US20140115726A1 - New tale-protein scaffolds and uses thereof - Google Patents

New tale-protein scaffolds and uses thereof Download PDF

Info

Publication number
US20140115726A1
US20140115726A1 US14/009,509 US201214009509A US2014115726A1 US 20140115726 A1 US20140115726 A1 US 20140115726A1 US 201214009509 A US201214009509 A US 201214009509A US 2014115726 A1 US2014115726 A1 US 2014115726A1
Authority
US
United States
Prior art keywords
domain
seq
recognizing
terminus
chimeric protein
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
US14/009,509
Other languages
English (en)
Inventor
Philippe Duchateau
Alexandre Juillerat
Julien Valton
Claudia Bertonati
Jean-Charles Epinat
George H. Silva
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 US14/009,509 priority Critical patent/US20140115726A1/en
Assigned to CELLECTIS, S.A. reassignment CELLECTIS, S.A. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: DUCHATEAU, PHILIPPE, JUILLERAT, ALEXANDRE, EPINAT, JEAN-CHARLES, BERTONATI, Claudia, VALTON, JULIEN, SILVA, GEORGE H.
Publication of US20140115726A1 publication Critical patent/US20140115726A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N9/00Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
    • C12N9/14Hydrolases (3)
    • C12N9/16Hydrolases (3) acting on ester bonds (3.1)
    • 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
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/195Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from bacteria
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/46Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • C07K14/47Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • C07K14/4701Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals not used
    • C07K14/4702Regulators; Modulating activity
    • C07K14/4703Inhibitors; Suppressors
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/10Processes for the isolation, preparation or purification of DNA or RNA
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • 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
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/10Cells modified by introduction of foreign genetic material
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N9/00Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
    • C12N9/14Hydrolases (3)
    • C12N9/16Hydrolases (3) acting on ester bonds (3.1)
    • C12N9/22Ribonucleases RNAses, DNAses
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/01Fusion polypeptide containing a localisation/targetting motif
    • C07K2319/07Fusion polypeptide containing a localisation/targetting motif containing a mitochondrial localisation signal
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/80Fusion polypeptide containing a DNA binding domain, e.g. Lacl or Tet-repressor
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12PFERMENTATION OR ENZYME-USING PROCESSES TO SYNTHESISE A DESIRED CHEMICAL COMPOUND OR COMPOSITION OR TO SEPARATE OPTICAL ISOMERS FROM A RACEMIC MIXTURE
    • C12P19/00Preparation of compounds containing saccharide radicals
    • C12P19/26Preparation of nitrogen-containing carbohydrates
    • C12P19/28N-glycosides
    • C12P19/30Nucleotides
    • C12P19/34Polynucleotides, e.g. nucleic acids, oligoribonucleotides

