases and transposases, can therefore be used as a tool for inserting a transgene into safe harbors, thereby avoiding the appearance of adverse events such as leukemia in the frame of gene therapy.
  • these meganucleases, as well as other enymes like integrases, recombinases and transposases can be used for inserting any transgene into the safe harbor starting from a single targeting construct irrespective of the sequence of the transgene.
  • the invention therefore relates to:
  • the term “endonuclease” refers to any wild-type or variant enzyme capable of catalyzing the hydrolysis (cleavage) of bonds between nucleic acids within of a DNA or RNA molecule, preferably a DNA molecule.
  • the endonucleases according to the present invention do not cleave the DNA or RNA molecule irrespective of its sequence, but recognize and cleave the DNA or RNA molecule at specific polynucleotide sequences, further referred to as “target sequences” or “target sites”.
  • Target sequences recognized and cleaved by an endonuclease according to the invention are referred to as target sequences according to the invention.
  • the endonuclease according to the invention can for example be a homing endonuclease (Paques et al. Curr Gen Ther. 2007 7:49-66), a chimeric Zinc-Finger nuclease (ZFN) resulting from the fusion of engineered zinc-finger domains with the catalytic domain of a restriction enzyme such as Fokl (Porteus et al. Nat. Biotechnol. 2005 23:967-973) or a chemical endonuclease (Arimondo et al. Mol Cell Biol. 2006 26:324-333; Simon et al. NAR 2008 36:3531-3538; Eisenschmidt et al.
  • a homing endonuclease Paques et al. Curr Gen Ther. 2007 7:49-66
  • ZFN Zinc-Finger nuclease
  • a chemical or peptidic cleaver is conjugated either to a polymer of nucleic acids or to another DNA recognizing a specific target sequence, thereby targeting the cleavage activity to a specific sequence.
  • the endonuclease according to the invention is preferably a homing endonuclease, also known under the name of meganuclease.
  • homing endonucleases are well-known to the art (see e.g. Stoddard, Quarterly Reviews of Biophysics, 2006, 38:49-95).
  • Homing endonucleases recognize a DNA target sequence and generate a single- or double-strand break.
  • Homing endonucleases are highly specific, recognizing DNA target sites ranging from 12 to 45 base pairs (bp) in length, usually ranging from 14 to 40 bp in length.
  • the homing endonuclease according to the invention may for example correspond to a LAGLIDADG endonuclease, to a HNH endonuclease, or to a GIY-YIG endonuclease.
  • endonuclease examples include I-Sce I, I-Chu I, I-Cre I, I-Csm I, PI-Sce I, PI-Tli I, PI-Mtu I, I-Ceu I, I-Sce II, I-Sce III, HO, PI-Civ I, PI-Ctr I, PI-Aae I, PI-Bsu I, PI-Dha I, PI-Dra I, PI-Mav I, PI-Mch I, PI-Mfu I, PI-Mfl I, PI-Mga I, PI-Mgo I, PI-Min I, PI-Mka I, PI-Mle I, PI-Mma I, PI-Msh I, PI-Msm I, PI-Mth I, PI-Mtu I, PI-Mxe I, PI-Npu I, PI-Pfu I,
  • the homing endonuclease according to the invention is a LAGLIDADG endonuclease such as I-SceI, I-CreI, I-CeuI, I-MsoI, and 1-DmoI.
  • said LAGLIDADG endonuclease is I-CreI.
  • Wild-type I-CreI is a homodimeric homing endonuclease that is capable of cleaving a 22 to 24 bp double-stranded target sequence.
  • the sequence of a wild-type monomer of I-CreI includes the sequence shown as SEQ ID NO: 1 (which corresponds to the I-CreI sequence of pdb accession number 1g9y) and the sequence shown in SwissProt Accession n° P05725 (in particular the sequence shown in version 73, last modified Nov. 3, 2009).
  • the I-CreI variants may comprise an additional alanine after the first methionine of the wild type I-CreI sequence, and three additional amino acid residues at the C-terminal extremity (see sequence of SEQ ID NO: 42 and FIG. 11 ). These three additional amino acid residues consist of two additional alanine residues and one aspartic acid residue after the final proline of the wild type I-CreI sequence. These additional residues do not affect the properties of the enzyme. For the sake of clarity, these additional residues do not affect the numbering of the residues in I-CreI or variants thereof. More specifically, the numbering used herein exclusively refers to the position of residues in the wild type I-CreI enzyme of SEQ ID NO: 1. For instance, the second residue of wild-type I-CreI is in fact the third residue of a variant of SEQ ID NO: 42 since this variant comprises an additional alanine after the first methionine.
  • I-CreI variants may be homodimers (meganuclease comprising two identical monomers), heterodimers (meganuclease comprising two non-identical monomers) and single-chains.
  • the invention encompasses both wild-type (naturally-occurring) and variant endonucleases.
  • the endonuclease according to the invention is a “variant” endonuclease, i.e. an endonuclease that does not naturally exist in nature and that is obtained by genetic engineering or by random mutagenesis.
  • the variant endonuclease according to the invention can for example be obtained by substitution of at least one residue in the amino acid sequence of a wild-type, naturally-occurring, endonuclease with a different amino acid. Said substitution(s) can for example be introduced by site-directed mutagenesis and/or by random mutagenesis.
  • such variant endonucleases remain functional, i.e. they retain the capacity of recognizing and specifically cleaving a target sequence.
  • the variant endonuclease according to the invention cleaves a target sequence that is different from the target sequence of the corresponding wild-type endonuclease.
  • the target sequence of a variant I-CreI endonuclease is different from the sequence of SEQ ID NO: 4.
  • the present invention is based on the finding that such variant endonucleases with novel specificities can be used for inserting a gene into a “safe harbor” locus of the genome of a cell, tissue or individual.
  • locus is the specific physical location of a DNA sequence (e.g. of a gene) on a chromosome. As used in this specification, the term “locus” usually refers to the specific physical location of an endonuclease's target sequence on a chromosome. Such a locus, which comprises a target sequence that is recognized and cleaved by an endonuclease according to the invention, is referred to as “locus according to the invention”.
  • insertion into a safe harbor locus should have no impact on the expression of other genes. Testing these properties is a multi-step process, and a first pre-screening of candidate safe harbor loci by bioinformatic means is desirable. One can thus first identify loci in which targeted insertion is unlikely to result in insertional mutagenesis.
  • One of the major features of a locus according to the invention is that (i) it is located in a region wherein retroviral insertion was observed in a cell from a patient, in a gene therapy clinical trial, and (ii) said retroviral insertion has not been associated with a cancer or an abnormal cell proliferation.
  • one way to identify safe habor loci is to use the data generated by former gene therapy trials.
  • insertions of retroviral vector-borne transgenes next to the LMO2 and CCND2 genes have been shown to be associated with leukemia.
  • the follow up of vector insertions in patients have clearly demonstrated that cells carrying this insertion had outnumbered the other modified cells after a several years process (Hacein-Bey-Abina et al. Science 2003 302:415-9; Deichmann et al. J. of Clin. Invest. 2007 117:2225-32, Cavazzana-Calvo et al. Blood 2007 109:4575-4581).
  • RIS Retroviral Insertion Sites
  • the locus according to the invention is defined as a locus comprising a target sequence that is located at a distance of at most 200, 180, 150, 100 or 50 kb from a retroviral insertion site (RIS), said RIS being neither associated with cancer nor with abnormal cell proliferation.
  • RIS retroviral insertion site
  • Such loci are referred to as “safe harbor” loci according to the invention (or loci according to the invention), i.e. loci that are safe for insertion of transgenes.
  • RIS Riviral insertion sites
  • RIS a genomic site which was identified as an insertion site for a retroviral vector in a cell from a patient treated by gene therapy with said retroviral vector.
  • RIS are well-known to the art. They include but are not limited to those described in Schwarzwaelder et al. (J. Clin. Invest. 2007 117:2241), Deichmann et al. (J. of Clin. Invest. 2007 117:2225), Aiuti et al. (J. Clin. Invest. 2007 117:2233), Recchia et al. (PNAS 2006 103:1457) and Mavilio et al. (Nature Medicine 12:1397, 2006).
  • retroviral vector any vector derived from a virus from the retroviridae family.
  • the RIS according to the invention is neither associated with cancer nor with abnormal cell proliferation.
  • RIS known to be associated with leukemia or with abnormal cell proliferation are well known in the art and can easily be excluded by the skilled in the art.
  • Such RIS known to be associated with leukemia or with abnormal cell proliferation include, e.g., insertion sites next to the LMO2, CCND2, MDS1-EVI1, PRDM16, and SETBP1 genes.
  • the RIS used to define safe harbor loci have been identified in a clinical trial, with the transduced cells being stem cells.
  • the RIS can thus have been identified in cells from a patient treated by gene therapy by transduction of stem cells.
  • the RIS used to define safe harbor loci have been identified in a clinical trial for SCID patients, with the transduced cells being hematopoietic stem cells (HSCs).
  • HSCs hematopoietic stem cells
  • the RIS can thus have been identified in cells from a patient treated by gene therapy by transduction of hematopoietic stem cells.
  • CIS Common Integration sites
  • the target sequence according to the invention is not located in a CIS,
  • said target sequence or locus is preferably located at a distance of at least 50, 100 or 200 kb from a RIS being part of a common integration site (CIS).
  • CIS Common Integration site
  • CIS is meant a genomic region of 30 kb, 50 kb, 100 kb or 200 kb wherein RIS identified in clinical trials are overrepresented (assuming a random distribution of insertions).
  • Such CIS are well known in the art and are described in Schwarzwaelder et al. (J. Clin. Invest. 2007 117:2241), Deichmann et al. (J. of Clin. Invest. 2007 117:2225), Aiuti et al. (J. Clin. Invest. 2007 117:2233), Recchia et al. (PNAS 2006 103:1457), Mavilio et al. (Nature Medicine 12:1397, 2006) and Gabriel et al. (Nat. Med. 2009 15(12):143.
  • targeted integration into the locus according to the invention should not result in the disruption of essential functions in the targeted cell.
  • insertion into the locus according to the invention does preferably not substantially modify expression of genes located in the vicinity of the target sequence, for example of the nearest genes.
  • insertion of a genetic element into said locus does preferably not substantially modify the phenotype of said cell, tissue or individual (except for the phenotype due to expression of the genetic element).
  • phenotype is meant a cell's, a tissue's or an individual's observable traits. The phenotype includes e.g. the viability, the cellular proliferation and/or the growth rate.
  • the skilled in the art can easily verify that a locus is a safe harbor locus according to the invention e.g. by analyzing the expression pattern of adjacent genes, by carrying out micro-array studies of transcriptome and/or by characterizing proliferation and/or differentiation abnormalities (if any).
  • the locus according to the invention does not comprise any gene.
  • a locus that does not comprise any gene refers to a locus that does not comprise any referenced or known gene.
  • such a locus does not comprise any known gene according to sequence databases such as those available on the National Center for Biotechnology Information (NCBI) website. Therefore, the target sequence according to the invention and/or the locus according to the invention can advantageously be located at a distance of at least 1, 5, 10, 25, 50, 100, 180, 200, 250, 300, 400 or 500 kb from the nearest genes.
  • gene is meant the basic unit of heredity, consisting of a segment of DNA arranged in a linear manner along a chromosome, which codes for a specific protein or segment of protein.
  • a gene typically includes a promoter, a 5′ untranslated region, one or more coding sequences (exons), optionally introns, a 3′ untranslated region.
  • the gene may further comprise a terminator, enhancers and/or silencers.
  • nearest genes is meant the one, two or three genes that are located the closest to the target sequence, centromeric and telomeric to the target sequence respectively.
  • the locus according to the invention further allows stable expression of the transgene.
  • the target sequence according to the invention is only present once within the genome of said cell, tissue or individual.
  • a safe harbor locus according to the invention, one can then (i) either construct a variant endonuclease specifically recognizing and cleaving a target sequence located within said locus, e.g. as described in Examples 1, 2 and 5, or (ii) determine whether a known wild-type endonuclease is capable of cleaving a target sequence located within said locus.
  • the skilled in the art can insert therein a target sequence that is recognized and cleaved by a known wild-type or variant endonuclease.
  • the invention is drawn to a method for obtaining an endonuclease suitable for inserting a transgene into the genome of an individual, comprising the step of:
  • RIS being part of a CIS may be excluded, and/or the genomic region defined at step (b) may only extend 50 kb upstream and 50 kb downstream of said RIS, and/or the locus comprising the target sequence may not comprise any gene.
  • the locus according to the invention may for example correspond to any one of the SH3, SH4, SH6, SH12, SH13, SH19, SH20, SH21, SH33, SH7 or SH8 loci which are described in Tables A to C below.
  • Table A provides the location of the locus within the human genome, a target sequence comprised within the locus, the location of the closest RIS as well as the reference to a publication describing the RIS, and examples of endonucleases according to the invention that cleave the locus.
  • Table B provides information about the nearest genes that are located immediately upstream (at 5′) and downstream (at 3′) of the locus according to the invention.
  • the distance indicates the distance between the target sequence and the nearest coding sequence of the gene.
  • Table C and D provide similar information as Table B, but for the second nearest genes and for the third nearest genes, respectively.
  • Tables A′, B′, C′ and D′ provide updated information similar to that in Tables A, B, C and D, respectively, for some loci and associated examples of target sequences within these loci, namely SH3, SH4, SH6, SH8 and SH19. Updated localization information is given by reference to GRCh37/hg19 version of the human genome assembly.
  • the locus according to the invention may also correspond to any one of the SH18, SH31, SH38, SH39, SH41, SH42, SH43, SH44, SH45, SH46, SH47, SH48, SH49, SH50, SH51, SH52, SH70, SH71, SH72, SH73, SH74 and SH75 which are described in Tables A′′ to D′′ below.
  • Table A′′ provides the location of the locus within the human genome, a target sequence comprised within the locus, the location of the closest RIS as well as the reference to a publication describing the RIS, the distance between said target and the closest RIS and examples of endonucleases according to the invention that cleave the locus.
  • Table B′′ provides information about the nearest genes that are located immediately upstream (at 5′) and downstream (at 3′) of the locus according to the invention.
  • the distance indicates the distance between the target sequence and the nearest coding sequence of the gene.
  • Table C′′ and D′′ provide similar information as Table B′′, but for the second nearest genes and for the third nearest genes, respectively.
  • loci Locations of loci, targets in this loci and genes are given according to GRCh37/hg19 version of the human genome assembly.
  • the locus according to the invention may also correspond to any one of the SH101, SH106, SH107, SH102, SH105, SH103, SH104, SH113, SH109, SH112, SH108, SH110, SH114, SH116, SH111, SH115, SH121, SH120, SH122, SH117, SH118, SH119, SH123, SH126, SH128, SH129, SH124, SH131, SH125, SH127 and SH130 which are described in Tables E and F below.
  • Table E provides the location of the locus within the human genome, a target sequence comprised within the locus, the location of the closest RIS as well as the reference to a publication describing the RIS, the distance between said target and the closest RIS and examples of endonucleases according to the invention that cleave the locus.
  • Table F provides information about the nearest genes that are located immediately upstream (at 5′) and downstream (at 3′) of the locus according to the invention.
  • the distance indicates the distance between the target sequence and the nearest coding sequence of the gene.
  • loci Locations of loci, targets in this loci and genes are given in Tables E and F according to GRCh36.3/hg19 version of the human genome assembly.
  • the locus according to the invention may also correspond to any one of the SH125, SH127, SH130, SH102, SH105, SH103, SH104, SH117, SH118, SH119 and SH123 which are described in Table G below.
  • Table G provides examples of target sequences located in introns of genes which are mentioned and examples of endonucleases according to the invention that cleave said intronic locus.
  • the locus according to the invention may also contains any one of the SH11, SH12, SH13, SH17, SH19, SH20, SH21, SH23, SH33, SH34, SH40, SH53, SH54, SH55, SH56, SH57, SH58, SH59, SH60, SH61, SH62, SH65, SH67, SH68 and SH69 which are given in Tables H below.
  • Table H provides target sequences comprised within these loci as well as examples of endonucleases according to the invention that cleave these target sequences.
  • the locus according to the invention is the SH3 locus.
  • SH3 locus refers to the region of human chromosome 6 that is located at about 120 kb centromeric to the gene encoding the lymphocyte antigen 86 (see e.g.
  • the SH3 locus extends from position 6850510 to 6853677 of the sequence shown in NC 000006.11. It comprises a sequence of SEQ ID NO: 54.
  • the locus according to the invention is the SH4 locus.
  • the SH4 locus is defined herein as the region of human chromosome 7 that is located at about 320 kb telomeric to MyoD family inhibitor domain containing locus (MDFIC), or to the homologous region in another species (see e.g.
  • the SH4 locus extends from position 114972751 to 114976380 of the sequence shown in NC 000007.13. It comprises a sequence of SEQ ID NO: 55.
  • transgene refers to a sequence encoding a polypeptide.
  • the polypeptide encoded by the transgene is either not expressed, or expressed but not biologically active, in the cell, tissue or individual in which the transgene is inserted.
  • the transgene encodes a therapeutic polypeptide useful for the treatment of an individual.
  • the individual may be a human or non-human animal.
  • the individual is preferably a human.
  • the individual can be a non-human animal, preferably a vertebrate and/or a mammalian animal such as e.g. a mouse, a rat, a rabbit, a Chinese hamster, a Guinea pig or a monkey.
  • the cells and tissues according to the invention are preferably derived from such human or non-human animals.
  • variant endonuclease according to the invention can for example be derived:
  • the invention pertains to a dimeric I-CreI protein comprising or consisting of two monomers, each monomer comprising or consisting of a sequence at least 80%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% identical to SEQ ID NO: 1 or to SEQ ID NO: 42, wherein said dimeric I-CreI protein is capable of cleaving a target sequence located within a safe harbor locus.
  • the target sequence neither comprises nor consists of a sequence of SEQ ID NO: 4.
  • the dimeric I-CreI protein according to the invention is a heterodimeric protein.
  • a protein having a sequence at least, for example, 95% “identical” to a query sequence of the present invention it is intended that the sequence of the protein is identical to the query sequence except that the sequence may include up to five nucleotide mutations per each 100 amino acids of the query sequence.
  • up to 5% (5 of 100) of the amino acids of the sequence may be inserted, deleted, or replaced with another nucleotide.
  • the needle program is for example available on the ebi.ac.uk world wide web site.
  • the percentage of identity in accordance with the invention can thus be calculated using the EMBOSS::needle (global) program with a “Gap Open” parameter equal to 10.0, a “Gap Extend” parameter equal to 0.5, and a Blosum62 matrix.
  • Each monomer of the dimeric I-CreI protein according to the invention may for example comprise at least, at most or about 2, 5, 8, 10, 12, 15, 18, 20 or 25 mutations compared with the sequence of a wild-type monomer (SEQ ID NO: 1) or with a monomer of SEQ ID NO: 42.
  • the monomer according to the invention comprises a sequence that differs from SEQ ID NO: 1 or SEQ ID NO: 42 by at least, at most or about 2, 5, 8, 10, 12, 15, 18, 20, 25 or 30 mutations.
  • the mutation preferably corresponds to a substitution of one amino acid with another amino acid. Therefore, a preferred embodiment according to the invention is directed to a dimeric I-CreI protein comprising or consisting of two monomers comprising a sequence at least 80%, identical to SEQ ID NO: 1 or SEQ ID NO: 42, wherein said sequence only differs from SEQ ID NO: 1 or SEQ ID NO: 42 by the presence of amino acid substitutions.
  • the monomers of the dimeric I-CreI protein according to the invention are preferably derived from monomers comprising or consisting of the sequence of SEQ ID NO: 42.
  • the mutations are preferably located at positions of the I-CreI sequence that are involved in recognition of the target sequence. Indeed, introducing such mutations allow designing meganucleases with novel specificities.
  • the monomers may also have mutations corresponding to:
  • the monomers of the protein according to the invention may comprise one or more amino acids added at the NH 2 terminus and/or COOH terminus of the sequence, such as a Tag useful in purification of the protein, a propeptide and/or a nuclear localization signal.
  • the monomers of the protein according to the invention may comprise AAD amino acids added at the COOH terminus of the sequence of SEQ ID NO: 1, as is the case in a monomer of SEQ ID NO: 42.
  • a monomer comprising a 44A mutation refers to a I-CreI monomer in which the amino acid at position 44 of SEQ ID NO: 1 (i.e. a glutamine, Q) is replaced with an alanine (A).
  • this monomer differs from the wild-type I-CreI monomer of SEQ ID NO: 1 by at least the following amino acid substitution: Q44A.
  • the I-CreI monomer of SEQ ID NO: 42 comprises some additional amino acid residues compared to the I-CreI monomer of SEQ ID NO: 1 (see FIG. 11 ). Therefore, on SEQ ID NO: 42, the 44A mutation corresponds to a replacement of the glutamine at position 45 of SEQ ID NO: 42 with an alanine.
  • a monomer comprising 44A 54L 64A 70Q 75N 158R 162A mutations may for example have the sequence of SEQ ID NO: 57 (when this monomer is directly derived from a I-CreI monomer of SEQ ID NO: 1) or the sequence of SEQ ID NO: 58 (when this monomer is directly derived from a I-CreI monomer of SEQ ID NO: 42).
  • FIG. 12 shows an alignment between two such monomers, and indicates the position of the 44A 54L 64A 70Q 75N 158R and 162A mutations on these monomers.
  • dimeric I-CreI proteins capable of cleaving target sequences located in the SH3, SH4 or SH6 locus, are further described below.
  • the target sequence is located within the SH3 locus (defined hereabove).
  • the target sequence located within SH3 may for example comprise or consist of SEQ ID NO: 2, or of nucleotides 2 to 23 of SEQ ID NO: 2.
  • Example 1 discloses several examples of heterodimeric I-CreI proteins according to the invention capable of cleaving such a target sequence.
  • methods for constructing other such proteins are well-known in the art and include e.g. those described in PCT applications WO 2006/097784, WO 2006/097853 and WO 2009019614, and in Arnould et al. (J. Mol. Biol., 2006, 355:443-458).
  • the monomers of such a dimeric protein preferably comprise at least one, preferably at least 3, 4, 5 or 6, amino acid substitutions located at a position selected from the group consisting of positions 4, 24, 26, 28, 30, 32, 33, 38, 44, 50, 54, 57, 64, 66, 70, 71, 75, 77, 81, 86, 92, 105, 142, 151, 154, 158 and 162 of SEQ ID NO: 1, preferably positions 4, 30, 38, 44, 50, 54, 57, 64, 66, 70, 71, 75, 77, 81, 86, 92, 105, 142, 151, 154, 158 and 162 of SEQ ID NO: 1.
  • substitutions may for example be selected from the following substitutions: 4E, 30G, 38R, 44A, 50R, 54L, 57E, 64A, 66C, 70Q, 70D, 71 R, 75N, 75Y, 77V, 81T, 86D, 92R, 105A, 142R, 151A, 154G, 158R, 158W and 162A.
  • the dimeric protein may optionally comprise a mutation at position 1, however, such a mutation has no influence on cleavage activity or on cleavage specificity.
  • Such dimeric I-CreI proteins may for example comprise or consist of:
  • the dimeric I-CreI protein according the invention comprises or consists of:
  • the dimeric I-CreI protein according the invention comprises or consists of:
  • the dimeric I-CreI protein according the invention comprises or consists of:
  • the monomers of the dimeric I-CreI protein may also comprise additional mutations, for example allowing the obtention of an obligate heterodimer.
  • additional mutations are known to the skilled in the art and include those described in Fajardo-Sanchez et al. (Nucleic Acids Res. 2008 36:2163-73).
  • the above monomers are directly derived from a monomer of SEQ ID NO: 42, and differ from the sequence of SEQ ID NO: 42 only by the presence of the indicated mutations.
  • the target sequence is located within the SH4 locus (defined hereabove).
  • the target sequence located within SH4 may for example comprise or consist of SEQ ID NO: 3, or of nucleotides 2 to 23 of SEQ ID NO: 3.
  • Example 2 discloses several examples of dimeric I-CreI proteins according to the invention capable of cleaving such a target sequence.
  • the monomers of such a dimeric protein preferably comprise at least one, preferably at least 3, 4, 5 or 6, amino acid substitutions located at a position selected from the group consisting of positions 24, 44, 68, 70, 75 and 77 of SEQ ID NO: 1.
  • Said substitutions may for example be selected from the following substitutions: 24V, 44R, 44Y, 68Y, 68A, 70S, 70D, 75Y, 75N, 77R, 77N and 77V.
  • Such dimeric I-CreI proteins may for example comprise or consist of:
  • the dimeric I-CreI protein according the invention comprises or consists of:
  • the monomers of the dimeric I-CreI protein may also comprise additional mutations, for example allowing the obtention of an obligate heterodimer.
  • additional mutations are known to the skilled in the art and include those described in Fajardo-Sanchez et al. (Nucleic Acids Res. 2008 36:2163-73).
  • the above monomers are directly derived from a monomer of SEQ ID NO: 42, and differ from the sequence of SEQ ID NO: 42 only by the presence of the indicated mutations.
  • the target sequence is located within the SH6 locus (defined hereabove).
  • the target sequence located within SH6 may for example comprise or consist of SEQ ID NO: 59, or of nucleotides 2 to 23 of SEQ ID NO: 59.
  • Example 5 discloses several examples of dimeric I-CreI proteins according to the invention capable of cleaving such a target sequence.
  • the monomers of such a dimeric protein preferably comprise at least one, preferably at least 3, 4, 5 or 6, amino acid substitutions located at a position selected from the group consisting of positions 7, 24, 27, 28, 34, 40, 44, 68, 70, 75, 77, 81, 85, 96, 99, 103, 108, 111, 121, 132, 144 and 160 of SEQ ID NO: 1.
  • substitutions may for example be selected from the following substitutions: 7R, 24F, 27V, 28Q, 34R, 40R, 44A, 44K, 68T, 70L, 70G, 70S, 75N, 77V, 81T, 81V, 85R, 96R, 99R, 103T, 103S, 108V, 111H, 121E, 132V, 144S, 160R and 160E.
  • Such dimeric I-CreI proteins may for example comprise or consist of:
  • the dimeric I-CreI protein according the invention comprises or consists of:
  • the dimeric I-CreI protein according the invention comprises or consists of:
  • the monomers of the dimeric I-CreI protein may also comprise additional mutations, for example allowing the obtention of an obligate heterodimer.
  • additional mutations are known to the skilled in the art and include those described in Fajardo-Sanchez et al. (Nucleic Acids Res. 2008 36:2163-73).
  • the above monomers are directly derived from a monomer of SEQ ID NO: 42, and differ from the sequence of SEQ ID NO: 42 only by the presence of the indicated mutations.
  • Fusion proteins comprising the two monomers of a dimeric I-CreI protein fused together and retaining the biological activity of the parent dimeric I-CreI protein can be constructed (Grizot et al. NAR 2009 37:5405; Li et al. Nucleic Acids Res. 2009 37:1650-62; Epinat et al. Nucleic Acids Res. 2003 31:2952-62). Such fusion proteins are commonly referred to as “single-chain meganucleases”.
  • the invention further relates to a fusion protein comprising the two monomers of the dimeric I-CreI protein as defined hereabove, or biologically active fragments of such monomers.
  • a fusion protein comprising the two monomers of the dimeric I-CreI protein as defined hereabove, or biologically active fragments of such monomers.
  • the first and second monomers of a dimeric I-CreI protein as defined hereabove are fused together and are optionally connected to each other by a linker such as a peptidic linker.
  • the linker may for example comprise or consist of SEQ ID NO: 43 or SEQ ID NO: 326.
  • a fusion protein according to the invention is capable of cleaving a target sequence according to the invention, i.e., it is capable of cleaving the same target sequence as the dimeric I-CreI protein from which it is derived.
  • the single chain meganuclease of the present invention further comprises obligate heterodimer mutations as described above so as to obtain single chain obligate heterodimer meganuclease variants.
  • This design greatly decreases off-site cleavage and toxicity while enhancing efficacy.
  • the structure and stability of this single-chain molecule are very similar to those of the dimeric variants and this molecule appears to be monomeric in solution. In all respects, this single-chain molecule performs as well as I-SceI considered to be gold standard in terms of specificity. These properties place this new generation of meganucleases among the best molecular scissors available for genome surgery strategies and should facilitate gene correction therapy for monogenetic diseases, such as for example severe combined immunodeficiency (SCID), while potentially avoiding the deleterious effects of previous gene therapy approaches.
  • SCID severe combined immunodeficiency
  • the C-terminal monomer may comprise the K7E and K96E mutations
  • the N-terminal monomer may comprise the E8K, E61 R and G19S mutations.
  • Examples 1, 2 and 5 disclose several examples of such fusion proteins according to the invention.
  • the fusion protein according to the invention comprises or consists of a sequence at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to any one of SEQ ID Nos. 25-40 and 76-96, or to a fragment of at least 50, 100, 150 or 200 amino acids thereof.
  • the endonuclease according to the invention is preferably introduced to said cell, tissue or animal as a nucleic acid molecule rather than as a protein.
  • the invention pertains to a nucleic acid encoding the endonuclease according to the invention, e.g. encoding a dimeric I-CreI protein or a fusion protein described hereabove.
  • the endonuclease is a dimeric I-CreI protein
  • said nucleic acid comprises at least two coding sequences, one for each monomer.
  • the endonuclease is a fusion protein
  • said nucleic acid comprises at least one coding sequence.
  • the endonuclease protein can be combined with a variety of cell-penetrating peptide leading to a recombinant protein; such combined molecules are able to enter target cells at much higher levels of efficiency than the endonuclease alone.
  • vector comprising such a nucleic acid according to the invention.
  • vector is meant a nucleic acid molecule capable of transporting another nucleic acid to which it has been linked.
  • Vectors which can be used in the present invention includes but is not limited to viral vectors, plasmids and YACs, which may consist of chromosomal, non chromosomal, semisynthetic or synthetic nucleic acids.
  • Preferred vectors are those capable of autonomous replication (episomal vector) and/or expression of nucleic acids to which they are linked (expression vectors). Large numbers of suitable vectors are known to those of skill in the art and commercially available.
  • the vector is a viral vector such as e.g. a vector derived from a retrovirus, an adenovirus, a parvovirus (e.g. an adeno-associated viruses), a coronavirus, a negative strand RNA virus (e.g. an orthomyxovirus such as influenza virus, a rhabdovirus such as rabies and vesicular stomatitis virus, a paramyxovirus such as measles and Sendai virus), a positive strand RNA virus such as picornavirus and alphavirus, or a double-stranded DNA virus such as adenovirus, herpesvirus (e.g.
  • a viral vector such as e.g. a vector derived from a retrovirus, an adenovirus, a parvovirus (e.g. an adeno-associated viruses), a coronavirus, a negative strand RNA virus (e.g. an orthomyxovirus such as influenza virus
  • Herpes Simplex virus types 1 and 2 Epstein-Barr virus, cytomegalovirus
  • poxvirus e.g. vaccinia, fowlpox and canarypox
  • Preferred vectors include lentiviral vectors, and particularly self-inactivacting lentiviral vectors.
  • the vector can also comprise elements such as:
  • said vector is an “expression vector”, i.e. a vector in which at least one coding sequence is operatively linked to transcriptional and translational control elements.
  • the nucleic acid encoding the endonuclease according to the invention e.g. encoding the dimeric I-CreI protein or the fusion protein described hereabove
  • the vector according to the invention comprises a targeting construct comprising a transgene and two sequences homologous to the genomic sequence flanking the target sequence as defined herein (e.g. the target sequence of SEQ ID NO: 2 or 3).
  • the genomic sequences flanking the target sequence are preferably immediately adjacent to the target site.
  • Such targeting constructs are well-known to the skilled in the art.
  • such constructs typically comprise a first sequence that is homologous to the upstream (5′) genomic sequence flanking the target sequence, the transgene to be inserted, and a second fragment that is homologous to the downstream (3′) genomic sequence flanking the target sequence.
  • homologous is intended a sequence with enough identity to another one to lead to a homologous recombination between sequences, more particularly having at least 95% identity, preferably 97% identity and more preferably 99% identity to each other.
  • homologous sequences of at least 50 bp, preferably more than 100 bp and more preferably more than 200 bp are used. Therefore, the targeting DNA construct is preferably from 200 pb to 6000 pb, more preferably from 1000 pb to 2000 pb. Indeed, shared DNA homologies are located in regions flanking upstream and downstream the site of the break and the DNA sequence to be introduced should be located between the two arms.
  • the targeting construct may also comprise a positive selection marker between the two homology arms and eventually a negative selection marker upstream of the first homology arm or downstream of the second homology arm.
  • the marker(s) allow(s) the selection of cells having inserted the sequence of interest by homologous recombination at the target site.
  • nucleic acid encoding the endonuclease according to the invention and the targeting construct can also be located on two separate vectors. Therefore, the invention also pertains to a combination of two vectors, namely:
  • the vectors and combinations described hereabove can for example be used as a medicament.
  • these vectors and combinations can be used in gene therapy.
  • the invention relates to a vector or combination according to the invention for use as a medicament.
  • the transgene encodes a therapeutic polypeptide.
  • diseases that may be treated by gene therapy using the vectors and combinations according to the invention include but are not limited to X-SCID, SCID, epidermolysis bullosa, leber amaurosis, hemophilia, thalassemia, fanconi anemia and muscular dystrophy.
  • the transgene encodes the following therapeutic polypeptides, respectively: IL2RG, GI7A1, Rp 65, Blood factors VIII and IX, haemoglobin A and B, Fanc-A, Fanc-C (or other Fanconi Anemia related genes), Dystrophine.
  • the invention further relates to a pharmaceutical composition
  • a pharmaceutical composition comprising the vectors and combinations according to the invention and a pharmaceutically active carrier.
  • the invention also relates to a method of treating an individual by gene therapy comprising administering an effective amount of a vector or combination according to the invention to an individual in need thereof.
  • an effective amount is meant an amount sufficient to achieve insertion of the transgene into the genome of the individual to be treated. Such concentrations can be routinely determined by those of skilled in the art.
  • subject in need thereof is meant an individual suffering from or susceptible of suffering from a genetic disease that can be treated or prevented by insertion of the transgene.
  • the individuals to be treated in the frame of the invention are preferably human beings.
  • vectors and combinations described hereabove not only find use in gene therapy but also in non pharmaceutical uses such as, e.g., production of animal models and production of recombinant cell lines expressing a protein of interest.
  • the invention relates to:
  • the above use or method aims at inserting a transgene encoding a protein of interest into the genome of a cell order to obtain a recombinant cell line for protein production.
  • Suitable cells for constructing recombinant cell lines for protein production include but are not limited to human (e.g. PER.C6 or HEK), Chinese Ovary hamster (CHO) and mouse (NSE0) cells.
  • the above use aims at making a non-human animal model of a hereditary disorder.
  • the invention is also directed to a non-human transgenic animal comprising a nucleic acid, an expression vector or a combination according to the invention in its genome.
  • FIG. 1 represents target sequences of meganucleases described in Example 1.
  • FIGS. 2 and 3 represent SCOH SH3 meganucleases vs. I-SceI and SCOH-RAG DNA dose response in CHO.
  • FIG. 4 represents target sequences of meganucleases described in Example 2.
  • FIGS. 5 and 6 represent SCOH SH4 meganucleases vs. I-SceI and SCOH-RAG DNA dose response in CHO.
  • FIG. 7 represents a scheme of the mechanism leading to the generation of small deletions and insertions (InDel) during repair of double-strand break by non homologous end-joining (NHEJ).
  • FIG. 8 represents the insertion sites upon cleavage with SH3 or SH4 meganucleases.
  • FIG. 9 represents target sequences of meganucleases described in Example 5.
  • FIG. 10 represents SCOH SH6 meganucleases vs. I-SceI and SCOH-RAG DNA dose response in CHO.
  • FIG. 11 represents a sequence alignment between a I-CreI monomer of SEQ ID NO: 1 and a I-CreI monomer of SEQ ID NO: 42.
  • FIG. 12 represents a sequence alignment between a I-CreI monomer of SEQ ID NO: 1 and two I-CreI monomers comprising 44A 54L 64A 70Q 75N 158R and 162A mutations.
  • the first one (SEQ ID NO: 57) is directly derived from SEQ ID NO: 1 and the second one (SEQ ID NO: 58) is directly derived from SEQ ID NO: 42.
  • FIGS. 13 to 17 illustrate examples 6 to 9.
  • SEQ ID NO: 1 shows the amino acid sequence of a wild-type I-CreI monomer.
  • SEQ ID NO: 2 shows the sequence of a target sequence according to the invention that is located within the SH3 locus.
  • SEQ ID NO: 3 shows the sequence of a target sequence according to the invention that is located within the SH4 locus.
  • SEQ ID NO: 4 shows the sequence of the target sequence of the wild-type I-CreI homodimeric protein.
  • SEQ ID Nos. 5 to 10 represent sequences shown on FIG. 1 .
  • SEQ ID Nos. 11 to 15 represent oligonucleotides, primers and linkers used in Example 1.
  • SEQ ID Nos. 16 to 19 represent sequences shown on FIG. 4 .
  • SEQ ID Nos. 20 to 24 represent oligonucleotides, primers and linkers used in Example 2.
  • SEQ ID Nos. 25 to 32 represent the single-chain meganucleases constructed in Example 1, referred to as SCOH-SH3-b56-A, SCOH-SH3-b56-B, SCOH-SH3-b56-C, SCOH-SH3-b56-D, SCOH-SH3-b1-A, SCOH-SH3-b1-B, SCOH-SH3-b1-C and SCOH-SH3-b1-D respectively.
  • SEQ ID Nos. 33 to 40 represent the single-chain meganucleases constructed in Example 2, referred to as SCOH-SH4-b56-A, SCOH-SH4-b56-B, SCOH-SH4-b56-C, SCOH-SH4-b56-D, SCOH-SH4-b1-A, SCOH-SH4-b1-B, SCOH-SH4-b1-C and SCOH-SH4-b1-D respectively.
  • SEQ ID NO: 41 represents the positive control SCOH-RAG.
  • SEQ ID NO: 42 shows the amino acid sequence of a I-CreI monomer with an additional alanine at position 2, and with three additional residues after the final proline.
  • SEQ ID NO: 43 shows the amino acid sequence of the RM2 linker.
  • SEQ ID Nos. 44 to 49 represent oligonucleotides, primers and linkers used in Example 3.
  • SEQ ID Nos. 50 to 53 represent oligonucleotides, primers and linkers used in Example 4.
  • SEQ ID Nos. 54 to 55 show sequences comprised in the SH3, SH4 and SH6 loci, respectively.
  • SEQ ID NO: 57 shows a monomer derived from a monomer of SEQ ID NO: 1 that comprises 44A 54L 64A 70Q 75N 158R 162A mutations.
  • SEQ ID NO: 58 shows a monomer derived from a monomer of SEQ ID NO: 42 that comprises 44A 54L 64A 70Q 75N 158R 162A mutations.
  • SEQ ID NO: 59 shows the sequence of a target sequence according to the invention that is located within the SH6 locus.
  • SEQ ID Nos. 60 to 64 represent sequences shown on FIG. 9 .
  • SEQ ID Nos. 65 to 75 represent oligonucleotides, primers and linkers used in Example 5.
  • SEQ ID Nos. 76 to 85 represent the single-chain meganucleases constructed in Example 5, referred to as SCOH-SH6-b1-B, SCOH-SH6-b1-C, SCOH-SH6-b1-C, QCSH61-A01, QCSH61-E01, QCSH61-H0, QCSH62-A02, QCSH61-H01b, QCSH61-H01c and QCSH61-H01d respectively.
  • SEQ ID Nos. 86 to 96 represent the single-chain meganucleases capable of cleaving the SH7 locus (SEQ ID Nos. 86 and 87), SH8 locus (SEQ ID NO: 88), the SH12 locus (SEQ ID NO: 89), the SH13 locus (SEQ ID NO: 90), the SH19 locus (SEQ ID NO: 91), the SH20 locus (SEQ ID NO: 92), the SH21 locus (SEQ ID Nos. 93 to 95) and the SH33 locus (SEQ ID NO: 96).
  • SEQ ID Nos. 97 to 104 represent sequences comprised within the SH12, SH13, SH19, SH20, SH21, SH33, SH7 and SH8 loci, respectively.
  • SEQ ID Nos. 105 to 325 represent sequences disclosed in Examples 6 to 9 and/or in any one of Tables A′, A′′, E, G and H.
  • SEQ ID NO: 326 shows the amino acid sequence of the BQY linker.
  • I-CreI variants were constructed by genetic engineering of I-CreI monomers of SEQ ID NO: 42.
  • SH3 is a locus comprising a 24 bp non-palindromic target (SEQ ID NO: 2) that is present on chromosome 6. As shown in Table A, SH3 is located in the vicinity of a RIS disclosed in Deichmann et al. (J. of Clin. Invest. 2007 117:2225). The SH3 sequence is not included in any of the CIS described in Deichmann et al.
  • I-CreI heterodimers capable of cleaving a target sequence of SEQ ID NO: 2 were identified using methods derived from those described in Chames et al. (Nucleic Acids Res., 2005, 33, e178), Arnould et al. (J. Mol. Biol., 2006, 355, 443-458), Smith et al. (Nucleic Acids Res., 2006, 34, e149), Arnould et al. (Arnould et al. J Mol. Biol. 2007 371:49-65). Some of these heterodimers were then cloned into mammalian expression vectors for assessing SH3 cleavage in CHO cells.
  • I-CreI variants potentially cleaving the SH3 target sequence in heterodimeric form were constructed by genetic engineering. Pairs of such variants were then co-expressed in yeast. Upon co-expression, one obtains three molecular species, namely two homodimers and one heterodimer. It was then determined whether the heterodimers were capable of cutting the SH3 target sequence of SEQ ID NO: 2.
  • the SH3 sequence is partially a combination of the 10AAT_P (SEQ ID NO: 5), 5AAG_P (SEQ ID NO: 6), 10AGG_P (SEQ ID NO: 7) and 5TTT_P (SEQ ID NO: 8) target sequences which are shown on FIG. 1 .
  • These sequences are cleaved by mega-nucleases obtained as described in International PCT applications WO 2006/097784 and WO 2006/097853, Arnould et al. (J. Mol. Biol., 2006, 355, 443-458) and Smith et al. (Nucleic Acids Res., 2006).
  • SH3 should be cleaved by combinatorial variants resulting from these previously identified meganucleases.
  • SH3.3 and SH3.4 Two palindromic targets, SH3.3 and SH3.4, were derived from SH3 ( FIG. 1 ). Since SH3.3 and SH3.4 are palindromic, they should be cleaved by homodimeric proteins. Therefore, homodimeric I-CreI variants cleaving either the SH3.3 palindromic target sequence of SEQ ID NO: 9 or the SH3.4 palindromic target sequence of SEQ ID NO: 10 were constructed using methods derived from those described in Chames et al. (Nucleic Acids Res., 2005, 33, e178), Arnould et al. (J. Mol. Biol., 2006, 355, 443-458), Smith et al. (Nucleic Acids Res., 2006, 34, e149) and Arnould et al. (Arnould et al. J Mol Biol. 2007 371:49-65).
  • Yeast reporter vector was transformed into the FYBL2-7B Saccharomyces cerevisiae strain having the following genotype: MAT a, ura3 ⁇ 851, trp1 ⁇ 63, leu2 ⁇ 1, lys2A202.
  • the resulting strain corresponds to a reporter strain (MilleGen).
  • the open reading frames coding for the variants cleaving the SH3.4 or the SH3.3 sequence were cloned in the pCLS542 expression vector and in the pCLS1107 expression vector, respectively.
  • Yeast DNA from these variants was extracted using standard protocols and was used to transform E. coli .
  • the resulting plasmids were then used to co-transform yeast. Transformants were selected on synthetic medium lacking leucine and containing G418.
  • Mating was performed using a colony gridder (QpixII, Genetix). Variants were gridded on nylon filters covering YPD plates, using a low gridding density (4-6 spots/cm 2 ). A second gridding process was performed on the same filters to spot a second layer consisting of different reporter-harboring yeast strains for each target. Membranes were placed on solid agar YPD rich medium, and incubated at 30° C. for one night, to allow mating. Next, filters were transferred to synthetic medium, lacking leucine and tryptophan, adding G418, with galactose (2%) as a carbon source, and incubated for five days at 37° C., to select for diploids carrying the expression and target vectors.
  • I-CreI variants able to efficiently cleave the SH3 target in yeast when forming heterodimers are described hereabove in example 1.1.
  • the efficiency of some of these variants was compared using an extrachromosomal assay in CHO cells.
  • the screen in CHO cells is a single-strand annealing (SSA) based assay where cleavage of the target by the meganucleases induces homologous recombination and expression of a LagoZ reporter gene (a derivative of the bacterial lacZ gene).
  • SSA single-strand annealing
  • oligonucleotide corresponding to the SH3 target sequence flanked by gateway cloning sequences was ordered from PROLIGO (SEQ ID NO: 12; TGGCATACAAGTTTCCAATACAAGGTACAAAGTCCTGACAATCGTCTGTCA).
  • Double-stranded target DNA generated by PCR amplification of the single stranded oligonucleotide, was cloned using the Gateway protocol (INVITROGEN) into the pCLS1058 CHO reporter vector. Cloned target was verified by sequencing (MILLEGEN).
  • CHO K1 cells were transfected with Polyfect® transfection reagent according to the supplier's protocol (QIAGEN). 72 hours after transfection, culture medium was removed and 150 ⁇ l of lysis/revelation buffer for ⁇ -galactosidase liquid assay was added (typically 1 liter of buffer contained 100 ml of lysis buffer (Tris-HCl 10 mM pH7.5, NaCl 150 mM, Triton X100 0.1%, BSA 0.1 mg/ml, protease inhibitors), 10 ml of Mg 100 ⁇ buffer (MgCl 2 100 mM, ⁇ -mercaptoethanol 35%), 110 ml ONPG 8 mg/ml and 780 ml of sodium phosphate 0.1M pH7.5). After incubation at 37° C., OD was measured at 420 nm. The entire process was performed on an automated Velocity11 BioCel platform.
  • I-CreI variants were assayed together as heterodimers against the SH3 target in the CHO extrachromosomal assay.
  • Table II shows the functional combinations obtained for nine heterodimers.
  • SH3.3-MA is a SH3.3 cutter that bears the following mutations in comparison with the I-CreI wild type sequence: 44A 54L 70Q 75Y 92R 158R 162A.
  • SH3.4-M1 is a SH3.4 cutter that bears the following mutations in comparison with the I-CreI wild type sequence: 30G 38R 70D 75N 86D.
  • Single chain constructs were engineered using the linker RM2 of SEQ ID NO: 15 (AAGGSDKYNQALSKYNQALSKYNQALSGGGGS), thus resulting in the production of the single chain molecule: MA-linkerRM2-M1.
  • the G195 mutation was introduced in the C-terminal M1 variant.
  • mutations K7E, K96E were introduced into the MA variant and mutations E8K, E61 R into the M1 variant to create the single chain molecule: MA (K7E K96E)-linkerRM2-M1 (E8K E61R G195) that is further called SCOH-SH3-b1 scaffold.
  • I132V Isoleucine 132 with Valine
  • SCOH-SH3-b56 scaffold based on the best variants cleaving SH3.3 (44A 54L 70Q 75Y 92R 158R 162A) and SH3.4 (30G 38R, 50R 70D 75N 142R) as homodimers, respectively.
  • CHO K1 cells were transfected as described in example 1.2. 72 hours after transfection, culture medium was removed and 150 ⁇ l of lysis/revelation buffer for 1-galactosidase liquid assay was added. After incubation at 37° C., OD was measured at 420 nm. The entire process is performed on an automated Velocity11 BioCel platform. Per assay, 150 ng of target vector was cotransfected with an increasing quantity of variant DNA from 3.12 to 25 ng (25 ng of single chain DNA corresponding to 12.5 ng+12.5 ng of heterodimer DNA). Finally, the transfected DNA variant DNA quantity was 3.12 ng, 6.25 ng, 12.5 ng and 25 ng. The total amount of transfected DNA was completed to 175 ng (target DNA, variant DNA, carrier DNA) using an empty vector (pCLS0002).
  • the activity of the single chain molecules against the SH3 target was monitored using the previously described CHO assay along with our internal control SCOH-RAG and I-Sce I meganucleases. All comparisons were done at 3.12 ng, 6.25 ng, 12.5 ng, and 25 ng transfected variant DNA ( FIGS. 2 and 3 ). All the single molecules displayed SH3 target cleavage activity in CHO assay as listed in Table IV.
  • SCOH-SH3-b1-C has a similar profile, and is even more active than. Its activity reaches the maxima at the lowest DNA quantity transfected from low quantity to high quantity.
  • the molecule SCOH-SH3-b56-A has a maximal activity at higher DNA doses but reaches equivalent level of activity of SCOH-SH3-b1-C and our internal standard.
  • SH4 is a locus that is present on chromosome 7.
  • the SH4 locus comprises a 24 bp non-palindromic sequence of SEQ ID NO: 3.
  • SH4 is located in the vicinity a RIS disclosed in Schwarzwaelder et al. (J. Clin. Invest. 2007 117:2241).
  • the SH4 sequence is not included in any of the CIS described in Deichman et al.
  • Example 2 Experiments similar to those described hereabove in Example 1 were carried out to identify I-CreI heterodimers and single-chain meganucleases capable of cleaving a target sequence of SEQ ID NO: 3.
  • I-CreI variants potentially cleaving the SH4 target sequence in heterodimeric form were constructed by genetic engineering. Pairs of such variants were then co-expressed in yeast. Upon co-expression, one obtains three molecular species, namely two homodimers and one heterodimer. It was then determined whether the heterodimers were capable of cutting the SH4 target sequence of SEQ ID NO: 3.
  • the SH4 sequence is partially a combination of the 10AAA_P (SEQ ID NO: 4), 5ACT_P (SEQ ID NO: 16), 10AAA_P (SEQ ID NO: 4), 5GGT_P (SEQ ID NO: 17) targets shown on FIG. 4 .
  • These sequences are cleaved by previously identified mega-nucleases, obtained as described in International PCT Applications WO 2006/097784 and WO 2006/097853; Arnould et al., J. Mol. Biol., 2006, 355, 443-458; Smith et al., Nucleic Acids Res., 2006.
  • SH4 should be cleaved by combinatorial variants resulting from these previously identified meganucleases.
  • the screening procedure was performed using methods derived from those described in Chames et al. (Nucleic Acids Res., 2005, 33, e178), Arnould et al. (J. Mol. Biol., 2006, 355, 443-458), Smith et al. (Nucleic Acids Res., 2006, 34, e149) and Arnould et al. (Arnould et al. J Mol Biol. 2007 371:49-65) on the two following palindromic sequences: the SH4.3 sequence of SEQ ID NO: 18 and the SH4.4 sequence of SEQ ID NO: 19.
  • Example 1.1 The experimental procedure is as described in Example 1.1, with the exception that an oligonucleotide corresponding to the SH4 target sequence of SEQ ID NO: 20 (5′-TGGCATACAAGTTTTTAAAACACTGTACACCATTTTGACAATCGTCTGTCA-3′) was used.
  • Yeast DNA from variants cleaving the SH4.3 and SH4.4 target in the pCLS542 and pCLS1107 expression vectors was extracted using standard protocols and was used to transform E. coli .
  • the resulting plasmid DNA was then used to co-transform yeast strain. Transformants were selected on synthetic medium lacking leucine and containing G418.
  • Mating was performed using a colony gridder (QpixII, Genetix). Variants were gridded on nylon filters covering YPD plates, using a low gridding density (4-6 spots/cm 2 ). A second gridding process was performed on the same filters to spot a second layer consisting of different reporter-harboring yeast strains for each target. Membranes were placed on solid agar YPD rich medium, and incubated at 30° C. for one night, to allow mating. Next, filters were transferred to synthetic medium, lacking leucine and tryptophan, adding G418, with galactose (2%) as a carbon source, and incubated for five days at 37° C., to select for diploids carrying the expression and target vectors.
  • the screen in CHO cells is a single-strand annealing (SSA) based assay where cleavage of the target by the meganucleases induces homologous recombination and expression of a LagoZ reporter gene (a derivative of the bacterial lacZ gene).
  • SSA single-strand annealing
  • the target was cloned as follows.
  • Double-stranded target DNA generated by PCR amplification of the single stranded oligonucleotide, was cloned using the Gateway protocol (INVITROGEN) into CHO reporter vector (pCLS1058). The cloned fragment was verified by sequencing (MILLEGEN).
  • ORFs of I-CreI variants cleaving the SH4.5 and SH4.6 targets obtained hereabove were sub-cloned in pCLS2437.
  • ORFs were amplified by PCR on yeast DNA using primers of SEQ ID NO: 22 and 23 (5′-AAAAAGCAGGCTGGCGCGCCTACACAGCGGCCTTGCCACCATG-3′ and 5′-AGAAAGCTGGGTGCTAGCGCTCGAGTTATCAGTCGG-3′) primers.
  • PCR products were cloned in the CHO expression vector pCLS2437 using the AscI and NheI restrictions sites for internal fragment replacement. Selected clones resulting from ligation and E. coli transformation steps were verified by sequencing (MILLEGEN).
  • CHO K1 cells were transfected with Polyfect® transfection reagent according to the supplier's protocol (QIAGEN). 72 hours after transfection, culture medium was removed and 150 ⁇ l of lysis/revelation buffer for ⁇ -galactosidase liquid assay was added (typically 1 liter of buffer contained: 100 ml of lysis buffer (Tris-HCl 10 mM pH7.5, NaCl 150 mM, Triton X100 0.1%, BSA 0.1 mg/ml, protease inhibitors), 10 ml of Mg 100 ⁇ buffer (MgCl 2 100 mM, ⁇ -mercaptoethanol 35%), 110 ml ONPG 8 mg/ml and 780 ml of sodium phosphate 0.1M pH7.5).
  • lysis buffer Tris-HCl 10 mM pH7.5, NaCl 150 mM, Triton X100 0.1%, BSA 0.1 mg/ml, protease inhibitors
  • Mg 100 ⁇ buffer MgCl 2
  • Example 2.1 The four variants shown in Table VI and described herebaove in Example 2.1, were selected for further analysis.
  • I-CreI variants were cloned in pCLS2437. Then, I-CreI variants cleaving the SH4.3 or SH4.4 targets were assayed together as heterodimers against the SH4 target in the CHO extrachromosomal assay. Analysis of the efficiencies of cleavage and recombination of the SH4 sequence demonstrates that all tested combinations of I-CreI variants were able to transpose their cleavage activity from yeast to CHO cells without additional mutation (Table VII).
  • SH4.3-MA is a SH4.3 cutter that bears the following mutations in comparison with the I-CreI wild type sequence: 24V 44R 68Y 70S 75Y 77N.
  • SH4.4-M2 is a SH4.4 cutter that bears the following mutations in comparison with the I-CreI wild type sequence: 24V 44Y 70S 77V.
  • the Isoleucine 132 to Valine (I132V) mutation was introduced into the coding sequence of either, one, none or both N-terminal and C-terminal protein fragment.
  • the design of the derived single chain constructs is shown in Table VIII.
  • the single chain constructs were tested in CHO for their ability to induce cleavage of the SH4 target.
  • CHO K1 cells were transfected as described hereabove. 72 hours after transfection, culture medium was removed and 150 ⁇ l of lysis/revelation buffer for ⁇ -galactosidase liquid assay was added. After incubation at 37° C., OD was measured at 420 nm. The entire process is performed on an automated Velocity11 BioCel platform. Per assay, 150 ng of target vector was cotransfected with an increasing quantity of variant DNA from 3.12 to 25 ng (25 ng of single chain DNA corresponding to 12.5 ng+12.5 ng of heterodimer DNA). Finally, the transfected DNA variant DNA quantity was 3.12 ng, 6.25 ng, 12.5 ng and 25 ng. The total amount of transfected DNA was completed to 175 ng (target DNA, variant DNA, carrier DNA) using an empty vector (pCLS0002).
  • the single chain molecules described in Table VIII were monitored for their activity against the SH4 target using the previously described CHO assay by comparison to our internal control SCOH-RAG and I-Sce I meganucleases. All activity evaluation was done upon DNA transfected dose of 3.12 ng, 6.25 ng, 12.5 ng, and 25 ng. All single chain molecules were displaying activity on SH4 target as reported in Table VIII.
  • SCOH-SH4-b1C shows an activity level within the same range as the internal standard SCOH-RAG (: its activity increases from low quantity to high quantity. At the assayed DNA trasfected doses, its activity is superior to that of SCOH-SH4-B56A.
  • I-CreI variants able to efficiently cleave the SH3 and SH4 targets in yeast and in mammalian cells have been identified in Examples 1 and 2.
  • the efficiency of the SH3 and SH4 meganucleases to cleave their endogenous DNA target sequences was next tested. This example will demonstrate that meganucleases engineered to cleave the SH3 and SH4 target sequences cleave their cognate endogenous sites in human cells.
  • NHEJ non homologous end-joining
  • Single Chain I-CreI variants for SH3 and SH4 cloned in the pCLS1853 plasmid were used for this experiment.
  • the day previous experiment cells from the human embryonic kidney cell line, 293-H (Invitrogen) were seeded in a 10 cm dish at density of 1.2 10 6 cells/dish. The following day, cells were transfected with 3 ⁇ g of an empty plasmid or a meganuclease-expressing plasmid using lipofectamine (Invitrogen). 72 hours after transfection, cells were collected and diluted (dilution 1/20) in fresh culture medium. After 7 days of culture, cells were collected and genomic DNA extracted.
  • genomic DNA 200 ng of genomic DNA were used to amplify the endogenous locus surrounding the meganuclease cleavage site by PCR amplification.
  • a 377 bp fragment corresponding to the SH3 locus was amplified using specific PCR primers A (SEQ ID NO 44; 5′-tgggggtcttactctgtttccc-3′) and B (SEQ ID NO 45; 5′-aggagagtccttctttggcc-3′).
  • a 396 bp fragment corresponding to the SH4 locus was amplified using PCR primers C (SEQ ID NO 46; 5′-gagtgatagcataatgaaaacc-3′) and D (SEQ ID NO 47; 5′-ctcaccataagtcaactgtctc-3′).
  • PCR amplification was performed to obtain a fragment flanked by specific adaptator sequences (SEQ ID NO 48; 5′-CCATCTCATCCCTGCGTGTCTCCGACTCAG-3′ and SEQ ID NO: 49 5′-CCTATCCCCTGTGTGCCTTGGCAGTCTCAG-3′) provided by the company offering sequencing service (GATC Biotech AG, Germany) on the 454 sequencing system (454 Life Sciences).
  • FIG. 8 the analysis of the size of the DNA insertion or deletion sequences revealed a similar type of events with a predominance of small insertions ( ⁇ 5 bp) and of small deletions ( ⁇ 10 bp).
  • the vector comprising a targeting construct was the pCLS3777 or pCLS3778 plasmid containing a 2.8 kb sequence consisting of an exogenous DNA sequence, flanked by two sequences homologous to the human SH3 or SH4 loci.
  • the sequences homologous to the human SH3 or SH4 loci had a length of 1.5 kb.
  • Cleavage of the native SH3 or SH4 loci by the meganuclease yields a substrate for homologous recombination, which may use the donor repair plasmid as a repair matrix.
  • the frequency with which targeted integration occurs at the SH3 or SH4 loci is indicative of the cleavage efficiency of the genomic SH3 or SH4 target site.
  • Example 4.1 Material and Methods
  • the meganucleases used in this example are SCOH-SH3-b1-C and SCOH-SH4-b1-C cloned in a mammalian expression vector, resulting in plasmid pCLS2697 and pCLS2705, respectively.
  • the donor plasmid contained:
  • the left and right homology arms were inserted upstream (using an AscI site) and downstream (using a SbfI site), respectively, of an exogenous 2.8 kb DNA fragment containing two CMV promoters and a neomycin resistance gene.
  • the resulting plasmids are referred to as pCLS3777 (for SH3) and pCLS3778 (for SH4).
  • Human embryonic kidney 293H cells (Invitrogen) were plated at a density of 1 ⁇ 10 6 cells per 10 cm dish in complete medium (DMEM supplemented with 2 mM L-glutamine, penicillin (100 UI/ml), streptomycin (100 ⁇ g/ml), amphotericin B (Fongizone) (0.25 ⁇ g/ml) (Invitrogen-Life Science) and 10% FBS).
  • complete medium DMEM supplemented with 2 mM L-glutamine, penicillin (100 UI/ml), streptomycin (100 ⁇ g/ml), amphotericin B (Fongizone) (0.25 ⁇ g/ml) (Invitrogen-Life Science) and 10% FBS.
  • Lipofectamine 2000 transfection reagent (Invitrogen) according to the supplier's protocol. Briefly, 2 ⁇ g of the donor plasmid was co-transfected with 3 ⁇ g of single-chain meganuclease expression vector
  • genomic DNA extraction was performed with the ZR-96 genomic DNA kit (Zymo research) according to the supplier's protocol.
  • the gene targeting frequency was determined by PCR on genomic DNA using the following primers: 5′-CTGTGTGCTATGATCTTGCC-3′ (SH3 GHGF4; SEQ ID NO: 50) and 5′-CCTGTCTCTTGATCAGATCC-3′ (NeoR2; SEQ ID NO: 51) for SH3, and 5′-GTGGCCTCTCAGTCTGTTTA-3′ (SH4 GHGF2; SEQ ID NO: 52) and 5′-AGTCATAGCCGAATAGCCTC-3′ (NeoR5; SEQ ID NO: 53) for SH4.
  • the PCRs result in a 2500 bp (SH3) or a 2268 bp (SH4) gene targeting specific PCR product.
  • the SH3 GHGF4 and SH4 GHGF2 primers are forward primers located upstream of the left homology arms of the donor repair plasmids.
  • the NeoR primers are reverse primers located in the exogenous DNA inserted between the two homology arms of the donor repair plasmid.
  • Human embryonic kidney 293H cells were co-transfected with a plasmid expressing one of the two single-chain SH3 or SH4 meganucleases and the donor repair plasmid pCLS3777 or pCLS3778. As a control for spontaneous recombination, 293H cells were also transfected with the donor repair plasmid alone. The cells were then plated at 10 or 100 cells per well in 96-well microplates. Genomic DNA derived from these cells was analyzed for gene targeting by PCR as described in Material and Methods.
  • SH6 is a locus comprising a 24 bp non-palindromic target (TTAATACCCCGTACCTAATATTGC, SEQ ID NO: 59) that is present on chromosome 21.
  • SH6 is located in the vicinity of a RIS disclosed in Schwarzwaelder et al. (J Clin Invest 2007:2241-9). The SH6 sequence is not included in any of the CIS described in Deichman et al.
  • I-CreI variants potentially cleaving the SH6 target sequence in heterodimeric form were constructed by genetic engineering. Pairs of such variants were then co-expressed in yeast. Upon co-expression, one obtains three molecular species, namely two homodimers and one heterodimer. It was then determined whether the heterodimers were capable of cutting the SH6 target sequence of SEQ ID NO: 59.
  • the SH6 sequence is partially a combination of the 10AAT_P (SEQ ID NO: 60), 5CCC_P (SEQ ID NO: 61), 10AAT_P (SEQ ID NO: 60), 5TAG_P (SEQ ID NO: 62) target sequences which are shown on FIG. 9 .
  • These sequences are cleaved by mega-nucleases obtained as described in International PCT applications WO 2006/097784 and WO 2006/097853, Arnould et al. (J. Mol. Biol., 2006, 355, 443-458) and Smith et al. (Nucleic Acids Res., 2006).
  • SH6 should be cleaved by combinatorial variants resulting from these previously identified meganucleases.
  • SH6.3 and SH6.4 Two palindromic targets, SH6.3 and SH6.4, were derived from SH6 ( FIG. 9 ). Since SH6.3 and SH6.4 are palindromic, they should be cleaved by homodimeric proteins. Therefore, homodimeric I-CreI variants cleaving either the SH6.3 palindromic target sequence of SEQ ID NO: 63 or the SH6.4 palindromic target sequence of SEQ ID NO: 64 were constructed using methods derived from those described in Chames et al. (Nucleic Acids Res., 2005, 33, e178), Arnould et al. (J. Mol. Biol., 2006, 355, 443-458), Smith et al. (Nucleic Acids Res., 2006, 34, e149) and Arnould et al. (Arnould et al. J Mol Biol. 2007 371:49-65).
  • Example 1.1 The experimental procedure is as described in Example 1.1., with the exception that an oligonucleotide corresponding to the SH6 target sequence (5′-TGGCATACAAGTTTTTAATACCCCGTACCTAATATTGCCAATCGTCTGTCA-3′ (SEQ ID NO: 65) was used.
  • Yeast DNA was extracted from variants cleaving the SH6.3 and SH6.4 targets in the pCLS542 and pCLS1107 expression vectors using standard protocols and was used to transform E. coli . Transformants were selected on synthetic medium lacking leucine and containing G418.
  • Mating was performed using a colony gridder (QpixII, Genetix). Variants were gridded on nylon filters covering YPD plates, using a low gridding density (4-6 spots/cm 2 ). A second gridding process was performed on the same filters to spot a second layer consisting of different reporter-harboring yeast strains for each target. Membranes were placed on solid agar YPD rich medium, and incubated at 30° C. for one night, to allow mating. Next, filters were transferred to synthetic medium, lacking leucine and tryptophan, adding G418, with galactose (2%) as a carbon source, and incubated for five days at 37° C., to select for diploids carrying the expression and target vectors.
  • I-CreI variants able to efficiently cleave the SH6 target in yeast when forming heterodimers are described hereabove in example 5.1.
  • the efficiency of some of these variants was compared using an extrachromosomal assay in CHO cells.
  • the screen in CHO cells is a single-strand annealing (SSA) based assay where cleavage of the target by the meganucleases induces homologous recombination and expression of a LagoZ reporter gene (a derivative of the bacterial lacZ gene).
  • SSA single-strand annealing
  • oligonucleotide corresponding to the SH6 target sequence flanked by gateway cloning sequence was ordered from PROLIGO 5′-TGGCATACAAGTTTTTAATACCCCGTACCTAATATTGCCAATCGTCTGTCA-3′ (SEQ ID NO: 65).
  • Double-stranded target DNA, generated by PCR amplification of the single stranded oligonucleotide was cloned using the Gateway protocol (INVITROGEN) into CHO reporter vector (pCLS1058). Cloned target was verified by sequencing (MILLEGEN).
  • ORF of I-CreI variants cleaving the SH6.3 and SH6.4 targets identified in example 5.1 were sub-cloned in pCLS2437.
  • ORFs were amplified by PCR on yeast DNA using the following primers: 5′-AAAAAGCAGGCTGGCGCGCCTACACAGCGGCCTTGCCACCATG-3′ (SEQ ID NO: 66) and 5′-AGAAAGCTGGGTGCTAGCGCTCGAGTTATCAGTCGG-3′ (SEQ ID NO: 67) primers.
  • PCR products were cloned in the CHO expression vector pCLS2437 using the AscI and XhoI for internal fragment replacement. Selected clones resulting from ligation and E. coli transformation steps were verified by sequencing (MILLEGEN).
  • CHO K1 cells were transfected with Polyfect® transfection reagent according to the supplier's protocol (QIAGEN). 72 hours after transfection, culture medium was removed and 150 ⁇ l of lysis/revelation buffer for ⁇ -galactosidase liquid assay was added (typically 1 liter of buffer contained: 100 ml of lysis buffer (Tris-HCl 10 mM pH7.5, NaCl 150 mM, Triton X100 0.1%, BSA 0.1 mg/ml, protease inhibitors), 10 ml of Mg 100 ⁇ buffer (MgCl 2 100 mM, ⁇ -mercaptoethanol 35%), 110 ml ONPG 8 mg/ml and 780 ml of sodium phosphate 0.1M pH7.5).
  • lysis buffer Tris-HCl 10 mM pH7.5, NaCl 150 mM, Triton X100 0.1%, BSA 0.1 mg/ml, protease inhibitors
  • Mg 100 ⁇ buffer MgCl 2
  • the monomer capable of cleaving SH6.3 comprised the following mutations: 44K 70S 75N (referred to as SH6-3-M1-44K 70S 75N) and the monomer capable of cleaving SH6.4 comprised the following mutations: 28Q 40R 44A 70L 75N 96R 111H 144S (referred to as SH6-4-MB-28Q 40R 44A 70L 75N 96R 111 H 144S).
  • M1 ⁇ MA SH6 heterodimer gives high cleavage activity in yeast.
  • M1 is a SH6.3 cutter that bears the following mutations in comparison with the I-CreI wild type sequence: 44K 70S 75N.
  • MA is a SH6.4 cutter that bears the following mutations in comparison with the I-CreI wild type sequence: 7R 28Q 40R 44A 70L 75N 103T 121E 132V 160R.
  • Single chain constructs were engineered using the linker RM2 (AAGGSDKYNQALSKYNQALSKYNQALSGGGGS; SEQ ID NO: 15) resulting in the production of the single chain molecule: MA-RM2-M1.
  • the G19S mutation was introduced in the C-terminal M1 mutant.
  • mutations K96E was introduced into the MA mutant and mutations E8K, E61 R into the M1 mutant to create the single chain molecule: MA(K96E)-RM2-MA(E8K E61R) that is called further SCOH-SH6 b1 scaffold.
  • Additional mutations were introduced by use of the QuikChange Multi Site-Directed Mutagenesis Kit from Stratagene/Agilent technologies Inc according to the manufacturer's instructions.
  • a first set of oligonucleotides was used to introduce the mutations in the part of the single chain molecule corresponding to the first monomer.
  • a second set of oligonucleotides was designed to introduce the same mutations specifically in the second part of the single chain molecule corresponding to the second monomer as shown in (see Table XII).
  • Isolated clones obtained at the term of this process were sequenced to confirm the specific mutation profiles obtained. Profiles of interest were then tested in CHO SSA assay in comparison with the initial construct as described.
  • CHO K1 cells were transfected as described above. 72 hours after transfection, culture medium was removed and 150 ⁇ l of lysis/revelation buffer for ⁇ -galactosidase liquid assay was added. After incubation at 37° C., OD was measured at 420 nm. The entire process is performed on an automated Velocity11 BioCel platform.
  • target vector 150 ng was cotransfected with an increasing quantity of variant DNA from 3.12 ng to 25 ng (25 ng of single chain DNA corresponding to 12.5 ng+12.5 ng of heterodimer DNA). Finally, the transfected DNA variant DNA quantity was 3.12 ng, 6.25 ng, 12.5 ng and 25 ng. The total amount of transfected DNA was completed to 175 ng (target DNA, variant DNA, carrier DNA) using empty vector (pCLS0001).
  • the activity of the SCOH-SH6-b1-C (pCLS2796) and SCOH-SH6-b1-B-(pCLS2928) single chain molecules (see Table XIII) against the SH6 target was monitored using the previously described CHO assay by comparison to the SH6.3-M1 ⁇ SH6.4-MB forming heterodimer and our internal control SCOH-RAG and I-Sce I meganucleases. All comparisons were done at 3.12 ng, 6.25 ng, 12.5 ng, and 25 ng transfected variant DNA ( FIG. 10 ).
  • the two single chain meganucleases were able to cleave more efficiently the SH6 target than the starting heterodimer.
  • the activity of the best molecule, SCOH-SH6-b1-C was further improved by introduction additional mutations among those described above in a new bath of meganucleases.
  • the meganucleases used in this example are SCOH-QCSH6-H01 (SEQ ID NO: 81) or SCOH-QC-SH6-H01-V2-7E-70R75D (SEQ ID NO: 85) cloned in a mammalian expression vector, resulting in plasmid pCLS3690 ( FIG. 13 ) and pCLS4373 respectively.
  • the donor plasmid contains a PCR generated 1517 bp fragment of the SH6 locus (position 18437771 to 18439287 on chromosome 21, NC — 000021.8) as the left homology arm and a 1571 bp fragment of the SH6 locus (position 18439343 to 18440846 on chromosome 21, NC — 000021.8) as the right homology arm.
  • the left and right homology arms were inserted upstream (using an AscI site) and downstream (using a SbfI site), respectively, of an exogenous 2.8 kb DNA fragment containing two CMV promoters and a neomycin resistance gene.
  • the resulting plasmid is pCLS3779 ( FIG. 13 ; SEQ ID NO: 279).
  • Human embryonic kidney 293H cells (Invitrogen) were plated at a density of 1 ⁇ 10 6 cells per 10 cm dish in complete medium (DMEM supplemented with 2 mM L-glutamine, penicillin (100 UI/ml), streptomycin (100 ⁇ g/ml), amphotericin B (Fongizone) (0.25 ⁇ g/ml) (Invitrogen-Life Science) and 10% FBS).
  • complete medium DMEM supplemented with 2 mM L-glutamine, penicillin (100 UI/ml), streptomycin (100 ⁇ g/ml), amphotericin B (Fongizone) (0.25 ⁇ g/ml) (Invitrogen-Life Science) and 10% FBS.
  • Lipofectamine 2000 transfection reagent (Invitrogen) according to the supplier's protocol. Briefly, 2 ⁇ g of the donor plasmid was co-transfected with 3 ⁇ g of single-chain meganuclease expression vector
  • the frequency of gene targeting was determined by PCR on genomic DNA using the primers SH6 GHGF3: 5′-CAATGGAGTTTTGGAGCCAC-3′ (SEQ ID NO: 280) and NeoR9: 5′-ATCAGAGCAGCCGATTGTCT-3′ (SEQ ID NO: 281).
  • the PCRs result in a 2300 bp gene targeting specific PCR product ( FIG. 14 ).
  • the SH6 GHGF3 primer is a forward primer located upstream of the left homology arms of the donor repair plasmids.
  • the NeoR9 primer is a reverse primer located in the exogenous DNA inserted between the two homology arms of the donor repair plasmid.
  • Human embryonic kidney 293H cells were co-transfected with 2 vectors: a plasmid expressing one of the two single-chain SH6 meganucleases and the donor repair plasmid pCLS3779 ( FIG. 13 ; SEQ ID NO: 279).
  • 293H cells were also transfected with the donor repair plasmid alone. The cells were then plated at 10 or 100 cells per well in 96-well microplates or at 300 cells per 10 cm-dishes and 2 weeks later clonal colonies were isolated and plated in 96-well microplates. Genomic DNA derived from these cells was analyzed for gene targeting by PCR as described in Material and Methods.
  • the meganuclease used in this example is SCOH-QCSH6-H01 (SEQ ID NO: 81) cloned in a mammalian expression vector, resulting in plasmid pCLS3690.
  • the donor plasmid contains a PCR generated 1517 bp fragment of the SH6 locus (position 18437771 to 18439287 on chromosome 21, NC — 000021.8) as the left homology arm and a 1571 bp fragment of the SH6 locus (position 18439343 to 18440846 on chromosome 21, NC — 000021.8) as the right homology arm.
  • the left and right homology arms were inserted upstream (using an AscI site) and downstream (using a SbfI site), respectively, of an exogenous 2.8 kb DNA fragment containing two CMV promoters and a neomycin resistance gene.
  • the resulting plasmid is pCLS3779 ( FIG. 13 ; SEQ ID NO: 279).
  • Human embryonic kidney 293H cells (Invitrogen) were plated at a density of 1 ⁇ 10 6 cells per 10 cm dish in complete medium (DMEM supplemented with 2 mM L-glutamine, penicillin (100 UI/ml), streptomycin (100 ⁇ g/ml), amphotericin B (Fongizone) (0.25 ⁇ g/ml) (Invitrogen-Life Science) and 10% FBS).
  • complete medium DMEM supplemented with 2 mM L-glutamine, penicillin (100 UI/ml), streptomycin (100 ⁇ g/ml), amphotericin B (Fongizone) (0.25 ⁇ g/ml) (Invitrogen-Life Science) and 10% FBS.
  • Lipofectamine 2000 transfection reagent (Invitrogen) according to the supplier's protocol. Briefly, 2 ⁇ g of the donor plasmid was co-transfected with 3 ⁇ g of single-chain meganuclease expression vector
  • Gene targeting was determined by PCR on genomic DNA using the primers SH6 GHGF3: 5′-CAATGGAGTTTTGGAGCCAC-3′ (SEQ ID NO: 280) and NeoR9: 5′-ATCAGAGCAGCCGATTGTCT-3′ (SEQ ID NO: 281).
  • the PCRs result in a 2300 bp gene targeting specific PCR product ( FIG. 14 ).
  • the SH6 GHGF3 primer is a forward primer located upstream of the left homology arms of the donor repair plasmids.
  • the NeoR9 primer is a reverse primer located in the exogenous DNA inserted between the two homology arms of the donor repair plasmid.
  • Genomic DNA from cellular clones was digested with StuI or HindIII restriction enzymes (New England Biolabs), separated by electrophoresis on a 0.8% agarose gela and transferred onto a nitrocellulose membrane.
  • a DNA probe was prepared from 25 ng of a DNA fragment homologous to the Neomycin resistance gene with 32 P-radiolabeled dCTP and Rediprime II random prime labelling system (GE Healthcare) according to supplier's protocol and added to the nitrocellulose membrane tha had preincubated in hybridization buffer (NaPi 20 mM, 7% SDS, 1 mM EDTA). After overnight incubation at 65° C., the membrane was washed and exposed to a radiography film. The size of expected bands on the radiograph are 5.3 kb for StuI digestion and 6.8 kb for HindIII digestion ( FIG. 15 ).
  • Human embryonic kidney 293H cells were co-transfected with 2 vectors: a plasmid expressing one of the two single-chain SH6 meganucleases and the donor repair plasmid pCLS3779. The cells were then plated at 300 cells per 10-cm dish and 2 weeks later clonal colonies were isolated and plated in 96-well microplates. Genomic DNA derived from these cells was analyzed for gene targeting by PCR as described in Material and Methods. Genomic DNA was then used to validate targeted integration by southern blot analysis. The clones number 7 and 8 showed bands of the expected size whereas negative control clones number 5 and 6 did not ( FIG. 16 ).
  • sh6 locus To validate the capacity of sh6 locus to support transgene integration without disturbing the expression of neighboring genes, gene targeting experiments were conducted with a repair plasmid containing a 2.8 kb exogenous DNA fragment and cellular clones were identified that contained the targeted integration. The expression of genes upstream and downstream of the sh6 integration site was measured and compared to that of cellular clones that had not undergone targeted integration.
  • the meganucleases used in this example is SCOH-QCSH6-H01 (SEQ ID NO:81) cloned in a mammalian expression vector, resulting in plasmid pCLS3690.
  • the donor plasmid contains a PCR generated 1517 bp fragment of the SH6 locus (position 18437771 to 18439287 on chromosome 21, NC — 000021.8) as the left homology arm and a 1571 bp fragment of the SH6 locus (position 18439343 to 18440846 on chromosome 21, NC — 000021.8) as the right homology arm.
  • the left and right homology arms were inserted upstream (using an AscI site) and downstream (using a SbfI site), respectively, of an exogenous 2.8 kb DNA fragment containing two CMV promoters and a neomycin resistance gene.
  • the resulting plasmid is pCLS3779 ( FIG. 13 ; SEQ ID NO: 279).
  • Human embryonic kidney 293H cells (Invitrogen) were plated at a density of 1 ⁇ 10 6 cells per 10 cm dish in complete medium (DMEM supplemented with 2 mM L-glutamine, penicillin (100 UI/ml), streptomycin (100 ⁇ g/ml), amphotericin B (Fongizone) (0.25 ⁇ g/ml) (Invitrogen-Life Science) and 10% FBS).
  • complete medium DMEM supplemented with 2 mM L-glutamine, penicillin (100 UI/ml), streptomycin (100 ⁇ g/ml), amphotericin B (Fongizone) (0.25 ⁇ g/ml) (Invitrogen-Life Science) and 10% FBS.
  • Lipofectamine 2000 transfection reagent (Invitrogen) according to the supplier's protocol. Briefly, 2 ⁇ g of the donor plasmid was co-transfected with 3 ⁇ g of single-chain meganuclease expression vector
  • Gene targeting was determined by PCR on genomic DNA using the primers SH6 GHGF3: 5′-CAATGGAGTTTTGGAGCCAC-3′ (SEQ ID NO: 280) and NeoR9: 5′-ATCAGAGCAGCCGATTGTCT-3′ (SEQ ID NO: 281).
  • the PCRs result in a 2300 bp gene targeting specific PCR product ( Figure XX).
  • the SH6 GHGF3 primer (SEQ ID NO: 280) is a forward primer located upstream of the left homology arms of the donor repair plasmids.
  • the NeoR9 primer (SEQ ID NO: 281) is a reverse primer located in the exogenous DNA inserted between the two homology arms of the donor repair plasmid.
  • the threshold cycles (Ct) were determined with Stratagene software on fluorescence (dRn) after normalization by the ROX reference dye.
  • the intensity of gene expression was calculated using the formula 2 Ct(HPRT)-Ct(Gene) , the expression of the housekeeping gene HPRT being used as an internal normalizing factor.
  • Human embryonic kidney 293H cells were co-transfected with 2 vectors: a plasmid expressing one of the three single-chain SH6 meganucleases and the donor repair plasmid pCLS3779. The cells were then plated at 300 cells per 10-cm dish and 2 weeks later clonal colonies were isolated and plated in 96-well microplates. Genomic DNA derived from these cells was analyzed for gene targeting by PCR as described in Material and Methods. RNA was isolated from clones showing targeted integration and negative controls. Quantitative RT-PCR was performed to measure expression of genes surrounding the locus of targeted integration. The data are presented in FIG.
  • the coding sequences for the meganucleases used in this example were cloned in a mammalian expression vector, resulting in the plasmids listed in table XVI.
  • Human embryonic kidney 293H cells (Invitrogen) were plated at a density of 1 ⁇ 10 6 cells per 10 cm dish in complete medium (DMEM supplemented with 2 mM L-glutamine, penicillin (100 UI/ml), streptomycin (100 ⁇ g/ml), amphotericin B (Fongizone) (0.25 ⁇ g/ml) (Invitrogen-Life Science) and 10% FBS).
  • complete medium DMEM supplemented with 2 mM L-glutamine, penicillin (100 UI/ml), streptomycin (100 ⁇ g/ml), amphotericin B (Fongizone) (0.25 ⁇ g/ml) (Invitrogen-Life Science) and 10% FBS.
  • DMEM complete medium
  • penicillin 100 UI/ml
  • streptomycin 100 ⁇ g/ml
  • Amphotericin B Feongizone
  • FBS FBS
  • cells were transfected with 3 ⁇ g of single-chain
  • the frequency of mutagenesis was determined by deep sequencing analysis. Oligonucleotides were designed for PCR amplification of a DNA fragment surrounding each safe harbour target and are listed in table XVII.
  • Nucleotides were added to obtain a fragment flanked by specific adaptator sequences (5′-CCATCTCATCCCTGCGTGTCTCCGACTCAG-3′; SEQ ID NO 324) and (5′-CCTATCCCCTGTGTGCCTTGGCAGTCTCAG-3′; SEQ ID NO 325) provided by the company offering sequencing service (GATC Biotech AG, Germany) on the 454 sequencing system (454 Life Sciences). An average of 18,000 sequences was obtained from pools of 2 to 3 amplicons (500 ng each). After sequencing, different samples were identified based on barcode sequences introduced in the first of the above adaptators.
  • Human embryonic kidney 293H cells were transfected with a plasmid expressing a single-chain safe harbor meganuclease. After 2 to 6 days of incubation at 37° C., genomic DNA was isolated and PCR was used to amplify the genomic sequence surrounding the meganuclease target site. Sequences were then analyzed for the presence of insertions or deletions events (InDel) in the cleavage site of each safe harbor target. Results are summarized in table XVIII.
  • Examples 1, 2, 3 and 5 demonstrate that both I-CreI heterodimeric proteins and single-chain meganucleases capable of cleaving the SH3, the SH4 and the SH6 loci can be obtained. Moreover, these endonucleases are capable of cleaving these loci with a strong cleavage activity.
  • Example 4 demonstrates that single-chain meganucleases capable of cleaving the SH3 and the SH4 loci allow efficiently inserting a transgene into a target site of a human cell.
  • endonucleases can thus advantageously be used to insert a transgene into the SH3, the SH4 loci or the SH6 loci of an individual.
  • Example 6 demonstrates that at least two single chain molecules according to the invention are capable of inducing high levels of gene targeting at an endogenous sh6 locus.
  • Example 7 demonstrates that targeted integration a locus can support functional transgene expression.
  • Example 8 demonstrates that a targeted integration at a locus does not substantially modify expression of five genes located in the vicinity of the target sequence.
  • Example 9 demonstrates mutagenesis frequencies for different meganucleases targeting safe harbor sequences, which are indicative of the cleavage efficiency of the genomic target site by said meganucleases.

