WO2020068643A1 - Crispr à spécificité améliorée - Google Patents

Crispr à spécificité améliorée Download PDF

Info

Publication number
WO2020068643A1
WO2020068643A1 PCT/US2019/052390 US2019052390W WO2020068643A1 WO 2020068643 A1 WO2020068643 A1 WO 2020068643A1 US 2019052390 W US2019052390 W US 2019052390W WO 2020068643 A1 WO2020068643 A1 WO 2020068643A1
Authority
WO
WIPO (PCT)
Prior art keywords
virus
grnas
casy
composition
nucleic acid
Prior art date
Application number
PCT/US2019/052390
Other languages
English (en)
Inventor
Thomas MALCOLM
Original Assignee
Excision Biotherapeutics, Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from US16/141,407 external-priority patent/US20190071673A1/en
Application filed by Excision Biotherapeutics, Inc. filed Critical Excision Biotherapeutics, Inc.
Publication of WO2020068643A1 publication Critical patent/WO2020068643A1/fr

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
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • 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
    • C12N15/1131Non-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 viruses
    • 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
    • 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]

Definitions

  • the present invention relates to compositions and methods for delivering gene therapeutics. More specifically, the present invention relates to compositions and treatments for excising viruses from infected host cells and inactivating viruses with chemically altered compositions.
  • nucleases Gene editing allows DNA or RNA to be inserted, deleted, or replaced in an organism's genome by the use of nucleases.
  • nucleases There are several types of nucleases currently used, including meganucleases, zinc finger nucleases, transcription activator-like effector-based nucleases (TALENs), and clustered regularly interspaced short palindromic repeats (CRISPR)-Cas nucleases. These nucleases can create site-specific double strand breaks of the DNA in order to edit the DNA.
  • TALENs transcription activator-like effector-based nucleases
  • CRISPR clustered regularly interspaced short palindromic repeats
  • meganucleases have very long recognition sequences and are very specific to DNA. While meganucleases are less toxic than other gene editors, they are expensive to construct, as not many are known and mutagenesis must be used to create variants that recognize specific sequences.
  • Both zinc-finger and TALEN nucleases are non-specific for DNA but can be linked to DNA sequence recognizing peptides. However, each of these nucleases can produce off-target effects and cytotoxicity, and require time to create the DNA sequence recognizing peptides.
  • CRISPR-Cas nucleases are derived from prokaryotic systems and can use the Cas9 nuclease, the Cpfl nuclease, or other Cas nucleases for DNA editing.
  • CRISPR is an adaptive immune system found in many microbial organisms. While the CRISPR system was not well understood, it was found that there were genes associated to the CRISPR regions that coded for exonucleases and/or helicases, called CRISPR-associated proteins (Cas).
  • CRISPR-associated proteins Cas.
  • Several different types of Cas proteins were found, some using multi protein complexes (Type I), some using singe effector proteins with a universal tracrRNA and crRNA specific for a target DNA sequence (Type II), and some found in archea (Type III).
  • Cas9 (a Type II Cas protein) was discovered when the bacteria Streptococcus thermophilus was being studied and an unusual CRISPR locus was found (Bolotin, et al. 2005). It was also found that the spacers share a common sequence at one end (the protospacer adjacent motif PAM), and is used for target sequence recognition. Cas9 was not found with a screen but by examining a specific bacteria.
  • U.S. Patent Application Serial No. 14/838,057 to Khalili, et al. discloses a method of inactivating a proviral DNA integrated into the genome of a host cell latently infected with a retrovirus, by treating the host cell with a composition comprising a Clustered Regularly Interspaced Short Palindromic Repeat (CRISPR)-associated endonuclease, and two or more different guide RNAs (gRNAs), wherein each of the at least two gRNAs is complementary to a different target nucleic acid sequence in a long terminal repeat (LTR) of the proviral DNA; and inactivating the proviral DNA.
  • a composition is also provided for inactivating proviral DNA. Delivery of the CRISPR-associated endonuclease and gRNAs can be by various expression vectors, such as plasmid vectors, lentiviral vectors, adenoviral vectors, or adeno-associated virus vectors.
  • Viruses replicate by one of two cycles, either the lytic cycle or the lysogenic cycle.
  • the lytic cycle first the virus penetrates a host cell and releases its own nucleic acid.
  • the host cell's metabolic machinery is used to replicate the viral nucleic acid and accumulate the virus within the host cell. Once enough virions are produced within the host cell, the host cell bursts (lysis) and the virions go on to infect additional cells. Lytic viruses can integrate viral DNA into the host genome as well as be non-integrated where lysis does not occur over the period of the infection of the cell.
  • Lytic viruses include John Cunningham virus (JCV), hepatitis A, and various herpesviruses.
  • CCV John Cunningham virus
  • hepatitis A hepatitis A
  • various herpesviruses In the lysogenic cycle, virion DNA is integrated into the host cell, and when the host cell reproduces, the virion DNA is copied into the resulting cells from cell division. In the lysogenic cycle, the host cell does not burst.
  • Lysogenic viruses include hepatitis B, Zika virus, and HIV. Viruses such as lambda phage can switch between lytic and lysogenic cycles.
  • RNA-based RNA-targeting approach can allow temporary changes that can be adjusted up or down, and with greater specificity and functionality than existing methods for RNA interference. Specifically, it can address RNA embedded viral infections and resulting disease.
  • the study reports the identification and functional characterization of C2c2, an RNA-guided enzyme capable of targeting and degrading RNA.
  • C2c2 the first naturally-occurring CRISPR system that targets only RNA to have been identified, discovered by this collaborative group in October 2015— helps protect bacteria against viral infection. They demonstrate that C2c2 can be programmed to cleave particular RNA sequences in bacterial cells, which would make it an important addition to the molecular biology toolbox.
  • the RNA-focused action of C2c2 complements the CRISPR-Cas9 system, which targets DNA, the genomic blueprint for cellular identity and function.
  • the ability to target only RNA which helps carry out the genomic instructions, offers the ability to specifically manipulate RNA in a high-throughput manner— and manipulate gene function more broadly. This has the potential to accelerate progress to understand, treat and prevent disease.
  • compositions can be used to target RNA, such as siRNA/miRNA/shRNA/RNAi which do not use a nuclease based mechanism, and therefore one or more are utilized for the degradative silencing on viral RNA transcripts (non-coding or coding).
  • CRISPR enzymes In using CRISPR enzymes in therapeutics, it is important that the enzymes have specificity and not generate off-target effects, such as by cutting or mutating the wrong target. Off-target effects, even with low frequency of occurance, can lead to genetic instability and disruption of gene function in normal genes. Human genetic variability can also alter the enzyme specificity.
  • Several methods have been used to improve specificity of CRISPR enzymes.
  • the PAM and sgRNA used in CRISPR are involved in specificity, and it has been found that the nucleotides directly before the PAM can affect specificity. It has been found that adding two guanosines to the 5' end of sgRNA as well as truncated sgRNAs can increase specificity.
  • dCas9-Fokl fusion proteins have also been used to increase specifity. It has also been suggested that the exposure time of a subject's cells to enzyme activity be controlled. The exposure time can be controlled through several methods: 1) the addition of a nuclease inhibitor, or 2) controlled expression of the therapeutic nuclease or gRNAs from a regulated promoter (regulated by an antibiotic like tetracycline for example - in the presence/absence of tetracycline the expression of the nuclease/gRNAs can be turned on or off).
  • the drawback for the inhibitor approach is that it adds an extra step to the therapeutic process and much more experimentation would be required to show that the inhibitor itself is safe to use in humans, and also in combination with the therapeutic nuclease/gRNA.
  • the drawback for the tetracycline (or other small molecule-type 'switch') approaches is that tetracycline would need to be taken along with the therapeutic nuclease/gRNA deliverable plasmid. Dosing would be difficult to determine on a per patient basis. These methods do not adequately solve the problems of off-target effects. [00013] There remains a need for additional CRISPR enzymes for use in gene editing that can effectively target virus DNA or RNA. There also remains a need for CRISPR enzymes that have improved specificity with a target virus.
  • the present invention provides for a composition for treating a lysogenic virus including a vector encoding two or more gene editors chosen from the group consisting of gene editors that target viral DNA, gene editors that target viral RNA, and combinations thereof, wherein the gene editor that targets viral DNA includes at least two gRNAs having at least one modified nucleic acid.
  • the present invention also provides for a composition for treating a lytic virus, including a vector encoding isolated nucleic acid encoding at least one gene editor that targets viral DNA and a viral RNA targeting composition, wherein the at least one gene editor that targets viral DNA includes at least two gRNAs having at least one modified nucleic acid.
  • the present invention also provides for a composition for treating both lysogenic and lytic viruses, including a vector encoding isolated nucleic acid encoding two or more gene editors that target viral RNA, chosen from the group consisting of CRISPR-associated nucleases, Argonaute endonuclease gDNAs, C2c2, C2cl, c2c3, RNase P RNA, and combinations thereof, wherein the at two or more gene editors that target viral RNA include at least two gRNAs having at least one modified nucleic acid.
  • the present invention provides for a composition for treating lytic viruses, including a vector encoding isolated nucleic acid encoding two or more gene editors that target viral RNA and a viral RNA targeting composition, wherein the at two or more gene editors that target viral RNA include at least two gRNAs having at least one modified nucleic acid.
  • the present invention also provides for a method of increasing specificity of gene editors in treating an individual for a virus by modifying at least one nucleic acid of at least one gRNA in a gene editor composition, administering the gene editor composition to an individual having a virus, and increasing the specificity of the gene editor to a target in the virus.
  • the present invention provides for a method of treating a lysogenic virus, by administering a composition including a vector encoding isolated nucleic acid encoding two or more gene editors chosen from the group consisting of gene editors that target viral DNA, gene editors that target viral RNA, and combinations thereof to an individual having a lysogenic virus, wherein the gene editors that target viral DNA include at least two gRNAs having at least one modified nucleic acid, and inactivating the lysogenic virus.
  • the present invention also provides for a method for treating a lytic virus, by administering a composition including a vector encoding isolated nucleic acid encoding at least one gene editor that targets viral DNA and a viral RNA targeting composition to an individual having a lytic virus, wherein the gene editor that targets viral DNA includes at least two gRNAs having at least one modified nucleic acid, and inactivating the lytic virus.
  • the present invention also provides for a method for treating both lysogenic and lytic viruses, by administering a composition including a vector encoding isolated nucleic acid encoding two or more gene editors that target viral RNA, chosen from the group consisting of CRISPR-associated nucleases, Argonaute endonuclease gDNAs, C2c2, RNase P RNA, and combinations thereof to an individual having a lysogenic virus and lytic virus, wherein the gene editor that targets viral RNA includes at least two gRNAs having at least one modified nucleic acid, and inactivating the lysogenic virus and lytic virus.
  • a composition including a vector encoding isolated nucleic acid encoding two or more gene editors that target viral RNA, chosen from the group consisting of CRISPR-associated nucleases, Argonaute endonuclease gDNAs, C2c2, RNase P RNA, and combinations thereof to an individual having a lysogenic virus and lytic virus, wherein the
  • the present invention provides for a method for treating lytic viruses, by administering a composition including a vector encoding isolated nucleic acid encoding two or more gene editors that target viral RNA and a viral RNA targeting composition to an individual having a lytic virus, wherein the gene editor that targets viral RNA includes at least two gRNAs having at least one modified nucleic acid, and inactivating the lytic virus.
