WO2017062754A1 - Compositions and methods for enhancing crispr activity by polq inhibition - Google Patents

Compositions and methods for enhancing crispr activity by polq inhibition Download PDF

Info

Publication number
WO2017062754A1
WO2017062754A1 PCT/US2016/055967 US2016055967W WO2017062754A1 WO 2017062754 A1 WO2017062754 A1 WO 2017062754A1 US 2016055967 W US2016055967 W US 2016055967W WO 2017062754 A1 WO2017062754 A1 WO 2017062754A1
Authority
WO
WIPO (PCT)
Prior art keywords
polq
cas9
cells
cell
kit
Prior art date
Application number
PCT/US2016/055967
Other languages
French (fr)
Inventor
Agnel SFEIR
Pedro MATEOS-GOMEZ
Original Assignee
New York University
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 New York University filed Critical New York University
Priority to US15/766,256 priority Critical patent/US10760081B2/en
Publication of WO2017062754A1 publication Critical patent/WO2017062754A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/113Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing
    • C12N15/1137Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing against enzymes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7088Compounds having three or more nucleosides or nucleotides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/43Enzymes; Proenzymes; Derivatives thereof
    • A61K38/45Transferases (2)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
    • 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
    • C12N15/102Mutagenizing nucleic acids
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/87Introduction of foreign genetic material using processes not otherwise provided for, e.g. co-transformation
    • C12N15/90Stable introduction of foreign DNA into chromosome
    • C12N15/902Stable introduction of foreign DNA into chromosome using homologous recombination
    • C12N15/907Stable introduction of foreign DNA into chromosome using homologous recombination in mammalian cells
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N9/00Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
    • C12N9/14Hydrolases (3)
    • C12N9/16Hydrolases (3) acting on ester bonds (3.1)
    • C12N9/22Ribonucleases RNAses, DNAses
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12YENZYMES
    • C12Y207/00Transferases transferring phosphorus-containing groups (2.7)
    • C12Y207/07Nucleotidyltransferases (2.7.7)
    • C12Y207/07007DNA-directed DNA polymerase (2.7.7.7), i.e. DNA replicase
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/10Type of nucleic acid
    • C12N2310/20Type of nucleic acid involving clustered regularly interspaced short palindromic repeats [CRISPRs]
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2510/00Genetically modified 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
    • C12N2800/00Nucleic acids vectors
    • C12N2800/80Vectors containing sites for inducing double-stranded breaks, e.g. meganuclease restriction sites