Abstract

The present invention concerns the endonucleases capable of cleaving a target sequence located in a “safe harbor loci”, i.e. a loci allowing safe expression of a transgene. The present invention further concerns the use of such endonucleases for inserting transgenes into a cell, tissue or individual.

Description

  • The present invention concerns the endonucleases capable of cleaving a target sequence located in a “safe harbor loci”, i.e. a loci allowing safe expression of a transgene. The present invention further concerns the use of such endonucleases for inserting transgenes into a cell, tissue or organism.
  • Meganucleases
  • Meganucleases, also referred to as homing endonucleases, were the first endonucleases used to induce double-strand breaks and recombination in living cells (Rouet et al. PNAS 1994 91:6064-6068; Rouet et al. Mol Cell Biol. 1994 14:8096-8106; Choulika et al. Mol Cell Biol. 1995 15:1968-1973; Puchta et al. PNAS 1996 93:5055-5060). However, their use has long been limited by their narrow specificity. Although several hundred natural meganucleases had been identified over the past years, this diversity was still largely insufficient to address genome complexity, and the probability of finding a meganuclease cleavage site within a gene of interest is still extremely low. These findings highlighted the need for artificial endonucleases with tailored specificities, cleaving chosen sequences with the same selectivity as natural endonucleases.
  • Meganucleases have emerged as scaffolds of choice for deriving genome engineering tools cutting a desired target sequence (Paques et al. Curr Gen Ther. 2007 7:49-66). Combinatorial assembly processes allowing to engineer meganucleases with modified specificities has been described by Arnould et al. J Mol. Biol. 2006 355:443-458; Arnould et al. J Mol. Biol. 2007 371:49-65; Smith et al. NAR 2006 34:e149; Grizot et al. NAR 2009 37:5405). Briefly, these processes rely on the identifications of locally engineered variants with a substrate specificity that differs from the substrate specificity of the wild-type meganuclease by only a few nucleotides. Up to four sets of mutations identified in such proteins can then be assembled in new proteins in order to generate new meganucleases with entirely redesigned binding interface.
  • These processes require two steps, wherein different sets of mutations are first assembled into homodimeric variants cleaving palindromic targets. Two homodimers can then be co-expressed in order to generate heterodimeric meganucleases cleaving the chosen non palindromic target. The first step of this process remains the most challenging one, and one cannot know in advance whether a meganuclease cleaving a given locus could be obtained with absolute certainty. Indeed, not all sequences are equally likely to be cleaved by engineered meganucleases, and in certain cases, meganuclease engineering could prove difficult (Galetto et al. Expert Opin Biol Ther. 2009 9:1289-303).
  • Other Enzymes Suitable for Site-Specific Genome Modifications
  • Specialized enzymes like integrases, recombinases, transposases and endonucleases have been proposed for site-specific genome modifications. For years, the use of these enzymes remained limited, due to the challenge of retargeting their natural specificities towards desired target sites. Indeed, the target sites of these proteins, or sequences with a sufficient degree of sequence identity, should be present in the sequences neighboring the mutations to be corrected, or within the gene to be inactivated, which is usually not the case, except in the case of pre-engineered sequences. The main challenge that would allow the use of these DNA modifying enzymes in gene therapy relies on the possibility of redesigning their DNA binding properties. Many strategies have been developed, aiming to obtain artificial proteins with tailored substrate specificities,
  • The integrase from the Streptomyces phage PhiC31 was used early for targeted gene transfer in an endogenous locus. This enzyme mediates recombination of the phage genome into the bacterial chromosome through a site-specific reaction between the phage attachment site (attP) and the bacterial attachment site (attB) (Kuhstoss et al. J Mol Biol 1991 222:897-908; Rausch et al. NAR 1991 19:5187-5189). This can occur from plasmids carrying attB sites into native genomic sequences harboring partial identity with attP, called pseudo attP sites (attP′). The PhiC31 integrase has been used to transfer several transgenes, including hFIX, in the human genome (Olivares et al. Nat Biotech 2002 20:1124-1128; Ginsburg et al. Adv Genet. 2005 54:179-187; Calos Curr Gene Ther 2006 6:633-645; Chalberg et al. J Mol Biol 2006 357:28-48; Aneja et al. J Gene Med 2007 9:967-975). The drawback here is that the site where integration can occur cannot be chosen (Chalberg et al. J Mol Biol 2006 357:28-48), and one has to rely on pseudo attP sites within the human genome loci, for precise integration. Whereas a major integration site is found on chromosome 19, hundreds other integration loci have been identified (Chalberg et al. J Mol Biol 2006 357:28-48). In recent work, the PhiC31 integrase was mutated in order to increase efficiency and specificity for integration at an attP′ site, paving the way for the development of engineered integrases that target chosen sites (Keravala et al. Mol Ther 2009 17:112-120). However, development of engineered integrases has lagged behind similar efforts focused on targeted recombinase and endonuclease systems.
  • Site-specific recombinases, such as the Cre recombinase from bacteriophage P1, or the Flp protein from Saccharomyces cerevisiae have been used to induce recombination between pre-engineered sequences containing their cognate sites. The Cre recombinase recognizes and mediates recombination between two identical 34 bp sites known as loxP (Abremski et al. Cell 1983 32:1301-1311). For many years, a limitation of Cre derived recombinases has been that repeated loxP, or pseudo loxP sites, must be present in order to allow DNA integration between these two sites. However, directed evolution of the DNA binding interface of this molecule has been used to create recombinases with new specificities (Buchholz et al. Nat Biotech 2001 19:1047-1052; Santoro et al. PNAS 2002 99:4185-4190). The Cre recombinase system has also been useful in providing a framework for the use of DNA targeting enzymes to induce the excision of viral sequences. Indeed, work with a retroviral Moloney murine leukemia virus vector system has shown that, when loxP sites are introduced in the LTR of an integrative retroviral vector, the expression of Cre can result in the deletion of all the sequences between the two loxP sites (Choulika et al. J Virol 1996 70:1792-1798). More recently, an engineered Cre recombinase variant has been used to excise an HIV type 1 provirus (Sarkar et al. Science 2007 316:1912-1915) from cells. The recombinase was redesigned to target the proviral LTRs, and used to induce the excision of all intervening sequences. Engineering attempts have also been made with the Flp recombinase, targeting the FRT (Flp Recombination Target) sequence (Buchholzt et al. Nat Biotech 1998 16:657-662), and variants recognizing non-native Flp recombination targets have been obtained (Voziyanov et al. J Mol Biol 2003 326:65-76). However, there is no example of targeted insertion in a non-pre-engineered locus with such enzymes today.
  • Transposons such as Piggy Back and Sleeping Beauty can provide efficient tools for insertion of sequences into vertebrate cells and have been proposed as an alternative to viral mediated gene delivery to achieve long-lasting expression (Izsvak et al. Mol ther 2004 9:147-156; Ivics et al. Curr Gene Ther 2006 6:593-607; Mates et al. Nat Genet. 2009 41:753-761). Transposons are a natural means of gene delivery in which a DNA sequence present in a DNA molecule is inserted in another location, through the action of the transposase. An engineered SB transposase, called SB100X was recently shown to increase the efficiency of the process (Mates et al. Nat Genet. 2009 41:753-761). Transposition is random on a genomic level (for example, SB integrates into TA dinucleotides (Vigdal et al. J Mol Biol 2002 323:441-452), and should therefore not be considered as tools for targeted approaches. However, further work has shown the possibility of chromosomal transposition mediated by engineered transposases in human cells, by fusing the transposase catalytic domain to specific DNA binding domains (Ivics, et al. Mol Ther 2007 15:1137-1144), paving the way for the development of a new category of targeted tools.
  • Gene Therapy
  • The successful treatment of several X-SCID patients by gene therapy nearly 10 years ago was one of the most significant milestones in the field of gene therapy. This tremendous achievement was followed by significant success in other clinical trials addressing different diseases, including another form of SCID, Epidermolysis Bullosa and Leber Amaurosis and others. However, these initial successes have long been overshadowed by a series of serious adverse events, i.e. the appearance of leukemia in X-SCID treated patients (Hacein-Bey-Abina et al. Science 2003 302:415-419; Hacein-Bey-Abina et al. J Clin Invest. 2008 118:3132-3142; Howe et al. J Clin Invest. 2008 118:3143-3150). All cases of leukemia, but one, could eventually be treated by chemotherapy, and the approach appears globally as a success, but these serious adverse effects highlighted the major risks of current gene therapy approaches.
  • There is thus a need in the art for a safe method for inserting a gene into the genome of a subject.
  • Most of the gene therapy protocols that are being developed these days for the treatment of inherited diseases are based on the complementation of a variant allele by an additional and functional copy of the disease-causing gene. In non-dividing tissues, such as retina, delivering this copy can be accomplished using a non integrative vector, derived for example, from an Adeno Associated Virus (AAV). However, when targeting stem cells, such as hematopoietic stem cells (HSCs), whose fate is to proliferate, persistent expression becomes an issue, and there is a need for integrative vectors. Retroviral vectors, which integrate in the genome and replicate with the hosts' chromosomes, have proved efficient for this purpose, but the random nature of their insertion has raised various concerns, all linked with gene expression. The cases of leukemia observed in the X-SCID trials were clearly linked to the activation of a proto-oncogene in the vicinity of the integration sites. In addition, inappropriate expression of the transgene could result in metabolic or immunological problems. Finally, insertion could result in the knock-out of endogenous genes.
  • Site-specific integration would be a promising alternative to random integration of viral vectors since it could alleviate the risks of insertional mutagenesis (Kolb et al. Trends Biotechnol. 2005 23:399-406; Porteus et al. Nat. Biotechnol. 2005 23:967-973; Paques et al. Curr Gen Ther. 2007 7:49-66). However, it is relatively tedious to engineer tools for targeted recombination. In addition, each tool has its intrinsic properties in terms of activity and specificity.
  • Therefore, there is a need in the art for a tool allowing the targeted insertion of transgenes into loci of the genome that can be considered as “safe harbors” for gene addition. In addition, it would be extremely advantageous if this tool could be used for inserting transgenes irrespective of their sequences, thereby allowing the treatment of numerous diseases by gene therapy using a same tool. Moreover, it would be extremely advantageous if this tool allowed inserting transgenes into the genome with a high efficacy, and led to stable expression of the transgene at high levels.
  • SUMMARY OF THE INVENTION
  • The invention is notably drawn to the following embodiments:
  • Embodiment 1
  • A variant endonuclease capable of cleaving a target sequence for use in inserting a transgene into the genome of an individual, wherein
      • i. said genome comprises a locus comprising said target sequence; and
      • ii. said target sequence is located at a distance of at most 200 kb from a retroviral insertion site (RIS), wherein said RIS is neither associated with cancer nor with abnormal cell proliferation.
    Embodiment 2
  • The endonuclease according to embodiment 1, wherein insertion of said transgene does not substantially modify expression of genes located in the vicinity of the target sequence.
  • Embodiment 3
  • The endonuclease according to embodiment 1 or 2, wherein said target sequence is located at a distance of at least 100 kb from the nearest genes.
  • Embodiment 4
  • The endonuclease according to any one of embodiments 1 to 3, wherein said RIS has been identified in cells from a patient treated by gene therapy by transduction of stem cells.
  • Embodiment 5
  • The endonuclease according to any one of embodiments 1 to 3, wherein said RIS has been identified in cells from a patient treated by gene therapy by transduction of hematopoietic stem cells.
  • Embodiment 6
  • The endonuclease according to any one of embodiments 1 to 5, wherein said endonuclease is a homing endonuclease.
  • Embodiment 7
  • The endonuclease according to embodiment 6, wherein said homing endonuclease is a member of the family of LAGLIDADG endonucleases.
  • Embodiment 8
  • The endonuclease according to embodiment 7, wherein said member of the family of LAGLIDADG endonucleases is I-CreI.
  • Embodiment 9
  • The endonuclease according to any one of embodiments 1 to 8, wherein said locus is selected from the SH3 locus on human chromosome 6p25.1, the SH4 locus on human chromosome 7q31.2, the SH6 locus on human chromosome 21q21.1, the SH12 locus on human chromosome 13q34, the SH13 locus on human chromosome 3p12.2, the SH19 locus on human chromosome 22, the SH20 locus on human chromosome 12q21.2, the SH21 locus on human chromosome 3p24.1, the SH33 locus on human chromosome 6p12.2, the SH7 locus on human chromosome 2p16.1 and the SH8 locus on human chromosome 5.
  • Embodiment 10
  • In vitro or ex vivo use of an endonuclease as defined in any one of embodiments 1 to 9 for inserting a transgene into the genome of a cell or a tissue.
  • Embodiment 11
  • A variant dimeric I-CreI protein comprising two monomers that comprise a sequence at least 80% identical to SEQ ID NO: 1 or SEQ ID NO: 42, wherein:
      • i. said dimeric I-CreI protein is capable of cleaving a target sequence located within a locus of an individual, said target sequence being located at a distance of at most 200 kb from a retroviral insertion site (RIS), and said RIS being neither associated with cancer nor with abnormal cell proliferation; and
      • ii. said target sequence does not comprise a sequence of SEQ ID NO: 4.
    Embodiment 12
  • The dimeric I-CreI protein according to embodiment 11, wherein said dimeric I-CreI protein is capable of cleaving a target sequence located within the SH3 locus on human chromosome 6p25.1.
  • Embodiment 13
  • The dimeric I-CreI protein according to embodiment 12, wherein said target sequence comprises the sequence of SEQ ID NO: 2.
  • Embodiment 14
  • The dimeric I-CreI protein according to embodiment 12 or 13, wherein said protein comprises:
      • a) a first monomer that comprises amino acid substitutions at positions 30, 38, 70 and 75 of SEQ ID NO: 1; and
      • b) a second monomer that comprises amino acid substitutions at positions 44, 54, 70 and 75 of SEQ ID NO: 1.
    Embodiment 15
  • The dimeric I-CreI protein according to embodiment 14, wherein said polypeptide comprises:
      • a) a first monomer comprising 30G 38R 70D 75N 86D mutations;
      • b) a second monomer selected from the group consisting of:
        • i. a monomer comprising 44A 54L 64A 70Q 75N 158R 162A mutations;
        • ii. a monomer comprising 44A 54L 70Q 75Y 92R 158R 162A mutations;
        • iii. a monomer comprising 4E 44A 54L 64A 70Q 75N 158R 162A mutations;
        • iv. a monomer comprising 44A 54L 64A 70Q 75N 158W 162A mutations;
        • v. a monomer comprising 44A 54L 70Q 75N mutations;
        • vi. a monomer comprising 44A 54L 57E 70Q 75N 158R 162A mutations; and
        • vii. a monomer comprising 44V 54L 70Q 75N 77V mutations;
    Embodiment 16
  • The dimeric I-CreI protein according to embodiment 14, wherein said polypeptide comprises:
      • a) a first monomer comprising 30G 38R 70D 75N 81T 154G mutations;
      • b) a second monomer selected from the group consisting of:
        • i. a monomer comprising 44A 54L 70Q 75N 105A 158R 162A mutations;
        • ii. a monomer comprising 44A 54L 64A 70Q 75N 158R 162A mutations;
        • iii. a monomer comprising 4E 44A 54L 64A 70Q 75N 158R 162A mutations;
        • iv. a monomer comprising 44A 54L 64A 70Q 75N 158W 162A mutations;
        • v. a monomer comprising 44A 54L 70Q 75N mutations; and
        • vi. a monomer comprising 44V 54L 70Q 75N 77V mutations;
    Embodiment 17
  • The dimeric I-CreI protein according to embodiment 14, wherein said polypeptide comprises:
      • a) a first monomer comprising 30G 38R 50R 70D 75N 142R mutations;
      • b) a second monomer selected from the group consisting of:
        • i. a monomer comprising 44A 54L 70Q 75N 105A 158R 162A mutations;
        • ii. a monomer comprising 44A 54L 64A 70Q 75N 158R 162A mutations;
        • iii. a monomer comprising 44A 54L 70Q 75Y 92R 158R 162A mutations;
        • iv. a monomer comprising 4E 44A 54L 64A 70Q 75N 158R 162A mutations;
        • v. a monomer comprising 44A 54L 64A 70Q 75N 158W 162A mutations;
        • vi. a monomer comprising 44A 54L 66C 70Q 71 R 75N 151A 158R 162A mutations;
        • vii. a monomer comprising 44A 54L 70Q 75N mutations;
        • viii. a monomer comprising 44A 54L 57E 70Q 75N 158R 162A mutations; and
        • ix. a monomer comprising 44V 54L 70Q 75N 77V mutations;
    Embodiment 18
  • The dimeric I-CreI protein according to embodiment 11, wherein said dimeric I-CreI protein is capable of cleaving a target sequence located within the SH4 locus on human chromosome 7q31.2.
  • Embodiment 19
  • The dimeric I-CreI protein according to embodiment 18, wherein said target sequence comprises the sequence of SEQ ID NO: 3.
  • Embodiment 20
  • The dimeric I-CreI protein according to embodiment 18 or 19, wherein said protein comprises:
      • a) a first monomer that comprises amino acid substitutions at positions 24, 70, 75 and 77 of SEQ ID NO: 1; and
      • b) a second monomer that comprises amino acid substitutions at positions 24, 44 and 70 of SEQ ID NO: 1.
    Embodiment 21
  • The dimeric I-CreI protein according to embodiment 20, wherein said polypeptide comprises:
      • a) a first monomer selected from the group consisting of:
        • i. a monomer comprising 24V 44R 68Y 70S 75Y 77N mutations;
        • ii. a monomer comprising 24V 68A 70S 75N 77R mutations; and
        • iii. a monomer comprising 24V 70D 75N 77R mutations;
      • b) a second monomer selected from the group consisting of:
        • i. a monomer comprising 24V 44Y 70S mutations; and
        • ii. a monomer comprising 24V 44Y 70S 77V mutations.
    Embodiment 22
  • The dimeric I-CreI protein according to embodiment 11, wherein said dimeric I-CreI protein is capable of cleaving a target sequence located within the SH6 locus on human chromosome 21q21.1.
  • Embodiment 23
  • The dimeric I-CreI protein according to embodiment 22, wherein said target sequence comprises the sequence of SEQ ID NO: 59.
  • Embodiment 24
  • The dimeric I-CreI protein according to embodiment 22 or 23, wherein said protein comprises:
      • a) a first monomer that comprises amino acid substitutions at positions 44, and optionally at positions 70 and/or 75 of SEQ ID NO: 1; and
      • b) a second monomer that comprises amino acid substitutions at positions 28, 40, 44, 70 and 75 of SEQ ID NO: 1.
    Embodiment 25
  • The dimeric I-CreI protein according to embodiment 24, wherein said polypeptide comprises:
      • a) a first monomer comprising 44K 68T 70G 75N mutations; and
      • b) a second monomer selected from the group consisting of:
        • i. a monomer comprising 28Q 40R 44A 70L 75N 96R 111H 144S mutations;
        • ii. a monomer comprising 7R 28Q 40R 44A 70L 75N 85R 103T mutations;
        • iii. a monomer comprising 28Q 40R 44A 70L 75N 103S mutations;
        • iv. a monomer comprising 24F 27V 28Q 40R 44A 70L 75N 99R mutations;
        • v. a monomer comprising 7R 28Q 40R 44A 70L 75N 81T mutations;
        • vi. a monomer comprising 7R 28Q 40R 44A 70L 75N 77V mutations;
        • vii. a monomer comprising 7R 28Q 40R 44A 70L 75N 103T 121E 132V 160R mutations;
        • viii. a monomer comprising 28Q 40R 44A 70L 75N mutations;
        • ix. a monomer comprising 7R 28Q 40R 44A 70L 75N 103T mutations; and
        • x. a monomer comprising 28Q 34R 40R 44A 70L 75N 81V 103T 108V 160E mutations.
    Embodiment 26
  • The dimeric I-CreI protein according to embodiment 24, wherein said polypeptide comprises:
      • a) a first monomer comprising a 44K mutation, and optionally 70S and/or 75N mutations; and
      • b) a second monomer selected from the group consisting of:
        • i. a monomer comprising 28Q 40R 44A 70L 75N 96R 111H 144S mutations;
        • ii. a monomer comprising 7R 28Q 40R 44A 70L 75N 85R 103T mutations;
        • iii. a monomer comprising 28Q 40R 44A 70L 75N 103S mutations;
        • iv. a monomer comprising 24F 27V 28Q 40R 44A 70L 75N 99R mutations;
        • v. a monomer comprising 7R 28Q 40R 44A 70L 75N 81T mutations;
        • vi. a monomer comprising 7R 28Q 40R 44A 70L 75N 103T 121E 132V 160R mutations;
        • vii. a monomer comprising 7R 28Q 40R 44A 70L 75N 103T mutations; and
        • viii. a monomer comprising 28Q 34R 40R 44A 70L 75N 81V 103T 108V 160E mutations.
    Embodiment 27
  • A fusion protein comprising the monomers of the dimeric I-CreI protein according to any one of embodiments 11 to 26.
  • Embodiment 28
  • The fusion protein according to embodiment 27, wherein said monomers are connected by a peptidic linker comprising a sequence of SEQ ID NO: 43.
  • Embodiment 29
  • The fusion protein according to embodiment 27 or 28, wherein the C-terminal monomer further comprises K7E and K96E mutations, and wherein the N-terminal monomer further comprises E8K, E61R and G19S mutations.
  • Embodiment 30
  • The fusion protein according to any one of embodiments 27 to 29, wherein said fusion protein comprises a sequence selected from the group consisting of SEQ ID Nos. 25-40 and 76-96.
  • Embodiment 31
  • A nucleic acid encoding the endonuclease according to any one of embodiments 1-9 or the protein according to any one of embodiments 11 to 30.
  • Embodiment 32
  • An expression vector comprising the nucleic acid according to embodiment 31.
  • Embodiment 33
  • The expression vector according to embodiment 32, further comprising a targeting construct comprising a transgene and two sequences homologous to the genomic sequence flanking a target sequence recognized by the endonuclease as defined in one of embodiments 1-9 or by the protein as defined in any one of embodiments 11 to 30.
  • Embodiment 34
  • The expression vector of embodiment 33, wherein said transgene encodes a therapeutic polypeptide.
  • Embodiment 35
  • The expression vector according to any one of embodiments 32 to 34 for use in gene therapy.
  • Embodiment 36
  • A combination of:
      • an expression vector according to embodiment 32; and
      • a vector comprising a targeting construct comprising a transgene and two sequences homologous to the genomic sequence of a target sequence a recognized by the endonuclease as defined in one of embodiments 1-9 or by the protein as defined in any one of embodiments 11 to 30.
    Embodiment 37
  • A pharmaceutical composition comprising the expression vector as defined in any one of embodiments 32 to 34 or the combination as defined in embodiment 36 and a pharmaceutically active carrier.
  • Embodiment 38
  • A method of treating an individual by gene therapy comprising administering an effective amount of the expression vector as defined in any one of embodiments 32 to 34 or of the combination as defined in embodiment 36 to an individual in need thereof.
  • Embodiment 39
  • A method for obtaining an endonuclease suitable for inserting a transgene into the genome of an individual, comprising the step of:
      • a) selecting, within the genome of said individual, a retroviral insertion site (RIS) that is neither associated with cancer nor with abnormal cell proliferation;
      • b) defining a genomic region extending 200 kb upstream and 200 kb downstream of said RIS; and
      • c) identifying a wild-type endonuclease or constructing a variant endonuclease capable of cleaving a target sequence located within said genomic region.
    Embodiment 40
  • Use of the endonuclease according to any one of embodiments 1 to 9, or of the protein according to any one of embodiments 11 to 30, or of the nucleic acid according to embodiment 31, or of the expression vector according to any one of embodiments 32 to 34, or of the combination according to embodiment 36, for inserting a transgene into the genome of a cell, tissue or non-human animal, wherein said use is not therapeutic.
  • Embodiment 41
  • The use of embodiment 40, for making a non-human animal model of a hereditary disorder.
  • Embodiment 42
  • The use of embodiment 40, for producing a recombinant protein.
  • Embodiment 43
  • A non-human transgenic animal comprising a nucleic acid according to embodiment 31, or an expression vector according to any one of embodiments 32-34, or a combination according to embodiment 36 in its genome.
  • DETAILED DESCRIPTION OF THE INVENTION
  • The inventors have identified “safe harbors” loci within the genome allowing safe expression of a transgene through targeted insertion wherein (i) said loci are close to a retroviral insertion site identified in a cell from a patient treated by gene therapy, and (ii) said retroviral insertion are not associated with cancer or abnormal cell proliferation. As immediately apparent from the following description and examples, the safe harbor loci according to the invention may either be located within the intron of a gene, or within an intergenic region.
  • In particular, the inventors have found that endonucleases could be engineered in such a way as to target said safe harbors for gene addition.
  • More specifically, the inventors have engineered several I-CreI meganucleases that are capable of recognizing and cleaving target sequences located within different safe harbors loci, for instance the SH6, the SH3 locus, the SH4 locus, the SH12 locus, the SH13 locus, the SH19, the SH20 locus, the SH21 locus, the SH33 locus, the SH7 locus, the SH8 locus, the SH18 locus, the SH31 locus, the SH38 locus, the SH39 locus, the SH41 locus, the SH42 locus, the SH43 locus, the SH44 locus, the SH45 locus, the SH46 locus, the SH47 locus, the SH48 locus, the SH49 locus, the SH50 locus, the SH51 locus, the SH52 locus, the SH70 locus, the SH71 locus, the SH72 locus, the SH73 locus, the SH74 locus, the SH75 locus, the SH101 locus, the SH106 locus, the SH107 locus, the SH102 locus, the SH105 locus, the SH103 locus, the SH104 locus, the SH113 locus, the SH109 locus, the SH112 locus, the SH108 locus, the SH110 locus, the SH114 locus, the SH116 locus, the SH111 locus, the SH115 locus, the SH121 locus, the SH120 locus, the SH122 locus, the SH117 locus, the SH118 locus, the SH119 locus, the SH123 locus, the SH126 locus, the SH128 locus, the SH129 locus, the SH124 locus, the SH131 locus, the SH125 locus, the SH127 locus, the SH130 locus, the SH11 locus, the SH17 locus, the SH23 locus, the SH34 locus, the SH40 locus, the SH53 locus, the SH54 locus, the SH55 locus, the SH56 locus, the SH57 locus, the SH58 locus, the SH59 locus, the SH60 locus, the SH61 locus, the SH62 locus, the SH65 locus, the SH67 locus, the SH68 locus and the SH69 locus that are further described herein.
  • It has further been shown that these meganucleases can cleave their target sequences efficiently.
  • These meganucleases, as well as other enymes like integrases, recombinases and transposases, can therefore be used as a tool for inserting a transgene into safe harbors, thereby avoiding the appearance of adverse events such as leukemia in the frame of gene therapy. In addition, these meganucleases, as well as other enymes like integrases, recombinases and transposases can be used for inserting any transgene into the safe harbor starting from a single targeting construct irrespective of the sequence of the transgene.
  • Endonucleases According to the Invention and Uses Thereof.
  • The invention therefore relates to:
      • an endonuclease capable of cleaving a target sequence for use in inserting a transgene into the genome of an individual, wherein (i) said genome comprises a locus comprising said target sequence, and (ii) said target sequence is located at a distance of at most 200 kb from a retroviral insertion site (RIS), wherein said RIS is neither associated with cancer nor with abnormal cell proliferation.
      • an in vitro or ex vivo use of an endonuclease capable of cleaving a target sequence for inserting a transgene into the genome of a cell or a tissue, (i) said genome comprises a locus comprising said target sequence, and (ii) said target sequence is located at a distance of at most 200 kb from a retroviral insertion site (RIS), wherein said RIS is neither associated with cancer nor with abnormal cell proliferation.
      • a method for inserting a transgene into the genome of an individual comprising the steps of (i) providing an endonuclease capable of cleaving a target sequence, wherein said genome comprises a locus comprising said target sequence, and said target sequence is located at a distance of at most 200 kb from a retroviral insertion site (RIS) that is neither associated with cancer nor with abnormal cell proliferation; (ii) contacting an individual with a transgene and with said endonuclease, whereby said transgene is inserted into said locus of the genome of the individual.
  • As used herein, the term “endonuclease” refers to any wild-type or variant enzyme capable of catalyzing the hydrolysis (cleavage) of bonds between nucleic acids within of a DNA or RNA molecule, preferably a DNA molecule. The endonucleases according to the present invention do not cleave the DNA or RNA molecule irrespective of its sequence, but recognize and cleave the DNA or RNA molecule at specific polynucleotide sequences, further referred to as “target sequences” or “target sites”. Target sequences recognized and cleaved by an endonuclease according to the invention are referred to as target sequences according to the invention.
  • The endonuclease according to the invention can for example be a homing endonuclease (Paques et al. Curr Gen Ther. 2007 7:49-66), a chimeric Zinc-Finger nuclease (ZFN) resulting from the fusion of engineered zinc-finger domains with the catalytic domain of a restriction enzyme such as Fokl (Porteus et al. Nat. Biotechnol. 2005 23:967-973) or a chemical endonuclease (Arimondo et al. Mol Cell Biol. 2006 26:324-333; Simon et al. NAR 2008 36:3531-3538; Eisenschmidt et al. NAR 2005 33:7039-7047; Cannata et al. PNAS 2008 105:9576-9581). In chemical endonucleases, a chemical or peptidic cleaver is conjugated either to a polymer of nucleic acids or to another DNA recognizing a specific target sequence, thereby targeting the cleavage activity to a specific sequence.
  • The endonuclease according to the invention is preferably a homing endonuclease, also known under the name of meganuclease. Such homing endonucleases are well-known to the art (see e.g. Stoddard, Quarterly Reviews of Biophysics, 2006, 38:49-95). Homing endonucleases recognize a DNA target sequence and generate a single- or double-strand break. Homing endonucleases are highly specific, recognizing DNA target sites ranging from 12 to 45 base pairs (bp) in length, usually ranging from 14 to 40 bp in length. The homing endonuclease according to the invention may for example correspond to a LAGLIDADG endonuclease, to a HNH endonuclease, or to a GIY-YIG endonuclease. Examples of such endonuclease include I-Sce I, I-Chu I, I-Cre I, I-Csm I, PI-Sce I, PI-Tli I, PI-Mtu I, I-Ceu I, I-Sce II, I-Sce III, HO, PI-Civ I, PI-Ctr I, PI-Aae I, PI-Bsu I, PI-Dha I, PI-Dra I, PI-Mav I, PI-Mch I, PI-Mfu I, PI-Mfl I, PI-Mga I, PI-Mgo I, PI-Min I, PI-Mka I, PI-Mle I, PI-Mma I, PI-Msh I, PI-Msm I, PI-Mth I, PI-Mtu I, PI-Mxe I, PI-Npu I, PI-Pfu I, PI-Rma I, PI-Spb I, PI-Ssp I, PI-Fac I, PI-Mja I, PI-Pho I, PI-Tag I, PI-Thy I, PI-Tko I, PI-Tsp I, I-MsoI.
  • In a preferred embodiment, the homing endonuclease according to the invention is a LAGLIDADG endonuclease such as I-SceI, I-CreI, I-CeuI, I-MsoI, and 1-DmoI.
  • In a most preferred embodiment, said LAGLIDADG endonuclease is I-CreI. Wild-type I-CreI is a homodimeric homing endonuclease that is capable of cleaving a 22 to 24 bp double-stranded target sequence. The sequence of a wild-type monomer of I-CreI includes the sequence shown as SEQ ID NO: 1 (which corresponds to the I-CreI sequence of pdb accession number 1g9y) and the sequence shown in SwissProt Accession n° P05725 (in particular the sequence shown in version 73, last modified Nov. 3, 2009).
  • In the present patent application, the I-CreI variants may comprise an additional alanine after the first methionine of the wild type I-CreI sequence, and three additional amino acid residues at the C-terminal extremity (see sequence of SEQ ID NO: 42 and FIG. 11). These three additional amino acid residues consist of two additional alanine residues and one aspartic acid residue after the final proline of the wild type I-CreI sequence. These additional residues do not affect the properties of the enzyme. For the sake of clarity, these additional residues do not affect the numbering of the residues in I-CreI or variants thereof. More specifically, the numbering used herein exclusively refers to the position of residues in the wild type I-CreI enzyme of SEQ ID NO: 1. For instance, the second residue of wild-type I-CreI is in fact the third residue of a variant of SEQ ID NO: 42 since this variant comprises an additional alanine after the first methionine.
  • In the present application, I-CreI variants may be homodimers (meganuclease comprising two identical monomers), heterodimers (meganuclease comprising two non-identical monomers) and single-chains.
  • The invention encompasses both wild-type (naturally-occurring) and variant endonucleases. In a preferred embodiment, the endonuclease according to the invention is a “variant” endonuclease, i.e. an endonuclease that does not naturally exist in nature and that is obtained by genetic engineering or by random mutagenesis. The variant endonuclease according to the invention can for example be obtained by substitution of at least one residue in the amino acid sequence of a wild-type, naturally-occurring, endonuclease with a different amino acid. Said substitution(s) can for example be introduced by site-directed mutagenesis and/or by random mutagenesis. In the frame of the present invention, such variant endonucleases remain functional, i.e. they retain the capacity of recognizing and specifically cleaving a target sequence.
  • The variant endonuclease according to the invention cleaves a target sequence that is different from the target sequence of the corresponding wild-type endonuclease. For example, the target sequence of a variant I-CreI endonuclease is different from the sequence of SEQ ID NO: 4. Methods for obtaining such variant endonucleases with novel specificities are well-known in the art.
  • The present invention is based on the finding that such variant endonucleases with novel specificities can be used for inserting a gene into a “safe harbor” locus of the genome of a cell, tissue or individual.
  • As used herein, the term “locus” is the specific physical location of a DNA sequence (e.g. of a gene) on a chromosome. As used in this specification, the term “locus” usually refers to the specific physical location of an endonuclease's target sequence on a chromosome. Such a locus, which comprises a target sequence that is recognized and cleaved by an endonuclease according to the invention, is referred to as “locus according to the invention”.
  • Ideally, insertion into a safe harbor locus should have no impact on the expression of other genes. Testing these properties is a multi-step process, and a first pre-screening of candidate safe harbor loci by bioinformatic means is desirable. One can thus first identify loci in which targeted insertion is unlikely to result in insertional mutagenesis.
  • One of the major features of a locus according to the invention is that (i) it is located in a region wherein retroviral insertion was observed in a cell from a patient, in a gene therapy clinical trial, and (ii) said retroviral insertion has not been associated with a cancer or an abnormal cell proliferation.
  • Indeed, one way to identify safe habor loci according to the invention is to use the data generated by former gene therapy trials. In the X-SCID trial, insertions of retroviral vector-borne transgenes next to the LMO2 and CCND2 genes have been shown to be associated with leukemia. The follow up of vector insertions in patients have clearly demonstrated that cells carrying this insertion had outnumbered the other modified cells after a several years process (Hacein-Bey-Abina et al. Science 2003 302:415-9; Deichmann et al. J. of Clin. Invest. 2007 117:2225-32, Cavazzana-Calvo et al. Blood 2007 109:4575-4581). In another clinical trial, insertion in several loci were found to trigger a high proliferation rate in two patients (Ott et al. Nat Med 2006 12:401-9). In these cases, proliferation seemed to be a consequence of the insertional activation of the MDS1-EVI1, PRDM16, or SETBP1 genes. Although malignancy was not observed initially, EVII activation eventually resulted in myelodysplasia in both patients (Stein et al., Nat. Med. 2010 16: 198-205). More generally, even if non oncogenic, cell proliferation resulting from activation of a gene close to the insert could represent a first step towards malignancy, and therefore lead to potential problems in terms of safety. In order to better understand the pattern of viral vector integration, and its potential consequences on the fate of transformed cells, several large scale studies of Retroviral Insertion Sites (RIS) have been conducted in patients from gene therapy trials (Mavilio et al., Nat Med 2006:1397-1402; Recchia et al. PNAS 2006:1457-62; Aiuti, et al. J Clin Invest 2007:2233-40; Schwarzwaelder et al. J Clin Invest 2007:2241-9; Deichmann et al. J Clin Invest 2007:2225-32). RIS which are not associated with leukemia or with abnormal cell proliferation can be considered as safe harbors. Therefore, the locus according to the invention preferably overlaps or is close to a RIS identified in a clinical trial, and yet not associated with cancer or abnormal cell proliferation.