  • the present invention provides for a method of treating lysogenic viruses, by administering a composition including a vector encoding isolated nucleic acid encoding a Cas9 nuclease that is engineered to prevent off-target effects and at least two gRNAs having at least one modified nucleic acid, and inactivating the lysogenic virus.
  • FIGURE 1 is a picture of lytic and lysogenic virus within a cell and at which point CRISPR Cas9 can be used and at which point RNA targeting systems can be used;
  • FIGURE 2 is a chart of various Archaea Cas9 effectors, CasY.l - CasY.6 effectors, and CasX effectors of the present invention.
  • FIGURE BA is a representation of unmodified RNA
  • FIGURE 3B is a representation of LNA
  • FIGURE 3C is a representation of BNA nc .
  • the present invention is generally directed to compositions and methods for treating lysogenic and lytic viruses with various gene editing systems and enzyme effectors.
  • the compositions can treat both lysogenic viruses and lytic viruses, or optionally viruses that use both methods of replication.
  • the compositions preferably include nucleic acid modifications that increase specificity to a target viral genome, such as bridged nucleic acids, further described below.
  • the nucleic acid modifications to the gRNA allow for tighter and therefore more specific binding of the nuclease to its target sequence, thereby offering more flexibility to additional viral genetic targets that would otherwise not be considered. The modifications are unexpected because the gRNAs are designed and modified chemically to increase specificity and reduce off-target effects.
  • the term "vector” includes cloning and expression vectors, as well as viral vectors and integrating vectors.
  • An "expression vector” is a vector that includes a regulatory region. Vectors are also further described below.
  • lentiviral vector includes both integrating and non-integrating lentiviral vectors.
  • Viruses replicate by one of two cycles, either the lytic cycle or the lysogenic cycle.
  • the lytic cycle first the virus penetrates a host cell and releases its own nucleic acid.
  • the host cell's metabolic machinery is used to replicate the viral nucleic acid and accumulate the virus within the host cell. Once enough virions are produced within the host cell, the host cell bursts (lysis) and the virions go on to infect additional cells. Lytic viruses can integrate viral DNA into the host genome as well as be non-integrated where lysis does not occur over the period of the infection of the cell.
  • Lysogenic virus refers to a virus that replicates by the lysogenic cycle (i.e. does not cause the host cell to burst and integrates viral nucleic acid into the host cell DNA).
  • the lysogenic virus can mainly replicate by the lysogenic cycle but sometimes replicate by the lytic cycle.
  • virion DNA is integrated into the host cell, and when the host cell reproduces, the virion DNA is copied into the resulting cells from cell division. In the lysogenic cycle, the host cell does not burst.
  • Lysogenic virus refers to a virus that replicates by the lytic cycle (i.e. causes the host cell to burst after an accumulation of virus within the cell).
  • the lytic virus can mainly replicate by the lytic cycle but sometimes replicate by the lysogenic cycle.
  • Nucleic acid refers to both RNA and DNA, including cDNA, genomic DNA, synthetic DNA, and DNA (or RNA) containing nucleic acid analogs, any of which may encode a polypeptide of the invention and all of which are encompassed by the invention.
  • Polynucleotides can have essentially any three-dimensional structure.
  • a nucleic acid can be double-stranded or single- stranded (i.e., a sense strand or an antisense strand).
  • Non-limiting examples of polynucleotides include genes, gene fragments, exons, introns, messenger RNA (mRNA) and portions thereof, transfer RNA, ribosomal RNA, siRNA, micro-RNA, short hairpin RNA (shRNA), interfering RNA (RNAi), ribozymes, cDNA, recombinant polynucleotides, branched polynucleotides, plasmids, vectors, isolated DNA of any sequence, isolated RNA of any sequence, nucleic acid probes, and primers, as well as nucleic acid analogs.
  • Nucleic acids can encode a fragment of a naturally occurring Cas9 or a biologically active variant thereof and at least two gRNAs where in the gRNAs are complementary to a sequence in a virus.
  • An "isolated" nucleic acid can be, for example, a naturally-occurring DNA molecule or a fragment thereof, provided that at least one of the nucleic acid sequences normally found immediately flanking that DNA molecule in a naturally-occurring genome is removed or absent.
  • an isolated nucleic acid includes, without limitation, a DNA molecule that exists as a separate molecule, independent of other sequences (e.g., a chemically synthesized nucleic acid, or a cDNA or genomic DNA fragment produced by the polymerase chain reaction (PCR) or restriction endonuclease treatment).
  • An isolated nucleic acid also refers to a DNA molecule that is incorporated into a vector, an autonomously replicating plasmid, a virus, or into the genomic DNA of a prokaryote or eukaryote.
  • an isolated nucleic acid can include an engineered nucleic acid such as a DNA molecule that is part of a hybrid or fusion nucleic acid.
  • Isolated nucleic acid molecules can be produced by standard techniques. For example, polymerase chain reaction (PCR) techniques can be used to obtain an isolated nucleic acid containing a nucleotide sequence described herein, including nucleotide sequences encoding a polypeptide described herein. PCR can be used to amplify specific sequences from DNA as well as RNA, including sequences from total genomic DNA or total cellular RNA. Various PCR methods are described in, for example, PCR Primer: A Laboratory Manual, Dieffenbach and Dveksler, eds., Cold Spring Harbor Laboratory Press, 1995.
  • sequence information from the ends of the region of interest or beyond is employed to design oligonucleotide primers that are identical or similar in sequence to opposite strands of the template to be amplified.
  • Various PCR strategies also are available by which site-specific nucleotide sequence modifications can be introduced into a template nucleic acid.
  • Isolated nucleic acids also can be chemically synthesized, either as a single nucleic acid molecule (e.g., using automated DNA synthesis in the 3' to 5' direction using phosphoramidite technology) or as a series of oligonucleotides.
  • one or more pairs of long oligonucleotides e.g., >50-100 nucleotides
  • each pair containing a short segment of complementarity e.g., about 15 nucleotides
  • DNA polymerase is used to extend the oligonucleotides, resulting in a single, double-stranded nucleic acid molecule per oligonucleotide pair, which then can be ligated into a vector.
  • Isolated nucleic acids of the invention also can be obtained by mutagenesis of, e.g., a naturally occurring portion of a Cas9-encoding DNA (in accordance with, for example, the formula above).
  • Cloaked refers to a gene editing composition that has been modified or altered chemically at immunogenic sites to prevent inducing an immunogenic response when administered. Cloaking can include changing proteins, DNA sequences, or RNA sequences.
  • the cloaked gene editors can include introducing glycosylation, and eliminating oxidative sites ((IFN -la includes more glycosylation than IRNb-lb which has increased immunogenicity, Ratanji, et al. J Immunotoxicol, 2014 Apr 11(2):99-109).
  • Cloaking gene editors can further include removing or changing proteins that generate non-natural amino acids, such as isoaspartic acid, selenocysteine, or pyrrolysine.
  • gRNA refers to guide RNA.
  • the gRNAs in the CRISPR Cas9 systems and other CRISPR nucleases herein are used for the excision of viral genome segments and hence the crippling disruption of the virus' capability to replicate/produce protein. This is accomplished by using two or more specifically designed gRNAs to avoid the issues seen with single gRNAs such as viral escape or mutations.
  • the gRNA can be a sequence complimentary to a coding or a non-coding sequence and can be tailored to the particular virus to be targeted.
  • the gRNA can be a sequence complimentary to a protein coding sequence, for example, a sequence encoding one or more viral structural proteins, (e.g., gag, pol, env and tat).
  • the gRNA sequence can be a sense or anti-sense sequence. It should be understood that when a gene editor composition is administered herein, preferably this includes two or more gRNAs.
  • the gRNAs used in the present invention preferably include various modified nucleic acids that enhance the specificity of the gene editing composition.
  • Cromwell, et al. incorporation of bridged nucleic acids into CRISPR RNAs improves Cas9 endonuclease specificity, Nature Communications 9:1448 (2018)) showed that incorporation of next-generation bridged nucleic acids (2',4'-BNA NC [N-Me]) and locked nucleic acids (LNA) at specific locations in CRISPR-RNAs (crRNAs) broadly reduced off-target DNA cleavage by Cas9 in vitro and in cells by several orders of magnitude.
  • next-generation bridged nucleic acids (2',4'-BNA NC [N-Me]
  • LNA locked nucleic acids
  • the gRNA of the present invention can include one or more bridged nucleic acids to increase their specificity.
  • the bridged nucleic acids can be locked nucleic acids (LNAs) that are conformationally restricted RNA nucleotides in which the 2' oxygen in the ribose forms a covalent bond to the 4' carbon, inducing N-type (CB'-endo) sugar puckering and a preference for an A-form helix.
  • LNAs locked nucleic acids
  • CB'-endo N-type sugar puckering
  • the LNAs have better base stacking and thermal stability compared to RNA and this provides high efficiency in binding and improved mismatch discrimination.
  • the bridged nucleic acids can also be N-methyl substituted (2',4'-BNA NC [N-Me]) to provide greater conformational flexibility and nuclease resistance, as well as less toxicity as compared to LNAs.
  • a representation of unmodified RNA is shown in FIGURE BA, an example of LNA is shown in FIGURE 3B, and an example of BNA NC is shown in FIGURE 3C.
  • the bridged nucleic acids can be located at any suitable site in the gRNA.
  • the bridged nucleic acids can be located at sites in the gRNA that are associated with mismatches, and the sites can be particular to the gRNA being used.
  • One, two, three, four, or more bridged nucleic acids can be incorporated into the gRNAs.
  • the gRNA of the present invention can also or alternatively include chemical modifications such as with 2'-fluoro-ribose or 2' -O-methyl 3' phosphorothioate (MS), or any other modification that can increase the specificity and decrease off-targeting effects.
  • the gRNAs including modified nucleic acids can be used with any of the gene editing nucleases further described below, such as Argonaute proteins, RNase P RNA, C2cl, C2c2, C2c3, Cas9, Cpfl, TevCas9, Archaea Cas9, CasY.l, CasY.2, CasY.3, CasY.4, CasY.5, CasY.6, and CasX.
  • Argonaute protein refers to proteins of the PIWI protein superfamily that contain a PIWI (P element-induced wimpy testis) domain, a MID (middle) domain, a PAZ (Piwi- Argonaute-Zwille) domain and an N-terminal domain.
  • Argonaute proteins are capable of binding small RNAs, such as microRNAs, small interfering RNAs (siRNAs), and Piwi-interacting RNAs. Argonaute proteins can be guided to target sequences with these RNAs in order to cleave mRNA, inhibit translation, or induce mRNA degradation in the target sequence.
  • Argonaute proteins There are several different human Argonaute proteins, including AGOl, AG02, AG03, and AG04 that associate with small RNAs.
  • AG02 has slicer ability, i.e. acts as an endonuclease.
  • Argonaute proteins can be used for gene editing. Endonucleases from the Argonaute protein family (from Natronobacterium gregoryi Argonaute) also use oligonucleotides as guides to degrade invasive genomes. Work by Gao et al has shown that the Natronobacterium gregoryi Argonaute (NgAgo) is a DNA-guided endonuclease suitable for genome editing in human cells.
  • Natronobacterium gregoryi Argonaute Natronobacterium gregoryi Argonaute
  • NgAgo binds 5' phosphorylatedsingle-stranded guide DNA (gDNA) of ⁇ 24 nucleotides, efficiently creates site-specific DNA double-strand breaks when loaded with the gDNA.
  • the NgAgo-gDNA system does not require a protospacer-adjacent motif (PAM), as does Cas9, and preliminary characterization suggests a low tolerance to guide-target mismatches and high efficiency in editing (G+C)-rich genomic targets.
  • the Argonaute protein endonucleases used in the present invention can also be Rhodobacter sphaeroides Argonaute (RsArgo).
  • RsArgo can provide stable interaction with target DNA strands and guide RNA, as it is able to maintain base-pairing in the 3'-region of the guide RNA between the N-terminal and PIWI domains. RsArgo is also able to specifically recognize the 5' base-U of guide RNA, and the duplex-recognition loop of the PAZ domain with guide RNA can be important in DNA silencing activity.
  • Other prokaryotic Argonaute proteins pAgos
  • the Argonaute proteins can be derived from Arabidopsis thaliana, D.
  • Argonaute proteins can also be used that are endo-nucleolytically inactive but post-translational modifications can be made to the conserved catalytic residues in order to activate them as endonucleases. Any of the above argonaute protein endonucleases can be in cloaked form.