Definitions

  • the present invention relates to new Transcription Activator-Like Effector proteins and more particularly new Transcription Activator-Like Effector Nucleases (TALENs) that can efficiently target and process nucleic acids.
  • the present invention also concerns methods to use these new Transcription Activator-Like Effector proteins.
  • the present invention also relates to vectors, compositions and kits in which Transcription Activator-Like Effector proteins of the present invention are used.
  • TAL effectors Transcription activator-like effectors
  • Natural TAL effectors are produced by phytopathogenic bacteria and function upon infection as transcription activators of plant genes [for review see (Bogdanove, Schornack et al. 2010; Christian, Cermak et al. 2010)]. Since the isolation of the first TAL effectors gene (Bonas, Stall et al. 1989), the presence of repetitive motifs nearly identical within the central domain has been questioned. Today this central domain has been shown to be responsible for DNA recognition through a new type of DNA-binding domain (Boch, Scholze et al. 2009; Moscou and Bogdanove 2009). Each repeat is made usually of 33 or 34 amino acids and mediates the recognition of 1 nucleotide of the DNA target through 2 critical amino acids located at positions 12 and 13 in each repeat.
  • RVD right-variable di-residue
  • TALEN TAL Nuclease
  • the targeted sequence must start by a T, then T and A should be disfavored at position 1 and 2 respectively. They found also a strong bias towards the RVD-NG at the last position of the repeat arrays. Finally, the target DNA should have a low G content (9 ⁇ 8%). The robustness of these rules is not yet established. So far, the most established limitations when one want to design a TALEN against a chosen sequence is the presence of a T at the beginning of each target DNA sequence. Actually this essential T is not imposed by a specific RVD as it interacts directly with the N-terminal domain of the protein, and thus is not governed by the so called RVD code.
  • ZFN are classically heterodimeric proteins that bind two DNA sequences separated by 6 bp.
  • the TALEN described so far were all designed following the same architecture i.e. TALEN acts as heterodimers proteins in which the nuclease catalytic domain such Fok1 is fused to the TALE C-terminal region.
  • TALEN acts as heterodimers proteins in which the nuclease catalytic domain such Fok1 is fused to the TALE C-terminal region.
  • the optimal length of the DNA sequence separating the two binding domains had to be determined.
  • ZFNs DNA targets contain almost exclusively 6 bp intervening sequences, TALEN appears to tolerate a much more wide range of DNA length and as expected, appears to be dependent on the TALE scaffold used.
  • a spacer length below 10 nucleotides did not allowed efficient cleavage in vitro as did the C-terminal truncation bearing the 95 first amino acids of the C-terminal domain.
  • TALEN bearing the 28 first residues of the C-terminal domain showed nuclease activity in vitro when tested on target comprising spacer from 10 to 24 bps, with a maximal activity for spacer of 12-13 bps. Sequences narrower than 8 bp did not allow significant cleavage activity. Even though some guidance's were described (Cermak, Doyle et al. 2011) to help designing active TALEN, too few data are available today to confirm their benefits.
  • the inventors have developed a new type of TAL effector proteins and particularly a new type of TALEN that can be engineered to specifically recognize and process target nucleic acid efficiently, overpassing the actual limitations.
  • the present invention relates to new Transcription Activator-Like Effector proteins and more particularly new Transcription Activator-Like Effector Nucleases (TALENs) that can efficiently target and process nucleic acids.
  • the present invention also concerns methods to use these new Transcription Activator-Like Effector proteins for various applications.
  • the present invention also concerns the creation of functional single-polypeptide fusion proteins, i.e chimeric proteins derived from a Transcription Activator-Like Effector for simple and efficient vectorization.
  • the present invention also relates to vectors, compositions and kits in which chimeric proteins of the present invention derived of Transcription Activator-Like Effector are used.
  • FIG. 1 General description of a chimeric protein according to the present invention.
  • FIG. 2 Activity of AvrBs3-derived TALEN in yeast (30° C.); AvrBs3-derived TALEN is represented by a black bar and negative control (empty vector) and positive control (I-SceI meganuclease) are represented by grey bars. Activities are normalized to the positive control.
  • FIG. 3 Activity of AVRBs3-derived TALEN in mammalian cells (Extrachromosomic assay in CHO-K1); AvrBs3-derived TALEN is represented by a black curve, negative control (empty vector) by a dark grey curve and positive control (I-SceI meganuclease) by a light grey curve
  • FIG. 4 Activity of N152 AvrBs3-derived TALEN in yeast (30° C.); truncated variants is represented by a grey bars and AvrBs3-derived TALEN (control wt) is represented by a black bar. Activities are normalized to the AvrBs3-derived TALEN (Control wt) (SEQ ID NO: 5) activity on its 15 bp target (Avr15) (SEQ ID NO: 6).
  • FIG. 5 Impact of truncations of the TALE C-terminal domain on AvrBs3-derived TALEN in yeast; truncated variants are represented by grey bars and AvrBs3-derived TALEN (control wt) is represented by a black bar; Activities are normalized to the AvrBs3-derived TALEN (Control wt) (SEQ ID NO: 5) activity on its 15 bp target (Avr15) (SEQ ID NO: 6).
  • FIG. 6 Activity in yeast of AvrBs3-derived TALENs comprising truncations of the C-terminal domain against targets of various spacer lengths. Effect of spacer length on: A, AVRBS3 TALEN having the C-terminal domain truncated after position E886 (C0) (SEQ ID NO: 19); C0 truncated variant is represented by grey bars and AvrBs3-derived TALEN is represented by black bars. Activities are normalized to the AvrBs3-derived TALEN (Control wt) activity on its 15 bp target (Avr15). * indicates no detectable activity.
  • C AVRBS3 TALEN having the C-terminal domain truncated after position G914 (C28) (SEQ ID NO: 21); C28 truncated variant is represented by grey bars and AvrBs3-derived TALEN is represented by black bars.
  • Activities are normalized to the AvrBs3-derived TALEN (Control wt) activity on its 15 bp target (Avr15). * indicates no activity over negative control.
  • D AVRBS3 TALEN having the C-terminal domain truncated after position L926 (C40) (SEQ ID NO: 22); C40 truncated variant is represented by grey bars and AvrBs3-derived TALEN is represented by black bars. Activities are normalized to the AvrBs3-derived TALEN (Control wt) activity on its 15 bp target (Avr15). * indicates no activity over negative control.
  • F AVRBS3 TALEN having the C-terminal domain truncated after position T1003 (C115) (SEQ ID NO: 24); C115 truncated variant is represented by grey bars and AvrBs3-derived TALEN is represented by black bars.
  • Activities are normalized to the AvrBs3-derived TALEN (Control wt) activity on its 15 bp target (Avr15). * indicates no activity over negative control.
  • G AVRBS3 TALEN having the C-terminal domain truncated after position E1057 (C172) (SEQ ID NO: 25); C172 truncated variant is represented by grey bars and AvrBs3-derived TALEN is represented by black bars. Activities are normalized to the AvrBs3-derived TALEN (Control wt) activity on its 15 bp target (Avr15). * indicates no activity over negative control.
  • FIG. 7 Impact of nucleotide identity at position n of the target on the AvrBs3-derived TALEN activity (in yeast); Control target Avr15 with a T at position n is represented by a black bar. Activities are normalized to the AvrBs3-derived TALEN (Control wt) (SEQ ID NO: 5) activity on its 15 bp target (Avr15) (SEQ ID NO: 6).
  • FIG. 8 Activity of engineered TALENs in mammalian cells (Extrachromosomic assay in CHO-K1); DMDT2.1 TALEN (SEQ ID NO: 180, SEQ ID NO: 186, SEQ ID NO: 189; example 6) is represented by a dark grey curve, ILRGT2.1 TALEN (SEQ ID NO: 181, SEQ ID NO: 187, SEQ ID NO: 190; example 6) is represented by a black curve, HBBT1.1 TALEN (SEQ ID NO: 182, SEQ ID NO: 188, SEQ ID NO: 192; example 6) is represented by a dark grey curve, negative control (empty vector) by a light grey dashed curve and positive control (I-SceI meganuclease) by a black dashed curve.
  • ILRGT2.1 TALEN SEQ ID NO: 181, SEQ ID NO: 187, SEQ ID NO: 190; example 6
  • HBBT1.1 TALEN SEQ ID NO: 182, S
  • FIG. 9 Schematic of chimeric protein configurations according to the invention.
  • FIG. 10 Schematic of the method for optimizing the control of double-stranded break activity of a chimeric protein according to the invention.
  • FIG. 11 Schematic of the method for increasing the number of targets that can be reach by a chimeric protein according to the invention.
  • FIG. 12 Activity of TALE-AvrBs3::TevI in yeast (37° C.).
  • the negative control consists in a TALEN without any RVDs.
  • n.d. indicates no detectable activity
  • + indicates an activity over 0.3 in yeast assay
  • +++ indicates an activity over 0.7 in yeast assay (International PCT Applications WO 2004/067736 and in Epinat, Arnould et al. 2003; Chames, Epinat et al. 2005; Arnould, Chames et al. 2006; Smith, Grizot et al. 2006).
  • FIG. 13 Activity of TALE-AvrBs3::TevI in mammalian cells.
  • pCLS8993 SEQ ID NO: 457
  • pCLS8994 SEQ ID NO: 458
  • Negative control empty vector
  • I-SceI meganuclease I-SceI meganuclease
  • FIG. 14 Activity of TALE-AvrBs3::NucA in yeast (37° C.).
  • the negative control is a target lacking a recognition site (neg. ctrl.: SEQ ID NO: 472).
  • Compact is a target having only one recognition site (SEQ ID NO: 468).
  • n.d. indicates no detectable activity, + indicates an activity over 0.3 in yeast assay at 37° C.; ++ indicates an activity over 0.5 in yeast assay at 37° C. and +++ indicates an activity over 0.7 in yeast assay at 37° C.
  • International PCT Applications WO 2004/067736 and in Epinat, Arnould et al. 2003; Chames, Epinat et al. 2005; Arnould, Chames et al. 2006; Smith, Grizot et al. 2006).
  • FIG. 15 Activity of TALE-AvrBs3::ColE7 in yeast (37° C.).
  • the negative control is a target lacking a recognition site (neg. ctrl.: SEQ ID NO: 472).
  • Compact is a target having only one recognition site (SEQ ID NO: 468).
  • n.d. indicates no detectable activity, + indicates an activity over 0.3 in yeast assay at 37° C.; ++ indicates an activity over 0.5 in yeast assay at 37° C. and +++ indicates an activity over 0.7 in yeast assay at 37° C.
  • International PCT Applications WO 2004/067736 and in Epinat, Arnould et al. 2003; Chames, Epinat et al. 2005; Arnould, Chames et al. 2006; Smith, Grizot et al. 2006).
  • FIGS. 16 to 19 Activity of asymmetrical TALENs in yeast (37° C.). n.d. indicates no detectable activity at 37° C., +/ ⁇ indicated an activity above 0.3 in yeast assay at 37° C.; + indicated an activity over 0.3 in yeast assay at 37° C.; ++ indicated an activity over 0.5 in yeast assay at 37° C.; +++ indicated an activity over 0.75 in yeast assay at 37° C. (International PCT Applications WO 2004/067736 and in Epinat, Arnould et al. 2003; Chames, Epinat et al. 2005; Arnould, Chames et al. 2006; Smith, Grizot et al. 2006).
  • FIG. 20 Activity of TALE-AvrBs3::EndoT7 in yeast (37° C.). n.d. indicates no detectable activity at 37° C., + indicated an activity over 0.3 in yeast assay at 37° C.; ++ indicated an activity over 0.5 in yeast assay at 37° C.; +++ indicated an activity over 0.75 in yeast assay at 37° C. (International PCT Applications WO 2004/067736 and in Epinat, Arnould et al. 2003; Chames, Epinat et al. 2005; Arnould, Chames et al. 2006; Smith, Grizot et al. 2006).
  • FIG. 21 Activity of colE7 (SEQ ID NO: 340), EndoT7 (SEQ ID NO: 363) and I-TevI (SEQ ID NO: 349) catalytic heads containing TALEN with various polypeptide linker in yeast (37° C.).
  • Compact is a target having only one recognition site (SEQ ID NO: 224).
  • n.d. indicates no detectable activity at 37° C., + indicated an activity over 0.3 in yeast assay at 37° C.; ++ indicated an activity over 0.5 in yeast assay at 37° C.; +++ indicated an activity over 0.75 in yeast assay at 37° C.
  • International PCT Applications WO 2004/067736 and in Epinat, Arnould et al. 2003; Chames, Epinat et al. 2005; Arnould, Chames et al. 2006; Smith, Grizot et al. 2006).
  • FIG. 22 Activity of TALEN having various polypeptide linkers (37° C.).
  • the negative control is a target lacking recognition sites.
  • n.d. indicates no detectable activity at 37° C., +/ ⁇ indicated an activity above 0.7 in yeast assay at 37° C.; +++ indicated an activity over 0.70 in yeast assay at 37° C.
  • FIG. 23 Tal repeats arrays assembly and subcloning into yeast expression plasmids.
  • A Legend of materials used for TAL repeat assembly.
  • B immobilization of the first biotinylated TAL repeat fragment on a streptavidin coated solid support and ligation to a second TAL repeat harboring SfaNI compatible overhangs (BbvI overhangs displayed in red).
  • C consecutive ligation/restriction of TAL repeats to generate the complete TAL repeats array.
  • D SfaNI digestion of the TAL repeats array.
  • E BbvI digestion and recovery of the TAL repeats array.
  • FIG. 24 Influence of TAL repeat number on TALEN activity. TALEN activities of 52 different TALENs (SEQ ID NO: 507-558) bearing from 9.5 to 15.5 TAL repeats were determined. The mean values of TALEN activities are displayed as a function of TAL repeat number; error bars represent the standard deviation of activities of TALENs bearing the same number of TAL repeats.
  • FIG. 25 Influence of different single protein/DNA mismatches at position N-1 or N-2 on TALEN activity as a function of TAL repeat number.
  • RAGT2.4 TALENs bearing 15.5 or 11.5 Tal repeats (SEQ ID NO: 617 or 622) along with their respective DNA targets (top and bottom respectively).
  • FIG. 26 Influence of C-terminal domain substitution by polypeptide linkers 8, 27 and 35 on AvrBs3 TALEN nuclease activity in yeast.
  • AvrBs3 TALENs bearing polypeptide linkers 8, 27 and 35 (SEQ ID: 141, 160 and 168) as C-terminal domain were assayed toward AvrBs3 homodimeric targets bearing from 5 to 30 bp DNA spacer.
  • Their yeast activities are displayed as a function of spacer length.
  • FIG. 27 Activities for novel variations of the TALE::FokI scaffold.
  • the negative control consists in a TALEN without any RVDs. n.d. indicates no detectable activity, + indicates an activity between 0.3 and 0.5 in our assay, ++ indicates an activity between 0.5 and 0.7 in our assay and +++ indicates an activity over 0.7 in our assay (International PCT Applications WO 2004/067736 and in Epinat, Arnould et al. 2003; Chames, Epinat et al. 2005; Arnould, Chames et al. 2006; Smith, Grizot et al. 2006).
  • FIG. 28 Activities of combination TALE::FokI and FokI::TALE in yeast.
  • the negative control consists in a TALEN without any RVDs. n.d. indicates no detectable activity, ++ indicates an activity between 0.5 and 0.7 in our assay and +++ indicates an activity over 0.7 in our assay (International PCT Applications WO 2004/067736 and in Epinat, Arnould et al. 2003; Chames, Epinat et al. 2005; Arnould, Chames et al. 2006; Smith, Grizot et al. 2006).
  • Table 3 List of AvrBs3 targets with various spacer lengths (SEQ ID NO: 220 to 255).
  • Table 4 List of AvrBs3 targets with all combination at position n (SEQ ID NO: 171 to 174).
  • Table 5 List of AvrBs3/RAGT2 (heterodimer) targets with various spacer lengths (SEQ ID NO: 256 to 291).
  • Table 6 Activities of 27 custom TALEN tested as homodimers, in four different scaffolds (full wt C-terminal domain, C0 truncated C-terminal domain, C11 truncated C-terminal domain and C40 truncated C-terminal domain); (n.a: non available; n.d: non detected) (SEQ ID NO: 26 to 133).
  • Table 7 Binding and target sequences of 27 custom TALEN (homodimers) (SEQ ID NO: 193 to 219).
  • Table 8 List of AvrBs3 targets with various spacer lengths (SEQ ID NO: 220 to 255) including a target with only one recognition site (compact, SEQ ID NO: 468) and a negative control target (neg. ctrl., SEQ ID NO: 472) consisting in a target without any recognition site.
  • Table 9 List of AvrBs3 targets containing two identical recognition sequences juxtaposed with the 5′ ends proximal and separated by “spacer” DNA ranging from 5 to 35 bps (SEQ ID NO: 629 to 659).
  • Table 10 List of RagT2-R/AvrBs3 hybrid targets contain two different recognition sequences juxtaposed with the 3′ end of the first (RagT2-R) proximal to the 5′ end of the second (AvrBs3) and separated by “spacer” DNA ranging from 5 to 40 bps (SEQ ID NO: 666 to 701),
  • the present invention relates to new Transcription Activator-Like Effector proteins and more particularly new Transcription Activator-Like Effector Nucleases (TALENs) that can efficiently target and process nucleic acids.
  • TALENs Transcription Activator-Like Effector Nucleases
  • the present invention relates to a chimeric protein derived from a Transcription Activator-Like Effector (TALE) comprising:
  • said chimeric protein further comprises a peptidic linker to fuse said catalytic domain to said core scaffold.
  • a chimeric protein constituted by a core scaffold comprising a DNA binding domain and a protein domain to process a nucleic acid target sequence.
  • a chimeric protein derived from a Transcription Activator-Like Effector (TALE) comprising:
  • the chimeric protein of the present invention is derived from any naturally occurring TAL effectors, such as those described in Bogdanove et al. (Boch and Bonas 2010; Bogdanove, Schornack et al. 2010) and listed in Boch et al. (Boch and Bonas 2010).
  • the chimeric protein of the present invention is derived from any TAL effectors of plant pathogenic bacteria in the genus Xanthomonas as listed in Boch et al (Boch and Bonas 2010) as a non-limiting example.
  • only one part of the core scaffold is derived from a TAL effector; as an illustrative example, only said set of Repeat Variable Dipeptide regions is derived from a TAL effector.
  • each RVD of said core scaffold is made of 30 to 42 amino acids, more preferably 33 or 34 wherein two critical amino acids located at positions 12 and 13 mediates the recognition of one nucleotide of said nucleic acid target sequence; equivalent two critical amino acids can be located at positions other than 12 and 13 specially in RVDs taller than 33 or 34 amino acids long.
  • RVDs associated with recognition of the different nucleotides are HD for recognizing C, NG for recognizing T, NI for recognizing A, NN for recognizing G or A, NS for recognizing A, C, G or T, HG for recognizing T, IG for recognizing T, NK for recognizing G, HA for recognizing C, ND for recognizing C, HI for recognizing C, HN for recognizing G, NA for recognizing G, SN for recognizing G or A and YG for recognizing T, TL for recognizing A, VT for recognizing A or G and SW for recognizing A.
  • RVDs associated with recognition of the nucleotides C, T, A, G/A and G respectively are selected from the group consisting of NN or NK for recognizing G, HD for recognizing C, NG for recognizing T and NI for recognizing A, TL for recognizing A, VT for recognizing A or G and SW for recognizing A.
  • RVDS associated with recognition of the nucleotide C are selected from the group consisting of N*
  • RVDS associated with recognition of the nucleotide T are selected from the group consisting of N* and H*, where * denotes a gap in the repeat sequence that corresponds to a lack of amino acid residue at the second position of the RVD.
  • critical amino acids 12 and 13 can be mutated towards other amino acid residues in order to modulate their specificity towards nucleotides A, T, C and G and in particular to enhance this specificity.
  • other amino acid residues is intended any of the twenty natural amino acid residues or unnatural amino acids derivatives.
  • said core scaffold of the present invention comprises between 8 and 30 RVDs. More preferably, said core scaffold of the present invention comprises between 8 and 20 RVDs; again more preferably 15 RVDs.
  • said core scaffold comprises an additional single truncated RVD made of 20 amino acids located at the C-terminus of said set of RVDs, i.e. an additional C-terminal half-RVD.
  • said core scaffold of the present invention comprises between 8.5 and 30.5 RVDs, “0.5” referring to previously mentioned half-RVD (or terminal RVD, or half-repeat). More preferably, said core scaffold of the present invention comprises between 8.5 and 20.5 RVDs, again more preferably, 15.5 RVDs.
  • said half-RVD is in a core scaffold context which allows a lack of specificity of said half-RVD toward nucleotides A, C, G, T. In a more preferred embodiment, said half-RVD is absent.
  • said core scaffold of the present invention comprises RVDs of different origins.
  • said core scaffold comprises RVDs originating from different naturally occurring TAL effectors.
  • internal structure of some RVDs of the core scaffold of the present invention are constituted by structures or sequences originated from different naturally occurring TAL effectors.
  • said core scaffold of the present invention comprises RVDs-like domains.
  • RVDs-like domains have a sequence different from naturally occurring RVDs but have the same function and/or global structure within said core scaffold of the present invention.
  • said RVDs-like domains are protein domains selected from the group consisting of Puf RNA binding protein or Ankyrin super-family.
  • Non-limiting examples of such proteins from which RVDs-like domain can be derived are given by SEQ ID NO: 398 and SEQ ID NO: 399 respectively corresponding to proteins fem-3 and aRep.
  • said core scaffold of the chimeric protein of the present invention comprises a mix of naturally occurring RVDs structures and RVDs-like domains.
  • said core scaffold of the present invention is entirely composed by RVDs-like domains that are not originated from pathogenic organisms.
  • said core scaffold of the chimeric protein of the present invention is totally artificial, i.e. without any RVDs-like domains derived from naturally occurring TAL effectors.
  • Such mutations can also overcome nucleic acid modifications such as DNA alkylation (acetylation, methylation) as a non-limiting example; in this case, the core scaffold of the chimeric protein of the invention can have a higher selectivity for a methylated or unmethylated target sequence; in other words, said core scaffold can tolerate nucleic acid methylation or can be specific of a methylated target sequence.
  • VVAIA and LLPVL amino acids motifs in RVDs may be of structural importance for hydrophobic contact between helices of adjacent RVD and may represent good candidate motifs to mutate for modulating the intra RVD interactions or interdependency between adjacent RVDs in a set of repeated variable domains and thus the affinity and specificity of subsequent TALENs.
  • RVDs variants mutated at these residues to modify interactions between adjacent RVDs resulting in a core scaffold of the present invention with more rigidity.
  • More or less rigidity in core scaffolds of the present invention allows to enhance or decrease protein-protein interactions within the structure of the scaffold, particularly when adjacent RVDs in said core scaffold are from different origins; also, it allows to modify protein-nucleic acid interactions between said core scaffold of the chimeric protein of the present invention and its nucleic acid target. Modifications of protein-protein or protein-nucleic acid interactions can be quantified by measuring biochemical constants [affinity (Ka)/dissociation (Kd)/turn over (Kcat) constants] associated with such interactions/reactions.
  • said core scaffold of the present invention comprises one additional domain at the N-terminus of said set of Repeat Variable Dipeptide regions.
  • said additional N-terminus domain is derived from the N-terminus domain of a naturally occurring TAL effector.
  • said additional N-terminus domain is the full-length N-terminus domain of a naturally occurring TAL effector N-terminus domain.
  • said additional N-terminus domain is a variant of a naturally occurring Tal effector.
  • said additional domain is a truncated variant of a naturally occurring TAL effector N-terminus domain.
  • said additional domain is a truncated version of AvrBs3 TAL effector.
  • said truncated version lacks at its C-terminus extremity the peptidic sequence that confers specificity to position 0 of the recognized and bound sequence, i.e. the “RVD0” repeat, named for a postulated 0 th repeat that has only weak sequence similarity but a predicted structural similarity to the repeats in Bogdanove et al. (Bogdanove 2010 current opinion).
  • said truncated version lacks at least one amino acid residue selected from the group consisting of the 152 first N-terminal amino acids residues. In another embodiment, said truncated version lacks more than the first 152 amino acids residues.
  • said additional N-terminus domain is a non-TAL effector originating domain.
  • said additional N-terminus domain is derived from a protein having non-specific nucleic acid binding characteristics.
  • said additional N-terminus domain is derived from a protein having non-specific DNA binding characteristics.
  • said additional N-terminus domain is derived from a protein having non-specific RNA binding characteristics.
  • said additional N-terminus domain is derived from a protein having specific nucleic acid binding characteristics, such as non-limiting examples, meganucleases or zinc-finger proteins or derivatives of those such as variants with only DNA binding activity.
  • said additional N-terminus domain is a chimeric domain comprising a TAL effector originating subdomain and a non-TAL effector originating subdomain.
  • said additional N-terminus domain is a variant increasing the affinity of said core scaffold of the chimeric protein of the present invention toward its binding nucleic acid sequence.
  • said additional N-terminus domain is a variant which allows overcoming sequence constraints associated with said RVD0, i.e. the necessity to have a T as the first base on binding nucleic acid sequence.
  • said additional N-terminus domain is a variant which allows changing this sequence constraint to A, G or C respectively.
  • said additional N-terminus domain is a variant which allow suppressing the sequence constraints associated with RVD0.
  • said additional N-terminus domain of the core scaffold of the chimeric protein of the present invention comprises a localization sequence (or signal) which allows targeting said chimeric protein toward a given organelle within an organism, a tissue or a cell.
  • localization signals are nuclear localization signals, chloroplastic localization signals or mitochondrial localization signals.
  • said additional N-terminus domain can comprise a nuclear export signal having the opposite effect of a nuclear localization signal to help targeting organelles such as chloroplasts or mitochondria.
  • additional N-terminus domains with a combination of several localization signals can be as a non-limiting example a nuclear localization signal and a tissue-specific signal to help addressing said chimeric protein of the present invention in the nuclear of tissue specific cells.
  • said additional N-terminal domain can be fused with a protein domain, a protein module, an antibody (or part of it) or a tag of interest, well-known in the art, for a specific application.
  • said additional N-terminal domain can be linked with a chemical molecule such as a small compound of interest for a defined application.
  • said additional domain at the N-terminus of said set of Repeat Variable Dipeptide regions of said core scaffold of the present invention is selected from the group consisting of SEQ ID NO: 292, SEQ ID NO: 293 and SEQ ID NO: 401 or derivatives thereof.
  • said core scaffold of the present invention comprises one additional domain at the C-terminus of said set of Repeat Variable Dipeptide regions.
  • said additional C-terminus domain is derived from the C-terminus domain of a naturally occurring TAL effector.
  • said additional C-terminus domain is the full-length C-terminus domain of a naturally occurring TAL effector.
  • said additional C-terminus domain is a variant of a naturally occurring Tal effector C-terminus domain.
  • said additional domain is a truncated variant of a naturally occurring TAL effector C-terminus domain.
  • said truncated version is a C-terminus domain without Activation Domain (SEQ ID NO: 400 and 402).
  • said additional domain is a truncated version of AvrBs3 TAL effector. In another embodiment, said additional domain is truncated after position E886 (C0). In another embodiment, said additional domain is truncated after position P897 (C11; SEQ ID NO: 295). In another embodiment, said additional domain is truncated after position G914 (C28; SEQ ID NO: 296). In another embodiment, said additional domain is truncated after position L926 (C40; SEQ ID NO: 297). In another embodiment, said additional domain is truncated after position D950 (C64; SEQ ID NO: 298).
  • said additional domain is truncated after position R1000 (C115; SEQ ID NO: 299). In another embodiment, said additional domain is truncated after position D1059 (C172; SEQ ID NO: 300) (amino acid numbering refers to C-terminus domain of AvrBs3 TAL effector).
  • said additional C-terminus domain is a non-TAL effector originating domain.
  • said additional C-terminus domain is derived from a protein having non-specific nucleic acid binding characteristics.
  • said additional C-terminus domain is derived from a protein having non-specific DNA binding characteristics.
  • said additional C-terminus domain is derived from a protein having non-specific RNA binding characteristics.
  • said additional C-terminus domain is derived from a protein having specific nucleic acid binding characteristics, such as non-limiting examples, meganucleases or zinc-finger proteins or derivatives of those such as variants with only DNA binding activity.
  • said additional C-terminus domain is a chimeric domain comprising a TAL effector originating subdomain and a non-TAL effector originating subdomain.
  • said additional C-terminus domain is a variant increasing the affinity of said core scaffold of the chimeric protein of the present invention toward its binding nucleic acid sequence.
  • said additional C-terminus domain of the core scaffold of the chimeric protein of the present invention comprises a localization sequence (or signal) which allows targeting said chimeric protein toward a given organelle within an organism, a tissue or a cell.
  • localization signals are nuclear localization signals, chloroplastic localization signals or mitochondrial localization signals.
  • said additional C-terminus domain can comprise a nuclear export signal having the opposite effect of a nuclear localization signal to help targeting organelles such as chloroplasts or mitochondria.
  • additional C-terminus domains with a combination of several localization signals can be as a non-limiting example a nuclear localization signal and a tissue-specific signal to help addressing said chimeric protein of the present invention in the nuclear of tissue specific cells.
  • said additional C-terminal domain can be fused with a protein domain, a protein module, an antibody (or part of it) or a tag of interest, well-known in the art, for a specific application.
  • said additional C-terminal domain can be linked with a chemical molecule such as a small compound of interest for a defined application.
  • said additional domain at the C-terminus of said set of Repeat Variable Dipeptide regions of said core scaffold of the present invention is selected from the group consisting of SEQ ID NO: 295 to 300, SEQ ID NO: 400 and SEQ ID NO: 402 or derivatives thereof.