Definitions

  • the present disclosure relates generally to enhancing the efficiency of clustered regularly interspaced short palindromic repeats (CRISPR)-based editing, and more particularly to enhancing the activity of Cas9 in mammalian cells by inhibiting POLQ, an alternative nonhomologous end joining (HEJ) factor, during Cas9 editing.
  • CRISPR clustered regularly interspaced short palindromic repeats
  • HEJ alternative nonhomologous end joining
  • CRISPR-based editing of DNA including editing of the genome of mammalian cells, has gained widespread notoriety for its potentially significant power to engineer genomes for a multitude of purposes.
  • compositions and methods to improve CRISPR-based genome editing The present disclosure meets this need.
  • Embodiments include performing Cas9 DNA editing in cells in which pol ⁇ ("POLQ") enzyme and/or its function is reduced and/or eliminated.
  • POLQ pol ⁇
  • the disclosure is applicable to Cas9 nuclease and Cas9 nickase CRISPR editing.
  • the disclosure comprises increasing homozygous targeting, such as when using Cas9 nickase, and increasing both homozygous and heterozygous targeting, such as when Cas9 nuclease is used.
  • Methods of the disclosure can be performed using cells that have been engineered to express a Cas9, and one or more CRISPR guide RNAs (gRNA) directed to a target DNA sequence in the cell(s).
  • the cells can be engineered to express the Cas9 and gRNAs, as well as other polynucleotides/proteins using a variety of methods that are known in the art.
  • the disclosure comprises introducing into cells a polynucleotide (a mutation template) for modifying a target DNA sequence via CRISPR editing.
  • the cells are engineered to express Cas9 and the gRNA and/or the mutation template from a single expression vector, or by distinct expression vectors.
  • POLQ enzyme and/or its function is reduced and/or eliminated in the cells where CRISPR editing is performed. This can be achieved by
  • the agent capable of inhibiting POLQ expression comprises a polynucleotide directed to a polynucleotide in the cell that encodes POLQ (a POLQ targeting polynucleotide).
  • the POLQ targeting polynucleotide is selected from the group consisting of an antisense oligonucleotide, an siRNA, an shRNA, a polynucleotide encoding an shRNA, or a ribozyme, and combinations thereof.
  • the disclosure includes allowing expression of the Cas9 such that the target DNA sequence is modified, wherein the modified DNA sequence comprises an insertion of at least a segment of the mutation template into the target DNA sequence. Detectable markers can also be inserted.
  • kits for use in methods of the invention can comprise a polynucleotide encoding a Cas9 and an agent for use in inhibiting POLQ.
  • kits for use in methods of the invention can comprise a polynucleotide encoding a Cas9 and an agent for use in inhibiting POLQ.
  • the disclosure provides a kit comprising an expression vector configured for expressing a mutation template for use in CRISPR chromosome editing, and further comprises an agent for use in inhibiting POLQ.
  • CCE embryonic stem cells CCE a mES cell line derived from 129/Sv mouse strain; Robertson E, Bradley A, Kuehn M, Evans M, Nature. 1986 Oct 2-8; 323(6087):445-8.
  • Three independent population of CCE mES cells, with distinct ZsGreen intensity were isolated (highlighted in 1, 2 and 3 circles).
  • Group 1 depicts non-targeted cells
  • Group 2 represents cells carrying heterozygous Sox2 cells and in group 3 both alleles of Sox 2 were targeted.
  • the control gRNAs were designed to target two independent loci in the cells (H3F3B and Rosa26, 5'-CCCGTATCCGGCGAGCCAAC-3' (SEQ ID NO:8) and 5 ' -ACTCC AGTCTTTCT AGAAGA-3 ' (SEQ ID NO: 9), respectively) to which, the donor plasmid has no homology.
  • ID) Left depicts results of F ACS analysis for ZsGreen positive mES cells (POLQ WT and null, derived from 6.Cg-Polq tmlJc /J mouse) treated with the indicated Cas9 nuclease and the donor plasmid.
  • the Cas9 plasmid also encodes the gRNA that targets Hsp90abl .
  • Figure 2 Representative sequences used to illustrate embodiments of this disclosure. Boxes indicate segments of the sequences as labeled. The sequence includes the region within the sox2 gene that was subject to cleavage by Cas9- nuclease and by Cas9 nickase (D10A). The lower part shows the template used for the genome editing.
  • the Guide RNA for Sox2 is SEQ ID NO: 1 (provided as a cDNA sequence).
  • the Sox2 amino acid sequence is SEQ ID NO:2.
  • the DNA sequence of the Template plasmid for Sox2 is SEQ ID NO:3; the amino acid sequence for the Template plasmid for Sox2 is SEQ ID NO:4. Dots signify a stop codon and are present in 3' untranslated sequences.
  • Figure 3 Representative sequences used to illustrate embodiments of this disclosure; the DNA sequence is SEQ ID NO:5. Boxes indicate segments of the sequences as labeled and show primer sequences that were used to perform genotyping PCR and confirm whether edited Sox2 cells were homozygous or heterozygous.
  • FIG. 4 Representative sequences used to illustrate embodiments of this disclosure. Boxes indicate segments of the sequences as labeled. The sequence contains the sequence that was subject to cleavage by Cas9- nuclease and by Cas9 nickase (D10A) in hsp90abl.
  • the Guide RNAs for Hsp90abl sequence is SEQ ID NO:6.
  • the lower part shows the sequence of the donor template used for the genome editing.
  • the DNA sequence for the Template for Hsp90abl is SEQ ID NO:7. DESCRIPTION
  • the present disclosure includes all DNA sequences described herein, their complementary sequences, and the RNA equivalents of the DNA sequences, wherein each T is replaced by a U.
  • compositions and methods for enhancing the present disclosure provides compositions and methods for enhancing the present disclosure
  • the present disclosure comprises improving Cas9 mediated DNA editing in mammalian cells by concurrent inhibition of POLQ.
  • the disclosure comprises use of Cas9 nickase and/or Cas9 nuclease.
  • the Cas9 nickase is a mutated version of a wild-type Cas9 nuclease.
  • the nickase generates a single-stranded break (i.e., a nick) at a location based on a guide RNA as further described herein, while wild type Cas9 nuclease generates a blunt double stranded DNA break.
  • the CRISPR/Cas enzyme is the Cas9 enzyme having the amino acid sequence encoded by Streptococcus pyogenes Cas9.
  • improving Cas9 editing comprises increasing the number of chromosomes and/or the number of cells in which a chromosome is edited by the Cas9 relative to a control.
  • the control comprises an amount of Cas9 editing without concurrent suppression of POLQ.
  • control can comprise the amount of a DNA segment integrated into a chromosome using Cas9 editing as described herein without concurrent suppression of POLQ.
  • the disclosure comprises using Cas9 nickase in CRISPR-mediated DNA editing in conjunction with POLQ suppression to increase homozygous editing relative to a suitable control.
  • the disclosure comprises using Cas9 nuclease in CRISPR-mediated DNA editing in conjunction with POLQ suppression to increase both homozygous and heterozygous editing relative to a suitable control.
  • comparisons of editing by Cas9 nickase to editing by Cas9 nuclease in conjunction with POLQ suppression can be made to determine differences in heterozygous and homozygous editing.
  • the disclosure includes methods that pertain to populations of cells, whereby the percentage of cells in the population that comprise a homozygous or a heterozygous mutation is increased. Aspects of this and other embodiments of the disclosure can be related to any suitable reference, such as control value obtained directly or derived from editing performed without POLQ inhibition/suppression, i.e., editing performed in the presence of wild type amounts of POLQ.
  • methods of the disclosure comprise allowing expression of the Cas9 in the presence of a suitable gRNA such that a target DNA sequence is modified by Cas9 in a mammalian cell in which POLQ is inhibited.
  • the targeted DNA can be any desired DNA sequence, provided it has the requisite sequence to be subject to cleavage by Cas9.
  • targeting a particular DNA sequence is facilitated at least in part by selection of a suitable guide RNA (gRNA) sequence.
  • gRNA guide RNA
  • Cas9 "targeting" means directing the Cas9 to a particular DNA sequence, i.e., a target locus, in a chromosome, and can thereby introduce an intended mutation that is present in a mutation template as further described below.
  • a targeting RNA comprising a gRNA sequence
  • the sequence of the targeting RNA has a segment that is the same as or complementarity to any CRISPR site in the target gene or other chromosomal region.
  • a suitable target sequence generally comprises a specific sequence on its 3' end referred to as a protospacer adjacent motif or "PAM".
  • PAM protospacer adjacent motif
  • the target sequence conforms to the well-known N12-20NGG motif, wherein the NGG is the PAM sequence.
  • the gRNA will comprise a segment that is from 12-20 nucleotides in length which is the same as or complementary to a DNA target sequence (a spacer) in the target DNA sequence.
  • the 12-20 nucleotides directed to the spacer sequence will be present in the gRNA, regardless of whether the gRNA is comprised within, for example, a crRNA.
  • a separate trans- activating crRNA can be used to assist in maturation of an RNA that is targeted to a particular gene.
  • CRISPR system Introduction of a CRISPR system according to this disclosure into cells will result in binding of a targeting RNA/Cas9 complex to the target sequence so that the Cas9 can act on and cleave the target, and thus result in an alteration selected from a variety of
  • the cleaved sequence can be repaired by end joining DNA repair, which can result in either insertions or deletions at the break site, or by using a repair template to introduce mutations, or to introduce an inserted sequence.
  • the disclosure includes administering an inhibitor of DNA-dependent protein kinase (DNA-PK) such that the classical NHEJ pathway of double DSB repair is inhibited.
  • DNA-PK inhibitors are known in the art and are commercially available; in a non-limiting example NU7441 is used.
  • a polynucleotide comprising or consisting of a gRNA can be introduced to a mammalian cell encoded by the same expression vector that also encodes the Cas9 enzyme, or it can be expressed from a distinct vector, or it can be introduced as RNA.
  • any suitable expression vector can be used, and suitable expression vectors for introducing Cas9 encoding sequences, as well as gRNA sequences, as well as donor templates, are publicly available.
  • the expression vector is a plasmid, or is a modified viral vector.
  • the polynucleotide encoding the CRISPR system is transiently present in a cell.
  • the polynucleotide encoding the CRISPR system is stably present in the cell, and/or may be integrated into a chromosome.
  • Expression vectors can be introduced into cells using any suitable technique and delivery system, many of which are known in the art and include but are not limited to electroporation, lipid-based transfection systems, standard plasmid transformation approaches, such as by using competent cells, phage or viral transduction, microinjection, including direct injection of a CRISPR vector or by introducing RNA itself.
  • expression systems/plasmids for use in CRISPR editing that include Cas9 nickase or Cas9 nuclease are publicly available from, for example, ADDGENE (Cambridge, MA, USA).
  • an agent that inhibits POLQ, a Cas9 and a suitable gRNA targeting a desired DNA sequence are introduced into an individual for a prophylactic/and or therapeutic purpose.
  • the disclosure includes pharmaceutical formulations comprising the CRISPR system reagents and/or POLQ inhibitors as described herein, and methods of administering them.
  • a Cas9, a gRNA, a mutation template, and combinations thereof can be introduced into cells which already have reduced or eliminated POLQ expression and/or function.
  • a donor template for use in modifying a chromosome at a target locus
  • a donor template can also be considered a DNA mutation template.
  • the DNA mutation template can comprise a DNA segment having any nucleotide length and homology with either DNA strand of the double-stranded chromosome segment comprising a target locus, so long as the length and sequence identity are adequate to introduce the intended mutation into the target locus via functioning of the CRISPR-Cas system described herein.
  • the DNA mutation template is a single- stranded oligo DNA nucleotide (ssODN).
  • the DNA mutation template is a double-stranded (ds) template.
  • the DNA mutation template is provided as an episomal element, such as a plasmid or PCR product.
  • the DNA mutation template is a ssODN having a length of from 20-2000 nucleotides (or base pairs if double-stranded), and in either case includes all integers from and including 20-2000, and all ranges there between.
  • the homology of at least a segment of the DNA mutation template to the segment of the chromosome comprising the target locus comprises from 90%- 100% identity with a contiguous segment of the chromosome, inclusive, and including all integers and ranges between 90%- 100%.
  • the mutation is flanked by homologous sequence segments with similarity above at least 80%.
  • the DNA mutation template can comprise regions that have less than 80%, or even no homology to the segment of the chromosome that comprises the target locus, and such regions can even be the majority of the template, so long as there is adequate homology between the DNA mutation template and the chromosome to participate in homologous recombination of a portion of the chromosome and the template that contains the mutation to be introduced.
  • the degree of identity between homologous segments can be calculated exclusive of the mutation to be introduced.
  • the DNA mutation template can comprise the intended mutation to be inserted into the chromosome.