  • Human WRN is a RecQ helicase encoded by the Werner syndrome gene. It is implicated in genome maintenance, including replication, recombination, excision repair and DNA damage response. These genetic processes and expression of WRN are concomitantly upregulated in many types of cancers. Therefore, it has been proposed that targeted destruction of this helicase could be useful for elimination of cancer cells. Reports have applied the external guide sequence (EGS) approach in directing an RNase P RNA to efficiently cleave the WRN mRNA in cultured human cell lines, thus abolishing translation and activity of this distinctive 3'-5' DNA helicase-nuclease. RNase P RNA in cloaked form is another potential endonuclease for use with the present invention.
  • EGS external guide sequence
  • C2c2 The Class 2 type Vl-A CRISPR/Cas effector "C2c2" demonstrates an RNA-guided RNase function.
  • C2c2 from the bacterium Leptotrichia shohii provides interference against RNA phage.
  • C2c2 In vitro biochemical analysis show that C2c2 is guided by a single crRNA and can be programmed to cleave ssRNA targets carrying complementary protospacers.
  • C2c2 can be programmed to knock down specific mRNAs. Cleavage is mediated by catalytic residues in the two conserved HEPN domains, mutations in which generate catalytically inactive RNA-binding proteins.
  • RNA-focused action of C2c2 complements the CRISPR-Cas9 system, which targets DNA, the genomic blueprint for cellular identity and function.
  • the ability to target only RNA, which helps carry out the genomic instructions, offers the ability to specifically manipulate RNA in a high-throughput manner— and manipulate gene function more broadly.
  • C2cl Another Class 2 type V-B CRISPR/Cas effector "C2cl” can also be used in the present invention for editing DNA.
  • C2cl contains RuvC-like endonuclease domains related distantly to Cpfl (described below).
  • C2cl can target and cleave both strands of target DNA site-specifically. According to Yang, et al.
  • C2cl- mediated cleavage results in a staggered seven-nucleotide break of target DNA
  • crRNA adopts a pre ordered five-nucleotide A-form seed sequence in the binary complex, with release of an inserted tryptophan, facilitating zippering up of 20-bp guide RNA:target DNA heteroduplex on ternary complex formation, and that the PAM-interacting cleft adopts a "locked" conformation on ternary complex formation.
  • C2cl can be in a cloaked form.
  • C2c3 is a gene editor effecor of type V-C that is distantly related to C2cl, and also contains RuvC-like nuclease domains. C2c3 is also similar to the CasY.l - CasY.6 group described below. C2c3 can be in a cloaked form.
  • CRISPR Cas9 refers to Clustered Regularly Interspaced Short Palindromic Repeat (CRISPR)-associated endonuclease Cas9.
  • CRISPR/Cas loci encode RNA-guided adaptive immune systems against mobile genetic elements (viruses, transposable elements and conjugative plasmids).
  • Three types (l-lll) of CRISPR systems have been identified.
  • CRISPR clusters contain spacers, the sequences complementary to antecedent mobile elements.
  • CRISPR clusters are transcribed and processed into mature CRISPR (Clustered Regularly Interspaced Short Palindromic Repeats) RNA (crRNA).
  • the CRISPR-associated endonuclease belongs to the type II CRISPR/Cas system and has strong endonuclease activity to cut target DNA.
  • Cas9 is guided by a mature crRNA that contains about 20 base pairs (bp) of unique target sequence (called spacer) and a trans-activated small RNA (tracrRNA) that serves as a guide for ribonuclease Ill-aided processing of pre-crRNA.
  • spacer unique target sequence
  • tracrRNA trans-activated small RNA
  • the crRNA:tracrRNA duplex directs Cas9 to target DNA via complementary base pairing between the spacer on the crRNA and the complementary sequence (called protospacer) on the target DNA.
  • Cas9 recognizes a trinucleotide (NGG) protospacer adjacent motif (PAM) to specify the cut site (the 3rd nucleotide from PAM).
  • the crRNA and tracrRNA can be expressed separately or engineered into an artificial fusion small guide RNA (sgRNA) via a synthetic stem loop (AGAAAU) to mimic the natural crRNA/tracrRNA duplex.
  • sgRNA like shRNA, can be synthesized or in vitro transcribed for direct RNA transfection or expressed from U6 or Hl-promoted RNA expression vector, although cleavage efficiencies of the artificial sgRNA are lower than those for systems with the crRNA and tracrRNA expressed separately.
  • Any of the Cas9 endonucleases can be in a cloaked form.
  • CRISPR/Cpfl is a DNA-editing technology analogous to the CRISPR/Cas9 system, characterized in 2015 by Feng Zhang's group from the Broad Institute and MIT.
  • Cpfl is an RNA-guided endonuclease of a class II CRISPR/Cas system. This acquired immune mechanism is found in Prevotella and Francisella bacteria. It prevents genetic damage from viruses.
  • Cpfl genes are associated with the
  • CRISPR locus coding for an endonuclease that use a guide RNA to find and cleave viral DNA.
  • Cpfl is a smaller and simpler endonuclease than Cas9, overcoming some of the CRISPR/Cas9 system limitations.
  • CRISPR/Cpfl could have multiple applications, including treatment of genetic illnesses and degenerative conditions.
  • Agonaute is another potential gene editing system. Cpfl can be in a cloaked form.
  • a CRISPR/TevCas9 system can also be used.
  • CRISPR/Cas9 DNA repair systems in the cells of an organism will repair the site of the cut.
  • the TevCas9 enzyme was developed to cut DNA at two sites of the target so that it is harder for the cells' DNA repair systems to repair the cuts (Wolfs, et al., Biasing genome-editing events toward precise length deletions with an RNA-guided TevCas9 dual nuclease, PNAS, doi:10.1073).
  • the TevCas9 nuclease is a fusion of a l-Tevi nuclease domain to Cas9. TevCas9 can be in a cloaked form.
  • the Cas9 nuclease can have a nucleotide sequence identical to the wild type Streptococcus pyrogenes sequence.
  • the CRISPR-associated endonuclease can be a sequence from other species, for example other Streptococcus species, such as thermophilus; Psuedomona aeruginosa, Escherichia coli, or other sequenced bacteria genomes and archaea, or other prokaryotic microorganisms.
  • the wild type Streptococcus pyrogenes Cas9 sequence can be modified.
  • the nucleic acid sequence can be codon optimized for efficient expression in mammalian cells, i.e., "humanized.”
  • a humanized Cas9 nuclease sequence can be for example, the Cas9 nuclease sequence encoded by any of the expression vectors listed in Genbank accession numbers KM099231.1 Gl:669193757; KM099232.1 Gl :669193761; or KM099233.1 Gl:669193765.
  • the Cas9 nuclease sequence can be for example, the sequence contained within a commercially available vector such as PX330 or PX260 from Addgene (Cambridge, MA).
  • the Cas9 endonuclease can have an amino acid sequence that is a variant or a fragment of any of the Cas9 endonuclease sequences of Genbank accession numbers KM099231.1 Gl:669193757; KM099232.1
  • the Cas9 nucleotide sequence can be modified to encode biologically active variants of Cas9, and these variants can have or can include, for example, an amino acid sequence that differs from a wild type Cas9 by virtue of containing one or more mutations (e.g., an addition, deletion, or substitution mutation or a combination of such mutations).
  • One or more of the substitution mutations can be a substitution (e.g., a conservative amino acid substitution).
  • a biologically active variant of a Cas9 polypeptide can have an amino acid sequence with at least or about 50% sequence identity (e.g., at least or about 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 97%, 98%, or 99% sequence identity) to a wild type Cas9 polypeptide.
  • Conservative amino acid substitutions typically include substitutions within the following groups: glycine and alanine; valine, isoleucine, and leucine; aspartic acid and glutamic acid; asparagine, glutamine, serine and threonine; lysine, histidine and arginine; and phenylalanine and tyrosine.
  • the amino acid residues in the Cas9 amino acid sequence can be non-naturally occurring amino acid residues.
  • Naturally occurring amino acid residues include those naturally encoded by the genetic code as well as non-standard amino acids (e.g., amino acids having the D-configuration instead of the L-configuration).
  • the present peptides can also include amino acid residues that are modified versions of standard residues (e.g. pyrrolysine can be used in place of lysine and selenocysteine can be used in place of cysteine).
  • Non-naturally occurring amino acid residues are those that have not been found in nature, but that conform to the basic formula of an amino acid and can be incorporated into a peptide.
  • RNA-guided endonuclease Cas9 has emerged as a versatile genome-editing platform, some have reported that the size of the commonly used Cas9 from Streptococcus pyogenes (SpCas9) limits its utility for basic research and therapeutic applications that use the highly versatile adeno-associated virus (AAV) delivery vehicle. Accordingly, the six smaller Cas9 orthologues have been used and reports have shown that Cas9 from Staphylococcus aureus (SaCas9) can edit the genome with efficiencies similar to those of SpCas9, while being more than 1 kilobase shorter. SaCas9 is 105S bp, whereas SpCas9 is 1S58 bp.
  • the Cas9 nuclease sequence can be a mutated sequence.
  • the Cas9 nuclease can be mutated in the conserved HN H and RuvC domains, which are involved in strand specific cleavage.
  • an aspartate-to- alanine (D10A) mutation in the RuvC catalytic domain allows the Cas9 nickase mutant (Cas9n) to nick rather than cleave DNA to yield single-stranded breaks, and the subsequent preferential repair through HDR can potentially decrease the frequency of unwanted indel mutations from off-target double- stranded breaks.
  • mutations of the gene editor effector sequence can minimize or prevent off-targeting.
  • the gene editor effector can also be Archaea Cas9.
  • the size of Archaea Cas9 is 950aa ARMAN 1 and 967aa ARMAN 4.
  • the Archaea Cas9 can be derived from ARMAN-1 (Condidotus Micrarchaeum acidiphilum ARMAN-1) or ARMAN-4 ( Candidatus Parvarchaeum acidiphilum ARMAN-4).
  • Two examples of Archaea Cas9 are provided in FIGURE 2, derived from ARMAN-1 and ARMAN-4.
  • the sequences for ARMAN 1 and ARMAN 4 are below.
  • the Archaea Cas9 can be in a cloaked form.
  • ARMAN 1 nucleic acid sequence
  • the gene editor effector can also be CasX, examples of which are shown in FIGURE 2.
  • CasX has a TTC PAM at the 5' end (similar to Cpfl).
  • the TTC PAM can have limitations in viral genomes that are GC rich, but not so much in those that are GC poor.
  • the size of CasX (986 bp), smaller than other type V proteins, provides the potential for four gRNA plus one siRNA in a delivery plasmid.
  • CasX can be derived from Deltaproteobacteria or Planctomycetes. The sequences for these CasX effectors are below. CasX is preferably in a cloaked form.
  • the gene editor effector can also be CasY.l - CasY.6, exam ples of which are shown in FIGURE 2.
  • CasY.l - CasY.6 has TA PAM, and a shorter PAM sequence can be useful as there are less targeting limitations.
  • the size of CasY.l - CasY.6 (1125 bp) provides the potential for two gRNA plus one siRNA or four gRNA in a delivery plasmid.
  • CasY.l - CasY.6 can be derived from phyla radiation (CPR) bacteria, such as, but not limited to, katanobacteria, vogelbacteria, parcubacteria, komeilibacteria, or kerfeldbacteria The sequences for CasY.l - CasY.6 are below. CasY.l - CasY.6 can be in a cloaked form .
  • CPR phyla radiation
  • Tev is an RNA-guided dual active site nuclease that generates two noncompatible DNA breaks at a target site, effectively deleting the majority of the target site such that it cannot be regenerated.
  • the present invention provides for a composition for treating a lysogenic virus (budding virus) including a vector encoding two or more CRISPR-associated nucleases such as Cas9, Cpfl, C2cl, C2c3, TevCas9, Archaea Cas9, CasY.l - CasY.6, and CasX gRNAs, Argonaute endonuclease gDNAs and other gene editors that target viral DNA, and RNA editors such as C2c2, or any other composition that targets RNA such as siRNA/miRNA/shRNAs/RNAi.
  • Any of the gene editor compositions include at least two gRNAs that have at least one modified nucleic acid as described above.
  • the composition includes isolated nucleic acid encoding a CRISPR-associated endonuclease (Cas9 or any other described above) and two or more gRNAs that are complementary to a target sequence in a lysogenic virus.
  • Each gRNA can be complimentary to a different sequence within the lysogenic virus.
  • the composition removes the replication critical segment of the viral genome (DNA) (or RNA using RNA editors such as C2c2) within the genome itself and translation products using RNA editors such as C2c2.
  • the entire viral genome can be excised from the host cell infected with virus. Alternatively, additions, deletions, or mutations can be made in the genome of the virus.
  • the composition can optionally include other CRISPR or gene editing systems that target DNA.
  • the gRNAs are designed to be the most optimal in safety to provide no off target effects and no viral escape.
  • the composition can treat any virus in the tables below that are indicated as having a lysogenic replication cycle, and is especially useful for retroviruses.
  • the composition can be delivered by a vector or any other method as described below.
  • the present invention also provides for a composition for treating a lytic virus, including a vector encoding two or more CRISPR-associated nucleases such as Cas9, Cpfl, C2cl, C2c3, TevCas9, Archaea Cas9, CasY.l - CasY.6, and CasX gRNAs, Argonaute endonuclease gDNAs and other gene editors for targeting viral DNA genomes for the excision of viral genes in virus that are lysogenic and either 1) small interfering RNA (siRNA)/microRNA (miRNA), short hairpin RNA, and interfering RNA (RNAi) (for RNA interference) that target critical RNAs (viral mRNA) that translate (non-coding or coding) viral proteins involved with the formation of viral proteins and/or virions or 2) CRISPR-associated nucleases such as Cas9, Cpfl, C2cl, C2c3, TevCas9, Archa