  • More specifically, the locus according to the invention is defined as a locus comprising a target sequence that is located at a distance of at most 200, 180, 150, 100 or 50 kb from a retroviral insertion site (RIS), said RIS being neither associated with cancer nor with abnormal cell proliferation. Such loci are referred to as “safe harbor” loci according to the invention (or loci according to the invention), i.e. loci that are safe for insertion of transgenes.
  • By “Retroviral insertion sites” (RIS) is meant a genomic site which was identified as an insertion site for a retroviral vector in a cell from a patient treated by gene therapy with said retroviral vector. Such RIS are well-known to the art. They include but are not limited to those described in Schwarzwaelder et al. (J. Clin. Invest. 2007 117:2241), Deichmann et al. (J. of Clin. Invest. 2007 117:2225), Aiuti et al. (J. Clin. Invest. 2007 117:2233), Recchia et al. (PNAS 2006 103:1457) and Mavilio et al. (Nature Medicine 12:1397, 2006).
  • By “retroviral vector” is meant any vector derived from a virus from the retroviridae family.
  • The RIS according to the invention is neither associated with cancer nor with abnormal cell proliferation. RIS known to be associated with leukemia or with abnormal cell proliferation are well known in the art and can easily be excluded by the skilled in the art. Such RIS known to be associated with leukemia or with abnormal cell proliferation include, e.g., insertion sites next to the LMO2, CCND2, MDS1-EVI1, PRDM16, and SETBP1 genes.
  • In a more preferred embodiment according to the invention, the RIS used to define safe harbor loci have been identified in a clinical trial, with the transduced cells being stem cells. The RIS can thus have been identified in cells from a patient treated by gene therapy by transduction of stem cells.
  • In another most preferred embodiment according to the invention, the RIS used to define safe harbor loci have been identified in a clinical trial for SCID patients, with the transduced cells being hematopoietic stem cells (HSCs). The RIS can thus have been identified in cells from a patient treated by gene therapy by transduction of hematopoietic stem cells.
  • Furthermore, more stringent criteria for definition of a RIS according to the invention can be used.
  • Among RIS, Common Integration sites (CIS) are loci in which the statistical over representation of RIS could be interpreted as the consequence of cell high proliferation rate upon insertion. (Mikkers et al., 2003, Nat. Genet. 32:153; Lund et al., 2002, Nat. Genet. 32:160; Hemati et al. 2004, PLOS Biol. 2:e423; Suzuki et al., 2002, Nat. Genet. 32:166-174; Deichman et al. J. of Clin. Invest. 2007 117:2225-32). For example, Deichman et al. (J. of Clin. Invest. 2007 117:2225-32) made a survey of RIS from 9X-SCID patients treated by gene therapy, and found 572 unique RIS that could be mapped unequivocally to the human genome. Among them, they defined CIS of second, third, fourth, fifth, and higher order. CIS of second orders were defined by the occurrence of two retroviral insertions within a 30 kb distance, CIS of third, fourth and fifth order by the occurrence of 3, 4 or 5 insertions within 50, 100 or 200 kb, respectively. 122 RIS were found in 47 different CIS loci, 33-fold the value expected under random distribution of the RIS. Eleven CIS were found to localize next to proto-oncogenes, including ZNF217, VAV-3, CCND2, LMO2, MDS1, BCL2L1, NOTCH2, SOCS2, RUNX1, RUNX3, and SEPT6.
  • To ensure maximal safety, it could be preferred to avoid RIS located within CIS. Therefore, in a preferred embodiment according to the invention, the target sequence according to the invention is not located in a CIS, In addition, said target sequence or locus is preferably located at a distance of at least 50, 100 or 200 kb from a RIS being part of a common integration site (CIS).
  • By “Common Integration site” (CIS) is meant a genomic region of 30 kb, 50 kb, 100 kb or 200 kb wherein RIS identified in clinical trials are overrepresented (assuming a random distribution of insertions). Such CIS are well known in the art and are described in Schwarzwaelder et al. (J. Clin. Invest. 2007 117:2241), Deichmann et al. (J. of Clin. Invest. 2007 117:2225), Aiuti et al. (J. Clin. Invest. 2007 117:2233), Recchia et al. (PNAS 2006 103:1457), Mavilio et al. (Nature Medicine 12:1397, 2006) and Gabriel et al. (Nat. Med. 2009 15(12):143.
  • In addition to be close to a RIS, targeted integration into the locus according to the invention should not result in the disruption of essential functions in the targeted cell.
  • Therefore, in a specific embodiment according to the invention, insertion into the locus according to the invention does preferably not substantially modify expression of genes located in the vicinity of the target sequence, for example of the nearest genes.
  • In addition, in another specific embodiment, insertion of a genetic element into said locus does preferably not substantially modify the phenotype of said cell, tissue or individual (except for the phenotype due to expression of the genetic element). By “phenotype” is meant a cell's, a tissue's or an individual's observable traits. The phenotype includes e.g. the viability, the cellular proliferation and/or the growth rate. The skilled in the art can easily verify that a locus is a safe harbor locus according to the invention e.g. by analyzing the expression pattern of adjacent genes, by carrying out micro-array studies of transcriptome and/or by characterizing proliferation and/or differentiation abnormalities (if any).
  • In still another specific embodiment, the locus according to the invention does not comprise any gene. A locus that does not comprise any gene refers to a locus that does not comprise any referenced or known gene. In other terms, such a locus does not comprise any known gene according to sequence databases such as those available on the National Center for Biotechnology Information (NCBI) website. Therefore, the target sequence according to the invention and/or the locus according to the invention can advantageously be located at a distance of at least 1, 5, 10, 25, 50, 100, 180, 200, 250, 300, 400 or 500 kb from the nearest genes.
  • By “gene” is meant the basic unit of heredity, consisting of a segment of DNA arranged in a linear manner along a chromosome, which codes for a specific protein or segment of protein. A gene typically includes a promoter, a 5′ untranslated region, one or more coding sequences (exons), optionally introns, a 3′ untranslated region. The gene may further comprise a terminator, enhancers and/or silencers.
  • By “nearest genes” is meant the one, two or three genes that are located the closest to the target sequence, centromeric and telomeric to the target sequence respectively.
  • In a preferred embodiment, the locus according to the invention further allows stable expression of the transgene.
  • In another preferred embodiment, the target sequence according to the invention is only present once within the genome of said cell, tissue or individual.
  • Once such a safe harbor locus according to the invention has been selected, one can then (i) either construct a variant endonuclease specifically recognizing and cleaving a target sequence located within said locus, e.g. as described in Examples 1, 2 and 5, or (ii) determine whether a known wild-type endonuclease is capable of cleaving a target sequence located within said locus. Alternatively, once a safe harbor locus according to the invention has been selected, the skilled in the art can insert therein a target sequence that is recognized and cleaved by a known wild-type or variant endonuclease.
  • Therefore, the invention is drawn to a method for obtaining an endonuclease suitable for inserting a transgene into the genome of an individual, comprising the step of:
      • a) selecting and/or identifying, within the genome of said individual, a retroviral insertion site (RIS) that is neither associated with cancer nor with abnormal cell proliferation;
      • b) defining a genomic region extending 200 kb upstream and 200 kb downstream of said RIS; and
      • c) identifying a wild-type endonuclease or constructing a variant endonuclease capable of cleaving a target sequence located within said genomic region.
        Such an endonuclease allows safely inserting a transgene into the genome of the cell, tissue or individual, for example without substantially modifying (i) expression of the nearest genes, and/or (ii) the cellular proliferation and/or the growth rate of the cell, tissue or individual.
  • All criteria presented hereabove in connection with the locus according to the invention can of course be applied when carried out the above method. For example, RIS being part of a CIS may be excluded, and/or the genomic region defined at step (b) may only extend 50 kb upstream and 50 kb downstream of said RIS, and/or the locus comprising the target sequence may not comprise any gene.
  • The locus according to the invention may for example correspond to any one of the SH3, SH4, SH6, SH12, SH13, SH19, SH20, SH21, SH33, SH7 or SH8 loci which are described in Tables A to C below.
  • Table A provides the location of the locus within the human genome, a target sequence comprised within the locus, the location of the closest RIS as well as the reference to a publication describing the RIS, and examples of endonucleases according to the invention that cleave the locus.
  • Table B provides information about the nearest genes that are located immediately upstream (at 5′) and downstream (at 3′) of the locus according to the invention. The distance indicates the distance between the target sequence and the nearest coding sequence of the gene.
  • Table C and D provide similar information as Table B, but for the second nearest genes and for the third nearest genes, respectively.
  • Tables A′, B′, C′ and D′ provide updated information similar to that in Tables A, B, C and D, respectively, for some loci and associated examples of target sequences within these loci, namely SH3, SH4, SH6, SH8 and SH19. Updated localization information is given by reference to GRCh37/hg19 version of the human genome assembly.
  • The locus according to the invention may also correspond to any one of the SH18, SH31, SH38, SH39, SH41, SH42, SH43, SH44, SH45, SH46, SH47, SH48, SH49, SH50, SH51, SH52, SH70, SH71, SH72, SH73, SH74 and SH75 which are described in Tables A″ to D″ below.
  • Table A″ provides the location of the locus within the human genome, a target sequence comprised within the locus, the location of the closest RIS as well as the reference to a publication describing the RIS, the distance between said target and the closest RIS and examples of endonucleases according to the invention that cleave the locus.
  • Table B″ provides information about the nearest genes that are located immediately upstream (at 5′) and downstream (at 3′) of the locus according to the invention. The distance indicates the distance between the target sequence and the nearest coding sequence of the gene.
  • Table C″ and D″ provide similar information as Table B″, but for the second nearest genes and for the third nearest genes, respectively.
  • Locations of loci, targets in this loci and genes are given according to GRCh37/hg19 version of the human genome assembly.
  • TABLE A
    Example of Target Cleaved by
    Sequence SEQ Close to  RIS meganucleases
    Human Comprised within  ID a RIS at described (examples) of
    Name chromosome locus the locus: NO: position: in: SEQ ID NO:
    SH3  6 6p25.1 CCAATACAAGGTACAAAG 54 6837845 Deichmann, 2007 25-32
    TCCTGA
    SH4
     7 7q31.2 TTAAAACACTGTACACCA 55 114606124 Schwarzwaelder, 2007 33-40
    TTTTGA
    SH6 21 21q21.1 TTAATACCCCGTACCTAA 59 17265069 Schwarzwaelder, 2007 76-85
    TATTGC
    SH12 13 13q34 ATAAAACAAGTCACGTTA 97 109463429 Mavilio, 2006 89
    TTTTGG
    SH13
     3 3p12.2 ATTACACTCTTTAAGTGA 98 80607284 Recchia, 2006 90
    TTTTAA
    SH19
    22 chr22 GCAAAACATTGTAAGACA 99 46815611 Aiuti, 2007 91
    TGCCAA
    SH20
    12 12q21.2 GCTGGCTGCTTCACATTG 100 74339720 Mavilio, 2006 92
    GAGAGA
    SH21
     3 3p24.1 TAGAAATCTGTTAAAAGA 101 31235316 Deichmann, 2007 93-95
    GATGAT
    SH33
     6 6p12.2 TTTTCATCACTTAAAGTG 102 50055278 Recchia, 2006 96
    TTTTAA
    SH7
     2 2p16.1 ACAACACTTTGTGAGACG 103 58962165 Deichmann, 2007 86-87
    TCTAAG
    SH8
     5 chr5 ACAATCTGAGGTAAGTAA 104 20572231 Aiuti, 2007 88
    TACTGA
  • TABLE A′
    Example of RIS Cleaved 
    Target position by mega-
    Human Sequence Close  according  nucleases
    chro- Comprised SEQ to a RIS  to RIS RIS (examples)
    mo-  within  ID at GRCh37/ Distance described of SEQ ID
    Name some locus the locus: NO:  position: hg19 (bases) in: NO:
    SH3  6 6p25.1 CCAATACAAGGTAC  54 6837845 6892846 40782 Deichmann, 25-32
    AAAGTCCTGA 2007
    SH4  7 7q31.2 TTAAAACACTGTAC  55 114606124 115051621 77337 Schwarzwaelder, 33-40
    ACCATTTTGA 2007
    SH6 21 21q21.1 TTAATACCCCGTAC  59 17265069 18343198 96099 Schwarzwaelder, 76-85
    CTAATATTGC 2007
    SH8  5 chr5 ACAATCTGAGGTAA 104 20572231 20536474 50714 Aiuti, 2007 88
    GTAATACTGA
    SH19
    22 chr22 GCAAAACATTGTAA  99 46815611 20536474 97664 Aiuti, 2007 91
    GACATGCCAA
  • TABLE A″
    Target Example of 
    position Target Sequence
    Human on Comprised within 
    Name chromosome chromosome the locus: SEQ ID NO:
    SH18  5 20634138 CTTACCCCACGTACCACAGACTGT 105
    SH31 14 65874037 TTGTAATGTCTTACAAGGTTTTAA 106
    SH38 10 3983262 CTGGGATGTCTCACGACAGCATGG 107
    SH39 11 104531937 TCCTTCTGTCTTAAGAGATTTATC 108
    SH41  5 18182572 CCTCTCTTAGGTGAGACGGTACAT 109
    SH42  5 20466837 TATATCCCATGTGAGACATGCAGT 110
    SH43 18 37446750 TAAATACGTCTTACATTATTTTGC 111
    SH44  6 147302518 AAGAAATGTCTCACAGAATTTTAC 112
    SH45  8 24854461 CAGATATGTCTTAAAATGTCACTG 113
    SH46 19 12036102 ACCAGATGTCGTGAGACGGGGGAG 114
    SH47  8 25002335 GCAGGCTTATTCACCAGGGTTTAC 115
    SH48 10 101896036 TTGAAATTAGTTACAGGAGGTTAT 116
    SH49 13 68191409 ATAATACAATTTACCTAATCCTAT 117
    SH50  1 47411545 CCCGGCCCCTTTAATCCATCTTAA 118
    SH51 21 30011146 TTGAGCTCACTCACATGGTCTCAG 119
    SH52 12 76131166 CTCCACTGTCTTACCTAATCCAGC 120
    SH70 12 796917 CATGTATGATTTACATCGGTTTGA 121
    SH71  2 231579954 GTTGTATTATTTACCTCAGATGAA 122
    SH72  6 25192217 TTTGGATGCTGTAAAGAATTTCCT 123
    SH73  8 78807830 ATAAAACGACTTACAAGGTCTGAA 124
    SH74 19 29033855 TTCAGATCTCGTACAGGGGATGAC 125
    SH75  8 114771707 CTGCCATAGGGTAACTGAGTCAAT 126
    RIS Cleaved
    position by mega-
    according nucleases
    Close to  to RIS (example)
    a RIS at GRCh37/ RIS described of SEQ
    Name position: hg19 distance in: ID NO: Plasmids
    SH18 20536474 20536474 97664 Aiuti, 2007 127 pCLS5518
    128 pCLS5519
    129 pCLS5520
    130 pCLS5521
    SH31 64841555 65771802 102235 Recchia, 2006 131 pCLS3904
    132 pCLS4076
    SH38 3929865 3939865 43397 Mavilio F, 2006
    SH39 104003035 104465318 66619 Schwartzwaelder,  133 pCLS6038
    2007 134 pCLS6039
    SH41 18180277 18134776 47796 Schwartzwaelder,  135 pCLS5187
    2007 136 pCLS5188
    SH42 20581361 20535860 69023 Schwartzwaelder,  137 pCLS5549
    2007 138 pCLS5550
    SH43 35630950 37378963 67787 Schwartzwaelder,  139 pCLS5594
    2007 140 pCLS5595
    SH44 147201063 147220493 82025 Schwartzwaelder,  141 pCLS5868
    2007 142 pCLS5869
    SH45 24923302 24867385 12924 Mavilio F, 2006
    SH46 11713157 11852157 183945 Mavilio F, 2006
    SH47 24923302 24867385 134950 Mavilio F, 2006
    SH48 101755754 101765764 130272 Mavilio F, 2006
    SH49 65947183 68149182 42227 Schwartzwaelder, 
    2007
    SH50 46928138 47216118 195427 Mavilio F, 2006
    SH51 28929744 30007873 3273 Mavilio F, 2006
    SH52 74339720 76053453 77713 Mavilio F, 2006 143 pCLS5870
    144 pCLS5871
    SH70 708202 837941 41024 Recchia, 2006 145 pCLS5957
    SH71 231351771 231526266 53688 Recchia, 2006 146 pCLS5958
    SH72 25101289 24993310 198907 Recchia, 2006 147 pCLS5959
    SH73 78989339 78939377 131547 Deichmann, 2007 148 pCLS5960
    SH74   33661180 28969340 64515 Deichmann, 2007 149 pCLS5961
    SH75  114711413 114754830 16877 Deichmann, 2007 150 pCLS5962
  • TABLE B
    Dist Dist
    Left Gene Left Right Gene Right
    Name Left Gene1 Description1 Kb1 Right Gene1 Description1 Kb1
    SH3 LY86 MD-1, RP105- 197 RREB1 ras responsive 330
    associated element binding
    protein
    1
    isoform 1
    SH4 MDFIC MyoD family 318 TFEC transcription 606
    inhibitor domain factor
    containing EC isoform b
    protein isoform
    p40
    SH6 C21orf34 hypothetical 675 CXADR coxsackie virus 446
    protein and adenovirus
    LOC388815 receptor
    isoform b precursor
    SH12 LOC728767 hypothetical 41 COL4A1 alpha 1 type IV 302
    protein collagen
    preproprotein
    preproprotein
    SH13 ROBO1 roundabout 1 919 LOC728290 hypothetical 484
    isoform a protein
    SH19 LOC100289420 hypothetical 1106 FAM19A5 family with 208
    protein sequence
    XP_002343824 similarity 19
    (chemokine
    (C-C motif)-
    like), member
    A5 isoform 1
    SH20 KRR1 HIV-1 rev 120 LOC100289143 hypothetical 307
    binding protein 2 protein
    XP_002343241
    SH21 GADL1 glutamate 236 STT3B source of 402
    decarboxylase- immunodominant
    like 1 MHC-associated
    peptides
    SH33 DEFB133 beta-defensin 7 DEFB114 beta-defensin 114 4
    133
    SH7 FANCL Fanconi anemia, 685 LOC730134 similar to 312
    complementation hCG1815165
    group L isoform 2
    SH8 CDH18 cadherin 18, 647 LOC100288118 hypothetical 988
    type 2 protein
    preproprotein XP_002342537
    preproprotein
  • TABLE B′
    Dist Dist
    Left Gene Left Right Gene Right
    Name Left Gene1 Description 1 Kb1 Right Gene1 Description1 Kb1
    SH3 LOC652960 na 56 RREB1 ras responsive 256
    element
    binding protein
    1 isoform 2
    SH4 MDFIC MyoD family 315 LOC100287693 na 162
    inhibitor domain
    containing
    protein isoform
    p40
    SH6 RPS26P5 na 945 RPL39P40 na 433
    SH8 NUP50P3 na 179 LOC728411 na 973
    SH19 LOC100289420 hypothetical 1105 FAM19A5 family with 208
    protein sequence
    XP_002343824 similarity 19
    (chemokine
    (C-C motif)-
    like), member
    A5 isoform
    2
  • TABLE B″
    Dist Dist
    Left Gene Left Right Gene Right
    Name Left Gene1 Description1 Kb1 Right Gene1 Description1 Kb1
    SH18 NUP50P3 na 328 LOC728411 na 825
    SH31 PTBP1P na 127 LOC645431 na 3
    SH38 LOC727894 hypothetical 5 LOC100128356 na 498
    protein
    SH39 DDI1 DDI1, DNA-damage 622 CASP12 na 225
    inducible 1,
    homolog 1
    SH41 RPL36AP21 na 132 RPL32P14 na 858
    SH42 NUP50P3 na 160 LOC728411 na 992
    SH43 RPL7AP66 na 531 RPL17P45 na 277
    SH44 LOC729176 na 177 STXBP5 syntaxin binding 222
    protein 5
    (tomosyn)
    isoform a
    SH45 NEFL neurofilament, 40 DOCK5 dedicator of 187
    light cytokinesis 5
    polypeptide 68 kDa
    SH46 VN2R15P na 9 VN2R21P na 27
    SH47 NEFL neurofilament, 188 DOCK5 dedicator of 39
    light cytokinesis 5
    polypeptide 68 kDa
    SH48 LOC644566 na 18 LOC644573 na 6
    SH49 RPSAP53 na 349 LOC390411 na 214
    SH50 CYP4A11 cytochrome P450, 4 CYP4X1 cytochrome 77
    family 4, P450, family 4,
    subfamily A, subfamily X,
    polypeptide 11 polypeptide 1
    SH51 NCRNA00161 na 98 N6AMT1 N-6 adenine- 233
    specific DNA
    methyltransferase
    1 isoform 1
    SH52 RPL10P13 na 48 LOC100289143 hypothetical 201
    protein
    XP_002343241
    SH70 LOC100049716 na 41 LOC100132369 hypothetical 64
    protein
    SH71 LOC646839 na 141 ITM2C integral 149
    membrane
    protein 2C
    isoform 3
    SH72 LOC100132239 na 38 LOC100129757 na 26
    SH73 LOC100289199 na 878 PKIA cAMP-dependent 620
    protein kinase
    inhibitor alpha
    isoform 7
    SH74 LOC100131694 na 558 LOC100129507 na 184
    SH75 RPL18P7 na 382 TRPS1 zinc finger 1648
    transcription
    factor TRPS1
  • TABLE C
    Dist Dist
    Left Gene Left Right Gene Right
    Name Left Gene2 Description2 Kb2 Right Gene2 Description2 Kb2
    SH3 F13A1 coagulation 533 LOC100288758 hypothetical 378
    factor XIII A1 protein
    subunit XP_002342653
    precursor
    SH4 FOXP2 forkhead box P2 644 TES testin isoform 1 876
    isoform III
    SH6 C21orf34 hypothetical 996 BTG3 B-cell 527
    protein translocation
    LOC388815 gene
    3 isoform b
    isoform a
    SH12 IRS2 insulin receptor 63 COL4A2 alpha 2 type IV 459
    substrate 2 collagen
    preproprotein
    preproprotein
    SH13 ROBO2 roundabout, 2863 GBE1 glucan 982
    axon guidance (1,4-alpha-),
    receptor, branching
    homolog 2 enzyme 1
    isoform ROBO2a
    SH19 TBC1D22A TBC1 domain 1108 FAM19A5 family with 295
    family, member sequence
    22A similarity 19
    (chemokine
    (C-C motif)-
    like), member
    A5 isoform 2
    SH20 GLIPR1 GLI 133 LOC100131830 hypothetical 382
    pathogenesis- protein
    related 1
    precursor
    SH21 TGFBR2 transforming 439 OSBPL10 oxysterol- 532
    growth factor, binding
    beta receptor II protein-like
    isoform A protein 10
    precursor
    SH33 CRISP1 acidic epididymal 99 DEFB113 beta-defensin 113 13
    glycoprotein-like
    1 isoform 2
    precursor
    SH7 VRK2 vaccinia related 767 BCL11A B-cell CLL/ 1526
    kinase 2 isoform 6 lymphoma 11A
    isoform
    3
    SH8 LOC391769 similar to 2830 CDH12 cadherin 12, 1266
    HIStone family type 2
    member (his-72) preproprotein
    preproprotein
  • TABLE C′
    Dist Dist
    Left Gene Left Right Gene Right
    Name Left Gene2 Description2 Kb2 Right Gene2 Description2 Kb2
    SH3 LY86 MD-1, RP105- 196 LOC100288758 hypothetical 376
    associated protein
    XP_002342653
    SH4 FOXP2 forkhead box P2 643 TFEC transcription 600
    isoform III factor
    EC isoform a
    SH6 VDAC2P na 971 CXADR coxsackie virus 446
    and adenovirus
    receptor precursor
    SH8 CDH18 cadherin 18, 646 LOC100288118 hypothetical 987
    type 2 protein
    preproprotein XP_002342537
    preproprotein
    SH19 TBC1D22A TBC1 domain 1107 LOC100128946 hypothetical 614
    family, member protein
    22A
  • TABLE C″
    Dist Dist
    Left Gene Left Right Gene Right
    Name Left Gene2 Description2 Kb2 Right Gene2 Description2 Kb2
    SH18 CDH18 cadherin 18, 794 LOC100288118 hypothetical 839
    type 2 protein
    preproprotein XP_002342537
    preproprotein
    SH31 RPL36AP2 na 137 FUT8 fucosyltransferase 3
    8 isoform c
    SH38 LOC100130652 hypothetical 112 LOC100216001 na 709
    protein
    SH39 PDGFD platelet derived 496 LOC643733 na 242
    growth factor D
    isoform 1
    precursor
    SH41 LOC100133112 na 488 LOC646273 na 1050
    SH42 CDH18 cadherin 18, 627 LOC100288118 hypothetical 1006
    type 2 protein
    preproprotein XP_002342537
    preproprotein
    SH43 LOC647946 na 114 KC6 na 1613
    SH44 C6orf103 hypothetical 165 LOC442266 na 425
    protein
    LOC79747
    SH45 LOC100129717 na 40 GNRH1 gonadotropin- 422
    releasing
    hormone 1
    precursor
    SH46 ZNF69 zinc finger 10 ZNF763 zinc finger 39
    protein 69 protein
    440 like
    SH47 LOC100129717 na 188 GNRH1 gonadotropin- 274
    releasing
    hormone 1
    precursor
    SH48 CPN1 carboxypeptidase 54 ERLIN1 ER lipid raft 13
    N, polypeptide 1 associated 1
    precursor
    SH49 LOC730236 hypothetical 385 OR7E111P na 284
    protein
    SH50 CYP4Z2P na 45 CYP4Z1 cytochrome P450 121
    4Z1
    SH51 C21orf94 na 615 HSPD1P7 na 248
    SH52 LOC100129649 na 135 LOC100131830 hypothetical 276
    protein
    SH70 NINJ2 ninjurin 2 24 WNK1 WNK lysine 65
    deficient protein
    kinase 1
    SH71 HMGB1L3 na 199 GPR55 G protein-coupled 192
    receptor 55
    SH72 NUP50P2 na 50 RPL21P68 na 69
    SH73 PXMP3 peroxin 2 895 FAM164A hypothetical 770
    protein
    LOC51101
    SH74 LOC100132081 na 640 LOC148145 na 422
    SH75 LOC100289099 na 1220 EIF3H eukaryotic 2885
    translation
    initiation factors,
    subunits gamma,
    40 kDa
  • TABLE D
    Dist Dist
    Left Gene Left Right Gene Right
    Name Left Gene3 Description3 Kb3 Right Gene3 Description3 Kb3
    SH3 NRN1 neuritin 845 LOC100288790 hypothetical 417
    precursor protein
    XP_002342654
    SH4 FOXP2 forkhead box P2 644 TES testin isoform 2 900
    isoform II
    SH6 USP25 ubiquitin 1189 C21orf91 early 726
    specific undifferentiated
    peptidase
    25 retina and lens
    isoform
    2
    SH12 MYO16 myosin heavy 642 RAB20 RAB20, member 675
    chain Myr 8 RAS oncogene
    family
    SH13 ROBO2 roundabout, 2863 LOC100289598 hypothetical 4448
    axon guidance protein
    receptor, XP_002342405
    homolog
    2
    isoform ROBO2b
    SH19 CERK ceramide kinase 1543 LOC100128946 hypothetical 616
    protein
    SH20 GLIPR1L2 GLI 209 PHLDA1 pleckstrin 398
    pathogenesis- homology-like
    related 1 like 2 domain, family A,
    member 1
    SH21 RBMS3 RNA binding 1127 ZNF860 zinc finger 859
    motif, single protein 860
    stranded
    interacting
    protein 3
    isoform 1
    SH33 CRISP1 acidic epididymal 99 DEFB110 beta-defensin 110 53
    glycoprotein-like
    1 isoform 1
    precursor
    SH7 VRK2 vaccinia related 767 BCL11A B-cell 1526
    kinase 2 isoform CLL/lymphoma
    2 11A isoform 2
    SH8 LOC391767 similar to TBP- 2851 PRDM9 PR domain 3023
    associated factor containing 9
    11
  • TABLE D′
    Dist Dist
    Left Gene Left Right Gene Right
    Name Left Gene3 Description3 Kb3 Right Gene3 Description3 Kb3
    SH3 LOC643875 na 316 LOC100288790 hypothetical 416
    protein
    XP_002342654
    SH4 RPL36P13 na 1036 TES testin isoform 2 876
    SH6 C21orf34 hypothetical 459 BTG3 B-cell 526
    protein translocation
    LOC388815 gene
    3 isoform a
    isoform b
    SH8 LOC646273 na 1251 GUSBP1 na 1005
    SH19 CERK ceramide kinase 1542 LOC100287247 hypothetical 768
    protein
    XP_002343807
  • TABLE D″
    Dist Dist
    Left Gene Left Right Gene Right
    Name Left Gene3 Description3 Kb3 Right Gene3 Description3 Kb3
    SH18 LOC646273 na 1399 GUSBP1 na 857
    SH31 RPL21P7 na 139 RPL21P8 na 60
    SH38 KLF6 Kruppel-like 155 LOC338588 na 715
    factor 6
    SH39 LOC100190922 na 1031 CASP4 caspase 4 281
    isoform gamma
    precursor
    SH41 LOC391769 similar to 526 CDH18 cadherin 18, 1290
    HIStone family type 2
    member (his-72) preproprotein
    preproprotein
    SH42 LOC646273 na 1232 GUSBP1 na 1024
    SH43 RPL12P40 na 2193 NPM1P1 na 1922
    SH44 RAB32 RAB32, 426 SAMD5 sterile alpha 527
    member RAS motif domain
    oncogene family containing 5
    SH45 LOC100289018 hypothetical 81 KCTD9 potassium 430
    protein channel
    XP_002342868 tetramerisation
    domain
    containing 9
    SH46 VN2R14P na 53 ZNF433 zinc finger 89
    protein 433
    SH47 LOC100289018 hypothetical 229 KCTD9 potassium 283
    protein channel
    XP_002342868 tetramerisation
    domain
    containing 9
    SH48 NCRNA00093 na 177 CHUK conserved helix- 52
    loop-helix
    ubiquitous kinase
    SH49 PCDH9 protocadherin 9 386 OR7E33P na 293
    isoform 1
    precursor
    SH50 LOC100132680 na 45 LOC100132432 na 123
    SH51 NCRNA00113 na 887 LOC391276 na 262
    SH52 KRR1 HIV-1 rev binding 225 PHLDA1 pleckstrin 288
    protein 2 homology-like
    domain, family A,
    member 1
    SH70 B4GALNT3 beta 1,4-N-acetyl- 125 HSN2 hereditary 179
    galactosaminyl- sensory
    transferase- neuropathy, type
    transferase-III II
    SH71 SP100 nuclear antigen 169 LOC100289170 na 232
    Sp100 isoform 2
    SH72 CMAH na 54 LOC100128495 na 80
    SH73 ZFHX4 zinc finger 1028 IL7 interleukin 7 837
    homeodomain 4 precursor
    SH74 LOC642290 na 715 UQCRFS1 ubiquinol- 664
    cytochrome c
    reductase,
    Rieske iron-sulfur
    polypeptide 1
    SH75 CSMD3 CUB and Sushi 322 UTP23 UTP23, small 3007
    multiple domains 3 subunit (SSU)
    isoform 2 processome
    component,
    homolog
  • The locus according to the invention may also correspond to any one of the SH101, SH106, SH107, SH102, SH105, SH103, SH104, SH113, SH109, SH112, SH108, SH110, SH114, SH116, SH111, SH115, SH121, SH120, SH122, SH117, SH118, SH119, SH123, SH126, SH128, SH129, SH124, SH131, SH125, SH127 and SH130 which are described in Tables E and F below.
  • Table E provides the location of the locus within the human genome, a target sequence comprised within the locus, the location of the closest RIS as well as the reference to a publication describing the RIS, the distance between said target and the closest RIS and examples of endonucleases according to the invention that cleave the locus.
  • Table F provides information about the nearest genes that are located immediately upstream (at 5′) and downstream (at 3′) of the locus according to the invention. The distance indicates the distance between the target sequence and the nearest coding sequence of the gene.
  • Locations of loci, targets in this loci and genes are given in Tables E and F according to GRCh36.3/hg19 version of the human genome assembly.
  • TABLE E
    Target
    position
    on
    chromosome
    Human (start; Example of Target Sequence SEQ ID
    Name chromosome V36.3) Comprised within the locus: NO:
    SH101  3 72293606 CCTACACCCTGTAAGATGGCTAGT 151
    SH106 13 103230446 CTAAAATCATGTAAGTTGTATTAT 152
    SH107 13 103240747 TAAACATTTTGTACAGAATCTCAG 153
    SH102  4 143846381 ATGAGATAATGTACAAGGTTTTGT 154
    SH105 12 64610385 CAGGGACTATTTACAAAAGATTGA 155
    SH103  4 143907910 CCAAACCTAGGTAAGAGATATGAA 156
    SH104  7 131856646 TATAGATCAAGTAACAAGTGTAAT 157
    SH113  8 66935276 TTTTACTGTCTTACCTAGTTTTGC 158
    SH109  3 72674929 TCAATCTCACTTACAAAGTTGTGA 159
    SH112  7 127627660 CTAGGATGTAGTACAGGGTGCTAT 160
    SH108  3 173734739 AATATCTCATGTAACACATATTGC 161
    SH110  5 14051421 TTACTCCCATTTACAAGAGCAGAG 162
    SH114 10 11537739 ACCAGACCTTGTAAGTTATACAGA 163
    SH116 21 14663030 ATAAAATAAGTTACAGAGTTACAA 164
    SH111  7 127808719 ACTTCCTGTTTTACAAGGTGTAAT 165
    SH115 12 95084648 CCTGGATATGTTACAACAGAAAGC 166
    SH121  8 8897353 TTTCTCTCAGGTAAAACAGTCCAC 167
    SH120  8 24344273 GTAAGCTATTGTAAGAAATGCAAG 168
    SH122 17 58931643 ATGAGATGATGTACAAAGTCCTAG 169
    SH117  1 223618330 ACTGTATTTTGTAAAGTGTCCCTC 170
    SH118  4 8209666 TCTTCATGTTGTACCTTGTCCCCT 171
    SH119  5 138660535 ATCATCTGAGGTAAAGAGTTCTGA 172
    SH123 19 40227362 GCTCTCTCTGGTACCTGATAGTGA 173
    SH126  2 194307577 ACAAACTCTTTTACGGGATTCAGG 174
    SH128  2 193954229 TTCACATGCTTTACGAAAGTTAGC 175
    SH129  2 194043922 CCTACATTTCGTAAGACATCTATT 176
    SH124  4 159540469 GCAAACTGTGGTACCTAGGCCCGT 177
    SH131  1 201630446 TCGAGCCACTGTACCTAGTTTTGT 178
    SH125 17 10025853 ACAGGATCCAGTAAAGGAGCCGGC 179
    SH127  2 20001992 GCTGTACTATTTACGGTATTCAAT 180
    SH130 16 56151416 ATAAACTTCGGTAAGACATCTCAA 181
    RIS Cleaved
    position by mega-
    according nucleases
    to RIS RIS (examples)
    GRCh36.3/ Distance described of SEQ ID
    Name hg19 (bases) in: NO: Plasmids
    SH101 72478871 185265 Gabriel et al, 2009 182 pCLS7518
    SH106 103311358 80912 Gabriel et al, 2009 183 pCLS7523
    SH107 103311358 70611 Gabriel et al, 2009 184 pCLS7524
    SH102 143708544 137837 Gabriel et al, 2009 185 pCLS7519
    SH105 64560662 49723 Gabriel et al, 2009 186 pCLS7522
    SH103 143708544 199366 Gabriel et al, 2009 187 pCLS7520
    SH104 131765633 91013 Gabriel et al, 2009  188 pCLS7521
    SH113 67019410 84134 Gabriel et al, 2009  189 pCLS7530
    SH109 72478871 196058 Gabriel et al, 2009  190 pCLS7526
    SH112 127698957 71297 Gabriel et al, 2009  191 pCLS7529
    SH108 173720808 13931 Gabriel et al, 2009  192 pCLS7525
    SH110 14197567 146146 Gabriel et al, 2009  193 pCLS7527
    SH114 11694871 157132 Gabriel et al, 2009  194 pCLS7531
    SH116 14814623 151593 Gabriel et al, 2009  195 pCLS7533
    SH111 127698957 109762 Gabriel et al, 2009  196 pCLS7528
    SH115 95131508 46860 Gabriel et al, 2009  197 pCLS7532
    SH121 8837115 60238 Gabriel et al, 2009  198 pCLS7538
    SH120 24200341 143932 Gabriel et al, 2009  199 pCLS7537
    SH122 59056021 124378 Gabriel et al, 2009  200 pCLS7539
    SH117 223700385 82055 Gabriel et al, 2009  201 pCLS7534
    SH118 8250751 41085 Gabriel et al, 2009  202 pCLS7535
    SH119 138751654 91119 Gabriel et al, 2009  203 pCLS7536
    SH123  40144506 82856 Gabriel et al, 2009  204 pCLS7540
    SH126 194148379 159198 Gabriel et al, 2009  205 pCLS7543
    SH128 194148379 194150 Gabriel et al, 2009  206 pCLS7545
    SH129 194148379 104457 Gabriel et al, 2009  207 pCLS7546
    208 pCLS7547
    SH124 159391564 148905 Gabriel et al, 2009  209 pCLS7541
    SH131 201525001 105445 Gabriel et al, 2009  210 pCLS7549
    SH125 9964030 61823 Gabriel et al, 2009 211 pCLS7542
    SH127  20112551 110559 Gabriel et al, 2009  212 pCLS7544
    SH130  56136054 15362 Gabriel et al, 2009  213 pCLS7548
  • TABLE F
    Dist
    Left Dist Right
    Name Left Gene1 Kb1 Right Gene1 Kb1
    SH101 PROK2 380 RYPB 213
    SH106 SLC10A2 713 DAOA 1500
    SH107 SLC10A2 724 DAOA 1500
    SH113 PDE7A 19 DNAJC5B 161
    SH109 RYBP 96 SHQ1 208
    SH112 SND1 100 LEP 41
    SH108 TNFSF10 11 AADACL1 96
    SH110 DNAH5 54 TRIO 146
    SH114 CUGBP2 120 USP6NL 5
    SH116 ABCC13 66 HSPA13 3
    SH111 PRRT4 25 IMPDH1 11
    SH115 LTA4H 151 ELK3 27
    SH121 MFHAS1 110 ERI1 0.37
    SH120 ADAMDEC1 25 ADAM7 10
    SH122 ACE 3 KCNH6 24
    SH126 TMEFF2 1500 SLC39A10 2000
    SH128 TMEFF2 1400 SLC39A10 2100
    SH129 TMEFF2 1300 SLC39A10 2200
    SH124 TMEM144 145 RXFP1 122
    SH131 FMOD 44 PRELP 81
  • The locus according to the invention may also correspond to any one of the SH125, SH127, SH130, SH102, SH105, SH103, SH104, SH117, SH118, SH119 and SH123 which are described in Table G below.
  • Table G provides examples of target sequences located in introns of genes which are mentioned and examples of endonucleases according to the invention that cleave said intronic locus.
  • TABLE G
    Example of Target
    Sequence
    Comprised within   Hit
    Name the locus: position Gene Intron
    SH125 ACAGGATCCAGTAA intronic GAS7 1
    AGGAGCCGGC
    SH127 GCTGTACTATTTACG intronic WDR35 18
    GTATTCAAT
    SH130 ATAAACTTCGGTAAG intronic GPR114 1
    ACATCTCAA
    SH102 ATGAGATAATGTACA intronic INPP4B 2
    AGGTTTTGT
    SH105 CAGGGACTATTTACA intronic HMGA2 3
    AAAGATTGA
    SH103 CCAAACCTAGGTAA intronic INPP4B 1
    GAGATATGAA
    SH104 TATAGATCAAGTAAC intronic PLXNA4 1
    AAGTGTAAT
    SH117 ACTGTATTTTGTAAA intronic DNAH14 76
    GTGTCCCTC
    SH118 TCTTCATGTTGTACC intronic ABLIM2 1
    TTGTCCCCT
    SH119 ATCATCTGAGGTAAA intronic MATR3 5
    GAGTTCTGA
    SH123 GCTCTCTCTGGTAC intronic HPN 3
    CTGATAGTGA
  • The locus according to the invention may also contains any one of the SH11, SH12, SH13, SH17, SH19, SH20, SH21, SH23, SH33, SH34, SH40, SH53, SH54, SH55, SH56, SH57, SH58, SH59, SH60, SH61, SH62, SH65, SH67, SH68 and SH69 which are given in Tables H below.
  • Table H provides target sequences comprised within these loci as well as examples of endonucleases according to the invention that cleave these target sequences.
  • TABLE H
    Cleaved by
    SEQ meganucleases
    ID (examples) of
    Name Sequence NO: SEQ ID NO: Plasmids
    SH11 AGAAGCCCAGGTAAAACAGCCTGG 214 235 pCLS3895
    236 pCLS4664
    SH12 ATAAAACAAGTCACGTTATTTTGG 215 237 pCLS3896
    238 pCLS3915
    239 pCLS6445
    SH13 ATTACACTCTTTAAGTGATTTTAA 216 240 pCLS3897
    241 pCLS6446
    SH17 CTAGGCTGGATTACAGCGGCTTGA 217 242 pCLS3898
    SH19 GCAAAACATTGTAAGACATGCCAA 218 243 pCLS3899
    244 pCLS7278
    245 pCLS7279
    SH20 GCTGGCTGCTTCACATTGGAGAGA 219 246 pCLS3900
    SH21 TAGAAATCTGTTAAAAGAGATGAT 220 247 pCLS3901
    248 pCLS4666
    249 pCLS4667
    SH23 TCAAACCATTGTACTCCAGCCTGG 221 250 pCLS3902
    251 pCLS6447
    SH33 TTTTCATCACTTAAAGTGTTTTAA 222 252 pCLS3905
    253 pCLS4077
    254 pCLS4668
    255 pCLS4669
    SH34 TTTTCCTGTCTTACCAGGTTTTGT 223 256 pCLS3906
    SH40 GTCTTCTGTCTTAAGACATAAAAT 224 257 pCLS5427
    258 pCLS5565
    259 pCLS5566
    SH53 GTAAAATGGATTAAAAGAGGGAAG 225 260 pCLS4773
    SH54 CCAAAACACGTTAAAAAAGTTTAA 226 261 pCLS4774
    SH55 ATAATATTCTGTGACTCATGGCAA 227 262 pCLS4775
    SH56 AGTAGATCTTTTAAAAGATTTTAA 228 263 pCLS4776
    SH57 ATAAAACCACTTAAGACATAGGAA 229 264 pCLS4777
    SH58 ACTTGCTGTCTTAACAGAGAAGAT 230 265 pCLS4778
    SH59 ATGTACCTCTTTAAAACAGATGAA 231 266 pCLS4779
    SH60 CTCTTCTCCTGTGACAGAGTTCTG 232 267 pCLS4780
    SH61 TCCAGCCCCTGTGACAGAGTGAGA 233 268 pCLS5333
    SH62 ACAAAATATTTTAAGGGAGCCAAA 234 269 pCLS5334
    270 pCLS5335
    SH65 CTCACCTGTCTCACAAGGGAGGGA 271 275 pCLS5336
    SH67 CTACTACCATGTGACTGGTTGTAG 272 276 pCLS5337
    SH68 GCTGCACGTTTTACATGAGAGTAA 273 277 pCLS5955
    SH69 TCAGACTTCTTTACCTCATTTGAT 274 278 pCLS5956
  • In a specific embodiment, the locus according to the invention is the SH3 locus. The term “SH3 locus” refers to the region of human chromosome 6 that is located at about 120 kb centromeric to the gene encoding the lymphocyte antigen 86 (see e.g. the world wide web site ncbi.nlm.nih.gov/projects/mapview/maps.cgi?TAXID=9606&CHR=6&MAPS=ideogr%2Ccn tg-r%2CugHs%2Cgenes&BEG=6432845&END=7232845&thmb=on, which shows the 6,430K-7,230K region of chromosome 6), and to homologous regions in other species. More precisely, the SH3 locus extends from position 6850510 to 6853677 of the sequence shown in NC 000006.11. It comprises a sequence of SEQ ID NO: 54.
  • In another specific embodiment, the locus according to the invention is the SH4 locus. The SH4 locus is defined herein as the region of human chromosome 7 that is located at about 320 kb telomeric to MyoD family inhibitor domain containing locus (MDFIC), or to the homologous region in another species (see e.g. the world wide web site ncbi.nlm.nih.gov/projects/mapview/maps.cgi?TAXID=9606&CHR=7&MAPS=ideogr,cntg-r,ugHs,genes[113908811.00%3A114908811.00]&CMD=DN, which shows the 114,660K-115,660K region of chromosome 7). More precisely, the SH4 locus extends from position 114972751 to 114976380 of the sequence shown in NC 000007.13. It comprises a sequence of SEQ ID NO: 55.
  • As used herein, the term “transgene” refers to a sequence encoding a polypeptide. Preferably, the polypeptide encoded by the transgene is either not expressed, or expressed but not biologically active, in the cell, tissue or individual in which the transgene is inserted. Most preferably, the transgene encodes a therapeutic polypeptide useful for the treatment of an individual.
  • In the frame of the present invention, the individual may be a human or non-human animal. The individual is preferably a human. Alternatively, the individual can be a non-human animal, preferably a vertebrate and/or a mammalian animal such as e.g. a mouse, a rat, a rabbit, a Chinese hamster, a Guinea pig or a monkey. The cells and tissues according to the invention are preferably derived from such human or non-human animals.