  • said core scaffold of the chimeric protein according to the present invention comprises two additional domains respectively at the N-terminus and at the C-terminus of said set of Repeat Variable Dipeptide regions, as previously described.
  • said chimeric protein according to the present invention comprises at least one peptidic linker to fuse a protein domain to said core scaffold previously described.
  • said peptidic linker is flexible.
  • said peptidic linker is structured.
  • said peptidic linker sequence is selected from the group consisting of NFS1, NFS2, CFS1, RM2, BQY, QGPSG, LGPDGRKA, 1a8h — 1, 1 dnpA — 1, 1 d8cA — 2, 1 ckqA — 3, 1sbp — 1, 1 ev7A — 1, 1 alo — 3, 1amf — 1, 1 adjA — 3, 1 fcdC — 1, 1 al3 — 2, 1 g3p — 1, 1acc — 3, 1 ahjB — 1, 1acc — 1, 1 af7 — 1, 1 heiA — 1, 1bia — 2, 1 igtB — 1, 1 nfkA — 1, 1 au7A — 1, 1 bpoB — 1, 1 b0 pA — 2, 1 c05A — 2, 1gcb — 1, 1 bt3A — 1, 1 b3oB — 2, 16
  • the peptidic linker that can link said core scaffold to said protein domain of the chimeric protein according to the present invention can be selected from the group consisting of TAL1 to TAL37 (SEQ ID NO: 134 to SEQ ID NO: 170).
  • TAL1 to TAL37 SEQ ID NO: 134 to SEQ ID NO: 170.
  • a peptidic linker is not needed to fuse said core scaffold with said protein domain in order to obtain a chimeric protein according to the present invention.
  • more than one linker is needed to fuse several protein domains with said core scaffold according to the present invention.
  • two, three or four linkers can be used in the same chimeric protein according to the present invention.
  • said peptidic linker contains one or several active domains which allow its deployment under stimulation.
  • said peptidic linker can contain a calmodulin domain that changes its conformation under calcium stimulation; other protein domains changing their conformation under a specific metabolite interaction can be used.
  • such peptidic linker according to the present invention can contain a light sensitive domain which allows a change in peptidic linker structure from a folded inactive state toward an unfolded active state under light stimulation for example, or reverse.
  • Said peptidic linker can for example contain a first light-sensitive protein switch comprising a phytochrome-chromophore complex and a Phytochrome Interaction Factor (PIF) i.e. a second protein able to reversibly interact with said phytochrome-chromophore complex depending on the light activation/desactivation state.
  • PAF Phytochrome Interaction Factor
  • Other examples of active linkers can use small molecules such as Chemical Inducers of Dimerization (CID).
  • said chimeric protein according to the present invention comprises at least one protein domain or catalytic domain to process said nucleic acid target sequence.
  • the catalytic domain that is capable of processing said nucleic acid target sequence, when fused to said core scaffold according to the present invention is fused to the N-terminus part of said core scaffold.
  • said catalytic domain is fused to the C-terminus part of said core scaffold.
  • two catalytic domains are fused to both N-terminus part of said core scaffold and C-terminus of said core scaffold.
  • one or several catalytic domains to said core scaffold wherein said core scaffold comprises or not an additional domain at its N-terminus and/or at its C-terminus.
  • one or several peptidic linkers can be added for said fusions between the different domains of the chimeric protein according to the present invention.
  • several catalytic domains and several peptidic linkers is intended two or three or four or five as non-limiting examples.
  • said catalytic domain has an activity selected from the group consisting of nuclease activity, polymerase activity, kinase activity, phosphatase activity, methylase activity, topoisomerase activity, integrase activity, transposase activity, ligase activity, helicase activity, recombinase activity.
  • the catalytic domain fused to the core scaffold of the present invention can be a transcription activator or repressor (i.e. a transcription regulator), or a protein that interacts with or modifies other proteins implicated in DNA processing.
  • a transcription activator or repressor i.e. a transcription regulator
  • Non-limiting examples of DNA processing activities of said chimeric protein of the present invention include, for example, creating or modifying epigenetic regulatory elements, making site-specific insertions, deletions, or repairs in DNA, controlling gene expression, and modifying chromatin structure.
  • said catalytic domain has an endonuclease activity.
  • said protein domain has an exonuclease activity.
  • said catalytic domain is selected from the group consisting of proteins MmeI, Colicin-E7 (CEA7_ECOLX), Colicin-E9, APFL, EndA, Endo I (END1_ECOLI), Human Endo G (NUCG_HUMAN), Bovine Endo G (NUCG_BOVIN), R.HinP1I, I-BasI, I-BmoI, I-HmuI, I-TevI, I-TevII, I-TevIII, I-TwoI, R.MspI, R.MvaI, NucA, NucM, Vvn, Vvn_CLS, Staphylococcal nuclease (NUC_STAAU), Staphylococcal nuclease (NUC_STAHY),
  • the chimeric protein according to the present invention comprises a catalytic domain that is a polypeptide comprising an amino acid sequence having at least 80%, preferably at least 90%, more preferably at least 95% identity with any of SEQ ID NO: 339 to 397 and SEQ ID NO: 598-599.
  • said catalytic domain of the chimeric protein of the invention has an identity with I-TevI (SEQ ID NO: 349). In another embodiment, said catalytic domain has an identity with I-TevI (SEQ ID NO: 349) and is fused to the N-terminal domain of said core scaffold. In another embodiment, said catalytic domain has an identity with I-TevI (SEQ ID NO: 349) and is fused to the C-terminal domain of said core scaffold. In another embodiment, said chimeric protein according to the present invention acts as a monomer wherein two of said monomer respectively bind one nucleic acid sequence adjacent to a nucleic acid target sequence thereby together processing said nucleic target sequence.
  • said chimeric protein comprises a catalytic domain having identity with I-TevI (SEQ ID NO: 349) and fused to the C-terminal domain of said core scaffold and acts as a monomer wherein two of said monomer respectively bind one nucleic acid sequence adjacent to a nucleic acid target sequence thereby together processing said nucleic target sequence.
  • the first and the second monomers have the same amino acid sequence or not.
  • said chimeric protein comprises a catalytic domain having identity with I-TevI (SEQ ID NO: 349) and fused to the C-terminal domain of said core scaffold acting as a first monomer binding one nucleic acid sequence adjacent to a nucleic acid target sequence and wherein a second chimeric protein monomer comprising I-TevI (SEQ ID NO: 349) or derivatives thereof fused to the N-terminus of said core scaffold acts as a second monomer binding another nucleic acid sequence adjacent to said nucleic acid target sequence, thereby together processing said nucleic target sequence.
  • said catalytic domain of the chimeric protein of the invention has an identity with NucA (SEQ ID NO: 355). In another embodiment, said catalytic domain has an identity with NucA (SEQ ID NO: 355) and is fused to the N-terminal domain of said core scaffold. In another embodiment, said catalytic domain has an identity with NucA (SEQ ID NO: 355) and is fused to the C-terminal domain of said core scaffold. In another embodiment, said catalytic domain of the chimeric protein of the invention has an identity with ColE7 (SEQ ID NO: 340). In another embodiment, said catalytic domain has an identity with ColE7 (SEQ ID NO: 340) and is fused to the N-terminal domain of said core scaffold. In another embodiment, said catalytic domain has an identity with ColE7 (SEQ ID NO: 340) and is fused to the C-terminal domain of said core scaffold.
  • said catalytic domain of the chimeric protein of the invention has an identity with FokI (SEQ ID NO: 600) and is fused to the N-terminal domain of said core scaffold.
  • said additional catalytic domain at the N-terminus of said core scaffold comprises an amino acid sequence having at least 80%, preferably at least 90%, more preferably at least 95% identity with Fok-I (SEQ ID NO: 600).
  • any combinations of two catalytic domains selected from the group consisting of proteins MmeI, Colicin-E7 (CEA7_ECOLX), Colicin-E9, APFL, EndA, Endo I (END1_ECOLI), Human Endo G (NUCG_HUMAN), Bovine Endo G (NUCG_BOVIN), R.HinP1I, I-BasI, I-BmoI, I-HmuI, I-TevI, I-TevII, I-TevIII, I-TwoI, R.MspI, R.MvaI, NucA, NucM, Vvn, Vvn_CLS, Staphylococcal nuclease (NUC_STAAU), Staphylococcal nuclease (NUC_STAHY), Micrococcal nuclease (NUC_SHIFL), Endonuclease yncB, Endodeoxyribonuclea
  • I-HmuI protein domain can be fused to the N-terminus part of said core scaffold and ColE7 protein domain can be fused to the C-terminus part of said core scaffold.
  • I-TevI protein domain can be fused to the N-terminus part of said core scaffold and ColE7 protein domain can be fused to the C-terminus part of said core scaffold.
  • it can be envisioned to fuse one protein domain between two core scaffolds according to the invention, each one comprising at least one set of RVDs. In this last case, the number of RVDs for each engineered core TALE scaffold can be the same or not.
  • the chimeric protein according to the present invention comprises, catalytic domains respectively fused to the C-terminus and to the N-terminus of said core scaffold and selected from those having identity with:
  • the chimeric protein according to the present invention comprises, catalytic domains respectively fused to the C-terminus and to the N-terminus of said core scaffold and selected from those having identity with:
  • said catalytic domain of the chimeric protein of the invention has an identity with FokI (SEQ ID NO: 600) which is fused to the N-terminal domain of said core scaffold, wherein said chimeric protein acts as a monomer and wherein a second monomer binds another nucleic acid sequence adjacent to a nucleic acid target sequence thereby together processing said nucleic acid target sequence.
  • the first and the second monomers have the same amino acid sequence or not.
  • said catalytic domain of the chimeric protein of the invention has an identity with FokI (SEQ ID NO: 600) which is fused to the N-terminal domain of said core scaffold, wherein said chimeric protein acts as a monomer and wherein a second monomer, comprising a catalytic domain having an identity with FokI (SEQ ID NO: 600) which is fused to the C-terminal domain of said core scaffold, binds another nucleic acid sequence adjacent to a nucleic acid target sequence thereby together processing said nucleic acid target sequence.
  • FokI SEQ ID NO: 600
  • the chimeric protein according to the invention can comprise from its N-terminus toward its C-terminus: A first set of RVDs, a first linker, a protein domain, a second linker, a second set of RVDs.
  • the chimeric protein according to the invention can comprise a N-terminus domain as previously mentioned, a first set of RVDs, a first linker, a protein domain, a second linker, a second set of RVDs and a C-terminus domain as previously mentioned (as illustrated on FIG. 9B ).
  • linkers used in this case of chimeric protein according to the present invention can be active linkers comprising active domains which allow a change of their structure under appropriate stimulation.
  • the chimeric protein comprises a core scaffold with any combination between an additional C-terminus domain according to the invention and an additional N-terminus domain according to the invention.
  • said protein domain within the chimeric protein according to the present invention is a first protein subdomain interacting with a second protein subdomain to form said catalytic entity able to process said nucleic acid target sequence.
  • said first protein subdomain is selected from some of the group listed in Table 2 (SEQ ID NO: 339 to 397), such as MmeI (SEQ ID NO: 339), R.PleI (SEQ ID NO: 369), MlyI (SEQ ID NO: 370) as non-limiting examples, a functional mutant, a variant or derivatives of these protein subdomains thereof.
  • said second protein subdomain is selected from some of the group listed in Table 2 (SEQ ID NO: 339 to 397 and SEQ ID NO: 598-599), such as MmeI (SEQ ID NO: 339), R.PleI (SEQ ID NO: 369), MlyI (SEQ ID NO: 370) as non-limiting examples, a functional mutant, a variant or derivatives of these protein subdomains thereof.
  • said protein domain within the chimeric protein according to the present invention is a first protein subdomain interacting with a second protein subdomain to form a protein entity catalytically active.
  • Said first protein subdomain can be used to measure, quantify or provoke protein-protein interactions at said nucleic target sequence according to the present invention.
  • Said first protein subdomain can be a protein module or protein subdomain known to mediate protein-protein interaction in cell signaling.
  • Said first protein subdomain can be used for diagnosis, analytical or therapeutic applications.
  • Said protein entity can be a reporter protein such as a fluorescent protein, luciferase, ⁇ -galactosidase as non-limiting examples.
  • a first part of the reporter protein can be fused to said first protein subdomain according to the present invention and a second part of the reporter protein can be fused to said second protein subdomain, wherein said reporter protein is only active when said first and second protein subdomains according to the present invention interact.
  • said first protein subdomain or the protein entity resulting from the interaction between first and second subdomains can be used as intracellular sensor for calcium level, pH, redox environment as non-limiting examples.
  • said protein domain or subdomains are usable for applications such as Fluorescence Resonance Energy Transfer (FRET) as a non-limiting example.
  • said protein domain or subdomains are coupled to a dye.
  • said protein domain within the chimeric protein according to the present invention can be an inactive subdomain and can react with more than one protein domain or subdomain to form an active protein entity, i.e. said active entity can be comprising two, three, four or several subdomains and being an enzyme or a fluorescent protein as non-limiting example.
  • said active entity formed with the chimeric protein according to the present invention can interact or react with another protein or protein domain having a different activity.
  • said active entity formed with the chimeric protein according to the present invention can be associated within, or located 5′ or located 3′ regarding the nucleic acid target sequence location with another protein or protein domain having a different activity in order to process said nucleic acid target sequence; as a non-limiting example, said chimeric protein according to the present invention can comprise a protein domain with a cleavase activity for its nucleic acid target sequence and can be associated with an exonuclease activity to increase the mutagenesis rate at its nucleic acid target sequence location.
  • said second protein subdomain interacting with said first protein subdomain to form said catalytic entity able to process said nucleic acid target sequence according to the present invention is fused to a core scaffold comprising a set of Repeat Variable Dipeptide regions (RVDs) able to bind a second unique nucleic sequence adjacent to said nucleic acid target sequence wherein each RVD comprises a pair of amino acids responsible for recognizing one nucleotide selected from the group consisting of HD for recognizing C, NG for recognizing T, NI for recognizing A, NN for recognizing G or A, NS for recognizing A, C, G or T, HG for recognizing T, IG for recognizing T, NK for recognizing G, HA for recognizing C, ND for recognizing C, HI for recognizing C, HN for recognizing G, NA for recognizing G, SN for recognizing G or A and YG for recognizing T, TL for recognizing A, VT for recognizing A or G and SW for recognizing A.
  • RVDs associated with recognition of the nucleotides C, T, A, G/A and G respectively are selected from the group consisting of NN or NK for recognizing G, HD for recognizing C, NG for recognizing T and NI for recognizing A, TL for recognizing A, VT for recognizing A or G and SW for recognizing A.
  • RVDS associated with recognition of the nucleotide C are selected from the group consisting of N*
  • RVDS associated with recognition of the nucleotide T are selected from the group consisting of N* and H*, where * denotes a gap in the repeat sequence that corresponds to a lack of amino acid residue at the second position of the RVD.
  • critical amino acids 12 and 13 can be mutated towards other amino acid residues in order to modulate their specificity towards nucleotides A, T, C and G and in particular to enhance this specificity.
  • other amino acid residues is intended any of the twenty natural amino acid residues or unnatural amino acids derivatives.
  • said core scaffold fused to said second protein subdomain can have the same scope of characteristics that those previously listed to describe the chimeric protein according to the present invention, regarding the origin of said core scaffold, the number of RVDs comprised in said core scaffold, the nature of those RVDs (natural, artificial or RVDs-like domains), the existence of additional N-terminus or C-terminus or both domains on this core scaffold, the existence of one or several localization signals on this core scaffold, the existence of one or several peptidic linkers on this core scaffold to fuse one or several protein domains on this core scaffold.
  • said chimeric protein according to the present invention can function as a dimer wherein a first and a second monomer are derived from a Transcription Activator-Like Effector (TALE).
  • TALE Transcription Activator-Like Effector
  • said chimeric protein according to the present invention can function as a dimer wherein said first monomer comprises:
  • RVDs associated with recognition of the nucleotides C, T, A, G/A and G respectively are selected from the group consisting of NN or NK for recognizing G, HD for recognizing C, NG for recognizing T and NI for recognizing A, TL for recognizing A, VT for recognizing A or G and SW for recognizing A.
  • RVDS associated with recognition of the nucleotide C are selected from the group consisting of N*
  • RVDS associated with recognition of the nucleotide T are selected from the group consisting of N* and H*, where * denotes a gap in the repeat sequence that corresponds to a lack of amino acid residue at the second position of the RVD.
  • critical amino acids 12 and 13 in said first and second monomers can be mutated towards other amino acid residues in order to modulate their specificity towards nucleotides A, T, C and G and in particular to enhance this specificity.
  • other amino acid residues is intended any of the twenty natural amino acid residues or unnatural amino acids derivatives.
  • said core scaffolds of the first and second monomers can have the same scope of characteristics that those previously listed to describe the chimeric protein according to the present invention, regarding the origin of said core scaffolds, the number of RVDs comprised in said core scaffolds, the nature of those RVDs (natural, artificial or RVDs-like domains), the existence of additional N-terminus or C-terminus or both domains on these core scaffolds, the existence of one or several localization signals on these core scaffolds, the existence of one or several peptidic linkers on these core scaffolds to fuse one or several protein domains on these core scaffolds.
  • At least one monomer is selected from the group consisting of SEQ ID NO: 19 to 133, SEQ ID NO: 180-182, and SEQ ID NO: 186-188, a functional mutant, a variant or a derivative thereof.
  • said first and second monomers are selected from the group consisting of SEQ ID NO: 19 to 133, SEQ ID NO: 180-182, and SEQ ID NO: 186-188, functional mutants, variants or derivatives thereof.
  • said first and second monomers are fused by a peptidic linker forming a single polypeptide chain for simple and efficient vectorization.
  • said peptidic linker contains one or several active domains which allow its deployment under stimulation, as previously mentioned.
  • said first and second monomers have the same amino acid sequences and recognize the same nucleic acid sequence adjacent to said nucleic target sequence. In another embodiment, said first and second monomers have different amino acid sequences and recognize the same nucleic acid sequence adjacent to said nucleic target sequence, i.e first and second monomers are isoschizomers. In another embodiment, said first and second monomers have the same amino acid sequences and recognize different nucleic acid sequences adjacent to said nucleic target sequence because of TAL code degeneracy. In another embodiment, said first and second monomers have different amino acid sequences and recognize different nucleic acid sequences adjacent to said nucleic target sequence.
  • said chimeric protein according to the present invention binds a first and a second nucleic acid sequences which are on the same nucleic acid strand adjacent of said nucleic acid target sequence. In another embodiment, said chimeric protein according to the present invention binds a first and a second nucleic acid sequences which are adjacent to said nucleic acid target sequence but not on the same nucleic acid strand. In another embodiment, said chimeric protein according to the present invention binds a first and a second nucleic acid sequences which are located 5′ of said nucleic acid target sequence. In another embodiment, said chimeric protein according to the present invention binds a first and a second nucleic acid sequences which are located 3′ of said nucleic acid target sequence. In another embodiment, said chimeric protein according to the present invention binds a first nucleic sequence which is 5′ located of said nucleic acid sequence target and a second nucleic acid sequence which is 3′ located of said nucleic acid target sequence. In another embodiment, said
  • said chimeric protein according to the present invention binds a first and a second nucleic acid sequences which are adjacent to said nucleic acid target sequence and separated by a nucleic acid sequence (i.e. the spacer) of 5-40 base pairs (bp), i.e. the spacer length.
  • said chimeric protein according to the present invention binds a first and a second nucleic acid sequences which are adjacent to said nucleic acid target sequence and separated by a spacer of 8 bp length.
  • FIG. 9 Some structures of chimeric dimer proteins according to the invention are given on FIG. 9 .
  • said chimeric dimer protein according to the present invention can be associated with a third chimeric protein comprising:
  • Said third chimeric protein can have the same scope of characteristics that those previously listed regarding the origin of said core scaffold, the number of RVDs comprised in said core scaffold, the nature of those RVDs (natural, artificial or RVDs-like domains), the existence of additional N-terminus or C-terminus or both domains on this core scaffold, the existence of one or several localization signals on this core scaffold, the existence of one or several peptidic linkers on this core scaffold to fuse one or several protein domains on this core scaffold and the nature and characteristics of its protein(s) domain(s) or subdomain(s).
  • said third chimeric protein binds a nucleic acid sequence within the nucleic acid target sequence to process by said first and second monomers previously described.
  • said third chimeric protein can have a binding sequence within the spacer separating the first and a second nucleic acid sequences recognized and bound by the first and second monomers of the chimeric dimer protein according to the invention.
  • said third chimeric protein comprises a protein domain with a catalytic activity to process nucleic acid target sequence that is different from that of the catalytically active entity formed by the protein subdomains of respective first and second monomers of the chimeric dimer protein according to the present invention.
  • first and second monomer protein subdomains can form a catalytically active entity with a cleavase activity towards the nucleic acid target sequence and said third chimeric protein can comprise a protein domain with an exonuclease activity to increase the mutagenesis rate a the nucleic acid target sequence location.
  • said third chimeric protein binds a nucleic acid sequence located 5′ regarding the nucleic acid target sequence to process by said first and second monomers previously described.
  • said third chimeric protein binds a nucleic acid sequence located 3′ regarding the nucleic acid target sequence to process by said first and second monomers previously described.
  • said chimeric dimer protein according to the present invention can be associated with a core scaffold comprising a set of Repeat Variable Dipeptide regions (RVDs) able to bind a nucleic acid sequence adjacent to a nucleic acid target sequence to process wherein each RVD comprises a pair of amino acids responsible for recognizing one nucleotide selected from the group consisting of HD for recognizing C, NG for recognizing T, NI for recognizing A, NN for recognizing G or A, NS for recognizing A, C, G or T, HG for recognizing T, IG for recognizing T, NK for recognizing G, HA for recognizing C, ND for recognizing C, HI for recognizing C, HN for recognizing G, NA for recognizing G, SN for recognizing G or A, YG for recognizing T, TL for recognizing A, VT for recognizing A or G, SW for recognizing A, N* for recognizing C or T and H* for recognizing T (where * denotes a gap
  • said core scaffold associated with said chimeric dimer protein according to the present invention can have a binding sequence within the spacer separating the first and a second nucleic acid sequences recognized and bound by the first and second monomers of the chimeric dimer protein according to the invention.
  • said core scaffold associated with said chimeric dimer protein according to the present invention allows to control the processing activity of said chimeric dimer protein according to the present invention on its nucleic acid target sequence.
  • said core scaffold associated with said chimeric dimer protein according to the present invention allows to block the access of said chimeric dimer protein according to the present invention on its nucleic acid target sequence.
  • the expression of said core scaffold to control the processing activity of said chimeric dimer protein can be a cell-cycle or tissue dependent expression, allowing a cell-cycle or tissue dependent control of said chimeric dimer protein activity towards its nucleic acid target sequence.
  • a blocking core scaffold can also be used in combination with a chimeric protein according to the invention wherein said core scaffold binds the nucleic acid target sequence of said chimeric protein according to the invention to allow a control, a cell-cycle or time dependent control of said chimeric protein activity towards its nucleic acid sequence.
  • said chimeric protein according to the present invention can function as a trimer wherein a third monomer is derived from a Transcription Activator-Like Effector (TALE) and comprises:
  • TALE Transcription Activator-Like Effector
  • Said third monomer core can have the same scope of characteristics that those previously listed to describe a chimeric protein according to the present invention, regarding the origin of said core scaffold, the number of RVDs comprised in said core scaffold, the nature of those RVDs (natural, artificial or RVDs-like domains), the existence of additional N-terminus or C-terminus or both domains on this core scaffold, the existence of one or several localization signals on this core scaffold, the existence of one or several peptidic linkers on this core scaffold to fuse one or several protein domains on this core scaffold and the nature and characteristics of its protein(s) domain(s) or subdomain(s).
  • said third monomer binds a nucleic acid sequence within the nucleic acid target sequence to process by said first and second monomers previously described.
  • said third monomer can have a binding sequence within the spacer separating the first and a second nucleic acid sequences recognized and bound by the first and second monomers of the chimeric trimer protein according to the invention.
  • said third monomer binds a nucleic acid sequence located 5′ regarding the nucleic acid target sequence to process by said first and second monomers previously described.
  • said third monomer binds a nucleic acid sequence located 3′ regarding the nucleic acid target sequence to process by said first and second monomers previously described.
  • said third protein subdomain can be used to measure, quantify or provoke protein-protein interactions at said nucleic target sequence according to the present invention.
  • Said third protein subdomain can be a protein module or protein subdomain known to mediate protein-protein interaction in cell signaling.
  • Said third protein subdomain can be used for diagnosis, analytical or therapeutic applications.
  • Said third protein subdomain can be usable for applications such as Fluorescence Resonance Energy Transfer (FRET) as non-limiting example.
  • FRET Fluorescence Resonance Energy Transfer
  • said third protein subdomain is usable in FRET as a donor molecule, subdomains of first and second monomers being successive and compatible acceptor molecules.
  • said third protein subdomain is usable in FRET as an acceptor molecule, one of the subdomains of first and second monomers being respectively successive and compatible donor and acceptor molecules.
  • Said third protein subdomain can be coupled to a dye.
  • first, second and third subdomains of respective monomers constituting the chimeric trimer protein according to the invention can be a successive and compatible trio of “dyes” usable in FRET applications, more specific than the usual FRET applications using two successive and compatible dyes.
  • a recombinant polynucleotide encoding a chimeric protein or a monomer of a chimeric dimer protein as previously described according to the present invention.
  • Is also encompassed a vector comprising a recombinant polynucleotide encoding a chimeric protein or a monomer of a chimeric dimer protein as previously described according to the present invention.
  • Is also encompassed a host cell which comprises a vector and/or a recombinant polynucleotide encoding a chimeric protein or a monomer of a chimeric dimer protein as previously described according to the present invention.
  • non-human transgenic animal comprising a vector and/or a recombinant polynucleotide encoding a chimeric protein or a monomer of a chimeric dimer protein as previously described according to the present invention.
  • transgenic plant comprising a vector and/or a recombinant polynucleotide encoding a chimeric protein or a monomer of a chimeric dimer protein as previously described according to the present invention.
  • the present invention also relates to a kit comprising a chimeric protein or a monomer of a chimeric dimer protein according to the present invention or a vector and/or a recombinant polynucleotide encoding a chimeric protein or a monomer of a chimeric dimer protein as previously described according to the present invention and instructions for use said kit.