  • the composition includes isolated nucleic acid encoding a CRISPR-associated endonuclease (Cas9), two or more gRNAs that are complementary to a target DNA sequence in a virus, and either the siRNA/miRNA/shRNAs/RNAi or CRISPR-associated nucleases such as Cas9, Cpfl, C2cl, C2c3, TevCas9, Archaea Cas9, CasY.l - CasY.6, and CasX gRNAs, Argonaute endonuclease gDNAs and other gene editors that are complementary to a target RNA sequence in the virus.
  • Each gRNA can be complimentary to a different sequence within the virus.
  • the composition can additionally include any other cloaked CRISPR or gene editing systems that target viral DNA genomes and excise segments of those genomes.
  • This co- therapeutic is useful in treating individuals infected with lytic viruses that Cas9 systems alone cannot treat.
  • lytic and lysogenic viruses need to be treated in different ways.
  • CRISPR Cas9 is usually used to target DNA
  • this gene editing system can be designed to target RNA within the virus instead in order to target lytic viruses.
  • Nelles, et al. shows that RNA-targeting Cas9 was able to bind mRNAs. Any of the lytic viruses listed in the tables below can be targeted with this composition.
  • the composition can be delivered by a vector or any other method as described below.
  • the siRNA and C2c2 in the compositions herein are targeted to a particular gene in a virus or gene mRNA.
  • the siRNA can have a first strand of a duplex substantially identical to the nucleotide sequence of a portion of the viral gene or gene mRNA sequence.
  • the second strand of the siRNA duplex is complementary to both the first strand of the siRNA duplex and to the same portion of the viral gene mRNA.
  • Isolated siRNA can include short double-stranded RNA from about 17 nucleotides to about 29 nucleotides in length, preferably from about 19 to about 25 nucleotides in length, that are targeted to the target mRNA.
  • the siRNA's comprise a sense RNA strand and a complementary antisense RNA strand annealed together by standard Watson-Crick base-pairing interactions.
  • the sense strand comprises a nucleic acid sequence which is substantially identical to a target sequence contained within the target mRNA.
  • the siRNA of the invention can be obtained using a number of techniques known to those of skill in the art.
  • the siRNA can be chemically synthesized or recombinantly produced using methods known in the art, such as the Drosophila in vitro system described in U.S. published application 2002/0086B56 of Tuschl et al., the entire disclosure of which is herein incorporated by reference.
  • the siRNA of the invention are chemically synthesized using appropriately protected ribonucleoside phosphoramidites and a conventional DNA/RNA synthesizer.
  • the siRNA can be synthesized as two separate, complementary RNA molecules, or as a single RNA molecule with two complementary regions.
  • Commercial suppliers of synthetic RNA molecules or synthesis reagents include Proligo (Hamburg, Germany), Dharmacon Research (Lafayette, Colo., USA), Pierce Chemical (part of Perbio Science, Rockford, III., USA), Glen Research (Sterling, Va., USA), ChemGenes (Ashland, Mass., USA) and Cruachem (Glasgow, UK).
  • siRNA can also be expressed from recombinant circular or linear DNA plasmids using any suitable promoter.
  • suitable promoters for expressing siRNA of the invention from a plasmid include, for example, the U6 or HI RNA pol III promoter sequences and the cytomegalovirus promoter. Selection of other suitable promoters is within the skill in the art.
  • the recombinant plasmids of the invention can also comprise inducible or regulatable promoters for expression of the siRNA in a particular tissue or in a particular intracellular environment.
  • the siRNA expressed from recombinant plasmids can either be isolated from cultured cell expression systems by standard techniques, or can be expressed intracellularly.
  • siRNA of the invention can be expressed from a recombinant plasmid either as two separate, complementary RNA molecules, or as a single RNA molecule with two complementary regions.
  • siRNA can be useful in targeting JC Virus, BKV, or SV40 polyomaviruses (U.S. Patent Application Publication No. 2007/0249552 to Khalili, et al.), wherein siRNA is used which targets JCV agnoprotein gene or large T antigen gene mRNA and wherein the sense RNA strand comprises a nucleotide sequence substantially identical to a target sequence of about 19 to about 25 contiguous nucleotides in agnoprotein gene or large T antigen gene mRNA.
  • the present invention also provides for a composition for treating both lysogenic and lytic viruses, including a vector encoding two or more CRISPR-associated nucleases such as Cas9, Cpfl, C2cl, C2c3, TevCas9, Archaea Cas9, CasY.l - CasY.6, and CasX gRNAs, Argonaute endonuclease gDNAs, C2c2, C2cl, and other gene editors that target viral RNA.
  • Any of the gene editor compositions include at least two gRNAs that have at least one modified nucleic acid as described above.
  • the composition includes isolated nucleic acid encoding a CRISPR-associated endonuclease (Cas9) and two or more gRNAs that are complementary to a target RNA sequence in a virus.
  • Each gRNA can be complimentary to a different sequence within the virus.
  • the composition can additionally include any other CRISPR or gene editing systems that target viral RNA genomes and excise segments of those genomes. This composition can target viruses that have both lysogenic and lytic replication, as listed in the tables below.
  • the present invention provides for a composition for treating lytic viruses, including a vector encoding two or more CRISPR-associated nucleases such as Cas9, Cpfl, C2cl, C2c3, TevCas9, Archaea Cas9, CasY.l - CasY.6, and CasX gRNAs, Argonaute endonuclease gDNAs and other gene editors and siRNA/miRNAs/shRNAs/RNAi (RNA interference) that target critical RNAs (viral mRNA) that translate (non-coding or coding) viral proteins involved with the formation of viral proteins and/or virions.
  • CRISPR-associated nucleases such as Cas9, Cpfl, C2cl, C2c3, TevCas9, Archaea Cas9, CasY.l - CasY.6, and CasX gRNAs
  • Argonaute endonuclease gDNAs and other gene editors and siRNA
  • any of the gene editor compositions include at least two gRNAs that have at least one modified nucleic acid as described above.
  • the composition includes isolated nucleic acid encoding a CRISPR- associated endonuclease (Cas9 or any other described above) and two or more gRNAs that are complementary to a target RNA sequence in a lytic virus.
  • Each gRNA can be complimentary to a different sequence within the lytic virus.
  • the composition can optionally include other CRISPR or gene editing systems that target viral RNA genomes and excise segments of those genomes for disruption in lytic viruses.
  • compositions and methods of the present invention can be targeted by the compositions and methods of the present invention. Depending on whether they are lytic or lysogenic, different compositions and methods can be used as appropriate.
  • TABLE 2 lists viruses in the picornaviridae/hepeviridae/flaviviridae families and their method of replication.
  • the composition particularly useful in treating Hepatitis D is one that targets Hepatitis B as well, such as two or more CRISPR-associated nucleases such as Cas9, Cpfl, C2cl, C2c3, TevCas9, Archaea Cas9, CasY.l - CasY.6, and CasX gRNAs, Argonaute endonuclease gDNAs and other gene editors to treat the lysogenic virus and siRNAs/miRNAs/shRNAs/RNAi to treat the lytic virus.
  • CRISPR-associated nucleases such as Cas9, Cpfl, C2cl, C2c3, TevCas9, Archaea Cas9, CasY.l - CasY.6, and CasX gRNAs
  • Argonaute endonuclease gDNAs and other gene editors to treat the lysogenic virus and siRNAs/miRNAs/shRNAs/RNAi to treat
  • TABLE 3 lists viruses in the herpesviridae family and their method of replication.
  • TABLE 4 lists viruses in the orthomyxoviridae family and their method of replication.
  • TABLE 5 lists viruses in the retroviridae family and their method of replication.
  • TABLE 6 lists viruses in the papillomaviridae family and their method of replication.
  • TABLE 8 lists viruses in the reoviridae family and their method of replication.
  • TABLE 9 lists viruses in the rhabdoviridae family and their method of replication.
  • TABLE 10 lists viruses in the bunyanviridae family and their method of replication.
  • TABLE 11 lists viruses in the arenaviridae family and their method of replication.
  • TABLE 12 lists viruses in the filoviridae family and their method of replication.
  • compositions of the present invention can be used to treat either active or latent viruses.
  • the compositions of the present invention can be used to treat individuals in which latent virus is present but the individual has not yet presented symptoms of the virus.
  • the compositions can target virus in any cells in the individual, such as, but not limited to, CD4+ lymphocytes, macrophages, fibroblasts, monocytes, T lymphocytes, B lymphocytes, natural killer cells, dendritic cells such as
  • Langerhans cells and follicular dendritic cells hematopoietic stem cells, endothelial cells, brain microglial cells, and gastrointestinal epithelial cells.
  • the CRISPR endonuclease when any of the compositions are contained within an expression vector, can be encoded by the same nucleic acid or vector as the gRNA sequences. Alternatively or in addition, the CRISPR endonuclease can be encoded in a physically separate nucleic acid from the gRNA sequences or in a separate vector. It should be understood that because the gRNAs in the present invention are chemically modified, and then generally desalted and purified using HPLC, they may not necessarily be expressed from the same therapeutic plasmid that encodes the nuclease. Therefore, the BNA/LNA/other modified gRNAs may be delivered 'off-plasmid' or separately (packaged separately). However, with appropriate enzymes, the nucleases and gRNAs can also be included in the same plasmid.
  • vectors containing nucleic acids such as those described herein also are provided.
  • a “vector” is a replicon, such as a plasmid, phage, or cosmid, into which another DNA segment may be inserted so as to bring about the replication of the inserted segment.
  • a vector is capable of replication when associated with the proper control elements.
  • Suitable vector backbones include, for example, those routinely used in the art such as plasmids, viruses, artificial chromosomes, BACs, YACs, or PACs.
  • the term “vector” includes cloning and expression vectors, as well as viral vectors and integrating vectors.
  • An “expression vector” is a vector that includes a regulatory region.
  • the vectors provided herein also can include, for example, origins of replication, scaffold attachment regions (SARs), and/or markers.
  • a marker gene can confer a selectable phenotype on a host cell.
  • a marker can confer biocide resistance, such as resistance to an antibiotic (e.g., kanamycin, G418, bleomycin, or hygromycin).
  • an expression vector can include a tag sequence designed to facilitate manipulation or detection (e.g., purification or localization) of the expressed polypeptide.
  • Tag sequences such as green fluorescent protein (GFP), glutathione S- transferase (GST), polyhistidine, c-myc, hemagglutinin, or FlagTM tag (Kodak, New Haven, CT) sequences typically are expressed as a fusion with the encoded polypeptide.
  • GFP green fluorescent protein
  • GST glutathione S- transferase
  • polyhistidine polyhistidine
  • c-myc hemagglutinin
  • hemagglutinin or FlagTM tag (Kodak, New Haven, CT) sequences
  • FlagTM tag Kodak, New Haven, CT sequences
  • Additional expression vectors also can include, for example, segments of chromosomal, non-chromosomal and synthetic DNA sequences.
  • Suitable vectors include derivatives of SV40 and known bacterial plasmids, e.g., E.
  • phage DNAs e.g., the numerous derivatives of phage 1, e.g., NM989, and other phage DNA, e.g., M13 and filamentous
  • Yeast expression systems can also be used.
  • the non-fusion pYES2 vector [Xba ⁇ , Sph ⁇ , Sho ⁇ , Not ⁇ , GstXI, EcoR ⁇ , SstXI, Bam l, Sod, Kpnl, and Hind ⁇ cloning sites; Invitrogen) or the fusion pYESHisA, B, C [Xba ⁇ , Sph ⁇ , Sho ⁇ , Not ⁇ , SstXI, fcoRI, Bam l, Sod, Kpn ⁇ , and Hind ⁇ cloning sites, N- terminal peptide purified with ProBond resin and cleaved with enterokinase; Invitrogen), to mention just two, can be employed according to the invention.
  • a yeast two-hybrid expression system can also be prepared in accordance with the invention.
  • the vector can also include a regulatory region.
  • regulatory region refers to nucleotide sequences that influence transcription or translation initiation and rate, and stability and/or mobility of a transcription or translation product. Regulatory regions include, without limitation, promoter sequences, enhancer sequences, response elements, protein recognition sites, inducible elements, protein binding sequences, 5' and 3' untranslated regions (UTRs), transcriptional start sites, termination sequences, polyadenylation sequences, nuclear localization signals, and introns.
  • operably linked refers to positioning of a regulatory region and a sequence to be transcribed in a nucleic acid so as to influence transcription or translation of such a sequence.
  • the translation initiation site of the translational reading frame of the polypeptide is typically positioned between one and about fifty nucleotides downstream of the promoter.