  • the intended mutation can comprise or consist of a single nucleotide, or more than one nucleotide, and can be present in a protein-coding or non-protein coding portion of a chromosome, or may be present in a regulatory control element, including but not necessarily limited to a promoter or enhancer element, a splice junction, etc.
  • the intended mutation may also be present in a DNA sequence that encodes for transcription of an RNA that is not an mRNA, such as a microRNA, tRNA, rRNA, etc.
  • the intended mutation may introduce or remove a nucleotide that undergoes an epigenetic modification, such as a cytosine that is known to be methylated, or de-methylated during any of a variety of biological processes.
  • an epigenetic modification such as a cytosine that is known to be methylated, or de-methylated during any of a variety of biological processes.
  • the intended mutation can comprise or consist of a point mutation, an insertion, or a deletion, and will be dictated by the sequence of the mutation template and gRNA (so long as the other
  • the intended mutation comprises an insertion or deletion.
  • the mutation comprises up to 20% of the nucleotides in the DNA mutation template.
  • the POLQ can be inhibited using any suitable approach, including but not limited to small molecule inhibitors, or by using polynucleotide targeting agents that will affect POLQ expression. Further, at least one anti-POLQ monoclonal antibody is commercially available, from, for example, SIGMA-ALDRICH.
  • inhibiting POLQ comprises either inhibiting specifically or selectively inhibiting its enzymatic activity, or disrupting its expression, or a combination thereof.
  • inhibiting POLQ comprises introducing into the cells a polynucleotide that can inhibit translation of POLQ mRNA, and/or can participate in and/or facilitate RNAi-mediated reduction of POLQ mRNA.
  • an antisense polynucleotide is used to inhibit translation of POLQ mRNA.
  • Antisense nucleic acids can be DNA or RNA molecules that are complementary to at least a portion of the POLQ mRNA. In embodiments, oligomers of about fifteen nucleotides, and/or those that hybridize to the AUG initiation codon may be particularly efficient.
  • the polynucleotides described herein for use in targeting POLQ mRNA can in certain embodiments be modified, such as to be resistant to nucleases.
  • RNAi-mediated reduction in POLQ mRNA includes RNAi-mediated reduction in POLQ mRNA.
  • RNAi-based inhibition can be achieved using any suitable RNA polynucleotide that is targeted to POLQ mRNA.
  • a single stranded or double stranded RNA, wherein at least one strand is complementary to the POLQ mRNA can be introduced into the cell to promote RNAi -based degradation of POLQ mRNA.
  • microRNA (miRNA) targeted to the POLQ mRNA can be used.
  • a ribozyme that can specifically cleave POLQ mRNA can be used.
  • small interfering RNA siRNA
  • siRNA (or ribozymes) can be introduced directly, for example, as a double stranded siRNA complex, or by using a modified expression vector, such as a lentiviral vector, to produce an shRNA.
  • shRNAs adopt a typical hairpin secondary structure that contains a paired sense and antisense portion, and a short loop sequence between the paired sense and antisense portions.
  • shRNA is delivered to the cytoplasm where it is processed by DICER into siRNAs.
  • siRNA is recognized by RNA-induced silencing complex (RISC), and once incorporated into RISC, siRNAs facilitate cleavage and degradation of targeted mRNA.
  • RISC RNA-induced silencing complex
  • an shRNA polynucleotide used to suppress POLQ expression can comprise or consist of between 45-100 nucleotides, inclusive, and including all integers between 45 and 100.
  • the portion of the shRNA that is complementary to the POLQ mRNA can be from 21-29 nucleotides, inclusive, and including all integers between 21 and 29.
  • any suitable vector including but not necessarily limited to lentiviral vectors, can be made and used according to standard techniques, given the benefit of the present disclosure.
  • suitable vectors expressing shRNAs targeted to many human mRNAs are commercially available.
  • shRNA constructs provided in retroviral vectors are available from ORIGENE, (Rockville, MD, USA), and Applied Biological Materials (ABM) Inc., (Richmond, B.C., Canada), and can be included in methods and kits of this disclosure.
  • RNAi-mediated approaches for decreasing POLQ have already been demonstrated and can be adapted for use in embodiments of the present invention, given the benefit of this disclosure.
  • Mateos- Gomez et al. (Mammalian polymerase ⁇ promotes alternative NHEJ and suppresses
  • shRNA targeting POLQ sequences can comprise thesh Polq-1 (mouse sequence): 5 '-CGGCGGAGTATGAGAACTATT-3 ' (SEQ ID NO: 10); sh Polq-2 (mouse sequence): 5'- CC AGGAATC AAAGACGAC AAT-3 ' (SEQ ID NO: 11); sh Polq-3 (mouse sequence): 5'- CCTGGCTGAATGCTGAACTTT-3 ' (SEQ ID NO: 12); and sh POLQ (human sequence): 5'- CGGGCCTCTTT AGAT ATAAAT-3 ' (SEQ ID NO: 13) that are demonstrated in Mateos-Gomez et al. It will be recognized that the shRNA itself comprises the RNA equivalent of the DNA sequences. It will also be recognized that minor variations in these sequences that do not reduce shRNA targeting of the mRNA can be made and are included within the scope of this disclosure.
  • custom siRNAs or shRNA can be obtained from publicly available sources.
  • Lentiviruses and other suitable retroviral constructs are capable of stably and permanently infecting target cells, such as by integrating into a chromosome.
  • the disclosure includes disrupting the POLQ gene such that
  • the POLQ gene can be disrupted by targeted mutagenesis.
  • the present disclosure provides for replacement of the POLQ gene with a sequence encoding a detectable marker, such as a fluorescent protein, or integrating such a sequence into the POLQ gene, thereby disrupting it, or integrating such a sequence elsewhere in the genome of the mammalian cells. By replacing POLQ or integrating a sequence encoding a detectable protein into it the disclosure provides for marking cells that do not express POLQ.
  • the disclosure includes inhibiting POLQ prior to introducing the Cas9 and gRNA sequence, or concurrently introducing a POLQ inhibitor to the cell with the Cas9-nickase and gRNA sequence.
  • the DNA that is selected to by modified using compositions and methods of this disclosure can be any suitable target DNA sequence.
  • modification of the DNA confers a change in phenotype of the cell, such as a change in morphology, growth rate, expression of a detectable or selectable marker, or the modification is lethal to the cell.
  • the DNA sequence that is modified is present on a chromosome, or is present on an extra-chromosomal element, including but not limited to a plasmid.
  • the modification can comprise insertion of a detectable marker into a DNA element in a cell.
  • the disclosure includes modification of DNA in one or more cells, and can further comprise identification of one or more cells in which the
  • the mammalian cells can be any mammalian cells, including but not limited to stem cells, such as totipotent, pluripotent, and multipotent cells.
  • the cells are hematopoietic cells.
  • the cells are embryonic stem cells, or adult stem cells.
  • the cells are epidermal stem cells or epithelial stem cells, or neuronal precursor cells.
  • the cells are differentiated cells when the intended mutation is introduced.
  • the cells are mammalian cells.
  • the cells are human, or are non-human animal cells.
  • the disclosure includes progeny of any such cells, and non-human mammals comprising a cell or progeny of a cell made as described herein.
  • the cells modified according to this disclosure are not worm cells, including but not limited to cells of any nematode. In an embodiment, the cells modified according to this disclosure are not C. elegans cells.
  • the disclosure includes obtaining cells from an individual, modifying the cells ex vivo as described herein, and reintroducing the cells and/or their progeny into the individual for prophylaxis and/or therapy of a condition, disease or disorder, or to treat an injury, trauma or anatomical defect.
  • the cells modified ex vivo as described herein are used autologously.
  • the cells are provided as cell lines.
  • the cells are engineered to produce a protein or other compound; the cells themselves or the protein or compound they produce in certain implementations may be useful for prophylactic or therapeutic applications.
  • the mutation introduced into cells according to this disclosure is a homozygous dominant or homozygous recessive, or heterozygous dominant or heterozygous recessive mutation, that is correlated with a phenotype or condition, and is thus useful for modeling such phenotype or condition.
  • the disclosure includes identifying and separating cells that are heterozygous or are homozygous for a mutation introduced according to this disclosure. Such cells can be separated from one another, and from cells into which no mutation has been introduced. Identification and separation can be achieved using any suitable approach, including sequencing individual cells and expanding clones using routine approaches, or by including and detecting detectable markers, and/or other indicia of the mutation. In embodiments, the disclosure includes expanding separated cells to obtain a clonal population of the cells, and can further comprise promoting and/or allowing the cells to differentiate. Thus, the disclosure includes fully or partially differentiated cells that are homozygous or
  • compositions that include a cellular component, wherein the cellular component comprises or consists of cells modified according to this disclosure.
  • the disclosure include a kit comprising a polynucleotides encoding a Cas9, and may further comprise an agent capable of inhibiting POLQ, or a polynucleotide that encodes a polynucleotide capable of disrupting POLQ expression.
  • the polynucleotide capable of disrupting POLQ expression is a commercially available viral vector that encodes an shRNA targeted to POLQ mRNA.
  • the kit can further comprised a guide RNA or a polynucleotide encoding the guide RNA, or a vector adapted to insert and express a polynucleotide comprising a suitable gRNA segment.
  • the kit can further comprised a polynucleotide or vector that is adaptable for expressing a mutation template.
  • This Example provides: first, a demonstration of inserting into CCE mouse embryonic stem cells (CCE mESCs) a detectable marker (ZsGreen - a green fluorescent protein) at the end of the sox2 gene. A P2A sequence (57 nucleotides) followed by ZsGreen (696 nt) gene was inserted preceding the STOP codon of sox2. Second, a demonstration of inserting into mouse embryonic stem cells (mESCs) and mouse embryonic fibroblast (MEFs) a detectable marker (ZsGreen - a green fluorescent protein) at the end of the hsp90abl gene.
  • CCE mouse embryonic stem cells CCE mouse embryonic stem cells
  • ZsGreen - a green fluorescent protein a detectable marker at the end of the sox2 gene.
  • Hsp90abl using pSL301 as a vector, to which the following three segments were introduced.
  • 1- The last 83 lnt or 700nt of Sox2 or Hsp90abl genes without the STOP codon (used as a 5' homology arms in the donor plasmid), fused to 2- P2A-ZsGreen and followed by 3- the first 801 nt of the 3'UTR of Sox2 or the 700nt after the STOP codon of Hsp90abl respectively (used as a 3' homology arm in the donor plasmid).
  • Cas9 nuclease and nickase D10A were expressed from publicly available expression vectors.
  • pX330-U6-Chimeric_BB-CBh-hSpCas9 (Addgene plasmid #42230)
  • pX335-U6-Chimeric_BB-CBh-hSpCas9n(D10A) (Addgene Plasmid #42335)
  • a Puromycin-cassette was inserted for selection, which serves to enrich for ES cells expressing the Cas9.
  • gRNA was expressed (5'-CAGCCCTCACATGTGCGACA-3' (SEQ ID NO: 14) for Sox2 and 5' - CGATGAGGATGCCTCGCGC A-3 ' (SEQ ID NO: 15) for Hsp90abl) and two gRNAs with the nickase version (5 ' -TT AATGGCCGTGCCGGGC AC-3 ' (SEQ ID NO: 16; 5'-
  • GGGGAT ACT AT AC AGAGGGC-3 ' (SEQ ID NO: 18) were used to generate ends with 3' overhangs, and the couple 5' - CCCTCTGTATAGTATCCCCG-3 ' (SEQ ID NO: 19) and 5 '- ATCCACCTCTTCCATGCGCG-3' (SEQ ID NO:20) were used to generate ends with 5' overhangs).
  • both gRNAs were cloned in tandem U6 promoter-gRNA-l-U6 promoter-gRNA-2 using a Kpnl restriction site located in the px335 plasmid downstream of the gRNA cloning site.
  • mice cells were CCE mES cells, mES cells and MEFs (all three WT and
  • Lipofectamine® 3000 was obtained from Life Technologies.
  • the culture media for CCE mESCs contained 500ml of DMEM and 90ml of ESC qualified FBS, supplemented according to manufacturer with non-essential amino acids, L-glutamine, penicillin/streptomycin, B-mercaptoethanol and LIF protein.
  • the culture media for mESCs contained 500ml of DMEM and 90ml of ESC qualified FBS, supplemented according to manufacturer with non-essential amino acids, L-glutamine, penicillin/streptomycin, B-mercaptoethanol, LIF protein,
  • the culture media for MEFs contained 500ml of DMEM and 50ml of FBS, supplemented according to
  • DNA-PK inhibitor was NU7441 (luM) was included to block classical- HEJ.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Genetics & Genomics (AREA)
  • Engineering & Computer Science (AREA)
  • Organic Chemistry (AREA)
  • Molecular Biology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Zoology (AREA)
  • Wood Science & Technology (AREA)
  • General Health & Medical Sciences (AREA)
  • Biomedical Technology (AREA)
  • Biotechnology (AREA)
  • General Engineering & Computer Science (AREA)
  • Biochemistry (AREA)
  • Medicinal Chemistry (AREA)
  • Microbiology (AREA)
  • Biophysics (AREA)
  • Plant Pathology (AREA)
  • Physics & Mathematics (AREA)
  • Public Health (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Veterinary Medicine (AREA)
  • Animal Behavior & Ethology (AREA)
  • Epidemiology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Cell Biology (AREA)
  • Crystallography & Structural Chemistry (AREA)
  • Virology (AREA)
  • Mycology (AREA)
  • Immunology (AREA)
  • Toxicology (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)