  • the present invention also relates to a composition comprising a chimeric protein or a monomer of a chimeric dimer protein according to the present invention or a vector and/or a recombinant polynucleotide encoding a chimeric protein or a monomer of a chimeric dimer protein as previously described according to the present invention and a carrier. More preferably, is a pharmaceutical composition comprising a chimeric protein or a monomer of a chimeric dimer protein according to the present invention or a vector and for a recombinant polynucleotide encoding a chimeric protein or a monomer of a chimeric dimer protein as previously described according to the present invention and a pharmaceutically active carrier.
  • the chimeric protein or a monomer of a chimeric dimer protein according to the present invention and a pharmaceutically acceptable excipient are administered in a therapeutically effective amount.
  • Such a combination is said to be administered in a “therapeutically effective amount” if the amount administered is physiologically significant.
  • An agent is physiologically significant if its presence results in a detectable change in the physiology of the recipient.
  • an agent is physiologically significant if its presence results in a decrease in the severity of one or more symptoms of the targeted disease and in a genome correction of the lesion or abnormality.
  • Vectors comprising targeting DNA and/or nucleic acid encoding chimeric protein or a monomer of a chimeric dimer protein according to the present invention can be introduced into a cell by a variety of methods (e.g., injection, direct uptake, projectile bombardment, liposomes, electroporation). Chimeric proteins or monomers of chimeric dimer proteins according to the present invention can be stably or transiently expressed into cells using expression vectors. Techniques of expression in eukaryotic cells are well known to those in the art. (See Current Protocols in Human Genetics: Chapter 12 “Vectors For Gene Therapy” & Chapter 13 “Delivery Systems for Gene Therapy”).
  • the chimeric protein or a monomer of a chimeric dimer protein according to the present invention is substantially non-immunogenic, i.e., engender little or no adverse immunological response.
  • a variety of methods for ameliorating or eliminating deleterious immunological reactions of this sort can be used in accordance with the invention.
  • the chimeric protein or a monomer of a chimeric dimer protein according to the present invention is substantially free of N-formyl methionine.
  • Another way to avoid unwanted immunological reactions is to conjugate the chimeric protein or a monomer of a chimeric dimer protein according to the present invention to polyethylene glycol (“PEG”) or polypropylene glycol (“PPG”) (preferably of 500 to 20,000 daltons average molecular weight (MW)).
  • PEG polyethylene glycol
  • PPG polypropylene glycol
  • Conjugation with PEG or PPG can provide non-immunogenic, physiologically active, water soluble chimeric proteins or monomers of chimeric dimer proteins conjugates with anti-viral activity. Similar methods also using a polyethylene-polypropylene glycol copolymer are described in Saifer et al. (U.S. Pat. No. 5,006,333).
  • the present invention also relates to methods for use of said chimeric protein or a monomer of a chimeric dimer protein according to the invention for various applications ranging from targeted DNA cleavage to targeted gene regulation.
  • catalytical activities such as nuclease activity (endonuclease and exonuclease), polymerase activity, kinase activity, phosphatase activity, methylase activity, topoisomerase activity, integrase activity, transposase activity, ligase activity, helicase activity, recombinase activity]
  • nuclease activity encoded DNA clease activity
  • phosphatase activity phosphatase activity
  • methylase activity topoisomerase activity
  • integrase activity transposase activity
  • ligase activity helicase activity
  • recombinase activity the number and the combinations of several protein domains fused to said core scaffold, chimeric protein or a
  • the present invention relates to a method for increasing targeted HR (and mutagenesis via NHEJ) when Double-Strand break activity is promoted in a chimeric protein or a monomer of a chimeric dimer protein according to the present invention targeting a DNA target sequence according to the invention.
  • the addition of at least two catalytically active cleavase domains according to the invention allows to increase Double-strand break-induced mutagenesis by leading to a loss of genetic information and preventing any scarless re-ligation of targeted genomic locus of interest by NHEJ.
  • the present invention relates to a method for increasing targeted HR in a more conservative fashion (with less mutagenesis via NHEJ) when Single-Strand Break activity is promoted in a chimeric protein or a monomer of a chimeric dimer protein according to the present invention targeting a DNA target sequence according to the invention.
  • the present invention relates to a method for increasing excision of a single-strand of DNA spanning the binding region of a chimeric protein or a monomer of a chimeric dimer protein according to the present invention when both one cleavase enhancer domain and one nickase enhancer domain, respectively, are fused to both N-terminus and C-terminus of at least one of the core scaffold of a chimeric protein or a monomer of a chimeric dimer protein according to the present invention.
  • the present invention relates to a method for treatment of a genetic disease caused by a mutation in a specific single double-stranded DNA target sequence in a gene, comprising administering to a subject in need thereof an effective amount of a chimeric protein or a monomer of a chimeric dimer protein, according to the present invention, a functional mutant, a variant or a derivative thereof.
  • said chimeric protein, a functional mutant, a variant or a derivative thereof for treatment of a genetic disease is independent of single-nucleotide polymorphisms (SNPs) that occur in the respective genomes of subjects in need thereof, due to TALE code degeneracy.
  • SNPs single-nucleotide polymorphisms
  • the present invention relates to a method for treatment of a genetic disease caused by a mutation in a nucleic acid target sequence, comprising administering to a subject in need thereof, in order to cure said genetic disease, an effective amount of a chimeric protein or a monomer of a chimeric dimer protein, according to the present invention, a functional mutant, a variant or a derivative thereof wherein said chimeric protein overcomes the genomic variations of subjects due to SNPs.
  • Said method of the present invention allows the treatment of said genetic disease by constructing and administering one unique chimeric protein according to the invention to every subjects in need thereof, whatever SNPs profiles around said mutation responsible for genetic disease in these subjects.
  • said method of the present invention avoids the need to construct and administer one personalized chimeric protein for each subject in need thereof that takes into account each SNP profile around the mutation to cure.
  • said unique chimeric protein to cure said genetic disease according to the present invention can comprise degenerated RVDs in its core scaffold such as NN for recognizing G or A, NS for recognizing A, C, G or T or SN for recognizing G or A.
  • said method of the present invention allows to treat this genetic disease by constructing and administering a unique chimeric protein according to the present invention wherein said core scaffold of said chimeric protein comprises a SN-type RVD for recognizing either G, either A, present in the genome of every subjects at SNPs location, in order to bind said genomic sequence around said SNP and treat said genetic disease.
  • said method can be used to overcome interspecies sequence variations.
  • the present invention relates to a method for inserting a transgene into a specific single double-stranded DNA target sequence of a genomic locus of a cell, tissue or non-human animal, or a plant wherein at least one chimeric protein or a monomer of a chimeric dimer protein of the present invention is transitory or not introduced into said cell, tissue, non-human animal or plant.
  • the present invention relates to a method for optimizing the control of nucleic acid processing activity of a chimeric protein (or a monomer of a chimeric dimer protein according to the present invention) within its nucleic acid target sequence.
  • the present invention relates to a method for optimizing the control of nucleic acid processing activity of a chimeric protein (or a monomer of a chimeric dimer protein according to the present invention) within its nucleic acid target sequence by associating an optimum core scaffold structure to an optimum spacer length.
  • the present invention relates to a method for optimizing the control of nucleic acid processing activity of a chimeric protein (or a monomer of a chimeric dimer protein according to the present invention) within its nucleic acid target sequence by associating an optimum C-terminal truncation of the core scaffold structure to an optimum spacer length.
  • the present invention relates to a method for optimizing the control of nucleic acid processing activity of a chimeric protein (or a monomer of a chimeric dimer protein according to the present invention) within its nucleic acid target sequence by associating an optimum N-terminal truncation of the core scaffold structure to an optimum spacer length.
  • the present invention relates to a method for optimizing the control of nucleic acid processing activity of a chimeric protein (or a monomer of a chimeric dimer protein according to the present invention) within its nucleic acid target sequence by associating two optimum C-terminal truncations of the core scaffold structure to an optimum spacer length as illustrated on FIGS. 10 and 11 .
  • the present invention relates to a method for optimizing the control of nucleic acid processing activity of a chimeric protein (or a monomer of a chimeric dimer protein according to the present invention) within its nucleic acid target sequence by associating two optimum N-terminal truncations of the core scaffold structure to an optimum spacer length.
  • the present invention relates to a method for optimizing the control of double-stranded break activity of a chimeric protein (or a monomer of a chimeric dimer protein according to the present invention) within its nucleic acid target sequence.
  • the present invention relates to a method for optimizing the control of double-stranded break activity of a chimeric protein (or a monomer of a chimeric dimer protein according to the present invention) within its nucleic acid target sequence by associating an optimum core scaffold structure to an optimum spacer length.
  • the present invention relates to a method for optimizing the control of double-stranded break activity of a chimeric protein (or a monomer of a chimeric dimer protein according to the present invention) within its nucleic acid target sequence by associating an optimum C-terminal truncation of the core scaffold structure to an optimum spacer length.
  • the present invention relates to a method for optimizing the control of double-stranded break activity of a chimeric protein (or a monomer of a chimeric dimer protein according to the present invention) within its nucleic acid target sequence by associating two optimum C-terminal truncations of the core scaffold structure to an optimum spacer length.
  • the present invention relates to a method for optimizing the control of double-stranded break activity of a chimeric protein (or a monomer of a chimeric dimer protein according to the present invention) within its nucleic acid target sequence by associating two different optimum C-terminal truncations of the core scaffold structure to an optimum spacer length.
  • the present invention relates to a method for optimizing the control of double-stranded break activity of a chimeric protein (or a monomer of a chimeric dimer protein according to the present invention) within its nucleic acid target sequence by associating an optimum N-terminal truncation of the core scaffold structure to an optimum spacer length.
  • the present invention relates to a method for optimizing the control of double-stranded break activity of a chimeric protein (or a monomer of a chimeric dimer protein according to the present invention) within its nucleic acid target sequence by associating two optimum N-terminal truncations of the core scaffold structure to an optimum spacer length.
  • the present invention relates to a method for optimizing the control of double-stranded break activity of a chimeric protein (or a monomer of a chimeric dimer protein according to the present invention) within its nucleic acid target sequence by associating two different optimum N-terminal truncations of the core scaffold structure to an optimum spacer length.
  • the present invention relates to a method for increasing the number of targets that can be reach by a chimeric protein of the present invention, in a locus of a given genome, by associating to a spacer length imposed by the sequence of said locus of a given genome an optimal C-terminal truncation of the core scaffold of said chimeric protein as illustrated on FIG. 11 .
  • the present invention relates to a method for increasing the number of targets that can be reach by a chimeric protein of the present invention, in a locus of a given genome, by associating to a spacer length imposed by the sequence of said locus of a given genome an optimal C-terminal truncation of the core scaffold of said chimeric protein, wherein said spacer length can be comprised between 5 and 40 bp.
  • the present invention relates to a method for increasing the number of targets that can be reach by a chimeric protein of the present invention, in a locus of a given genome, by associating to a spacer length imposed by the sequence of said locus of a given genome an optimal C-terminal truncation of the core scaffold of said chimeric protein, wherein said spacer length can be comprised between 8 and 40 bp.
  • the present invention relates to a method for increasing the number of targets that can be reach by a chimeric protein of the present invention, in a locus of a given genome, by associating to a spacer length imposed by the binding site within said locus an optimal C-terminal truncation of the core scaffold of said chimeric protein.
  • the present invention relates to a method for increasing the number of targets that can be reach by a chimeric protein of the present invention, in a locus of a given genome, by associating to a spacer length imposed by the binding site within said locus, wherein said sequence of said locus is poor in T, an optimal C-terminal truncation of the core scaffold of said chimeric protein.
  • the present invention relates to a method for optimizing the control of double-stranded break location of a chimeric protein (or a monomer of a chimeric dimer protein according to the present invention) within its nucleic acid target sequence by associating two optimum C-terminal truncations of the core scaffold structure to an optimum spacer length wherein said association allows placing the cleavage site at a more desired location within said spacer as illustrated on FIG. 10 .
  • said association between two optimum C-terminal truncations of the core scaffold structure and an optimum spacer length allows placing the cleavage site at a more desired location that is not in the center of said spacer as illustrated on FIGS. 10 B and C.
  • said association between two optimum C-terminal truncations of the core scaffold structure and an optimum spacer length allows placing the cleavage site at a more desired location that is in the left part (i.e 5′ located regarding the center of the spacer) of said spacer as illustrated on FIG. 10 C.
  • said association between two optimum C-terminal truncations of the core scaffold structure and an optimum spacer length allows placing the cleavage site at a more desired location that is in the right part (i.e 3′ located regarding the center of the spacer) of said spacer as illustrated on FIG. 10 B.
  • said association between two optimum C-terminal truncations of the core scaffold structure and an optimum spacer length allows placing the cleavage site at the center of said spacer as illustrated on FIG. 10 A.
  • the present invention relates to a method for increasing the number of targets that can be reach by a chimeric protein of the present invention, in a locus of a given genome, by associating to a spacer length imposed by the sequence of said locus of a given genome an optimal N-terminal truncation of the core scaffold of said chimeric protein.
  • the present invention relates to a method for increasing the number of targets that can be reach by a chimeric protein of the present invention, in a locus of a given genome, by associating to a spacer length imposed by the sequence of said locus of a given genome an optimal N-terminal truncation of the core scaffold of said chimeric protein, wherein said spacer length can be comprised between 5 and 40 bp.
  • the present invention relates to a method for increasing the number of targets that can be reach by a chimeric protein of the present invention, in a locus of a given genome, by associating to a spacer length imposed by the sequence of said locus of a given genome an optimal N-terminal truncation of the core scaffold of said chimeric protein, wherein said spacer length can be comprised between 8 and 40 bp.
  • the present invention relates to a method for increasing the number of targets that can be reach by a chimeric protein of the present invention, in a locus of a given genome, by associating to a spacer length imposed by the binding site within said locus an optimal N-terminal truncation of the core scaffold of said chimeric protein.
  • the present invention relates to a method for increasing the number of targets that can be reach by a chimeric protein of the present invention, in a locus of a given genome, by associating to a spacer length imposed by the binding site within said locus, wherein said sequence of said locus is poor in T, an optimal N-terminal truncation of the core scaffold of said chimeric protein.
  • the present invention relates to a method for optimizing the control of double-stranded break location of a chimeric protein (or a monomer of a chimeric dimer protein according to the present invention) within its nucleic acid target sequence by associating two optimum N-terminal truncations of the core scaffold structure to an optimum spacer length wherein said association allows placing the cleavage site at a more desired location within said spacer.
  • said association between two optimum N-terminal truncations of the core scaffold structure and an optimum spacer length allows placing the cleavage site at a more desired location that is not in the center of said spacer.
  • said association between two optimum N-terminal truncations of the core scaffold structure and an optimum spacer length allows placing the cleavage site at a more desired location that is in the left part (i.e 5′ located regarding the center of the spacer) of said spacer.
  • said association between two optimum N-terminal truncations of the core scaffold structure and an optimum spacer length allows placing the cleavage site at a more desired location that is in the right part (i.e 3′ located regarding the center of the spacer) of said spacer.
  • said association between two optimum N-terminal truncations of the core scaffold structure and an optimum spacer length allows placing the cleavage site at the center of said spacer.
  • the present invention relates to a method for optimizing the control of nucleic acid processing activity, the double-stranded break activity of a chimeric protein (or a monomer of a chimeric dimer protein according to the present invention) within its nucleic acid target sequence, by respectively associating two optimum N-terminal and C-terminal truncations of the core scaffold structure to an optimum spacer length.
  • the present invention relates to a method for optimizing the control of double-stranded break location of a chimeric protein (or a monomer of a chimeric dimer protein according to the present invention) within its nucleic acid target sequence by respectively associating two optimum N-terminal and C-terminal truncations of the core scaffold structure to an optimum spacer length wherein said association allows placing the cleavage site at a more desired location within said spacer
  • Amino acid residues in a polypeptide sequence are designated herein according to the one-letter code, in which, for example, Q means Gln or Glutamine residue, R means Arg or Arginine residue and D means Asp or Aspartic acid residue.
  • Amino acid substitution means the replacement of one amino acid residue with another, for instance the replacement of an Arginine residue with a Glutamine residue in a peptide sequence is an amino acid substitution.
  • DNA or nucleic acid processing activity refers to a particular/given enzymatic activity of a protein domain comprised in a chimeric protein according to the invention such as in the expression “a protein domain to process said nucleic acid target sequence”.
  • Said DNA or nucleic acid processing activity can refer to a cleavage activity, either a cleavase activity either a nickase activity, more broadly a nuclease activity but also a polymerase activity, a kinase activity, a phosphatase activity, a methylase activity, a topoisomerase activity, an integrase activity, a transposase activity, a ligase, a helicase or recombinase activity as non-limiting examples.
  • Nucleotides are designated as follows: one-letter code is used for designating the base of a nucleoside: a is adenine, t is thymine, c is cytosine, and g is guanine.
  • r represents g or a (purine nucleotides)
  • k represents g or t
  • s represents g or c
  • w represents a or t
  • m represents a or c
  • y represents t or c (pyrimidine nucleotides)
  • d represents g, a or t
  • v represents g, a or c
  • b represents g, t or c
  • h represents a, t or c
  • n represents g, a, t or c.
  • variant by “variant”, “chimeric protein variant” or “TALEN variant”, it is intended a chimeric protein, a chimeric protein derived from a Transcription Activator-like Effector (TALE) or a TALEN obtained by replacement of at least one residue in the amino acid sequence of the parent chimeric protein, parent chimeric protein derived from a Transcription Activator-like Effector (TALE) or parent TALEN with at least a different amino acid.
  • TALE Transcription Activator-like Effector
  • TALE Transcription Activator-like Effector
  • peptide linker or “peptidic linker” it is intended to mean a peptide sequence which allows the connection of different monomers or different parts comprised in a fusion protein such as between a core scaffold and a protein domain in a chimeric protein according to the present invention and which allows the adoption of a correct conformation for said fusion protein activity and/or specificity.
  • Peptide linkers can be of various sizes, from 3 amino acids to 50 amino acids as a non limiting indicative range. Peptide linkers can also be qualified as structured or unstructured. Peptide linkers can be qualified as active linkers when they comprise active domains that are able to change their structural conformation under appropriate stimulation.
  • one cell type related to the chosen cell type or organism is intended a cell type or an organism sharing characteristics with said chosen cell type or said chosen organism; this cell type or organism related to the chosen cell type or organism, can be derived from said chosen cell type or organism or not.
  • subdomain it is intended a protein subdomain or a protein part that interacts with another protein subdomain or protein part to form an active entity and/or a catalytic active entity possibly bearing nucleic acid or DNA processing activity of said chimeric protein according to the invention.
  • targeting DNA construct/minimal repair matrix/repair matrix it is intended to mean a DNA construct comprising a first and second portion that are homologous to regions 5′ and 3′ of the DNA target in situ.
  • the DNA construct also comprises a third portion positioned between the first and second portion which comprise some homology with the corresponding DNA sequence in situ or alternatively comprise no homology with the regions 5′ and 3′ of the DNA target in situ.
  • a homologous recombination event is stimulated between the genome containing the targeted gene comprised in the locus of interest and the repair matrix, wherein the genomic sequence containing the DNA target is replaced by the third portion of the repair matrix and a variable part of the first and second portions of the repair matrix.
  • mutant is intended a catalytically active mutant of a protein or a protein domain; such mutant can have the same activity compared to its parent protein or protein domain or additional properties.
  • This definition applies to chimeric proteins or protein domains that constitute chimeric proteins according to the present invention.
  • derivatives of these proteins or protein domains that comprise the entirety or part of these proteins or protein domains fused to other proteic or chemical parts such as tags, antibodies, polyethylene glycol as non-limiting examples.
  • single polypeptide chain is used to qualify a chimeric protein according to the invention which functions as a dimer wherein one first monomer and one second monomer are fused by a peptidic linker.
  • DNA target is intended a polynucleotide sequence that can be processed by a chimeric protein according to the present invention.
  • DNA target refers to a specific DNA location, preferably a genomic location in a cell, but also a portion of genetic material that can exist independently to the main body of genetic material such as plasmids, episomes, virus, transposons or in organelles such as mitochondria or chloroplasts as non-limiting examples.
  • the nucleic acid target sequence is defined by the 5′ to 3′ sequence of one strand of said target, as indicate above for Avr15 (SEQ ID NO: 6) as a non-limiting example.
  • Adjacent is used to distinguish between 1) the nucleic acid sequence recognized and bound by a set of specific RVDs comprised in the core scaffold of said chimeric protein according to the invention and 2) the nucleic acid target sequence to be processed by said chimeric protein according to the invention, said nucleic sequences 1) and 2) being adjacent.
  • said chimeric protein according to the invention functions as a dimer comprising respectively two monomers
  • the term adjacent is equally used to qualify the nucleic acid target sequence regarding the first nucleic acid sequence recognized and bound by the first monomer and to qualify the nucleic acid target sequence regarding the second nucleic acid sequence recognized and bound by the second monomer.
  • the nucleic acid sequence adjacent to the nucleic acid target sequence is meant the recognition/binding site of said chimeric protein according to the invention.
  • delivery vector or “delivery vectors” is intended any delivery vector which can be used in the present invention to put into cell contact (i.e “contacting”) or deliver inside cells or subcellular compartments agents/chemicals and molecules (proteins or nucleic acids) needed in the present invention. It includes, but is not limited to liposomal delivery vectors, viral delivery vectors, drug delivery vectors, chemical carriers, polymeric carriers, lipoplexes, polyplexes, dendrimers, microbubbles (ultrasound contrast agents), nanoparticles, emulsions or other appropriate transfer vectors. These delivery vectors allow delivery of molecules, chemicals, macromolecules (genes, proteins), or other vectors such as plasmids, peptides developed by Diatos. In these cases, delivery vectors are molecule carriers. By “delivery vector” or “delivery vectors” is also intended delivery methods to perform transfection.
  • vector refers to a nucleic acid molecule capable of transporting another nucleic acid to which it has been linked.
  • a “vector” in the present invention includes, but is not limited to, a viral vector, a plasmid, a RNA vector or a linear or circular DNA or RNA molecule which may consists of a chromosomal, non chromosomal, semi-synthetic 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.
  • Viral vectors include retrovirus, adenovirus, parvovirus (e.g. adenoassociated viruses), coronavirus, negative strand RNA viruses such as orthomyxovirus (e.g., influenza virus), rhabdovirus (e.g., rabies and vesicular stomatitis virus), paramyxovirus (e.g.
  • RNA viruses such as picornavirus and alphavirus
  • double-stranded DNA viruses including adenovirus, herpesvirus (e.g., Herpes Simplex virus types 1 and 2, Epstein-Barr virus, cytomega-lovirus), and poxvirus (e.g., vaccinia, fowlpox and canarypox).
  • herpesvirus e.g., Herpes Simplex virus types 1 and 2, Epstein-Barr virus, cytomega-lovirus
  • poxvirus e.g., vaccinia, fowlpox and canarypox
  • Other viruses include Norwalk virus, togavirus, flavivirus, reoviruses, papovavirus, hepadnavirus, and hepatitis virus, for example.
  • retroviruses examples include: avian leukosis-sarcoma, mammalian C-type, B-type viruses, D type viruses, HTLV-BLV group, lentivirus, spumavirus (Coffin, J. M., Retroviridae: The viruses and their replication, In Fundamental Virology, Third Edition, B. N. Fields, et al., Eds., Lippincott-Raven Publishers, Philadelphia, 1996).
  • lentiviral vector HIV-Based lentiviral vectors that are very promising for gene delivery because of their relatively large packaging capacity, reduced immunogenicity and their ability to stably transduce with high efficiency a large range of different cell types.
  • Lentiviral vectors are usually generated following transient transfection of three (packaging, envelope and transfer) or more plasmids into producer cells.
  • lentiviral vectors enter the target cell through the interaction of viral surface glycoproteins with receptors on the cell surface.
  • the viral RNA undergoes reverse transcription, which is mediated by the viral reverse transcriptase complex.
  • the product of reverse transcription is a double-stranded linear viral DNA, which is the substrate for viral integration in the DNA of infected cells.
  • integrated lentiviral vectors or LV
  • integrated lentiviral vectors or LV
  • non integrative lentiviral vectors or NILV is meant efficient gene delivery vectors that do not integrate the genome of a target cell through the action of the virus integrase.
  • One type of preferred vector is an episome, i.e., a nucleic acid capable of extra-chromosomal replication.
  • Preferred vectors are those capable of autonomous replication and/or expression of nucleic acids to which they are linked.
  • Vectors capable of directing the expression of genes to which they are operatively linked are referred to herein as “expression vectors.
  • a vector according to the present invention comprises, but is not limited to, a YAC (yeast artificial chromosome), a BAC (bacterial artificial), a baculovirus vector, a phage, a phagemid, a cosmid, a viral vector, a plasmid, a RNA vector or a linear or circular DNA or RNA molecule which may consist of chromosomal, non chromosomal, semi-synthetic or synthetic DNA.
  • expression vectors of utility in recombinant DNA techniques are often in the form of “plasmids” which refer generally to circular double stranded DNA loops which, in their vector form are not bound to the chromosome.
  • Vectors can comprise selectable markers, for example: neomycin phosphotransferase, histidinol dehydrogenase, dihydrofolate reductase, hygromycin phosphotransferase, herpes simplex virus thymidine kinase, adenosine deaminase, glutamine synthetase, and hypoxanthine-guanine phosphoribosyl transferase for eukaryotic cell culture; TRP1 for S. cerevisiae ; tetracyclin, rifampicin or ampicillin resistance in E. coli .