  • a promoter can, however, be positioned as much as about 5,000 nucleotides upstream of the translation initiation site or about 2,000 nucleotides upstream of the transcription start site.
  • a promoter typically comprises at least a core (basal) promoter.
  • a promoter also may include at least one control element, such as an enhancer sequence, an upstream element or an upstream activation region (UAR).
  • control element such as an enhancer sequence, an upstream element or an upstream activation region (UAR).
  • the choice of promoters to be included depends upon several factors, including, but not limited to, efficiency, selectability, inducibility, desired expression level, and cell- or tissue-preferential expression. It is a routine matter for one of skill in the art to modulate the expression of a coding sequence by appropriately selecting and positioning promoters and other regulatory regions relative to the coding sequence.
  • Vectors include, for example, viral vectors (such as adenoviruses (“Ad”), adeno-associated viruses (AAV), and vesicular stomatitis virus (VSV) and retroviruses), liposomes and other lipid- containing complexes, and other macromolecular complexes capable of mediating delivery of a polynucleotide to a host cell.
  • viral vectors such as adenoviruses (“Ad"), adeno-associated viruses (AAV), and vesicular stomatitis virus (VSV) and retroviruses
  • liposomes and other lipid- containing complexes such as liposomes and other lipid- containing complexes
  • other macromolecular complexes capable of mediating delivery of a polynucleotide to a host cell.
  • Vectors can also comprise other components or functionalities that further modulate gene delivery and/or gene expression, or that otherwise provide beneficial properties to the targeted cells.
  • such other components include, for example, components that influence binding or targeting to cells (including components that mediate cell-type or tissue-specific binding); components that influence uptake of the vector nucleic acid by the cell; components that influence localization of the polynucleotide within the cell after uptake (such as agents mediating nuclear localization); and components that influence expression of the polynucleotide.
  • Such components also might include markers, such as detectable and/or selectable markers that can be used to detect or select for cells that have taken up and are expressing the nucleic acid delivered by the vector.
  • Such components can be provided as a natural feature of the vector (such as the use of certain viral vectors which have components or functionalities mediating binding and uptake), or vectors can be modified to provide such functionalities.
  • Other vectors include those described by Chen et al; BioTechniques, 34: 167-171 (2003). A large variety of such vectors are known in the art and are generally available.
  • a "recombinant viral vector” refers to a viral vector comprising one or more heterologous gene products or sequences. Since many viral vectors exhibit size-constraints associated with packaging, the heterologous gene products or sequences are typically introduced by replacing one or more portions of the viral genome. Such viruses may become replication-defective, requiring the deleted function(s) to be provided in trans during viral replication and encapsidation (by using, e.g., a helper virus or a packaging cell line carrying gene products necessary for replication and/or encapsidation).
  • Suitable nucleic acid delivery systems include recombinant viral vector, typically sequence from at least one of an adenovirus, adenovirus-associated virus (AAV), helper-dependent adenovirus, retrovirus, or hemagglutinating virus of Japan-liposome (HVJ) complex.
  • the viral vector comprises a strong eukaryotic promoter operably linked to the polynucleotide e.g., a cytomegalovirus (CMV) promoter.
  • CMV cytomegalovirus
  • the recombinant viral vector can include one or more of the polynucleotides therein, preferably about one polynucleotide.
  • the viral vector used in the invention methods has a pfu (plague forming units) of from about 10 s to about 5x 10 10 pfu.
  • pfu plaque forming units
  • use of between from about 0.1 nanograms to about 4000 micrograms will often be useful e.g., about 1 nanogram to about 100 micrograms.
  • Retroviral vectors include Moloney murine leukemia viruses and HIV-based viruses.
  • One HIV-based viral vector comprises at least two vectors wherein the gag and pol genes are from an HIV genome and the env gene is from another virus.
  • DNA viral vectors include pox vectors such as orthopox or avipox vectors, herpesvirus vectors such as a herpes simplex I virus (HSV) vector [Geller, A. I. et al., J. Neurochem, 64: 487 (1995); Lim, F., et al., in DNA Cloning: Mammalian Systems, D. Glover, Ed. (Oxford Univ.
  • HSV herpes simplex I virus
  • Pox viral vectors introduce the gene into the cells cytoplasm.
  • Avipox virus vectors result in only a short term expression of the nucleic acid.
  • Adenovirus vectors, adeno-associated virus vectors and herpes simplex virus (HSV) vectors may be an indication for some invention embodiments.
  • the adenovirus vector results in a shorter term expression (e.g., less than about a month) than adeno- associated virus, in some embodiments, may exhibit much longer expression.
  • the particular vector chosen will depend upon the target cell and the condition being treated. The selection of appropriate promoters can readily be accomplished.
  • An example of a suitable promoter is the 763-base-pair cytomegalovirus (CMV) promoter.
  • Suitable promoters which may be used for gene expression include, but are not limited to, the Rous sarcoma virus (RSV) (Davis, et al., Hum Gene Ther 4:151 (1993)), the SV40 early promoter region, the herpes thymidine kinase promoter, the regulatory sequences of the metallothionein (MMT) gene, prokaryotic expression vectors such as the b-lactamase promoter, the tac promoter, promoter elements from yeast or other fungi such as the GAL4 promoter, the ADH (alcohol dehydrogenase) promoter, PGK (phosphoglycerol kinase) promoter, alkaline phosphatase promoter; and the animal transcriptional control regions, which exhibit tissue specificity and have been utilized in transgenic animals: elastase I gene control region which is active in pancreatic acinar cells, insulin gene control region which is active in pancreatic beta cells, immunoglobulin gene control region
  • Certain proteins can expressed using their native promoter.
  • Other elements that can enhance expression can also be included such as an enhancer or a system that results in high levels of expression such as a tat gene and tar element.
  • This cassette can then be inserted into a vector, e.g., a plasmid vector such as, pUC19, pUC118, pBR322, or other known plasmid vectors, that includes, for example, an E. coli origin of replication. See, Sambrook, et al., Molecular Cloning: A Laboratory Manual, Cold Spring Harbor Laboratory press, (1989).
  • the plasmid vector may also include a selectable marker such as the b-lactamase gene for ampicillin resistance, provided that the marker polypeptide does not adversely affect the metabolism of the organism being treated.
  • the cassette can also be bound to a nucleic acid binding moiety in a synthetic delivery system, such as the system disclosed in WO 95/22618.
  • the polynucleotides of the invention can also be used with a microdelivery vehicle such as cationic liposomes and adenoviral vectors.
  • a microdelivery vehicle such as cationic liposomes and adenoviral vectors.
  • Replication-defective recombinant adenoviral vectors can be produced in accordance with known techniques. See, Quantin, et al., Proc. Notl. Acad. Sci. USA, 89:2581-2584 (1992); Stratford- Perricadet, et al., J. Clin. Invest., 90:626-630 (1992); and Rosenfeld, et al., Cell, 68:143-155 (1992).
  • Another delivery method is to use single stranded DNA producing vectors which can produce the expressed products intracellularly. See for example, Chen et al, BioTechniques, 34: 167-171 (2003), which is incorporated herein, by reference, in its entirety.
  • compositions of the present invention can be prepared in a variety of ways known to one of ordinary skill in the art. Regardless of their original source or the manner in which they are obtained, the compositions of the invention can be formulated in accordance with their use.
  • the nucleic acids and vectors described above can be formulated within compositions for application to cells in tissue culture or for administration to a patient or subject.
  • Any of the pharmaceutical compositions of the invention can be formulated for use in the preparation of a medicament, and particular uses are indicated below in the context of treatment, e.g., the treatment of a subject having a virus or at risk for contracting a virus.
  • any of the nucleic acids and vectors can be administered in the form of pharmaceutical compositions.
  • compositions can be prepared in a manner well known in the pharmaceutical art, and can be administered by a variety of routes, depending upon whether local or systemic treatment is desired and upon the area to be treated. Administration may be topical (including ophthalmic and to mucous membranes including intranasal, vaginal and rectal delivery), pulmonary (e.g., by inhalation or insufflation of powders or aerosols, including by nebulizer; intratracheal, intranasal, epidermal and transdermal), ocular, oral or parenteral.
  • topical including ophthalmic and to mucous membranes including intranasal, vaginal and rectal delivery
  • pulmonary e.g., by inhalation or insufflation of powders or aerosols, including by nebulizer; intratracheal, intranasal, epidermal and transdermal
  • ocular oral or parenteral.
  • Methods for ocular delivery can include topical administration (eye drops), subconjunctival, periocular or intravitreal injection or introduction by balloon catheter or ophthalmic inserts surgically placed in the conjunctival sac.
  • Parenteral administration includes intravenous, intra-arterial, subcutaneous, intraperitoneal or intramuscular injection or infusion; or intracranial, e.g., intrathecal or intraventricular administration.
  • Parenteral administration can be in the form of a single bolus dose, or may be, for example, by a continuous perfusion pump.
  • compositions and formulations for topical administration may include transdermal patches, ointments, lotions, creams, gels, drops, suppositories, sprays, liquids, powders, and the like.
  • Conventional pharmaceutical carriers, aqueous, powder or oily bases, thickeners and the like may be necessary or desirable.
  • compositions which contain, as the active ingredient, nucleic acids and vectors described herein in combination with one or more pharmaceutically acceptable carriers.
  • pharmaceutically acceptable refer to molecular entities and compositions that do not produce an adverse, allergic or other untoward reaction when administered to an animal or a human, as appropriate.
  • the methods and compositions disclosed herein can be applied to a wide range of species, e.g., humans, non-human primates (e.g., monkeys), horses or other livestock, dogs, cats, ferrets or other mammals kept as pets, rats, mice, or other laboratory animals.
  • compositions of the invention includes any and all solvents, dispersion media, coatings, antibacterial, isotonic and absorption delaying agents, buffers, excipients, binders, lubricants, gels, surfactants and the like, that may be used as media for a pharmaceutically acceptable substance.
  • the active ingredient is typically mixed with an excipient, diluted by an excipient or enclosed within such a carrier in the form of, for example, a capsule, tablet, sachet, paper, or other container.
  • the excipient when it serves as a diluent, it can be a solid, semisolid, or liquid material (e.g., normal saline), which acts as a vehicle, carrier or medium for the active ingredient.
  • the compositions can be in the form of tablets, pills, powders, lozenges, sachets, cachets, elixirs, suspensions, emulsions, solutions, syrups, aerosols (as a solid or in a liquid medium), lotions, creams, ointments, gels, soft and hard gelatin capsules, suppositories, sterile injectable solutions, and sterile packaged powders.
  • the type of diluent can vary depending upon the intended route of administration.
  • the resulting compositions can include additional agents, such as preservatives.
  • the carrier can be, or can include, a lipid-based or polymer-based colloid.
  • the carrier material can be a colloid formulated as a liposome, a hydrogel, a microparticle, a nanoparticle, or a block copolymer micelle.
  • the carrier material can form a capsule, and that material may be a polymer-based colloid.
  • the nucleic acid sequences of the invention can be delivered to an appropriate cell of a subject. This can be achieved by, for example, the use of a polymeric, biodegradable microparticle or microcapsule delivery vehicle, sized to optimize phagocytosis by phagocytic cells such as macrophages.
  • a polymeric, biodegradable microparticle or microcapsule delivery vehicle sized to optimize phagocytosis by phagocytic cells such as macrophages.
  • PLGA poly-lacto-co-glycolide
  • the polynucleotide is encapsulated in these microparticles, which are taken up by macrophages and gradually biodegraded within the cell, thereby releasing the polynucleotide. Once released, the DNA is expressed within the cell.
  • a second type of microparticle is intended not to be taken up directly by cells, but rather to serve primarily as a slow-release reservoir of nucleic acid that is taken up by cells only upon release from the micro-particle through biodegradation.