  • Endonucleases According to the Invention that are Derived from I-CreI
  • The variant endonuclease according to the invention can for example be derived:
      • either from the wild-type I-CreI meganuclease, which is a homodimeric protein comprising two monomers, each of these monomers comprising a sequence of SEQ ID NO: 1 or the sequence shown in shown SwissProt Accession n ° P05725;
      • or from a I-CreI meganuclease comprising two monomers, each of these monomers comprising a sequence of SEQ ID NO: 42 Such a I-CreI meganuclease, which recognizes the wild-type target sequence, has been shown to be suitable for engineering endonucleases with novel specificities.
  • Therefore, the invention pertains to a dimeric I-CreI protein comprising or consisting of two monomers, each monomer comprising or consisting of a sequence at least 80%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% identical to SEQ ID NO: 1 or to SEQ ID NO: 42, wherein said dimeric I-CreI protein is capable of cleaving a target sequence located within a safe harbor locus.
  • Preferably, the target sequence neither comprises nor consists of a sequence of SEQ ID NO: 4.
  • Most preferably, the dimeric I-CreI protein according to the invention is a heterodimeric protein.
  • By a protein having a sequence at least, for example, 95% “identical” to a query sequence of the present invention, it is intended that the sequence of the protein is identical to the query sequence except that the sequence may include up to five nucleotide mutations per each 100 amino acids of the query sequence. In other words, to obtain a protein having a sequence at least 95% identical to a query sequence, up to 5% (5 of 100) of the amino acids of the sequence may be inserted, deleted, or replaced with another nucleotide. The <<needle>> program, which uses the Needleman-Wunsch global alignment algorithm (Needleman and Wunsch, 1970 J. Mol. Biol. 48:443-453) to find the optimum alignment (including gaps) of two sequences when considering their entire length, may for example be used. The needle program is for example available on the ebi.ac.uk world wide web site. The percentage of identity in accordance with the invention can thus be calculated using the EMBOSS::needle (global) program with a “Gap Open” parameter equal to 10.0, a “Gap Extend” parameter equal to 0.5, and a Blosum62 matrix.
  • Each monomer of the dimeric I-CreI protein according to the invention may for example comprise at least, at most or about 2, 5, 8, 10, 12, 15, 18, 20 or 25 mutations compared with the sequence of a wild-type monomer (SEQ ID NO: 1) or with a monomer of SEQ ID NO: 42. In other terms, the monomer according to the invention comprises a sequence that differs from SEQ ID NO: 1 or SEQ ID NO: 42 by at least, at most or about 2, 5, 8, 10, 12, 15, 18, 20, 25 or 30 mutations.
  • In the frame of the present invention, the mutation preferably corresponds to a substitution of one amino acid with another amino acid. Therefore, a preferred embodiment according to the invention is directed to a dimeric I-CreI protein comprising or consisting of two monomers comprising a sequence at least 80%, identical to SEQ ID NO: 1 or SEQ ID NO: 42, wherein said sequence only differs from SEQ ID NO: 1 or SEQ ID NO: 42 by the presence of amino acid substitutions.
  • The monomers of the dimeric I-CreI protein according to the invention are preferably derived from monomers comprising or consisting of the sequence of SEQ ID NO: 42.
  • The mutations are preferably located at positions of the I-CreI sequence that are involved in recognition of the target sequence. Indeed, introducing such mutations allow designing meganucleases with novel specificities.
  • In addition to such mutations, the monomers may also have mutations corresponding to:
      • mutations that improve the binding and/or the cleavage properties of the protein towards the target site, such as e.g. G195, G19A, F54L, S79G, E80K, F87L, V105A and/or I132V (see for example WO 2008/152524); and/or
      • mutations leading to the obtention of an obligate heterodimer (see for example WO 2008/093249 and Fajardo-Sanchez et al., Nucleic Acids Res. 2008 36:2163-73); and/or
      • mutations suitable for the generation of a fusion protein such as, e.g., the deletion of the five most N-terminal amino acid residues of SEQ ID NO: 1 in the C-terminal monomer of a fusion protein; and/or
      • a mutation consisting of the insertion of an alanine between the first and the second residue of SEQ ID NO: 1, as is the case in a monomer of SEQ ID NO: 42.
  • In addition to the sequence homologous to SEQ ID NO: 1 or SEQ ID NO: 42, the monomers of the protein according to the invention may comprise one or more amino acids added at the NH2 terminus and/or COOH terminus of the sequence, such as a Tag useful in purification of the protein, a propeptide and/or a nuclear localization signal. In particular, the monomers of the protein according to the invention may comprise AAD amino acids added at the COOH terminus of the sequence of SEQ ID NO: 1, as is the case in a monomer of SEQ ID NO: 42.
  • In the present specification, the mutations are indicated by the position on SEQ ID NO: 1 followed by the nature of the amino acid replacing the amino acid located at this position in SEQ ID NO: 1. For example, a monomer comprising a 44A mutation refers to a I-CreI monomer in which the amino acid at position 44 of SEQ ID NO: 1 (i.e. a glutamine, Q) is replaced with an alanine (A). Thus this monomer differs from the wild-type I-CreI monomer of SEQ ID NO: 1 by at least the following amino acid substitution: Q44A. As explained hereabove, the I-CreI monomer of SEQ ID NO: 42 comprises some additional amino acid residues compared to the I-CreI monomer of SEQ ID NO: 1 (see FIG. 11). Therefore, on SEQ ID NO: 42, the 44A mutation corresponds to a replacement of the glutamine at position 45 of SEQ ID NO: 42 with an alanine.
  • For the purpose of illustration, a monomer comprising 44A 54L 64A 70Q 75N 158R 162A mutations may for example have the sequence of SEQ ID NO: 57 (when this monomer is directly derived from a I-CreI monomer of SEQ ID NO: 1) or the sequence of SEQ ID NO: 58 (when this monomer is directly derived from a I-CreI monomer of SEQ ID NO: 42). FIG. 12 shows an alignment between two such monomers, and indicates the position of the 44A 54L 64A 70Q 75N 158R and 162A mutations on these monomers.
  • Examples of dimeric I-CreI proteins according to the invention, capable of cleaving target sequences located in the SH3, SH4 or SH6 locus, are further described below.
  • Dimeric I-CreI Protein According to the Invention Capable of Cleaving the SH3 Locus
  • In a preferred embodiment, the target sequence is located within the SH3 locus (defined hereabove). The target sequence located within SH3 may for example comprise or consist of SEQ ID NO: 2, or of nucleotides 2 to 23 of SEQ ID NO: 2. Example 1 discloses several examples of heterodimeric I-CreI proteins according to the invention capable of cleaving such a target sequence. In addition, methods for constructing other such proteins are well-known in the art and include e.g. those described in PCT applications WO 2006/097784, WO 2006/097853 and WO 2009019614, and in Arnould et al. (J. Mol. Biol., 2006, 355:443-458).
  • The monomers of such a dimeric protein preferably comprise at least one, preferably at least 3, 4, 5 or 6, amino acid substitutions located at a position selected from the group consisting of positions 4, 24, 26, 28, 30, 32, 33, 38, 44, 50, 54, 57, 64, 66, 70, 71, 75, 77, 81, 86, 92, 105, 142, 151, 154, 158 and 162 of SEQ ID NO: 1, preferably positions 4, 30, 38, 44, 50, 54, 57, 64, 66, 70, 71, 75, 77, 81, 86, 92, 105, 142, 151, 154, 158 and 162 of SEQ ID NO: 1. Said substitutions may for example be selected from the following substitutions: 4E, 30G, 38R, 44A, 50R, 54L, 57E, 64A, 66C, 70Q, 70D, 71 R, 75N, 75Y, 77V, 81T, 86D, 92R, 105A, 142R, 151A, 154G, 158R, 158W and 162A. The dimeric protein may optionally comprise a mutation at position 1, however, such a mutation has no influence on cleavage activity or on cleavage specificity.
  • Such dimeric I-CreI proteins may for example comprise or consist of:
      • a first monomer comprising at least one amino acid substitution compared to SEQ ID NO: 1, wherein said at least one amino acid substitution is located at a position selected from the group consisting of positions 30, 38, 50, 70, 75, 81, 86, 142 and 154 of SEQ ID NO: 1. Preferably, said first monomer comprises substitutions at positions 30, 38, 70 and 75 of SEQ ID NO: 1. Most preferably, said substitutions are selected from the following substitutions: 30G, 38R, 50R, 70D, 75N, 81T, 86D, 142R and 154G. Such a monomer may for example comprise at least 4, 5 or 6 mutations compared to SEQ ID NO: 1, and/or at most 4, 5, 6, 8, 10, 12 or 15 amino acid mutations compared to SEQ ID NO: 1; and
      • a second monomer comprising at least one amino acid substitution compared to SEQ ID NO: 1, wherein said at least one amino acid substitution is located at a position selected from the group consisting of positions 4, 44, 54, 57, 64, 66, 70, 71, 75, 77, 92, 105, 151, 158 and 162 of SEQ ID NO: 1. Preferably, said second monomer comprises substitutions at positions 44, 54, 70 and 75 of SEQ ID NO: 1. Most preferably, said substitutions are selected from the following substitutions: 4E, 44A, 54L, 57E, 64A, 66C, 70Q, 71R, 75N, 75Y, 77V, 92R, 105A, 151A 158R, 158W and 162A. Such a monomer may for example comprise at least 4, 5 or 6 mutations compared to SEQ ID NO: 1, and/or at most 4, 6, 8, 10, 12 or 15 amino acid mutations compared to SEQ ID NO: 1.
  • In a specific embodiment, the dimeric I-CreI protein according the invention comprises or consists of:
      • a) a first monomer comprising 30G 38R 70D 75N 86D mutations;
      • b) a second monomer selected from the group consisting of:
        • i. a monomer comprising 44A 54L 64A 70Q 75N 158R 162A mutations;
        • ii. a monomer comprising 44A 54L 70Q 75Y 92R 158R 162A mutations;
        • iii. a monomer comprising 4E 44A 54L 64A 70Q 75N 158R 162A mutations;
        • iv. a monomer comprising 44A 54L 64A 70Q 75N 158W 162A mutations;
        • v. a monomer comprising 44A 54L 70Q 75N mutations;
        • vi. a monomer comprising 44A 54L 57E 70Q 75N 158R 162A mutations; and
        • vii. a monomer comprising 44V 54L 70Q 75N 77V mutations;
  • In another specific embodiment, the dimeric I-CreI protein according the invention comprises or consists of:
      • a) a first monomer comprising 30G 38R 70D 75N 81T 154G mutations;
      • b) a second monomer selected from the group consisting of:
        • i. a monomer comprising 44A 54L 70Q 75N 105A 158R 162A mutations;
        • ii. a monomer comprising 44A 54L 64A 70Q 75N 158R 162A mutations;
        • iii. a monomer comprising 4E 44A 54L 64A 70Q 75N 158R 162A mutations;
        • iv. a monomer comprising 44A 54L 64A 70Q 75N 158W 162A mutations;
        • v. a monomer comprising 44A 54L 70Q 75N mutations; and
        • vi. a monomer comprising 44V 54L 70Q 75N 77V mutations;
  • In still another specific embodiment, the dimeric I-CreI protein according the invention comprises or consists of:
      • a) a first monomer comprising 30G 38R, 50R 70D 75N 142R mutations;
      • b) a second monomer selected from the group consisting of:
        • i. a monomer comprising 44A 54L 70Q 75N 105A 158R 162A mutations;
        • ii. a monomer comprising 44A 54L 64A 70Q 75N 158R 162A mutations;
        • iii. a monomer comprising 44A 54L 70Q 75Y 92R 158R 162A mutations;
        • iv. a monomer comprising 4E 44A 54L 64A 70Q 75N 158R 162A mutations;
        • v. a monomer comprising 44A 54L 64A 70Q 75N 158W 162A mutations;
        • vi. a monomer comprising 44A 54L 66C 70Q 71R 75N 151A 158R 162A mutations;
        • vii. a monomer comprising 44A 54L 70Q 75N mutations;
        • viii. a monomer comprising 44A 54L 57E 70Q 75N 158R 162A mutations; and
        • ix. a monomer comprising 44V 54L 70Q 75N 77V mutations.
  • The monomers of the dimeric I-CreI protein may also comprise additional mutations, for example allowing the obtention of an obligate heterodimer. Such mutations are known to the skilled in the art and include those described in Fajardo-Sanchez et al. (Nucleic Acids Res. 2008 36:2163-73).
  • In a specific embodiment, the above monomers are directly derived from a monomer of SEQ ID NO: 42, and differ from the sequence of SEQ ID NO: 42 only by the presence of the indicated mutations.
  • Dimeric I-CreI Protein According to the Invention Capable of Cleaving the SH4 Locus
  • In a preferred embodiment, the target sequence is located within the SH4 locus (defined hereabove). The target sequence located within SH4 may for example comprise or consist of SEQ ID NO: 3, or of nucleotides 2 to 23 of SEQ ID NO: 3. Example 2 discloses several examples of dimeric I-CreI proteins according to the invention capable of cleaving such a target sequence.
  • The monomers of such a dimeric protein preferably comprise at least one, preferably at least 3, 4, 5 or 6, amino acid substitutions located at a position selected from the group consisting of positions 24, 44, 68, 70, 75 and 77 of SEQ ID NO: 1. Said substitutions may for example be selected from the following substitutions: 24V, 44R, 44Y, 68Y, 68A, 70S, 70D, 75Y, 75N, 77R, 77N and 77V.
  • Such dimeric I-CreI proteins may for example comprise or consist of:
      • a first monomer comprising at least one amino acid substitution compared to SEQ ID NO: 1, wherein said at least one amino acid substitution is located at a position selected from the group consisting of positions 24, 44, 68, 70, 75 and 77 of SEQ ID NO: 1. Preferably, the first monomer comprises substitutions at positions 24, 70, 75 and 77 of SEQ ID NO: 1. Most preferably, said substitutions are selected from the following substitutions: 24V, 44R, 68Y, 68A, 70D, 70S, 75Y, 75N, 77N and 77R. Such a monomer may for example comprise at least 4, 5 or 6 mutations compared to SEQ ID NO: 1, and/or at most 4, 5, 6, 8, 10, 12 or 15 amino acid mutations compared to SEQ ID NO: 1; and
      • a second monomer comprising at least one amino acid substitution compared to SEQ ID NO: 1, wherein said at least one amino acid substitution is located at a position selected from the group consisting of positions 24, 44, 70 and 77 of SEQ ID NO: 1. Preferably, the second monomer comprises substitutions at positions 24, 44 and 70 of SEQ ID NO: 1. Most preferably, said substitutions are selected from the following substitutions: 24V, 44Y, 70S and 77V. Such a monomer may for example comprise at least 3 or 4 mutations compared to SEQ ID NO: 1, and/or at most 3, 4, 6, 8, 10, 12 or 15 amino acid mutations compared to SEQ ID NO: 1.
  • In a specific embodiment, the dimeric I-CreI protein according the invention comprises or consists of:
      • a) a first monomer selected from the group consisting of:
        • i. a monomer comprising 24V 44R 68Y 70S 75Y 77N mutations;
        • ii. a monomer comprising 24V 68A 70S 75N 77R mutations; and
        • iii. a monomer comprising 24V 70D 75N 77R mutations;
      • b) a second monomer selected from the group consisting of:
        • i. a monomer comprising 24V 44Y 70S mutations; and
        • ii. a monomer comprising 24V 44Y 70S 77V mutations.
  • The monomers of the dimeric I-CreI protein may also comprise additional mutations, for example allowing the obtention of an obligate heterodimer. Such mutations are known to the skilled in the art and include those described in Fajardo-Sanchez et al. (Nucleic Acids Res. 2008 36:2163-73).
  • In a specific embodiment, the above monomers are directly derived from a monomer of SEQ ID NO: 42, and differ from the sequence of SEQ ID NO: 42 only by the presence of the indicated mutations.
  • Dimeric I-CreI Protein According to the Invention Capable of Cleaving the SH6 Locus
  • In a preferred embodiment, the target sequence is located within the SH6 locus (defined hereabove). The target sequence located within SH6 may for example comprise or consist of SEQ ID NO: 59, or of nucleotides 2 to 23 of SEQ ID NO: 59. Example 5 discloses several examples of dimeric I-CreI proteins according to the invention capable of cleaving such a target sequence.
  • The monomers of such a dimeric protein preferably comprise at least one, preferably at least 3, 4, 5 or 6, amino acid substitutions located at a position selected from the group consisting of positions 7, 24, 27, 28, 34, 40, 44, 68, 70, 75, 77, 81, 85, 96, 99, 103, 108, 111, 121, 132, 144 and 160 of SEQ ID NO: 1. Said substitutions may for example be selected from the following substitutions: 7R, 24F, 27V, 28Q, 34R, 40R, 44A, 44K, 68T, 70L, 70G, 70S, 75N, 77V, 81T, 81V, 85R, 96R, 99R, 103T, 103S, 108V, 111H, 121E, 132V, 144S, 160R and 160E.
  • Such dimeric I-CreI proteins may for example comprise or consist of:
      • a first monomer comprising at least one amino acid substitution compared to SEQ ID NO: 1, wherein said at least one amino acid substitution is located at a position selected from the group consisting of positions 7, 24, 27, 28, 34, 40, 44, 70, 75, 77, 81, 85, 96, 99, 103, 108, 111, 121, 132, 144 and 160 of SEQ ID NO: 1. Preferably, the first monomer comprises substitutions at positions 28, 40, 44, 70 and 75 of SEQ ID NO: 1. Most preferably, said substitutions are selected from the following substitutions: 7R, 24F, 27V, 28Q, 34R, 40R, 44A, 70L, 75N, 77V, 81T, 81V, 85R, 96R, 99R, 103T, 103S, 108V, 111H, 121E, 132V, 144S and 160R et 160E. Such a monomer may for example comprise at least 5 or 6 mutations compared to SEQ ID NO: 1, and/or at most 5, 6, 8, 10, 12, 15 or 20 amino acid mutations compared to SEQ ID NO: 1; and
      • a second monomer comprising at least one amino acid substitution compared to SEQ ID NO: 1, wherein said at least one amino acid substitution is located at a position selected from the group consisting of positions 44, 68, 70 and 75 of SEQ ID NO: 1. Preferably, the second monomer comprises substitutions at positions 44, 70 and 75 of SEQ ID NO: 1. Most preferably, said substitutions are selected from the following substitutions: 44K, 68T, 70G, 70S and 75N. Such a monomer may for example comprise at least 3 or 4 mutations compared to SEQ ID NO: 1, and/or at most 3, 4, 6, 8, 10, 12 or 15 amino acid mutations compared to SEQ ID NO: 1.
  • In a specific embodiment, the dimeric I-CreI protein according the invention comprises or consists of:
      • a) a first monomer comprising 44K 68T 70G 75N mutations; and
      • b) a second monomer selected from the group consisting of:
        • i. a monomer comprising 28Q 40R 44A 70L 75N 96R 111H 144S mutations;
        • ii. a monomer comprising 7R 28Q 40R 44A 70L 75N 85R 103T mutations;
        • iii. a monomer comprising 28Q 40R 44A 70L 75N 103S mutations;
        • iv. a monomer comprising 24F 27V 28Q 40R 44A 70L 75N 99R mutations;
        • v. a monomer comprising 7R 28Q 40R 44A 70L 75N 81T mutations;
        • vi. a monomer comprising 7R 28Q 40R 44A 70L 75N 77V mutations;
        • vii. a monomer comprising 7R 28Q 40R 44A 70L 75N 103T 121E 132V 160R mutations;
        • viii. a monomer comprising 28Q 40R 44A 70L 75N mutations;
        • ix. a monomer comprising 7R 28Q 40R 44A 70L 75N 103T mutations; and
        • x. a monomer comprising 28Q 34R, 40R 44A 70L 75N 81V 103T 108V 160E mutations.
  • In another specific embodiment, the dimeric I-CreI protein according the invention comprises or consists of:
      • a) a first monomer comprising 44K 70S 75N mutations; and
      • b) a second monomer selected from the group consisting of:
        • i. a monomer comprising 28Q 40R 44A 70L 75N 96R 111H 144S mutations;
        • ii. a monomer comprising 7R 28Q 40R 44A 70L 75N 85R 103T mutations;
        • iii. a monomer comprising 28Q 40R 44A 70L 75N 103S mutations;
        • iv. a monomer comprising 24F 27V 28Q 40R 44A 70L 75N 99R mutations;
        • v. a monomer comprising 7R 28Q 40R 44A 70L 75N 81T mutations;
        • vi. a monomer comprising 7R 28Q 40R 44A 70L 75N 103T 121E 132V 160R mutations;
        • vii. a monomer comprising 7R 28Q 40R 44A 70L 75N 103T mutations; and
        • viii. a monomer comprising 28Q 34R, 40R 44A 70L 75N 81V 103T 108V 160E mutations.
  • The monomers of the dimeric I-CreI protein may also comprise additional mutations, for example allowing the obtention of an obligate heterodimer. Such mutations are known to the skilled in the art and include those described in Fajardo-Sanchez et al. (Nucleic Acids Res. 2008 36:2163-73).
  • In a specific embodiment, the above monomers are directly derived from a monomer of SEQ ID NO: 42, and differ from the sequence of SEQ ID NO: 42 only by the presence of the indicated mutations.
  • Fusion Proteins According to the Invention
  • Fusion proteins comprising the two monomers of a dimeric I-CreI protein fused together and retaining the biological activity of the parent dimeric I-CreI protein can be constructed (Grizot et al. NAR 2009 37:5405; Li et al. Nucleic Acids Res. 2009 37:1650-62; Epinat et al. Nucleic Acids Res. 2003 31:2952-62). Such fusion proteins are commonly referred to as “single-chain meganucleases”.
  • Therefore, the invention further relates to a fusion protein comprising the two monomers of the dimeric I-CreI protein as defined hereabove, or biologically active fragments of such monomers. In such a fusion protein, the first and second monomers of a dimeric I-CreI protein as defined hereabove are fused together and are optionally connected to each other by a linker such as a peptidic linker. The linker may for example comprise or consist of SEQ ID NO: 43 or SEQ ID NO: 326.
  • In the frame of the present invention, it is understood that such a fusion protein according to the invention is capable of cleaving a target sequence according to the invention, i.e., it is capable of cleaving the same target sequence as the dimeric I-CreI protein from which it is derived. The single chain meganuclease of the present invention further comprises obligate heterodimer mutations as described above so as to obtain single chain obligate heterodimer meganuclease variants.
  • In the first version of I-CreI single chain (Epinat et al. NAR 2003 3:2952-2962; WO 03/078619), the N-terminal monomer of the single-chain meganuclease consisted essentially of positions 1 to 93 of I-CreI amino acid sequence whereas the C-terminal (positions 8 to 163 of I-CreI amino acid sequence) was a nearly complete I-CreI monomer. More recently, a new way to design a single chain molecule derived from the I-CreI homodimeric meganuclease consisted in two nearly complete C-terminal and N-terminal I-CreI monomers (see, e.g. WO 2009/095793). This design greatly decreases off-site cleavage and toxicity while enhancing efficacy. The structure and stability of this single-chain molecule are very similar to those of the dimeric variants and this molecule appears to be monomeric in solution. In all respects, this single-chain molecule performs as well as I-SceI considered to be gold standard in terms of specificity. These properties place this new generation of meganucleases among the best molecular scissors available for genome surgery strategies and should facilitate gene correction therapy for monogenetic diseases, such as for example severe combined immunodeficiency (SCID), while potentially avoiding the deleterious effects of previous gene therapy approaches.
  • In addition to the mutations described hereabove, additional mutations may be introduced into the sequence of each of the two monomers of the fusion protein. For example, the C-terminal monomer may comprise the K7E and K96E mutations, and the N-terminal monomer may comprise the E8K, E61 R and G19S mutations.
  • Examples 1, 2 and 5 disclose several examples of such fusion proteins according to the invention.
  • In a specific embodiment, the fusion protein according to the invention comprises or consists of a sequence at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to any one of SEQ ID Nos. 25-40 and 76-96, or to a fragment of at least 50, 100, 150 or 200 amino acids thereof.
  • Nucleic Acids, Vectors and Combinations According to the Invention
  • When inserting a transgene into the genome of a cell, tissue or animal, the endonuclease according to the invention is preferably introduced to said cell, tissue or animal as a nucleic acid molecule rather than as a protein.
  • Therefore, the invention pertains to a nucleic acid encoding the endonuclease according to the invention, e.g. encoding a dimeric I-CreI protein or a fusion protein described hereabove. When the endonuclease is a dimeric I-CreI protein, said nucleic acid comprises at least two coding sequences, one for each monomer. When the endonuclease is a fusion protein, said nucleic acid comprises at least one coding sequence. The endonuclease protein can be combined with a variety of cell-penetrating peptide leading to a recombinant protein; such combined molecules are able to enter target cells at much higher levels of efficiency than the endonuclease alone. These cell-penetrating peptides were developed by Diatos S. A. (WO01/64738; WO05/016960; WO03/018636; WO05/018650; WO07/069,068). The applicant has previously shown that endonuclease cell-penetrating peptides combinations can enter target cells efficiently and that the internalized endonuclease can act upon the target cell genome so as to generate a DSB and in turn stimulate a homologous recombination event. The applicant has shown that the complex three dimensional structure of the endonuclease is not affected by the presence of the cell-penetrating peptide and that the all important specificity of the endonuclease also remains unaffected (data not shown).
  • Another aspect of the invention is a vector comprising such a nucleic acid according to the invention. By “vector” is meant a nucleic acid molecule capable of transporting another nucleic acid to which it has been linked.
  • Vectors which can be used in the present invention includes but is not limited to viral vectors, plasmids and YACs, which may consist of chromosomal, non chromosomal, semisynthetic or synthetic nucleic acids. Preferred vectors are those capable of autonomous replication (episomal vector) and/or expression of nucleic acids to which they are linked (expression vectors). Large numbers of suitable vectors are known to those of skill in the art and commercially available.
  • In a preferred embodiment, the vector is a viral vector such as e.g. a vector derived from a retrovirus, an adenovirus, a parvovirus (e.g. an adeno-associated viruses), a coronavirus, a negative strand RNA virus (e.g. an orthomyxovirus such as influenza virus, a rhabdovirus such as rabies and vesicular stomatitis virus, a paramyxovirus such as measles and Sendai virus), a positive strand RNA virus such as picornavirus and alphavirus, or a double-stranded DNA virus such as adenovirus, herpesvirus (e.g. Herpes Simplex virus types 1 and 2, Epstein-Barr virus, cytomegalovirus) and poxvirus (e.g. vaccinia, fowlpox and canarypox). Preferred vectors include lentiviral vectors, and particularly self-inactivacting lentiviral vectors.
  • In addition to the sequence coding for the endonuclease according to the invention, the vector can also comprise elements such as:
      • transcriptional and translational control elements such as promoters, enhancers, polyadenylation sites, terminations signals, introns, etc.;
      • a multiple cloning site;
      • a replication origin;
      • selection markers;
      • a transgene; and/or
      • a targeting construct comprising sequences sharing homologies with the region surrounding the genomic target site as defined herein.
  • In a preferred embodiment, said vector is an “expression vector”, i.e. a vector in which at least one coding sequence is operatively linked to transcriptional and translational control elements. In the frame of this embodiment, the nucleic acid encoding the endonuclease according to the invention (e.g. encoding the dimeric I-CreI protein or the fusion protein described hereabove) is operatively linked to transcriptional and translational control elements.
  • In a preferred embodiment, the vector according to the invention comprises a targeting construct comprising a transgene and two sequences homologous to the genomic sequence flanking the target sequence as defined herein (e.g. the target sequence of SEQ ID NO: 2 or 3). The genomic sequences flanking the target sequence are preferably immediately adjacent to the target site.
  • Such targeting constructs are well-known to the skilled in the art. For insertion of a transgene, such constructs typically comprise a first sequence that is homologous to the upstream (5′) genomic sequence flanking the target sequence, the transgene to be inserted, and a second fragment that is homologous to the downstream (3′) genomic sequence flanking the target sequence.
  • By “homologous” is intended a sequence with enough identity to another one to lead to a homologous recombination between sequences, more particularly having at least 95% identity, preferably 97% identity and more preferably 99% identity to each other.
  • Preferably, homologous sequences of at least 50 bp, preferably more than 100 bp and more preferably more than 200 bp are used. Therefore, the targeting DNA construct is preferably from 200 pb to 6000 pb, more preferably from 1000 pb to 2000 pb. Indeed, shared DNA homologies are located in regions flanking upstream and downstream the site of the break and the DNA sequence to be introduced should be located between the two arms.
  • The targeting construct may also comprise a positive selection marker between the two homology arms and eventually a negative selection marker upstream of the first homology arm or downstream of the second homology arm. The marker(s) allow(s) the selection of cells having inserted the sequence of interest by homologous recombination at the target site.
  • Methods for constructing targeting constructs suitable for inserting a transgene into the SH3 or SH4 locus are given in Example 4.
  • The nucleic acid encoding the endonuclease according to the invention and the targeting construct can also be located on two separate vectors. Therefore, the invention also pertains to a combination of two vectors, namely:
      • an expression vector according the invention; and
      • a vector comprising a targeting construct comprising a transgene and two sequences homologous to the genomic sequence of the target sequence according to the invention.
  • Pharmaceutical Uses According to the Invention
  • The vectors and combinations described hereabove can for example be used as a medicament. In particular, these vectors and combinations can be used in gene therapy.
  • Therefore, the invention relates to a vector or combination according to the invention for use as a medicament. In such vectors and combinations, the transgene encodes a therapeutic polypeptide.
  • In particular, diseases that may be treated by gene therapy using the vectors and combinations according to the invention include but are not limited to X-SCID, SCID, epidermolysis bullosa, leber amaurosis, hemophilia, thalassemia, fanconi anemia and muscular dystrophy.
  • In these diseases, the transgene encodes the following therapeutic polypeptides, respectively: IL2RG, GI7A1, Rp 65, Blood factors VIII and IX, haemoglobin A and B, Fanc-A, Fanc-C (or other Fanconi Anemia related genes), Dystrophine.
  • The invention further relates to a pharmaceutical composition comprising the vectors and combinations according to the invention and a pharmaceutically active carrier.
  • The invention also relates to a method of treating an individual by gene therapy comprising administering an effective amount of a vector or combination according to the invention to an individual in need thereof.
  • By “effective amount” is meant an amount sufficient to achieve insertion of the transgene into the genome of the individual to be treated. Such concentrations can be routinely determined by those of skilled in the art.
  • By “subject in need thereof” is meant an individual suffering from or susceptible of suffering from a genetic disease that can be treated or prevented by insertion of the transgene. The individuals to be treated in the frame of the invention are preferably human beings.
  • Non Pharmaceutical Uses According to the Invention
  • The vectors and combinations described hereabove not only find use in gene therapy but also in non pharmaceutical uses such as, e.g., production of animal models and production of recombinant cell lines expressing a protein of interest.
  • Therefore, the invention relates to:
      • the use of an endonuclease, nucleic acid, expression vector or combination according to the invention for inserting a transgene into the genome of a cell, tissue or non-human animal, wherein said use is not therapeutic.
      • a method of inserting a transgene into the genome of a cell, tissue or non-human animal, comprising the step of bringing said cell, tissue or non-human animal in contact with an endonuclease, nucleic acid, expression vector or combination according to the invention, thereby inserting said transgene into said genome.
  • In a preferred embodiment, the above use or method aims at inserting a transgene encoding a protein of interest into the genome of a cell order to obtain a recombinant cell line for protein production. Suitable cells for constructing recombinant cell lines for protein production include but are not limited to human (e.g. PER.C6 or HEK), Chinese Ovary hamster (CHO) and mouse (NSE0) cells.
  • In another preferred embodiment, the above use aims at making a non-human animal model of a hereditary disorder.
  • The invention is also directed to a non-human transgenic animal comprising a nucleic acid, an expression vector or a combination according to the invention in its genome.
  • All references cited herein, including journal articles or abstracts, published patent applications, issued patents or any other references, are entirely incorporated by reference herein, including all data, tables, figures and text presented in the cited references.
  • The invention will be further evaluated in view of the following examples and figures.
  • BRIEF DESCRIPTION OF THE FIGURES
  • FIG. 1 represents target sequences of meganucleases described in Example 1.
  • FIGS. 2 and 3 represent SCOH SH3 meganucleases vs. I-SceI and SCOH-RAG DNA dose response in CHO.
  • FIG. 4 represents target sequences of meganucleases described in Example 2.
  • FIGS. 5 and 6 represent SCOH SH4 meganucleases vs. I-SceI and SCOH-RAG DNA dose response in CHO.
  • FIG. 7 represents a scheme of the mechanism leading to the generation of small deletions and insertions (InDel) during repair of double-strand break by non homologous end-joining (NHEJ).
  • FIG. 8 represents the insertion sites upon cleavage with SH3 or SH4 meganucleases.
  • FIG. 9 represents target sequences of meganucleases described in Example 5.
  • FIG. 10 represents SCOH SH6 meganucleases vs. I-SceI and SCOH-RAG DNA dose response in CHO.
  • FIG. 11 represents a sequence alignment between a I-CreI monomer of SEQ ID NO: 1 and a I-CreI monomer of SEQ ID NO: 42.
  • FIG. 12 represents a sequence alignment between a I-CreI monomer of SEQ ID NO: 1 and two I-CreI monomers comprising 44A 54L 64A 70Q 75N 158R and 162A mutations. The first one (SEQ ID NO: 57) is directly derived from SEQ ID NO: 1 and the second one (SEQ ID NO: 58) is directly derived from SEQ ID NO: 42.
  • FIGS. 13 to 17 illustrate examples 6 to 9.
  • BRIEF DESCRIPTION OF THE SEQUENCES
  • SEQ ID NO: 1 shows the amino acid sequence of a wild-type I-CreI monomer.
  • SEQ ID NO: 2 shows the sequence of a target sequence according to the invention that is located within the SH3 locus.
  • SEQ ID NO: 3 shows the sequence of a target sequence according to the invention that is located within the SH4 locus.
  • SEQ ID NO: 4 shows the sequence of the target sequence of the wild-type I-CreI homodimeric protein.
  • SEQ ID Nos. 5 to 10 represent sequences shown on FIG. 1.
  • SEQ ID Nos. 11 to 15 represent oligonucleotides, primers and linkers used in Example 1.
  • SEQ ID Nos. 16 to 19 represent sequences shown on FIG. 4.
  • SEQ ID Nos. 20 to 24 represent oligonucleotides, primers and linkers used in Example 2.
  • SEQ ID Nos. 25 to 32 represent the single-chain meganucleases constructed in Example 1, referred to as SCOH-SH3-b56-A, SCOH-SH3-b56-B, SCOH-SH3-b56-C, SCOH-SH3-b56-D, SCOH-SH3-b1-A, SCOH-SH3-b1-B, SCOH-SH3-b1-C and SCOH-SH3-b1-D respectively.
  • SEQ ID Nos. 33 to 40 represent the single-chain meganucleases constructed in Example 2, referred to as SCOH-SH4-b56-A, SCOH-SH4-b56-B, SCOH-SH4-b56-C, SCOH-SH4-b56-D, SCOH-SH4-b1-A, SCOH-SH4-b1-B, SCOH-SH4-b1-C and SCOH-SH4-b1-D respectively.
  • SEQ ID NO: 41 represents the positive control SCOH-RAG.
  • SEQ ID NO: 42 shows the amino acid sequence of a I-CreI monomer with an additional alanine at position 2, and with three additional residues after the final proline.
  • SEQ ID NO: 43 shows the amino acid sequence of the RM2 linker.
  • SEQ ID Nos. 44 to 49 represent oligonucleotides, primers and linkers used in Example 3.
  • SEQ ID Nos. 50 to 53 represent oligonucleotides, primers and linkers used in Example 4.
  • SEQ ID Nos. 54 to 55 show sequences comprised in the SH3, SH4 and SH6 loci, respectively.
  • SEQ ID NO: 57 shows a monomer derived from a monomer of SEQ ID NO: 1 that comprises 44A 54L 64A 70Q 75N 158R 162A mutations.
  • SEQ ID NO: 58 shows a monomer derived from a monomer of SEQ ID NO: 42 that comprises 44A 54L 64A 70Q 75N 158R 162A mutations.
  • SEQ ID NO: 59 shows the sequence of a target sequence according to the invention that is located within the SH6 locus.
  • SEQ ID Nos. 60 to 64 represent sequences shown on FIG. 9.
  • SEQ ID Nos. 65 to 75 represent oligonucleotides, primers and linkers used in Example 5.
  • SEQ ID Nos. 76 to 85 represent the single-chain meganucleases constructed in Example 5, referred to as SCOH-SH6-b1-B, SCOH-SH6-b1-C, SCOH-SH6-b1-C, QCSH61-A01, QCSH61-E01, QCSH61-H0, QCSH62-A02, QCSH61-H01b, QCSH61-H01c and QCSH61-H01d respectively.
  • SEQ ID Nos. 86 to 96 represent the single-chain meganucleases capable of cleaving the SH7 locus (SEQ ID Nos. 86 and 87), SH8 locus (SEQ ID NO: 88), the SH12 locus (SEQ ID NO: 89), the SH13 locus (SEQ ID NO: 90), the SH19 locus (SEQ ID NO: 91), the SH20 locus (SEQ ID NO: 92), the SH21 locus (SEQ ID Nos. 93 to 95) and the SH33 locus (SEQ ID NO: 96).
  • SEQ ID Nos. 97 to 104 represent sequences comprised within the SH12, SH13, SH19, SH20, SH21, SH33, SH7 and SH8 loci, respectively.