  • selectable markers for example: neomycin phosphotransferase, histidinol dehydrogenase, dihydrofolate reductase, hygromycin phosphotransferase, herpes simplex virus thymidine kinase, adenosine deaminase
  • said vectors are expression vectors, wherein a sequence encoding a polypeptide of interest is placed under control of appropriate transcriptional and translational control elements to permit production or synthesis of said polypeptide. Therefore, said polynucleotide is comprised in an expression cassette. More particularly, the vector comprises a replication origin, a promoter operatively linked to said encoding polynucleotide, a ribosome binding site, a RNA-splicing site (when genomic DNA is used), a polyadenylation site and a transcription termination site. It also can comprise an enhancer or silencer elements. Selection of the promoter will depend upon the cell in which the polypeptide is expressed. Suitable promoters include tissue specific and/or inducible promoters.
  • inducible promoters are: eukaryotic metallothionine promoter which is induced by increased levels of heavy metals, prokaryotic lacZ promoter which is induced in response to isopropyl- ⁇ -D-thiogalacto-pyranoside (IPTG) and eukaryotic heat shock promoter which is induced by increased temperature.
  • tissue specific promoters are skeletal muscle creatine kinase, prostate-specific antigen (PSA), ⁇ -antitrypsin protease, human surfactant (SP) A and B proteins, ⁇ -casein and acidic whey protein genes.
  • Inducible promoters may be induced by pathogens or stress, more preferably by stress like cold, heat, UV light, or high ionic concentrations (reviewed in Potenza C et al. 2004, In vitro Cell Dev Biol 40:1-22). Inducible promoter may be induced by chemicals (reviewed in (Moore, Samalova et al. 2006); (Padidam 2003); (Wang, Zhou et al. 2003); (Zuo and Chua 2000).
  • Delivery vectors and vectors can be associated or combined with any cellular permeabilization techniques such as sonoporation or electroporation or derivatives of these techniques.
  • cell or cells any prokaryotic or eukaryotic living cells, cell lines derived from these organisms for in vitro cultures, primary cells from animal or plant origin.
  • primary cell or “primary cells” are intended cells taken directly from living tissue (i.e. biopsy material) and established for growth in vitro, that have undergone very few population doublings and are therefore more representative of the main functional components and characteristics of tissues from which they are derived from, in comparison to continuous tumorigenic or artificially immortalized cell lines. These cells thus represent a more valuable model to the in vivo state they refer to.
  • eukaryotic cells refer to a fungal, plant or animal cell or a cell line derived from the organisms listed below and established for in vitro culture. More preferably, the fungus is of the genus Aspergillus, Penicillium, Acremonium, Trichoderma, Chrysoporium, Mortierella, Kluyveromyces or Pichia ; More preferably, the fungus is of the species Aspergillus niger, Aspergillus nidulans, Aspergillus oryzae, Aspergillus terreus, Penicillium chrysogenum, Penicillium citrinum, Acremonium Chrysogenum, Trichoderma reesei, Mortierella alpine, Chrysosporium lucknowense, Kluyveromyces lactis, Pichia pastoris or Pichia ciferrii.
  • the plant is of the genus Arabidospis, Nicotiana, Solanum, lactuca, Brassica, Oryza, Asparagus, Pisum, Medicago, Zea, Hordeum, Secale, Triticum, Capsicum, Cucumis, Cucurbita, Citrullis, Citrus, Sorghum ; More preferably, the plant is of the species Arabidospis thaliana, Nicotiana tabaccum, Solanum lycopersicum, Solanum tuberosum, Solanum melongena, Solanum esculentum, Lactuca saliva, Brassica napus, Brassica oleracea, Brassica rapa, Oryza glaberrima, Oryza sativa, Asparagus officinalis, Pisum sativum, Medicago sativa, zea mays, Hordeum vulgare, Secale cereal, Triticum aestivum, Triticum durum, Capsicum sativus,
  • the animal cell is of the genus Homo, Rattus, Mus, Sus, Bos, Danio, Canis, Felis, Equus, Salmo, Oncorhynchus, Gallus, Meleagris, Drosophila, Caenorhabditis ; more preferably, the animal cell is of the species Homo sapiens, Rattus norvegicus, Mus musculus, Sus scrofa, Bos taurus, Danio rerio, Canis lupus, Felis catus, Equus caballus, Salmo salar, Oncorhynchus mykiss, Gallus gallus, Meleagris gallopavo, Drosophila melanogaster, Caenorhabditis elegans.
  • the cell can be a plant cell, a mammalian cell, a fish cell, an insect cell or cell lines derived from these organisms for in vitro cultures or primary cells taken directly from living tissue and established for in vitro culture.
  • cell lines can be selected from the group consisting of CHO-K1 cells; HEK293 cells; Caco2 cells; U2-OS cells; NIH 3T3 cells; NSO cells; SP2 cells; CHO-S cells; DG44 cells; K-562 cells, U-937 cells; MRC5 cells; IMR90 cells; Jurkat cells; HepG2 cells; HeLa cells; HT-1080 cells; HCT-116 cells; Hu-h7 cells; Huvec cells; Molt 4 cells.
  • All these cell lines can be modified by the method of the present invention to provide cell line models to produce, express, quantify, detect, study a gene or a protein of interest; these models can also be used to screen biologically active molecules of interest in research and production and various fields such as chemical, biofuels, therapeutics and agronomy as non-limiting examples.
  • homologous is intended a sequence with enough identity to another one to lead to homologous recombination between sequences, more particularly having at least 95% identity, preferably 97% identity and more preferably 99%.
  • identity refers to sequence identity between two nucleic acid molecules or polypeptides. Identity can be determined by comparing a position in each sequence which may be aligned for purposes of comparison. When a position in the compared sequence is occupied by the same base, then the molecules are identical at that position. A degree of similarity or identity between nucleic acid or amino acid sequences is a function of the number of identical or matching nucleotides at positions shared by the nucleic acid sequences. Various alignment algorithms and/or programs may be used to calculate the identity between two sequences, including FASTA, or BLAST which are available as a part of the GCG sequence analysis package (University of Wisconsin, Madison, Wis.), and can be used with, e.g., default setting.
  • mutation is intended the substitution, deletion, insertion of one or more nucleotides/amino acids in a polynucleotide (cDNA, gene) or a polypeptide sequence. Said mutation can affect the coding sequence of a gene or its regulatory sequence. It may also affect the structure of the genomic sequence or the structure/stability of the encoded mRNA.
  • double-strand break-induced mutagenesis refers to a mutagenesis event consecutive to an NHEJ event following an endonuclease-induced DSB, leading to insertion/deletion at the cleavage site of an endonuclease.
  • 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.
  • 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.
  • gene of interest refers to any nucleotide sequence encoding a known or putative gene product.
  • locus is the specific physical location of a DNA sequence (e.g. of a gene) on a chromosome.
  • locus usually refers to the specific physical location of a chimeric protein's nucleic target sequence on a chromosome.
  • locus which comprises a target sequence that is recognized and cleaved by a chimeric protein according to the invention, is referred to as “locus according to the invention”.
  • the expression “genomic locus of interest” is used to qualify a nucleic acid sequence in a genome that can be a putative target for a double-strand break according to the invention.
  • genomic locus of interest of the present invention can not only qualify a nucleic acid sequence that exists in the main body of genetic material (i.e. in a chromosome) of a cell but also a portion of genetic material that can exist independently to said main body of genetic material such as plasmids, episomes, virus, transposons or in organelles such as mitochondria or chloroplasts as non-limiting examples.
  • loss of genetic information is understood the elimination or addition of at least one given DNA fragment (at least one nucleotide) or sequence within the intervening sequence between at least two processing sites of the chimeric protein of the present invention or between two chimeric proteins according to the present invention.
  • This loss of genetic information can be, as a non-limiting example, the elimination of an intervening sequence between two processing sites of two chimeric proteins according to the present invention.
  • this loss of genetic information can also be an excision of a single-strand of DNA spanning the binding region of a chimeric protein according to the present invention
  • scarless re-ligation or “scarless religation” is intended the perfect re-ligation event, without loss of genetic information (no insertion/deletion events) of the DNA broken ends through NHEJ process after the creation of a double-strand break event.
  • Imprecise NHEJ is intended the re-ligation of nucleic acid ends generated by a DSB, with insertions or deletions of nucleotides. Imprecise NHEJ is an outcome and not a repair pathway and can result from different NHEJ pathways (Ku dependent or Ku independent as non-limiting examples).
  • fusion protein is intended the result of a well-known process in the art consisting in the joining of two or more genes which originally encode for separate proteins or part of them, the translation of said “fusion gene” resulting in a single polypeptide with functional properties derived from each of the original proteins.
  • chimeric protein any fusion protein comprising a core scaffold comprising a set of RVDs to bind a nucleic acid sequence and one protein domain to process a nucleic acid target sequence adjacent to said bound nucleic acid sequence.
  • Said chimeric protein according to the present invention can function as a dimer wherein each monomer (a monomer of a chimeric dimer protein in this case) constituting said chimeric dimer protein comprises a set of RVDs to bind a nucleic acid sequence and one protein domain to process a nucleic acid target sequence adjacent to said bound nucleic acid sequence.
  • protein domain or “catalytic domain” is meant the nucleic acid target sequence processing part of said chimeric protein according to the present invention.
  • Said protein domain or catalytic domain can provide any catalytical activity as classified and named according to the reaction they catalyze [Enzyme Commission number (EC number) at http://www.chem.qmul.ac.uk/iubmb/enzyme/)].
  • Said protein domain or catalytic domain can be a catalytically active entity by itself.
  • Said protein domain or catalytic domain can be a protein subdomain that needs to interact with another protein subdomain to form a dimeric protein domain active entity.
  • said protein domain or catalytic domain can be a first protein subdomain interacting with a second protein subdomain of another chimeric monomer protein according to the invention to form the catalytically active protein entity able to process the nucleic acid target sequence.
  • TALEN TALE-nuclease
  • TALEN Transcription Activator Like Effector
  • Said TALEN is a subclass of chimeric protein according to the present invention.
  • spacer is meant the nucleic acid area that separates the two nucleic acid sequences recognized and bound by each monomer constituting a chimeric dimer protein according to the invention.
  • spacer length is meant the nucleic acid distance that separates the two nucleic acid sequences recognized and bound by each monomer constituting a chimeric dimer protein according to the invention.
  • said nucleic acid target sequence of the chimeric protein according to the present invention can be encompassed in said spacer.
  • Said nucleic acid target sequence of the chimeric protein according to the present invention can be identical to said spacer.
  • Said nucleic acid target sequence of the chimeric protein according to the present invention can be different of said spacer.
  • phrases “selected from the group consisting of,” “chosen from,” and the like include mixtures of the specified materials.
  • the amino acid sequences of the N-terminal, C-terminal domains and RVDS were based on the AvrBs3 TAL (ref: GenBank: X16130.1, SEQ ID NO: 1).
  • Nter wt or WT Nter (SEQ ID NO: 292), i.e. corresponding to the N terminal domain of natural AvrBs3 (SEQ ID NO: 1) except an Ala residue in position 2]
  • Cter wt or WT Cter (SEQ ID NO: 400) lacking the activation domain of the C-terminal domain of natural AvrBs3 (SEQ ID NO: 1)
  • nuclease catalytic head were synthesized (TopGene Technologies) and subcloned into the pCLS0542 (SEQ ID NO: 2) yeast expression plasmid, using NcoI and EagI restriction enzymes, leading to the backbone plasmid pCLS7183 (referred as backbone wt, SEQ ID NO: 3).
  • the C-terminal and the N-terminal domains are separated by two BsmBI restriction sites.
  • the AvrBs3-derived set of repeat domains (RVDs) (SEQ ID NO: 4) was subcloned in the pCLS7183 using type IIs restriction enzymes BsmBI for the receiving plasmid and BbvI and SfaNI for the inserted RVD sequence, leading to pCLS7184 and subsequent AvrBs3-derived TALEN (referred as control wt, SEQ ID NO: 5). All the yeast target reporter plasmids containing the TALEN DNA target sequences were constructed as previously described (International PCT Applications WO 2004/067736 and in (Epinat, Arnould et al.
  • the AvrBs3-derived TALEN was tested at 30° C. in our yeast SSA assay previously described (International PCT Applications WO 2004/067736 and in (Epinat, Arnould et al. 2003; Chames, Epinat et al. 2005; Arnould, Chames et al. 2006; Smith, Grizot et al. 2006) as homodimer (two identical recognition sequences are placed facing each other on both DNA strands) on the target Avr15 (SEQ ID NO: 6, Table 3). TALEN cleavage activity levels on its respective target in yeast are shown on FIG. 2 .
  • the DNA encoding a HA tag, the N-terminal domain (referred as Nter wt), the C-terminal domain (referred as Cter wt) and the nuclease catalytic head were synthesized and subcloned into the pCLS1853 (SEQ ID NO: 7) mammalian expression plasmid, using AscI and XhoI restriction enzymes, leading to the backbone plasmid pCLS7111 (SEQ ID NO: 8).
  • the C-terminal and the N-terminal domains are separated by two BsmBI restriction sites.
  • the AvrBs3-derived set of repeat domains (SEQ ID NO: 4) was subcloned in the pCLS7111 using type IIs restriction enzymes BsmBI for the receiving plasmid and BbvI and SfaNI for the inserted RVD sequence, leading to pCLS7509 and subsequent TALEN (SEQ ID NO: 9).
  • All the mammalian target reporter plasmids containing the TALEN DNA target sequences were constructed using standard gateway Gateway protocol (INVITROGEN) into a CHO reporter vector (Grizot, Epinat et al.; Arnould, Chames et al. 2006).
  • Activity of AvrBs3-derived TALEN was tested in our extrachromosomal assay in mammalian cells (CHO K1) as homodimer (two identical recognition sequences are placed facing each other on both DNA strands) on the target Avr15 (SEQ ID NO: 6, Table 3).
  • CHO K1 cells were transfected in a 96-well plate format with 75 or 200 ng of target vector and an increasing quantity of each variant DNA from 0.7 to 25 ng, in the presence of PolyFect reagent (1 ⁇ L per well). The total amount of transfected DNA was completed to 125 or 250 ng (target DNA, variant DNA, carrier DNA) using an empty vector. 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 (Grizot, Epinat et al.).
  • TALEN cleavage activity levels on their respective targets in mammalian cells are shown on FIG. 3 .
  • Truncations of the first 153 (numbering based on SEQ ID NO:1)_amino acids residues of the N-terminal domain of the AvrBs3-derived TALEN (pCLS7184, SEQ ID NO: 5) were realized.
  • DNA sequence corresponding to amino acids D154 to N228 was amplified by PCR, using the backbone plasmid pCLS7183 (referred as backbone wt, SEQ ID NO: 3) as template, to add a NcoI restriction site in 5′ and a XmnI restriction site in 3′.
  • the PCR construct was subcloned in the TALEN yeast expression backbone (pCLS7183, SEQ ID NO: 3) to replace the sequence of the N-terminal domain (referred as Nter wt) comprised between the NcoI and XmnI restriction sites, leading to pCLS7724 (SEQ ID NO: 10). All DNA sequences were validated by sequencing.
  • the AvrBs3-derived set of repeat domains (SEQ ID NO: 4) was subcloned in the pCLS7724 using type IIs restriction enzymes BsmBI for the receiving plasmid and BbvI and SfaNI for the inserted RVD sequence, leading to pCLS7725 and subsequent TALEN (SEQ ID NO: 11).
  • This truncated variant was screened in our yeast SSA assay (see Example 1) as homodimer (two identical recognition sequences are placed facing each other on both DNA strands) on the target Avr15 (SEQ ID NO: 6, Table 3). Activity level of the truncated variant is shown on FIG. 4 .
  • Incremental truncation of the DNA coding for the N-terminal domain of the AvrBs3-derived TALEN (pCLS7184, SEQ ID NO: 5), starting from the 5′ of the coding strand, allows the identification of minimal N-terminal domains that could still lead active TALEN.
  • the complete sequence of the N-terminal domain of AvrBs3 is amplified by PCR. Restriction site BsmBI, XmnI and SacI or AatII are introduced, in this specific order at the 3′ of the coding strand. After digestion with SacI or AatII, incremental truncation of the 5′ is performed using a 3′ ⁇ 5′ exonuclease III.
  • RVDs The AvrBs3-derived set of repeat domains (RVDs) (SEQ ID NO: 4) or any other previously synthesized RVD sequence is subcloned in the previously constructed plasmids using type IIs restriction enzymes BsmBI for the receiving plasmid. All created constructions are screened in our yeast SSA assay (see Example 1) for activity and specificity toward the AvrBs3 target or any other targets.
  • a particular truncated variant is judged useful if it provides, a minimal 5% retention in activity of the starting AvrBs3-derived TALEN (SEQ ID NO: 5) on its Avr15 target (SEQ ID NO: 6), more preferably a minimal 10% retention, more preferably 20%, more preferably 30%, more preferably 40%, more preferably 50%, again more preferably a retention in activity greater than 50%.
  • a particular variant is judged useful if it provides, on any targets having a C, G or A at position 0, a minimal 5% retention in activity of the starting AvrBs3-derived TALEN (SEQ ID NO: 5) on its Avr15 target (SEQ ID NO: 6), more preferably a minimal 10% retention, more preferably 20%, more preferably 30%, more preferably 40%, more preferably 50%, again more preferably a retention in activity greater than 50%.
  • a particular variant is judged useful if it provides, on any or all targets, a minimal 5% retention in activity of the starting AvrBs3-derived TALEN (SEQ ID NO: 5) on its Avr15 target (SEQ ID NO: 6), more preferably a minimal 10% retention, more preferably 20%, more preferably 30%, more preferably 40%, more preferably 50%, again more preferably a retention in activity greater than 50%.
  • DNA sequences corresponding to truncations (numbering based on SEQ ID NO:1) after positions E886 (C0), P897 (C11), G914 (C28), L926 (C40), D950 (C64), R1000 (C115) and D1059 (C172) were amplified by PCR, using the backbone plasmid pCLS7183 (referred as backbone wt, SEQ ID NO: 3) as template, to add a XmnI restriction site in 5′ and a BamHI restriction site in 3′ (Protein sequences of truncated C-terminal domains C11 to C172 are respectively given in SEQ ID NO: 295 to 300).
  • PCR constructs were subcloned in the TALEN yeast expression backbone (pCLS7183, SEQ ID NO: 3) to replace the sequence of the full C-terminal domain, leading to pCLS7820, pCLS7802, pCLS7806, pCLS7808, pCLS7810, pCLS7812, pCLS7816 (SEQ ID NO: 12 to 18). All DNA sequences were validated by sequencing.
  • the AvrBs3-derived set of repeat domains (SEQ ID NO: 4) was subcloned in pCLS7820, pCLS7802, pCLS7806, pCLS7808, pCLS7810, pCLS7812, pCLS7816 (SEQ ID NO: 12 to 18) and using type IIs restriction enzymes (BsmBI for the receiving plasmid and BbvI and SfaNI for the inserted RVD sequence) leading to pCLS7821, pCLS7803, pCLS7807, pCLS7809, pCLS7811, pCLS7813, pCLS7817 and subsequent TALENs (SEQ ID NO: 19 to 25).
  • type IIs restriction enzymes BsmBI for the receiving plasmid and BbvI and SfaNI for the inserted RVD sequence
  • TAL effectors possess a characteristic truncated RVD (the so-called half repeat) at the end (C-terminus) of the repeat region.
  • This half repeat is supposed to target specifically the last base of the target sequenced.
  • an Avrbs3-derived TALEN (pCLS7184, SEQ ID NO: 5) was screened, in our yeast assay, for activity on four identical targets except for the last base (A, T, G or C, in the n position, SEQ ID NO: 171 to 174, Table 4). No significant differences in activity were observed on the four targets as shown on FIG. 7 indicating the lack of specificity of the last half RVD in this TALEN context.
  • linker library is then subcloned in pCLS7183 (SEQ ID NO: 3) via the XmaI and BamHI restriction sites to replace the C-terminal domain of the AvrBs3-derived TALEN (pCLS7184, SEQ ID NO: 5).
  • RVDs The AvrBs3-derived set of repeat domains (RVDs) or any other RVD sequences having or lacking the terminal half RVD is cloned in this backbone library plasmid as described in Example 3A and resulting clones are screened in our yeast SSA assay (see Example 1).
  • a particular polypeptide linker domain is judged useful if it provides a minimal 5% retention in activity of the starting AvrBs3-derived TALEN (SEQ ID NO: 5) on its Avr15 target (SEQ ID NO: 6), more preferably a minimal 10% retention, more preferably 20%, more preferably 30%, more preferably 40%, more preferably 50%, again more preferably a retention in activity greater than 50%.
  • polypeptide linker The DNA (so called “polypeptide linker”) coding for, seven different polypeptides (SEQ ID NO: 479 to 485) were prepared by PCR using standard molecular biology procedures. These linkers contain, at the DNA level, a XmaI and a BamHI restriction sites in their 5′ and 3′ ends respectively. These seven linkers were then subcloned individually into a XmaI and BamHI pre-digested backbone pCLS9943 (SEQ ID NO: 486) via the XmaI and BamHI restriction sites to create a new C-terminal domain linker scaffold (pCLS12233 to 12238 and pCLS12270, SEQ ID NO: 487 to 493).
  • This backbone, pCLS9943 contains an additional N-terminal NLS sequence followed by an HA tag and a C11 truncated C-terminal domain compared to the original pCLS7183.
  • the RVD arrays coding for RAGT2.3 (SEQ ID NO: 494) were subcloned individually in the pCLS12233 to pCLS12237 and pCLS12270 using type IIs restriction enzymes BsmBI for the receiving plasmid and BbvI and SfaNI for the inserted RVD sequence, leading to the seven constructs, pCLS12945 to pCLS12951 (SEQ ID NO: 495 to 501).
  • Linkers #8, #27 and #35 (SEQ ID NO: 141, 160 and 168 respectively) showed significant activity toward AvrBs3 target in our yeast assay ( FIG. 26 ).
  • Activity is detected for targets with a spacer ranging from 15 to 30 bp for the TALEN having the full C-terminal domain, activity is detected for targets with a spacer ranging from 8 to 30 bp for the TALEN having the truncation C0 or C11, activity is detected for targets with a spacer ranging from 9 bp to 30 bp for the TALEN having the truncation C28, activity is detected for targets with a spacer ranging from 11 to 30 bp for the TALEN having the truncation C40, activity is detected for targets with a spacer ranging from 12 bp to 30 bp for the TALEN having the truncation C64 and C115 and activity is detected for targets with a spacer ranging from 13 to 30 bp for the TALEN having the truncation C172.
  • heterodimeric TALEN requires such large pseudo palindromic binding sites, such sequences are unlikely to be naturally present in genomic target. Relationship between spacer length and C-terminal truncations has been studied. In such a case of heterodimeric targets, C-terminal truncations on both TALENs do not require being identical and asymmetrical truncations have been tested.
  • Results show that different truncations can be associated in the same TALEN, modulating the activity over the spacer length space.
  • Such kinds of architectures (asymmetrical) direct more precisely the cleavage on the target spacer, either on the left part or on the right part or on center part.
  • results allow optimizing the control of double-stranded break localization of a TALEN within its nucleic acid target sequence by associating two optimum C-terminal truncations of the core scaffold structure to an optimum spacer length wherein said association allows placing the cleavage site at a more desired location within said spacer.
  • Said association between two optimum C-terminal truncations of the core scaffold structure and an optimum spacer length allows placing the cleavage site at a more desired location that is in the center of said spacer or not i.e either in the left part (5′ located regarding the center of the spacer) of said spacer, either in the right part (3′ located regarding the center of the spacer) of said spacer.
  • TALEN couples are combinations of AvrBs3-derived (SEQ ID NO: 5) and RAGT2R (SEQ ID NO: 127) parent TALEN containing respectively C0, C11, C40, C117 and Cter WT C-terminal domains for AvrBs3-derived constructs and C0, C11, C40 and Cter WT C-terminal domains for RAGT2R constructs.
  • the DNA encoding a nuclear localization sequence NLS, either a HA tag or a S tag, the N-terminal domain, the C11 truncated C-terminal domain and a nuclease catalytic head was synthesized (TopGene Technologies) and subcloned into the pCLS1853 (SEQ ID NO: 7) mammalian expression plasmid, using AscI and XhoI restriction enzymes, leading to the backbone plasmids pCLS8425 (HA tag) and pCLS8429 (S tag) (SEQ ID NO: 175 and 176).
  • the C-terminal and the N-terminal domains are separated by two BsmBI restriction sites.
  • the set of repeat domains (RVDs) binding the left part of the DNA target sequence DMDT2.1, ILRGT2.1, and HBBT1.1 (SEQ ID NO: 189, SEQ ID NO: 190 and SEQ ID NO: 192) were subcloned in the pCLS8425 using type IIs restriction enzymes BsmBI for the receiving plasmid and BbvI and SfaNI for the inserted RVD sequence, leading to pCLS8453 (DMDT2.1 left; SEQ ID NO: 180), pCLS8445 (ILRGT2.1 left; SEQ ID NO: 181), and pCLS8461 (HBBT1.1 left; SEQ ID NO: 182) and subsequent left monomer TALENs (SEQ ID NO: 180 to 182).
  • RVDs The set of repeat domains (RVDs) binding the right part of the DNA target sequence DMDT2.1, ILRGT2.1, and HBBT1.1 (SEQ ID NO: 183 to 185) were subcloned in the pCLS8429 using type IIs restriction enzymes BsmBI for the receiving plasmids and BbvI and SfaNI for the inserted RVD sequences, leading to pCLS8457 (DMDT2.1 right; SEQ ID NO: 186), pCLS8449 (ILRGT2.1 left; SEQ ID NO: 187), and pCLS8465 (HBBT1.1 left; SEQ ID NO: 188) and subsequent TALEN (SEQ ID NO: 186 to 188).
  • the plasmids pair pCLS8453 and pCLS8457 were co-transformed, with its DMDT2.1 target into CHO-K1 cells in order to express the heterodimeric TALEN.
  • Activity of the TALEN was screened in our mammalian SSA assay (see example 1).
  • the plasmids pair pCLS8445 and pCLS8549 were co-transformed, with its ILRGT2.1 target, into CHO-K1 cells in order to express the heterodimeric TALEN.
  • Activity of the TALEN was screened in our mammalian SSA assay (see example 1).
  • the plasmids pair pCLS8461 and pCLS8465 were co-transformed, with its HBBT1.1 target, into CHO-K1 cells in order to express the heterodimeric TALEN.
  • Activity of the TALEN was screened in our mammalian SSA assay (see example 1).
  • TALENs activity levels in this assay indicate that they cleave their target sequence in the CHO mammalian cells ( FIG. 8 ).
  • the catalytic domain of I-TevI (SEQ ID NO: 349), a member of the GIY-YIG endonuclease family, was fused to a TAL backbone, composed of a N-terminal domain, a central core composed of RVDs and a C-terminal domain, to create a new class of TALEN (TALE::TevI). To distinguish the orientation (N-terminal vs.
  • construct names are written as either CD::TALE-RVD (catalytic domain is fused N-terminal to the TALE domain) or TALE-RVD::CD (catalytic domain is fused C-terminal to the TALE domain), where “-RVD” optionally designates the sequence recognized by the TALE domain and “CD” is the catalytic domain type.
  • TALE-AvrBs3::TevI novel TALE::TevI constructions that target AvrBs3 sequence for example, thus named TALE-AvrBs3::TevI.
  • a core TALE scaffold ST2 (SEQ ID NO: 464) onto which (a) different sets of RVD domains could be inserted to change DNA binding specificity, and; (b) a selection of I-TevI-derived catalytic domains could be attached, N- or C-terminal, to effect DNA cleavage (or nicking) was generated.
  • the sT2 truncated scaffold was generated by the PCR from a full-length core TALEN scaffold template (pCLS7183, SEQ ID NO: 3) using primers CMP_G061 (SEQ ID NO: 440) and CMP_G065 (SEQ ID NO: 441) and was cloned into vector pCLS7865 (SEQ ID NO: 442) to generate pCLS7865-cTAL11_CFS1 (pCLS9009, SEQ ID NO: 443) where CFS1 designates the amino acid sequence -GSSG- (with underlying restriction site BamHI and Kpn21 in the coding DNA to facilitate cloning).
  • the DNA sequence coding for the RVDs to target the AvrBs3 site was subcloned into both plasmids pCLS9010 (SEQ ID NO: 451) and pCLS9011 (SEQ ID NO: 452) using Type IIS restriction enzymes BsmBI for the receiving plasmid and BbvI and SfaNI for the inserted RVD sequence to create the subsequent TALE-AvrBs3::TevI constructs cT11Avr_TevD01 (pCLS9012, SEQ ID NO: 453) and cT11Avr_TevD02 (pCLS9013, SEQ ID NO: 454), respectively.
  • TALE-AvrBs3::TevI constructs were sequenced and the insert transferred to additional vectors as needed (see below and Example 7b).
  • TALE-AvrBs3::TevI yeast expression plasmids pCLS8523 (SEQ ID NO: 455) and pCLS8524 (SEQ ID NO: 456), were prepared by yeast in vivo cloning using plasmids pCLS9012 and pCLS9013 (SEQ ID NO: 453 and 454), respectively.
  • plasmid pCLS9012; SEQ ID NO: 453 or pCLS9013, SEQ ID NO: 454 linearized by digestion with BssHII and 1 ng of the pCLS0542 (SEQ ID NO: 2) plasmid DNA linearized by digestion with NcoI and EagI were used to transform the yeast S. cerevisiae strain FYC2-6A (MAT ⁇ , trp1 ⁇ 63, leu2 ⁇ 1, his3 ⁇ 200) using a high efficiency LiAc transformation protocol (Arnould et al. 2007).
  • yeast target reporter plasmids containing the TALEN DNA target sequences were constructed as previously described (International PCT Applications WO 2004/067736 and in Epinat, Arnould et al. 2003; Chames, Epinat et al. 2005; Arnould, Chames et al. 2006; Smith, Grizot et al. 2006).
  • TALE-AvrBs3::TevI constructs were tested in a yeast SSA assay as previously described (International PCT Applications WO 2004/067736 and in Epinat, Arnould et al. 2003; Chames, Epinat et al. 2005; Arnould, Chames et al. 2006; Smith, Grizot et al. 2006) on pseudo palindromic targets in order to compare activity with a standard TALE-AvrBs3::FokI TALEN, which requires two binding sites for activity.
  • AvrBS3 targets contain two identical recognition sequences juxtaposed with the 3′ ends proximal and separated by “spacer” DNA ranging from 5 to 40 bps (SEQ ID NO: 220 to 255, Table 3).
  • TALE-AvrBs3::TevI activity levels on their respective targets in yeast cells are shown on FIG. 12 .
  • Data summarized in FIG. 12 show that TALE-AvrBs3::TevI is active against several targets in Yeast.
  • DNA encoding the TALE-AvrBs3::TevI construct from either pCLS9012 (SEQ ID NO: 453) or pCLS9013 (SEQ ID NO: 454) was subcloned into the pCLS1853 (SEQ ID NO: 7) mammalian expression plasmid using AscI and XhoI restriction enzymes for the receiving plasmid and BssHII and XhoI restriction enzymes for the TALE-AvrBs3::TevI insert, leading to the mammalian expression plasmids pCLS8993 and pCLS8994 (SEQ ID NO: 457 and 458), respectively.