  • These polymeric particles should therefore be large enough to preclude phagocytosis (i.e., larger than 5pm and preferably larger than 20pm).
  • Another way to achieve uptake of the nucleic acid is using liposomes, prepared by standard methods.
  • the nucleic acids can be incorporated alone into these delivery vehicles or co-incorporated with tissue-specific antibodies, for example antibodies that target cell types that are commonly latently infected reservoirs of HIV infection, for example, brain macrophages, microglia, astrocytes, and gut- associated lymphoid cells.
  • a molecular complex composed of a plasmid or other vector attached to poly-L-lysine by electrostatic or covalent forces.
  • Poly-L-lysine binds to a ligand that can bind to a receptor on target cells.
  • Delivery of "naked DNA" i.e., without a delivery vehicle) to an intramuscular, intradermal, or subcutaneous site, is another means to achieve in vivo expression.
  • nucleic acid sequence encoding the an isolated nucleic acid sequence comprising a sequence encoding a CRISPR-associated endonuclease and a guide RNA is operatively linked to a promoter or enhancer-promoter combination. Promoters and enhancers are described above.
  • compositions of the invention can be formulated as a nanoparticle, for example, nanoparticles comprised of a core of high molecular weight linear polyethylenimine (LPEI) complexed with DNA and surrounded by a shell of polyethyleneglycol-modified (PEGylated) low molecular weight LPEI.
  • LPEI high molecular weight linear polyethylenimine
  • PEGylated polyethyleneglycol-modified
  • the nucleic acids and vectors may also be applied to a surface of a device (e.g., a catheter) or contained within a pump, patch, or other drug delivery device.
  • the nucleic acids and vectors of the invention can be administered alone, or in a mixture, in the presence of a pharmaceutically acceptable excipient or carrier (e.g., physiological saline).
  • a pharmaceutically acceptable excipient or carrier e.g., physiological saline
  • the excipient or carrier is selected on the basis of the mode and route of administration.
  • Suitable pharmaceutical carriers, as well as pharmaceutical necessities for use in pharmaceutical formulations, are described in Remington's Pharmaceutical Sciences (E. W. Martin), a well-known reference text in this field, and in the USP/NF (United States Pharmacopeia and the National Formulary).
  • the present invention provides for a method of increasing specificity of gene editors in treating an individual for a virus by modifying at least one nucleic acid of at least one gRNA in a gene editor composition, administering the gene editor composition to an individual having a virus, and increasing the specificity of the gene editor to a target in the virus.
  • modifying the nucleic acid of the gRNAs can increase the specificity of the gene editor.
  • the nucleic acid can be modified to a composition of locked nucleic acid, N-methyl substituted bridged nucleic acid, 2'- fluoro-ribose, 2'-0-methyl S' phosphorothioate, or combinations thereof.
  • the gene editor can be any of Argonaute proteins, RNase P RNA, C2cl, C2c2, C2cB, Cas9, Cpfl, TevCas9, Archaea Cas9, CasY.l,
  • the virus being treated can be any virus described herein.
  • the present invention provides for a method of treating a lysogenic virus, by administering a composition including two or more CRISPR-associated nucleases such as Cas9, Cpfl, C2cl, and TevCas9 gRNAs, Argonaute endonuclease gDNAs and other gene editors that target viral DNA to an individual having a lysogenic virus wherein the gene editors that target viral DNA include at least two gRNAs having at least one modified nucleic acid, and inactivating the lysogenic virus.
  • the lysogenic virus is integrated into the genome of the host cell and the composition inactivates the lysogenic virus by excising the viral DNA from the host cell.
  • the composition can include any of the properties as described above, such as being in isolated nucleic acid, be packaged in a vector delivery system, or include other CRISPR or gene editing systems that target DNA.
  • the lysogenic virus can be any listed in the tables above.
  • treatment can be in vivo (directly administering the composition) or ex vivo (for example, a cell or plurality of cells, or a tissue explant, can be removed from a subject having an viral infection and placed in culture, and then treated with the composition).
  • ex vivo for example, a cell or plurality of cells, or a tissue explant
  • the vector can deliver the compositions to a specific cell type.
  • the invention is not so limited however, and other methods of DNA delivery such as chemical transfection, using, for example calcium phosphate, DEAE dextran, liposomes, lipoplexes, surfactants, and perfluoro chemical liquids are also contemplated, as are physical delivery methods, such as electroporation, micro injection, ballistic particles, and "gene gun" systems.
  • the amount of the compositions administered is enough to inactivate all of the virus present in the individual.
  • An individual is effectively treated whenever a clinically beneficial result ensues. This may mean, for example, a complete resolution of the symptoms of a disease, a decrease in the severity of the symptoms of the disease, or a slowing of the disease's progression.
  • the present methods may also include a monitoring step to help optimize dosing and scheduling as well as predict outcome.
  • compositions described herein can be administered to any part of the host's body for subsequent delivery to a target cell.
  • a composition can be delivered to, without limitation, the brain, the cerebrospinal fluid, joints, nasal mucosa, blood, lungs, intestines, muscle tissues, skin, or the peritoneal cavity of a mammal.
  • routes of delivery a composition can be administered by intravenous, intracranial, intraperitoneal, intramuscular, subcutaneous, /ntramuscular, intrarectal, intravaginal, intrathecal, intratracheal, intradermal, or transdermal injection, by oral or nasal administration, or by gradual perfusion over time.
  • an aerosol preparation of a composition can be given to a host by inhalation.
  • the dosage required will depend on the route of administration, the nature of the formulation, the nature of the patient's illness, the patient's size, weight, surface area, age, and sex, other drugs being administered, and the judgment of the attending clinicians. Wide variations in the needed dosage are to be expected in view of the variety of cellular targets and the differing efficiencies of various routes of administration. Variations in these dosage levels can be adjusted using standard empirical routines for optimization, as is well understood in the art. Administrations can be single or multiple (e.g., 2- or 3-, 4-, 6-, 8-, 10-, 20-, 50-, 100-, 150-, or more fold). Encapsulation of the compounds in a suitable delivery vehicle (e.g., polymeric microparticles or implantable devices) may increase the efficiency of delivery.
  • a suitable delivery vehicle e.g., polymeric microparticles or implantable devices
  • the duration of treatment with any composition provided herein can be any length of time from as short as one day to as long as the life span of the host (e.g., many years).
  • a compound can be administered once a week (for, for example, 4 weeks to many months or years); once a month (for, for example, three to twelve months or for many years); or once a year for a period of 5 years, ten years, or longer.
  • the frequency of treatment can be variable.
  • the present compounds can be administered once (or twice, three times, etc.) daily, weekly, monthly, or yearly.
  • an effective amount of any composition provided herein can be administered to an individual in need of treatment.
  • the term "effective" as used herein refers to any amount that induces a desired response while not inducing significant toxicity in the patient. Such an amount can be determined by assessing a patient's response after administration of a known amount of a particular composition.
  • the level of toxicity if any, can be determined by assessing a patient's clinical symptoms before and after administering a known amount of a particular composition. It is noted that the effective amount of a particular composition administered to a patient can be adjusted according to a desired outcome as well as the patient's response and level of toxicity. Significant toxicity can vary for each particular patient and depends on multiple factors including, without limitation, the patient's disease state, age, and tolerance to side effects.
  • the present invention also provides for a method for treating a lytic virus, including administering a vector encoding two or more CRISPR-associated nucleases such as Cas9, Cpfl, C2cl, C2c3, TevCas9, Archaea Cas9, CasY.l - CasY.6, and CasX gRNAs, Argonaute endonuclease gDNAs and other gene editors that target viral DNA and a composition chosen from siRNAs/miRNAs/shRNAs/RNAi and CRISPR-associated nucleases such as Cas9, Cpfl, C2cl, C2c3, TevCas9, Archaea Cas9, CasY.l - CasY.6, and CasX gRNAs, Argonaute endonuclease gDNAs and other gene editors that target viral RNA to an individual having a lytic virus, wherein the gene editor that targets viral DNA includes at least two g
  • the composition inactivates the lytic virus by excising the viral DNA and RNA from the host cell.
  • the composition can include any of the properties as described above, such as being in isolated nucleic acid, be packaged in a vector delivery system, or include other CRISPR or gene editing systems that target DNA.
  • the lytic virus can be any listed in the tables above.
  • the gene editor that targets viral RNA can also include at least two gRNAs having at least one modified nucleic acid.
  • the present invention also provides for a method for treating both lysogenic and lytic viruses, by administering a composition including a vector encoding two or more CRISPR-associated nucleases such as Cas9, Cpfl, C2cl, C2c3, TevCas9, Archaea Cas9, CasY.l - CasY.6, and CasX gRNAs, Argonaute endonuclease gDNAs and other gene editors that target viral RNA to an individual having a lysogenic virus and lytic virus, wherein the gene editor that targets viral RNA includes at least two gRNAs having at least one modified nucleic acid, and inactivating the lysogenic virus and lytic virus.
  • CRISPR-associated nucleases such as Cas9, Cpfl, C2cl, C2c3, TevCas9, Archaea Cas9, CasY.l - CasY.6, and CasX gRNAs
  • the composition inactivates the viruses by excising the viral RNA from the host cell.
  • the composition can include any of the properties as described above, such as being in isolated nucleic acid, or include other CRISPR or gene editing systems that target RNA.
  • the lysogenic virus and lytic virus can be any listed in the tables above.
  • RNA-based genome that is non-integrating (not converted to DNA), yet contributes to lysogenic type replication cycle.
  • the viral genome can be eliminated.
  • the approach can be utilized to also target viral mRNA which occurs downstream (as the genome is translated).
  • the present invention provides for a method for treating lytic viruses, by administering a composition including a vector encoding two or more CRISPR-associated nucleases such as Cas9, Cpfl, C2cl, C2c3, TevCas9, Archaea Cas9, CasY.l - CasY.6, and CasX gRNAs, Argonaute endonuclease gDNAs and other gene editors that target viral RNA and siRNA/miRNAs/shRNAs/RNAi that target viral RNA to an individual having a lytic virus, wherein the gene editor that targets viral RNA includes at least two gRNAs having at least one modified nucleic acid, and inactivating the lytic virus.
  • CRISPR-associated nucleases such as Cas9, Cpfl, C2cl, C2c3, TevCas9, Archaea Cas9, CasY.l - CasY.6, and CasX gRNAs
  • the composition inactivates the lytic virus by excising the viral RNA from the host cell.
  • the composition can include any of the properties as described above, such as being in isolated nucleic acid, or include other CRISPR or gene editing systems that target RNA. Two or more gene editors will be utilized that can target RNA to excise the RNA-based viral genome and/or the viral mRNA that occurs downstream. In the case of siRNA/miRNA/shRNA/RNAi which do not use a nuclease based mechanism, one or more are utilized for the degradative silencing on viral RNA transcripts (non-coding or coding)
  • the lytic virus can be any listed in the tables above.
  • the present invention also provides for a method of treating lysogenic viruses, by administering a composition including a vector encoding isolated nucleic acid encoding a Cas9 nuclease that is engineered to prevent off-target effects (such as those described in TABLE 1 above) and at least two gRNAs having at least one modified nucleic acid, and inactivating the lysogenic virus.
  • the composition can include any of the properties as described above, such as being in isolated nucleic acid, be packaged in a vector delivery system, or include other CRISPR or gene editing systems that target DNA.
  • the lysogenic virus can be any listed in the tables above.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Genetics & Genomics (AREA)
  • Engineering & Computer Science (AREA)
  • Chemical & Material Sciences (AREA)
  • Biomedical Technology (AREA)
  • Organic Chemistry (AREA)
  • Zoology (AREA)
  • Wood Science & Technology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Molecular Biology (AREA)
  • General Engineering & Computer Science (AREA)
  • Biotechnology (AREA)
  • Microbiology (AREA)
  • Biochemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Plant Pathology (AREA)
  • Physics & Mathematics (AREA)
  • Biophysics (AREA)
  • Virology (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