  • SEQ ID Nos. 105 to 325 represent sequences disclosed in Examples 6 to 9 and/or in any one of Tables A′, A″, E, G and H.
  • SEQ ID NO: 326 shows the amino acid sequence of the BQY linker.
  • EXAMPLES
  • In the following examples, all the I-CreI variants were constructed by genetic engineering of I-CreI monomers of SEQ ID NO: 42.
  • Example 1 Engineering Meganucleases Targeting the SH3 Locus
  • SH3 is a locus comprising a 24 bp non-palindromic target (SEQ ID NO: 2) that is present on chromosome 6. As shown in Table A, SH3 is located in the vicinity of a RIS disclosed in Deichmann et al. (J. of Clin. Invest. 2007 117:2225). The SH3 sequence is not included in any of the CIS described in Deichmann et al.
  • I-CreI heterodimers capable of cleaving a target sequence of SEQ ID NO: 2 were identified using methods derived from those described in Chames et al. (Nucleic Acids Res., 2005, 33, e178), Arnould et al. (J. Mol. Biol., 2006, 355, 443-458), Smith et al. (Nucleic Acids Res., 2006, 34, e149), Arnould et al. (Arnould et al. J Mol. Biol. 2007 371:49-65). Some of these heterodimers were then cloned into mammalian expression vectors for assessing SH3 cleavage in CHO cells. These results were then utilized to design single-chain meganucleases directed against the target sequence of SEQ ID NO: 2. These single-chain meganucleases were cloned into mammalian expression vectors and tested for SH3 cleavage in CHO cells. Strong cleavage activity of the SH3 target could be observed for these single chain molecules in mammalian cells.
  • Example 1.1. Identification of Meganucleases Cleaving SH3
  • I-CreI variants potentially cleaving the SH3 target sequence in heterodimeric form were constructed by genetic engineering. Pairs of such variants were then co-expressed in yeast. Upon co-expression, one obtains three molecular species, namely two homodimers and one heterodimer. It was then determined whether the heterodimers were capable of cutting the SH3 target sequence of SEQ ID NO: 2.
  • a) Construction of Variants of the I-CreI Meganuclease Cleaving Palindromic Sequences Derived from the SH3 Target Sequence
  • The SH3 sequence is partially a combination of the 10AAT_P (SEQ ID NO: 5), 5AAG_P (SEQ ID NO: 6), 10AGG_P (SEQ ID NO: 7) and 5TTT_P (SEQ ID NO: 8) target sequences which are shown on FIG. 1. These sequences are cleaved by mega-nucleases obtained as described in International PCT applications WO 2006/097784 and WO 2006/097853, Arnould et al. (J. Mol. Biol., 2006, 355, 443-458) and Smith et al. (Nucleic Acids Res., 2006). Thus, SH3 should be cleaved by combinatorial variants resulting from these previously identified meganucleases.
  • Two palindromic targets, SH3.3 and SH3.4, were derived from SH3 (FIG. 1). Since SH3.3 and SH3.4 are palindromic, they should be cleaved by homodimeric proteins. Therefore, homodimeric I-CreI variants cleaving either the SH3.3 palindromic target sequence of SEQ ID NO: 9 or the SH3.4 palindromic target sequence of SEQ ID NO: 10 were constructed using methods derived from those described in Chames et al. (Nucleic Acids Res., 2005, 33, e178), Arnould et al. (J. Mol. Biol., 2006, 355, 443-458), Smith et al. (Nucleic Acids Res., 2006, 34, e149) and Arnould et al. (Arnould et al. J Mol Biol. 2007 371:49-65).
  • b) Construction of Target Vector
  • An oligonucleotide of SEQ ID NO: 11, corresponding to the SH3 target sequence flanked by gateway cloning sequences, was ordered from PROLIGO. This oligo has the following sequence: TGGCATACAAGTTTCCAATACAAGGTACAAAGTCCTGACAATCGTCTGTCA). Double-stranded target DNA, generated by PCR amplification of the single stranded oligonucleotide, was cloned into the pCLS1055 yeast reporter vector using the Gateway protocol (INVITROGEN).
  • Yeast reporter vector was transformed into the FYBL2-7B Saccharomyces cerevisiae strain having the following genotype: MAT a, ura3Δ851, trp1Δ63, leu2Δ1, lys2A202. The resulting strain corresponds to a reporter strain (MilleGen).
  • c) Co-Expression of Variants
  • The open reading frames coding for the variants cleaving the SH3.4 or the SH3.3 sequence were cloned in the pCLS542 expression vector and in the pCLS1107 expression vector, respectively. Yeast DNA from these variants was extracted using standard protocols and was used to transform E. coli. The resulting plasmids were then used to co-transform yeast. Transformants were selected on synthetic medium lacking leucine and containing G418.
  • d) Mating of Meganucleases Coexpressing Clones and Screening in Yeast
  • Mating was performed using a colony gridder (QpixII, Genetix). Variants were gridded on nylon filters covering YPD plates, using a low gridding density (4-6 spots/cm2). A second gridding process was performed on the same filters to spot a second layer consisting of different reporter-harboring yeast strains for each target. Membranes were placed on solid agar YPD rich medium, and incubated at 30° C. for one night, to allow mating. Next, filters were transferred to synthetic medium, lacking leucine and tryptophan, adding G418, with galactose (2%) as a carbon source, and incubated for five days at 37° C., to select for diploids carrying the expression and target vectors. After 5 days, filters were placed on solid agarose medium with 0.02% X-Gal in 0.5 M sodium phosphate buffer, pH 7.0, 0.1% SDS, 6% dimethyl formamide (DMF), 7 mM β-mercaptoethanol, 1% agarose, and incubated at 37° C., to monitor β-galactosidase activity. Results were analyzed by scanning and quantification was performed using an appropriate software.
  • e) Results
  • Co-expression of different variants resulted in cleavage of the SH3 target in 58 tested combinations. Functional combinations are summarized in Table I herebelow. In this table, “+” indicates a functional combination on the SH3 target sequence, i.e., the heterodimer is capable of cleaving the SH3 target sequence.
  • TABLE I
    Amino acids positions and residues
    of the I-Crel variants cleaving the SH3.3 target
    44A
    4E 1V 54L
    44A 44A 44A
    44A 44A 66C 44A
    54L 54L 54L
    54L 54L 70Q 54L
    70Q 64A 70Q 64A
    64A 71R 57E 44V
    75N 70Q 75Y 70Q 70Q 75N 44A 70Q 54L
    105A 75N 92R 75N 75N 151A 54L 75N 70Q
    158R 158R
    158R 158R 158W 158R 70Q 158R 75N
    162A 162A 162A 162A 162A 162A
    75N
    162A 77V
    Amino acids 30G 38R + + + + + + +
    positions and 70D 75N
    resdidues of the I- 86D
    Crel variants cleaving 30G 38R + + + + + +
    the SH3.4 target 70D 75N
    81T 154G
    30G 38R + + + + + + + + +
    50R 70D
    75N 142R
  • In conclusion, several heterodimeric I-CreI variants, capable of cleaving the SH3 target sequence in yeast, were identified.
  • Example 1.2. Validation of SH3 Target Cleavage in an Extrachromosomal Model in CHO Cells
  • I-CreI variants able to efficiently cleave the SH3 target in yeast when forming heterodimers are described hereabove in example 1.1. In order to identify heterodimers displaying maximal cleavage activity for the SH3 target in CHO cells, the efficiency of some of these variants was compared using an extrachromosomal assay in CHO cells. The screen in CHO cells is a single-strand annealing (SSA) based assay where cleavage of the target by the meganucleases induces homologous recombination and expression of a LagoZ reporter gene (a derivative of the bacterial lacZ gene).
  • a) Cloning of SH3 Target in a Vector for CHO Screen
  • An oligonucleotide corresponding to the SH3 target sequence flanked by gateway cloning sequences, was ordered from PROLIGO (SEQ ID NO: 12; TGGCATACAAGTTTCCAATACAAGGTACAAAGTCCTGACAATCGTCTGTCA). Double-stranded target DNA, generated by PCR amplification of the single stranded oligonucleotide, was cloned using the Gateway protocol (INVITROGEN) into the pCLS1058 CHO reporter vector. Cloned target was verified by sequencing (MILLEGEN).
  • b) Re-Cloning of Meganucleases
  • The open-reading frames coding for these variants identified in Table I hereabove sub-cloned into the pCLS2437 expression vector. ORFs were amplified by PCR on yeast DNA using primers of SEQ ID Nos. 13 and 14 (5′-AAAAAGCAGGCTGGCGCGCCTACACAGCGGCCTTGCCACCATG-3′ and 5′-AGAAAGCTGGGTGCTAGCGCTCGAGTTATCAGTCGG-3′). PCR products were cloned in the CHO expression vector pCLS2437 using the AscI and XhoI restriction enzymes for internal fragment replacement. Selected clones resulting from ligation and E. coli transformation steps were verified by sequencing (MILLEGEN).
  • c) Extrachromosomal Assay in Mammalian Cells
  • CHO K1 cells were transfected with Polyfect® transfection reagent according to the supplier's protocol (QIAGEN). 72 hours after transfection, culture medium was removed and 150 μl of lysis/revelation buffer for β-galactosidase liquid assay was added (typically 1 liter of buffer contained 100 ml of lysis buffer (Tris-HCl 10 mM pH7.5, NaCl 150 mM, Triton X100 0.1%, BSA 0.1 mg/ml, protease inhibitors), 10 ml of Mg 100× buffer (MgCl 2 100 mM, β-mercaptoethanol 35%), 110 ml ONPG 8 mg/ml and 780 ml of sodium phosphate 0.1M pH7.5). After incubation at 37° C., OD was measured at 420 nm. The entire process was performed on an automated Velocity11 BioCel platform.
  • Per assay, 150 ng of target vector was cotransfected with 12.5 ng of each one of both variants.
  • d) Results
  • The four following variants described in Table I were re-cloned into pCLS2437:
      • 44A 54L 70Q 75Y 92R 158R 162A (referred to as SH3.3-MA);
      • 1V 44A 54L 64A 70Q 75N 158W 162A (referred to as SH3.3-MB);
      • 30G 38R 70D 75N 86D (referred to as SH3.4-M1); and
      • 30G 38R 70D 75N 81T 154G (referred to as SH3.4-M2).
  • These I-CreI variants were assayed together as heterodimers against the SH3 target in the CHO extrachromosomal assay.
  • Table II shows the functional combinations obtained for nine heterodimers.
  • TABLE II
    Optimized variants cleaving SH3.3
    44A 54L 70Q 75Y 1V 44A 54L 64A 70Q
    92R 158R
    162A 75N 158W
    162A
    Optimized 30G 38R 70D + +
    variants 75N 86D
    cleaving 30G 38R 70D + +
    SH3.4 75N 81T 154G
  • Analysis of the efficiencies of cleavage and recombination of the SH3 sequence demonstrates that all of the four tested combinations of I-CreI variants were capable to transpose their cleavage activity from yeast to CHO cells without additional mutation.
  • Example 1.3. Covalent Assembly as Single Chain and Improvement of Meganucleases Cleaving SH3
  • Co-expression of the variants identified in example 1.1. leads to a high cleavage activity of the SH3 target in yeast. Some of the heterodimers have been validated for SH3 cleavage in a mammalian expression system (example 1.2.). One of them, shown in Table III, was selected for further optimization.
  • TABLE III
    Amino acids positions and residues
    SH3 variant of the I-CreI variants
    SH3.3- MA 44A 54L 70Q 75Y 92R 158R 162A
    SH3.4-M1 30G 38R 70D 75N 86D
  • The MA×M1 SH3 heterodimer gives high cleavage activity in yeast. SH3.3-MA is a SH3.3 cutter that bears the following mutations in comparison with the I-CreI wild type sequence: 44A 54L 70Q 75Y 92R 158R 162A. SH3.4-M1 is a SH3.4 cutter that bears the following mutations in comparison with the I-CreI wild type sequence: 30G 38R 70D 75N 86D.
  • Single chain constructs were engineered using the linker RM2 of SEQ ID NO: 15 (AAGGSDKYNQALSKYNQALSKYNQALSGGGGS), thus resulting in the production of the single chain molecule: MA-linkerRM2-M1. During this design step, the G195 mutation was introduced in the C-terminal M1 variant. In addition, mutations K7E, K96E were introduced into the MA variant and mutations E8K, E61 R into the M1 variant to create the single chain molecule: MA (K7E K96E)-linkerRM2-M1 (E8K E61R G195) that is further called SCOH-SH3-b1 scaffold. Some additional amino-acid substitutions have been found in previous studies to enhance the activity of I-CreI derivatives: the replacement of Isoleucine 132 with Valine (I132V) is one of them. The I132V mutation was introduced into either one, both or none of the coding sequence of N-terminal and C-terminal protein fragments.
  • The same strategy was applied to a second scaffold, termed SCOH-SH3-b56 scaffold, based on the best variants cleaving SH3.3 (44A 54L 70Q 75Y 92R 158R 162A) and SH3.4 (30G 38R, 50R 70D 75N 142R) as homodimers, respectively.
  • The resulting proteins are shown in Table IV below. All the single chain molecules were assayed in CHO for cleavage of the SH3 target.
  • a) Cloning of the Single Chain Molecule
  • A series of synthetic gene assembly was ordered to MWG-EUROFINS. Synthetic genes coding for the different single chain variants targeting SH3 were cloned in pCLS1853 using AscI and XhoI restriction sites.
  • b) Extrachromosomal Assay in Mammalian Cells
  • CHO K1 cells were transfected as described in example 1.2. 72 hours after transfection, culture medium was removed and 150 μl of lysis/revelation buffer for 1-galactosidase liquid assay was added. After incubation at 37° C., OD was measured at 420 nm. The entire process is performed on an automated Velocity11 BioCel platform. Per assay, 150 ng of target vector was cotransfected with an increasing quantity of variant DNA from 3.12 to 25 ng (25 ng of single chain DNA corresponding to 12.5 ng+12.5 ng of heterodimer DNA). Finally, the transfected DNA variant DNA quantity was 3.12 ng, 6.25 ng, 12.5 ng and 25 ng. The total amount of transfected DNA was completed to 175 ng (target DNA, variant DNA, carrier DNA) using an empty vector (pCLS0002).
  • d) Results
  • The activity of the single chain molecules against the SH3 target was monitored using the previously described CHO assay along with our internal control SCOH-RAG and I-Sce I meganucleases. All comparisons were done at 3.12 ng, 6.25 ng, 12.5 ng, and 25 ng transfected variant DNA (FIGS. 2 and 3). All the single molecules displayed SH3 target cleavage activity in CHO assay as listed in Table IV.
  • TABLE IV
    Cleavage
    Mutations on N- Mutations on C- SEQ ID of SH3 in
    Name terminal monomer terminal monomer No. CHO cells
    SCOH-SH3-b56- A 7E 44A 54L 70Q 75Y 8K 19S 30G 38R 25 +
    92R 96E 158R 162A 50R 61R 70D 75N
    142R
    SCOH-SH3-b56- B 7E 44A 54L 70Q 75Y 8K 19S 30G 38R 26 +
    92R 96E 132V 158R 50R 61R 70D 75N
    162A 142R
    SCOH-SH3-b56- C 7E 44A 54L 70Q 75Y 8K 19S 30G 38R 27 +
    92R 96E 132V 158R 50R 61R 70D 75N
    162A 132V 142R
    SCOH-SH3-b56- D 7E 44A 54L 70Q 75Y 8K 19S 30G 38R 28 +
    92R 96E 158R 162A 50R 61R 70D 75N
    132V 142R
    SCOH-SH3-b1- A 7E 44A 54L 70Q 75Y 8K 19S 30G 38R 29 +
    92R 96E 158R 162A 61R 70D 75N 86D
    SCOH-SH3-b1- B 7E 44A 54L 70Q 75Y 8K 19S 30G 38R 30 +
    92R 96E 132V 158R 61R 70D 75N 86D
    162A
    SCOH-SH3-b1- C 7E 44A 54L 70Q 75Y 8K 19S 30G 38R 31 +
    92R 96E 132V 158R 61R 70D 75N 86D
    162A 132V
    SCOH-SH3-b1- D 7E 44A 54L 70Q 75Y 8K 19S 30G 38R 32 +
    92R 96E 158R 162A 61R 70D 75N 86D
    132V
  • Variants shared specific behaviour upon assayed dose depending on the mutation profile they bear (FIGS. 2 and 3). For example, SCOH-SH3-b1-C has a similar profile, and is even more active than. Its activity reaches the maxima at the lowest DNA quantity transfected from low quantity to high quantity. In comparison with SCOH-SH3-b1-C, the molecule SCOH-SH3-b56-A has a maximal activity at higher DNA doses but reaches equivalent level of activity of SCOH-SH3-b1-C and our internal standard.
  • All of the variants described are active and can be used for inserting transgenes into the SH3 locus.
  • Example 2 Engineering Meganucleases Targeting the SH4 Locus
  • SH4 is a locus that is present on chromosome 7. The SH4 locus comprises a 24 bp non-palindromic sequence of SEQ ID NO: 3. As shown in Table A, SH4 is located in the vicinity a RIS disclosed in Schwarzwaelder et al. (J. Clin. Invest. 2007 117:2241). The SH4 sequence is not included in any of the CIS described in Deichman et al.
  • Experiments similar to those described hereabove in Example 1 were carried out to identify I-CreI heterodimers and single-chain meganucleases capable of cleaving a target sequence of SEQ ID NO: 3.
  • Example 2.1. Identification of Meganucleases Cleaving SH4
  • I-CreI variants potentially cleaving the SH4 target sequence in heterodimeric form were constructed by genetic engineering. Pairs of such variants were then co-expressed in yeast. Upon co-expression, one obtains three molecular species, namely two homodimers and one heterodimer. It was then determined whether the heterodimers were capable of cutting the SH4 target sequence of SEQ ID NO: 3.
  • a) Construction of Variants of the I-CreI Meganuclease Cleaving Palindromic Sequences Derived from the SH4 Target Sequence
  • The SH4 sequence is partially a combination of the 10AAA_P (SEQ ID NO: 4), 5ACT_P (SEQ ID NO: 16), 10AAA_P (SEQ ID NO: 4), 5GGT_P (SEQ ID NO: 17) targets shown on FIG. 4. These sequences are cleaved by previously identified mega-nucleases, obtained as described in International PCT Applications WO 2006/097784 and WO 2006/097853; Arnould et al., J. Mol. Biol., 2006, 355, 443-458; Smith et al., Nucleic Acids Res., 2006. Thus, SH4 should be cleaved by combinatorial variants resulting from these previously identified meganucleases.
  • The screening procedure was performed using methods derived from those described in Chames et al. (Nucleic Acids Res., 2005, 33, e178), Arnould et al. (J. Mol. Biol., 2006, 355, 443-458), Smith et al. (Nucleic Acids Res., 2006, 34, e149) and Arnould et al. (Arnould et al. J Mol Biol. 2007 371:49-65) on the two following palindromic sequences: the SH4.3 sequence of SEQ ID NO: 18 and the SH4.4 sequence of SEQ ID NO: 19.
  • b) Construction of Target Vector
  • The experimental procedure is as described in Example 1.1, with the exception that an oligonucleotide corresponding to the SH4 target sequence of SEQ ID NO: 20 (5′-TGGCATACAAGTTTTTAAAACACTGTACACCATTTTGACAATCGTCTGTCA-3′) was used.
  • c) Co-Expression of Variants
  • Yeast DNA from variants cleaving the SH4.3 and SH4.4 target in the pCLS542 and pCLS1107 expression vectors was extracted using standard protocols and was used to transform E. coli. The resulting plasmid DNA was then used to co-transform yeast strain. Transformants were selected on synthetic medium lacking leucine and containing G418.
  • d) Mating of Meganucleases Coexpressing Clones and Screening in Yeast
  • Mating was performed using a colony gridder (QpixII, Genetix). Variants were gridded on nylon filters covering YPD plates, using a low gridding density (4-6 spots/cm2). A second gridding process was performed on the same filters to spot a second layer consisting of different reporter-harboring yeast strains for each target. Membranes were placed on solid agar YPD rich medium, and incubated at 30° C. for one night, to allow mating. Next, filters were transferred to synthetic medium, lacking leucine and tryptophan, adding G418, with galactose (2%) as a carbon source, and incubated for five days at 37° C., to select for diploids carrying the expression and target vectors. After 5 days, filters were placed on solid agarose medium with 0.02% X-Gal in 0.5 M sodium phosphate buffer, pH 7.0, 0.1% SDS, 6% dimethyl formamide (DMF), 7 mM β-mercaptoethanol, 1% agarose, and incubated at 37° C., to monitor β-galactosidase activity. Results were analyzed by scanning and quantification was performed using appropriate software.
  • e) Results
  • Co-expression of variants cleaving the SH4.3 target and of variants cleaving the SH4.4 target resulted in cleavage of the SH4 target in 6 cases. Functional combinations are summarized in Table V.
  • TABLE V
    Amino acids positions and residues
    of the I-CreI variants cleaving the SH4.3 target
    24V 44R 68Y 70S 24V 68A 70S 75N 24V 70D 75N
    75Y 77N 77R 77R
    Amino acids positions and 24V 44Y 70S + + +
    resdidues 24V 44Y 70S + + +
    of I-CreI variants cleaving 77V
    the SH4.4 target
  • Example 2.2. Validation of SH4 Target Cleavage in an Extrachromosomal Model in CHO Cells
  • In order to identify heterodimers displaying maximal cleavage activity for the SH4 target in CHO cells, the efficiency of several combinations of variants to cut the SH4 target was assessed using an extrachromosomal assay in CHO cells. The screen in CHO cells is a single-strand annealing (SSA) based assay where cleavage of the target by the meganucleases induces homologous recombination and expression of a LagoZ reporter gene (a derivative of the bacterial lacZ gene).
  • a) Cloning of SH4 Target in a Vector for CHO Screen
  • The target was cloned as follows. An oligonucleotide of SEQ ID NO: 21, corresponding to the SH4 target sequence flanked by gateway cloning sequence, was ordered from PROLIGO (5′-TGGCATACAAGTTTTTAAAACACTGTACACCATTTTGACAATCGTCTGTCA-3′). Double-stranded target DNA, generated by PCR amplification of the single stranded oligonucleotide, was cloned using the Gateway protocol (INVITROGEN) into CHO reporter vector (pCLS1058). The cloned fragment was verified by sequencing (MILLEGEN).
  • b) Re-Cloning of Meganucleases
  • The ORFs of I-CreI variants cleaving the SH4.5 and SH4.6 targets obtained hereabove were sub-cloned in pCLS2437. ORFs were amplified by PCR on yeast DNA using primers of SEQ ID NO: 22 and 23 (5′-AAAAAGCAGGCTGGCGCGCCTACACAGCGGCCTTGCCACCATG-3′ and 5′-AGAAAGCTGGGTGCTAGCGCTCGAGTTATCAGTCGG-3′) primers. PCR products were cloned in the CHO expression vector pCLS2437 using the AscI and NheI restrictions sites for internal fragment replacement. Selected clones resulting from ligation and E. coli transformation steps were verified by sequencing (MILLEGEN).
  • c) Extrachromosomal Assay in Mammalian Cells
  • CHO K1 cells were transfected with Polyfect® transfection reagent according to the supplier's protocol (QIAGEN). 72 hours after transfection, culture medium was removed and 150 μl of lysis/revelation buffer for β-galactosidase liquid assay was added (typically 1 liter of buffer contained: 100 ml of lysis buffer (Tris-HCl 10 mM pH7.5, NaCl 150 mM, Triton X100 0.1%, BSA 0.1 mg/ml, protease inhibitors), 10 ml of Mg 100× buffer (MgCl 2 100 mM, β-mercaptoethanol 35%), 110 ml ONPG 8 mg/ml and 780 ml of sodium phosphate 0.1M pH7.5). After incubation at 37° C., OD was measured at 420 nm. The entire process is performed on an automated Velocity11 BioCel platform. Per assay, 150 ng of target vector was cotransfected with 12.5 ng of each one of both variants (12.5 ng of variant cleaving palindromic SH4.3 target and 12.5 ng of variant cleaving palindromic SH4.4 target).
  • d Results
  • The four variants shown in Table VI and described herebaove in Example 2.1, were selected for further analysis.
  • TABLE VI
    Amino acids positions and residues
    of the I-CreI variants
    SH4.3-MA 24V 44R 68Y 70S 75Y 77N
    SH4.3-MC 24V 68A 70S 75N 77R
    SH4.4-M1 24V 44Y 70S
    SH4.4-M2 24V 44Y 70S 77V
  • These variants were cloned in pCLS2437. Then, I-CreI variants cleaving the SH4.3 or SH4.4 targets were assayed together as heterodimers against the SH4 target in the CHO extrachromosomal assay. Analysis of the efficiencies of cleavage and recombination of the SH4 sequence demonstrates that all tested combinations of I-CreI variants were able to transpose their cleavage activity from yeast to CHO cells without additional mutation (Table VII).
  • TABLE VII
    Amino acids
    positions and residues
    of the I-CreI variants:
    variants cleaving SH4.3
    SH4.3-MA: SH4.3-MC:
    24V 44R 68Y 24V 68A
    70S 75Y 77N 70S 75N 77R
    Amino acids SH4.4-M1: + +
    positions and 24V 44Y 70S
    residues of the SH4.4-M2: + +
    I-CreI variants: 24V 44Y 70S 77V
    variants cleaving
    SH4.4
  • Example 2.3. Covalent Assembly as Single Chain and Improvement of Meganucleases Cleaving SH4 by Site-Directed Mutagenesis
  • Co-expression of the variants described in Example 2.1. leads to a high cleavage activity of the SH4 target in yeast. In addition, some of them have been validated for SH4 cleavage in a mammalian expression system (Example 2.2.).
  • The MA×M2 SH4 heterodimer gives high cleavage activity in yeast. SH4.3-MA is a SH4.3 cutter that bears the following mutations in comparison with the I-CreI wild type sequence: 24V 44R 68Y 70S 75Y 77N. SH4.4-M2 is a SH4.4 cutter that bears the following mutations in comparison with the I-CreI wild type sequence: 24V 44Y 70S 77V.
  • As described in example 1.3, single chain constructs were engineered using the linker RM2, thereby resulting in the production of a single chain molecule referred to as MA-LinkerRM2-M2. During this design step, the G19S mutation was introduced in the C-terminal M2 mutant. In addition, K7E and K96E mutations were introduced into the MA mutant, and E8K and E61R mutations into the M2 mutant in order to create a single chain molecule referred to as MA (K7E K96E)-linkerRM2-M2 (E8K E61R G19S) that is called further SCOH-SH4-b1 scaffold.
  • The Isoleucine 132 to Valine (I132V) mutation was introduced into the coding sequence of either, one, none or both N-terminal and C-terminal protein fragment.
  • The same strategy was applied to a second scaffold based on the good cutters on SH4.3 (44R 68Y 70S 75Y 77N) and SH4.4 (24V 44Y 70S 77V). This scaffold is further referred to as SCOH-SH4-b56 scaffold.
  • The design of the derived single chain constructs is shown in Table VIII. The single chain constructs were tested in CHO for their ability to induce cleavage of the SH4 target.
  • a) Cloning of the Single Chain Molecule
  • A series of synthetic gene assembly was performed to MWG-EUROFINS. Synthetic genes, coding for the different single chain variants targeting SH4, were cloned in pCLS1853 using AscI and XhoI restriction sites.
  • b) Extrachromosomal Assay in Mammalian Cells
  • CHO K1 cells were transfected as described hereabove. 72 hours after transfection, culture medium was removed and 150 μl of lysis/revelation buffer for β-galactosidase liquid assay was added. After incubation at 37° C., OD was measured at 420 nm. The entire process is performed on an automated Velocity11 BioCel platform. Per assay, 150 ng of target vector was cotransfected with an increasing quantity of variant DNA from 3.12 to 25 ng (25 ng of single chain DNA corresponding to 12.5 ng+12.5 ng of heterodimer DNA). Finally, the transfected DNA variant DNA quantity was 3.12 ng, 6.25 ng, 12.5 ng and 25 ng. The total amount of transfected DNA was completed to 175 ng (target DNA, variant DNA, carrier DNA) using an empty vector (pCLS0002).
  • c) Results
  • The single chain molecules described in Table VIII were monitored for their activity against the SH4 target using the previously described CHO assay by comparison to our internal control SCOH-RAG and I-Sce I meganucleases. All activity evaluation was done upon DNA transfected dose of 3.12 ng, 6.25 ng, 12.5 ng, and 25 ng. All single chain molecules were displaying activity on SH4 target as reported in Table VIII.
  • TABLE VIII
    Activity on
    SH4 target
    Mutations on N-terminal Mutations on C- SEQ ID in CHO
    Name monomer terminal monomer No. Assay
    SCOH- 7E 44R 68Y 70S 75Y 8K 19S 24V 44Y 61R 33 +
    SH4-b56-A 77N 96E 70S 77V
    SCOH- 7E 44R 68Y 70S 75Y 8K 19S 24V 44Y 61R 34 +
    SH4-b56-B 77N 96E 132V 70S 77V
    SCOH- 7E 44R 68Y 70S 75Y 8K 19S 24V 44Y 61R 35 +
    SH4-b56-C 77N 96E 132V 70S 77V 132V
    SCOH- 7E 44R 68Y 70S 75Y 8K 19S 24V 44Y 61R 36 +
    SH4-b56-D 77N 96E 70S 77V 132V
    SCOH- 7E 24V 44R 68Y 70S 8K 19S 24V 44Y 61R 37 +
    SH4-b1-A 75Y 77N 96E 70S 77V
    SCOH- 7E 24V 44R 68Y 70S 8K 19S 24V 44Y 61R 38 +
    SH4-b1-B 75Y 77N 96E 132V 70S 77V
    SCOH- 7E 24V 44R 68Y 70S 8K 19S 24V 44Y 61R 39 +
    SH4-b1-C 75Y 77N 96E 132V 70S 77V 132V
    SCOH- 7E 24V 44R 68Y 70S 8K 19S 24V 44Y 61R 40 +
    SH4-b1-D 75Y 77N 96E 70S 77V 132V
  • Variants shared specific behaviour upon assayed dose depending on the mutation profile they bear (FIGS. 5 and 6). For example, SCOH-SH4-b1C shows an activity level within the same range as the internal standard SCOH-RAG (: its activity increases from low quantity to high quantity. At the assayed DNA trasfected doses, its activity is superior to that of SCOH-SH4-B56A.
  • All of these variants are active at different levels of intensity and can thus be used for SH4 genome targeting.
  • Example 3 Detection of Cleavage Activity at the SH Loci in Human Cell Line
  • I-CreI variants able to efficiently cleave the SH3 and SH4 targets in yeast and in mammalian cells (CHO K1 cells) have been identified in Examples 1 and 2. The efficiency of the SH3 and SH4 meganucleases to cleave their endogenous DNA target sequences was next tested. This example will demonstrate that meganucleases engineered to cleave the SH3 and SH4 target sequences cleave their cognate endogenous sites in human cells.
  • Repair of double-strand break by non homologous end-joining (NHEJ) can generate small deletions and insertions (InDel) (FIG. 7). In nature, this error-prone mechanism can be deleterious for the cells survival but provides a rapid indicator of meganucleases activity at endogenous loci.
  • Example 3.1: Detection of Induced Mutagenesis at the Endogenous Site
  • The assays based on cleavage-induced recombination in mammal or yeast cells, which are used for screening variants with altered specificity, are described in International PCT Application WO 2004/067736; Epinat et al., Nucleic Acids Res., 2003, 31:2952-2962; Chames et al., Nucleic Acids Res., 2005, 33:e178, and Arnould et al., J. Mol. Biol., 2006, 355:443-458. These assays result in a functional LacZ reporter gene which can be monitored by standard methods.
  • Single Chain I-CreI variants for SH3 and SH4 cloned in the pCLS1853 plasmid were used for this experiment. The day previous experiment, cells from the human embryonic kidney cell line, 293-H (Invitrogen) were seeded in a 10 cm dish at density of 1.2 106 cells/dish. The following day, cells were transfected with 3 μg of an empty plasmid or a meganuclease-expressing plasmid using lipofectamine (Invitrogen). 72 hours after transfection, cells were collected and diluted (dilution 1/20) in fresh culture medium. After 7 days of culture, cells were collected and genomic DNA extracted.
  • 200 ng of genomic DNA were used to amplify the endogenous locus surrounding the meganuclease cleavage site by PCR amplification. A 377 bp fragment corresponding to the SH3 locus was amplified using specific PCR primers A (SEQ ID NO 44; 5′-tgggggtcttactctgtttccc-3′) and B (SEQ ID NO 45; 5′-aggagagtccttctttggcc-3′). A 396 bp fragment corresponding to the SH4 locus was amplified using PCR primers C (SEQ ID NO 46; 5′-gagtgatagcataatgaaaacc-3′) and D (SEQ ID NO 47; 5′-ctcaccataagtcaactgtctc-3′). PCR amplification was performed to obtain a fragment flanked by specific adaptator sequences (SEQ ID NO 48; 5′-CCATCTCATCCCTGCGTGTCTCCGACTCAG-3′ and SEQ ID NO: 49 5′-CCTATCCCCTGTGTGCCTTGGCAGTCTCAG-3′) provided by the company offering sequencing service (GATC Biotech AG, Germany) on the 454 sequencing system (454 Life Sciences). An average of 18,000 sequences was obtained from pools of 2 amplicons (500 ng each). After sequencing, different samples were identified based on barcode sequences introduced in the first of the above adaptators. Sequences were then analyzed for the presence of insertions or deletions in the cleavage site of SH3 or SH4 respectively.
  • Example 3.2: Results
  • TABLE IX
    Total InDel % of
    Vector sequence containing InDel
    expressing: number sequences events
    SH
    3 meganuclease 12841 56 0.44
    Empty 2153 1 0.05
    SH 4 meganuclease 8259 18 0.22
    Empty 12811 3 0.02
  • The analysis of the genomic DNA extracted from cells transfected with the meganuclease targeting the SH3 locus showed that 56 out of the 12841 analyzed sequences (0.44%) contained InDel events within the recognition site of SH3. Similarly, after transfection with the meganuclease targeting the SH4 locus, 18 out of the 8259 analyzed sequences (0.22%) contained InDel events within the recognition site of SH4.
  • Since small deletions or insertions could be related to PCR or sequencing artefacts, the same loci were analyzed after transfection with a plasmid that does not express the meganuclease. The analysis of the SH3 and SH4 loci revealed that virtually no InDel events could be detected. Indeed, only 0.05% (1/2153) and 0.02% (3/12811) of the analyzed sequences contained mutations.
  • Moreover, the analysis of the size of the DNA insertion or deletion sequences (FIG. 8) revealed a similar type of events with a predominance of small insertions (<5 bp) and of small deletions (<10 bp).
  • These data demonstrate that the meganucleases engineered to target respectively the SH3 or SH4 loci are active in human cells and can cleave their cognate endogenous sequence. Moreover, it shows that meganucleases have the ability to generate small InDel events within a sequence which would disrupt a gene ORF and thus inactivate the corresponding gene expression product.
  • Example 4 Gene Targeting at the Endogenous SH3 and SH4 Loci in Human Cells
  • To validate the cleavage activity of engineered single-chain SH3 and SH4 meganucleases, their ability to stimulate homologous recombination at the endogenous human SH3 and SH4 loci was next evaluated. Cells were transfected with mammalian expression plasmids for single chain molecules SCOH-SH3-b1-C or SCOH-SH4-b1-C and a vector comprising a targeting construct. The vector comprising a targeting construct (also referred to as “donor repair plasmid”) was the pCLS3777 or pCLS3778 plasmid containing a 2.8 kb sequence consisting of an exogenous DNA sequence, flanked by two sequences homologous to the human SH3 or SH4 loci. The sequences homologous to the human SH3 or SH4 loci had a length of 1.5 kb. Cleavage of the native SH3 or SH4 loci by the meganuclease yields a substrate for homologous recombination, which may use the donor repair plasmid as a repair matrix. Thus, the frequency with which targeted integration occurs at the SH3 or SH4 loci is indicative of the cleavage efficiency of the genomic SH3 or SH4 target site.
  • Example 4.1: Material and Methods
  • a) Meganuclease Expression Plasmids
  • The meganucleases used in this example are SCOH-SH3-b1-C and SCOH-SH4-b1-C cloned in a mammalian expression vector, resulting in plasmid pCLS2697 and pCLS2705, respectively.
  • b) Donor Repair Plasmids
  • For SH3 gene targeting experiments, the donor plasmid contained:
      • as the left homology arm: a PCR-generated fragment of the SH3 locus (position 6850510 to 6852051 on chromosome 6, NC000006.11). This fragment has a length of 1540 bp;
      • as the right homology arm: a fragment of the SH3 locus (position 6852107 to 6853677 on chromosome 6, NC000006.11). This fragment has a length of 1571 bp.
        For SH4 gene targeting experiments, the donor plasmid contained:
      • as the left homology arm: a PCR-generated fragment of the SH4 locus (position 114972751 to 114974269 on chromosome 7, NC000007.13). This fragment has a length of 1519 bp; and
      • as the right homology arm: a fragment of the SH4 locus (position 114974316 to 114976380 on chromosome 7, NC000007.13). This fragment has a length of 2065 bp.
  • For both SH3 and SH4, the left and right homology arms were inserted upstream (using an AscI site) and downstream (using a SbfI site), respectively, of an exogenous 2.8 kb DNA fragment containing two CMV promoters and a neomycin resistance gene. The resulting plasmids are referred to as pCLS3777 (for SH3) and pCLS3778 (for SH4).
  • c) Sh3 and Sh4 Gene Targeting Experiments
  • Human embryonic kidney 293H cells (Invitrogen) were plated at a density of 1×106 cells per 10 cm dish in complete medium (DMEM supplemented with 2 mM L-glutamine, penicillin (100 UI/ml), streptomycin (100 μg/ml), amphotericin B (Fongizone) (0.25 μg/ml) (Invitrogen-Life Science) and 10% FBS). The next day, cells were transfected with Lipofectamine 2000 transfection reagent (Invitrogen) according to the supplier's protocol. Briefly, 2 μg of the donor plasmid was co-transfected with 3 μg of single-chain meganuclease expression vectors. After 72 hours of incubation at 37° C., cells were trypsinized and plated in complete medium at 10 or 100 cells per well in 96-well plates.
  • Once cells were 80 to 100% confluent, genomic DNA extraction was performed with the ZR-96 genomic DNA kit (Zymo research) according to the supplier's protocol.
  • d) PCR Analysis of Gene Targeting Events
  • The gene targeting frequency was determined by PCR on genomic DNA using the following primers: 5′-CTGTGTGCTATGATCTTGCC-3′ (SH3 GHGF4; SEQ ID NO: 50) and 5′-CCTGTCTCTTGATCAGATCC-3′ (NeoR2; SEQ ID NO: 51) for SH3, and 5′-GTGGCCTCTCAGTCTGTTTA-3′ (SH4 GHGF2; SEQ ID NO: 52) and 5′-AGTCATAGCCGAATAGCCTC-3′ (NeoR5; SEQ ID NO: 53) for SH4. The PCRs result in a 2500 bp (SH3) or a 2268 bp (SH4) gene targeting specific PCR product. The SH3 GHGF4 and SH4 GHGF2 primers are forward primers located upstream of the left homology arms of the donor repair plasmids. The NeoR primers are reverse primers located in the exogenous DNA inserted between the two homology arms of the donor repair plasmid.
  • Example 4.2: Results
  • Human embryonic kidney 293H cells were co-transfected with a plasmid expressing one of the two single-chain SH3 or SH4 meganucleases and the donor repair plasmid pCLS3777 or pCLS3778. As a control for spontaneous recombination, 293H cells were also transfected with the donor repair plasmid alone. The cells were then plated at 10 or 100 cells per well in 96-well microplates. Genomic DNA derived from these cells was analyzed for gene targeting by PCR as described in Material and Methods.
  • In the absence of meganuclease (repair plasmid alone), no PCR positive signal was detected among the 22560 and 18800 cells (for SH3 and SH4, respectively) that were analyzed in pools of 10 or 100 cells.
  • In contrast to this, in the presence of the SH3 meganuclease, 12 positive clones were detected among the 18800 cells analyzed in pools of 100 cells, thereby indicating a frequency of recombination of 0.064%. In the presence of the SH4 meganuclease, 11 positives were detected among the 3760 cells analyzed in pools of 10 cells indicating a frequency of recombination of 0.29%. The results are presented in Table X below. The recombination frequencies indicated here are underestimated because not all plated cells start dividing again. Estimate survival upon plating can thus be estimated to be about 33%. Therefore, frequencies of recombination are probably underestimated by a 3-fold factor.
  • TABLE X
    Gene targeting
    Meganuclease Cells per well PCR+ events frequency
    SH3
    100 12/18800 0.064% 
    SH4
    10 11/3760  0.29%
    SH4
    100 15/18800 0.08%
    None (with SH3 100  0/18800 NA
    repair plasmid)
    None (with SH4 100  0/18800 NA
    repair plasmid)
    NA: not applicable