Abstract

Provided are approaches to improving CRISPR-based DNA editing by performing the editing in cells in which pol Θ ("POLQ") enzyme and/or its function is reduced. Approaches are provided that are applicable to Cas9 nuclease and Cas9 nickase CRISPR editing. Also provided are kits that can contain a polynucleotide encoding a Cas9 and an agent for use in inhibiting POLQ, and/or or an expression vector configured for expressing a mutation template for use in CRISPR chromosome editing and an agent for use in inhibiting POLQ.

Description

COMPOSITIONS AND METHODS FOR ENHANCING CRISPR ACTIVITY BY POLQ
INHIBITION
CROSS-REFERENCE TO RELATED APPLICATIONS
[0001] This application claims priority to U.S. provisional patent application no.
62/238,192, filed October 7, 2015.
FIELD
[0002] The present disclosure relates generally to enhancing the efficiency of clustered regularly interspaced short palindromic repeats (CRISPR)-based editing, and more particularly to enhancing the activity of Cas9 in mammalian cells by inhibiting POLQ, an alternative nonhomologous end joining ( HEJ) factor, during Cas9 editing.
BACKGROUND
[0003] CRISPR-based editing of DNA, including editing of the genome of mammalian cells, has gained widespread notoriety for its potentially significant power to engineer genomes for a multitude of purposes. However, there remains an ongoing need for compositions and methods to improve CRISPR-based genome editing. The present disclosure meets this need.
SUMMARY
[0004] The present disclosure provides approaches to improving CRISPR-based DNA editing. Embodiments include performing Cas9 DNA editing in cells in which pol Θ ("POLQ") enzyme and/or its function is reduced and/or eliminated. The disclosure is applicable to Cas9 nuclease and Cas9 nickase CRISPR editing. In certain non-limiting implementations the disclosure comprises increasing homozygous targeting, such as when using Cas9 nickase, and increasing both homozygous and heterozygous targeting, such as when Cas9 nuclease is used.
[0005] Methods of the disclosure can be performed using cells that have been engineered to express a Cas9, and one or more CRISPR guide RNAs (gRNA) directed to a target DNA sequence in the cell(s). The cells can be engineered to express the Cas9 and gRNAs, as well as other polynucleotides/proteins using a variety of methods that are known in the art. In certain aspects the disclosure comprises introducing into cells a polynucleotide (a mutation template) for modifying a target DNA sequence via CRISPR editing. In non-limiting examples the cells are engineered to express Cas9 and the gRNA and/or the mutation template from a single expression vector, or by distinct expression vectors.
[0006] In non-limiting examples POLQ enzyme and/or its function is reduced and/or eliminated in the cells where CRISPR editing is performed. This can be achieved by
introducing into the cells an agent capable of inhibiting expression of POLQ, and/or capable of inhibiting its function. In non-limiting embodiments the agent capable of inhibiting POLQ expression comprises a polynucleotide directed to a polynucleotide in the cell that encodes POLQ (a POLQ targeting polynucleotide). In certain implementations the POLQ targeting polynucleotide is selected from the group consisting of an antisense oligonucleotide, an siRNA, an shRNA, a polynucleotide encoding an shRNA, or a ribozyme, and combinations thereof. The disclosure includes allowing expression of the Cas9 such that the target DNA sequence is modified, wherein the modified DNA sequence comprises an insertion of at least a segment of the mutation template into the target DNA sequence. Detectable markers can also be inserted.
[0007] The disclosure is applicable to any mammalian cells and at any stage of cell development. In embodiments the cells are human cells, or are non-human mammal cells. The disclosure includes mammalian cells made by the methods described herein, their progeny, and non-human mammals comprising them. Also provided are kits for use in methods of the invention. The kits can comprise a polynucleotide encoding a Cas9 and an agent for use in inhibiting POLQ. In embodiments the disclosure provides a kit comprising an expression vector configured for expressing a mutation template for use in CRISPR chromosome editing, and further comprises an agent for use in inhibiting POLQ.
DESCRIPTION OF THE FIGURES
[0008] Figure 1 A) Flow cytometry analysis to quantify ZsGreen expression in mouse
CCE embryonic stem cells (mESc) (CCE a mES cell line derived from 129/Sv mouse strain; Robertson E, Bradley A, Kuehn M, Evans M, Nature. 1986 Oct 2-8; 323(6087):445-8. Keller G, Kennedy M, Papayannopoulou T, Wiles MV, Mol Cell Biol. 1993 Jan; 13(l):473-86. Numbers represent the percentage of cells in each quadrant. Three independent population of CCE mES cells, with distinct ZsGreen intensity were isolated (highlighted in 1, 2 and 3 circles). IB)
Genotyping PCR for Sox2 on DNA extracted from the three highlighted groups of cells (1 A) in POLQ proficient (POLQ+/+ ,WT) and PolQ deficient (PolQ-/-, Null) CCE mES cells. Group 1 depicts non-targeted cells, Group 2 represents cells carrying heterozygous Sox2 cells and in group 3 both alleles of Sox 2 were targeted. 1C) Left, graph depicting results of F ACS analysis for ZsGreen positive CCE mES cells (POLQ+/+ and PolQ-/-) treated with the indicated Cas9 nuclease plasmid (also encoding for a gRNA targeting the Sox2 gene) and a donor plasmid with the sequence to be inserted at the end of the gene. DNA-PK inhibitor was used to block repair by classical NHEJ (Non-Homologous End- Joining). Right, cells treated with Cas9-nickase containing plasmid that also encodes for two Sox2 gRNAs. Three independent experiments, each performed in duplicate. The control gRNAs were designed to target two independent loci in the cells (H3F3B and Rosa26, 5'-CCCGTATCCGGCGAGCCAAC-3' (SEQ ID NO:8) and 5 ' -ACTCC AGTCTTTCT AGAAGA-3 ' (SEQ ID NO: 9), respectively) to which, the donor plasmid has no homology. ID) Left, depicts results of F ACS analysis for ZsGreen positive mES cells (POLQ WT and null, derived from 6.Cg-PolqtmlJc /J mouse) treated with the indicated Cas9 nuclease and the donor plasmid. The Cas9 plasmid also encodes the gRNA that targets Hsp90abl . Right, cells treated with a Cas9-nickase containing plasmid that encodes for two Hsp90abl gRNAs. This experiment was performed with two different Cas9-nickase plasmids, one encoding two gRNAs that generates 3' overhangs after cleavage and other that generates 5' overhangs). Three independent experiments, each performed in duplicate. IE) Depicts results of FACS analysis for ZsGreen positive MEFs cells (POLQ WT and null, derived from B6.Cg- PolqtmlJcs/J mouse) treated with the indicated Cas9 as in Figure ID. Three independent experiments, each performed in duplicate.
[0009] Figure 2. Representative sequences used to illustrate embodiments of this disclosure. Boxes indicate segments of the sequences as labeled. The sequence includes the region within the sox2 gene that was subject to cleavage by Cas9- nuclease and by Cas9 nickase (D10A). The lower part shows the template used for the genome editing. In Figure 2 the Guide RNA for Sox2 is SEQ ID NO: 1 (provided as a cDNA sequence). The Sox2 amino acid sequence is SEQ ID NO:2. The DNA sequence of the Template plasmid for Sox2 is SEQ ID NO:3; the amino acid sequence for the Template plasmid for Sox2 is SEQ ID NO:4. Dots signify a stop codon and are present in 3' untranslated sequences.
[0010] Figure 3. Representative sequences used to illustrate embodiments of this disclosure; the DNA sequence is SEQ ID NO:5. Boxes indicate segments of the sequences as labeled and show primer sequences that were used to perform genotyping PCR and confirm whether edited Sox2 cells were homozygous or heterozygous.
[0011] Figure 4. Representative sequences used to illustrate embodiments of this disclosure. Boxes indicate segments of the sequences as labeled. The sequence contains the sequence that was subject to cleavage by Cas9- nuclease and by Cas9 nickase (D10A) in hsp90abl. The Guide RNAs for Hsp90abl sequence is SEQ ID NO:6. The lower part (Template for Hsp90abl) shows the sequence of the donor template used for the genome editing. The DNA sequence for the Template for Hsp90abl is SEQ ID NO:7. DESCRIPTION
[0012] Unless defined otherwise herein, all technical and scientific terms used in this disclosure have the same meaning as commonly understood by one of ordinary skill in the art to which this disclosure pertains.
[0013] Every numerical range given throughout this specification includes its upper and lower values, as well as every narrower numerical range that falls within it, as if such narrower numerical ranges were all expressly written herein.
[0014] The present disclosure includes all DNA sequences described herein, their complementary sequences, and the RNA equivalents of the DNA sequences, wherein each T is replaced by a U.
[0015] The present disclosure provides compositions and methods for enhancing
CRISPR-mediated DNA editing facilitated by a Cas9 enzyme in mammalian cells. The disclosure is demonstrated using a non-limiting proof of principle as described further in Example 1 below which shows that it is possible to improve gene targeting efficiency in mammalian cells by combining Cas9 mediated targeting with POLQ inhibition. This is in contrast to results previously described using C. elegans as a model, where deletion of POLQ slightly decreased targeting efficiency of Cas9 nuclease (Van Schendel, Robin et al.
"Polymerase 0 Is a Key Driver of Genome Evolution and of CRISPR/Cas9-Mediated
Mutagenesis." Nature Communications 6 (2015): 7394. PMC. June 16, 2015). Instead, as demonstrated in this disclosure, in mammalian cells the Cas9 targeting was enhanced by loss of the POLQ polymerase. Specifically, it was found: 1- that homozygous targeting (represented in Figure 1C) is more enriched than heterozygous targeting when Cas9 nickase was used in CCE mESCs to insert ZsGreen at the end of sox2; 2- that homozygous and heterozygous targeting is more enriched in POLQ null mESCs and MEFs when Cas9 nuclease was used to introduce a cassette coding for ZsGreen at the 3'UTR of hsp90abl. Thus, in various embodiments, the present disclosure comprises improving Cas9 mediated DNA editing in mammalian cells by concurrent inhibition of POLQ. In embodiments, the disclosure comprises use of Cas9 nickase and/or Cas9 nuclease. It will be recognized by those skilled in the art that the Cas9 nickase is a mutated version of a wild-type Cas9 nuclease. The nickase generates a single-stranded break (i.e., a nick) at a location based on a guide RNA as further described herein, while wild type Cas9 nuclease generates a blunt double stranded DNA break. In certain aspects, when the nickase is used with to gRNAs that introduce the nick close to one another in the DNA a double stranded break with overhangs can be generated. In embodiments, the CRISPR/Cas enzyme is the Cas9 enzyme having the amino acid sequence encoded by Streptococcus pyogenes Cas9. [0016] In certain embodiments, improving Cas9 editing comprises increasing the number of chromosomes and/or the number of cells in which a chromosome is edited by the Cas9 relative to a control. In an embodiment, the control comprises an amount of Cas9 editing without concurrent suppression of POLQ. In an embodiment the control can comprise the amount of a DNA segment integrated into a chromosome using Cas9 editing as described herein without concurrent suppression of POLQ. In embodiments, the disclosure comprises using Cas9 nickase in CRISPR-mediated DNA editing in conjunction with POLQ suppression to increase homozygous editing relative to a suitable control. In embodiments, the disclosure comprises using Cas9 nuclease in CRISPR-mediated DNA editing in conjunction with POLQ suppression to increase both homozygous and heterozygous editing relative to a suitable control. In certain implementations comparisons of editing by Cas9 nickase to editing by Cas9 nuclease in conjunction with POLQ suppression can be made to determine differences in heterozygous and homozygous editing. The disclosure includes methods that pertain to populations of cells, whereby the percentage of cells in the population that comprise a homozygous or a heterozygous mutation is increased. Aspects of this and other embodiments of the disclosure can be related to any suitable reference, such as control value obtained directly or derived from editing performed without POLQ inhibition/suppression, i.e., editing performed in the presence of wild type amounts of POLQ.
[0017] In embodiments, methods of the disclosure comprise allowing expression of the Cas9 in the presence of a suitable gRNA such that a target DNA sequence is modified by Cas9 in a mammalian cell in which POLQ is inhibited. The targeted DNA can be any desired DNA sequence, provided it has the requisite sequence to be subject to cleavage by Cas9. In embodiments, targeting a particular DNA sequence is facilitated at least in part by selection of a suitable guide RNA (gRNA) sequence. Thus, as used herein Cas9 "targeting" means directing the Cas9 to a particular DNA sequence, i.e., a target locus, in a chromosome, and can thereby introduce an intended mutation that is present in a mutation template as further described below. The skilled artisan will appreciate a multitude of ways that are known in the art to design and introduce a suitable gRNA sequence into any mammalian cell or population of mammalian cells. Thus, in embodiments, a targeting RNA comprising a gRNA sequence is used. In general, the sequence of the targeting RNA has a segment that is the same as or complementarity to any CRISPR site in the target gene or other chromosomal region. In this regard, a suitable target sequence generally comprises a specific sequence on its 3' end referred to as a protospacer adjacent motif or "PAM". In an embodiment the target sequence conforms to the well-known N12-20NGG motif, wherein the NGG is the PAM sequence. Thus, in embodiments, the gRNA will comprise a segment that is from 12-20 nucleotides in length which is the same as or complementary to a DNA target sequence (a spacer) in the target DNA sequence. The 12-20 nucleotides directed to the spacer sequence will be present in the gRNA, regardless of whether the gRNA is comprised within, for example, a crRNA. In embodiments, a separate trans- activating crRNA (tracrRNA) can be used to assist in maturation of an RNA that is targeted to a particular gene. Introduction of a CRISPR system according to this disclosure into cells will result in binding of a targeting RNA/Cas9 complex to the target sequence so that the Cas9 can act on and cleave the target, and thus result in an alteration selected from a variety of