L'invention concerne une composition pour un traitement contre un virus lysogène, comprenant un vecteur codant pour un acide nucléique isolé codant pour au moins deux éditeurs de gènes sélectionnés parmi des éditeurs de gènes qui ciblent l'ADN viral, des éditeurs de gènes qui ciblent l'ARN viral et des combinaisons de ceux-ci. L'invention concerne également une composition pour un traitement contre un virus lytique, comprenant un vecteur codant pour un acide nucléique isolé codant pour au moins un éditeur de gène qui cible l'ADN viral, et une composition de ciblage de l'ARN viral. L'invention concerne par ailleurs une composition pour un traitement à la fois contre un virus lysogène et un virus lytique, comprenant un vecteur codant pour un acide nucléique isolé codant pour au moins deux éditeurs de gènes qui ciblent l'ARN viral. L'invention concerne en outre une composition pour le traitement contre des virus lytiques. L'invention concerne également un procédé pour accroître la spécificité d'éditeurs de gènes dans le traitement d'un individu contre un virus. L'invention concerne enfin des méthodes de traitement contre un virus lysogène ou un virus lytique, par administration des compositions ci-dessus à un patient porteur d'un virus, de manière à inactiver le virus.
PCT/US2019/052390 2018-09-25 2019-09-23 Crispr à spécificité améliorée WO2020068643A1 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US16/141,407 US20190071673A1 (en) 2017-01-18 2018-09-25 CRISPRs WITH IMPROVED SPECIFICITY
US16/141,407 2018-09-25

Publications (1)

Publication Number Publication Date
WO2020068643A1 true WO2020068643A1 (fr) 2020-04-02

Family

ID=69950834

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2019/052390 WO2020068643A1 (fr) 2018-09-25 2019-09-23 Crispr à spécificité améliorée

Country Status (1)

Country Link
WO (1) WO2020068643A1 (fr)

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20170051276A1 (en) * 2013-03-14 2017-02-23 Caribou Biosciences, Inc. Compositions And Methods Of Nucleic Acid-Targeting Nucleic Acids
US20180119140A1 (en) * 2015-04-06 2018-05-03 The Board Of Trustees Of The Leland Stanford Junior University Chemically Modified Guide RNAs for CRISPR/CAS-Mediated Gene Regulation
US20180127783A1 (en) * 2013-11-07 2018-05-10 Editas Medicine, Inc. CRISPR-RELATED METHODS AND COMPOSITIONS WITH GOVERNING gRNAS
WO2018140269A1 (fr) * 2017-01-26 2018-08-02 Excision Biotherapeutics, Inc. Lentivirus et lentivirus non intégratif utilisés comme vecteurs viraux pour administrer une thérapie crispr

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20170051276A1 (en) * 2013-03-14 2017-02-23 Caribou Biosciences, Inc. Compositions And Methods Of Nucleic Acid-Targeting Nucleic Acids
US20180127783A1 (en) * 2013-11-07 2018-05-10 Editas Medicine, Inc. CRISPR-RELATED METHODS AND COMPOSITIONS WITH GOVERNING gRNAS
US20180119140A1 (en) * 2015-04-06 2018-05-03 The Board Of Trustees Of The Leland Stanford Junior University Chemically Modified Guide RNAs for CRISPR/CAS-Mediated Gene Regulation
WO2018140269A1 (fr) * 2017-01-26 2018-08-02 Excision Biotherapeutics, Inc. Lentivirus et lentivirus non intégratif utilisés comme vecteurs viraux pour administrer une thérapie crispr

Similar Documents

Publication Publication Date Title
US20180201921A1 (en) CRISPRs
US20230233654A1 (en) Gene editing methods and compositions for eliminating risk of jc virus activation and pml (progressive multifocal leukoencephalopathy) during immunosuppressive therapy
US20220290177A1 (en) Compositions and methods for excision with single grna
KR20230057487A (ko) 게놈 조정을 위한 방법 및 조성물
US20230048681A1 (en) Compositions and methods of treatment for lytic and lysogenic viruses
KR20230169449A (ko) Rna-가이드된 핵산 변형 효소 및 이의 사용 방법
WO2017192172A1 (fr) Éradication, guidée par l'arn, du virus varicelle-zona
WO2017142835A1 (fr) Excision de séquences d'acides nucléiques rétrovirales
US20180208914A1 (en) Lentivirus and non-integrating lentivirus as viral vector to deliver crispr therapeutic
EP3758714A1 (fr) Méthodes et compositions de traitement du syndrome d'angelman
WO2023096766A1 (fr) Méthodes de blocage d'une infection asfv par interruption d'interactions de récepteurs cellulaires et viraux
US20190071673A1 (en) CRISPRs WITH IMPROVED SPECIFICITY
US20190338315A1 (en) CLOAKED CRISPRs
WO2020068643A1 (fr) Crispr à spécificité améliorée
US20190336617A1 (en) CRISPRs IN SERIES TREATMENT
WO2020014703A1 (fr) Détection de protéines/immunoglobulines bactériennes destinée à une thérapie d'édition génique
US20240050547A1 (en) Methods of blocking / neutralizing asfv infection through interruption of cellular and viral receptor interactions
NZ747016A (en) Compositions and methods of treatment for lytic and lysogenic viruses
WO2024118959A1 (fr) Méthodes de blocage/neutralisation d'une infection à asfv par interruption d'interactions de récepteurs cellulaires et viraux
WO2021173998A2 (fr) Traitement du virus de la leucémie à lymphocytes t humains par édition de gènes
WO2018130518A1 (fr) Procédés et composition pharmaceutique d'induction de la sénescence dans des cellules cancéreuses

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: 19864880

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 19864880

Country of ref document: EP

Kind code of ref document: A1