  • These results demonstrate that the two single chain molecules SCOH-SH3-b1-C and SCOH-SH4-b1-C are capable of inducing high levels of gene targeting at the endogenous SH3 and SH4 locus, respectively.
  • Example 5 Engineering Meganucleases Targeting the SH6 Locus
  • SH6 is a locus comprising a 24 bp non-palindromic target (TTAATACCCCGTACCTAATATTGC, SEQ ID NO: 59) that is present on chromosome 21. SH6 is located in the vicinity of a RIS disclosed in Schwarzwaelder et al. (J Clin Invest 2007:2241-9). The SH6 sequence is not included in any of the CIS described in Deichman et al.
  • Example 5.1. Identification of Meganucleases Cleaving SH6
  • I-CreI variants potentially cleaving the SH6 target sequence in heterodimeric form were constructed by genetic engineering. Pairs of such variants were then co-expressed in yeast. Upon co-expression, one obtains three molecular species, namely two homodimers and one heterodimer. It was then determined whether the heterodimers were capable of cutting the SH6 target sequence of SEQ ID NO: 59.
  • a) Construction of Variants of the I-CreI Meganuclease Cleaving Palindromic Sequences Derived from the SH6 Target Sequence
  • The SH6 sequence is partially a combination of the 10AAT_P (SEQ ID NO: 60), 5CCC_P (SEQ ID NO: 61), 10AAT_P (SEQ ID NO: 60), 5TAG_P (SEQ ID NO: 62) target sequences which are shown on FIG. 9. These sequences are cleaved by mega-nucleases obtained as described in International PCT applications WO 2006/097784 and WO 2006/097853, Arnould et al. (J. Mol. Biol., 2006, 355, 443-458) and Smith et al. (Nucleic Acids Res., 2006). Thus, SH6 should be cleaved by combinatorial variants resulting from these previously identified meganucleases.
  • Two palindromic targets, SH6.3 and SH6.4, were derived from SH6 (FIG. 9). Since SH6.3 and SH6.4 are palindromic, they should be cleaved by homodimeric proteins. Therefore, homodimeric I-CreI variants cleaving either the SH6.3 palindromic target sequence of SEQ ID NO: 63 or the SH6.4 palindromic target sequence of SEQ ID NO: 64 were constructed using methods derived from those described in Chames et al. (Nucleic Acids Res., 2005, 33, e178), Arnould et al. (J. Mol. Biol., 2006, 355, 443-458), Smith et al. (Nucleic Acids Res., 2006, 34, e149) and Arnould et al. (Arnould et al. J Mol Biol. 2007 371:49-65).
  • b) Construction of Target Vector
  • The experimental procedure is as described in Example 1.1., with the exception that an oligonucleotide corresponding to the SH6 target sequence (5′-TGGCATACAAGTTTTTAATACCCCGTACCTAATATTGCCAATCGTCTGTCA-3′ (SEQ ID NO: 65) was used.
  • c) Co-Expression of Variants
  • Yeast DNA was extracted from variants cleaving the SH6.3 and SH6.4 targets in the pCLS542 and pCLS1107 expression vectors using standard protocols and was used to transform E. coli. Transformants were selected on synthetic medium lacking leucine and containing G418.
  • d) Mating of Meganucleases Coexpressing Clones and Screening in Yeast
  • Mating was performed using a colony gridder (QpixII, Genetix). Variants were gridded on nylon filters covering YPD plates, using a low gridding density (4-6 spots/cm2). A second gridding process was performed on the same filters to spot a second layer consisting of different reporter-harboring yeast strains for each target. Membranes were placed on solid agar YPD rich medium, and incubated at 30° C. for one night, to allow mating. Next, filters were transferred to synthetic medium, lacking leucine and tryptophan, adding G418, with galactose (2%) as a carbon source, and incubated for five days at 37° C., to select for diploids carrying the expression and target vectors. After 5 days, filters were placed on solid agarose medium with 0.02% X-Gal in 0.5 M sodium phosphate buffer, pH 7.0, 0.1% SDS, 6% dimethyl formamide (DMF), 7 mM β-mercaptoethanol, 1% agarose, and incubated at 37° C., to monitor β-galactosidase activity. Results were analyzed by scanning and quantification was performed using appropriate software.
  • e) Results
  • Co-expression of ten variants cleaving the SH6.4 target and of two variants cleaving the SH6.3 target resulted in cleavage of the SH6.1 target in all but two cases. These two cases corresponded in which double transformants were not obtained. Functional combinations are summarized in Table XI.
  • TABLE XI
    Amino acids positions and
    residues of the I-CreI variants
    cleaving the SH6.3 target
    44K 68T 70G
    75N 44K 70S 75N
    Amino acids 28Q 40R 44A 70L 75N 96R 111H + +
    positions and 144S
    residues 7R 28Q 40R 44A 70L 75N 85R + +
    of the I-CreI 103T
    variants cleaving 28Q 40R 44A 70L 75N 103S + +
    the SH6.4 target 24F 27V 28Q 40R 44A 70L 75N + +
    99R
    7R 28Q 40R 44A 70L 75N 81T + +
    7R 28Q 40R 44A 70L 75N 77V Not tested +
    7R 28Q 40R 44A 70L 75N 103T + +
    121E 132V 160R
    28Q 40R 44A 70L
    75N Not tested +
    7R 28Q 40R 44A 70L 75N 103T + +
    28Q 34R 40R 44A 70L 75N 81V + +
    103T 108V 160E
    + indicates a functional combination
  • Example 5.2. Validation of SH6 Target Cleavage in an Extrachromosomal Model in CHO Cells
  • I-CreI variants able to efficiently cleave the SH6 target in yeast when forming heterodimers are described hereabove in example 5.1. In order to identify heterodimers displaying maximal cleavage activity for the SH3 target in CHO cells, the efficiency of some of these variants was compared using an extrachromosomal assay in CHO cells. The screen in CHO cells is a single-strand annealing (SSA) based assay where cleavage of the target by the meganucleases induces homologous recombination and expression of a LagoZ reporter gene (a derivative of the bacterial lacZ gene).
  • a) Cloning of SH6 Target in a Vector for CHO Screen
  • The target was cloned as follows: oligonucleotide corresponding to the SH6 target sequence flanked by gateway cloning sequence was ordered from PROLIGO 5′-TGGCATACAAGTTTTTAATACCCCGTACCTAATATTGCCAATCGTCTGTCA-3′ (SEQ ID NO: 65). Double-stranded target DNA, generated by PCR amplification of the single stranded oligonucleotide, was cloned using the Gateway protocol (INVITROGEN) into CHO reporter vector (pCLS1058). Cloned target was verified by sequencing (MILLEGEN).
  • b) Re-Cloning of Meganucleases
  • The ORF of I-CreI variants cleaving the SH6.3 and SH6.4 targets identified in example 5.1 were sub-cloned in pCLS2437. ORFs were amplified by PCR on yeast DNA using the following primers: 5′-AAAAAGCAGGCTGGCGCGCCTACACAGCGGCCTTGCCACCATG-3′ (SEQ ID NO: 66) and 5′-AGAAAGCTGGGTGCTAGCGCTCGAGTTATCAGTCGG-3′ (SEQ ID NO: 67) primers. PCR products were cloned in the CHO expression vector pCLS2437 using the AscI and XhoI for internal fragment replacement. Selected clones resulting from ligation and E. coli transformation steps were verified by sequencing (MILLEGEN).
  • c) Extrachromosomal Assay in Mammalian Cells
  • CHO K1 cells were transfected with Polyfect® transfection reagent according to the supplier's protocol (QIAGEN). 72 hours after transfection, culture medium was removed and 150 μl of lysis/revelation buffer for β-galactosidase liquid assay was added (typically 1 liter of buffer contained: 100 ml of lysis buffer (Tris-HCl 10 mM pH7.5, NaCl 150 mM, Triton X100 0.1%, BSA 0.1 mg/ml, protease inhibitors), 10 ml of Mg 100× buffer (MgCl 2 100 mM, β-mercaptoethanol 35%), 110 ml ONPG 8 mg/ml and 780 ml of sodium phosphate 0.1M pH7.5). After incubation at 37° C., OD was measured at 420 nm. The entire process is performed on an automated Velocity11 BioCel platform. Per assay, 150 ng of target vector was cotransfected with 12.5 ng of each one of both variants (12.5 ng of variant cleaving palindromic SH6.3 target and 12.5 ng of variant cleaving palindromic SH6.4 target).
  • d) Results
  • One couple of variants forming an heterodimeric endonuclease able to cleave SH6 in yeast was chosen for confirmation in CHO using extrachromosomal assay in a transient transfection.
  • The monomer capable of cleaving SH6.3 comprised the following mutations: 44K 70S 75N (referred to as SH6-3-M1-44K 70S 75N) and the monomer capable of cleaving SH6.4 comprised the following mutations: 28Q 40R 44A 70L 75N 96R 111H 144S (referred to as SH6-4-MB-28Q 40R 44A 70L 75N 96R 111 H 144S).
  • Analysis of the efficiencies of cleavage and recombination of the SH6 sequence demonstrates that the tested combination of I-CreI variants was able to transpose its cleavage activity from yeast to CHO cells without additional mutation.
  • Example 5.3. Covalent Assembly as Single Chain and Improvement of Meganucleases Cleaving SH6
  • Co-expression of the cutter described in example 5.1 leads to a high cleavage activity of the SH6 target in yeast. One of them have been validated for SH6 cleavage in a mammalian expression system (example 5.2).
  • The M1×MA SH6 heterodimer gives high cleavage activity in yeast. M1 is a SH6.3 cutter that bears the following mutations in comparison with the I-CreI wild type sequence: 44K 70S 75N. MA is a SH6.4 cutter that bears the following mutations in comparison with the I-CreI wild type sequence: 7R 28Q 40R 44A 70L 75N 103T 121E 132V 160R.
  • Single chain constructs were engineered using the linker RM2 (AAGGSDKYNQALSKYNQALSKYNQALSGGGGS; SEQ ID NO: 15) resulting in the production of the single chain molecule: MA-RM2-M1. During this design step, the G19S mutation was introduced in the C-terminal M1 mutant. In addition, mutations K96E was introduced into the MA mutant and mutations E8K, E61 R into the M1 mutant to create the single chain molecule: MA(K96E)-RM2-MA(E8K E61R) that is called further SCOH-SH6 b1 scaffold.
  • Four additional amino-acid substitutions have been found in previous studies to enhance the activity of I-CreI derivatives: these mutations correspond to the replacement of Phenylalanine 54 with Leucine (F54L), Glutamic acid 80 with Lysine (E80K), Valine 105 with Alanine (V105A) and Isoleucine 132 with Valine (I132V). Some combinations were introduced into the coding sequence of N-terminal and C-terminal protein fragment, and the first batch of resulting proteins were assayed for their ability to induce cleavage of the SH6 target.
  • a) Introduction of Additional Mutations into the SC-OH Single Chain Construct
  • Additional mutations were introduced by use of the QuikChange Multi Site-Directed Mutagenesis Kit from Stratagene/Agilent technologies Inc according to the manufacturer's instructions. A first set of oligonucleotides was used to introduce the mutations in the part of the single chain molecule corresponding to the first monomer. A second set of oligonucleotides was designed to introduce the same mutations specifically in the second part of the single chain molecule corresponding to the second monomer as shown in (see Table XII).
  • TABLE XII
    SEQ ID NO: Name Sequence
    Oligonucleotides used for mutagenesis of the first monomer
    68 F54LFor ACCCAGCGCCGTTGGCTGCTGGACAAACTAGTG
    69 F54LRev CACTAGTTTGTCCAGCAGCCAACGGCGCTGGGT
    70 103T_105AFor AAACAGGCAACCCTGGCTCTGAAAATTATCGAA
    71 103T_105ARev TTCGATAATTTTCAGAGCCAGGGTTGCCTGTTT
    Oligonucleotides used for mutagenesis of the second monomer
    72 F54Lmono2_For CACAAAGAAGGTGGTTGTTGGACAAATTGGTT
    73 F54Lmono2_Rev AACCAATTTGTCCAACAACCACCTTCTTTGTG
    74 E80Kmono2_For TGTCTAAAATTAAGCCTCTTCATAACTTTCTC
    75 E80Kmono2_Rev GAGAAAGTTATGAAGAGGCTTAATTTTAGACA
  • Isolated clones obtained at the term of this process were sequenced to confirm the specific mutation profiles obtained. Profiles of interest were then tested in CHO SSA assay in comparison with the initial construct as described.
  • b) Extrachromosomal Assay in Mammalian Cells
  • CHO K1 cells were transfected as described above. 72 hours after transfection, culture medium was removed and 150 μl of lysis/revelation buffer for β-galactosidase liquid assay was added. After incubation at 37° C., OD was measured at 420 nm. The entire process is performed on an automated Velocity11 BioCel platform.
  • Per assay, 150 ng of target vector was cotransfected with an increasing quantity of variant DNA from 3.12 ng to 25 ng (25 ng of single chain DNA corresponding to 12.5 ng+12.5 ng of heterodimer DNA). Finally, the transfected DNA variant DNA quantity was 3.12 ng, 6.25 ng, 12.5 ng and 25 ng. The total amount of transfected DNA was completed to 175 ng (target DNA, variant DNA, carrier DNA) using empty vector (pCLS0001).
  • c) Results
  • The activity of the SCOH-SH6-b1-C (pCLS2796) and SCOH-SH6-b1-B-(pCLS2928) single chain molecules (see Table XIII) against the SH6 target was monitored using the previously described CHO assay by comparison to the SH6.3-M1×SH6.4-MB forming heterodimer and our internal control SCOH-RAG and I-Sce I meganucleases. All comparisons were done at 3.12 ng, 6.25 ng, 12.5 ng, and 25 ng transfected variant DNA (FIG. 10). The two single chain meganucleases were able to cleave more efficiently the SH6 target than the starting heterodimer. The activity of the best molecule, SCOH-SH6-b1-C, was further improved by introduction additional mutations among those described above in a new bath of meganucleases.
  • TABLE XIII
    Mutations SH6
    on SEQ cleavage
    Mutations on N-terminal C-terminal ID Activity in
    Name segment segment NO: CHO
    SCOH- 7R 28Q 40R 44A 70L 75N 8K 19S 44K 76 +
    SH6-b1-B 96E 103T 121E 132V 160R 61R 70S
    75N
    SCOH- 7R 28Q 40R 44A 70L 75N 8K 19S 44K 77 +
    SH6-b1-C 96E 103T 121E 132V 160R 61R 70S
    75N 132V
  • Additional mutations were further introduced into the single chain scaffold according material and method. The molecules obtained and tested are listed in Table XIV.
  • TABLE XIV
    SH6
    cleavage
    SEQ Activity
    Mutations on N- Mutations on C- ID in
    Name terminal segment terminal segment NO: CHO
    SCOH- 7R 28Q 40R 44A 70L 8K 19S 44K 61R 78 +
    SH6-b1-C 75N 96E 103T 121E 70S 75N 132V
    132V 160R
    QCSH61- 7R 28Q 40R 44A 70L 8K 19S 44K 61R 79 +
    A01 75N 96E 103T 105A 70S 75N 132V
    121E 132V 160R
    QCSH61- 7R 28Q 40R 44A 70L 8K 19S 44K 54L 80 +
    E01 75N 96E 103T 121E 61R 70S 75N
    132V 160R 132V
    QCSH61- 7R 28Q 40R 44A 70L 8K 19S 44K 54L 81 +
    H01a 75N 96E 103T 105A 61R 70S 75N
    121E 132V 160R 80K 132V
    QCSH61- 7E 28Q 40R 44A 70L 8K 19S 44K 54L 83 +
    H01b 75N 96E 103T 105A 61R 70S 75N
    121E 132 V160R 80K 132V
    QCSH61- 7R 28Q 40R 44A 70L 8K 19S 44K 54L 84 +
    H01c 75N 96E 103T 105A 61R 80K 132V
    121E 132V 160R
    QCSH61- 7E 28Q 40R 44A 70L 8K 19S 44K 54L 85 +
    H01d 75N 96E 103T 105A 61R 80K 132V
    121E 132V 160R
    QCSH62- 7R 28Q 40R 44A 54L 8K 19S 44K 61R 82 +
    A02 70L 75N 96E 103T 70S 75N 132V
    121E 132V 160R
  • All the variants were active in the described conditions and shared specific behaviour upon assayed dose depending on the mutation profile they bear (FIG. 10). For example, QCSH61-H01a, b, c, d have a similar profile to our internal standard SCOH-RAG. They are very active molecule even at low doses. All of these variants could be used for SH6 genome targeting.
  • Example 6 Gene Targeting at the Endogenous SH6 Loci in Human Cells
  • To validate the cleavage activity of engineered single-chain SH6 meganucleases, their ability to stimulate homologous recombination at the endogenous human SH6 loci was evaluated. Cells were transfected with mammalian expression plasmids for single chain molecules SCOH-QCSH6-H01 (SEQ ID NO: 81; pCLS3690) or SCOH-QC-SH6-H01-V2-7E-70R75D (SEQ ID NO: 85; pCLS4373) and the donor repair plasmid pCLS3779 (FIG. 13; SEQ ID NO: 279) containing 2.8 kb of exogenous DNA sequence flanked by two sequences, both 1.5 kb in length, homologous to the human SH6 locus. Cleavage of the native SH6 locus by the meganuclease yields a substrate for homologous recombination, which may use the donor repair plasmid containing 2.8 kb of exogenous DNA flanked by homology arms as a repair matrix. Thus, the frequency with which targeted integration occurs at the SH6 locus is indicative of the cleavage efficiency of the genomic SH6 target site.
  • Example 6.1. Materials and Methods
  • a) Meganuclease Expression Plasmids
  • The meganucleases used in this example are SCOH-QCSH6-H01 (SEQ ID NO: 81) or SCOH-QC-SH6-H01-V2-7E-70R75D (SEQ ID NO: 85) cloned in a mammalian expression vector, resulting in plasmid pCLS3690 (FIG. 13) and pCLS4373 respectively.
  • b) Donor Repair Plasmid
  • The donor plasmid contains a PCR generated 1517 bp fragment of the SH6 locus (position 18437771 to 18439287 on chromosome 21, NC000021.8) as the left homology arm and a 1571 bp fragment of the SH6 locus (position 18439343 to 18440846 on chromosome 21, NC000021.8) as the right homology arm. The left and right homology arms were inserted upstream (using an AscI site) and downstream (using a SbfI site), respectively, of an exogenous 2.8 kb DNA fragment containing two CMV promoters and a neomycin resistance gene. The resulting plasmid is pCLS3779 (FIG. 13; SEQ ID NO: 279).
  • c) Sh6 Gene Targeting Experiments
  • Human embryonic kidney 293H cells (Invitrogen) were plated at a density of 1×106 cells per 10 cm dish in complete medium (DMEM supplemented with 2 mM L-glutamine, penicillin (100 UI/ml), streptomycin (100 μg/ml), amphotericin B (Fongizone) (0.25 μg/ml) (Invitrogen-Life Science) and 10% FBS). The next day, cells were transfected with Lipofectamine 2000 transfection reagent (Invitrogen) according to the supplier's protocol. Briefly, 2 μg of the donor plasmid was co-transfected with 3 μg of single-chain meganuclease expression vectors. After 72 hours of incubation at 37° C., cells were trypsinized and plated in complete medium at 10 or 100 cells per well in 96-well plates. Alternatively, after 72 hours of incubation at 37° C., cells were trypsinized and plated in complete medium at 300 cells per dish in 10 cm-dishes. After 2 weeks of incubation at 37° C., individual clonal cellular colonies were picked and plated in complete medium in 96-well plates. Once cells were 80 to 100% confluent, genomic DNA extraction was performed with the ZR-96 genomic DNA kit (Zymo research) according to the supplier's protocol.
  • d) PCR Analysis of Gene Targeting Events
  • The frequency of gene targeting was determined by PCR on genomic DNA using the primers SH6 GHGF3: 5′-CAATGGAGTTTTGGAGCCAC-3′ (SEQ ID NO: 280) and NeoR9: 5′-ATCAGAGCAGCCGATTGTCT-3′ (SEQ ID NO: 281). The PCRs result in a 2300 bp gene targeting specific PCR product (FIG. 14). The SH6 GHGF3 primer is a forward primer located upstream of the left homology arms of the donor repair plasmids. The NeoR9 primer is a reverse primer located in the exogenous DNA inserted between the two homology arms of the donor repair plasmid.
  • Example 6.2. Results
  • Human embryonic kidney 293H cells were co-transfected with 2 vectors: a plasmid expressing one of the two single-chain SH6 meganucleases and the donor repair plasmid pCLS3779 (FIG. 13; SEQ ID NO: 279). As a control for spontaneous recombination, 293H cells were also transfected with the donor repair plasmid alone. The cells were then plated at 10 or 100 cells per well in 96-well microplates or at 300 cells per 10 cm-dishes and 2 weeks later clonal colonies were isolated and plated in 96-well microplates. Genomic DNA derived from these cells was analyzed for gene targeting by PCR as described in Material and Methods. In the absence of meganuclease (repair plasmid alone), 5 PCR positive signals were detected among the 67680 cells analyzed in pools of 10 or 100 cells indicating a frequency of spontaneous of recombination of 0.007%. In contrast, in the presence of the SCOH-QCSH6-H01 (SEQ ID NO: 81; pCLS3690) or SCOH-QC-SH6-H01-V2-7E-70R75D meganucleases (SEQ ID NO: 85; pCLS4773), 177 and 35 positives were detected among the 73320 and 18800 cells analyzed in pools of 10 or 100 cells indicating a frequency of recombination of 0.24% and 0.19% respectively. Results are presented in Table XV. These results demonstrate that the two single chain molecules SCOH-QCSH6-H01 (SEQ ID NO: 81; pCLS3690) and SCOH-QC-SH6-H01-V2-7E-70R75D (SEQ ID NO: 85; pCLS4773) are capable of inducing high levels of gene targeting at the endogenous sh6 locus.
  • TABLE XV
    Frequency of gene targeting events at the sh6 locus
    in human 293H cells
    Cells per Gene targeting
    Meganuclease well PCR+ events frequency
    SCOH-QCSH6-H01 100 151/65800 0.23%
    (SEQ ID NO: 81)
    SCOH-QC-SH6- 100  35/18800 0.19%
    H01-V2-7E-70R75D
    (SEQ ID NO: 85)
    None (with SH6 100  5/56400 0.009% 
    repair plasmid)
    SCOH-QCSH6-H01 10 26/7520 0.35%
    (SEQ ID NO: 81)
    None (with SH6 10  0/11280 NA
    repair plasmid)
    SCOH-QCSH6-H01 monoclonal 9/650 1.38%
    (SEQ ID NO: 81)
    SCOH-QC-SH6- monoclonal 2/116 1.72%
    H01-V2-7E-70R75D
    (SEQ ID NO: 85)
    None (with SH6 monoclonal 0/752 NA
    repair plasmid)
    NA: not applicable
  • Example 7 Transgene Expression after Gene Targeting at the Endogenous Sh6 Loci in Human Cells
  • To validate the capacity of sh6 locus to support transgene expression at sh6 locus cleavage activity of engineered single-chain SH6 meganucleases, gene targeting experiments were conducted with a repair plasmid containing a neomycin-resistance gene expression cassette and the ability of modified cells to grow in Neomycin-containing media was measured. The survival and growth of cells in the presence of Neomycin is dependent on the expression of the neomycin-resistance gene and is therefore indicative of transgene expression at the SH6 locus following targeted integration.
  • Example 7.1. Materials and Methods
  • a) Meganuclease Expression Plasmids
  • The meganuclease used in this example is SCOH-QCSH6-H01 (SEQ ID NO: 81) cloned in a mammalian expression vector, resulting in plasmid pCLS3690.
  • b) Donor Repair Plasmid
  • The donor plasmid contains a PCR generated 1517 bp fragment of the SH6 locus (position 18437771 to 18439287 on chromosome 21, NC000021.8) as the left homology arm and a 1571 bp fragment of the SH6 locus (position 18439343 to 18440846 on chromosome 21, NC000021.8) as the right homology arm. The left and right homology arms were inserted upstream (using an AscI site) and downstream (using a SbfI site), respectively, of an exogenous 2.8 kb DNA fragment containing two CMV promoters and a neomycin resistance gene. The resulting plasmid is pCLS3779 (FIG. 13; SEQ ID NO: 279).
  • c) Sh6 Gene Targeting Experiments
  • Human embryonic kidney 293H cells (Invitrogen) were plated at a density of 1×106 cells per 10 cm dish in complete medium (DMEM supplemented with 2 mM L-glutamine, penicillin (100 UI/ml), streptomycin (100 μg/ml), amphotericin B (Fongizone) (0.25 μg/ml) (Invitrogen-Life Science) and 10% FBS). The next day, cells were transfected with Lipofectamine 2000 transfection reagent (Invitrogen) according to the supplier's protocol. Briefly, 2 μg of the donor plasmid was co-transfected with 3 μg of single-chain meganuclease expression vectors. After 72 hours of incubation at 37° C., cells were trypsinized and plated in complete medium at 300 cells per dish in 10 cm-dishes. After 2 weeks of incubation at 37° C., individual clonal cellular colonies were picked and plated in complete medium in 96-well plates. After one week of incubation at 37° C., cells were trypsined, plated into 2 replicate 96-well plates and incubated at 37° C. Once cells were 80 to 100% confluent, genomic DNA extraction was performed on one of the replicate plate with the ZR-96 genomic DNA kit (Zymo research) according to the supplier's protocol. The other replicate was used to isolate gene-targeted clone and expand them.
  • d) PCR Identification of Gene Targeted Clones
  • Gene targeting was determined by PCR on genomic DNA using the primers SH6 GHGF3: 5′-CAATGGAGTTTTGGAGCCAC-3′ (SEQ ID NO: 280) and NeoR9: 5′-ATCAGAGCAGCCGATTGTCT-3′ (SEQ ID NO: 281). The PCRs result in a 2300 bp gene targeting specific PCR product (FIG. 14). The SH6 GHGF3 primer is a forward primer located upstream of the left homology arms of the donor repair plasmids. The NeoR9 primer is a reverse primer located in the exogenous DNA inserted between the two homology arms of the donor repair plasmid.
  • e) Validation of Targeted Integration by Southern Blot:
  • Genomic DNA from cellular clones was digested with StuI or HindIII restriction enzymes (New England Biolabs), separated by electrophoresis on a 0.8% agarose gela and transferred onto a nitrocellulose membrane. A DNA probe was prepared from 25 ng of a DNA fragment homologous to the Neomycin resistance gene with 32P-radiolabeled dCTP and Rediprime II random prime labelling system (GE Healthcare) according to supplier's protocol and added to the nitrocellulose membrane tha had preincubated in hybridization buffer (NaPi 20 mM, 7% SDS, 1 mM EDTA). After overnight incubation at 65° C., the membrane was washed and exposed to a radiography film. The size of expected bands on the radiograph are 5.3 kb for StuI digestion and 6.8 kb for HindIII digestion (FIG. 15).
  • f) Neomycin-Resistance Test:
  • Cellular clones identified by PCR as targeted at SH6 locus were plated at 300 cells per well in 96-well microplates in the presence of G418 antibiotics (PAA laboratories). After 10 days of incubation at 37° C., viability was measured using Vialight bioassay kit (Lonza) and a Victor luminescence reader (Perkin Elmer) according to supplier's protocol.
  • Example 7.2. Results
  • Human embryonic kidney 293H cells were co-transfected with 2 vectors: a plasmid expressing one of the two single-chain SH6 meganucleases and the donor repair plasmid pCLS3779. The cells were then plated at 300 cells per 10-cm dish and 2 weeks later clonal colonies were isolated and plated in 96-well microplates. Genomic DNA derived from these cells was analyzed for gene targeting by PCR as described in Material and Methods. Genomic DNA was then used to validate targeted integration by southern blot analysis. The clones number 7 and 8 showed bands of the expected size whereas negative control clones number 5 and 6 did not (FIG. 16). Those cellular clones were tested for their ability to survive in the presence of G418 (PAA laboratories). Only clones with targeted integration (number 7 and 8) showed resistance to G418 at concentrations superior to 0.4 mg/ml (FIG. 16). This indicates that targeted integration at sh6 locus can support functional transgene expression.
  • Example 8 Neighboring Gene Expression after Gene Targeting at the Endogenous sh6 Loci in Human Cells
  • To validate the capacity of sh6 locus to support transgene integration without disturbing the expression of neighboring genes, gene targeting experiments were conducted with a repair plasmid containing a 2.8 kb exogenous DNA fragment and cellular clones were identified that contained the targeted integration. The expression of genes upstream and downstream of the sh6 integration site was measured and compared to that of cellular clones that had not undergone targeted integration.
  • Example 8.1. Materials and Methods
  • a) Meganuclease Expression Plasmids
  • The meganucleases used in this example is SCOH-QCSH6-H01 (SEQ ID NO:81) cloned in a mammalian expression vector, resulting in plasmid pCLS3690.
  • b) Donor Repair Plasmid
  • The donor plasmid contains a PCR generated 1517 bp fragment of the SH6 locus (position 18437771 to 18439287 on chromosome 21, NC000021.8) as the left homology arm and a 1571 bp fragment of the SH6 locus (position 18439343 to 18440846 on chromosome 21, NC000021.8) as the right homology arm. The left and right homology arms were inserted upstream (using an AscI site) and downstream (using a SbfI site), respectively, of an exogenous 2.8 kb DNA fragment containing two CMV promoters and a neomycin resistance gene. The resulting plasmid is pCLS3779 (FIG. 13; SEQ ID NO: 279).
  • c) Sh6 Gene Targeting Experiments
  • Human embryonic kidney 293H cells (Invitrogen) were plated at a density of 1×106 cells per 10 cm dish in complete medium (DMEM supplemented with 2 mM L-glutamine, penicillin (100 UI/ml), streptomycin (100 μg/ml), amphotericin B (Fongizone) (0.25 μg/ml) (Invitrogen-Life Science) and 10% FBS). The next day, cells were transfected with Lipofectamine 2000 transfection reagent (Invitrogen) according to the supplier's protocol. Briefly, 2 μg of the donor plasmid was co-transfected with 3 μg of single-chain meganuclease expression vectors. After 72 hours of incubation at 37° C., cells were trypsinized and plated in complete medium at 300 cells per dish in 10 cm-dishes. After 2 weeks of incubation at 37° C., individual clonal cellular colonies were picked and plated in complete medium in 96-well plates. After one week of incubation at 37° C., cells were trypsined, plated into 2 replicate 96-well plates and incubated at 37° C. Once cells were 80 to 100% confluent, genomic DNA extraction was performed on one of the replicate plate with the ZR-96 genomic DNA kit (Zymo research) according to the supplier's protocol. The other replicate was used to isolate gene-targeted clone and expand them.
  • d) PCR Identification of Gene Targeted Clones
  • Gene targeting was determined by PCR on genomic DNA using the primers SH6 GHGF3: 5′-CAATGGAGTTTTGGAGCCAC-3′ (SEQ ID NO: 280) and NeoR9: 5′-ATCAGAGCAGCCGATTGTCT-3′ (SEQ ID NO: 281). The PCRs result in a 2300 bp gene targeting specific PCR product (Figure XX). The SH6 GHGF3 primer (SEQ ID NO: 280) is a forward primer located upstream of the left homology arms of the donor repair plasmids. The NeoR9 primer (SEQ ID NO: 281) is a reverse primer located in the exogenous DNA inserted between the two homology arms of the donor repair plasmid.
  • e) Expression of Genes Upstream and Downstream from Sh6 Locus:
  • Gene expression was measured by quantitative RT-PCR. RNA was isolated from subconfluent cellular clones using RNeasy RNA isolation kit (Qiagen) according to manufacturer's protocol. 3 μg of RNA was used to generate cDNA using Superscript III First-strand kit (Invitrogen). Quantitative PCR was performed on 10 ng of cDNA per 12 μl-reaction, in duplicate samples, using SYBR® Premix Ex Taq™ DNA Polymerase (Lonza) on Stratagene MPX3000 instrument. For each gene, the primers used are listed in the following table:
  • SEQ SEQ
    ID ID
    Gene Forward primer NO: Reverse primer NO:
    HPRT 5′-GCCAGACTTTGTTGGATTTG-3′ 282 5′-CTCTCATCTTAGGCTTTGTATTTTG-3′ 283
    USP25 5′-CAGAGGACATGATGAAGAATTGA-3′ 284 5′-CTCGATCCTCTCCAGATTCG-3′ 285
    NRIP1 5′-GCACTGTGGTCAGACTGCAT-3′ 286 5′-TTCCATCGCAATCAGAGAGA-3′ 287
    CXADR 5′-CTTATCATCTTTTGCTGTCG -3′ 288 5′-TACTGCCGATGTAGCTTCTG-3′ 289
    BTG3 5′-CCAGAAAAACCATCGAAAGG -3′ 290 5′-GGTCACTATACAAGATGCAGC-3′ 291
    C21orf91 5′-AAACACTCTCCTTCTGCCACA-3′ 292 5′-ATGGCCCCTTAATGATTTGG-3′ 293
  • The threshold cycles (Ct) were determined with Stratagene software on fluorescence (dRn) after normalization by the ROX reference dye. The intensity of gene expression was calculated using the formula 2Ct(HPRT)-Ct(Gene), the expression of the housekeeping gene HPRT being used as an internal normalizing factor.
  • Example 8.2. Results
  • Human embryonic kidney 293H cells were co-transfected with 2 vectors: a plasmid expressing one of the three single-chain SH6 meganucleases and the donor repair plasmid pCLS3779. The cells were then plated at 300 cells per 10-cm dish and 2 weeks later clonal colonies were isolated and plated in 96-well microplates. Genomic DNA derived from these cells was analyzed for gene targeting by PCR as described in Material and Methods. RNA was isolated from clones showing targeted integration and negative controls. Quantitative RT-PCR was performed to measure expression of genes surrounding the locus of targeted integration. The data are presented in FIG. 17 where the average intensity of duplicate samples is shown for 3 individual targeted clones (KI) and 3 individual non-targeted clones (WT) after normalization with the housekeeping gene HPRT. No significant difference is observed for each of the 5 genes measured, indicating that targeted integration at the sh6 locus has no consequence on the expression of neighboring genes.
  • Example 9 Mutagenesis at Endogenous Safe Harbor Loci in Human Cells
  • To validate the cleavage activity of engineered single-chain Safe Harbor meganucleases, their ability to stimulate mutagenesis at endogenous human safe harbor loci was evaluated. Cells were transfected with mammalian expression plasmids for single chain molecules. Cleavage of a native safe harbor locus by the meganuclease yields a substrate for non-homologous end joining, which is an error-prone process and can result in small insertion or deletions at the meganuclease target site. Thus, the frequency at which mutations occur at an endogenous safe harbor locus is indicative of the cleavage efficiency of the genomic target site by the meganuclease.
  • Example 9.1. Materials and Methods
  • a) Meganuclease Expression Plasmids
  • The coding sequences for the meganucleases used in this example were cloned in a mammalian expression vector, resulting in the plasmids listed in table XVI.
  • TABLE XVI
    Meganucleases targeting safe harbour sequences
    locus
    targeted meganuclease plasmid SEQ ID NO
    sh3 SCOH-SH3-b1-C pCLS2697 31
    sh4 SCOH-SH4-b1-C pCLS2705 39
    sh6 QCSH61-H01 pCLS3690 81
    sh6 QC-SH6- pCLS4373 85
    H01_V2_7E_70R75D
    sh6 QC-SH6-H01_7E pCLS4377 83
    sh6 SCOH-SH6-b12-G2_BQY pCLS6567 294
    sh6 SCOH-SH6-b11-G2.2_BQY pCLS6570 295
    sh8 SCOH-SH8 pCLS3894 88
    sh13 SCOH-SH13 pCLS3897 90
    sh18 SCOH-SH18-b11-C.2 pCLS5519 128
    sh19 SCOH-SH19 pCLS3899 91
    sh31 SCOH-SH31.2 pCLS4076 132
    sh39 SCOH-SH39-b11-C pCLS6038 133
    sh41 SCOH-SH41-b11-C pCLS5187 135
    sh42 SCOH-SH42-b11-C pCLS5549 137
    sh43 SCOH-SH43-b12-C pCLS5595 140
    sh44 SCOH-SH44-b11-C pCLS5868 141
    sh52 SCOH-SH52-b12-C pCLS5871 144
  • b) Safe Harbor Locus Mutagenesis Experiments
  • Human embryonic kidney 293H cells (Invitrogen) were plated at a density of 1×106 cells per 10 cm dish in complete medium (DMEM supplemented with 2 mM L-glutamine, penicillin (100 UI/ml), streptomycin (100 μg/ml), amphotericin B (Fongizone) (0.25 μg/ml) (Invitrogen-Life Science) and 10% FBS). The next day, cells were transfected with 3 μg of single-chain meganuclease expression vector using Lipofectamine 2000 transfection reagent (Invitrogen) according to the supplier's protocol. After 2 to 6 days of incubation at 37° C., cells were trypsinized and genomic DNA extraction was performed with the DNeasy blood and tissue kit (Qiagen) according to the supplier's protocol.
  • c) Deep Sequencing Analysis of Mutagenesis Events
  • The frequency of mutagenesis was determined by deep sequencing analysis. Oligonucleotides were designed for PCR amplification of a DNA fragment surrounding each safe harbour target and are listed in table XVII.
  • TABLE XVII
    PCR primers for mutagenesis analysis of safe harbour targets
    locus SEQ ID SEQ ID
    targeted forward primer NO reverse primer NO
    sh3 5′-TGGGGGTCTTACTCTGTTTC 296 5′-AGGAGAGTCCTTCTTTGGCCAA 297
    CCAG-3′ T-3′
    sh4 5′-GAGTGATAGCATAATGAAAA 298 5′-CTCACCATAAGTCAACTGTCTCA  299
    CCCA-3′ G-3′
    sh6 5′-TCTTTGTGTTTCCAAAGAGT  300 5′-GAATGGTCTGAAAATGGAGAGG 301
    TCCTTTGGCTTTCAC-3′ TTAAATGAGATTT-3′
    sh8 5′-ACTAAATATGTTAATTGTGT  302 5′-ATTGCTACTTCATTTGTTATGTT  303
    GTATACAGTTTTTGT-3′ AACTATGACATG-3′
    sh13 5′-TTTTTGTGGGTCCACAGTAG 304 5′-CAGTTGAACTCATGGATGTAGA 305
    GTGTATATATTTATGG-3′ GAGTAGAAGAATG-3′
    sh18 5′-GACCTGAAGCTCAGGTACT 306 5′-AGTGGTGGTAGGCAGGACAT-3′ 307
    T-3′
    sh19 5′-CTTAGGTAAACCTCAAAACA 308 5′-CTGCTAGAGCCCGTAATGTTTCA 309
    ACAAGAGAGGAGCAA-3′ ATCATAGTTATT-3′
    sh31 5′-TTCAGGTTAGGTGACCTTCA 310 5′-AAGACCAGGCTGGGCAACCATAG 311
    AACT-3′ C-3′
    sh39 5′-GAATAATGGAATAAACCCAG 312 5′-GTGTTCAAGGAAAATGGAGTGA 313
    AGAGAAACAGAG-3′ TATTAGGAAT-3′
    sh41 5′-GGAGATATCATTAAAAGAGG 314 5′-ATTACAATAGCCTTAGGAAACTA  315
    CATT-3′ G-3′
    sh42 5′-GAGTCACAGCCACCTTACAT 316 5′-AAGTAGAACACATTCCTATTTCC  317
    TTTACTTTTC-3′ ATTAAGT-3′
    sh43  5′-ATTAAGTACAAAATTTGGTCC 318 5′-AAAGTTGATTCATCTGAAACAT 319
    AAT-3′ G-3′
    sh44  5′-GCAGCGATCCATGGTGGAG 320 5′-TAACACAGGCTCATGTAGGT-3′ 321
    A-3′
    sh52   5′-ATGTTATTCGAGGACCCACT- 322 5′-GTGACAACTCTGCTAGAAGA-3′ 323
    3′
  • Nucleotides were added to obtain a fragment flanked by specific adaptator sequences (5′-CCATCTCATCCCTGCGTGTCTCCGACTCAG-3′; SEQ ID NO 324) and (5′-CCTATCCCCTGTGTGCCTTGGCAGTCTCAG-3′; SEQ ID NO 325) provided by the company offering sequencing service (GATC Biotech AG, Germany) on the 454 sequencing system (454 Life Sciences). An average of 18,000 sequences was obtained from pools of 2 to 3 amplicons (500 ng each). After sequencing, different samples were identified based on barcode sequences introduced in the first of the above adaptators.
  • Example 9.2. Results
  • Human embryonic kidney 293H cells were transfected with a plasmid expressing a single-chain safe harbor meganuclease. After 2 to 6 days of incubation at 37° C., genomic DNA was isolated and PCR was used to amplify the genomic sequence surrounding the meganuclease target site. Sequences were then analyzed for the presence of insertions or deletions events (InDel) in the cleavage site of each safe harbor target. Results are summarized in table XVIII.
  • TABLE XVIII
    Mutagenesis by meganucleases targeting safe harbor loci:
    locus Cleaved by meganucleases
    targeted of SEQ ID NO: Plasmids % InDels
    sh3 31 2697 0.8
    sh4 39 2705 0.2
    sh6 81 3690 0.6
    85 4373 3.5
    83 4377 1.5
    294 6567 1
    295 6570 3
    sh8 88 3894 0.5
    sh13 90 3897 1.5
    sh18 128 5519 1.2
    sh19 91 3899 0.9
    sh31 132 4076 5
    sh39 133 6038 1.5
    sh41 135 5187 0.4
    sh42 137 5549 0.7
    sh43 140 5595 0.4
    sh44 141 5868 3.6
    sh52 144 5871 3.2
  • Example 10 Conclusion
  • In conclusion, Examples 1, 2, 3 and 5 demonstrate that both I-CreI heterodimeric proteins and single-chain meganucleases capable of cleaving the SH3, the SH4 and the SH6 loci can be obtained. Moreover, these endonucleases are capable of cleaving these loci with a strong cleavage activity.
  • Example 4 demonstrates that single-chain meganucleases capable of cleaving the SH3 and the SH4 loci allow efficiently inserting a transgene into a target site of a human cell.
  • These endonucleases can thus advantageously be used to insert a transgene into the SH3, the SH4 loci or the SH6 loci of an individual.
  • Example 6 demonstrates that at least two single chain molecules according to the invention are capable of inducing high levels of gene targeting at an endogenous sh6 locus.
  • Example 7 demonstrates that targeted integration a locus can support functional transgene expression.
  • Example 8 demonstrates that a targeted integration at a locus does not substantially modify expression of five genes located in the vicinity of the target sequence.
  • Example 9 demonstrates mutagenesis frequencies for different meganucleases targeting safe harbor sequences, which are indicative of the cleavage efficiency of the genomic target site by said meganucleases.