  • All mammalian target reporter plasmids containing the TALEN DNA target sequences were constructed using the standard Gateway protocol (INVITROGEN) into a CHO reporter vector (Arnould, Chames et al. 2006, Grizot, Epinat et al. 2010).
  • the TALE-AvrBs3::TevI constructs were tested in an extrachromosomal assay in mammalian cells (CHO K1) on pseudo palindromic targets in order to compare activity with a standard TALE-AvrBs3::FokI TALEN, which requires two binding sites for activity.
  • AvrBS3 targets contain two identical recognition sequences juxtaposed with the 3′ ends proximal and separated by “spacer” DNA ranging from 5 to 40 bps (SEQ ID NO: 220 to 255, Table 3).
  • CHO K1 cells were transfected in a 96-well plate format with 75 ng of target vector and an increasing quantity of each variant DNA from 0.7 to 25 ng, in the presence of PolyFect reagent (1 ⁇ L per well). The total amount of transfected DNA was completed to 125 ng (target DNA, variant DNA, carrier DNA) using an empty vector. Seventy-two 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., optical density was measured at 420 nm. The entire process is performed on an automated Velocity11 BioCel platform (Grizot, Epinat et al. 2009).
  • TALE-AvrBs3::TevI constructs (12.5 ng DNA transfected) on the Avr15 target (SEQ ID NO: 230) are shown on FIG. 13 .
  • TALE-AvrBs3::TevI appears to be efficient to cleave the target sequence.
  • Variants differing by truncations of the C-terminal domain of the AvrBs3-derived TALEN are chosen as starting scaffolds.
  • a subset of these variants includes truncation after positions E886 (C0), P897 (C11), G914 (C28), L926 (C40), D950 (C64), R1000 (C115), D1059 (C172) (the protein domains of truncated C-terminal domains C11 to C172 are respectively given in SEQ ID NO: 295 to 300) and P1117 [also referred as Cter wt or WT Cter (SEQ ID NO: 400) lacking the activation domain of the C-terminal domain of natural AvrBs3 (SEQ ID NO: 1)].
  • the plasmids coding for the variant scaffolds containing the AvrBs3-derived N-terminal domain, the AvrBs3-derived set of repeat domains and the truncated AvrBs3-derived C-terminal domain [pCLS7821, pCLS7803, pCLS7807, pCLS7809, pCLS7811, pCLS7813, pCLS7817 (SEQ ID NO: 19 to 25) which are based on the pCLS7184 (SEQ ID NO: 5)] allow cloning of any catalytic domain in fusion to the C-terminal domain, using the restriction sites BamHI and EagI.
  • Variants of the catalytic domain of I-TevI are designed from the N-terminal region of I-TevI.
  • a subset of these variants includes truncations of the catalytic domain, the deletion-intolerant region of its linker, the deletion-tolerant region of its linker and its zinc finger (SEQ ID: 459 to 462) (Liu, dansereau et al. 2008).
  • the DNA corresponding to these variants of I-TevI is amplified by the PCR to introduce, at the DNA level, a BamHI (at the 5′ of the coding strand) and a EagI (at the 3′ of the coding strand) restriction site and, at the protein level, a linker (for example -SGGSGS- stretch, SEQ ID NO: 463) between the C terminal domain of the TAL and the variant of the catalytic domain of I-TevI.
  • the final TALE::TevI constructs are generated by insertion of the variant of I-TevI catalytic domains into the scaffold variants using BamHI and EagI and standard molecular biology procedures.
  • yeast target reporter plasmids containing the TALEN DNA target sequences were constructed as previously described (International PCT Applications WO 2004/067736 and in Epinat, Arnould et al. 2003; Chames, Epinat et al. 2005; Arnould, Chames et al. 2006; Smith, Grizot et al. 2006).
  • the TALE-AvrBs3::TevI constructs were tested in a yeast SSA assay as previously described (International PCT Applications WO 2004/067736 and in Epinat, Arnould et al. 2003; Chames, Epinat et al. 2005; Arnould, Chames et al.
  • AvrBS3 targets contain two identical recognition sequences juxtaposed with the 3′ ends proximal and separated by “spacer” DNA ranging from 5 to 40 bps (SEQ ID NO: 220 to 255, Table 3).
  • NucA (SEQ ID NO: 355), a nonspecific endonuclease from Anabaena sp., was fused to a TALE-derived scaffold (composed of a N-terminal domain, a central core composed of RVDs and a C-terminal domain) to create a new class of cTALEN (TALE::NucA). To distinguish the orientation (N-terminal vs.
  • construct names are written as either CD::TALE-RVD (catalytic domain is fused N-terminal to the TALE domain) or TALE-RVD::CD (catalytic domain is fused C-terminal to the TALE domain), where “-RVD” optionally designates the sequence recognized by the TALE domain and “CD” is the catalytic domain type.
  • TALE-AvrBs3::NucA novel TALE::NucA constructions that target for example the AvrBs3 sequence, and are thus named TALE-AvrBs3::NucA.
  • the wild-type NucA endonuclease can be inhibited by complex formation with the NuiA protein (SEQ ID NO: 473).
  • the NuiA protein can function as a protein domain to modulate the activity of NucA or TALE::NucA constructs.
  • a core TALE scaffold, sT2 (SEQ ID NO: 464) was selected onto which (a) different sets of RVD domains could be inserted to change DNA binding specificity, and; (b) a selection of NucA-derived catalytic domains could be attached, N- or C-terminal, to effect DNA cleavage (or nicking).
  • the sT2 truncated scaffold was generated by the PCR from a full-length core TALEN scaffold template (pCLS7183, SEQ ID NO: 3) using primers CMP_G061 (SEQ ID NO: 440) and CMP_G065 (SEQ ID NO: 441) and was cloned into vector pCLS7865 (SEQ ID NO: 442) to generate pCLS7865-cTAL11_CFS1 (pCLS9009, SEQ ID NO: 443), where CFS1 designates the amino acid sequence -GSSG- (with underlying restriction sites BamHI and Kpn2I in the coding DNA to facilitate cloning).
  • the NucA (SEQ ID NO: 355) catalytic domain corresponding to amino acid residues 25 to 274, was subcloned into the pCLS9009 backbone (SEQ ID NO: 443) by restriction and ligation using BamHI and EagI restriction sites, yielding pCLS7865-cT11_NucA (pCLS9937, SEQ ID NO: 465).
  • the fusion contains the dipeptide -GS- linking the TALE-derived DNA binding domain and NucA-derived catalytic domain.
  • the cloning step also brings at the amino acid level an AAD sequence at the Cter of the NucA catalytic domain.
  • the DNA sequence coding for the RVDs to target the AvrBs3 site was subcloned into plasmid pCLS9937 (SEQ ID NO: 465) using Type IIS restriction enzymes BsmBI for the receiving plasmid and BbvI and SfaNI for the inserted RVD sequence to create the subsequent TALE-AvrBs3::NucA construct cT11Avr_NucA (pCLS9938, SEQ ID NO: 466).
  • the TALE-AvrBs3::NucA construct was sequenced and the insert transferred to additional vectors as needed (see below).
  • the final TALE-AvrBs3::NucA yeast expression plasmid, pCLS9924 (SEQ ID NO: 467), was prepared by yeast in vivo cloning using plasmid pCLS9938 (SEQ ID NO: 466).
  • plasmid pCLS9938 approximately 40 ng of plasmid (pCLS9938) linearized by digestion with BssHII and 1 ng of the pCLS0542 (SEQ ID NO: 2) plasmid DNA linearized by digestion with NcoI and EagI were used to transform the yeast S. cerevisiae strain FYC2-6A (MAT ⁇ , trp1 ⁇ 63, leu2 ⁇ 1, his3 ⁇ 200) using a high efficiency LiAc transformation protocol (Arnould et al. 2007).
  • yeast target reporter plasmids containing the TALEN DNA target sequences were constructed as previously described (International PCT Applications WO 2004/067736 and in Epinat, Arnould et al. 2003; Chames, Epinat et al. 2005; Arnould, Chames et al. 2006; Smith, Grizot et al. 2006).
  • the TALE-AvrBs3::NucA construct was tested in a yeast SSA assay as previously described (International PCT Applications WO 2004/067736 and in Epinat, Arnould et al. 2003; Chames, Epinat et al. 2005; Arnould, Chames et al. 2006; Smith, Grizot et al. 2006) on pseudo palindromic targets in order to compare activity with a standard TALE-AvrBs3::FokI TALEN, which requires two binding sites for activity.
  • AvrBS3 targets contain two identical recognition sequences juxtaposed with the 3′ ends proximal and separated by “spacer” DNA ranging from 5 to 40 bps (SEQ ID NO: 220 to 255, Table 8).
  • constructs were tested on a target having only a single AvrBs3 recognition site (SEQ ID NO: 468; Table 8).
  • Variants differing by truncations of the C-terminal domain of the AvrBs3-derived TALEN are chosen as starting scaffolds.
  • a subset of these variants includes truncation after positions E886 (C0), P897 (C11), G914 (C28), L926 (C40), D950 (C64), R1000 (C115), D1059 (C172) (the protein domains of truncated C-terminal domains C11 to C172 are respectively given in SEQ ID NO: 295 to 300) and P1117 [also referred as Cter wt or WT Cter (SEQ ID NO: 400) lacking the activation domain of the C-terminal domain of natural AvrBs3 (SEQ ID NO: 1)].
  • the plasmids coding for the variant scaffolds containing the AvrBs3-derived N-terminal domain, the AvrBs3-derived set of repeat domains and the truncated AvrBs3-derived C-terminal domain [pCLS7821, pCLS7803, pCLS7807, pCLS7809, pCLS7811, pCLS7813, pCLS7817 (SEQ ID NO: 19 to 25) which are based on the pCLS7184 (SEQ ID NO: 5)] allow cloning of any catalytic domain in fusion to the C-terminal domain, using the restriction sites BamHI and EagI.
  • the DNA corresponding to amino acid residues 25 to 274 of NucA is amplified by the PCR to introduce, at the DNA level, a BamHI (at the 5′ of the coding strand) and a EagI (at the 3′ of the coding strand) restriction site and, at the protein level, a linker (for example -SGGSGS- stretch, SEQ ID NO: 463) between the C terminal domain of the TALE and the NucA catalytic domain.
  • the final TALE::NucA constructs are generated by insertion of the NucA catalytic domain into the scaffold variants using BamHI and EagI and standard molecular biology procedures.
  • the cloning step also brings at the amino acid level an AAD sequence at the Cter of the NucA catalytic domain.
  • yeast target reporter plasmids containing the TALEN DNA target sequences were constructed as previously described (International PCT Applications WO 2004/067736 and in Epinat, Arnould et al. 2003; Chames, Epinat et al. 2005; Arnould, Chames et al. 2006; Smith, Grizot et al. 2006).
  • TALE-AvrBs3::NucA constructs were tested in a yeast SSA assay as previously described (International PCT Applications WO 2004/067736 and in Epinat, Arnould et al. 2003; Chames, Epinat et al. 2005; Arnould, Chames et al. 2006; Smith, Grizot et al. 2006) on pseudo palindromic targets in order to compare activity with a standard TALE-AvrBs3::FokI TALEN, which requires two binding sites for activity.
  • AvrBs3 targets contain two identical recognition sequences juxtaposed with the 3′ ends proximal and separated by “spacer” DNA ranging from 5 to 40 bps (SEQ ID NO: 220 to 255, Table 8).
  • TALE-AvrBs3::NucA constructs were tested on a target having only a single AvrBs3 recognition site (SEQ ID NO: 468).
  • Data summarized in FIG. 14 show that TALE-AvrBs3::NucA constructs are active on all targets having at least one AvrBs3 recognition site, according to the chimeric protein of the present invention.
  • the catalytic domain of ColE7 (SEQ ID NO: 478 of ColE7 protein SEQ ID NO: 340), a nonspecific endonuclease from E. coli , was fused to a TALE-derived scaffold (composed of a N-terminal domain, a central core composed of RVDs and a C-terminal domain) to create a new class of cTALEN (TALE::ColE7).
  • TALE derived scaffold
  • construct names are written as either CD::TALE-RVD (catalytic domain is fused N-terminal to the TALE domain) or TALE-RVD::CD (catalytic domain is fused C-terminal to the TALE domain), where “-RVD” optionally designates the sequence recognized by the TALE domain and “CD” is the catalytic domain type.
  • TALE-AvrBs3::ColE7 novel TALE::ColE7 constructions that target for example the AvrBs3 sequence, and are thus named TALE-AvrBs3::ColE7.
  • the wild-type ColE7 endonuclease can be inhibited by complex formation with the Im7 immunity protein (SEQ ID NO: 474).
  • the Im7 protein can function as auxiliary protein domain to modulate the nuclease activity of ColE7 or TALE::ColE7 constructs.
  • a core TALE scaffold, sT2 (SEQ ID NO: 464) was selected onto which (a) different sets of RVD domains could be inserted to change DNA binding specificity, and; (b) a selection of ColE7-derived catalytic domains could be attached, N- or C-terminal, to effect DNA cleavage (or nicking).
  • the sT2 truncated scaffold was generated by the PCR from a full-length core TALEN scaffold template (pCLS7183, SEQ ID NO: 3) using primers CMP_G061 (SEQ ID NO: 440) and CMP_G065 (SEQ ID NO: 441) and was cloned into vector pCLS7865 (SEQ ID NO: 442) to generate pCLS7865-cTAL11_CFS1 (pCLS9009, SEQ ID NO: 443), where CFS1 designates the amino acid sequence -GSSG- (with underlying restriction sites BamHI and Kpn2I in the coding DNA to facilitate cloning).
  • the ColE7 (SEQ ID NO: 478) catalytic domain was subcloned into the pCLS9009 backbone by restriction and ligation using Kpn2I and EagI restriction sites, yielding pCLS7865-cT11_ColE7 (pCLS9939, SEQ ID NO: 475).
  • the fusion contains the dipeptide -GSSG- linking the TALE-derived DNA binding domain and ColE7-derived catalytic domain.
  • the DNA sequence coding for the RVDs to target the AvrBs3 site was subcloned into plasmid pCLS9939 (SEQ ID NO: 475) using Type IIS restriction enzymes BsmBI for the receiving plasmid and BbvI and SfaNI for the inserted RVD sequence to create the subsequent TALE-AvrBs3::ColE7 construct cT11Avr_ColE7 (pCLS9940, SEQ ID NO: 476).
  • the TALE-AvrBs3::ColE7 construct was sequenced and the insert transferred to additional vectors as needed (see below).
  • the final TALE-AvrBs3::ColE7 yeast expression plasmid, pCLS8589 was prepared by yeast in vivo cloning using plasmid pCLS9940 (SEQ ID NO: 476).
  • plasmid pCLS9940 approximately 40 ng of plasmid linearized by digestion with BssHII and 1 ng of the pCLS0542 (SEQ ID NO: 2) plasmid DNA linearized by digestion with NcoI and EagI were used to transform the yeast S. cerevisiae strain FYC2-6A (MAT ⁇ , trp1 ⁇ 63, leu2 ⁇ 1, his3 ⁇ 200) using a high efficiency LiAc transformation protocol (Arnould et al. 2007).
  • yeast target reporter plasmids containing the TALEN DNA target sequences were constructed as previously described (International PCT Applications WO 2004/067736 and in Epinat, Arnould et al. 2003; Chames, Epinat et al. 2005; Arnould, Chames et al. 2006; Smith, Grizot et al. 2006).
  • the TALE-AvrBs3::ColE7 construct was tested in a yeast SSA assay as previously described (International PCT Applications WO 2004/067736 and in Epinat, Arnould et al. 2003; Chames, Epinat et al. 2005; Arnould, Chames et al. 2006; Smith, Grizot et al. 2006) on pseudo palindromic targets in order to compare activity with a standard TALE-AvrBs3::FokI TALEN, which requires two binding sites for activity.
  • AvrBS3 targets contain two identical recognition sequences juxtaposed with the 3′ ends proximal and separated by “spacer” DNA ranging from 5 to 40 bps (SEQ ID NO: 220 to 255, Table 8).
  • constructs were tested on a target having only a single AvrBs3 recognition site (SEQ ID NO: 468, Table 8).
  • TALE-AvrBs3::ColE7 activity levels on the respective targets in yeast cells are shown on FIG. 15 .
  • Variants differing by truncations of the C-terminal domain of the AvrBs3-derived TALEN are chosen as starting scaffolds.
  • a subset of these variants includes truncation after positions E886 (C0), P897 (C11), G914 (C28), L926 (C40), D950 (C64), R1000 (C115), D1059 (C172) (the protein domains of truncated C-terminal domains C11 to C172 are respectively given in SEQ ID NO: 295 to 300) and P1117 [also referred as Cter wt or WT Cter (SEQ ID NO: 400) lacking the activation domain of the C-terminal domain of natural AvrBs3 (SEQ ID NO: 1)].
  • the plasmids coding for the variant scaffolds containing the AvrBs3-derived N-terminal domain, the AvrBs3-derived set of repeat domains and the truncated AvrBs3-derived C-terminal domain [pCLS7821, pCLS7803, pCLS7807, pCLS7809, pCLS7811, pCLS7813, pCLS7817 (SEQ ID NO: 19 to 25) which are based on the pCLS7184 (SEQ ID NO: 5)] allow cloning of any catalytic domain in fusion to the C-terminal domain, using the restriction sites BamHI and EagI.
  • the DNA corresponding to the catalytic domain of ColE7 is amplified by the PCR to introduce, at the DNA level, a BamHI (at the 5′ of the coding strand) and a EagI (at the 3′ of the coding strand) restriction site and, at the protein level, a linker (for example -SGGSGS- stretch, SEQ ID NO: 463) between the C terminal domain of the TALE and the ColE7 catalytic domain.
  • a linker for example -SGGSGS- stretch, SEQ ID NO: 463
  • variants of the ColE7 endonuclease domain that modulate catalytic activity can be generated having changes (individually or combined) at the following positions: D493, R496, K497, H545, N560 and H573 [positions refer to the amino acid sequence of the entire ColE7 protein (SEQ ID NO: 340)].
  • the final TALE::ColE7 constructs are generated by insertion of the ColE7 catalytic domain into the scaffold variants using BamHI and EagI and standard molecular biology procedures.
  • yeast target reporter plasmids containing the TALEN DNA target sequences were constructed as previously described (International PCT Applications WO 2004/067736 and in Epinat, Arnould et al. 2003; Chames, Epinat et al. 2005; Arnould, Chames et al. 2006; Smith, Grizot et al. 2006).
  • the TALE-AvrBs3::ColE7 constructs are tested in a yeast SSA assay as previously described (International PCT Applications WO 2004/067736 and in Epinat, Arnould et al. 2003; Chames, Epinat et al. 2005; Arnould, Chames et al. 2006; Smith, Grizot et al. 2006) on pseudo palindromic targets in order to compare activity with a standard TALE-AvrBs3::FokI TALEN, which requires two binding sites for activity.
  • AvrBS3 targets contain two identical recognition sequences juxtaposed with the 3′ ends proximal and separated by “spacer” DNA ranging from 5 to 40 bps (SEQ ID NO: 220 to 255, Table 8).
  • constructs were tested on a target having only a single AvrBs3 recognition site (SEQ ID NO: 468, Table 8).
  • Variants differing by truncations of the C-terminal domain of the AvrBs3-derived TALEN are chosen as starting scaffolds.
  • a subset of these variants includes truncation after positions E886 (C0), P897 (C11), G914 (C28), L926 (C40) and D950 (C64) (the protein domains of truncated C-terminal domains C11 to C64 are respectively given in SEQ ID NO: 295 to 298).
  • the plasmids coding for the variant scaffolds containing the AvrBs3-derived N-terminal domain, the AvrBs3-derived set of repeat domains and the truncated AvrBs3-derived C-terminal domain [pCLS7821, pCLS7803, pCLS7807, pCLS7809, pCLS7811, (SEQ ID NO: 19 to 23) which are based on the pCLS7184 (SEQ ID NO: 5)] allow cloning of any catalytic domain in fusion to the C-terminal domain, using the restriction sites BamHI and EagI.
  • the DNA corresponding to amino acid residues 2-149 of EndoT7 (SEQ ID NO: 363) is amplified by the PCR to introduce, at the DNA level, a BamHI (at the 5′ of the coding strand) and a EagI (at the 3′ of the coding strand) restriction site and, at the protein level, a linker (for example -SGGSGS- stretch, SEQ ID NO: 463) between the C terminal domain of the TALE and the EndoT7 catalytic domain.
  • the final TALE::EndoT7 constructs are generated by insertion of the EndoT7 catalytic domain into the scaffold variants using BamHI and EagI and standard molecular biology procedures.
  • yeast target reporter plasmids containing the TALEN DNA target sequences were constructed as previously described (International PCT Applications WO 2004/067736 and in Epinat, Arnould et al. 2003; Chames, Epinat et al. 2005; Arnould, Chames et al. 2006; Smith, Grizot et al. 2006).
  • the TALE-AvrBs3::EndoT7 constructs were tested in a yeast SSA assay as previously described (International PCT Applications WO 2004/067736 and in Epinat, Arnould et al. 2003; Chames, Epinat et al. 2005; Arnould, Chames et al. 2006; Smith, Grizot et al. 2006) on pseudo palindromic targets in order to compare activity with a standard TALE-AvrBs3::FokI TALEN, which requires two binding sites for activity.
  • AvrBs3 targets contain two identical recognition sequences juxtaposed with the 3′ ends proximal and separated by “spacer” DNA ranging from 5 to 40 bps (SEQ ID NO: 220 to 255, Table 8).
  • Data summarized in FIG. 20 show that TALE-AvrBs3::EndoT7 constructs are active on targets having two AvrBs3 recognition site, according to the chimeric protein of the present invention.
  • linker library is then subcloned in pCLS7183 (SEQ ID NO: 3) via the XmaI and BamHI restriction sites to replace the C-terminal domain of the AvrBs3-derived TALEN (pCLS7184, SEQ ID NO: 5).
  • the AvrBs3-derived set of repeat domains (RVDs) or any other RVD sequences having or lacking the terminal half RVD is cloned in this backbone library plasmid as described in Example 3A.
  • DNA from the library is obtained, after scrapping of the colonies from the Petri dishes, using standard miniprep techniques.
  • the FokI catalytic head is removed using BamHI and EagI restriction enzymes, the remaining backbone being purified using standard gel extraction techniques.
  • DNA coding for 3 catalytic heads presented in table 2 (SEQ ID NO: 340, 349 and 363) were amplified by the PCR to introduce, at the DNA level, a BamHI (at the 5′ of the coding strand) and a EagI (at the 3′ of the coding strand) restriction site and, at the protein level, a linker (for example -SGGSGS -stretch, SEQ ID NO: 463) between the C-terminal domain library and the catalytic head. After BamHI and EagI digestion and purification, the DNA coding for the different catalytic heads were individually subcloned into the library scaffold previously prepared.
  • DNA from the final library is obtained, after scrapping of the colonies from Petri dishes, using standard miniprep techniques and the resulting libraries are screened in our yeast SSA assay as previously described (International PCT Applications WO 2004/067736 and in Epinat, Arnould et al. 2003; Chames, Epinat et al. 2005; Arnould, Chames et al. 2006; Smith, Grizot et al. 2006) on pseudo palindromic targets in order to compare activity with a standard TALE-AvrBs3::FokI TALEN, which requires two binding sites for activity.
  • AvrBs3 targets contain two identical recognition sequences juxtaposed with the 3′ ends proximal and separated by “spacer” DNA containing 15, 18, 21 and 24 bps (SEQ ID NO: 230, 233, 236 and 239, Table 8).
  • constructs were tested on a target having only a single AvrBs3 recognition site (SEQ ID NO: 468). Data summarized in FIG.
  • linkers of SEQ ID NO: 147, 150, 156 and 162 for colE7 linkers of SEQ ID NO: 134, 153, 154, 157, 162 and 166 for I-TevI and linkers of SEQ ID NO: 134, 152, 153, 159 and 166 for EndoT7 being active on targets having one or two AvrBs3 recognition sites ( FIG. 21 ) according to the chimeric protein of the present invention.
  • the 52 different TAL repeats arrays containing from 9.5 to 15.5 Tal repeats were synthesized using a solid support method consisting in a sequential assembly of TAL repeats through consecutive restriction/ligation/washing steps as shown in FIG. 23 .
  • the first TAL repeat (SEQ ID NO: 584 encoding SEQ ID NO: 585) was immobilized on a solid support through biotin/streptavidin interaction, digested by SfaNI type IIS restriction endonuclease and then ligated to a second TAL repeat (SEQ ID NO: 586 encoding SEQ ID NO: 587) harboring SfaNI compatible overhangs at its 5′ end ( FIG.
  • the resulting TAL repeats array (i.e containing TAL repeats 1 and 2) was then used as template for subsequent additions of the appropriate TAL repeats (SEQ ID NO: 588-591, encoding SEQ ID NO: 592-595 for HD, NI, respectively targeting nucleotides C, A and NN, NG respectively targeting nucleotides G and T) to generate the complete TAL repeats arrays RAGT2.3 ( FIG. 23C ).
  • the complete TAL repeats array was finally digested by SfaNI to generate SfaNI overhangs at its 3′ end ( FIG. 23D ) and then striped of the solid support using BbvI type IIS restriction endonuclease ( FIG. 23E ).
  • the digested TAL repeats array was recovered and subcloned into yeast or mammalian expression plasmids harboring the Nterminal domain of AvrBs3 TAL effector and the forty first amino acids of its Cterminal domain fused to FokI type IIS restriction endonuclease (pCLS 7808, i.e. SEQ ID NO: 596 encoding SEQ ID NO: 597, FIG. 23F ).
  • pCLS7808 was derived from pCLS0542 (SEQ ID NO: 2) using NcoI and XhoI restriction sites.
  • TAL effector DNA binding domains are known to be highly specific with respect to their cognate target (refs: 3, 25). This has been demonstrated for different TAL DNA binding domains by independent studies. However, the influence of TAL repeat number on such specificity is unclear. To address this question in a systematic manner, RAGT2.3 and RAGT2.4 TALENs were chosen as models, then the number of their TAL repeats (15.5, 13.5, 11.5 9.5 TAL repeats were iteratively reduced, according to the assembly method described in example 12 and their TALEN activity were characterized toward their respective homodimeric target degenerated in positions N and N-1 ( FIG. 25 , SEQ ID NO: 602-615). For the sake of clarity, FIG.
  • FIG. 25A displays the different components of a TALEN including the N and C-terminal domains, the TAL DNA binding domain bearing the terminal half repeat and FokI catalytic domain.
  • the FIG. 25B displays the organization of the homodimeric RAGT2.4 TALEN targets (SEQ ID NO: 601) used for our experiments including the location of thymine T0 and the positions N and N-1 degenerated in this study.
  • the FIG. 25C displays two examples of TALEN bearing 15.5 or 11.5 Tal repeats along with their respective DNA targets (top and bottom respectively, SEQ ID NO: 601).
  • the catalytic domain of FokI (SEQ ID NO: 600), starting at residue P381, was fused to a TALE-derived scaffold (composed of a N-terminal domain, a central core composed of RVDs and a C-terminal domain) to create a half-TALEN.
  • a TALE-derived scaffold composed of a N-terminal domain, a central core composed of RVDs and a C-terminal domain
  • construct names are written as either CD::TALE-RVD (catalytic domain is fused N-terminal to the TALE domain) or TALE-RVD::CD (catalytic domain is fused C-terminal to the TALE domain), where “-RVD” optionally designates the sequence recognized by the TALE domain and “CD” is the catalytic domain type.
  • FokI::TALE constructions that either work together with other FokI::TALE constructions in a conventional “head-to-head” configuration or can be paired with TALE::FokI constructions in a novel “tail-to-head” configuration, allowing for targeting a single DNA strand (when considering the requisite T 0 as 5′ for target readout).
  • a core TALE scaffold, sT2 (SEQ ID NO: 464) was selected onto which (a) different sets of RVD domains could be inserted to change DNA binding specificity, and; (b) a selection of FokI-derived catalytic domains could be attached, N- or C-terminal, to effect DNA cleavage (or nicking).
  • pCLS7865-cTAL11_NFS1 (pCLS9008, SEQ ID NO: 624), where NFS1 designates the amino acid sequence -GSSG- (with underlying restriction sites BamHI and Kpn2I in the coding DNA to facilitate cloning)
  • pCLS7865-cTAL11_CFS1 (pCLS9009, SEQ ID NO: 660), where CFS1 designates the amino acid sequence -GSSG- (with underlying restriction sites BamHI and Kpn2I in the coding DNA to facilitate cloning).
  • the catalytic domain of FokI (SEQ ID NO: 600) was subcloned by restriction and ligation into pCLS9008 (SEQ ID NO: 624) using NcoI and BamHI restriction sites, yielding the construct FokI_cT11 (SEQ ID NO: 625).
  • the fusion contains the peptide -GSSG- linking the TALE-derived DNA binding domain and FokI derived catalytic domain.
  • the DNA sequence coding for the RVDs to target the AvrBS3 site was subcloned into the FokI_cT11 (SEQ ID NO: 625) scaffold using Type IIS restriction enzymes BsmBI for the receiving plasmid and BbvI and SfaNI for the inserted RVD sequence to create the subsequent FokI::TALE-AvrBs3 construct FokI_cT11Avr (SEQ ID NO: 627). This construct was sequenced and the insert transferred to additional vectors as needed.
  • the final FokI::TALE-AvrBS3 yeast expression plasmid, pCLS8674 (SEQ ID NO: 628), was prepared by restriction and ligation of the FokI_cT11Avr (SEQ ID NO: 627) insert into pCLS0542 (SEQ ID NO: 2) using NcoI and EagI restriction enzymes.
  • Plasmid pCLS8674 (SEQ ID NO: 628) was used to transform the yeast S. cerevisiae strain FYC2-6A (MAT ⁇ , trp1 ⁇ 63, leu2 ⁇ 1, his3 ⁇ 200) using a high efficiency LiAc transformation protocol (Arnould et al. 2007).
  • yeast target reporter plasmids containing the TALEN DNA target sequences were constructed as previously described (International PCT Applications WO 2004/067736 and in Epinat, Arnould et al. 2003; Chames, Epinat et al. 2005; Arnould, Chames et al. 2006; Smith, Grizot et al. 2006).
  • the FokI::TALE-AvrBs3 construct was tested in a yeast SSA assay as previously described (International PCT Applications WO 2004/067736 and in Epinat, Arnould et al. 2003; Chames, Epinat et al. 2005; Arnould, Chames et al. 2006; Smith, Grizot et al. 2006) on pseudo palindromic targets, since the construct requires two binding sites for activity.
  • AvrBs3 targets contain two identical recognition sequences juxtaposed with the 5′ ends proximal ( FIG. 9 D) and separated by “spacer” DNA ranging from 5 to 35 bps (SEQ ID NO: 629 to 659, Table 9).
  • FokI::TALE-AvrBs3 activity levels on the respective targets in yeast cells are shown in FIG. 27 .
  • the catalytic domain of FokI (SEQ ID NO: 600) was subcloned by restriction and ligation into pCLS9009 (SEQ ID NO: 660) using Kpn2I and EagI restriction sites, yielding the construct cT11_FokI (SEQ ID NO: G661).
  • the fusion contains the peptide -GSSG- linking the TALE-derived DNA binding domain and FokI derived catalytic domain.
  • the DNA sequence coding for the RVDs to target the RagT2-R site was subcloned into the cT11_FokI (SEQ ID NO: 661) scaffold using Type IIS restriction enzymes BsmBI for the receiving plasmid and BbvI and SfaNI for the inserted RVD sequence to create the subsequent TALE-RagT2-R::FokI construct cT11RagT2-R_FokI (SEQ ID NO: 663). This construct was sequenced and the insert transferred to additional vectors as needed.
  • the final TALE-RagT2-R::FokI yeast expression plasmid, pCLS9827 (SEQ ID NO: 664), was prepared by restriction and ligation of the cT11RagT2-R_FokI (SEQ ID NO: 663) insert into pCLS7763 (SEQ ID NO: 665) using NcoI and EagI restriction enzymes.
  • the plasmid pair pCLS9827 (SEQ ID NO: 664) and pCLS8674 (SEQ ID NO: 628) was then used in co-transformation experiments in the standard yeast SSA assay as previously described (International PCT Applications WO 2004/067736 and in Epinat, Arnould et al. 2003; Chames, Epinat et al. 2005; Arnould, Chames et al. 2006; Smith, Grizot et al. 2006).
  • yeast target reporter plasmids containing the TALEN DNA target sequences were constructed as previously described (International PCT Applications WO 2004/067736 and in Epinat, Arnould et al. 2003; Chames, Epinat et al. 2005; Arnould, Chames et al. 2006; Smith, Grizot et al. 2006).
  • the TALE-RagT2-R::FokI/FokI::TALE-AvrBs3 construct pairs were tested in a yeast SSA assay as previously described (International PCT Applications WO 2004/067736 and in Epinat, Arnould et al. 2003; Chames, Epinat et al. 2005; Arnould, Chames et al. 2006; Smith, Grizot et al. 2006) on asymmetric RagT2-R/AvrBs3 hybrid targets and compared with a parent standard TALEN (e.g. pCLS8674 (SEQ ID NO: 628) on appropriate pseudo palindromic targets (e.g. (SEQ ID NO: 629 to 659, Table 9).
  • TALEN e.g. pCLS8674 (SEQ ID NO: 628)
  • appropriate pseudo palindromic targets e.g. (SEQ ID NO: 629 to 659, Table 9).
  • RagT2-R/AvrBs3 hybrid targets contain two different recognition sequences juxtaposed with the 3′ end of the first (RagT2-R) proximal to the 5′ end of the second (AvrBs3) and separated by “spacer” DNA ranging from 5 to 40 bps (SEQ ID NO: 666 to 701, Table 10).
  • TALE-RagT2-R::FokI/FokI::TALE-AvrBs3 activity levels on the respective targets in yeast cells are shown in FIG. 28 .

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Engineering & Computer Science (AREA)
  • Genetics & Genomics (AREA)
  • Zoology (AREA)
  • Wood Science & Technology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Molecular Biology (AREA)
  • Biotechnology (AREA)
  • General Health & Medical Sciences (AREA)
  • Biomedical Technology (AREA)
  • Biochemistry (AREA)
  • General Engineering & Computer Science (AREA)
  • Microbiology (AREA)
  • Medicinal Chemistry (AREA)
  • Biophysics (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Physics & Mathematics (AREA)
  • Cell Biology (AREA)
  • Toxicology (AREA)
  • Plant Pathology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Public Health (AREA)
  • Immunology (AREA)
  • Animal Behavior & Ethology (AREA)
  • Crystallography & Structural Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Analytical Chemistry (AREA)
  • Veterinary Medicine (AREA)
  • Enzymes And Modification Thereof (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Peptides Or Proteins (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Breeding Of Plants And Reproduction By Means Of Culturing (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
US14/009,509 2011-04-05 2012-04-05 New tale-protein scaffolds and uses thereof Abandoned US20140115726A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US14/009,509 US20140115726A1 (en) 2011-04-05 2012-04-05 New tale-protein scaffolds and uses thereof

Applications Claiming Priority (9)

Application Number Priority Date Filing Date Title
US201161472065P 2011-04-05 2011-04-05
US201161496454P 2011-06-13 2011-06-13
US201161499047P 2011-06-20 2011-06-20
US201161499043P 2011-06-20 2011-06-20
US201161533098P 2011-09-09 2011-09-09
US201161533123P 2011-09-09 2011-09-09
US201161579544P 2011-12-22 2011-12-22
US14/009,509 US20140115726A1 (en) 2011-04-05 2012-04-05 New tale-protein scaffolds and uses thereof
PCT/US2012/032439 WO2012138939A1 (en) 2011-04-05 2012-04-05 New tale-protein scaffolds and uses thereof

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2012/032439 A-371-Of-International WO2012138939A1 (en) 2011-04-05 2012-04-05 New tale-protein scaffolds and uses thereof

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US16/237,901 Continuation US11136566B2 (en) 2011-04-05 2019-01-02 Tale-protein scaffolds and uses thereof

Publications (1)

Publication Number Publication Date
US20140115726A1 true US20140115726A1 (en) 2014-04-24

Family

ID=45976522

Family Applications (5)

Application Number Title Priority Date Filing Date
US13/440,940 Active 2033-07-20 US9315788B2 (en) 2011-04-05 2012-04-05 Method for the generation of compact TALE-nucleases and uses thereof
US14/009,509 Abandoned US20140115726A1 (en) 2011-04-05 2012-04-05 New tale-protein scaffolds and uses thereof
US15/069,672 Active 2032-08-12 US11198856B2 (en) 2011-04-05 2016-03-14 Method for the generation of compact tale-nucleases and uses thereof
US16/237,901 Active 2032-04-06 US11136566B2 (en) 2011-04-05 2019-01-02 Tale-protein scaffolds and uses thereof
US17/489,454 Pending US20220010292A1 (en) 2011-04-05 2021-09-29 Method for the generation of compact tale-nucleases and uses thereof

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US13/440,940 Active 2033-07-20 US9315788B2 (en) 2011-04-05 2012-04-05 Method for the generation of compact TALE-nucleases and uses thereof

Family Applications After (3)

Application Number Title Priority Date Filing Date
US15/069,672 Active 2032-08-12 US11198856B2 (en) 2011-04-05 2016-03-14 Method for the generation of compact tale-nucleases and uses thereof
US16/237,901 Active 2032-04-06 US11136566B2 (en) 2011-04-05 2019-01-02 Tale-protein scaffolds and uses thereof
US17/489,454 Pending US20220010292A1 (en) 2011-04-05 2021-09-29 Method for the generation of compact tale-nucleases and uses thereof

Country Status (13)

Country Link
US (5) US9315788B2 (de)
EP (3) EP3320910A1 (de)
JP (3) JP5996630B2 (de)
KR (1) KR101982360B1 (de)
CN (1) CN103608027B (de)
AU (2) AU2012240110B2 (de)
CA (2) CA3111953C (de)
DK (1) DK2694091T3 (de)
ES (1) ES2728436T3 (de)
HK (1) HK1253952A1 (de)
IL (1) IL228747B (de)
SG (2) SG194115A1 (de)
WO (2) WO2012138927A2 (de)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20150203871A1 (en) * 2012-06-05 2015-07-23 Cellectis Transcription Activator-Like Effector (TALE) Fusion Protein
US9834791B2 (en) 2013-11-07 2017-12-05 Editas Medicine, Inc. CRISPR-related methods and compositions with governing gRNAS

Families Citing this family (239)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP2342336B1 (de) 2008-09-05 2016-12-14 President and Fellows of Harvard College Kontinuierliche gerichtete evolution von proteinen und nukleinsäuren
WO2011159369A1 (en) * 2010-06-14 2011-12-22 Iowa State University Research Foundation, Inc. Nuclease activity of tal effector and foki fusion protein
US9394537B2 (en) 2010-12-22 2016-07-19 President And Fellows Of Harvard College Continuous directed evolution
EP3461896B1 (de) 2011-07-15 2023-11-29 The General Hospital Corporation Verfahren zur transkription einer aktivatorähnlichen effektoranordnung
EP3613852A3 (de) 2011-07-22 2020-04-22 President and Fellows of Harvard College Beurteilung und verbesserung einer nukleasespaltungsspezifität
WO2013043638A1 (en) * 2011-09-23 2013-03-28 Iowa State University Research Foundation, Inc. Monomer architecture of tal nuclease or zinc finger nuclease for dna modification
EP2776560A2 (de) 2011-11-07 2014-09-17 The University of Western Ontario Endonuklease zur gemonänderung
WO2013074999A1 (en) 2011-11-16 2013-05-23 Sangamo Biosciences, Inc. Modified dna-binding proteins and uses thereof
WO2013101877A2 (en) 2011-12-29 2013-07-04 Iowa State University Research Foundation, Inc. Genetically modified plants with resistance to xanthomonas and other bacterial plant pathogens
EP2825190B1 (de) * 2012-03-15 2016-11-30 Cellectis Di-rückstände mit variabler wiederholung zum anzielen von nukleotiden
US10704021B2 (en) 2012-03-15 2020-07-07 Flodesign Sonics, Inc. Acoustic perfusion devices
WO2013158309A2 (en) * 2012-04-18 2013-10-24 The Board Of Trustees Of The Leland Stanford Junior University Non-disruptive gene targeting
EP2841581B2 (de) 2012-04-23 2023-03-08 BASF Agricultural Solutions Seed US LLC Gezielte genomische veränderungen in pflanzen
WO2013176916A1 (en) 2012-05-25 2013-11-28 Roman Galetto Use of pre t alpha or functional variant thereof for expanding tcr alpha deficient t cells
US20150017136A1 (en) 2013-07-15 2015-01-15 Cellectis Methods for engineering allogeneic and highly active t cell for immunotherapy
US9890364B2 (en) 2012-05-29 2018-02-13 The General Hospital Corporation TAL-Tet1 fusion proteins and methods of use thereof
CA2877290A1 (en) 2012-06-19 2013-12-27 Daniel F. Voytas Gene targeting in plants using dna viruses
US9091827B2 (en) 2012-07-09 2015-07-28 Luxtera, Inc. Method and system for grating couplers incorporating perturbed waveguides
KR102141259B1 (ko) 2012-09-04 2020-08-05 셀렉티스 멀티―체인 키메라 항원 수용체 및 그것의 용도들
CN103668470B (zh) * 2012-09-12 2015-07-29 上海斯丹赛生物技术有限公司 一种dna文库及构建转录激活子样效应因子核酸酶质粒的方法
EP2906602B1 (de) 2012-10-12 2019-01-16 The General Hospital Corporation Transkriptionsaktivator-like-effektor-lysinspezifische demethylase-1 (lsd1)-fusionsproteine
CN110643600A (zh) 2012-10-23 2020-01-03 基因工具股份有限公司 用于切割靶dna的系统及其用途
CA2890160A1 (en) 2012-10-31 2014-05-08 Cellectis Coupling herbicide resistance with targeted insertion of transgenes in plants
RU2711249C2 (ru) * 2012-11-01 2020-01-15 Фэктор Байосайенс Инк. Способы и продукты для экспрессии белков в клетках
EP2920319B1 (de) 2012-11-16 2020-02-19 Poseida Therapeutics, Inc. Ortsspezifische enzyme und verfahren zur verwendung
US20160272980A1 (en) 2012-11-16 2016-09-22 Total Marketing Services Method for targeted modification of algae genomes
WO2014102688A1 (en) 2012-12-27 2014-07-03 Cellectis New design matrix for improvement of homology-directed gene targeting
WO2014118719A1 (en) 2013-02-01 2014-08-07 Cellectis Tevl chimeric endonuclease and their preferential cleavage sites
EP3623463B1 (de) 2013-02-07 2021-10-20 The General Hospital Corporation Tale-transkriptionsaktivatoren
WO2014128659A1 (en) 2013-02-21 2014-08-28 Cellectis Method to counter-select cells or organisms by linking loci to nuclease components
EP2971167B1 (de) * 2013-03-14 2019-07-31 Caribou Biosciences, Inc. Zusammensetzungen und verfahren zum targeting von nukleinsäuren durch nukleinsäuren
US9937207B2 (en) * 2013-03-21 2018-04-10 Sangamo Therapeutics, Inc. Targeted disruption of T cell receptor genes using talens
CA2908403A1 (en) 2013-04-02 2014-10-09 Bayer Cropscience Nv Targeted genome engineering in eukaryotes
WO2014169810A1 (zh) * 2013-04-16 2014-10-23 深圳华大基因科技服务有限公司 分离的寡核苷酸及其用途
CN105431532B (zh) 2013-05-13 2021-04-06 瑟勒提斯公司 Cd19特异性嵌合抗原受体及其用途
KR102220382B1 (ko) 2013-05-13 2021-02-25 셀렉티스 면역요법을 위한 매우 활성인 t 세포를 조작하는 방법
CA2913872C (en) * 2013-05-31 2022-01-18 Cellectis A laglidadg homing endonuclease cleaving the t-cell receptor alpha gene and uses thereof
CA2913871C (en) * 2013-05-31 2021-07-13 Cellectis A laglidadg homing endonuclease cleaving the c-c chemokine receptor type-5 (ccr5) gene and uses thereof
JP2016521561A (ja) * 2013-06-14 2016-07-25 セレクティス 植物における非トランスジェニックのゲノム編集のための方法
EP3013939A1 (de) * 2013-06-25 2016-05-04 Cellectis Modifizierte diatome für biokraftstoffproduktion
FR3008106A1 (fr) * 2013-07-05 2015-01-09 Agronomique Inst Nat Rech Methode d'induction d'une recombinaison meiotique ciblee
US10782479B2 (en) 2013-07-08 2020-09-22 Luxtera Llc Method and system for mode converters for grating couplers
US20150044192A1 (en) 2013-08-09 2015-02-12 President And Fellows Of Harvard College Methods for identifying a target site of a cas9 nuclease
US10006011B2 (en) * 2013-08-09 2018-06-26 Hiroshima University Polypeptide containing DNA-binding domain
US9359599B2 (en) * 2013-08-22 2016-06-07 President And Fellows Of Harvard College Engineered transcription activator-like effector (TALE) domains and uses thereof
US9322037B2 (en) 2013-09-06 2016-04-26 President And Fellows Of Harvard College Cas9-FokI fusion proteins and uses thereof
US9737604B2 (en) 2013-09-06 2017-08-22 President And Fellows Of Harvard College Use of cationic lipids to deliver CAS9
US9228207B2 (en) 2013-09-06 2016-01-05 President And Fellows Of Harvard College Switchable gRNAs comprising aptamers
WO2015042585A1 (en) * 2013-09-23 2015-03-26 Rensselaer Polytechnic Institute Nanoparticle-mediated gene delivery, genomic editing and ligand-targeted modification in various cell populations
US9476884B2 (en) 2013-10-04 2016-10-25 University Of Massachusetts Hybridization- independent labeling of repetitive DNA sequence in human chromosomes
CA2927965A1 (en) * 2013-10-25 2015-04-30 Cellectis Design of rare-cutting endonucleases for efficient and specific targeting dna sequences comprising highly repetitive motives
US9068179B1 (en) 2013-12-12 2015-06-30 President And Fellows Of Harvard College Methods for correcting presenilin point mutations
CN105939767B (zh) 2014-01-08 2018-04-06 弗洛设计声能学公司 具有双声电泳腔的声电泳装置
US10179911B2 (en) 2014-01-20 2019-01-15 President And Fellows Of Harvard College Negative selection and stringency modulation in continuous evolution systems
DK3105317T3 (en) 2014-02-14 2019-01-14 Cellectis Immunotherapy cells that are genetically engineered to target antigen on both immune cells and pathological cells
KR102228828B1 (ko) 2014-03-11 2021-03-16 셀렉티스 동종이형 이식에 양립성인 t-세포들을 만들어내는 방법
MX370272B (es) 2014-03-19 2019-12-09 Cellectis Receptores de antigeno quimerico especifico de cd123 para inmunoterapia de cancer.
CN106460009A (zh) 2014-03-21 2017-02-22 小利兰·斯坦福大学托管委员会 无核酸酶的基因组编辑
EP3129473B8 (de) 2014-04-11 2020-12-23 Cellectis Verfahren zur erzeugung von immunzellen, die gegen eine arginin- und/oder tryptophanverarmte mikroumgebung resistent sind
AU2015277180A1 (en) * 2014-06-17 2017-01-12 Poseida Therapeutics, Inc. A method for directing proteins to specific loci in the genome and uses thereof
ES2876925T3 (es) 2014-07-29 2021-11-15 Cellectis Receptores de antígeno quiméricos específicos para ROR1 (NTRKR1) para inmunoterapia contra el cáncer
AU2015298571B2 (en) 2014-07-30 2020-09-03 President And Fellows Of Harvard College Cas9 proteins including ligand-dependent inteins
EP3194432B1 (de) 2014-07-31 2019-04-10 Cellectis Ror1-spezifischer mehrkettiger chimärer antigenrezeptor
CA2959694C (en) 2014-09-04 2023-11-21 Cellectis Trophoblast glycoprotein (5t4, tpbg) specific chimeric antigen receptors for cancer immunotherapy
WO2016077052A2 (en) 2014-10-22 2016-05-19 President And Fellows Of Harvard College Evolution of proteases
US11014989B2 (en) 2015-01-26 2021-05-25 Cellectis Anti-CLL1 specific single-chain chimeric antigen receptors (scCARs) for cancer immunotherapy
CA2976376A1 (en) 2015-02-13 2016-08-18 Factor Bioscience Inc. Nucleic acid products and methods of administration thereof
US20160244784A1 (en) * 2015-02-15 2016-08-25 Massachusetts Institute Of Technology Population-Hastened Assembly Genetic Engineering
RU2017134441A (ru) 2015-03-11 2019-04-03 Селлектис Способы конструирования аллогенных т-клеток для повышения их персистенции и/или приживления у пациентов
WO2016168631A1 (en) 2015-04-17 2016-10-20 President And Fellows Of Harvard College Vector-based mutagenesis system
US11708572B2 (en) 2015-04-29 2023-07-25 Flodesign Sonics, Inc. Acoustic cell separation techniques and processes
US11377651B2 (en) 2016-10-19 2022-07-05 Flodesign Sonics, Inc. Cell therapy processes utilizing acoustophoresis
ES2905181T3 (es) * 2015-05-01 2022-04-07 Prec Biosciences Inc Deleción precisa de secuencias cromosómicas in vivo
US10752670B2 (en) 2015-05-20 2020-08-25 Cellectis Anti-GD3 specific chimeric antigen receptors for cancer immunotherapy
EP3929286A1 (de) 2015-06-17 2021-12-29 Poseida Therapeutics, Inc. Zusammensetzungen und verfahren zur führung von proteinen an spezifische loci im genom
EP3317399B1 (de) 2015-06-30 2024-06-26 Cellectis Verfahren zur verbesserung der funktionalität in einer nk-zelle durch geninaktivierung mit spezifischer endonuklease
US10392674B2 (en) 2015-07-22 2019-08-27 President And Fellows Of Harvard College Evolution of site-specific recombinases
US11524983B2 (en) 2015-07-23 2022-12-13 President And Fellows Of Harvard College Evolution of Bt toxins
WO2017019895A1 (en) 2015-07-30 2017-02-02 President And Fellows Of Harvard College Evolution of talens
CN104962523B (zh) 2015-08-07 2018-05-25 苏州大学 一种测定非同源末端连接修复活性的方法
CN108495641A (zh) 2015-08-11 2018-09-04 塞勒克提斯公司 用于靶向cd38抗原和用于cd38基因失活的工程化的用于免疫疗法的细胞
CN108513575A (zh) 2015-10-23 2018-09-07 哈佛大学的校长及成员们 核碱基编辑器及其用途
WO2017079428A1 (en) 2015-11-04 2017-05-11 President And Fellows Of Harvard College Site specific germline modification
EP3429634A1 (de) 2016-04-15 2019-01-23 Cellectis Verfahren zum engineering von prodrug-spezifischen überempfindlichen t-zellen zur immuntherapie durch genexpression
US10894093B2 (en) 2016-04-15 2021-01-19 Cellectis Method of engineering drug-specific hypersensitive t-cells for immunotherapy by gene inactivation
US11214789B2 (en) 2016-05-03 2022-01-04 Flodesign Sonics, Inc. Concentration and washing of particles with acoustics
EP4219731A3 (de) 2016-05-18 2023-08-09 Amyris, Inc. Zusammensetzungen und verfahren zur genomischen integration von nukleinsäuren in exogene landepads
MX2018014496A (es) 2016-05-26 2019-03-28 Nunhems Bv Plantas productoras de frutas sin semillas.
WO2018027078A1 (en) 2016-08-03 2018-02-08 President And Fellows Of Harard College Adenosine nucleobase editors and uses thereof
CA3033327A1 (en) 2016-08-09 2018-02-15 President And Fellows Of Harvard College Programmable cas9-recombinase fusion proteins and uses thereof
KR102607893B1 (ko) 2016-08-17 2023-12-01 몬산토 테크놀로지 엘엘씨 지베렐린 대사의 조작을 통해 저신장 식물의 수확량을 증가시키기 위한 방법 및 조성물
US10576167B2 (en) 2016-08-17 2020-03-03 Factor Bioscience Inc. Nucleic acid products and methods of administration thereof
WO2018039438A1 (en) 2016-08-24 2018-03-01 President And Fellows Of Harvard College Incorporation of unnatural amino acids into proteins using base editing
EP3757120B8 (de) 2016-10-04 2022-06-15 Precision Biosciences, Inc. Co-stimulierende domänen zur verwendung bei genetisch veränderten zellen
KR20240007715A (ko) 2016-10-14 2024-01-16 프레지던트 앤드 펠로우즈 오브 하바드 칼리지 핵염기 에디터의 aav 전달
AU2017378427A1 (en) 2016-12-14 2019-06-20 Ligandal, Inc. Methods and compositions for nucleic acid and protein payload delivery
WO2018115189A1 (en) 2016-12-21 2018-06-28 Cellectis Stably enginereed proteasome inhibitor resistant immune cells for immunotherapy
US10745677B2 (en) 2016-12-23 2020-08-18 President And Fellows Of Harvard College Editing of CCR5 receptor gene to protect against HIV infection
EP3592853A1 (de) 2017-03-09 2020-01-15 President and Fellows of Harvard College Unterdrückung von schmerzen durch geneditierung
WO2018162702A1 (en) 2017-03-10 2018-09-13 Institut National De La Sante Et De La Recherche Medicale (Inserm) Nuclease fusions for enhancing genome editing by homology-directed transgene integration
JP2020510439A (ja) 2017-03-10 2020-04-09 プレジデント アンド フェローズ オブ ハーバード カレッジ シトシンからグアニンへの塩基編集因子
JP6901293B2 (ja) * 2017-03-15 2021-07-14 株式会社三共 遊技用装置
SG11201908658TA (en) 2017-03-23 2019-10-30 Harvard College Nucleobase editors comprising nucleic acid programmable dna binding proteins
CA3058268A1 (en) 2017-03-31 2018-10-04 Cellectis Sa Universal anti-cd22 chimeric antigen receptor engineered immune cells
CA3062698A1 (en) 2017-05-08 2018-11-15 Precision Biosciences, Inc. Nucleic acid molecules encoding an engineered antigen receptor and an inhibitory nucleic acid molecule and methods of use thereof
WO2019103857A1 (en) 2017-11-22 2019-05-31 Iovance Biotherapeutics, Inc. Expansion of peripheral blood lymphocytes (pbls) from peripheral blood
US11560566B2 (en) 2017-05-12 2023-01-24 President And Fellows Of Harvard College Aptazyme-embedded guide RNAs for use with CRISPR-Cas9 in genome editing and transcriptional activation
CA3064014A1 (en) 2017-05-25 2018-11-29 Bluebird Bio, Inc. Cblb endonuclease variants, compositions, and methods of use
JP2020530277A (ja) 2017-06-30 2020-10-22 セレクティスCellectis 反復投与のための細胞免疫療法
US11447809B2 (en) 2017-07-06 2022-09-20 President And Fellows Of Harvard College Evolution of tRNA synthetases
US11732274B2 (en) 2017-07-28 2023-08-22 President And Fellows Of Harvard College Methods and compositions for evolving base editors using phage-assisted continuous evolution (PACE)
US11319532B2 (en) 2017-08-30 2022-05-03 President And Fellows Of Harvard College High efficiency base editors comprising Gam
US11624130B2 (en) 2017-09-18 2023-04-11 President And Fellows Of Harvard College Continuous evolution for stabilized proteins
WO2019060631A1 (en) * 2017-09-20 2019-03-28 Helix Nanotechnologies, Inc. EXPRESSION SYSTEMS THAT FACILITATE THE DELIVERY OF NUCLEIC ACIDS AND METHODS OF USE
WO2019070856A1 (en) 2017-10-03 2019-04-11 Precision Biosciences, Inc. MODIFIED EPIDERMAL GROWTH FACTOR RECEPTOR PEPTIDES FOR USE IN GENETICALLY MODIFIED CELLS
WO2019072824A1 (en) 2017-10-09 2019-04-18 Cellectis IMPROVED ANTI-CD123 CAR IN UNIVERSAL MODIFIED IMMUNE T LYMPHOCYTES
CN111757937A (zh) 2017-10-16 2020-10-09 布罗德研究所股份有限公司 腺苷碱基编辑器的用途
SG11202004457XA (en) 2017-11-17 2020-06-29 Iovance Biotherapeutics Inc Til expansion from fine needle aspirates and small biopsies
CA3085784A1 (en) 2017-12-14 2019-06-20 Flodesign Sonics, Inc. Acoustic transducer driver and controller
EP3501268B1 (de) 2017-12-22 2021-09-15 KWS SAAT SE & Co. KGaA Regeneration von pflanzen in der gegenwart von histondeacetylaseinhibitoren
EP3508581A1 (de) 2018-01-03 2019-07-10 Kws Saat Se Regenerierung von genetisch modifizierten pflanzen
AU2019207409B2 (en) 2018-01-12 2023-02-23 Basf Se Gene underlying the number of spikelets per spike qtl in wheat on chromosome 7a
WO2019149743A1 (en) 2018-01-30 2019-08-08 Cellectis Combination comprising allogeneic immune cells deficient for an antigen present on both t-cells and pathological cells and therapeutic antibody against said antigen
MX2020008560A (es) 2018-02-15 2020-10-12 Monsanto Technology Llc Composiciones y metodos para mejorar los rendimientos de cultivos mediante el apilamiento de rasgos.
EP3545756A1 (de) 2018-03-28 2019-10-02 KWS SAAT SE & Co. KGaA Regeneration von pflanzen in gegenwart von inhibitoren der histonmethyltransferase ezh2
SG11202009446TA (en) 2018-04-05 2020-10-29 Juno Therapeutics Inc T cells expressing a recombinant receptor, related polynucleotides and methods
MA52656A (fr) 2018-04-05 2021-02-17 Editas Medicine Inc Procédés de production de cellules exprimant un récepteur recombinant et compositions associées
CN112368003A (zh) 2018-04-27 2021-02-12 艾欧凡斯生物治疗公司 肿瘤浸润淋巴细胞的基因编辑及其在免疫治疗中的用途
MX2020011656A (es) 2018-05-02 2021-02-26 Cellectis Trigo manipulado con elevada fibra vegetal.
EP3567111A1 (de) 2018-05-09 2019-11-13 KWS SAAT SE & Co. KGaA Gen für resistenz gegen ein pathogen der gattung heterodera
WO2019241649A1 (en) 2018-06-14 2019-12-19 President And Fellows Of Harvard College Evolution of cytidine deaminases
EP3806619A1 (de) 2018-06-15 2021-04-21 Nunhems B.V. Kernlose wassermelonenpflanzen mit modifikationen in einem abc-transporter-gen
US20210254087A1 (en) 2018-06-15 2021-08-19 KWS SAAT SE & Co. KGaA Methods for enhancing genome engineering efficiency
US20220025388A1 (en) 2018-06-15 2022-01-27 KWS SAAT SE & Co. KGaA Methods for improving genome engineering and regeneration in plant
CA3103500A1 (en) 2018-06-15 2019-12-19 KWS SAAT SE & Co. KGaA Methods for improving genome engineering and regeneration in plant ii
EP3817767A1 (de) 2018-07-02 2021-05-12 Cellectis Chimäre antigen-rezeptoren (auto) exprimierende zellen und kombinationstherapie zur immuntherapie von patienten mit rezidivierender oder refraktärer aml mit genetischem risiko
EP3623379A1 (de) 2018-09-11 2020-03-18 KWS SAAT SE & Co. KGaA Gen zur modifizierung von rhizomania-virus (bnyvv)-resistenz
WO2020072059A1 (en) * 2018-10-04 2020-04-09 Bluebird Bio, Inc. Cblb endonuclease variants, compositions, and methods of use
US20220033775A1 (en) 2018-11-05 2022-02-03 Iovance Biotherapeutics, Inc. Expansion of tils utilizing akt pathways inhibitors
JP2022512899A (ja) 2018-11-05 2022-02-07 アイオバンス バイオセラピューティクス,インコーポレイテッド 抗pd-1抗体に対して不応性のnsclc患者の治療
AU2019374761A1 (en) 2018-11-05 2021-06-10 Iovance Biotherapeutics, Inc. Processes for production of tumor infiltrating lymphocytes and uses of the same in immunotherapy
TW202039829A (zh) 2018-11-05 2020-11-01 美商艾歐凡斯生物治療公司 改善之腫瘤反應性t細胞的選擇
US20220193131A1 (en) 2018-12-19 2022-06-23 Iovance Biotherapeutics, Inc. Methods of Expanding Tumor Infiltrating Lymphocytes Using Engineered Cytokine Receptor Pairs and Uses Thereof
EP3898661A1 (de) 2018-12-21 2021-10-27 Precision BioSciences, Inc. Genetische modifikation des hydroxysäure-oxidase-1-gens zur behandlung von primärer hyperoxalurie
WO2020157573A1 (en) 2019-01-29 2020-08-06 The University Of Warwick Methods for enhancing genome engineering efficiency
MX2021010288A (es) 2019-03-01 2021-09-23 Iovance Biotherapeutics Inc Expansion de linfocitos infiltrantes de tumores a partir de tumores liquidos y usos terapeuticos de los mismos.
EP3708651A1 (de) 2019-03-12 2020-09-16 KWS SAAT SE & Co. KGaA Verbesserung der pflanzenregeneration
DE112020001342T5 (de) 2019-03-19 2022-01-13 President and Fellows of Harvard College Verfahren und Zusammensetzungen zum Editing von Nukleotidsequenzen
US11427814B2 (en) 2019-03-26 2022-08-30 Encodia, Inc. Modified cleavases, uses thereof and related kits
AU2020247918B2 (en) * 2019-03-26 2022-06-30 Encodia, Inc. Modified cleavases, uses thereof and related kits
CA3135799A1 (en) 2019-04-03 2020-10-08 Precision Biosciences, Inc. Genetically-modified immune cells comprising a microrna-adapted shrna (shrnamir)
CA3136265A1 (en) 2019-04-05 2020-10-08 Precision Biosciences, Inc. Methods of preparing populations of genetically-modified immune cells
BR112021021200A2 (pt) 2019-05-01 2021-12-21 Juno Therapeutics Inc Células expressando um receptor quimérico de um locus cd247 modificado, polinucleotídeos relacionados e métodos
CA3136737A1 (en) 2019-05-01 2020-11-05 Juno Therapeutics, Inc. Cells expressing a recombinant receptor from a modified tgfbr2 locus, related polynucleotides and methods
EP3966334A1 (de) 2019-05-10 2022-03-16 Basf Se Regulatorische nukleinsäuremoleküle zur steigerung der genexpression bei pflanzen
US20220249559A1 (en) 2019-05-13 2022-08-11 Iovance Biotherapeutics, Inc. Methods and compositions for selecting tumor infiltrating lymphocytes and uses of the same in immunotherapy
EP3757219A1 (de) 2019-06-28 2020-12-30 KWS SAAT SE & Co. KGaA Verbesserte pflanzenregeneration und -umwandlung unter verwendung des grf1-booster-gens
US20220273715A1 (en) 2019-07-25 2022-09-01 Precision Biosciences, Inc. Compositions and methods for sequential stacking of nucleic acid sequences into a genomic locus
US10501404B1 (en) 2019-07-30 2019-12-10 Factor Bioscience Inc. Cationic lipids and transfection methods
US20220267732A1 (en) 2019-08-01 2022-08-25 Sana Biotechnology, Inc. Dux4 expressing cells and uses thereof
AU2020333851A1 (en) 2019-08-20 2022-03-31 Precision Biosciences, Inc. Lymphodepletion dosing regimens for cellular immunotherapies
WO2021035170A1 (en) 2019-08-21 2021-02-25 Precision Biosciences, Inc. Compositions and methods for tcr reprogramming using fusion proteins
EP4017508A1 (de) 2019-08-23 2022-06-29 Sana Biotechnology, Inc. Cd24+-exprimierende zellen und ihre verwendungen
AU2020344905A1 (en) 2019-09-12 2022-04-28 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
US20220389381A1 (en) 2019-10-25 2022-12-08 Iovance Biotherapeutics, Inc. Gene editing of tumor infiltrating lymphocytes and uses of same in immunotherapy
WO2021087305A1 (en) 2019-10-30 2021-05-06 Precision Biosciences, Inc. Cd20 chimeric antigen receptors and methods of use for immunotherapy
CA3157872A1 (en) 2019-11-12 2021-05-20 Otto Torjek Gene for resistance to a pathogen of the genus heterodera
US20230148071A1 (en) 2019-12-03 2023-05-11 Basf Se Regulatory nucleic acid molecules for enhancing gene expression in plants
WO2021113543A1 (en) 2019-12-06 2021-06-10 Precision Biosciences, Inc. Methods for cancer immunotherapy, using lymphodepletion regimens and cd19, cd20 or bcma allogeneic car t cells
CA3161104A1 (en) 2019-12-11 2021-06-17 Cecile Chartier-Courtaud Processes for the production of tumor infiltrating lymphocytes (tils) and methods of using the same
AU2021207810A1 (en) 2020-01-13 2022-08-04 Sana Biotechnology, Inc. Modification of blood type antigens
JP2023510916A (ja) 2020-01-17 2023-03-15 サナ バイオテクノロジー,インコーポレイテッド 遺伝子発現の制御のための安全スイッチ
WO2021158915A1 (en) 2020-02-06 2021-08-12 Precision Biosciences, Inc. Recombinant adeno-associated virus compositions and methods for producing and using the same
JP2023520997A (ja) 2020-03-25 2023-05-23 サナ バイオテクノロジー,インコーポレイテッド 神経学的障害及び神経学的病態の処置のための低免疫原性神経細胞
CN115697044A (zh) 2020-03-31 2023-02-03 艾洛生物系统有限公司 西瓜和其他葫芦科中内源罗汉果苷途径基因的调控
US20230172987A1 (en) 2020-05-04 2023-06-08 Iovance Biotherapeutics, Inc. Processes for production of tumor infiltrating lymphocytes and uses of the same in immunotherapy
EP4146804A1 (de) 2020-05-08 2023-03-15 The Broad Institute Inc. Verfahren und zusammensetzungen zur gleichzeitigen bearbeitung beider stränge einer doppelsträngigen zielnukleotidsequenz
WO2021231259A1 (en) 2020-05-11 2021-11-18 Precision Biosciences, Inc. Self-limiting viral vectors encoding nucleases
EP4149252A2 (de) * 2020-05-12 2023-03-22 Factor Bioscience Inc. Gentechnologisch hergestellte proteine
WO2021231661A2 (en) 2020-05-13 2021-11-18 Juno Therapeutics, Inc. Process for producing donor-batched cells expressing a recombinant receptor
WO2021260186A1 (en) 2020-06-26 2021-12-30 Juno Therapeutics Gmbh Engineered t cells conditionally expressing a recombinant receptor, related polynucleotides and methods
EP4192875A1 (de) 2020-08-10 2023-06-14 Precision BioSciences, Inc. Antikörper und fragmente spezifisch für b-zellreifungsantigen und verwendungen davon
WO2022036150A1 (en) 2020-08-13 2022-02-17 Sana Biotechnology, Inc. Methods of treating sensitized patients with hypoimmunogenic cells, and associated methods and compositions
WO2022076606A1 (en) 2020-10-06 2022-04-14 Iovance Biotherapeutics, Inc. Treatment of nsclc patients with tumor infiltrating lymphocyte therapies
US20230372397A1 (en) 2020-10-06 2023-11-23 Iovance Biotherapeutics, Inc. Treatment of nsclc patients with tumor infiltrating lymphocyte therapies
US20230365995A1 (en) 2020-10-07 2023-11-16 Precision Biosciences, Inc. Lipid nanoparticle compositions
WO2022087527A1 (en) 2020-10-23 2022-04-28 Elo Life Systems, Inc. Methods for producing vanilla plants with improved flavor and agronomic production
EP4240756A1 (de) 2020-11-04 2023-09-13 Juno Therapeutics, Inc. Zellen zur expression eines chimären rezeptors aus einem modifizierten invarianten kettenlocus der cd3-immunglobulin-superfamilie und zugehörige polynukleotide und verfahren
EP4262811A1 (de) 2020-12-17 2023-10-25 Iovance Biotherapeutics, Inc. Behandlung mit tumorinfiltrierenden lymphozytentherapien in kombination mit ctla-4- und pd-1-inhibitoren
WO2022133149A1 (en) 2020-12-17 2022-06-23 Iovance Biotherapeutics, Inc. Treatment of cancers with tumor infiltrating lymphocytes
EP4019638A1 (de) 2020-12-22 2022-06-29 KWS SAAT SE & Co. KGaA Förderung der regeneration und transformation in beta vulgaris
EP4019639A1 (de) 2020-12-22 2022-06-29 KWS SAAT SE & Co. KGaA Förderung der regeneration und transformation in beta vulgaris
IL303473A (en) 2020-12-31 2023-08-01 Sana Biotechnology Inc Methods and compositions for modulating CAR-T activity
EP4284823A1 (de) 2021-01-28 2023-12-06 Precision BioSciences, Inc. Modulation der tgf-beta-signalisierung in genetisch modifizierten eukaryotischen zellen
EP4284919A1 (de) 2021-01-29 2023-12-06 Iovance Biotherapeutics, Inc. Verfahren zur herstellung von modifizierten tumorinfiltrierenden lymphozyten und deren verwendung in der adoptiven zelltherapie
TW202304480A (zh) 2021-03-19 2023-02-01 美商艾歐凡斯生物治療公司 腫瘤浸潤淋巴球(til)中之與cd39/cd69選擇及基因剔除相關之til擴增之方法
WO2022204155A1 (en) 2021-03-23 2022-09-29 Iovance Biotherapeutics, Inc. Cish gene editing of tumor infiltrating lymphocytes and uses of same in immunotherapy
AU2022263418A1 (en) 2021-04-19 2023-10-26 Iovance Biotherapeutics, Inc. Chimeric costimulatory receptors, chemokine receptors, and the use of same in cellular immunotherapies
US20240141311A1 (en) 2021-04-22 2024-05-02 North Carolina State University Compositions and methods for generating male sterile plants
EP4340850A1 (de) 2021-05-17 2024-03-27 Iovance Biotherapeutics, Inc. Pd-1-geneditierte tumorinfiltrierende lymphozyten und verwendungen davon in der immuntherapie
BR112023024434A2 (pt) 2021-05-27 2024-02-20 Sana Biotechnology Inc Células hipoimunogênicas que compreendem hla-e ou hla-g geneticamente modificadas
AU2022309875A1 (en) 2021-07-14 2024-01-25 Sana Biotechnology, Inc. Altered expression of y chromosome-linked antigens in hypoimmunogenic cells
EP4373270A2 (de) 2021-07-22 2024-05-29 Iovance Biotherapeutics, Inc. Verfahren zur kryokonservierung von festen tumorfragmenten
CA3226942A1 (en) 2021-07-28 2023-02-02 Iovance Biotherapeutics, Inc. Treatment of cancer patients with tumor infiltrating lymphocyte therapies in combination with kras inhibitors
AU2022325231A1 (en) 2021-08-11 2024-02-08 Sana Biotechnology, Inc. Genetically modified cells for allogeneic cell therapy to reduce complement-mediated inflammatory reactions
AU2022325955A1 (en) 2021-08-11 2024-02-08 Sana Biotechnology, Inc. Genetically modified cells for allogeneic cell therapy to reduce instant blood mediated inflammatory reactions
EP4384544A1 (de) 2021-08-11 2024-06-19 Sana Biotechnology, Inc. Genetisch modifizierte zellen für allogene zelltherapie
IL310691A (en) 2021-08-11 2024-04-01 Sana Biotechnology Inc Genetically modified primary cells for allogeneic cell therapy
EP4398915A1 (de) 2021-09-09 2024-07-17 Iovance Biotherapeutics, Inc. Verfahren zur erzeugung von til-produkten mittels pd-1-talen-knockdown
WO2023064872A1 (en) 2021-10-14 2023-04-20 Precision Biosciences, Inc. Combinations of anti-bcma car t cells and gamma secretase inhibitors
AU2022371430A1 (en) 2021-10-19 2024-05-30 Precision Biosciences, Inc. Gene editing methods for treating alpha-1 antitrypsin (aat) deficiency
AR127482A1 (es) 2021-10-27 2024-01-31 Iovance Biotherapeutics Inc Sistemas y métodos para coordinar la fabricación de células para inmunoterapia específica de paciente
CN113980141B (zh) * 2021-10-27 2022-11-29 湖北大学 基于大肠杆菌素e家族dna酶的蛋白质复合物及其在人工蛋白支架中的应用
WO2023081767A1 (en) 2021-11-05 2023-05-11 Precision Biosciences, Inc. Methods for immunotherapy
WO2023081900A1 (en) 2021-11-08 2023-05-11 Juno Therapeutics, Inc. Engineered t cells expressing a recombinant t cell receptor (tcr) and related systems and methods
US11753677B2 (en) 2021-11-10 2023-09-12 Encodia, Inc. Methods for barcoding macromolecules in individual cells
WO2023091910A1 (en) 2021-11-16 2023-05-25 Precision Biosciences, Inc. Methods for cancer immunotherapy
AU2022395500A1 (en) 2021-11-23 2024-05-23 Cellectis Sa New tale protein scaffolds with improved on-target/off-target activity ratios
WO2023133525A1 (en) 2022-01-07 2023-07-13 Precision Biosciences, Inc. Optimized polynucleotides for protein expression
WO2023147488A1 (en) 2022-01-28 2023-08-03 Iovance Biotherapeutics, Inc. Cytokine associated tumor infiltrating lymphocytes compositions and methods
WO2023158836A1 (en) 2022-02-17 2023-08-24 Sana Biotechnology, Inc. Engineered cd47 proteins and uses thereof
WO2023173123A1 (en) 2022-03-11 2023-09-14 Sana Biotechnology, Inc. Genetically modified cells and compositions and uses thereof
WO2023183313A1 (en) 2022-03-22 2023-09-28 Sana Biotechnology, Inc. Engineering cells with a transgene in b2m or ciita locus and associated compositions and methods
WO2023196877A1 (en) 2022-04-06 2023-10-12 Iovance Biotherapeutics, Inc. Treatment of nsclc patients with tumor infiltrating lymphocyte therapies
WO2023201369A1 (en) 2022-04-15 2023-10-19 Iovance Biotherapeutics, Inc. Til expansion processes using specific cytokine combinations and/or akti treatment
WO2023220608A1 (en) 2022-05-10 2023-11-16 Iovance Biotherapeutics, Inc. Treatment of cancer patients with tumor infiltrating lymphocyte therapies in combination with an il-15r agonist
WO2024055017A1 (en) 2022-09-09 2024-03-14 Iovance Biotherapeutics, Inc. Processes for generating til products using pd-1/tigit talen double knockdown
WO2024055018A1 (en) 2022-09-09 2024-03-14 Iovance Biotherapeutics, Inc. Processes for generating til products using pd-1/tigit talen double knockdown
WO2024083579A1 (en) 2022-10-20 2024-04-25 Basf Se Regulatory nucleic acid molecules for enhancing gene expression in plants
WO2024097314A2 (en) 2022-11-02 2024-05-10 Sana Biotechnology, Inc. Methods and systems for determining donor cell features and formulating cell therapy products based on cell features
WO2024098027A1 (en) 2022-11-04 2024-05-10 Iovance Biotherapeutics, Inc. Methods for tumor infiltrating lymphocyte (til) expansion related to cd39/cd103 selection
WO2024098024A1 (en) 2022-11-04 2024-05-10 Iovance Biotherapeutics, Inc. Expansion of tumor infiltrating lymphocytes from liquid tumors and therapeutic uses thereof
WO2024100604A1 (en) 2022-11-09 2024-05-16 Juno Therapeutics Gmbh Methods for manufacturing engineered immune cells
WO2024112711A2 (en) 2022-11-21 2024-05-30 Iovance Biotherapeutics, Inc. Methods for assessing proliferation potency of gene-edited t cells
WO2024112571A2 (en) 2022-11-21 2024-05-30 Iovance Biotherapeutics, Inc. Two-dimensional processes for the expansion of tumor infiltrating lymphocytes and therapies therefrom
WO2024118836A1 (en) 2022-11-30 2024-06-06 Iovance Biotherapeutics, Inc. Processes for production of tumor infiltrating lymphocytes with shortened rep step
CN117344048A (zh) * 2023-09-28 2024-01-05 江苏省农业科学院 检测无毒基因Avr-Pik的RPA-LFD引物探针组、试剂盒及其应用
CN117821413A (zh) * 2023-12-15 2024-04-05 湖北大学 一种高进行性Pfu DNA聚合酶及其制备方法和应用

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8673557B2 (en) * 2011-02-28 2014-03-18 Seattle Children's Research Institute Coupling endonucleases with end-processing enzymes drives high efficiency gene disruption

Family Cites Families (28)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4179337A (en) 1973-07-20 1979-12-18 Davis Frank F Non-immunogenic polypeptides
US4683195A (en) 1986-01-30 1987-07-28 Cetus Corporation Process for amplifying, detecting, and/or-cloning nucleic acid sequences
US5006333A (en) 1987-08-03 1991-04-09 Ddi Pharmaceuticals, Inc. Conjugates of superoxide dismutase coupled to high molecular weight polyalkylene glycols
DE60316124T3 (de) 2002-03-15 2018-03-22 Cellectis Hybride and einzelkettige meganukleasen und deren anwendungen
WO2009095742A1 (en) 2008-01-31 2009-08-06 Cellectis New i-crei derived single-chain meganuclease and uses thereof
AU2003290518A1 (en) 2002-09-06 2004-04-23 Fred Hutchinson Cancer Research Center Methods and compositions concerning designed highly-specific nucleic acid binding proteins
AU2004208031B2 (en) 2003-01-28 2009-10-08 Cellectis Use of meganucleases for inducing homologous recombination ex vivo and in toto in vertebrate somatic tissues and application thereof.
WO2006097784A1 (en) 2005-03-15 2006-09-21 Cellectis I-crei meganuclease variants with modified specificity, method of preparation and uses thereof
WO2007034262A1 (en) 2005-09-19 2007-03-29 Cellectis Heterodimeric meganucleases and use thereof
US20110158974A1 (en) 2005-03-15 2011-06-30 Cellectis Heterodimeric Meganucleases and Use Thereof
WO2007049095A1 (en) 2005-10-25 2007-05-03 Cellectis Laglidadg homing endonuclease variants having mutations in two functional subdomains and use thereof
WO2007060495A1 (en) 2005-10-25 2007-05-31 Cellectis I-crei homing endonuclease variants having novel cleavage specificity and use thereof
WO2007093836A1 (en) 2006-02-13 2007-08-23 Cellectis Meganuclease variants cleaving a dna target sequence from a xp gene and uses thereof
WO2008010009A1 (en) 2006-07-18 2008-01-24 Cellectis Meganuclease variants cleaving a dna target sequence from a rag gene and uses thereof
BRPI0718747A2 (pt) 2006-11-14 2013-12-03 Cellectis Variantes meganuclease que clavam uma ou sequência alvo de dna a partir do gene hprt e usos das mesmas.
WO2008093152A1 (en) 2007-02-01 2008-08-07 Cellectis Obligate heterodimer meganucleases and uses thereof
WO2008102199A1 (en) 2007-02-20 2008-08-28 Cellectis Meganuclease variants cleaving a dna target sequence from the beta-2-microglobulin gene 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
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
EP2206723A1 (de) * 2009-01-12 2010-07-14 Bonas, Ulla Modulare DNA-bindende Domänen
US8586526B2 (en) * 2010-05-17 2013-11-19 Sangamo Biosciences, Inc. DNA-binding proteins and uses thereof
US8956828B2 (en) * 2009-11-10 2015-02-17 Sangamo Biosciences, Inc. Targeted disruption of T cell receptor genes using engineered zinc finger protein nucleases
EP2504430A4 (de) * 2009-11-27 2013-06-05 Basf Plant Science Co Gmbh Chimäre endonukleasen und ihre verwendung
CN102770539B (zh) * 2009-12-10 2016-08-03 明尼苏达大学董事会 Tal效应子介导的dna修饰
WO2011104382A1 (en) * 2010-02-26 2011-09-01 Cellectis Use of endonucleases for inserting transgenes into safe harbor loci
JP2013544082A (ja) * 2010-10-27 2013-12-12 セレクティス 二本鎖破断−誘導変異誘発の効率を増大させる方法
CA2913872C (en) * 2013-05-31 2022-01-18 Cellectis A laglidadg homing endonuclease cleaving the t-cell receptor alpha gene and uses thereof

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8673557B2 (en) * 2011-02-28 2014-03-18 Seattle Children's Research Institute Coupling endonucleases with end-processing enzymes drives high efficiency gene disruption

Non-Patent Citations (5)

* Cited by examiner, † Cited by third party
Title
Christian et al. (2010) Genetics, Vol. 186, 757-761 *
Li et al. (2011) Nucleic Acids Res., Vol. 39(1), 359-372. *
Looney et al. (1989) Gene, Vol. 80, 193-208 *
Mazur et al. (1999) J. Bio. Chem., Vol. 274, 19655-19660 *
West et al. (1989) J. Biol. Chem., Vol. 264, 10343-10346 *

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20150203871A1 (en) * 2012-06-05 2015-07-23 Cellectis Transcription Activator-Like Effector (TALE) Fusion Protein
US10815500B2 (en) * 2012-06-05 2020-10-27 Cellectis Transcription activator-like effector (TALE) fusion protein
US9834791B2 (en) 2013-11-07 2017-12-05 Editas Medicine, Inc. CRISPR-related methods and compositions with governing gRNAS
US10190137B2 (en) 2013-11-07 2019-01-29 Editas Medicine, Inc. CRISPR-related methods and compositions with governing gRNAS
US10640788B2 (en) 2013-11-07 2020-05-05 Editas Medicine, Inc. CRISPR-related methods and compositions with governing gRNAs
US11390887B2 (en) 2013-11-07 2022-07-19 Editas Medicine, Inc. CRISPR-related methods and compositions with governing gRNAS

Also Published As

Publication number Publication date
US11136566B2 (en) 2021-10-05
CA2832534C (en) 2022-01-04
US20190316100A1 (en) 2019-10-17
EP3320910A1 (de) 2018-05-16
WO2012138939A1 (en) 2012-10-11
CN103608027B (zh) 2016-01-20
AU2017248447A1 (en) 2017-11-02
IL228747B (en) 2020-04-30
DK2694091T3 (da) 2019-06-03
JP5996630B2 (ja) 2016-09-21
CA3111953C (en) 2023-10-24
KR101982360B1 (ko) 2019-05-24
EP2694091B1 (de) 2019-03-13
AU2012240110A1 (en) 2013-10-31
AU2017248447B2 (en) 2019-01-24
JP2017018125A (ja) 2017-01-26
US20130117869A1 (en) 2013-05-09
HK1253952A1 (zh) 2019-07-05
EP2694089B1 (de) 2024-06-05
IL228747A0 (en) 2013-12-31
EP2694091A2 (de) 2014-02-12
WO2012138927A2 (en) 2012-10-11
ES2728436T3 (es) 2019-10-24
US11198856B2 (en) 2021-12-14
WO2012138927A3 (en) 2013-04-25
US9315788B2 (en) 2016-04-19
US20220010292A1 (en) 2022-01-13
JP6839691B2 (ja) 2021-03-10
CA2832534A1 (en) 2012-10-11
EP2694089A1 (de) 2014-02-12
JP2014511698A (ja) 2014-05-19
AU2012240110B2 (en) 2017-08-03
CN103608027A (zh) 2014-02-26
US20160298098A1 (en) 2016-10-13
JP2019010119A (ja) 2019-01-24
JP6695239B2 (ja) 2020-05-20
CA3111953A1 (en) 2012-10-11
SG194115A1 (en) 2013-11-29
KR20140050602A (ko) 2014-04-29
SG10201602651YA (en) 2016-05-30

Similar Documents

Publication Publication Date Title
US11136566B2 (en) Tale-protein scaffolds and uses thereof
US11220683B2 (en) Method to overcome DNA chemical modifications sensitivity of engineered TALE DNA binding domains
US10815500B2 (en) Transcription activator-like effector (TALE) fusion protein
US20130337454A1 (en) Method for increasing the efficiency of double-strand break-induced mutagenesis
US11778993B2 (en) Repeat variable diresidues for targeting nucleotides
WO2012138901A1 (en) Method for enhancing rare-cutting endonuclease efficiency and uses thereof

Legal Events

Date Code Title Description
AS Assignment

Owner name: CELLECTIS, S.A., FRANCE

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:DUCHATEAU, PHILIPPE;JUILLERAT, ALEXANDRE;VALTON, JULIEN;AND OTHERS;SIGNING DATES FROM 20131030 TO 20131118;REEL/FRAME:031801/0952

STCB Information on status: application discontinuation

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