modifications. In one embodiment, the cleaved sequence can be repaired by end joining DNA repair, which can result in either insertions or deletions at the break site, or by using a repair template to introduce mutations, or to introduce an inserted sequence. In embodiments, the disclosure includes administering an inhibitor of DNA-dependent protein kinase (DNA-PK) such that the classical NHEJ pathway of double DSB repair is inhibited. A number of DNA-PK inhibitors are known in the art and are commercially available; in a non-limiting example NU7441 is used.
[0018] In embodiments, a polynucleotide comprising or consisting of a gRNA can be introduced to a mammalian cell encoded by the same expression vector that also encodes the Cas9 enzyme, or it can be expressed from a distinct vector, or it can be introduced as RNA. The same applies for introducing polynucleotides encoding the Cas9. In more detail, any suitable expression vector can be used, and suitable expression vectors for introducing Cas9 encoding sequences, as well as gRNA sequences, as well as donor templates, are publicly available. In embodiments, the expression vector is a plasmid, or is a modified viral vector. In embodiments, the polynucleotide encoding the CRISPR system is transiently present in a cell. In
embodiments, the polynucleotide encoding the CRISPR system is stably present in the cell, and/or may be integrated into a chromosome. Expression vectors can be introduced into cells using any suitable technique and delivery system, many of which are known in the art and include but are not limited to electroporation, lipid-based transfection systems, standard plasmid transformation approaches, such as by using competent cells, phage or viral transduction, microinjection, including direct injection of a CRISPR vector or by introducing RNA itself.
Additionally, expression systems/plasmids for use in CRISPR editing that include Cas9 nickase or Cas9 nuclease are publicly available from, for example, ADDGENE (Cambridge, MA, USA).
[0019] In embodiments, an agent that inhibits POLQ, a Cas9 and a suitable gRNA targeting a desired DNA sequence are introduced into an individual for a prophylactic/and or therapeutic purpose. Thus, the disclosure includes pharmaceutical formulations comprising the CRISPR system reagents and/or POLQ inhibitors as described herein, and methods of administering them. In certain approaches a Cas9, a gRNA, a mutation template, and combinations thereof, can be introduced into cells which already have reduced or eliminated POLQ expression and/or function.
[0020] In certain approaches the disclosure comprises use of a donor template for use in modifying a chromosome at a target locus, and thus a donor template can also be considered a DNA mutation template. The DNA mutation template can comprise a DNA segment having any nucleotide length and homology with either DNA strand of the double-stranded chromosome segment comprising a target locus, so long as the length and sequence identity are adequate to introduce the intended mutation into the target locus via functioning of the CRISPR-Cas system described herein. In embodiments, the DNA mutation template is a single- stranded oligo DNA nucleotide (ssODN). In embodiments, the DNA mutation template is a double-stranded (ds) template. In embodiments, the DNA mutation template is provided as an episomal element, such as a plasmid or PCR product. In certain embodiments, the DNA mutation template is a ssODN having a length of from 20-2000 nucleotides (or base pairs if double-stranded), and in either case includes all integers from and including 20-2000, and all ranges there between. In embodiments, the homology of at least a segment of the DNA mutation template to the segment of the chromosome comprising the target locus comprises from 90%- 100% identity with a contiguous segment of the chromosome, inclusive, and including all integers and ranges between 90%- 100%. In embodiments, the mutation is flanked by homologous sequence segments with similarity above at least 80%. It will be recognized however, that the DNA mutation template can comprise regions that have less than 80%, or even no homology to the segment of the chromosome that comprises the target locus, and such regions can even be the majority of the template, so long as there is adequate homology between the DNA mutation template and the chromosome to participate in homologous recombination of a portion of the chromosome and the template that contains the mutation to be introduced. The degree of identity between homologous segments can be calculated exclusive of the mutation to be introduced. The DNA mutation template can comprise the intended mutation to be inserted into the chromosome.
[0021] The intended mutation can comprise or consist of a single nucleotide, or more than one nucleotide, and can be present in a protein-coding or non-protein coding portion of a chromosome, or may be present in a regulatory control element, including but not necessarily limited to a promoter or enhancer element, a splice junction, etc. The intended mutation may also be present in a DNA sequence that encodes for transcription of an RNA that is not an mRNA, such as a microRNA, tRNA, rRNA, etc. The intended mutation may introduce or remove a nucleotide that undergoes an epigenetic modification, such as a cytosine that is known to be methylated, or de-methylated during any of a variety of biological processes. Thus, the intended mutation can comprise or consist of a point mutation, an insertion, or a deletion, and will be dictated by the sequence of the mutation template and gRNA (so long as the other
CRISPR-related elements, such as the NGG and PAM sequences are also present in appropriate proximity to the mutation site). In embodiments, the intended mutation comprises an insertion or deletion. In embodiments, the mutation comprises up to 20% of the nucleotides in the DNA mutation template. By introducing a mutation according to this disclosure, the characteristics of the resulting cells can be altered in a wide variety of ways, or the mutation can be silent, or its effects can be manifest conditioned upon the occurrence of a stimulus. In embodiments, the inserted mutation can convert the cell to a cell that is useful for modeling a disorder, and thus can be considered a pathogenic mutation. In embodiments, the mutation can comprise a selectable marker and/or a detectable marker.
[0022] The POLQ can be inhibited using any suitable approach, including but not limited to small molecule inhibitors, or by using polynucleotide targeting agents that will affect POLQ expression. Further, at least one anti-POLQ monoclonal antibody is commercially available, from, for example, SIGMA-ALDRICH. Thus, inhibiting POLQ comprises either inhibiting specifically or selectively inhibiting its enzymatic activity, or disrupting its expression, or a combination thereof. In embodiments, inhibiting POLQ comprises introducing into the cells a polynucleotide that can inhibit translation of POLQ mRNA, and/or can participate in and/or facilitate RNAi-mediated reduction of POLQ mRNA. The amino acid sequence of POLQ and its gene are known in the art. For example, the murine POLQ gene is publicly accessible under Gene ID: 77782 and the sequence of the human homologue is also known. In one embodiment, an antisense polynucleotide is used to inhibit translation of POLQ mRNA. Antisense nucleic acids can be DNA or RNA molecules that are complementary to at least a portion of the POLQ mRNA. In embodiments, oligomers of about fifteen nucleotides, and/or those that hybridize to the AUG initiation codon may be particularly efficient. The polynucleotides described herein for use in targeting POLQ mRNA can in certain embodiments be modified, such as to be resistant to nucleases.
[0023] In another aspect the disclosure includes RNAi-mediated reduction in POLQ mRNA. RNAi-based inhibition can be achieved using any suitable RNA polynucleotide that is targeted to POLQ mRNA. In embodiments, a single stranded or double stranded RNA, wherein at least one strand is complementary to the POLQ mRNA, can be introduced into the cell to promote RNAi -based degradation of POLQ mRNA. In another embodiment, microRNA (miRNA) targeted to the POLQ mRNA can be used. In another embodiment, a ribozyme that can specifically cleave POLQ mRNA can be used. In yet another embodiment, small interfering RNA (siRNA) can be used. siRNA (or ribozymes) can be introduced directly, for example, as a double stranded siRNA complex, or by using a modified expression vector, such as a lentiviral vector, to produce an shRNA. As is known in the art, shRNAs adopt a typical hairpin secondary structure that contains a paired sense and antisense portion, and a short loop sequence between the paired sense and antisense portions. shRNA is delivered to the cytoplasm where it is processed by DICER into siRNAs. siRNA is recognized by RNA-induced silencing complex (RISC), and once incorporated into RISC, siRNAs facilitate cleavage and degradation of targeted mRNA. In embodiments, an shRNA polynucleotide used to suppress POLQ expression can comprise or consist of between 45-100 nucleotides, inclusive, and including all integers between 45 and 100. The portion of the shRNA that is complementary to the POLQ mRNA can be from 21-29 nucleotides, inclusive, and including all integers between 21 and 29.
[0024] For delivering siRNA via shRNA, any suitable vector, including but not necessarily limited to lentiviral vectors, can be made and used according to standard techniques, given the benefit of the present disclosure. Further, suitable vectors expressing shRNAs targeted to many human mRNAs are commercially available. For example, several shRNA constructs provided in retroviral vectors are available from ORIGENE, (Rockville, MD, USA), and Applied Biological Materials (ABM) Inc., (Richmond, B.C., Canada), and can be included in methods and kits of this disclosure. Moreover, RNAi-mediated approaches for decreasing POLQ have already been demonstrated and can be adapted for use in embodiments of the present invention, given the benefit of this disclosure. In particular, as described in Mateos- Gomez et al., (Mammalian polymerase Θ promotes alternative NHEJ and suppresses
recombination, Nature, 2015 Feb 12;518(7538):254-7), from which the description of compositions and methods for inhibiting POLQ expression is incorporated herein by reference, delivery of shRNA targeted to POLQ mRNA efficiently reduces POLQ protein expression. Further, this is demonstrated in both mouse and human cells. Accordingly, in non-limiting embodiments, shRNA targeting POLQ sequences can comprise thesh Polq-1 (mouse sequence): 5 '-CGGCGGAGTATGAGAACTATT-3 ' (SEQ ID NO: 10); sh Polq-2 (mouse sequence): 5'- CC AGGAATC AAAGACGAC AAT-3 ' (SEQ ID NO: 11); sh Polq-3 (mouse sequence): 5'- CCTGGCTGAATGCTGAACTTT-3 ' (SEQ ID NO: 12); and sh POLQ (human sequence): 5'- CGGGCCTCTTT AGAT ATAAAT-3 ' (SEQ ID NO: 13) that are demonstrated in Mateos-Gomez et al. It will be recognized that the shRNA itself comprises the RNA equivalent of the DNA sequences. It will also be recognized that minor variations in these sequences that do not reduce shRNA targeting of the mRNA can be made and are included within the scope of this disclosure.
[0025] Additionally, custom siRNAs or shRNA can be obtained from publicly available sources. Lentiviruses and other suitable retroviral constructs are capable of stably and permanently infecting target cells, such as by integrating into a chromosome.
[0026] In another aspect, the disclosure includes disrupting the POLQ gene such that
POLQ mRNA and protein are not expressed. In one embodiment, the POLQ gene can be disrupted by targeted mutagenesis. In embodiments, the present disclosure provides for replacement of the POLQ gene with a sequence encoding a detectable marker, such as a fluorescent protein, or integrating such a sequence into the POLQ gene, thereby disrupting it, or integrating such a sequence elsewhere in the genome of the mammalian cells. By replacing POLQ or integrating a sequence encoding a detectable protein into it the disclosure provides for marking cells that do not express POLQ.
[0027] The disclosure includes inhibiting POLQ prior to introducing the Cas9 and gRNA sequence, or concurrently introducing a POLQ inhibitor to the cell with the Cas9-nickase and gRNA sequence.
[0028] As discussed above, the DNA that is selected to by modified using compositions and methods of this disclosure can be any suitable target DNA sequence. In embodiments, modification of the DNA confers a change in phenotype of the cell, such as a change in morphology, growth rate, expression of a detectable or selectable marker, or the modification is lethal to the cell. In embodiments, the DNA sequence that is modified is present on a chromosome, or is present on an extra-chromosomal element, including but not limited to a plasmid. In embodiments the modification can comprise insertion of a detectable marker into a DNA element in a cell. In embodiments, the disclosure includes modification of DNA in one or more cells, and can further comprise identification of one or more cells in which the
modification occurs, or identification of cells wherein the modification does not occur, and separating such cells from one another to provide a population of isolated cells that, for example, contain a DNA modification made by the Cas9. In embodiments, the mammalian cells can be any mammalian cells, including but not limited to stem cells, such as totipotent, pluripotent, and multipotent cells. In embodiments the cells are hematopoietic cells. In embodiments, the cells are embryonic stem cells, or adult stem cells. In embodiments, the cells are epidermal stem cells or epithelial stem cells, or neuronal precursor cells. In embodiments, the cells are differentiated cells when the intended mutation is introduced. In embodiments, the cells are mammalian cells. In embodiments, the cells are human, or are non-human animal cells. In embodiments the disclosure includes progeny of any such cells, and non-human mammals comprising a cell or progeny of a cell made as described herein. In certain embodiments the cells modified according to this disclosure are not worm cells, including but not limited to cells of any nematode. In an embodiment, the cells modified according to this disclosure are not C. elegans cells.
[0029] In embodiments, the disclosure includes obtaining cells from an individual, modifying the cells ex vivo as described herein, and reintroducing the cells and/or their progeny into the individual for prophylaxis and/or therapy of a condition, disease or disorder, or to treat an injury, trauma or anatomical defect. In embodiments, the cells modified ex vivo as described herein are used autologously. In embodiments, the cells are provided as cell lines. In
embodiments, the cells are engineered to produce a protein or other compound; the cells themselves or the protein or compound they produce in certain implementations may be useful for prophylactic or therapeutic applications.
[0030] In non-limiting examples, the mutation introduced into cells according to this disclosure is a homozygous dominant or homozygous recessive, or heterozygous dominant or heterozygous recessive mutation, that is correlated with a phenotype or condition, and is thus useful for modeling such phenotype or condition.
[0031] As described above, in embodiments, the disclosure includes identifying and separating cells that are heterozygous or are homozygous for a mutation introduced according to this disclosure. Such cells can be separated from one another, and from cells into which no mutation has been introduced. Identification and separation can be achieved using any suitable approach, including sequencing individual cells and expanding clones using routine approaches, or by including and detecting detectable markers, and/or other indicia of the mutation. In embodiments, the disclosure includes expanding separated cells to obtain a clonal population of the cells, and can further comprise promoting and/or allowing the cells to differentiate. Thus, the disclosure includes fully or partially differentiated cells that are homozygous or
heterozygous for the introduced intended mutation. In embodiments, the disclosure includes compositions that include a cellular component, wherein the cellular component comprises or consists of cells modified according to this disclosure.
[0032] In an embodiment, the disclosure include a kit comprising a polynucleotides encoding a Cas9, and may further comprise an agent capable of inhibiting POLQ, or a polynucleotide that encodes a polynucleotide capable of disrupting POLQ expression. In non- limiting examples, the polynucleotide capable of disrupting POLQ expression is a commercially available viral vector that encodes an shRNA targeted to POLQ mRNA. The kit can further comprised a guide RNA or a polynucleotide encoding the guide RNA, or a vector adapted to insert and express a polynucleotide comprising a suitable gRNA segment. The kit can further comprised a polynucleotide or vector that is adaptable for expressing a mutation template.
Example 1
[0033] The following specific Example is provided to illustrate the invention, but is not intended to be limiting in any way.
[0034] It will be apparent from this Example that a specific locus in cells with different genetic backgrounds was targeted, namely POLQ wild type and POLQ null backgrounds, using a ZsGreen reporter plasmid. Targeting was achieved with CRISPR/Cas9 editing tools, employing both the Cas9 nuclease and Cas9 nickase independently. To determine the efficiency of integration of the ZsGreen plasmid, the cells were subject to FACS sorting and the ZsGreen positive cells were quantified. In each case, a comparison of the % of ZsGreen+ cells in POLQ null to wild type cells was made. The results presented in Figure 1, with particular amino acid and nucleotide sequences identified in Figure 2 and 4. The results presented in Figure 1 show that POLQ inhibition had no impact on editing efficiency when using Cas9 nuclease to target sox2. In contrast, more efficient editing as evidenced by integration efficiency was achieved with Cas9 nickase with POLQ inhibition. Further, homozygous targeting (represented in Figure 1) is more enriched than heterozygous targeting. In the case of hsp90abl locus targeting (Figure 1), more homozygous and heterozygous editing was achieved with Cas9 nuclease in POLQ deficient mESCs and MEFs. Thus, this Example demonstrates the principle that improved Cas9 editing efficiency can be obtained by combining Cas9 with POLQ inhibition in mammalian cells. The improved efficiency of Cas9-Nuclease or Nickase targeting appears to be locus specific .
[0035] This Example provides: first, a demonstration of inserting into CCE mouse embryonic stem cells (CCE mESCs) a detectable marker (ZsGreen - a green fluorescent protein) at the end of the sox2 gene. A P2A sequence (57 nucleotides) followed by ZsGreen (696 nt) gene was inserted preceding the STOP codon of sox2. Second, a demonstration of inserting into mouse embryonic stem cells (mESCs) and mouse embryonic fibroblast (MEFs) a detectable marker (ZsGreen - a green fluorescent protein) at the end of the hsp90abl gene. A P2A sequence (57 nucleotides) followed by ZsGreen (696 nt) gene was inserted preceding the STOP codon of hsp90abl. Therefore, in the case of correct targeting, the Sox2 or the Hsp90abl and ZsGreen proteins will be translated as distinct proteins from a single mRNA, transcription of which is controlled by the sox2 or hsp90abl promoters respectively. The experiments were performed in POLQ WT and POLQ null mouse cells. [0036] To perform the experiment, the results of which are illustrated graphically in
Figure 1, and performed using the representative sequences shown in Figure 2 and 4, the following experimental approach and protocol was used. Those skilled in the art will recognize given the benefit of the present disclosure that alternative techniques and reagents could also be used.
[0037] Reagents
[0038] Two donor plasmids were generated as template for the targeting of Sox2 and
Hsp90abl using pSL301 as a vector, to which the following three segments were introduced. 1- The last 83 lnt or 700nt of Sox2 or Hsp90abl genes without the STOP codon (used as a 5' homology arms in the donor plasmid), fused to 2- P2A-ZsGreen and followed by 3- the first 801 nt of the 3'UTR of Sox2 or the 700nt after the STOP codon of Hsp90abl respectively (used as a 3' homology arm in the donor plasmid).
[0039] Cas9 nuclease and nickase D10A were expressed from publicly available expression vectors. In particular, pX330-U6-Chimeric_BB-CBh-hSpCas9 (Addgene plasmid #42230) and pX335-U6-Chimeric_BB-CBh-hSpCas9n(D10A) (Addgene Plasmid #42335), respectively. In both plasmids a Puromycin-cassette was inserted for selection, which serves to enrich for ES cells expressing the Cas9. With the Cas9 nuclease version one guide RNA
(gRNA) was expressed (5'-CAGCCCTCACATGTGCGACA-3' (SEQ ID NO: 14) for Sox2 and 5' - CGATGAGGATGCCTCGCGC A-3 ' (SEQ ID NO: 15) for Hsp90abl) and two gRNAs with the nickase version (5 ' -TT AATGGCCGTGCCGGGC AC-3 ' (SEQ ID NO: 16; 5'-
GTGAGGGCTGGACTGCGAAC-3 ' (SEQ ID NO: 16) for the Sox2 that after cleavage generate ends with 5' overhangs; For the Hsp90abl targeting the couple 5'- CGATGAGGATGCCTCGCGC A-3 ' (SEQ ID NO: 17) and 5'-
GGGGAT ACT AT AC AGAGGGC-3 ' (SEQ ID NO: 18) were used to generate ends with 3' overhangs, and the couple 5' - CCCTCTGTATAGTATCCCCG-3 ' (SEQ ID NO: 19) and 5 '- ATCCACCTCTTCCATGCGCG-3' (SEQ ID NO:20) were used to generate ends with 5' overhangs). The gRNAs were designed using crispr.mit.edu/ and cloned using www.genome- engineering.org/crispr/?page_id=23. In the case of the nickase both gRNAs were cloned in tandem U6 promoter-gRNA-l-U6 promoter-gRNA-2 using a Kpnl restriction site located in the px335 plasmid downstream of the gRNA cloning site.
[0040] The mouse cells were CCE mES cells, mES cells and MEFs (all three WT and
POLQ null). Lipofectamine® 3000 was obtained from Life Technologies. The culture media for CCE mESCs contained 500ml of DMEM and 90ml of ESC qualified FBS, supplemented according to manufacturer with non-essential amino acids, L-glutamine, penicillin/streptomycin, B-mercaptoethanol and LIF protein. The culture media for mESCs contained 500ml of DMEM and 90ml of ESC qualified FBS, supplemented according to manufacturer with non-essential amino acids, L-glutamine, penicillin/streptomycin, B-mercaptoethanol, LIF protein,
PD03259010 (MEK inhibitor) luM and CHIR99021 (GSK inhibitor) 3uM. The culture media for MEFs contained 500ml of DMEM and 50ml of FBS, supplemented according to
manufacturer with non-essential amino acids, L-glutamine, penicillin/streptomycin.
[0041] DNA-PK inhibitor was NU7441 (luM) was included to block classical- HEJ.
[0042] The following protocol was used to obtain the results summarized in Figure 1.
[0043] Transfecting plasmids into CCE mES cells:
[0044] Add 7.5ul of Lipofectamine® 3000 to 50ul of Opti-MEM® Reduced Serum
Medium, incubate 5 min at RT.
[0045] Add 500ng of Cas9 plasmid and 2000ng of donor plasmid to 50ul of Opti-MEM and then 5ul of P3000 reagent.
[0046] Mix both solutions and incubate RT at least 5 min.
[0047] Detach the CCE mES cells and obtain a single cell suspension.
[0048] Count the cells and take 1 million. Spin down 4 min 300g.
[0049] Aspirate the media and break up the cell pellet by manipulating the tube.
[0050] Add the solution (00030) to the cells, gently pipette up and down to make sure all cells are well resuspended. Incubate 10-12 min. Shake the tube to avoid cells precipitation.
[0051] After incubation, add 4ml of medium and plate them in a 6cm plate.
[0052] Place cells in the incubator overnight and change medium the following morning.
Add puromycin to a final concentration of 2.5 ug/ml.
[0053] After 30 hours of selection with puromycin (approximately 48 hour after transfection) to enrich for Cas9 expressing cells, the medium was replaced with non-puromycin containing media and the selected cells were allowed to grow.
[0054] For cells treated with DNA-PK inhibitor, the inhibitor was added in parallel to puromycin selection.
[0055] For 8 days medium was changed on a daily basis and the cells were passaged as required.
[0056] Harvest the cells and prepare them for flow cytometry to quantify the percentage of cells stably expressing ZsGreen. FACS Signal is gated as low and high Zsgreen, which is likely to reflect one and two copies of the gene. Results are depicted in Figure 1.
[0057] Transfecting plasmids into mES cells and MEFs:
[0058] Add 3ul of Lipofectamine® 3000 to 50ul of Opti-MEM® Reduced Serum Medium, incubate 5 min at RT.
[0059] Add 200ng of Cas9 plasmid and 800ng of donor plasmid to 50ul of Opti-MEM and then 2ul of P3000 reagent.
[0060] Mix both solutions and incubate RT at least 5 min.
[0061] Detach the mES cells or the MEFs and obtain a single cell suspension.
[0062] Count the cells and take 1 million. Spin down 4 min 300g.
[0063] Aspirate the media and break up the cell pellet by manipulating the tube.
[0064] Add the solution (00044) to the cells, gently pipette up and down to make sure all cells are well resuspended. Incubate 10-12 min. Shake the tube to avoid cells precipitation.
[0065] After incubation, add 1 ml of medium and plate them in a well of a 12 well plate.
[0066] Place cells in the incubator overnight and change medium the following morning.
Add puromycin to a final concentration of 0.5 ug/ml for mESCs and 2ug/ml for MEFs.
[0067] For mESCs and MEFs respectively, after 48 or 72 hours of selection with puromycin to enrich for Cas9 expressing cells, the medium was replaced with non-puromycin containing media and the selected cells were allowed to grow.
[0068] For 8 days medium was changed on a daily basis and the cells were passaged as required.
[0069] Harvest the cells and prepare them for flow cytometry to quantify the percentage of cells stably expressing ZsGreen. FACS Signal is gated as low and high Zsgreen, which is likely to reflect one and two copies of the gene. Results are depicted in Figure 1.
[0070] While the invention has been described through specific embodiments, routine modifications will be apparent to those skilled in the art and such modifications are intended to be within the scope of the present invention.

Claims

What is claimed is:
1. A method for modification of a target DNA sequence in a chromosome of a mammalian cell comprising introducing into the cell one or more polynucleotides encoding: i) a clustered regularly interspaced short palindromic repeats (CRISPR) associated enzyme (Cas9): and ii) a CRISPR guide RNA (gRNA) directed to the target DNA sequence in the cell; the method further comprising introducing into the cell an agent capable of inhibiting pol Θ) ("POLQ").
2. The method of claim 1, further comprising introducing into the cell a polynucleotide for modifying the target DNA sequence ("mutation template").
3. The method of claim 1 wherein the Cas9 and the gRNA are encoded on a single expression plasmid.
4. The method of claim 1 wherein the Cas9 and the gRNA are encoded on distinct expression plasmids.
5. The method of claim 1, wherein the mammalian cell comprises a stem cell.
6. The method of claim 1, wherein the Cas9 is a Cas9 nickase.
7. The method of claim 1, wherein the Cas9 is a Cas9 nuclease.
8. The method of claim 1, wherein the agent capable of inhibiting POLQ comprises a polynucleotide directed to a polynucleotide in the cell that encodes POLQ (POLQ targeting polynucleotide).
9. The method of claim 8, wherein the POLQ targeting polynucleotide is selected from the group consisting of an antisense oligonucleotide, an siRNA, an shRNA, a polynucleotide encoding an shRNA, a polynucleotide encoding a ribozyme, and combinations thereof.
10. The method of any one of claims 1-9, further comprising allowing expression of the Cas9 such that the target DNA sequence is modified, wherein the modified DNA sequence comprises an insertion of at least a segment of the mutation template into the target DNA sequence.
11. The method of claim 10, wherein the insertion mutation template encodes a detectable marker.
12. The method of claim 10, wherein the cell is a non-human mammalian cell.
13. A modified mammalian cell made by a process of any one of claims 1-9.
14. A modified mammalian cell made by a process of claim 10.
15. A non-human mammal comprising a cell of claim 10 or progeny of a cell of claim 10.
16. A kit comprising a polynucleotide encoding a Cas9, the kit further comprising an agent for use in inhibiting POLQ.
17. The kit of claim 16, wherein the agent for use in inhibiting POLQ is a POLQ targeting polynucleotide.
18. The kit of claim 16, further comprising a guide RNA or a polynucleotide encoding the guide RNA.
19. The kit of claim 16, further comprising an expression vector configured for expressing a mutation template.
20. The kit of any one of claims 16-20, wherein the Cas9 is a Cas9 nickase.
21. The kit of any one of claims 16-20, wherein the Cas9 is a Cas9 nuclease.
22. A kit comprising an expression vector configured for expressing a mutation template for use in clustered regularly interspaced short palindromic repeats (CRISPR) chromosome editing, the kit further comprising an agent for use in inhibiting POLQ.
23. The kit of claim 22, wherein the agent for use in inhibiting POLQ is a POLQ targeting polynucleotide.
PCT/US2016/055967 2015-10-07 2016-10-07 Compositions and methods for enhancing crispr activity by polq inhibition WO2017062754A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US15/766,256 US10760081B2 (en) 2015-10-07 2016-10-07 Compositions and methods for enhancing CRISPR activity by POLQ inhibition

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201562238192P 2015-10-07 2015-10-07
US62/238,192 2015-10-07

Publications (1)

Publication Number Publication Date
WO2017062754A1 true WO2017062754A1 (en) 2017-04-13

Family

ID=58488546

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2016/055967 WO2017062754A1 (en) 2015-10-07 2016-10-07 Compositions and methods for enhancing crispr activity by polq inhibition

Country Status (2)

Country Link
US (1) US10760081B2 (en)
WO (1) WO2017062754A1 (en)

Cited By (41)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9999671B2 (en) 2013-09-06 2018-06-19 President And Fellows Of Harvard College Delivery of negatively charged proteins using cationic lipids
US10077453B2 (en) 2014-07-30 2018-09-18 President And Fellows Of Harvard College CAS9 proteins including ligand-dependent inteins
US10113163B2 (en) 2016-08-03 2018-10-30 President And Fellows Of Harvard College Adenosine nucleobase editors and uses thereof
US10167457B2 (en) 2015-10-23 2019-01-01 President And Fellows Of Harvard College Nucleobase editors and uses thereof
WO2019086460A1 (en) 2017-10-30 2019-05-09 Kws Saat Se New strategies for precision genome editing
US10323236B2 (en) 2011-07-22 2019-06-18 President And Fellows Of Harvard College Evaluation and improvement of nuclease cleavage specificity
US10465176B2 (en) 2013-12-12 2019-11-05 President And Fellows Of Harvard College Cas variants for gene editing
WO2019234132A1 (en) * 2018-06-05 2019-12-12 KWS SAAT SE & Co. KGaA Base editing in polymerase theta deficient plants
WO2019234129A1 (en) * 2018-06-05 2019-12-12 KWS SAAT SE & Co. KGaA Haploid induction with modified dna-repair
US10508298B2 (en) 2013-08-09 2019-12-17 President And Fellows Of Harvard College Methods for identifying a target site of a CAS9 nuclease
WO2020030924A1 (en) 2018-08-10 2020-02-13 Artios Pharma Limited Thiazoleureas as anticancer agents
WO2020030925A1 (en) 2018-08-10 2020-02-13 Artios Pharma Limited Heterocyclic substituted ureas, for use against cancer
US10597679B2 (en) 2013-09-06 2020-03-24 President And Fellows Of Harvard College Switchable Cas9 nucleases and uses thereof
US10745677B2 (en) 2016-12-23 2020-08-18 President And Fellows Of Harvard College Editing of CCR5 receptor gene to protect against HIV infection
US10858639B2 (en) 2013-09-06 2020-12-08 President And Fellows Of Harvard College CAS9 variants and uses thereof
WO2021028670A1 (en) 2019-08-09 2021-02-18 Artios Pharma Limited Deuterated compounds for use in the treatment of cancer
WO2021028643A1 (en) 2019-08-09 2021-02-18 Artios Pharma Limited Heterocyclic compounds for use in the treatment of cancer
WO2021123785A1 (en) 2019-12-17 2021-06-24 Artios Pharma Limited Dna polymerase theta inhibitors
US11046948B2 (en) 2013-08-22 2021-06-29 President And Fellows Of Harvard College Engineered transcription activator-like effector (TALE) domains and uses thereof
US11236313B2 (en) 2016-04-13 2022-02-01 Editas Medicine, Inc. Cas9 fusion molecules, gene editing systems, and methods of use thereof
US11268082B2 (en) 2017-03-23 2022-03-08 President And Fellows Of Harvard College Nucleobase editors comprising nucleic acid programmable DNA binding proteins
US11306324B2 (en) 2016-10-14 2022-04-19 President And Fellows Of Harvard College AAV delivery of nucleobase editors
US11319532B2 (en) 2017-08-30 2022-05-03 President And Fellows Of Harvard College High efficiency base editors comprising Gam
WO2022167817A1 (en) 2021-02-07 2022-08-11 Artios Pharma Limited Novel process
US11447770B1 (en) 2019-03-19 2022-09-20 The Broad Institute, Inc. Methods and compositions for prime editing nucleotide sequences
US11542496B2 (en) 2017-03-10 2023-01-03 President And Fellows Of Harvard College Cytosine to guanine base editor
US11542509B2 (en) 2016-08-24 2023-01-03 President And Fellows Of Harvard College Incorporation of unnatural amino acids into proteins using base editing
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
US11597924B2 (en) 2016-03-25 2023-03-07 Editas Medicine, Inc. Genome editing systems comprising repair-modulating enzyme molecules and methods of their use
WO2023067353A1 (en) 2021-10-21 2023-04-27 Artios Pharma Limited Heterocyclic compounds for use in the treatment of cancer
WO2023067354A1 (en) 2021-10-21 2023-04-27 Artios Pharma Limited Heterocyclic compounds for use in the treatment of cancer
WO2023067356A1 (en) 2021-10-21 2023-04-27 Artios Pharma Limited Heterocyclic compounds for use in the treatment of cancer
US11661590B2 (en) 2016-08-09 2023-05-30 President And Fellows Of Harvard College Programmable CAS9-recombinase fusion proteins and uses thereof
US11667911B2 (en) 2015-09-24 2023-06-06 Editas Medicine, Inc. Use of exonucleases to improve CRISPR/CAS-mediated genome editing
US11680268B2 (en) 2014-11-07 2023-06-20 Editas Medicine, Inc. Methods for improving CRISPR/Cas-mediated genome-editing
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)
US11795443B2 (en) 2017-10-16 2023-10-24 The Broad Institute, Inc. Uses of adenosine base editors
US11866726B2 (en) 2017-07-14 2024-01-09 Editas Medicine, Inc. Systems and methods for targeted integration and genome editing and detection thereof using integrated priming sites
US11898179B2 (en) 2017-03-09 2024-02-13 President And Fellows Of Harvard College Suppression of pain by gene editing
US11912985B2 (en) 2020-05-08 2024-02-27 The Broad Institute, Inc. Methods and compositions for simultaneous editing of both strands of a target double-stranded nucleotide sequence
US11999947B2 (en) 2023-02-24 2024-06-04 President And Fellows Of Harvard College Adenosine nucleobase editors and uses thereof

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20120021935A1 (en) * 2009-11-13 2012-01-26 Centre National De La Recherche Scientifique (Cnrs) Signature for the diagnosis of cancer aggressiveness and genetic instability
US20150020223A1 (en) * 2012-12-12 2015-01-15 The Broad Institute Inc. Delivery, engineering and optimization of systems, methods and compositions for sequence manipulation and therapeutic applications
US20150232881A1 (en) * 2013-11-07 2015-08-20 Editas Medicine, Inc. CRISPR-RELATED METHODS AND COMPOSITIONS WITH GOVERNING gRNAS

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20120021935A1 (en) * 2009-11-13 2012-01-26 Centre National De La Recherche Scientifique (Cnrs) Signature for the diagnosis of cancer aggressiveness and genetic instability
US20150020223A1 (en) * 2012-12-12 2015-01-15 The Broad Institute Inc. Delivery, engineering and optimization of systems, methods and compositions for sequence manipulation and therapeutic applications
US20150232881A1 (en) * 2013-11-07 2015-08-20 Editas Medicine, Inc. CRISPR-RELATED METHODS AND COMPOSITIONS WITH GOVERNING gRNAS

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
CECCALDI ET AL.: "Homologous-recombination-deficient tumours are dependent on Poltheta-mediated repair", NATURE, vol. 518, February 2015 (2015-02-01), pages 1 - 32, XP055374850 *
MALI ET AL.: "RNA-guided human genome engineering via Cas9", SCIENCE, vol. 339, 3 January 2013 (2013-01-03), pages 823 - 826, XP055153369 *
MATEOS-GOMEZ ET AL.: "Mammalian polymerase theta promotes alternative NHEJ and suppresses recombination", NATURE, vol. 518, 2 February 2015 (2015-02-02), pages 254 - 257, XP055374846 *

Cited By (58)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10323236B2 (en) 2011-07-22 2019-06-18 President And Fellows Of Harvard College Evaluation and improvement of nuclease cleavage specificity
US11920181B2 (en) 2013-08-09 2024-03-05 President And Fellows Of Harvard College Nuclease profiling system
US10954548B2 (en) 2013-08-09 2021-03-23 President And Fellows Of Harvard College Nuclease profiling system
US10508298B2 (en) 2013-08-09 2019-12-17 President And Fellows Of Harvard College Methods for identifying a target site of a CAS9 nuclease
US11046948B2 (en) 2013-08-22 2021-06-29 President And Fellows Of Harvard College Engineered transcription activator-like effector (TALE) domains and uses thereof
US9999671B2 (en) 2013-09-06 2018-06-19 President And Fellows Of Harvard College Delivery of negatively charged proteins using cationic lipids
US10858639B2 (en) 2013-09-06 2020-12-08 President And Fellows Of Harvard College CAS9 variants and uses thereof
US10912833B2 (en) 2013-09-06 2021-02-09 President And Fellows Of Harvard College Delivery of negatively charged proteins using cationic lipids
US11299755B2 (en) 2013-09-06 2022-04-12 President And Fellows Of Harvard College Switchable CAS9 nucleases and uses thereof
US10597679B2 (en) 2013-09-06 2020-03-24 President And Fellows Of Harvard College Switchable Cas9 nucleases and uses thereof
US10682410B2 (en) 2013-09-06 2020-06-16 President And Fellows Of Harvard College Delivery system for functional nucleases
US11053481B2 (en) 2013-12-12 2021-07-06 President And Fellows Of Harvard College Fusions of Cas9 domains and nucleic acid-editing domains
US11124782B2 (en) 2013-12-12 2021-09-21 President And Fellows Of Harvard College Cas variants for gene editing
US10465176B2 (en) 2013-12-12 2019-11-05 President And Fellows Of Harvard College Cas variants for gene editing
US11578343B2 (en) 2014-07-30 2023-02-14 President And Fellows Of Harvard College CAS9 proteins including ligand-dependent inteins
US10704062B2 (en) 2014-07-30 2020-07-07 President And Fellows Of Harvard College CAS9 proteins including ligand-dependent inteins
US10077453B2 (en) 2014-07-30 2018-09-18 President And Fellows Of Harvard College CAS9 proteins including ligand-dependent inteins
US11680268B2 (en) 2014-11-07 2023-06-20 Editas Medicine, Inc. Methods for improving CRISPR/Cas-mediated genome-editing
US11667911B2 (en) 2015-09-24 2023-06-06 Editas Medicine, Inc. Use of exonucleases to improve CRISPR/CAS-mediated genome editing
US10167457B2 (en) 2015-10-23 2019-01-01 President And Fellows Of Harvard College Nucleobase editors and uses thereof
US11214780B2 (en) 2015-10-23 2022-01-04 President And Fellows Of Harvard College Nucleobase editors and uses thereof
US11597924B2 (en) 2016-03-25 2023-03-07 Editas Medicine, Inc. Genome editing systems comprising repair-modulating enzyme molecules and methods of their use
US11236313B2 (en) 2016-04-13 2022-02-01 Editas Medicine, Inc. Cas9 fusion molecules, gene editing systems, and methods of use thereof
US10947530B2 (en) 2016-08-03 2021-03-16 President And Fellows Of Harvard College Adenosine nucleobase editors and uses thereof
US10113163B2 (en) 2016-08-03 2018-10-30 President And Fellows Of Harvard College Adenosine nucleobase editors and uses thereof
US11702651B2 (en) 2016-08-03 2023-07-18 President And Fellows Of Harvard College Adenosine nucleobase editors and uses thereof
US11661590B2 (en) 2016-08-09 2023-05-30 President And Fellows Of Harvard College Programmable CAS9-recombinase fusion proteins and uses thereof
US11542509B2 (en) 2016-08-24 2023-01-03 President And Fellows Of Harvard College Incorporation of unnatural amino acids into proteins using base editing
US11306324B2 (en) 2016-10-14 2022-04-19 President And Fellows Of Harvard College AAV delivery of nucleobase editors
US10745677B2 (en) 2016-12-23 2020-08-18 President And Fellows Of Harvard College Editing of CCR5 receptor gene to protect against HIV infection
US11820969B2 (en) 2016-12-23 2023-11-21 President And Fellows Of Harvard College Editing of CCR2 receptor gene to protect against HIV infection
US11898179B2 (en) 2017-03-09 2024-02-13 President And Fellows Of Harvard College Suppression of pain by gene editing
US11542496B2 (en) 2017-03-10 2023-01-03 President And Fellows Of Harvard College Cytosine to guanine base editor
US11268082B2 (en) 2017-03-23 2022-03-08 President And Fellows Of Harvard College Nucleobase editors comprising nucleic acid programmable DNA binding proteins
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
US11866726B2 (en) 2017-07-14 2024-01-09 Editas Medicine, Inc. Systems and methods for targeted integration and genome editing and detection thereof using integrated priming sites
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
US11932884B2 (en) 2017-08-30 2024-03-19 President And Fellows Of Harvard College High efficiency base editors comprising Gam
US11795443B2 (en) 2017-10-16 2023-10-24 The Broad Institute, Inc. Uses of adenosine base editors
CN111542610A (en) * 2017-10-30 2020-08-14 科沃施种子欧洲股份两合公司 Novel strategy for precise genome editing
WO2019086460A1 (en) 2017-10-30 2019-05-09 Kws Saat Se New strategies for precision genome editing
WO2019234129A1 (en) * 2018-06-05 2019-12-12 KWS SAAT SE & Co. KGaA Haploid induction with modified dna-repair
WO2019234132A1 (en) * 2018-06-05 2019-12-12 KWS SAAT SE & Co. KGaA Base editing in polymerase theta deficient plants
WO2020030924A1 (en) 2018-08-10 2020-02-13 Artios Pharma Limited Thiazoleureas as anticancer agents
WO2020030925A1 (en) 2018-08-10 2020-02-13 Artios Pharma Limited Heterocyclic substituted ureas, for use against cancer
US11643652B2 (en) 2019-03-19 2023-05-09 The Broad Institute, Inc. Methods and compositions for prime editing nucleotide sequences
US11795452B2 (en) 2019-03-19 2023-10-24 The Broad Institute, Inc. Methods and compositions for prime editing nucleotide sequences
US11447770B1 (en) 2019-03-19 2022-09-20 The Broad Institute, Inc. Methods and compositions for prime editing nucleotide sequences
WO2021028670A1 (en) 2019-08-09 2021-02-18 Artios Pharma Limited Deuterated compounds for use in the treatment of cancer
WO2021028643A1 (en) 2019-08-09 2021-02-18 Artios Pharma Limited Heterocyclic compounds for use in the treatment of cancer
WO2021123785A1 (en) 2019-12-17 2021-06-24 Artios Pharma Limited Dna polymerase theta inhibitors
US11912985B2 (en) 2020-05-08 2024-02-27 The Broad Institute, Inc. Methods and compositions for simultaneous editing of both strands of a target double-stranded nucleotide sequence
WO2022167817A1 (en) 2021-02-07 2022-08-11 Artios Pharma Limited Novel process
WO2023067356A1 (en) 2021-10-21 2023-04-27 Artios Pharma Limited Heterocyclic compounds for use in the treatment of cancer
WO2023067354A1 (en) 2021-10-21 2023-04-27 Artios Pharma Limited Heterocyclic compounds for use in the treatment of cancer
WO2023067353A1 (en) 2021-10-21 2023-04-27 Artios Pharma Limited Heterocyclic compounds for use in the treatment of cancer
US11999947B2 (en) 2023-02-24 2024-06-04 President And Fellows Of Harvard College Adenosine nucleobase editors and uses thereof

Also Published As

Publication number Publication date
US10760081B2 (en) 2020-09-01
US20180305697A1 (en) 2018-10-25

Similar Documents

Publication Publication Date Title
US10760081B2 (en) Compositions and methods for enhancing CRISPR activity by POLQ inhibition
Yuan et al. Genetic modulation of RNA splicing with a CRISPR-guided cytidine deaminase
Arbab et al. Cloning-free CRISPR
CN107208078B (en) Methods and compositions for targeted genetic modification using paired guide RNAs
US20190330603A1 (en) Crispr-cas system, materials and methods
Jafarnejad et al. Translational control of ERK signaling through miRNA/4EHP-directed silencing
CA3136735A1 (en) Methods and compositions for editing rnas
WO2017181107A2 (en) Modified cpf1 mrna, modified guide rna, and uses thereof
US20150299702A1 (en) Circular rna for inhibition of microrna
US20130164846A1 (en) Rna molecules and uses thereof
WO2018033110A1 (en) Method for repairing gene point mutation
Yoshimoto et al. Biosynthesis of circular RNA ciRS-7/CDR1as is mediated by mammalian-wide interspersed repeats
WO2018089437A1 (en) Compositions and methods for scarless genome editing
Jain et al. Distinct functions of argonaute slicer in siRNA maturation and heterochromatin formation
CA3206081A1 (en) Gene transcription framework, vector system, genome sequence editing method and application
JP2020037599A (en) Production and utilization of novel therapeutic anti-cancer agents
TW201735932A (en) Use of microRNA precursors as drugs for inducing CD34-positive adult stem cell expansion
JP2023544987A (en) Prime editing-based gene editing composition with improved editing efficiency and its uses
Sasaki et al. Oocyte-specific gene knockdown by intronic artificial microRNAs driven by Zp3 transcription in mice
EP4230738A1 (en) Prime editing-based gene editing composition with enhanced editing efficiency and use thereof
US20050079614A1 (en) RNAs able to modulate chromatin silencing
CN114072500B (en) Method for controlling microRNA expression
US20220333129A1 (en) A nucleic acid delivery vector comprising a circular single stranded polynucleotide
WO2023169482A1 (en) Modified crispr-based gene editing system and methods of use
Chavali et al. Functional categories of RNA regulation

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 16854414

Country of ref document: EP

Kind code of ref document: A1

WWE Wipo information: entry into national phase

Ref document number: 15766256

Country of ref document: US

NENP Non-entry into the national phase

Ref country code: DE

32PN Ep: public notification in the ep bulletin as address of the adressee cannot be established

Free format text: NOTING OF LOSS OF RIGHTS PURSUANT TO RULE 112(1) EPC (EPO FORM 1205A DATED 14/08/2018)

122 Ep: pct application non-entry in european phase

Ref document number: 16854414

Country of ref document: EP

Kind code of ref document: A1