Claims (36)

1. A variant endonuclease capable of cleaving a target sequence for use in inserting a transgene into a the genome of an individual, wherein
i. said genome comprises a locus comprising said target sequence; and
ii. said target sequence is located at a distance of at most 200 kb from a retroviral insertion site (RIS), wherein said RIS is neither associated with cancer nor with abnormal cell proliferation.
2. The endonuclease according to claim 1, wherein insertion of said transgene does not substantially modify expression of genes located in the vicinity of the target sequence.
3. The endonuclease according to claim 1, wherein said target sequence is located at a distance of at least 100 kb from the nearest genes.
4. The endonuclease according to claim 1, wherein said endonuclease is a homing endonuclease.
5. The endonuclease according to claim 1, wherein said endonuclease is capable of cleaving a target sequence located within a locus selected from the group consisting of the SH6 locus on human chromosome 21q21.1, the SH3 locus on human chromosome 6p25.1, the SH4 locus on human chromosome 7q31.2, the SH12 locus on human chromosome 13q34, the SH13 locus on human chromosome 3p12.2, the SH19 locus on human chromosome 22, the SH20 locus on human chromosome 12q21.2, the SH21 locus on human chromosome 3p24.1, the SH33 locus on human chromosome 6p12.2, the SH7 locus on human chromosome 2p16.1, the SH8 locus on human chromosome 5, the SH18 locus, the SH31 locus, the SH38 locus, the SH39 locus, the SH41 locus, the SH42 locus, the SH43 locus, the SH44 locus, the SH45 locus, the SH46 locus, the SH47 locus, the SH48 locus, the SH49 locus, the SH50 locus, the SH51 locus, the SH52 locus, the SH70 locus, the SH71 locus, the SH72 locus, the SH73 locus, the SH74 locus, the SH75 locus, the SH101 locus, the SH106 locus, the SH107 locus, the SH102 locus, the SH105 locus, the SH103 locus, the SH104 locus, the SH113 locus, the SH109 locus, the SH112 locus, the SH108 locus, the SH110 locus, the SH114 locus, the SH116 locus, the SH111 locus, the SH115 locus, the SH121 locus, the SH120 locus, the SH122 locus, the SH117 locus, the SH118 locus, the SH119 locus, the SH123 locus, the SH126 locus, the SH128 locus, the SH129 locus, the SH124 locus, the SH131 locus, the SH125 locus, the SH127 locus, the SH130 locus, the SH11 locus, the SH17 locus, the SH23 locus, the SH34 locus, the SH40 locus, the SH53 locus, the SH54 locus, the SH55 locus, the SH56 locus, the SH57 locus, the SH58 locus, the SH59 locus, the SH60 locus, the SH61 locus, the SH62 locus, the SH65 locus, the SH67 locus, the SH68 locus and the SH69 locus.
6. A variant dimeric I-CreI protein comprising two monomers that each comprises a sequence at least 80% identical to SEQ ID NO: 1 or SEQ ID NO: 42, wherein:
i. said dimeric I-CreI protein is capable of cleaving a target sequence located within a locus of an individual, said target sequence being located at a distance of at most 200 kb from a retroviral insertion site (RIS), and said RIS being neither associated with cancer nor with abnormal cell proliferation; and
ii. said target sequence does not comprise a sequence of SEQ ID NO: 4.
7. The dimeric I-CreI protein according to claim 6, wherein said dimeric I-CreI protein is capable of cleaving a target sequence located within a locus selected from the group consisting of the SH6 locus on human chromosome 21q21.1, the SH3 locus on human chromosome 6p25.1, the SH4 locus on human chromosome 7q31.2, the SH12 locus on human chromosome 13q34, the SH13 locus on human chromosome 3p12.2, the SH19 locus on human chromosome 22, the SH20 locus on human chromosome 12q21.2, the SH21 locus on human chromosome 3p24.1, the SH33 locus on human chromosome 6p12.2, the SH7 locus on human chromosome 2p16.1, the SH8 locus on human chromosome 5, the SH18 locus, the SH31 locus, the SH38 locus, the SH39 locus, the SH41 locus, the SH42 locus, the SH43 locus, the SH44 locus, the SH45 locus, the SH46 locus, the SH47 locus, the SH48 locus, the SH49 locus, the SH50 locus, the SH51 locus, the SH52 locus, the SH70 locus, the SH71 locus, the SH72 locus, the SH73 locus, the SH74 locus, the SH75 locus, the SH101 locus, the SH106 locus, the SH107 locus, the SH102 locus, the SH105 locus, the SH103 locus, the SH104 locus, the SH113 locus, the SH109 locus, the SH112 locus, the SH108 locus, the SH110 locus, the SH114 locus, the SH116 locus, the SH111 locus, the SH115 locus, the SH121 locus, the SH120 locus, the SH122 locus, the SH117 locus, the SH118 locus, the SH119 locus, the SH123 locus, the SH126 locus, the SH128 locus, the SH129 locus, the SH124 locus, the SH131 locus, the SH125 locus, the SH127 locus, the SH130 locus, the SH11 locus, the SH17 locus, the SH23 locus, the SH34 locus, the SH40 locus, the SH53 locus, the SH54 locus, the SH55 locus, the SH56 locus, the SH57 locus, the SH58 locus, the SH59 locus, the SH60 locus, the SH61 locus, the SH62 locus, the SH65 locus, the SH67 locus, the SH68 locus and the SH69 locus.
8. The dimeric I-CreI protein according to claim 6, wherein said dimeric I-CreI protein is capable of cleaving a target sequence located within the SH6 locus on human chromosome 21 q21.1,
9. The dimeric I-CreI protein according to claim 8, wherein said target sequence comprises the sequence of SEQ ID NO: 59.
10. A fusion protein comprising the monomers of the dimeric I-CreI protein as defined in claim 6.
11. The fusion protein according to claim 10, wherein said fusion protein comprises a sequence selected from the group consisting of SEQ ID Nos. 81, 82-85, 294, 295, 76-80, 25-40, 86-96, 127-150, 182-213, 235-270 and 275-278.
12. A nucleic acid encoding:
a) a variant endonuclease capable of cleaving a target sequence for use in inserting a transgene into a genome of an individual, wherein:
i) said genome comprises a locus comprising said target sequence; and
ii) said target sequence is located at a distance of at most 200 kb from a retroviral insertion site (RIS), wherein said RIS is neither associated with cancer nor with abnormal cell proliferation; or
b) a variant dimeric I-CreI protein according to claim 6.
13. An expression vector comprising the nucleic acid as defined in claim 12.
14. The expression vector according to claim 13, further comprising a targeting construct comprising a transgene and two sequences homologous to the genomic sequence flanking a target sequence recognized by the endonuclease.
15. A combination of:
an expression vector as defined in claim 13; and
a vector comprising a targeting construct comprising a transgene and two sequences homologous to the genomic sequence of a target sequence recognized by the endonuclease.
16. A pharmaceutical composition comprising the expression vector as defined in claim 14, and a pharmaceutically acceptable carrier.
17. A method of inserting a transgene into a genome of a cell, tissue or non-human animal comprising administering to said cell, tissue or non-human animal an endonuclease according to claim 1.
18. A method of making a non-human animal model of a hereditary disorder comprising the method of claim 17.
19. A method of producing a recombinant protein comprising the method of claim 17.
20. A method for obtaining an endonuclease suitable for inserting a transgene into the genome of an individual, comprising the step of:
a) selecting, within the genome of said individual, a retroviral insertion site (RIS) that is neither associated with cancer nor with abnormal cell proliferation;
b) defining a genomic region extending 200 kb upstream and 200 kb downstream of said RIS; and
c) identifying a wild-type endonuclease or constructing a variant endonuclease capable of cleaving a target sequence located within said genomic region.
21. A pharmaceutical composition comprising the combination as defined in claim 15 and a pharmaceutically acceptable carrier.
22. A method of inserting a transgene into a genome of a cell, tissue or non-human animal comprising administering to said cell, tissue or non-human animal a variant according to claim 6.
23. A method of inserting a transgene into a genome of a cell, tissue or non-human animal comprising administering to said cell, tissue or non-human animal a nucleic acid according to claim 12.
24. A method of inserting a transgene into a genome of a cell, tissue or non-human animal comprising administering to said cell, tissue or non-human animal an expression vector according to claim 13.
25. A method of inserting a transgene into a genome of a cell, tissue or non-human animal comprising administering to said cell, tissue or non-human animal an expression vector according to claim 14.
26. A method of inserting a transgene into a genome of a cell, tissue or non-human animal comprising administering to said cell, tissue or non-human animal a combination according to claim 15.
27. A method of making a non-human animal model of a hereditary disorder comprising the method of claim 22.
28. A method of making a non-human animal model of a hereditary disorder comprising the method of claim 23.
29. A method of making a non-human animal model of a hereditary disorder comprising the method of claim 24.
30. A method of making a non-human animal model of a hereditary disorder comprising the method of claim 25.
31. A method of making a non-human animal model of a hereditary disorder comprising the method of claim 26.
32. A method of producing a recombinant protein comprising the method of claim 22.
33. A method of producing a recombinant protein comprising the method of claim 23.
34. A method of producing a recombinant protein comprising the method of claim 24.
35. A method of producing a recombinant protein comprising the method of claim 25.
36. A method of producing a recombinant protein comprising the method of claim 26.
US13/702,066 2010-02-26 2011-02-28 Use of endonucleases for inserting transgenes into safe harbor loci Abandoned US20130227715A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US13/702,066 US20130227715A1 (en) 2010-02-26 2011-02-28 Use of endonucleases for inserting transgenes into safe harbor loci

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
US30850910P 2010-02-26 2010-02-26
EP10305202.3 2010-02-26
EP10305202 2010-02-26
US13/702,066 US20130227715A1 (en) 2010-02-26 2011-02-28 Use of endonucleases for inserting transgenes into safe harbor loci
PCT/EP2011/052916 WO2011104382A1 (en) 2010-02-26 2011-02-28 Use of endonucleases for inserting transgenes into safe harbor loci

Publications (1)

Publication Number Publication Date
US20130227715A1 true US20130227715A1 (en) 2013-08-29

Family

ID=44063552

Family Applications (2)

Application Number Title Priority Date Filing Date
US13/036,343 Expired - Fee Related US9290748B2 (en) 2010-02-26 2011-02-28 Use of endonucleases for inserting transgenes into safe harbor loci
US13/702,066 Abandoned US20130227715A1 (en) 2010-02-26 2011-02-28 Use of endonucleases for inserting transgenes into safe harbor loci

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US13/036,343 Expired - Fee Related US9290748B2 (en) 2010-02-26 2011-02-28 Use of endonucleases for inserting transgenes into safe harbor loci

Country Status (7)

Country Link
US (2) US9290748B2 (en)
EP (1) EP2539445B1 (en)
JP (1) JP2013520190A (en)
CN (1) CN103097527A (en)
AU (1) AU2011219716A1 (en)
CA (1) CA2791116A1 (en)
WO (1) WO2011104382A1 (en)

Cited By (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20110173710A1 (en) * 2007-12-13 2011-07-14 Cellectis Chimeric meganuclease enzymes and uses thereof
US20110207199A1 (en) * 2007-08-03 2011-08-25 Cellectis Novel method to generate meganucleases with altered characteristics
US20110225664A1 (en) * 2008-09-08 2011-09-15 Cellectis Meganuclease variants cleaving a dna target sequence from a glutamine synthetase gene and uses thereof
US8802437B2 (en) 2009-09-24 2014-08-12 Cellectis Meganuclease reagents of uses thereof for treating genetic diseases caused by frame shift/non sense mutations
US9044492B2 (en) 2011-02-04 2015-06-02 Cellectis Sa Method for modulating the efficiency of double-strand break-induced mutagenesis
US9365864B2 (en) 2008-10-23 2016-06-14 Cellectis Meganuclease recombination system
WO2019169233A1 (en) * 2018-03-02 2019-09-06 Generation Bio Co. Closed-ended dna (cedna) vectors for insertion of transgenes at genomic safe harbors (gsh) in humans and murine genomes

Families Citing this family (21)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2012017329A2 (en) * 2010-08-02 2012-02-09 Cellectis S.A. Method for targeted genomic events in algae
US20140115726A1 (en) * 2011-04-05 2014-04-24 Cellectis New tale-protein scaffolds and uses thereof
AU2013355327A1 (en) * 2012-12-05 2015-06-11 Sangamo Therapeutics, Inc. Methods and compositions for regulation of metabolic disorders
WO2014183066A2 (en) 2013-05-10 2014-11-13 Whitehead Institute For Biomedical Research Protein modification of living cells using sortase
WO2014182700A1 (en) * 2013-05-10 2014-11-13 Sangamo Biosciences, Inc. Delivery methods and compositions for nuclease-mediated genome engineering
KR20160021812A (en) * 2013-06-19 2016-02-26 시그마-알드리치 컴퍼니., 엘엘씨 Targeted integration
WO2015073867A1 (en) * 2013-11-15 2015-05-21 The United States Of America, As Represented By The Secretary, Department Of Health & Human Services Engineering neural stem cells using homologous recombination
US10370680B2 (en) 2014-02-24 2019-08-06 Sangamo Therapeutics, Inc. Method of treating factor IX deficiency using nuclease-mediated targeted integration
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
CN107429261A (en) 2014-12-15 2017-12-01 应用干细胞有限公司 Produce single or multiplexed reporters method and compositions rapidly in cell
CA3138329A1 (en) 2019-05-10 2020-11-19 Basf Se Regulatory nucleic acid molecules for enhancing gene expression in plants
EP4028530A1 (en) 2019-09-12 2022-07-20 Basf Se Regulatory nucleic acid molecules for enhancing gene expression in plants
WO2021069387A1 (en) 2019-10-07 2021-04-15 Basf Se Regulatory nucleic acid molecules for enhancing gene expression in plants
CA3159805A1 (en) 2019-12-03 2021-06-10 Frank Meulewaeter Regulatory nucleic acid molecules for enhancing gene expression in plants
US11332744B1 (en) 2020-10-26 2022-05-17 Arsenal Biosciences, Inc. Safe harbor loci
EP4301853A1 (en) * 2021-03-02 2024-01-10 President and Fellows of Harvard College Compositions and methods for human genomic safe harbor site integration
CN115485367B (en) * 2021-04-16 2023-09-12 杭州启函生物科技有限公司 Safe harbor locus for cell engineering
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
WO2023141602A2 (en) 2022-01-21 2023-07-27 Renagade Therapeutics Management Inc. Engineered retrons and methods of use
WO2024044723A1 (en) 2022-08-25 2024-02-29 Renagade Therapeutics Management Inc. Engineered retrons and methods of use
WO2024054006A1 (en) * 2022-09-05 2024-03-14 주식회사 에피바이오텍 Novel genomic safe harbor and use thereof

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2007014275A2 (en) * 2005-07-26 2007-02-01 Sangamo Biosciences, Inc. Targeted integration and expression of exogenous nucleic acid sequences

Family Cites Families (24)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
FR2805821B1 (en) 2000-03-01 2004-01-16 Diatos AMINO ACID SEQUENCES TO FACILITATE THE PENETRATION OF A SUBSTANCE OF INTEREST WITHIN CELLS AND / OR CELL CORES
US7049286B2 (en) 2001-08-30 2006-05-23 Diatos, S.A. Insulin conjugates and methods of use thereof
US8206965B2 (en) 2002-03-15 2012-06-26 Cellectis S.A. Hybrid and single chain meganucleases and use thereof
WO2009095742A1 (en) 2008-01-31 2009-08-06 Cellectis New i-crei derived single-chain meganuclease and uses thereof
US20060206949A1 (en) 2003-01-28 2006-09-14 Sylvain Arnould Custom-made meganuclease and use thereof
BRPI0413521A (en) 2003-08-14 2006-10-10 Diatos amino acid sequences that facilitate the penetration of a substance of interest into cells and / or cell nuclei
US7402436B2 (en) * 2004-05-03 2008-07-22 City Of Hope Lentiviral vectors for site-specific gene insertion
ATE466933T1 (en) 2005-03-15 2010-05-15 Cellectis I-CREI MEGANUCLEASE VARIANTS WITH MODIFIED SPECIFICITY AND METHOD FOR THEIR PRODUCTION AND USE
WO2006097784A1 (en) 2005-03-15 2006-09-21 Cellectis I-crei meganuclease variants with modified specificity, method of preparation and uses thereof
CA2626262C (en) 2005-10-18 2015-09-08 Homme W. Hellinga Rationally-designed meganucleases with altered sequence specificity and dna-binding affinity
WO2007049095A1 (en) * 2005-10-25 2007-05-03 Cellectis Laglidadg homing endonuclease variants having mutations in two functional subdomains and use thereof
CA2633063A1 (en) 2005-12-16 2007-06-21 Diatos Cell penetrating peptide conjugates for delivering of nucleic acids intocells
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
US20100146651A1 (en) * 2006-11-14 2010-06-10 Cellectis Meganuclease variants cleaving a dna target sequence from the hprt gene and uses thereof
WO2008093152A1 (en) 2007-02-01 2008-08-07 Cellectis Obligate heterodimer meganucleases and uses thereof
WO2008149176A1 (en) * 2007-06-06 2008-12-11 Cellectis Meganuclease variants cleaving a dna target sequence from the mouse rosa26 locus and uses thereof
CN101784658A (en) 2007-06-06 2010-07-21 赛莱克蒂斯公司 Enhancing is from the method for the nicking activity of the meganuclease of I-CreI
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
WO2009114321A2 (en) 2008-03-11 2009-09-17 Precision Biosciencs, Inc. Rationally-designed meganucleases for maize genome engineering
JP2011519558A (en) 2008-04-28 2011-07-14 プレシジョン バイオサイエンシズ,インク. A rationally designed fusion molecule of DNA-binding protein and effector domain
EP2329017A2 (en) * 2008-08-04 2011-06-08 Cellectis Novel method to generate meganucleases with altered characteristics
WO2010026443A1 (en) * 2008-09-08 2010-03-11 Cellectis Meganuclease variants cleaving a dna target sequence from a glutamine synthetase gene and uses thereof

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2007014275A2 (en) * 2005-07-26 2007-02-01 Sangamo Biosciences, Inc. Targeted integration and expression of exogenous nucleic acid sequences

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
Chalberg TW et al. Integration Specificity of Phage phiC31 Integrase in the Human Genome. Journal of Molecular Biology. 2006. 357, 28-48. *

Cited By (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20110207199A1 (en) * 2007-08-03 2011-08-25 Cellectis Novel method to generate meganucleases with altered characteristics
US20110173710A1 (en) * 2007-12-13 2011-07-14 Cellectis Chimeric meganuclease enzymes and uses thereof
US20110225664A1 (en) * 2008-09-08 2011-09-15 Cellectis Meganuclease variants cleaving a dna target sequence from a glutamine synthetase gene and uses thereof
US9273296B2 (en) 2008-09-08 2016-03-01 Cellectis Meganuclease variants cleaving a DNA target sequence from a glutamine synthetase gene and uses thereof
US9365864B2 (en) 2008-10-23 2016-06-14 Cellectis Meganuclease recombination system
US8802437B2 (en) 2009-09-24 2014-08-12 Cellectis Meganuclease reagents of uses thereof for treating genetic diseases caused by frame shift/non sense mutations
US9044492B2 (en) 2011-02-04 2015-06-02 Cellectis Sa Method for modulating the efficiency of double-strand break-induced mutagenesis
WO2019169233A1 (en) * 2018-03-02 2019-09-06 Generation Bio Co. Closed-ended dna (cedna) vectors for insertion of transgenes at genomic safe harbors (gsh) in humans and murine genomes

Also Published As

Publication number Publication date
US9290748B2 (en) 2016-03-22
CN103097527A (en) 2013-05-08
US20110239319A1 (en) 2011-09-29
WO2011104382A1 (en) 2011-09-01
JP2013520190A (en) 2013-06-06
AU2011219716A1 (en) 2012-09-27
EP2539445A1 (en) 2013-01-02
CA2791116A1 (en) 2011-09-01
EP2539445B1 (en) 2018-03-21

Similar Documents

Publication Publication Date Title
US9290748B2 (en) Use of endonucleases for inserting transgenes into safe harbor loci
EP2121004B1 (en) Obligate heterodimer meganucleases and uses thereof
JP6695239B2 (en) Method for producing compact TALE-nuclease and use thereof
US10006052B2 (en) Laglidadg homing endonuclease cleaving the C-C chemokine receptor type-5 (CCR5) gene and uses thereof
US20140112904A9 (en) Method for enhancing the cleavage activity of i-crei derived meganucleases
US20110158974A1 (en) Heterodimeric Meganucleases and Use Thereof
US20130059387A1 (en) Meganuclease variants cleaving a dna target sequence from the hprt gene and uses thereof
US20130183282A1 (en) Meganuclease variants cleaving a DNA target sequence from the rhodopsin gene and uses thereof
US20130045539A1 (en) Meganuclease recombination system
JP2013544082A (en) Method for increasing the efficiency of double-strand break-induced mutagenesis
JP2010524500A (en) Targeted integration into the PPP1R12C locus
WO2009001159A1 (en) Method for enhancing the cleavage activity of i-crei derived meganucleases
AU2003298574A1 (en) Use of chimeric nucleases to stimulate gene targeting
WO2012007848A2 (en) Meganuclease variants cleaving a dna target sequence in the was gene and uses thereof

Legal Events

Date Code Title Description
AS Assignment

Owner name: CELLECTIS, FRANCE

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:DANOS, OLIVIER;DUCLERT, AYMERIC;REEL/FRAME:030311/0622

Effective date: 20130122

Owner name: CELLECTIS, FRANCE

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:DANOS, OLIVIER;DUCLERT, AYMERIC;REEL/FRAME:030311/0951

Effective date: 20130122

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION