Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
CLOAKED CRISPRS
Document Type and Number:
WIPO Patent Application WO/2019/213062
Kind Code:
A1
Abstract:
A composition including an isolated cloaked gene editor. A composition for treating a lysogenic virus, including a vector encoding isolated nucleic acid encoding two or more cloaked gene editors chosen from cloaked gene editors that target viral DNA, cloaked gene editors that target viral RNA, and combinations thereof. A composition for treating a lytic virus, including a vector encoding isolated nucleic acid encoding at least one cloaked gene editor that targets viral DNA and a cloaked viral RNA targeting composition. A composition for treating both lysogenic and lytic viruses, including a vector encoding isolated nucleic acid encoding two or more cloaked gene editors that target viral RNA. A composition for treating lytic viruses. A method of preventing antibody neutralizing effects with gene editors in humans. Methods of treating a lysogenic virus or a lytic virus, by administering the above compositions to an individual having a virus and inactivating the virus.

Inventors:
MALCOLM THOMAS (US)
MITCHELL DAVID (US)
Application Number:
PCT/US2019/029882
Publication Date:
November 07, 2019
Filing Date:
April 30, 2019
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
MALCOLM THOMAS (US)
MITCHELL DAVID (US)
International Classes:
C12N15/90
Domestic Patent References:
WO2017210380A12017-12-07
Other References:
KATREKAR ET AL.: "Oligonucleotide conjugated multifunctional adeno-associated viruses", SCIENTIFIC REPORTS, vol. 8, no. 1, 26 February 2018 (2018-02-26), pages 3589-1 - 3589-8, XP055648766
Attorney, Agent or Firm:
KOHN, Kenneth I. (US)
Download PDF:
Claims:
CLAIMS

What is claimed is:

1. A composition comprising an isolated cloaked gene editor.

2. The composition of claim 1, wherein said cloaked gene editor is chosen from the group consisting of cloaked Argonaute proteins, cloaked RNase P RNA, cloaked C2cl, cloaked C2c2, cloaked C2c3, cloaked Cas9, cloaked Cpfl, cloaked TevCas9, cloaked Archaea Cas9, cloaked CasY.l, cloaked CasY.2, cloaked CasY.3, cloaked CasY.4, cloaked CasY.5, cloaked CasY.6, and cloaked CasX.

3. The composition of claim 1, wherein said isolated cloaked gene editor includes chemical changes chosen from the group consisting of introducing glycosylation, eliminating oxidative sites, changing proteins that generate non-natural amino acids, and combinations thereof.

4. A composition for treating a lysogenic virus, comprising a vector encoding isolated nucleic acid encoding two or more cloaked gene editors chosen from the group consisting of cloaked gene editors that target viral DNA, cloaked gene editors that target viral RNA, and combinations thereof.

5. The composition of claim 4, wherein said cloaked gene editors that target viral DNA are chosen from the group consisting of cloaked CRISPR-associated nucleases and cloaked Argonaute endonuclease gDNAs.

6. The composition of claim 5, wherein said cloaked CRISPR-associated nucleases are chosen from the group consisting of cloaked Cas9 gRNAs, cloaked Cpfl gRNAs, cloaked C2cl gRNAs, cloaked C2c3 gRNAs, cloaked TevCas9 gRNAs, cloaked Archaea Cas9 gRNAs, cloaked CasY.l gRNAs, cloaked CasY.2 gRNAs, cloaked CasY.3 gRNAs, cloaked CasY.4 gRNAs, cloaked CasY.5 gRNAs, cloaked CasY.6 gRNAs, and cloaked CasX gRNAs.

7. The composition of claim 4, wherein said cloaked gene editors that target viral RNA are chosen from the group consisting of cloaked C2c2 and cloaked RNase P RNA.

8. The composition of claim 4, wherein said composition removes a replication critical segment of the viral DNA or RNA.

9. The composition of claim 4, wherein said composition excises an entire viral genome of said lysogenic virus from a host cell.

10. The composition of claim 4, wherein said lysogenic virus is chosen from the group consisting of hepatitis A, hepatitis B, hepatitis D, HSV-1, HSV-2, cytomegalovirus, Epstein-Barr virus, Varicella Zoster virus, HIV1, HIV2, HTLV1, HTLV2, Rous Sarcoma virus, HPV virus, yellow fever, zika, dengue, West Nile, Japanese encephalitis, lyssa virus, vesiculovirus, cytohabdovirus, Hantaan virus, Rift Valley virus, Bunyamwera virus, Lassa virus, Junin virus, Machupo virus, Sabia virus, Tacaribe virus, Flexal virus, Whitewater Arroyo virus, ebola, Marburg virus, JC virus, and BK virus.

11. A composition for treating a lytic virus, comprising a vector encoding isolated nucleic acid encoding at least one cloaked gene editor that targets viral DNA and a cloaked viral RNA targeting composition.

12. The composition of claim 11, wherein said cloaked gene editor that targets viral DNA is chosen from the group consisting of cloaked CRISPR-associated nucleases and cloaked Argonaute endonuclease gDNAs.

13. The composition of claim 12, wherein said cloaked CRISPR-associated nucleases are chosen from the group consisting of cloaked Cas9 gRNAs, cloaked Cpfl gRNAs, cloaked C2cl gRNAs, cloaked C2c3 gRNAs, cloaked TevCas9 gRNAs, cloaked Archaea Cas9 gRNAs, cloaked CasY.l gRNAs, cloaked CasY.2 gRNAs, cloaked CasY.3 gRNAs, cloaked CasY.4 gRNAs, cloaked CasY.5 gRNAs, cloaked CasY.6 gRNAs, and cloaked CasX gRNAs.

14. The composition of claim 11, wherein said cloaked viral RNA targeting composition is chosen from the group consisting of siRNAs, miRNAs, shRNAs, RNAi, cloaked CRISPR-associated nucleases, cloaked Argonaute endonuclease gDNAs, cloaked C2c2, and cloaked RNase P RNA.

15. The composition of claim 11, wherein said composition removes a replication critical segment of the viral DNA or RNA.

16. The composition of claim 11, wherein said composition excises an entire viral genome of said lytic virus from a host cell.

17. The composition of claim 11, wherein said lytic virus is chosen from the group consisting of hepatitis A, hepatitis C, hepatitis D, coxsachievirus, HSV-1, HSV-2, cytomegalovirus, Epstein-Barr virus, varicella zoster virus, HIV1, HIV2, HTLV1, HTLV2, Rous Sarcoma virus, rota, seadornvirus, coltivirus, JC virus, and BK virus.

18. A composition for treating both lysogenic and lytic viruses, comprising a vector encoding isolated nucleic acid encoding two or more cloaked gene editors that target viral RNA, chosen from the group consisting of cloaked CRISPR-associated nucleases, cloaked Argonaute endonuclease gDNAs, cloaked C2c2, cloaked RNase P RNA, and combinations thereof.

19. The composition of claim 18, wherein said cloaked CRISPR-associated nucleases are chosen from the group consisting of cloaked Cas9 gRNAs, cloaked Cpfl gRNAs, cloaked C2cl gRNAs, cloaked C2c3 gRNAs, cloaked TevCas9 gRNAs, cloaked Archaea Cas9 gRNAs, cloaked CasY.l gRNAs, cloaked CasY.2 gRNAs, cloaked CasY.3 gRNAs, cloaked CasY.4 gRNAs, cloaked CasY.5 gRNAs, cloaked CasY.6 gRNAs, and cloaked CasX gRNAs.

20. The composition of claim 18, wherein said composition removes a replication critical segment of the viral RNA.

21. The composition of claim 18, wherein said composition excises an entire viral genome of said lysogenic and lytic virus from a host cell.

22. The composition of claim 18, wherein said lysogenic and lytic virus is chosen from the group consisting of hepatitis A, hepatitis C, hepatitis D, HSV-1, HSV-2, cytomegalovirus, Epstein-Barr virus, varicella zoster virus, HIV1, HIV2, HTLV1, HTLV2, Rous Sarcoma virus, JC virus, and BK virus.

23. A composition for treating lytic viruses, comprising a vector encoding isolated nucleic acid encoding two or more cloaked gene editors that target viral RNA and a cloaked viral RNA targeting composition.

24. The composition of claim 23, wherein said gene editors that target viral RNA are chosen from the group consisting of cloaked CRISPR-associated nucleases and cloaked Argonaute endonuclease gDNAs.

25. The composition of claim 22, wherein said cloaked CRISPR-associated nucleases are chosen from the group consisting of cloaked Cas9 gRNAs, cloaked Cpfl gRNAs, cloaked C2cl gRNAs, cloaked C2c3 gRNAs, cloaked TevCas9 gRNAs, cloaked Archaea Cas9 gRNAs, cloaked CasY.l gRNAs, cloaked CasY.2 gRNAs, cloaked CasY.3 gRNAs, cloaked CasY.4 gRNAs, cloaked CasY.5 gRNAs, cloaked CasY.6 gRNAs, and cloaked CasX gRNAs.

26. The composition of claim 23, wherein said cloaked viral RNA targeting composition is chosen from the group consisting of siRNAs, miRNAs, shRNAs, RNAi, cloaked C2c2, and cloaked RNase P RNA.

27. The composition of claim 23, wherein said composition removes a replication critical segment of the viral RNA.

28. The composition of claim 23, wherein said composition excises an entire viral genome of said lytic virus from a host cell.

29. The composition of claim 23, wherein said lytic virus is chosen from the group consisting of hepatitis A, hepatitis C, hepatitis D, coxsachievirus, HSV-1, HSV-2, cytomegalovirus, Epstein-Barr virus, varicella zoster virus, HIV1, HIV2, HTLV1, HTLV2, Rous Sarcoma virus, rota, seadornvirus, coltivirus, JC virus, and BK virus.

BO. A method of preventing antibody neutralizing effects with gene editors in humans, including the steps of:

cloaking a gene editor; and

administering the cloaked gene editor to a human without generating antibodies to the cloaked gene editor.

31. The method of claim 30, wherein the gene editor is chosen from the group consisting of cloaked Argonaute proteins, cloaked RNase P RNA, cloaked C2cl, cloaked C2c2, cloaked C2c3, cloaked Cas9, cloaked Cpfl, cloaked TevCas9, cloaked Archaea Cas9, cloaked CasY.l, cloaked CasY.2, cloaked CasY.3, cloaked CasY.4, cloaked CasY.5, cloaked CasY.6, and cloaked CasX.

32. The method of claim 30, wherein said cloaking step is further defined as introducing chemical changes to the gene editor chosen from the group consisting of introducing glycosylation, eliminating oxidative sites, changing proteins that generate non-natural amino acids, and combinations thereof.

33. A method of treating a lysogenic virus, including the steps of:

administering a composition including a vector encoding isolated nucleic acid encoding two or more cloaked gene editors chosen from the group consisting of cloaked gene editors that target viral DNA, cloaked gene editors that target viral RNA, and combinations thereof to an individual having a lysogenic virus; and

inactivating the lysogenic virus.

34. The method of claim 33, wherein the cloaked gene editors that target viral DNA are chosen from the group consisting of cloaked CRISPR-associated nucleases and cloaked Argonaute endonuclease gDNAs.

35. The method of claim 34, wherein the cloaked CRISPR-associated nucleases are chosen from the group consisting of cloaked Cas9 gRNAs, cloaked Cpfl gRNAs, cloaked C2cl gRNAs, cloaked C2c3 gRNAs, cloaked TevCas9 gRNAs, cloaked Archaea Cas9 gRNAs, cloaked CasY.l gRNAs, cloaked CasY.2 gRNAs, cloaked CasY.3 gRNAs, cloaked CasY.4 gRNAs, cloaked CasY.5 gRNAs, cloaked CasY.6 gRNAs, and cloaked CasX gRNAs.

36. The method of claim 33, wherein the cloaked gene editors that target viral RNA are chosen from the group consisting of humanizes C2c2 and cloaked RNase P RNA.

37. The method of claim 33, wherein said inactivating step includes removing a replication critical segment of the viral DNA or RNA.

38. The method of claim 33, wherein said inactivating step includes excising an entire viral genome of the lysogenic virus from a host cell.

39. The method of claim 33, wherein the lysogenic virus is chosen from the group consisting of hepatitis A, hepatitis B, hepatitis D, HSV-1, HSV-2, cytomegalovirus, Epstein-Barr virus, Varicella Zoster virus, HIV1, HIV2, HTLV1, HTLV2, Rous Sarcoma virus, HPV virus, yellow fever, zika, dengue, West Nile, Japanese encephalitis, lyssa virus, vesiculovirus, cytohabdovirus, Hantaan virus, Rift Valley virus, Bunyamwera virus, Lassa virus, Junin virus, Machupo virus, Sabia virus, Tacaribe virus, Flexal virus, Whitewater Arroyo virus, ebola, Marburg virus, JC virus, and BK virus.

40. A method for treating a lytic virus, including the steps of:

administering a composition including a vector encoding isolated nucleic acid encoding at least one cloaked gene editor that targets viral DNA and a cloaked viral RNA targeting composition to an individual having a lytic virus; and

inactivating the lytic virus.

41. The method of claim 40, wherein the cloaked gene editor that targets viral DNA is chosen from the group consisting of cloaked CRISPR-associated nucleases and cloaked Argonaute endonuclease gDNAs.

42. The method of claim 41, wherein the cloaked CRISPR-associated nucleases are chosen from the group consisting of cloaked Cas9 gRNAs, cloaked Cpfl gRNAs, cloaked C2cl gRNAs, cloaked C2c3 gRNAs, cloaked TevCas9 gRNAs, cloaked Archaea Cas9 gRNAs, cloaked CasY.l gRNAs, cloaked CasY.2 gRNAs, cloaked CasY.3 gRNAs, cloaked CasY.4 gRNAs, cloaked CasY.5 gRNAs, cloaked CasY.6 gRNAs, and cloaked CasX gRNAs.

43. The method of claim 40, wherein the cloaked viral RNA targeting composition is chosen from the group consisting of siRNAs, miRNAs, shRNAs, RNAi, cloaked CRISPR-associated nucleases, cloaked Argonaute endonuclease gDNAs, cloaked C2c2, and cloaked RNase P RNA.

44. The method of claim 40, wherein said inactivating step includes removing a replication critical segment of the viral DNA or RNA.

45. The method of claim 40, wherein said inactivating step includes excising an entire viral genome of the lytic virus from a host cell.

46. The method of claim 40, wherein the lytic virus is chosen from the group consisting of hepatitis A, hepatitis C, hepatitis D, coxsachievirus, HSV-1, HSV-2, cytomegalovirus, Epstein-Barr virus, varicella zoster virus, HIV1, HIV2, HTLV1, HTLV2, Rous Sarcoma virus, rota, seadornvirus, coltivirus, JC virus, and BK virus.

47. A method for treating both lysogenic and lytic viruses, including the steps of:

administering a composition including a vector encoding isolated nucleic acid encoding two or more cloaked gene editors that target viral RNA, chosen from the group consisting of cloaked CRISPR-associated nucleases, cloaked Argonaute endonuclease gDNAs, cloaked C2c2, cloaked RNase P RNA, and combinations thereof to an individual having a lysogenic virus and lytic virus; and

inactivating the lysogenic virus and lytic virus.

48. The method of claim 47, wherein said cloaked CRISPR-associated nucleases are chosen from the group consisting of cloaked Cas9 gRNAs, cloaked Cpfl gRNAs, cloaked C2cl gRNAs, cloaked C2c3 gRNAs, cloaked TevCas9 gRNAs, cloaked Archaea Cas9 gRNAs, cloaked CasY.l gRNAs, cloaked CasY.2 gRNAs, cloaked CasY.3 gRNAs, cloaked CasY.4 gRNAs, cloaked CasY.5 gRNAs, cloaked CasY.6 gRNAs, and cloaked CasX gRNAs.

49. The method of claim 47, wherein said inactivating step includes removing a replication critical segment of the viral RNA.

50. The method of claim 47, wherein said inactivating step includes excising an entire viral genome of the lysogenic and lytic virus from a host cell.

51. The method of claim 47, wherein the lysogenic and lytic virus is chosen from the group consisting of hepatitis A, hepatitis C, hepatitis D, HSV-1, HSV-2, cytomegalovirus, Epstein-Barr virus, varicella zoster virus, HIV1, HIV2, HTLV1, HTLV2, Rous Sarcoma virus, JC virus, and BK virus.

52. A method for treating lytic viruses, including the steps of:

administering a composition including a vector encoding isolated nucleic acid encoding two or more cloaked gene editors that target viral RNA and a cloaked viral RNA targeting composition to an individual having a lytic virus; and

inactivating the lytic virus.

53. The method of claim 52, wherein the cloaked gene editors that target viral RNA are chosen from the group consisting of cloaked CRISPR-associated nucleases and cloaked Argonaute endonuclease gDNAs.

54. The method of claim 53, wherein the cloaked CRISPR-associated nucleases are chosen from the group consisting of cloaked Cas9 gRNAs, cloaked Cpfl gRNAs, cloaked C2cl gRNAs, cloaked C2c3 gRNAs, cloaked TevCas9 gRNAs, cloaked Archaea Cas9 gRNAs, cloaked CasY.l gRNAs, cloaked CasY.2 gRNAs, cloaked CasY.3 gRNAs, cloaked CasY.4 gRNAs, cloaked CasY.5 gRNAs, cloaked CasY.6 gRNAs, and cloaked CasX gRNAs.

55. The method of claim 52, wherein the cloaked viral RNA targeting composition is chosen from the group consisting of siRNAs, miRNAs, shRNAs, RNAi, cloaked C2c2, and cloaked RNase P RNA.

56. The method of claim 52, wherein said inactivating step includes removing a replication critical segment of the viral RNA.

57. The method of claim 52, wherein said inactivating step includes excising an entire viral genome of the lytic virus from a host cell.

58. The method of claim 48, wherein the lytic virus is chosen from the group consisting of hepatitis A, hepatitis C, hepatitis D, coxsachievirus, HSV-1, HSV-2, cytomegalovirus, Epstein-Barr virus, varicella zoster virus, HIV1, HIV2, HTLV1, HTLV2, Rous Sarcoma virus, rota, seadornvirus, coltivirus, JC virus, and BK virus.

59. A method of screening for a cloaked editor, including the steps of:

identifying antigen epitopes on a gene editor that react with human IgG and/or IgM immunoglobulins;

identifying amino acid residues of the gene editor that affect antigen-epitope recognition but have no affect on the gene editor gRNA binding, DNA association, and/or DNA nuclease activity through a yeast colony analysis; and

performing genetic engineering on epitope sequences identified to limit antigenicity.

Description:
CLOAKED CRISPRs

BACKGROUND OF THE INVENTION

1. TECHNICAL FIELD

[0001] 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.

2. BACKGROUND ART

[0002] Gene editing allows DNA or RNA to be inserted, deleted, or replaced in an organism's genome by the use of 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.

[0003] 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.

[0004] 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.

[0005] 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). 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 bacterium.

[0006] 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.

[0007] Viruses replicate by one of two cycles, either the lytic cycle or the lysogenic cycle. In the lytic cycle, first the virus penetrates a host cell and releases its own nucleic acid. Next, 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.

[0008] Lytic viruses include John Cunningham virus (JCV), hepatitis A, and 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.

[0009] While the methods and compositions described above are useful in treating lysogenic viruses that have been integrated into the genome of a host cell, gene editing systems are not able to effectively treat lytic viruses. Treating a lytic virus will result in inefficient clearance of the virus if solely using this system unless inhibitor drugs are available to suppress viral expression, as in the case of HIV. Most viruses presently lack targeted inhibitor drugs. In particular, the CRISPR-associated nuclease cannot access viral nucleic acid that is contained within the virion (that is, protected by capsid or envelope proteins for example).

[00010] Researchers from the Broad Institute of MIT and Harvard, Massachusetts Institute of Technology, the National Institutes of Health, Rutgers University- New Brunswick and the Skolkovo Institute of Science and Technology have characterized a new CRISPR system that targets RNA, rather than DNA. This approach has the potential to open an additional avenue in cellular manipulation relating to editing RNA. Whereas DNA editing makes permanent changes to the genome of a cell, the CRISPR-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.

[00011] The findings reveal that 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. Other 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).

[00012] Antibodies are large Y-shaped proteins produced by the body's immune system after detection of antigens, i.e. any numerous foreign substances, including bacteria, fungi, parasites, viruses, and chemicals. Antibodies elicit the body's immune response to the antigens. An antibody has structure that is specific for an epitope on an antigen that allows the antibody to bind with the antigen thereby forming an immune complex. The binding can neutralize the antigen or tag it for destruction by the body.

[00013] Charlesworth, et al. report that anti-Cas9 antibodies were found in human serum for SaCas9 (S. aureus Cas9) and for SpCas9 (S. pyrogenes Cas9), as well as anti-SaCas9 T-cells (Identification of Pre-Existing Adaptive Immunity to Cas9 Protein in Humans, Jan. 5, 2018, bioRxiv). This shows that there can be pre-existing immune responses to Cas9 because of previous exposure of humans to the bacteria S. aureus and S. pyrogenes. Therefore, administration of Cas9 to humans for various treatments could pose a problem with neutralizing antibody effects. Neutralizing antibodies defend cells in the body from antigens or foreign matter by neutralizing any effects the antigen may have. Several existing treatments have been found to have a neutralizing antibody effect. For example, it has been found that any positive biological effects of administration of PCSK9 are negated because neutralizing antibodies attack the PCSK9 antibodies. Neutralizing antibody response has also been found with IFN-b treatment for MS patients, with patients receiving lower and less frequent doses having lower neutralizing antibody titers (Freedman, Medscape Neurology, Sept. 30, 2003). This can especially be an issue with antibodies derived from sources other than human, such as from mice or bacteria. Such antibodies, while they can be humanized, remain different enough that they can induce neutralizing antibodies in the body.

[00014] 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 will not induce a neutralizing antibody effect in the body of the subject being treated. SUMMARY OF THE INVENTION

[00015] The present invention provides for a composition including an isolated cloaked gene editor.

[00016] 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 cloaked gene editors that target viral DNA, cloaked gene editors that target viral RNA, and combinations thereof.

[00017] The present invention also provides for a composition for treating a lytic virus, including a vector encoding isolated nucleic acid encoding at least one cloaked gene editor that targets viral DNA and a cloaked viral RNA targeting composition.

[00018] 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 cloaked gene editors that target viral RNA, chosen from the group consisting of cloaked CRISPR-associated nucleases, cloaked Argonaute endonuclease gDNAs, cloaked C2c2, cloaked C2cl, cloaked c2c3, cloaked RNase P RNA, and combinations thereof.

[00019] The present invention provides for a composition for treating lytic viruses, including a vector encoding isolated nucleic acid encoding two or more cloaked gene editors that target viral RNA and a cloaked viral RNA targeting composition.

[00020] The present invention also provides for a method of preventing antibody neutralizing effects with gene editors in humans, by cloaking a gene editor, and administering the cloaked gene editor to a human without generating antibodies to the cloaked gene editor.

[00021] 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 cloaked gene editors chosen from the group consisting of cloaked gene editors that target viral DNA, cloaked gene editors that target viral RNA, and combinations thereof to an individual having a lysogenic virus and inactivating the lysogenic virus.

[00022] 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 cloaked gene editor that targets viral DNA and a cloaked viral RNA targeting composition to an individual having a lytic virus and inactivating the lytic virus.

[00023] 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 cloaked gene editors that target viral RNA, chosen from the group consisting of cloaked CRISPR- associated nucleases, cloaked Argonaute endonuclease gDNAs, cloaked C2c2, cloaked RNase P RNA, and combinations thereof to an individual having a lysogenic virus and lytic virus, and inactivating the lysogenic virus and lytic virus. [00024] 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 cloaked gene editors that target viral RNA and a cloaked viral RNA targeting composition to an individual having a lytic virus and inactivating the lytic virus.

[00025] The present invention also provides for a method of screening for a cloaked editor, by identifying antigen epitopes on a gene editor that react with human IgG and/or IgM immunoglobulins, identifying amino acid residues of the gene editor that affect antigen-epitope recognition but have no effect on the gene editor gRNA binding, DNA association, and/or DNA nuclease activity through a yeast colony analysis, and performing genetic engineering on epitope sequences identified to limit antigenicity.

DESCRIPTION OF THE DRAWINGS

[00026] Other advantages of the present invention are readily appreciated as the same becomes better understood by reference to the following detailed description when considered in connection with the accompanying drawings wherein:

[00027] 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; and

[00028] FIGURE 2 is a chart of various Archaea Cas9 effectors, CasY.l - CasY.6 effectors, and CasX effectors of the present invention.

DETAILED DESCRIPTION OF THE INVENTION

[00029] 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 are also cloaked to reduce antibody neutralizing effects.

[00030] The term "cloaked" as used herein 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. For example, 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. Cloaking of the gene editors herein renders the gene editors less likely to generate antibodies against them while still maintaining their activity. Cloaked gene editors are particularly useful when exposing humans to rare bacterial strains.

[00031] The present invention can use a method of screening to identify a cloaked gene editor as well with the following steps, which are further detailed in Example 1 below. The method most generally includes identifying antigen epitopes on a gene editor that react with human IgG and/or IgM immunoglobulins, identifying amino acid residues of the gene editor that affect antigen-epitope recognition but have no effect on the gene editor gRNA binding, DNA association, and/or DNA nuclease activity through a yeast colony analysis, and performing genetic engineering on epitope sequences identified to limit antigenicity.

[00032] In identifying the antigen epitopes, the gene editor is expressed in human extracts. Optionally, gene editor enzymes can also be isolated from bacterial extracts. The gene editor is cleaved with proteases and incubated with human IgG or human IgM agarose beads, the beads are washed multiple times, and the bound peptides are eluted with a low pH solution. The solution is neutralized, and mass spectrometry is performed to identify at least one epitope. Next epitope deleted constructs of the gene editor are constructed, the constructs are expressed in vitro, separated by SDS-PAGE, transferred to nitrocellulose, and probed with human serum and compared to full length gene editor in vitro translated protein. If no protein band is detected from a deletion construct compared to detection of wild type SaCas9 protein band, a candidate epitope is identified. Then the epitope sequence can be genetically engineered to limit its antigenicity (at this point or after identifying the effects of gRNA binding, DNA association, and/or DNA nuclease activity with the yeast colony analysis (which is further described below)).

[00033] 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.

[00034] The term "lentiviral vector" includes both integrating and non-integrating lentiviral vectors.

[00035] Viruses replicate by one of two cycles, either the lytic cycle or the lysogenic cycle. In the lytic cycle, first the virus penetrates a host cell and releases its own nucleic acid. Next, 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. Viruses such as lambda phage can switch between lytic and lysogenic cycles.

[00036] "Lysogenic virus" as used herein, 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. 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.

[00037] "Lytic virus" as used herein 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.

[00038] "gRNA" as used herein 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 gRNA.

[00039] "Nucleic acid" as used herein, 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. In the context of the present invention, 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.

[00040] 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. Thus, 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. In addition, 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. A nucleic acid existing among many (e.g., dozens, or hundreds to millions) of other nucleic acids within, for example, cDNA libraries or genomic libraries, or gel slices containing a genomic DNA restriction digest, is not an isolated nucleic acid.

[00041] 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. Generally, 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.

[00042] 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. For example, one or more pairs of long oligonucleotides (e.g., >50-100 nucleotides) can be synthesized that contain the desired sequence, with each pair containing a short segment of complementarity (e.g., about 15 nucleotides) such that a duplex is formed when the oligonucleotide pair is annealed. 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).

[00043] There are many different cloaked gene editors (CRISPR systems or others) and enzyme effectors that can be used with the methods and compositions of the present invention to target either DNA or RNA in viruses. These include cloaked Argonaute proteins, cloaked RNase P RNA, cloaked C2cl, cloaked C2c2, cloaked C2c3, various cloaked Cas9 enzymes, cloaked Cpfl, cloaked TevCas9, cloaked Archaea Cas9, cloaked CasY.l - CasY.6 effectors, and cloaked CasX effectors. Each of these are further described below. The present invention also provides for cloaked versions of these gene editors.

[00044] "Argonaute protein" as used herein, 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. 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. 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) can also be used in DNA interference and cleavage. The Argonaute proteins can be derived from Arabidopsis thaliana, D. melanogaster, Aquifex aeolicus, Thermus thermophiles, Pyrococcus furiosus, Thermus thermophilus JL- 18, Thermus thermophilus strain HB27, Aquifex aeolicus strain VF5, Archaeoglobus fulgidus, Anoxybacillus flavithermus, Halogeometricum borinquense, Microsystis aeruginosa, Clostridium bartlettii, Halorubrum lacusprofundi, Thermosynechococcus elongatus, and Synechococcus elongatus. 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.

[00045] 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.

[00046] 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. In vitro biochemical analysis show that C2c2 is guided by a single crRNA and can be programmed to cleave ssRNA targets carrying complementary protospacers. In bacteria, 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. 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. These results demonstrate the capability of C2c2 as a new RNA-targeting tools. C2c2 is preferably in a cloaked form.

[00047] 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. (PAM-Dependent Target DNA Recognition and Cleavage by C2cl CRISPR-Cas Endonuclease, Cell, 2016 Dec 15; 167(7):1814-1828)), a crystal structure confirms Alicyclobacillus acidoterrestris C2cl (AacC2cl) binds to sgRNA as a binary complex and targets DNAs as ternary complexes, thereby capturing catalytically competent conformations of AacC2cl with both target and non-target DNA strands independently positioned within a single RuvC catalytic pocket. Yang, et al. confirms that 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 is preferably in a cloaked form.

[00048] C2c3 is a gene editor effector 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 is preferably in a cloaked form.

[00049] "CRISPR Cas9" as used herein refers to Clustered Regularly Interspaced Short Palindromic Repeat (CRISPR)-associated endonuclease Cas9. In bacteria the 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, Cas9, 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. 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. Such 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 are preferably in cloaked form.

[00050] 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. As referenced above, Agonaute is another potential gene editing system. Cpfl is preferably in cloaked form.

[00051] A CRISPR/TevCas9 system can also be used. In some cases it has been shown that once CRISPR/Cas9 cuts DNA in one spot, 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 is preferably in a cloaked form.

[00052] The Cas9 nuclease can have a nucleotide sequence identical to the wild type Streptococcus pyrogenes sequence. In some embodiments, 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. Alternatively, 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. Alternatively, 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). In some embodiments, 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 Gl:669193761; or KM099233.1 Gl :669193765 or Cas9 amino acid sequence of PX330 or PX260

(Addgene, Cambridge, MA). 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). For example, 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. These include D-alloisoleucine (2R,3S)-2-amino-3-methylpentanoic acid and L-cyclopentyl glycine (S)-2-amino-2-cyclopentyl acetic acid. For other examples, one can consult textbooks or the worldwide web (a site is currently maintained by the California Institute of Technology and displays structures of non-natural amino acids that have been successfully incorporated into functional proteins). The Cas-9 can also be any shown in TABLE 1 below.

TABLE 1

[00053] Although the 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 1053 bp, whereas SpCas9 is 1358 bp.

[00054] The Cas9 nuclease sequence, or any of the gene editor effector sequences described herein, can be a mutated sequence. For example the Cas9 nuclease can be mutated in the conserved HNH and RuvC domains, which are involved in strand specific cleavage. For example, 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. In general, mutations of the gene editor effector sequence can minimize or prevent off-targeting.

[00055] 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. Preferably, the Archaea Cas9 is cloaked. [00056] ARMAN 1 amino acid sequence 950aa

[00057] (SEQ ID NO: 1):

[00058] MRDSITAPRYSSALAARI KEFNSAFKLGI DLGTKTGGVALVKDN KVLLAKTFLDYHKQTLEERRI HRRNRRSRL

ARRKRIARLRSWI LRQKIYGKQLPDPYKIKKMQLPNGVRKGENWI DLVVSGRDLSPEAFVRAITLIFQKRGQRYEEVAKEI

EEMSYKEFSTHI KALTSVTEEEFTALAAEIERRQDVVDTDKEAERYTQLSELLSKVSESKSESKDRAQRKED LGKVVNAFCS

AHRI EDKDKWCKELMKLLDRPVRHARFLNKVLIRCNICDRATPKKSRPDVRELLYFDTVRN FLKAGRVEQNPDVISYYKKI

YMDAEVI RVKI LNKEKLTDEDKKQKRKLASELNRYKNKEYVTDAQKKMQEQLKTLLFMKLTGRSRYCMAHL KERAAGK

DVEEGLHGVVQKRHDRNIAQRN HDLRVI NLI ESLLFDQNKSLSDAI RKNGLMYVTI EAPEPKTKHAKKGAAVVRDPRKL

KEKLFDDQNGVCIYTGLQLDKLEISKYEKDHI FPDSRDGPSI RDNLVLTTKEI NSDKGDRTPWEWMHDN PEKWKAFERR

VAEFYKKGRIN ERKRELLLNKGTEYPGDNPTELARGGARVNN FITEFN DRLKTHGVQELQTI FERN KPIVQVVRGEETQR

LRRQWNALNQNFI PLKDRAMSFNHAEDAAIAASMPPKFWREQIYRTAWHFGPSGN ERPDFALAELAPQWN DFFMT

KGGPI IAVLGKTKYSWKHSII DDTIYKPFSKSAYYVGIYKKPNAITSNAIKVLRPKLLNGEHTMSKNAKYYHQKIGN ERFLM

KSQKGGSIITVKPHDGPEKVLQISPTYECAVLTKHDGKI IVKFKPIKPLRDMYARGVIKAMDKELETSLSSMSKHAKYKELH

THDI IYLPATKKHVDGYFIITKLSAKHGIKALPESMVKVKYTQIGSENNSEVKLTKPKPEITLD SEDITN IYNFTR

[00059] ARMAN 1 nucleic acid sequence

[00060] (SEQ ID NO: 2):

[00061 ] atga gagactctat tactgcacct agatacagct ccgctcttgc cgccagaata aaggagttta attctgcttt caagttagga atcgacctag gaacaaaaac cggcggcgta gcactggtaa aagacaacaa agtgctgctc gctaagacat tcctcgatta ccataaacaa acactggagg aaaggaggat ccatagaaga aacagaagga gcaggctagc caggcggaag aggattgctc ggctgcgatc atggatactc agacagaaga tttatggcaa gcagcttcct gacccataca aaatcaaaaa aatgcagttg cctaatggtg tacgaaaagg ggaaaactgg attgacctgg tagtttctgg acgggacctt tcaccagaag ccttcgtgcg tgcaataact ctgatattcc aaaagagagg gcaaagatat gaagaagtgg ccaaagagat agaagaaatg agttacaagg aatttagtac tcacataaaa gccctgacat ccgttactga agaagaattt actgctctgg cagcagagat agaacggagg caggatgtgg ttgacacaga caaggaggcc gaacgctata cccaattgtc tgagttgctc tccaaggtct cagaaagcaa atctgaatct aaagacagag cgcagcgtaa ggaggatctc ggaaaggtgg tgaacgcttt ctgcagtgct catcgtatcg aagacaagga taaatggtgt aaagaactta tgaaattact agacagacca gtcagacacg ctaggttcct taacaaagta ctgatacgtt gcaatatctg cgatagggca acccctaaga aatccagacc tgacgtgagg gaactgctat attttgacac agtaagaaac ttcttgaagg ctggaagagt ggagcaaaac ccagacgtta ttagttacta taaaaaaatt tatatggatg cagaagtaat cagggtcaaa attctgaata aggaaaagct gactgatgag gacaaaaagc aaaagaggaa attagcgagc gaacttaaca ggtacaaaaa caaagaatac gtgactgatg cgcagaagaa gatgcaagag caacttaaga cattgctgtt catgaagctg acaggcaggt ctagatactg catggctcat cttaaggaaa gggcagcagg caaagatgta gaagaaggac ttcatggcgt tgtgcagaaa agacacgaca ggaacatagc acagcgcaat cacgacttac gtgtgattaa tcttattgag agtctgcttt tcgaccaaaa caaatcgctc tccgatgcaa taaggaagaa cgggttaatg tatgttacta ttgaggctcc agagccaaag actaagcacg caaagaaagg cgcagctgtg gtaagggatc ccagaaagtt gaaggagaag ttgtttgatg atcaaaacgg cgtttgcata tatacgggct tgcagttaga caaattagag ataagtaaat acgagaagga ccatatcttt ccagattcaa gggatggacc atctatcagg gacaatcttg tactcactac aaaagagata aattcagaca aaggcgatag gaccccatgg gaatggatgc atgataaccc agaaaaatgg aaagcgttcg agagaagagt cgcagaattc tataagaaag gcagaataaa tgagaggaaa agagaactcc tattaaacaa aggcactgaa taccctggcg ataacccgac tgagctggcg cggggaggcg cccgtgttaa caactttatt actgaattta atgaccgcct caaaacgcat ggagtccagg aactgcagac catctttgag cgtaacaaac caatagtgca ggtagtcagg ggtgaagaaa cgcagcgtct gcgcagacaa tggaatgcac taaaccagaa tttcatacca ctaaaggaca gggcaatgtc gttcaaccac gctgaagacg cagccatagc agcaagcatg ccaccaaaat tctggaggga gcagatatac cgtactgcgt ggcactttgg acctagtgga aatgagagac cggactttgc tttggcagaa ttggcgccac aatggaatga cttctttatg actaagggcg gtccaataat agcagtgctg ggcaaaacga agtatagttg gaagcacagc ataattgatg acactatata caagccattc agcaaaagtg cttactatgt tgggatatac aaaaagccga acgccatcac gtccaatgct ataaaagtct taaggccaaa actcttaaat ggcgaacata caatgtctaa gaatgcaaag tattatcatc agaagattgg taatgagcgc ttcctcatga aatctcagaa aggtggatcg ataattacag taaaaccaca cgacggaccg gaaaaagtgc ttcaaatcag ccctacatat gaatgcgcag tccttactaa gcatgacggt aaaataatag tcaaatttaa accaataaag ccgctacggg acatgtatgc ccgcggtgtg attaaagcca tggacaaaga gcttgaaaca agcctctcta gcatgagtaa acacgctaag tacaaggagt tacacactca tgatatcata tatctgcctg ctacaaagaa gcacgtagat ggctacttca taataaccaa actaagtgcg aaacatggca taaaagcact ccccgaaagc atggttaaag tcaagtatac tcaaattggg agtgaaaaca atagtgaagt gaagcttacc aaaccaaaac cagagataac tttggatagt gaagatatta caaacatata taatttcacc cgctaag

[00062] ARMAN 4 amino acid sequence 967aa

[00063] (SEQ ID NO: 3):

[00064] MLGSSRYLRYN LTSFEGKEPFLIMGYYKEYNKELSSKAQKEFNDQISEFNSYYKLGIDLGDKTGIAIVKGN KII L

AKTLIDLHSQKLDKRREARRNRRTRLSRKKRLARLRSWVMRQKVGNQRLPDPYKI MHDNKYWSIYN KSNSAN KKNWI

DLLIHSNSLSADDFVRGLTNFRKRGYLAFKYLSRLSDKEFEKYI DNLKPPISKYEYDEDLEELSSRVENGEI EEKKFEGLKNKL

DKI DKESKDFQVKQREEVKKELEDLVDLFAKSVDNKI DKARWKRELNN LLDKKVRKIRFDN RFILKCKIKGCN KNTPKKEK

VRDFELKMVLN NARSDYQISDEDLNSFRNEVI NI FQKKENLKKGELKGVTI EDLRKQLN KTFN KAKI KKGI REQIRSIVFEKI

SGRSKFCKEHLKEFSEKPAPSDRI NYGVNSAREQHDFRVLNFI DKKIFKDKU DPSKLRYITI ESPEPETEKLEKGQISEKSFET

LKEKLAKETGGIDIYTGEKLKKDFEIEHIFPRARMGPSIRENEVASN LETNKEKADRTPWEWFGQDEKRWSEFEKRVNSL

YSKKKISERKREILLN KSNEYPGLNPTELSRI PSTLSDFVESI RKMFVKYGYEEPQTLVQKGKPI IQWRGRDTQALRWRW

HALDSNI IPEKDRKSSFNHAEDAVIAACMPPYYLRQKIFREEAKI KRKVSN KEKEVTRPDMPTKKIAPNWSEFMKTRN EP

VI EVIGKVKPSWKNSI MDQTFYKYLLKPFKDNU KIPNVKNTYKWIGVNGQTDSLSLPSKVLSISNKKVDSSTVLLVHDKK

GGKRNWVPKSIGGLLVYITPKDGPKRIVQVKPATQGLLIYRN EDGRVDAVREFIN PVI EMYNNGKLAFVEKENEEELLKY

FNLLEKGQKFERI RRYDMITYNSKFYYVTKI NKN HRVTIQEESKIKAESDKVKSSSGKEYTRKETEELSLQKLAELISI

[00065] ARMAN 4 nucleic acid sequence

[00066] (SEQ ID NO: 4):

[00067] at gttaggctcc agcaggtacc tccgttataa cctaacctcg tttgaaggca aggagccatt tttaataatg ggatattaca aagagtataa taaggaatta agttccaaag ctcaaaaaga atttaatgat caaatttctg aatttaattc gtattacaaa ctaggtatag atctcggaga taaaacagga attgcaatcg taaagggcaa caaaataatc ctagcaaaaa cactaattga tttgcattcc caaaaattag ataaaagaag ggaagctaga agaaatagaa gaactcggct ttccagaaag aaaaggcttg cgagattaag atcgtgggta atgcgtcaga aagttggcaa tcaaagactt cccgatccat ataaaataat gcatgacaat aagtactggt ctatatataa taagagtaat tctgcaaata aaaagaattg gatagatctg ttaatccaca gtaactcttt atcagcagac gattttgtta gaggcttaac tataattttc agaaaaagag gctatttagc atttaagtat ctttcaaggt taagcgataa ggaatttgaa aaatacatag ataacttaaa accacctata agcaaatacg agtatgatga ggatttagaa gaattatcaa gcagggttga aaatggggaa atagaggaaa agaaattcga aggcttaaag aataagctag ataaaataga caaagaatct aaagactttc aagtaaagca aagagaagaa gtaaaaaagg aactggaaga cttagttgat ttgtttgcta aatcagttga taataaaata gataaagcta ggtggaaaag ggagctaaat aatttattgg ataagaaagt aaggaaaata cggtttgaca accgctttat tttgaagtgc aaaattaagg gctgtaacaa gaatactcca aagaaagaga aggtcagaga ttttgaattg aagatggttt taaataatgc tagaagcgat tatcagattt ctgatgagga tttaaactct tttagaaatg aagtaataaa tatatttcaa aagaaggaaa acttaaagaa aggagagctg aaaggagtta ctattgaaga tttgagaaag cagcttaata aaacttttaa taaagccaag attaaaaaag ggataaggga gcagataagg tctatcgtgt ttgaaaaaat tagtggaagg agtaaattct gcaaagaaca tctaaaagaa ttttctgaga agccggctcc ttctgacagg attaattatg gggttaattc agcaagagaa caacatgatt ttagagtctt aaatttcata gataaaaaaa tattcaaaga taagttgata gatccctcaa aattgaggta tataactatt gaatctccag aaccagaaac agagaagttg gaaaaaggtc aaatatcaga gaagagcttc gaaacattga aagaaaaatt ggctaaagaa acaggtggta ttgatatata cactggtgaa aaattaaaga aagactttga aatagagcac atattcccaa gagcaaggat ggggccttct ataagggaaa acgaagtagc atcaaatctg gaaacaaata aggaaaaggc cgatagaact ccttgggaat ggtttgggca agatgaaaaa agatggtcag agtttgagaa aagagttaat tctctttata gtaaaaagaa aatatcagag agaaaaagag aaattttgtt aaataagagt aatgaatatc cgggattaaa ccctacagaa ctaagtagaa tacctagtac gctgagcgac ttcgttgaga gtataagaaa aatgtttgtt aagtatggct atgaagagcc tcaaactttg gttcaaaaag gaaaaccgat aatacaagtt gttagaggca gagacacaca agctttgagg tggagatggc atgcattaga tagtaatata ataccagaaa aggacaggaa aagttcattt aatcacgctg aagatgcagt tattgccgcc tgtatgccac cttactatct caggcaaaaa atatttagag aagaagcaaa aataaaaaga aaagtaagca ataaggaaaa ggaagttaca cggcctgaca tgcctactaa aaagatagct ccgaactggt cggaatttat gaaaactaga aatgagccgg ttattgaagt aataggaaaa gttaagccaa gctggaaaaa cagcataatg gatcaaacat tttataaata tcttttgaag ccatttaaag ataacctgat aaaaataccc aacgttaaaa atacatacaa gtggatagga gttaatggac aaactgattc attatccctc ccgagtaagg tcttatctat ctctaataaa aaggttgatt cttctacagt tcttcttgtg catgataaga agggtggtaa gcggaattgg gtacctaaaa gtataggggg tttgttggta tatataactc ctaaagacgg gccgaaaaga atagttcaag taaagccagc aactcagggt ttgttaatat atagaaatga agatggcaga gtagatgctg taagagagtt cataaatcca gtgatagaaa tgtataataa tggcaaattg gcatttgtag aaaaagaaaa tgaagaagag cttttgaaat attttaattt gctggaaaaa ggtcaaaaat ttgaaagaat aagacggtat gatatgataa cctacaatag taaattttac tatgtaacaa aaataaacaa gaatcacaga gttactatac aagaagagtc taagataaaa gcagaatcag acaaagttaa gtcctcttca ggcaaagagt atactcgtaa ggaaaccgag gaattatcac ttcaaaaatt agcggaatta attagtatat aaaa

[00068] 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.

[00069] CasX.l Planctomycetes amino acid sequence 978aa

[00070] (SEQ ID NO: 5):

[00071 ] MQEIKRIN KIRRRLVKDSNTKKAGKTGPMKTLLVRVMTPDLRERLENLRKKPEN IPQPISNTSRAN LNKLLTD

YTEMKKAILHVYWEEFQKDPVGLMSRVAQPAPKNI DQRKLI PVKDGNERLTSSGFACSQCCQPLYVYKLEQVN DKGKP

HTNYFGRCNVSEHERLI LLSPHKPEAN DELVTYSLGKFGQRALDFYSIHVTRESNHPVKPLEQIGGNSCASGPVGKALSD

ACMGAVASFLTKYQDII LEHQKVI KKNEKRLANLKDIASANGLAFPKITLPPQPHTKEGI EAYNNVVAQIVIWVNLN LWQ

KLKIGRDEAKPLQRLKGFPSFPLVERQANEVDWWDMVCNVKKLI NEKKEDGKVFWQN LAGYKRQEALLPYLSSEEDRK

KGKKFARYQFGDLLLHLEKKHGEDWGKVYDEAWERIDKKVEGLSKHIKLEEERRSED AQSKAALTDWLRAKASFVI EGL

KEADKDEFCRCELKLQKWYGDLRGKPFAIEAENSILDISGFSKQYNCAFIWQKDGVK KLN LYLII NYFKGGKLRFKKI KPEA

FEAN RFYTVI NKKSGEIVPMEVN FNFDDPN LII LPLAFGKRQGREFIWNDLLSLETGSLKLANGRVI EKTLYN RRTRQDEP

ALFVALTFERREVLDSSN IKPMN LIGIDRGEN IPAVIALTDPEGCPLSRFKDSLGNPTHILRIGESYKEKQRTIQAAKEVEQR

RAGGYSRKYASKAKN LADDMVRNTARDLLYYAVTQDAMLI FEN LSRGFGRQGKRTFMAERQYTRMEDWLTAKLAYE

GLPSKTYLSKTLAQYTSKTCSNCGFTITSADYDRVLEKLKKTATGWMTTINGKELKV EGQITYYNRYKRQNVVKDLSVELD

RLSEESVN NDISSWTKGRSGEALSLLKKRFSHRPVQEKFVCLNCGFETHADEQAALNIARSWLFLRSQ EYKKYQTNKTTG

NTDKRAFVETWQSFYRKKLKEVWKPAV

[00072] CasX.l Planctomycetes nucleic acid sequence

[00073] (SEQ ID NO: 6):

[00074] atgct tcttatttat cggagatatc ttcaaacacc atcaacatgg caatggtgaa ccattaatat tctttgatgc ttcttattta tcggagatat cttcaaacat tgcccatttt acaggcatat cttctggctc tttgatgctt cttatttatc ggagatatct tcaaacgtaa tgtattgaga aagacatcaa gattagataa ctttgatgct tcttatttat cggagatatc ttcaaacaca gaaacctgca aagattgtat atatataagc tttgatgctt cttatttatc ggagatatct tcaaacgata cgtattttag cccgtctatt tggggattaa ctttgatgct tcttatttat cggagatatc ttcaaacccc gcatatccag atttttcaat gacttctgga aattgtattt tcaatatttt acaagttgcg gaggatacct ttaataattt agcagagtta cgcactgtaa acctgttctt ctcacaaaaa gctttaacat cagattttca aagaacttct tatgtaattt ataagaatct aaaaaaacag ctctgggttt gcatccagaa ctctccgata aataagcgct ttacccatac gacatagtcg ctggtgatgg ctctcaaagt aatgagataa aagcgccagt aataatttac tattcacaaa tcctttcgtc aagcttaaaa tcaatcaaag accatatccc cttcattcca aatagcagcg cttccgtacc tttctatccg ttcatatatc tcctctgaga gaggataaat taccagactt atagagccat ccataaatcc tttttcttta aggttgagct ttagatcagc ccaccttgct tttgaaaggt taaactcaaa gacagaatat tgaatccgaa caccataggc ttccagaagt ttaactaacc gtgccctgac cttatcatct tcaatatcat aacaaatgag atgtcgcatt ttaaagctct ataggcttat aacattccct atcatcttga atatgctggc taaacaacct aacctgccgc tcaactgcgt gctgatacgt tattgattgg ataagtaaat tggttttctg ctcatctacc ttaaagaatt gatgccattt tttgattact tttggatagg catccttatt cagccaaaca cctttttggt cagtttcttt cctgaaatcg tctgtatcca cttcccttct atttatcaaa ttgatcacaa aacggtcagc caacggccgc cactcctcca gaagatcgca tattaaagag ggacgaccat aatagacgtc atgcaagtaa ccaaaggccg ggtcaaaacc gacgagtaat gcagtcgaat gtatttcgtt gaacaggagg gtgtagataa ggctcatcat ggcgttgatt tcatcctcag gaggtctctt ggtacggcgc acaaaaacaa agcttggatg ctttaagata gccgaaaaat tgccataata ctgccttgtt gttgcgcctt ctattccacg caaggtctct aaatcagtga cggcgttgat ttcggtacac tcgattctca aaccaagtct atatttatca agtaatgatt gctggttttt gatcttaccg gcaacgatac tttttgcaat ttcaagtttt ttgtggggat caaaatgctt atgaatttgc gcccgacgaa taaacagatt tttgacgggt tcaaattgaa ggctcccttg atattcccat ctgccgctaa agaaatgtat cggtatagat tattctctgc aaaggctaat aacacggcta tcgagggtaa cccggccaac taccacgata tcttttacct tcattgcggg aatcttctgc cccttctctt cattgtcctt ttttatgaga aatgcccgac cacgacaatc caaaatgaat tcatcacccg tgagatagag ggttatcctg tcggttatag cggtcatcag taagcctttt atttttctaa ccaagtattg aaggaagaca cgattcacta tactggcact gcggacacct atggtcatca accttgggaa acctgcttat atcaaaggac aagaagcagt ctcgcagatt tgtaacaact tctacacaac gcactttcag ggttttatct ataacaattt ctttccgtct ccgtgtttca cagaaaaata tttcaccaac tggtatattg acattataca tctcttcaag gcaaattgcc tgtaacccaa tctgaacgtg gaagttctca aaatccctta ccttccctgt ctttgtttcg ataggaatcg gtatcccatc cctccactcg ataaggtctg cccggcctgc caaaccgagc ttattgctgt aaagatacac gcctgttacc tgcttacaat cagggcagct tctctgcgat gatttatcca ccgccctgtg cgcgtgtatg gcctctgtaa agtggatgct cttagccata ttacgccgtt ctccaacaaa ggcataccat gcattgcgcg gacaatagat tgactccatt accgtgctga tgtgcaatat cagacggctg gtttccatac ttctttgagc ttctttctgt aaaaggattg ccatgtttca acaaatgccc ttttgtcagt atttccggtc gttttattgg tttgatacttcttatattct tgagaacgga gaaagagcca cgaccttgca atattcagtg ctgcttgttc gtctgcatgg gtttcaaaac cacagttcag gcaaacaaac ttttcctgca ccggcctgtg actaaatctc ttttttagca gagataaagc ttcaccactg cggccttttg tccaactaga aatatcatta tttaccgact cttccgaaag tctatccagc tctacagaga ggtcttttac cacattctgc cttttatacc ggttatagta tgttatctgt ccttcaactt ttaactcttt tccattgatt gtagtcatcc atccagtagc cgtcttcttg agcttttcga gcaccctgtc ataatctgca cttgtgattg taaaaccaca attagaacat gtctttgagg tatactgtgc cagagtcttt gaaagatagg tttttgatgg cagaccttca taggcaagct ttgcagtcag ccagtcttcc atcctcgtgt actgcctttc cgccataaaa gtcctcttgc cttgtctacc aaaaccgcgg gaaagatttt caaaaatgag cattgcatct tgagtaacag cataatataa gaggtcacga gctgtatttc ttaccatatc gtccgccaga ttcttcgcct ttgatgcata ttttctcgaa tatccgcctg cccgcctttg ttcaacttct ttagcagcct gaatagtccg ttgtttttcc ttataacttt ctcctattcg caaaatatgc gttggattgc ccaatgaatc tttgaatctt gacaaggggc atccttccgg gtctgttaat gctatgactg ccgggatatt ttctccccgg tctattccta tcagattcat cggttttata ttcgatgagt caagcacctc tcttctttca aatgtcaggg caacaaaaag tgctggttca tcctgtctcg tccttctgtt atagagcgtt ttttcaataa ccctgccatt ggcgagtttc aatgaacccg tctcaaggct caataggtcg ttccagataa actccctccc ctgccttttt ccaaaggcca aaggcagaat tatcaaattc gggtcatcaa aattgaagtt gacctccata ggcacaatct caccgctttt tttattaatt actgtataaa acctatttgc ttcaaaagct tctggcttga tttttttgaa gcgtagctta ccacctttga agtaatttat tattaaataa agatttaact tctttacgcc gtctttctgc catataaatg cacaattata ctgtttagaa aatccgctta tatctaaaat gctgttctct gcttctatag caaatggttt tcctctcaaa tctccatacc acttttgaag ctttaactca cacctgcaaa actcatcctt atcagcttct ttgagccctt caataacaaa agaggccttt gccctgagcc aatcagtgag ggcagccttt gattgagcat cttcagacct tctttcttcc tccaacttta tgtgcttact cagaccttca acttttttat ctattctttc ccatgcctca tcataaactt tgccccaatc ttcaccgtgt ttcttttcaa ggtgaagcaa aaggtcacca aactgataac gcgcaaactt ttttcctttt ttacggtctt cttcagacga aagatatgga agcaaggctt cctgcctttt atatccagca agattttgcc agaagacctt cccgtcctct ttcttttcgt taatcaactt tttgacatta cagaccatat cccaccaatc aacctcattc gcctggcgtt caacaagagg gaaggacgga aaacccttaa gccgctgtaa gggctttgcc tcatccctgc caattttgag tttctgccaa agattcaggt ttacccagat cactatctga gcaacaacat tgttataagc ttcaatccct tcttttgtat gcggttgcgg tggaagagtg attttaggaa atgcaagccc gtttgcactt gctatatcct ttagatttgc caatctcttt tcgttttttt ttataacctt ttggtgttcg aggatgatgt cctggtactt tgtaaggaaa ctggctactg ctcccataca ggcatcagat aaagccttac caacgggacc acttgcgcag ctattgccac cgatctgttc tagcggcttt acaggatggt tcgattctct tgttacgtgg attgaataaa agtccaatgc cctttgaccg aacttcccca acgaatacgt tactagctcg tcatttgcct ccggtttatg cggcgagagc aatatcaaac gttcatgctc ggagacatta caacggccaa agtaatttgt atggggctta cccttgtcat tcacttgttc aagcttataa acatagaggg gttgacagca ctgagaacag gcaaatccag aacttgttag tctctcattt ccgtccttca ccggaatcaa ttttctctga tcaatattct tgggcgctgg ttgtgcaacc ctgctcatca atccgacagg gtctttttgg aactcttccc aataaacatg caggattgct ttcttcattt ccgtatagtc agtgaggagt ttatttaaat ttgcacgtga agtatttgaa atgggctgag gaatgttttc cggctttttg cgaagattct ctaacctttc tctcaggtca ggtgtcataa cccgaacgag caaggttttc atagggccgg ttttgccggc ttttttcgtg ttgctatcct ttaccaatct ccttcgtatt ttatttatcc tttttatttc ctgcatcttt

[00075] CasX.l Deltaproteobacteria amino acid sequence 986aa

[00076] (SEQ ID NO: 7):

[00077] MEKRIN KIRKKLSADNATKPVSRSGPMKTLLVRVMTDDLKKRLEKRRKKPEVMPQVISNNAAN NLRMLLD

DYTKMKEAI LQVYWQEFKDDHVGLMCKFAQPASKKIDQNKLKPEMDEKGNLTTAGFACSQCGQPLFVYK LEQVSEKG

KAYTNYFGRCNVAEHEKLILLAQLKPEKDSDEAVTYSLGKFGQRALDFYSI HVTKESTHPVKPLAQIAGNRYASGPVGKAL

SDACMGTIASFLSKYQDII IEHQKVVKGNQKRLESLRELAGKENLEYPSVTLPPQPHTKEGVDAYN EVIARVRMWVNLN

LWQKLKLSRDDAKPLLRLKGFPSFPVVERREN EVDWWNTIN EVKKLIDAKRDMGRVFWSGVTAEKRNTILEGYNYLPN

EN DHKKREGSLEN PKKPAKRQFGDLLLYLEKKYAGDWGKVFDEAWERIDKKIAGLTSHIEREEARNAEDAQSK AVLTD

WLRAKASFVLERLKEMDEKEFYACEIQLQKWYGDLRGNPFAVEAENRVVDISGFSIG SDGHSIQYRN LLAWKYLENGKR

EFYLLMNYGKKGRIRFTDGTDIKKSGKWQGLLYGGGKAKVIDLTFDPDDEQLII LPLAFGTRQGREFIWN DLLSLETGLI K

LANGRVIEKTIYN KKIGRDEPALFVALTFERREVVDPSNI KPVN LIGVDRGEN I PAVIALTDPEGCPLPEFKDSSGGPTDI LR

IGEGYKEKQRAIQAAKEVEQRRAGGYSRKFASKSRN LADDMVRNSARDLFYHAVTHDAVLVFEN LSRGFGRQGKRTF

MTERQYTKMEDWLTAKLAYEGLTSKTYLSKTLAQYTSKTCSNCGFTITTADYDGMLV RLKKTSDGWATTLN NKELKAE

GQITYYNRYKRQTVEKELSAELDRLSEESGN NDISKWTKGRRDEALFLLKKRFSHRPVQEQFVCLDCGHEVHADEQAAL

N IARSWLFLNSNSTEFKSYKSGKQPFVGAWQAFYKRRLKEVWKPNA

[00078] CasX.l Deltaproteobacteria nucleic acid sequence

[00079] (SEQ ID NO: 8):

[00080] at ggaaaagaga ataaacaaga tacgaaagaa actatcggcc gataatgcca caaagcctgt gagcaggagc ggccccatga aaacactcct tgtccgggtc atgacggacg acttgaaaaa aagactggag aagcgtcgga aaaagccgga agttatgccg caggttattt caaataacgc agcaaacaat cttagaatgc tccttgatga ctatacaaag atgaaggagg cgatactaca agtttactgg caggaattta aggacgacca tgtgggcttg atgtgcaaat ttgcccagcc tgcttccaaa aaaattgacc agaacaaact aaaaccggaa atggatgaaa aaggaaatct aacaactgcc ggttttgcat gttctcaatg cggtcagccg ctatttgttt ataagcttga acaggtgagt gaaaaaggca aggcttatac aaattacttc ggccggtgta atgtggccga gcatgagaaa ttgattcttc ttgctcaatt aaaacctgaa aaagacagtg acgaagcagt gacatactcc cttggcaaat tcggccagag ggcattggac ttttattcaa tccacgtaac aaaagaatcc acccatccag taaagcccct ggcacagatt gcgggcaacc gctatgcaag cggacctgtt ggcaaggccc tttccgatgc ctgtatgggc actatagcca gttttctttc gaaatatcaa gacatcatca tagaacatca aaaggttgtg aagggtaatc aaaagaggtt agagagtctc agggaattgg cagggaaaga aaatcttgag tacccatcgg ttacactgcc gccgcagccg catacgaaag aaggggttga cgcttataac gaagttattg caagggtacg tatgtgggtt aatcttaatc tgtggcaaaa gctgaagctc agccgtgatg acgcaaaacc gctactgcgg ctaaaaggat tcccatcttt ccctgttgtg gagcggcgtg aaaacgaagt tgactggtgg aatacgatta atgaagtaaa aaaactgatt gacgctaaac gagatatggg acgggtattc tggagcggcg ttaccgcaga aaagagaaat accatccttg aaggatacaa ctatctgcca aatgagaatg accataaaaa gagagagggc agtttggaaa accctaagaa gcctgccaaa cgccagtttg gagacctctt gctgtatctt gaaaagaaat atgccggaga ctggggaaag gtcttcgatg aggcatggga gaggatagat aagaaaatag ccggactcac aagccatata gagcgcgaag aagcaagaaa cgcggaagac gctcaatcca aagccgtact tacagactgg ctaagggcaa aggcatcatt tgttcttgaa agactgaagg aaatggatga aaaggaattc tatgcgtgtg aaatccaact tcaaaaatgg tatggcgatc ttcgaggcaa cccgtttgcc gttgaagctg agaatagagt tgttgatata agcgggtttt ctatcggaag cgatggccat tcaatccaat acagaaatct ccttgcctgg aaatatctgg agaacggcaa gcgtgaattc tatctgttaa tgaattatgg caagaaaggg cgcatcagat ttacagatgg aacagatatt aaaaagagcg gcaaatggca gggactatta tatggcggtg gcaaggcaaa ggttattgat ctgactttcg accccgatga tgaacagttg ataatcctgc cgctggcctt tggcacaagg caaggccgcg agtttatctg gaacgatttg ctgagtcttg aaacaggcct gataaagctc gcaaacggaa gagttatcga aaaaacaatc tataacaaaa aaatagggcg ggatgaaccg gctctattcg ttgccttaac atttgagcgc cgggaagttg ttgatccatc aaatataaag cctgtaaacc ttataggcgt tgaccgcggc gaaaacatcc cggcggttat tgcattgaca gaccctgaag gttgtccttt accggaattc aaggattcat cagggggccc aacagacatc ctgcgaatag gagaaggata taaggaaaag cagagggcta ttcaggcagc aaaggaggta gagcaaaggc gggctggcgg ttattcacgg aagtttgcat ccaagtcgag gaacctggcg gacgacatgg tgagaaattc agcgcgagac cttttttacc atgccgttac ccacgatgcc gtccttgtct ttgaaaacct gagcaggggt tttggaaggc agggcaaaag gaccttcatg acggaaagac aatatacaaa gatggaagac tggctgacag cgaagctcgc atacgaaggt cttacgtcaa aaacctacct ttcaaagacg ctggcgcaat atacgtcaaa aacatgctcc aactgcgggt ttactataac gactgccgat tatgacggga tgttggtaag gcttaaaaag acttctgatg gatgggcaac taccctcaac aacaaagaat taaaagccga aggccagata acgtattata accggtataa aaggcaaacc gtggaaaaag aactctccgc agagcttgac aggctttcag aagagtcggg caataatgat atttctaagt ggaccaaggg tcgccgggac gaggcattat ttttgttaaa gaaaagattc agccatcggc ctgttcagga acagtttgtt tgcctcgatt gcggccatga agtccacgcc gatgaacagg cagccttgaa tattgcaagg tcatggcttt ttctaaactc aaattcaaca gaattcaaaa gttataaatc gggtaaacag cccttcgttg gtgcttggca ggccttttac aaaaggaggc ttaaagaggt atggaagccc aacgcctgat

[00081] The gene editor effector can also be CasY.l - CasY.6, examples 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 are preferably in a cloaked form.

[00082] CasY.l Candidatus katanobacteria amino acid sequence 1125aa

[00083] (SEQ ID NO: 9):

[00084] MRKKLFKGYI LHN KRLVYTGKAAI RSI KYPLVAPN KTALN NLSEKIIYDYEHLFGPLNVASYARNSN RYSLVDF

WIDSLRAGVIWQSKSTSLIDLISKLEGSKSPSEKI FEQIDFELKNKLDKEQFKDI ILLNTGIRSSSNVRSLRGRFLKCFKEEFRD

TEEVIACVDKWSKDLIVEGKSILVSKQFLYWEEEFGI KIFPHFKDN HDLPKLTFFVEPSLEFSPHLPLANCLERLKKFDISRES

LLGLDN NFSAFSNYFN ELFN LLSRGEIKKIVTAVLAVSKSWENEPELEKRLHFLSEKAKLLGYPKLTSSWADYRMIIGGK IKS

WHSNYTEQLIKVREDLKKHQIALDKLQEDLKKVVDSSLREQI EAQREALLPLLDTMLKEKDFSDDLELYRFI LSDFKSLLNG

SYQRYIQTEEERKEDRDVTKKYKDLYSNLRN IPRFFGESKKEQFN KFIN KSLPTI DVGLKILEDIRNALETVSVRKPPSITEEY

VTKQLEKLSRKYKI NAFNSNRFKQITEQVLRKYNNGELPKISEVFYRYPRESHVAI RILPVKISN PRKDISYLLDKYQISPDWK

NSN PGEVVDLIEIYKLTLGWLLSCNKDFSMDFSSYDLKLFPEAASLI KNFGSCLSGYYLSKMIFNCITSEIKGMITLYTRDKF

VVRYVTQMIGSNQKFPLLCLVGEKQTKNFSRNWGVLI EEKGDLGEEKNQEKCLIFKDKTDFAKAKEVEI FKNN IWRIRTS

KYQIQFLNRLFKKTKEWDLMNLVLSEPSLVLEEEWGVSWDKDKLLPLLKKEKSCEER LYYSLPLN LVPATDYKEQSAEIEQ

RNTYLGLDVGEFGVAYAVVRIVRDRIELLSWGFLKDPALRKIRERVQDMKKKQVMAV FSSSSTAVARVREMAI HSLRN

QIHSIALAYKAKI IYEISISNFETGGN RMAKIYRSIKVSDVYRESGADTLVSEMIWGKKN KQMGNHISSYATSYTCCNCART

PFELVIDN DKEYEKGGDEFIFNVGDEKKVRGFLQKSLLGKTIKGKEVLKSI KEYARPPIREVLLEGEDVEQLLKRRGNSYIYR

CPFCGYKTDADIQAALN IACRGYISDNAKDAVKEGERKLDYI LEVRKLWEKNGAVLRSAKFL

[00085] CasY.l Candidatus katanobacteria nucleic acid sequence

[00086] (SEQ ID NO: 10):

[00087] at gcgcaaaaaa ttgtttaagg gttacatttt acataataag aggcttgtat atacaggtaa agctgcaata cgttctatta aatatccatt agtcgctcca aataaaacag ccttaaacaa tttatcagaa aagataattt atgattatga gcatttattc ggacctttaa atgtggctag ctatgcaaga aattcaaaca ggtacagcct tgtggatttt tggatagata gcttgcgagc aggtgtaatt tggcaaagca aaagtacttc gctaattgat ttgataagta agctagaagg atctaaatcc ccatcagaaa agatatttga acaaatagat tttgagctaa aaaataagtt ggataaagag caattcaaag atattattct tcttaataca ggaattcgtt ctagcagtaa tgttcgcagt ttgagggggc gctttctaaa gtgttttaaa gaggaattta gagataccga agaggttatc gcctgtgtag ataaatggag caaggacctt atcgtagagg gtaaaagtat actagtgagt aaacagtttc tttattggga agaagagttt ggtattaaaa tttttcctca ttttaaagat aatcacgatt taccaaaact aacttttttt gtggagcctt ccttggaatt tagtccgcac ctccctttag ccaactgtct tgagcgtttg aaaaaattcg atatttcgcg tgaaagtttg ctcgggttag acaataattt ttcggccttt tctaattatt tcaatgagct ttttaactta ttgtccaggg gggagattaa aaagattgta acagctgtcc ttgctgtttc taaatcgtgg gagaatgagc cagaattgga aaagcgctta cattttttga gtgagaaggc aaagttatta gggtacccta agcttacttc ttcgtgggcg gattatagaa tgattattgg cggaaaaatt aaatcttggc attctaacta taccgaacaa ttaataaaag ttagagagga cttaaagaaa catcaaatcg cccttgataa attacaggaa gatttaaaaa aagtagtaga tagctcttta agagaacaaa tagaagctca acgagaagct ttgcttcctt tgcttgatac catgttaaaa gaaaaagatt tttccgatga tttagagctt tacagattta tcttgtcaga ttttaagagt ttgttaaatg ggtcttatca aagatatatt caaacagaag aggagagaaa ggaggacaga gatgttacca aaaaatataa agatttatat agtaatttgc gcaacatacc tagatttttt ggggaaagta aaaaggaaca attcaataaa tttataaata aatctctccc gaccatagat gttggtttaa aaatacttga ggatattcgt aatgctctag aaactgtaag tgttcgcaaa cccccttcaa taacagaaga gtatgtaaca aagcaacttg agaagttaag tagaaagtac aaaattaacg cctttaattc aaacagattt aaacaaataa ctgaacaggt gctcagaaaa tataataacg gagaactacc aaagatctcg gaggtttttt atagataccc gagagaatct catgtggcta taagaatatt acctgttaaa ataagcaatc caagaaagga tatatcttat cttctcgaca aatatcaaat tagccccgac tggaaaaaca gtaacccagg agaagttgta gatttgatag agatatataa attgacattg ggttggctct tgagttgtaa caaggatttt tcgatggatt tttcatcgta tgacttgaaa ctcttcccag aagccgcttc cctcataaaa aattttggct cttgcttgag tggttactat ttaagcaaaa tgatatttaa ttgcataacc agtgaaataa aggggatgat tactttatat actagagaca agtttgttgt tagatatgtt acacaaatga taggtagcaa tcagaaattt cctttgttat gtttggtggg agagaaacag actaaaaact tttctcgcaa ctggggtgta ttgatagaag agaagggaga tttgggggag gaaaaaaacc aggaaaaatg tttgatattt aaggataaaa cagattttgc taaagctaaa gaagtagaaa tttttaaaaa taatatttgg cgtatcagaa cctctaagta ccaaatccaa tttttgaata ggctttttaa gaaaaccaaa gaatgggatt taatgaatct tgtattgagc gagcctagct tagtattgga ggaggaatgg ggtgtttcgt gggataaaga taaactttta cctttactga agaaagaaaa atcttgcgaa gaaagattat attactcact tccccttaac ttggtgcctg ccacagatta taaggagcaa tctgcagaaa tagagcaaag gaatacatat ttgggtttgg atgttggaga atttggtgtt gcctatgcag tggtaagaat agtaagggac agaatagagc ttctgtcctg gggattcctt aaggacccag ctcttcgaaa aataagagag cgtgtacagg atatgaagaa aaagcaggta atggcagtat tttctagctc ttccacagct gtcgcgcgag tacgagaaat ggctatacac tctttaagaa atcaaattca tagcattgct ttggcgtata aagcaaagat aatttatgag atatctataa gcaattttga gacaggtggt aatagaatgg ctaaaatata ccgatctata aaggtttcag atgtttatag ggagagtggt gcggataccc tagtttcaga gatgatctgg ggcaaaaaga ataagcaaat gggaaaccat atatcttcct atgcgacaag ttacacttgt tgcaattgtg caagaacccc ttttgaactt gttatagata atgacaagga atatgaaaag ggaggcgacg aatttatttt taatgttggc gatgaaaaga aggtaagggg gtttttacaa aagagtctgt taggaaaaac aattaaaggg aaggaagtgt tgaagtctat aaaagagtac gcaaggccgc ctataaggga agtcttgctt gaaggagaag atgtagagca gttgttgaag aggagaggaa atagctatat ttatagatgc cctttttgtg gatataaaac tgatgcggat attcaagcgg cgttgaatat agcttgtagg ggatatattt cggataacgc aaaggatgct gtgaaggaag gagaaagaaa attagattac attttggaag ttagaaaatt gtgggagaag aatggagctg ttttgagaag cgccaaattt ttatagtt

[00088] CasY.2 Candidatus vogelbacteria amino acid sequence 1226aa

[00089] (SEQ ID NO: 11):

[00090] MQKVRKTLSEVHKN PYGTKVRNAKTGYSLQI ERLSYTGKEGMRSFKIPLENKNKEVFDEFVKKIRN DYISQV

GLLN LSDWYEHYQEKQEHYSLADFWLDSLRAGVIFAHKETEIKN LISKI RGDKSIVDKFNASIKKKHADLYALVDI KALYDF

LTSDARRGLKTEEEFFNSKRNTLFPKFRKKDN KAVDLWVKKFIGLDN KDKLN FTKKFIGFDPN PQI KYDHTFFFHQDIN F

DLERITTPKELISTYKKFLGKN KDLYGSDETTEDQLKMVLGFHN NHGAFSKYFNASLEAFRGRDNSLVEQII NNSPYWNS

HRKELEKRII FLQVQSKKI KETELGKPHEYLASFGGKFESWVSNYLRQEEEVKRQLFGYEEN KKGQKKFIVGN KQELDKI IR

GTDEYEI KAISKETIGLTQKCLKLLEQLKDSVDDYTLSLYRQLIVELRIRLNVEFQETYPELIGKSE KDKEKDAKN KRADKRYP

QIFKDIKLI PN FLGETKQMVYKKFI RSADILYEGIN FIDQIDKQITQN LLPCFKN DKERIEFTEKQFETLRRKYYLMNSSRFHH

VI EGII NN RKLI EMKKRENSELKTFSDSKFVLSKLFLKKGKKYENEVYYTFYI N PKARDQRRI KIVLDINGN NSVGI LQDLVQ

KLKPKWDDII KKNDMGELI DAIEIEKVRLGILIALYCEHKFKIKKELLSLDLFASAYQYLELEDDPEELSGTN LGRFLQSLVCSE I KGAI NKISRTEYI ERYTVQPMNTEKNYPLLI NKEGKATWHIAAKDDLSKKKGGGTVAMNQKIGKNFFGKQDYKTVFML

QDKRFDLLTSKYHLQFLSKTLDTGGGSWWKN KNI DLNLSSYSFIFEQKVKVEWDLTNLDHPI KIKPSENSDDRRLFVSIPF

VI KPKQTKRKDLQTRVNYMGI DIGEYGLAWTII NI DLKN KKIN KISKQGFIYEPLTHKVRDYVATIKDNQVRGTFGMPDTK

LARLRENAITSLRNQVHDIAMRYDAKPVYEFEISNFETGSNKVKVIYDSVKRADIGR GQN NTEADNTEVN LVWGKTSKQ

FGSQIGAYATSYICSFCGYSPYYEFENSKSGDEEGARDN LYQMKKLSRPSLEDFLQGN PVYKTFRDFDKYKNDQRLQKTG

DKDGEWKTHRGNTAIYACQKCRHISDADIQASYWIALKQVVRDFYKDKEMDGDLIQG DN KDKRKVNELNRLIGVHKD

VPI IN KN LITSLDI NLL

[00091 ] CasY.2 Candidatus vogelbacteria nucleic acid sequence

[00092] (SEQ ID NO: 12):

[00093] a tggtattagg ttttcataat aatcacggcg ctttttctaa gtatttcaac gcgagcttgg aagcttttag ggggagagac aactccttgg ttgaacaaat aattaataat tctccttact ggaatagcca tcggaaagaa ttggaaaaga gaatcatttt tttgcaagtt cagtctaaaa aaataaaaga gaccgaactg ggaaagcctc acgagtatct tgcgagtttt ggcgggaagt ttgaatcttg ggtttcaaac tatttacgtc aggaagaaga ggtcaaacgt caactttttg gttatgagga gaataaaaaa ggccagaaaa aatttatcgt gggcaacaaa caagagctag ataaaatcat cagagggaca gatgagtatg agattaaagc gatttctaag gaaaccattg gacttactca gaaatgttta aaattacttg aacaactaaa agatagtgtc gatgattata cacttagcct atatcggcaa ctcatagtcg aattgagaat cagactgaat gttgaattcc aagaaactta tccggaatta atcggtaaga gtgagaaaga taaagaaaaa gatgcgaaaa ataaacgggc agacaagcgt tacccgcaaa tttttaagga tataaaatta atccccaatt ttctcggtga aacgaaacaa atggtatata agaaatttat tcgttccgct gacatccttt atgaaggaat aaattttatc gaccagatcg ataaacagat tactcaaaat ttgttgcctt gttttaagaa cgacaaggaa cggattgaat ttaccgaaaa acaatttgaa actttacggc gaaaatacta tctgatgaat agttcccgtt ttcaccatgt tattgaagga ataatcaata ataggaaact tattgaaatg aaaaagagag aaaatagcga gttgaaaact ttctccgata gtaagtttgt tttatctaag ctttttctta aaaaaggcaa aaaatatgaa aatgaggtct attatacttt ttatataaat ccgaaagctc gtgaccagcg acggataaaa attgttcttg atataaatgg gaacaattca gtcggaattt tacaagatct tgtccaaaag ttgaaaccaa aatgggacga catcataaag aaaaatgata tgggagaatt aatcgatgca atcgagattg agaaagtccg gctcggcatc ttgatagcgt tatactgtga gcataaattc aaaattaaaa aagaactctt gtcattagat ttgtttgcca gtgcctatca atatctagaa ttggaagatg accctgaaga actttctggg acaaacctag gtcggttttt acaatccttg gtctgctccg aaattaaagg tgcgattaat aaaataagca ggacagaata tatagagcgg tatactgtcc agccgatgaa tacggagaaa aactatcctt tactcatcaa taaggaggga aaagccactt ggcatattgc tgctaaggat gacttgtcca agaagaaggg tgggggcact gtcgctatga atcaaaaaat cggcaagaat ttttttggga aacaagatta taaaactgtg tttatgcttc aggataagcg gtttgatcta ctaacctcaa agtatcactt gcagttttta tctaaaactc ttgatactgg tggagggtct tggtggaaaa acaaaaatat tgatttaaat ttaagctctt attctttcat tttcgaacaa aaagtaaaag tcgaatggga tttaaccaat cttgaccatc ctataaagat taagcctagc gagaacagtg atgatagaag gcttttcgta tccattcctt ttgttattaa accgaaacag acaaaaagaa aggatttgca aactcgagtc aattatatgg ggattgatat cggagaatat ggtttggctt ggacaattat taatattgat ttaaagaata aaaaaataaa taagatttca aaacaaggtt tcatctatga gccgttgaca cataaagtgc gcgattatgt tgctaccatt aaagataatc aggttagagg aacttttggc atgcctgata cgaaactagc cagattgcga gaaaatgcca ttaccagctt gcgcaatcaa gtgcatgata ttgctatgcg ctatgacgcc aaaccggtat atgaatttga aatttccaat tttgaaacgg ggtctaataa agtgaaagta atttatgatt cggttaagcg agctgatatc ggccgaggcc agaataatac cgaagcagac aatactgagg ttaatcttgt ctgggggaag acaagcaaac aatttggcag tcaaatcggc gcttatgcga caagttacat ctgttcattt tgtggttatt ctccatatta tgaatttgaa aattctaagt cgggagatga agaaggggct agagataatc tatatcagat gaagaaattg agtcgcccct ctcttgaaga tttcctccaa ggaaatccgg tttataagac atttagggat tttgataagt ataaaaacga tcaacggttg caaaagacgg gtgataaaga tggtgaatgg aaaacacaca gagggaatac tgcaatatac gcctgtcaaa agtgtagaca tatctctgat gcggatatcc aagcatcata ttggattgct ttgaagcaag ttgtaagaga tttttataaa gacaaagaga tggatggtga tttgattcaa ggagataata aagacaagag aaaagtaaac gagcttaata gacttattgg agtacataaa gatgtgccta taataaataa aaatttaata acatcactcg acataaactt actataga

[00094] CasY.3 Candidatus vogelbacteria amino acid sequence 1200aa

[00095] (SEQID NO: 13):

[00096] MKAKKSFYNQKRKFGKRGYRLHDERIAYSGGIGSMRSIKYELKDSYGIAGLRNRIADATI SDNKWLYGNINLN

DYLEWRSSKTDKQIEDGDRESSLLGFWLEALRLGFVFSKQSHAPNDFNETALQDLFE TLDDDLKHVLDRKKWCDFIKIGT

PKTNDQGRLKKQIKNLLKGNKREEIEKTLNESDDELKEKINRIADVFAKNKSDKYTI FKLDKPNTEKYPRINDVQVAFFCHP

DFEEITERDRTKTLDLIINRFNKRYEITENKKDDKTSNRMALYSLNQGYIPRVLNDL FLFVKDNEDDFSQFLSDLENFFSFS

NEQIKIIKERLKKLKKYAEPIPGKPQLADKWDDYASDFGGKLESWYSNRIEKLKKIP ESVSDLRNNLEKIRNVLKKQNNASK

ILELSQKIIEYIRDYGVSFEKPEIIKFSWINKTKDGQKKVFYVAKMADREFIEKLDL WMADLRSQLNEYNQDNKVSFKKKG

KKIEELGVLDFALNKAKKNKSTKNENGWQQKLSESIQSAPLFFGEGNRVRNEEVYNL KDLLFSEIKNVENILMSSEAEDLK

NIKIEYKEDGAKKGNYVLNVLARFYARFNEDGYGGWNKVKTVLENIAREAGTDFSKY GNNNNRNAGRFYLNGRERQV

FTLIKFEKSITVEKILELVKLPSLLDEAYRDLVNENKNHKLRDVIQLSKTIMALVLS HSDKEKQIGGNYIHSKLSGYNALISKR

DFISRYSVQTTNGTQCKLAIGKGKSKKGNEIDRYFYAFQFFKNDDSKINLKVIKNNS HKNIDFNDNENKINALQVYSSNY

QIQFLDWFFEKHQGKKTSLEVGGSFTIAEKSLTIDWSGSNPRVGFKRSDTEEKRVFV SQPFTLIPDDEDKERRKERMIKTK

NRFIGIDIGEYGLAWSLIEVDNGDKNNRGIRQLESGFITDNQQQVLKKNVKSWRQNQ IRQTFTSPDTKIARLRESLIGSY

KNQLESLMVAKKANLSFEYEVSGFEVGGKRVAKIYDSIKRGSVRKKDNNSQNDQSWG KKGINEWSFETTAAGTSQFCT

HCKRWSSLAIVDIEEYELKDYNDNLFKVKINDGEVRLLGKKGWRSGEKIKGKELFGP VKDAMRPNVDGLGMKIVKRKYL

KLDLRDWVSRYGNMAIFICPYVDCHHISHADKQAAFNIAVRGYLKSVNPDRAIKHGD KGLSRDFLCQEEGKLNFEQIGL

L

[00097] CasY.3 Candidatus vogelbacteria nucleic acid sequence

[00098] (SEQID NO: 14):

[00099] atgaaa gctaaaaaaa gtttttataa tcaaaagcgg aagttcggta aaagaggtta tcgtcttcac gatgaacgta tcgcgtattc aggagggatt ggatcgatgc gatctattaa atatgaattg aaggattcgt atggaattgc tgggcttcgt aatcgaatcg ctgacgcaac tatttctgat aataagtggc tgtacgggaa tataaatcta aatgattatt tagagtggcg atcttcaaag actgacaaac agattgaaga cggagaccga gaatcatcac tcctgggttt ttggctggaa gcgttacgac tgggattcgt gttttcaaaa caatctcatg ctccgaatga ttttaacgag accgctctac aagatttgtt tgaaactctt gatgatgatt tgaaacatgt tcttgatagg aaaaaatggt gtgactttat caagatagga acacctaaga caaatgacca aggtcgttta aaaaaacaaa tcaagaattt gttaaaagga aacaagagag aggaaattga aaaaactctc aatgaatcag acgatgaatt gaaagagaaa ataaacagaa ttgccgatgt ttttgcaaaa aataagtctg ataaatacac aattttcaaa ttagataaac ccaatacgga aaaatacccc agaatcaacg atgttcaggt ggcgtttttt tgtcatcccg attttgagga aattacagaa cgagatagaa caaagactct agatctgatc attaatcggt ttaataagag atatgaaatt accgaaaata aaaaagatga caaaacttca aacaggatgg ccttgtattc cttgaaccag ggctatattc ctcgcgtcct gaatgattta ttcttgtttg tcaaagacaa tgaggatgat tttagtcagt ttttatctga tttggagaat ttcttctctt tttccaacga acaaattaaa ataataaagg aaaggttaaa aaaacttaaa aaatatgctg aaccaattcc cggaaagccg caacttgctg ataaatggga cgattatgct tctgattttg gcggtaaatt ggaaagctgg tactccaatc gaatagagaa attaaagaag attccggaaa gcgtttccga tctgcggaat aatttggaaa agatacgcaa tgttttaaaa aaacaaaata atgcatctaa aatcctggag ttatctcaaa agatcattga atacatcaga gattatggag tttcttttga aaagccggag ataattaagt tcagctggat aaataagacg aaggatggtc agaaaaaagt tttctatgtt gcgaaaatgg cggatagaga attcatagaa aagcttgatt tatggatggc tgatttacgc agtcaattaa atgaatacaa tcaagataat aaagtttctt tcaaaaagaa aggtaaaaaa atagaagagc tcggtgtctt ggattttgct cttaataaag cgaaaaaaaa taaaagtaca aaaaatgaaa atggctggca acaaaaattg tcagaatcta ttcaatctgc cccgttattt tttggcgaag ggaatcgtgt acgaaatgaa gaagtttata atttgaagga ccttctgttt tcagaaatca agaatgttga aaatatttta atgagctcgg aagcggaaga cttaaaaaat ataaaaattg aatataaaga agatggcgcg aaaaaaggga actatgtctt gaatgtcttg gctagatttt acgcgagatt caatgaggat ggctatggtg gttggaacaa agtaaaaacc gttttggaaa atattgcccg agaggcgggg actgattttt caaaatatgg aaataataac aatagaaatg ccggcagatt ttatctaaac ggccgcgaac gacaagtttt tactctaatc aagtttgaaa aaagtatcac ggtggaaaaa atacttgaat tggtaaaatt acctagccta cttgatgaag cgtatagaga tttagtcaac gaaaataaaa atcataaatt acgcgacgta attcaattga gcaagacaat tatggctctg gttttatctc attctgataa agaaaaacaa attggaggaa attatatcca tagtaaattg agcggataca atgcgcttat ttcaaagcga gattttatct cgcggtatag cgtgcaaacg accaacggaa ctcaatgtaa attagccata ggaaaaggca aaagcaaaaa aggtaatgaa attgacaggt atttctacgc ttttcaattt tttaagaatg acgacagcaa aattaattta aaggtaatca aaaataattc gcataaaaac atcgatttca acgacaatga aaataaaatt aacgcattgc aagtgtattc atcaaactat cagattcaat tcttagactg gttttttgaa aaacatcaag ggaagaaaac atcgctcgag gtcggcggat cttttaccat cgccgaaaag agtttgacaa tagactggtc ggggagtaat ccgagagtcg gttttaaaag aagcgacacg gaagaaaaga gggtttttgt ctcgcaacca tttacattaa taccagacga tgaagacaaa gagcgtcgta aagaaagaat gataaagacg aaaaaccgtt ttatcggtat cgatatcggt gaatatggtc tggcttggag tctaatcgaa gtggacaatg gagataaaaa taatagagga attagacaac ttgagagcgg ttttattaca gacaatcagc agcaagtctt aaagaaaaac gtaaaatcct ggaggcaaaa ccaaattcgt caaacgttta cttcaccaga cacaaaaatt gctcgtcttc gtgaaagttt gatcggaagt tacaaaaatc aactggaaag tctgatggtt gctaaaaaag caaatcttag ttttgaatac gaagtttccg ggtttgaagt tgggggaaag agggttgcaa aaatatacga tagtataaag cgtgggtcgg tgcgtaaaaa ggataataac tcacaaaatg atcaaagttg gggtaaaaag ggaattaatg agtggtcatt cgagacgacg gctgccggaa catcgcaatt ttgtactcat tgcaagcggt ggagcagttt agcgatagta gatattgaag aatatgaatt aaaagattac aacgataatt tatttaaggt aaaaattaat gatggtgaag ttcgtctcct tggtaagaaa ggttggagat ccggcgaaaa gatcaaaggg aaagaattat ttggtcccgt caaagacgca atgcgcccaa atgttgacgg actagggatg aaaattgtaa aaagaaaata tctaaaactt gatctccgcg attgggtttc aagatatggg aatatggcta ttttcatctg tccttatgtc gattgccacc atatctctca tgcggataaa caagctgctt ttaatattgc cgtgcgaggg tatttgaaaa gcgttaatcc tgacagagca ataaaacacg gagataaagg tttgtctagg gactttttgt gccaagaaga gggtaagctt aattttgaac aaatagggtt attatgaa [000100] CasY.4 Candidatus parcubacteria amino acid sequence 1210aa

[000101] (SEQ ID NO: 15):

[000102] MSKRHPRISGVKGYRLHAQRLEYTGKSGAMRTIKYPLYSSPSGGRTVPREIVSAIN DDYVGLYGLSN FDDLY

NAEKRN EEKVYSVLDFWYDCVQYGAVFSYTAPGLLKNVAEVRGGSYELTKTLKGSHLYDELQI DKVI KFLNKKEISRANG

SLDKLKKDII DCFKAEYRERHKDQCNKLADDI KNAKKDAGASLGERQKKLFRDFFGISEQSENDKPSFTNPLN LTCCLLPF

DTVNN N RNRGEVLFN KLKEYAQKLDKNEGSLEMWEYIGIGNSGTAFSN FLGEGFLGRLREN KITELKKAMMDITDAW

RGQEQEEELEKRLRILAALTIKLREPKFDNHWGGYRSDI NGKLSSWLQNYI NQTVKIKEDLKGHKKDLKKAKEMIN RFGE

SDTKEEAVVSSLLESIEKIVPDDSADDEKPDI PAIAIYRRFLSDGRLTLN RFVQREDVQEALIKERLEAEKKKKPKKRKKKSD

AEDEKETIDFKELFPHLAKPLKLVPN FYGDSKRELYKKYKNAAIYTDALWKAVEKIYKSAFSSSLKNSFFDTDFDKDFFIKRL

QKIFSVYRRFNTDKWKPIVKNSFAPYCDIVSLAEN EVLYKPKQSRSRKSAAIDKNRVRLPSTEN IAKAGIALARELSVAGFD

WKDLLKKEEHEEYIDLI ELHKTALALLLAVTETQLDISALDFVENGTVKDFMKTRDGN LVLEGRFLEMFSQSIVFSELRGLA

GLMSRKEFITRSAIQTMNGKQAELLYIPHEFQSAKITTPKEMSRAFLDLAPAEFATS LEPESLSEKSLLKLKQMRYYPHYFG

YELTRTGQGI DGGVAENALRLEKSPVKKREIKCKQYKTLGRGQNKIVLYVRSSYYQTQFLEWFLHRPKNV QTDVAVSGSF

LIDEKKVKTRWNYDALTVALEPVSGSERVFVSQPFTI FPEKSAEEEGQRYLGI DIGEYGIAYTALEITGDSAKI LDQN FISDP

QLKTLREEVKGLKLDQRRGTFAMPSTKIARI RESLVHSLRNRI HHLALKHKAKIVYELEVSRFEEGKQKIKKVYATLKKADV

YSEIDADKN LQTTVWGKLAVASEISASYTSQFCGACKKLWRAEMQVDETITTQELIGTVRVI KGGTLI DAIKDFMRPPI FD

EN DTPFPKYRDFCDKHHISKKMRGNSCLFICPFCRANADADIQASQTIALLRYVKEEKKVED YFERFRKLKNI KVLGQMK

Kl

[000103] CasY.4 Candidatus parcubacteria nucleic acid sequence

[000104] (SEQ ID NO: 16):

[000105] atgagtaagc gacatcctag aattagcggc gtaaaagggt accgtttgca tgcgcaacgg ctggaatata ccggcaaaag tggggcaatg cgaacgatta aatatcctct ttattcatct ccgagcggtg gaagaacggt tccgcgcgag atagtttcag caatcaatga tgattatgta gggctgtacg gtttgagtaa ttttgacgat ctgtataatg cggaaaagcg caacgaagaa aaggtctact cggttttaga tttttggtac gactgcgtcc aatacggcgc ggttttttcg tatacagcgc cgggtctttt gaaaaatgtt gccgaagttc gcgggggaag ctacgaactt acaaaaacgc ttaaagggag ccatttatat gatgaattgc aaattgataa agtaattaaa tttttgaata aaaaagaaat ttcgcgagca aacggatcgc ttgataaact gaagaaagac atcattgatt gcttcaaagc agaatatcgg gaacgacata aagatcaatg caataaactg gctgatgata ttaaaaatgc aaaaaaagac gcgggagctt ctttagggga gcgtcaaaaa aaattatttc gcgatttttt tggaatttca gagcagtctg aaaatgataa accgtctttt actaatccgc taaacttaac ctgctgttta ttgccttttg acacagtgaa taacaacaga aaccgcggcg aagttttgtt taacaagctc aaggaatatg ctcaaaaatt ggataaaaac gaagggtcgc ttgaaatgtg ggaatatatt ggcatcggga acagcggcac tgccttttct aattttttag gagaagggtt tttgggcaga ttgcgcgaga ataaaattac agagctgaaa aaagccatga tggatattac agatgcatgg cgtgggcagg aacaggaaga agagttagaa aaacgtctgc ggatacttgc cgcgcttacc ataaaattgc gcgagccgaa atttgacaac cactggggag ggtatcgcag tgatataaac ggcaaattat ctagctggct tcagaattac ataaatcaaa cagtcaaaat caaagaggac ttaaagggac acaaaaagga cctgaaaaaa gcgaaagaga tgataaatag gtttggggaa agcgacacaa aggaagaggc ggttgtttca tctttgcttg aaagcattga aaaaattgtt cctgatgata gcgctgatga cgagaaaccc gatattccag ctattgctat ctatcgccgc tttctttcgg atggacgatt aacattgaat cgctttgtcc aaagagaaga tgtgcaagag gcgctgataa aagaaagatt ggaagcggag aaaaagaaaa aaccgaaaaa gcgaaaaaag aaaagtgacg ctgaagatga aaaagaaaca attgacttca aggagttatt tcctcatctt gccaaaccat taaaattggt gccaaacttt tacggcgaca gtaagcgtga gctgtacaag aaatataaga acgccgctat ttatacagat gctctgtgga aagcagtgga aaaaatatac aaaagcgcgt tctcgtcgtc tctaaaaaat tcattttttg atacagattt tgataaagat ttttttatta agcggcttca gaaaattttt tcggtttatc gtcggtttaa tacagacaaa tggaaaccga ttgtgaaaaa ctctttcgcg ccctattgcg acatcgtctc acttgcggag aatgaagttt tgtataaacc gaaacagtcg cgcagtagaa aatctgccgc gattgataaa aacagagtgc gtctcccttc cactgaaaat atcgcaaaag ctggcattgc cctcgcgcgg gagctttcag tcgcaggatt tgactggaaa gatttgttaa aaaaagagga gcatgaagaa tacattgatc tcatagaatt gcacaaaacc gcgcttgcgc ttcttcttgc cgtaacagaa acacagcttg acataagcgc gttggatttt gtagaaaatg ggacggtcaa ggattttatg aaaacgcggg acggcaatct ggttttggaa gggcgtttcc ttgaaatgtt ctcgcagtca attgtgtttt cagaattgcg cgggcttgcg ggtttaatga gccgcaagga atttatcact cgctccgcga ttcaaactat gaacggcaaa caggcggagc ttctctacat tccgcatgaa ttccaatcgg caaaaattac aacgccaaag gaaatgagca gggcgtttct tgaccttgcg cccgcggaat ttgctacatc gcttgagcca gaatcgcttt cggagaagtc attattgaaa ttgaagcaga tgcggtacta tccgcattat tttggatatg agcttacgcg aacaggacag gggattgatg gtggagtcgc ggaaaatgcg ttacgacttg agaagtcgcc agtaaaaaaa cgagagataa aatgcaaaca gtataaaact ttgggacgcg gacaaaataa aatagtgtta tatgtccgca gttcttatta tcagacgcaa tttttggaat ggtttttgca tcggccgaaa aacgttcaaa ccgatgttgc ggttagcggt tcgtttctta tcgacgaaaa gaaagtaaaa actcgctgga attatgacgc gcttacagtc gcgcttgaac cagtttccgg aagcgagcgg gtctttgtct cacagccgtt tactattttt ccggaaaaaa gcgcagagga agaaggacag aggtatcttg gcatagacat cggcgaatac ggcattgcgt atactgcgct tgagataact ggcgacagtg caaagattct tgatcaaaat tttatttcag acccccagct taaaactctg cgcgaggagg tcaaaggatt aaaacttgac caaaggcgcg ggacatttgc catgccaagc acgaaaatcg cccgcatccg cgaaagcctt gtgcatagtt tgcggaaccg catacatcat cttgcgttaa agcacaaagc aaagattgtg tatgaattgg aagtgtcgcg ttttgaagag ggaaagcaaa aaattaagaa agtctacgct acgttaaaaa aagcggatgt gtattcagaa attgacgcgg ataaaaattt acaaacgaca gtatggggaa aattggccgt tgcaagcgaa atcagcgcaa gctatacaag ccagttttgt ggtgcgtgta aaaaattgtg gcgggcggaa atgcaggttg acgaaacaat tacaacccaa gaactaatcg gcacagttag agtcataaaa gggggcactc ttattgacgc gataaaggat tttatgcgcc cgccgatttt tgacgaaaat gacactccat ttccaaaata tagagacttt tgcgacaagc atcacatttc caaaaaaatg cgtggaaaca gctgtttgtt catttgtcca ttctgccgcg caaacgcgga tgctgatatt caagcaagcc aaacaattgc gcttttaagg tatgttaagg aagagaaaaa ggtagaggac tactttgaac gatttagaaa gctaaaaaac attaaagtgc tcggacagat gaagaaaata tgatag

[000106] CasY.5 Candidatus komeilibacteria amino acid sequence 1192aa

[000107] (SEQ ID NO: 17):

[000108] MAESKQMQCRKCGASMKYEVIGLGKKSCRYMCPDCGN HTSARKIQN KKKRDKKYGSASKAQSQRIAVA GALYPDKKVQTI KTYKYPADLNGEVHDRGVAEKIEQAIQEDEIGLLGPSSEYACWIASQKQSEPYSVVDFWF DAVCAGG VFAYSGARLLSTVLQLSGEESVLRAALASSPFVDDI NLAQAEKFLAVSRRTGQDKLGKRIGECFAEGRLEALGIKDRMREF VQAI DVAQTAGQRFAAKLKI FGISQMPEAKQWNN DSGLTVCI LPDYYVPEEN RADQLVVLLRRLREIAYCMGIEDEAGF EHLGIDPGALSNFSNGN PKRGFLGRLLNN DIIALANN MSAMTPYWEGRKGELI ERLAWLKHRAEGLYLKEPHFGNSWA DHRSRIFSRIAGWLSGCAGKLKIAKDQISGVRTDLFLLKRLLDAVPQSAPSPDFIASISA LDRFLEAAESSQDPAEQVRALY

AFHLNAPAVRSIANKAVQRSDSQEWLI KELDAVDHLEFN KAFPFFSDTGKKKKKGANSNGAPSEEEYTETESIQQPEDA

EQEVNGQEGNGASKNQKKFQRI PRFFGEGSRSEYRILTEAPQYFDMFCNN MRAI FMQLESQPRKAPRDFKCFLQNRL

QKLYKQTFLNARSN KCRALLESVLISWGEFYTYGAN EKKFRLRHEASERSSDPDYVVQQALEIARRLFLFGFEWRDCSAG

ERVDLVEIHKKAISFLLAITQAEVSVGSYNWLGNSTVSRYLSVAGTDTLYGTQLEEF LNATVLSQMRGLAIRLSSQELKDG

FDVQLESSCQDN LQHLLVYRASRDLAACKRATCPAELDPKI LVLPAGAFIASVMKMIERGDEPLAGAYLRHRPHSFGWQ

I RVRGVAEVGMDQGTALAFQKPTESEPFKIKPFSAQYGPVLWLNSSSYSQSQYLDGFLSQP KNWSMRVLPQAGSVRV

EQRVALIWN LQAGKMRLERSGARAFFMPVPFSFRPSGSGDEAVLAPN RYLGLFPHSGGIEYAVVDVLDSAGFKI LERGT

IAVNGFSQKRGERQEEAHREKQRRGISDIGRKKPVQAEVDAAN ELHRKYTDVATRLGCRIVVQWAPQPKPGTAPTAQ

TVYARAVRTEAPRSGNQEDHARMKSSWGYTWSTYWEKRKPEDILGISTQVYWTGGIG ESCPAVAVALLGHIRATSTQ

TEWEKEEVVFGRLKKFFPS

[000109] CasY.5 Candidatus komeilibacteria nucleic acid sequence

[000110] (SEQ ID NO: 18):

[000111] accaaccacc tattgcgtct ttttcgctca ttttagcaaa agtggctgtc tagacataca ggtggaaagg tgagagtaaa gacatggcct gaatagcgtc ctcgtcctcg tctagacata caggtggaaa ggtgagagta aagaccggag cactcatcct ctcactctat tttgtctaga catacaggtg gaaaggtgag agtaaagaca aaccgtgcca cactaaaccg atgagtctag acatacaggt ggaaaggtga gagtaaagac tcaagtaact acctgttctt tcacaagtct agacatacag gtggaaaggt gagagtaaag actcaagtaa ctacctgttc tttcacaagt ctagacctgc aggtggtaag gtgagagtaa agactcaagt aactacctgt tctttcacaa gtctagacct gcaggtggta aggtgagagt aaagactttt atcctcctct ctatgcttct gagtctagac atttaggtgg aaaggtgaga gtaaagactt gtggagatcc atgaacttcg gcagtctaga cctgcaggtg gaaaggtgag agtaaagacg tccttcacac gatcttcctc tgttagtcta ggcctgcagg tggaaaggtg agagtaaaga cgcataagcg taattgaagc tctctccggt ccagaccttg tcgcgcttgt gttgcgacaa aggcggagtc cgcaataagt tctttttaca atgttttttc cataaaaccg atacaatcaa gtatcggttt tgcttttttt atgaaaatat gttatgctat gtgctcaaat aaaaatatca ataaaatagc gtttttttga taatttatcg ctaaaattat acataatcac gcaacattgc cattctcaca caggagaaaa gtcatggcag aaagcaagca gatgcaatgc cgcaagtgcg gcgcaagcat gaagtatgaa gtaattggat tgggcaagaa gtcatgcaga tatatgtgcc cagattgcgg caatcacacc agcgcgcgca agattcagaa caagaaaaag cgcgacaaaa agtatggatc cgcaagcaaa gcgcagagcc agaggatagc tgtggctggc gcgctttatc cagacaaaaa agtgcagacc ataaagacct acaaataccc agcggatctg aatggcgaag ttcatgacag aggcgtcgca gagaagattg agcaggcgat tcaggaagat gagatcggcc tgcttggccc gtccagcgaa tacgcttgct ggattgcttc acaaaaacaa agcgagccgt attcagttgt agatttttgg tttgacgcgg tgtgcgcagg cggagtattc gcgtattctg gcgcgcgcct gctttccaca gtcctccagt tgagtggcga ggaaagcgtt ttgcgcgctg ctttagcatc tagcccgttt gtagatgaca ttaatttggc gcaagcggaa aagttcctag ccgttagccg gcgcacaggc caagataagc taggcaagcg cattggagaa tgtttcgcgg aaggccggct tgaagcgctt ggcatcaaag atcgcatgcg cgaattcgtg caagcgattg atgtggccca aaccgcgggc cagcggttcg cggccaagct aaagatattc ggcatcagtc agatgcctga agccaagcaa tggaacaatg attccgggct cactgtatgt attttgccgg attattatgt cccggaagaa aaccgcgcgg accagctggt tgttttgctt cggcgcttac gcgagatcgc gtattgcatg ggaattgagg atgaagcagg atttgagcat ctaggcattg accctggcgc tctttccaat ttttccaatg gcaatccaaa gcgaggattt ctcggccgcc tgctcaataa tgacattata gcgctggcaa acaacatgtc agccatgacg ccgtattggg aaggcagaaa aggcgagttg attgagcgcc ttgcatggct taaacatcgc gctgaaggat tgtatttgaa agagccacat ttcggcaact cctgggcaga ccaccgcagc aggattttca gtcgcattgc gggctggctt tccggatgcg cgggcaagct caagattgcc aaggatcaga tttcaggcgt gcgtacggat ttgtttctgc tcaagcgcct tctggatgcg gtaccgcaaa gcgcgccgtc gccggacttt attgcttcca tcagcgcgct ggatcggttt ttggaagcgg cagaaagcag ccaggatccg gcagaacagg tacgcgcttt gtacgcgttt catctgaacg cgcctgcggt ccgatccatc gccaacaagg cggtacagag gtctgattcc caggagtggc ttatcaagga actggatgct gtagatcacc ttgaattcaa caaagcattt ccgttttttt cggatacagg aaagaaaaag aagaaaggag cgaatagcaa cggagcgcct tctgaagaag aatacacgga aacagaatcc attcaacaac cagaagatgc agagcaggaa gtgaatggtc aagaaggaaa tggcgcttca aagaaccaga aaaagtttca gcgcattcct cgatttttcg gggaagggtc aaggagtgag tatcgaattt taacagaagc gccgcaatat tttgacatgt tctgcaataa tatgcgcgcg atctttatgc agctagagag tcagccgcgc aaggcgcctc gtgatttcaa atgctttctg cagaatcgtt tgcagaagct ttacaagcaa acctttctca atgctcgcag taataaatgc cgcgcgcttc tggaatccgt ccttatttca tggggagaat tttatactta tggcgcgaat gaaaagaagt ttcgtctgcg ccatgaagcg agcgagcgca gctcggatcc ggactatgtg gttcagcagg cattggaaat cgcgcgccgg cttttcttgt tcggatttga gtggcgcgat tgctctgctg gagagcgcgt ggatttggtt gaaatccaca aaaaagcaat ctcatttttg cttgcaatca ctcaggccga ggtttcagtt ggttcctata actggcttgg gaatagcacc gtgagccggt atctttcggt tgctggcaca gacacattgt acggcactca actggaggag tttttgaacg ccacagtgct ttcacagatg cgtgggctgg cgattcggct ttcatctcag gagttaaaag acggatttga tgttcagttg gagagttcgt gccaggacaa tctccagcat ctgctggtgt atcgcgcttc gcgcgacttg gctgcgtgca aacgcgctac atgcccggct gaattggatc cgaaaattct tgttctgccg gctggtgcgt ttatcgcgag cgtaatgaaa atgattgagc gtggcgatga accattagca ggcgcgtatt tgcgtcatcg gccgcattca ttcggctggc agatacgggt tcgtggagtg gcggaagtag gcatggatca gggcacagcg ctagcattcc agaagccgac tgaatcagag ccgtttaaaa taaagccgtt ttccgctcaa tacggcccag tactttggct taattcttca tcctatagcc agagccagta tctggatgga tttttaagcc agccaaagaa ttggtctatg cgggtgctac ctcaagccgg atcagtgcgc gtggaacagc gcgttgctct gatatggaat ttgcaggcag gcaagatgcg gctggagcgc tctggagcgc gcgcgttttt catgccagtg ccattcagct tcaggccgtc tggttcagga gatgaagcag tattggcgcc gaatcggtac ttgggacttt ttccgcattc cggaggaata gaatacgcgg tggtggatgt attagattcc gcgggtttca aaattcttga gcgcggtacg attgcggtaa atggcttttc ccagaagcgc ggcgaacgcc aagaggaggc acacagagaa aaacagagac gcggaatttc tgatataggc cgcaagaagc cggtgcaagc tgaagttgac gcagccaatg aattgcaccg caaatacacc gatgttgcca ctcgtttagg gtgcagaatt gtggttcagt gggcgcccca gccaaagccg ggcacagcgc cgaccgcgca aacagtatac gcgcgcgcag tgcggaccga agcgccgcga tctggaaatc aagaggatca tgctcgtatg aaatcctctt ggggatatac ctggagcacc tattgggaga agcgcaaacc agaggatatt ttgggcatct caacccaagt atactggacc ggcggtatag gcgagtcatg tcccgcagtc gcggttgcgc ttttggggca cattagggca acatccactc aaactgaatg ggaaaaagag gaggttgtat tcggtcgact gaagaagttc tttccaagct agacgatctt tttaaaaact gggctgctgg ctatcgtatg gtcagtagct cttatttttt tacttgatat atggtattat

[000112] CasY.6 Candidatus kerfeldbacteria amino acid sequence 1287aa

[000113] (SEQ ID NO: 19):

MKRI LNSLKVAALRLLFRGKGSELVKTVKYPLVSPVQGAVEELAEAIRHDN LHLFGQKEIVDLMEKDEGTQVYSVVDFW

LDTLRLGMFFSPSANALKITLGKFNSDQVSPFRKVLEQSPFFLAGRLKVEPAERI LSVEI RKIGKREN RVENYAADVETCFI GQLSSDEKQSIQKLANDIWDSKDHEEQRMLKADFFAIPLI KDPKAVTEEDPEN ETAGKQKPLELCVCLVPELYTRGFGSI

ADFLVQRLTLLRDKMSTDTAEDCLEYVGI EEEKGNGMNSLLGTFLKN LQGDGFEQI FQFMLGSYVGWQGKEDVLRERL

DLLAEKVKRLPKPKFAGEWSGHRMFLHGQLKSWSSN FFRLFNETRELLESI KSDIQHATMLISYVEEKGGYHPQLLSQYR

KLMEQLPALRTKVLDPEI EMTHMSEAVRSYI MIHKSVAGFLPDLLESLDRDKDREFLLSI FPRIPKI DKKTKEIVAWELPGE

PEEGYLFTAN NLFRN FLEN PKHVPRFMAERIPEDWTRLRSAPVWFDGMVKQWQKVVNQLVESPGALYQFNESFLRQ

RLQAMLTVYKRDLQTEKFLKLLADVCRPLVDFFGLGGN DII FKSCQDPRKQWQTVI PLSVPADVYTACEGLAI RLRETLG

FEWKN LKGHEREDFLRLHQLLGNLLFWIRDAKLVVKLEDWMN NPCVQEYVEARKAI DLPLEI FGFEVPIFLNGYLFSELR

QLELLLRRKSVMTSYSVKTTGSPN RLFQLVYLPLN PSDPEKKNSNN FQERLDTPTGLSRRFLDLTLDAFAGKLLTDPVTQE

LKTMAGFYDHLFGFKLPCKLAAMSN HPGSSSKMVVLAKPKKGVASN IGFEPI PDPAHPVFRVRSSWPELKYLEGLLYLPE

DTPLTIELAETSVSCQSVSSVAFDLKN LTTILGRVGEFRVTADQPFKLTPI I PEKEESFIGKTYLGLDAGERSGVGFAIVTVD

GDGYEVQRLGVHEDTQLMALQQVASKSLKEPVFQPLRKGTFRQQERIRKSLRGCYWN FYHALMI KYRAKVVHEESVG

SSGLVGQWLRAFQKDLKKADVLPKKGGKNGVDKKKRESSAQDTLWGGAFSKKEEQQI AFEVQAAGSSQFCLKCGWW

FQLGMREVNRVQESGVVLDWNRSIVTFLIESSGEKVYGFSPQQLEKGFRPDIETFKK MVRDFMRPPMFDRKGRPAAA

YERFVLGRRHRRYRFDKVFEERFGRSALFICPRVGCGN FDHSSEQSAVVLALIGYIADKEGMSGKKLVYVRLAELMAEW

KLKKLERSRVEEQSSAQ

[000114] CasY.6 Candidatus kerfeldbacteria nucleic acid sequence

[000115] (SEQ ID NO: 20):

[000116] atgaagag aattctgaac agtctgaaag ttgctgcctt gagacttctg tttcgaggca aaggttctga attagtgaag acagtcaaat atccattggt ttccccggtt caaggcgcgg ttgaagaact tgctgaagca attcggcacg acaacctgca cctttttggg cagaaggaaa tagtggatct tatggagaaa gacgaaggaa cccaggtgta ttcggttgtg gatttttggt tggataccct gcgtttaggg atgtttttct caccatcagc gaatgcgttg aaaatcacgc tgggaaaatt caattctgat caggtttcac cttttcgtaa ggttttggag cagtcacctt tttttcttgc gggtcgcttg aaggttgaac ctgcggaaag gatactttct gttgaaatca gaaagattgg taaaagagaa aacagagttg agaactatgc cgccgatgtg gagacatgct tcattggtca gctttcttca gatgagaaac agagtatcca gaagctggca aatgatatct gggatagcaa ggatcatgag gaacagagaa tgttgaaggc ggattttttt gctatacctc ttataaaaga ccccaaagct gtcacagaag aagatcctga aaatgaaacg gcgggaaaac agaaaccgct tgaattatgt gtttgtcttg ttcctgagtt gtatacccga ggtttcggct ccattgctga ttttctggtt cagcgactta ccttgctgcg tgacaaaatg agtaccgaca cggcggaaga ttgcctcgag tatgttggca ttgaggaaga aaaaggcaat ggaatgaatt ccttgctcgg cacttttttg aagaacctgc agggtgatgg ttttgaacag atttttcagt ttatgcttgg gtcttatgtt ggctggcagg ggaaggaaga tgtactgcgc gaacgattgg atttgctggc cgaaaaagtc aaaagattac caaagccaaa atttgccgga gaatggagtg gtcatcgtat gtttctccat ggtcagctga aaagctggtc gtcgaatttc ttccgtcttt ttaatgagac gcgggaactt ctggaaagta tcaagagtga tattcaacat gccaccatgc tcattagcta tgtggaagag aaaggaggct atcatccaca gctgttgagt cagtatcgga agttaatgga acaattaccg gcgttgcgga ctaaggtttt ggatcctgag attgagatga cgcatatgtc cgaggctgtt cgaagttaca ttatgataca caagtctgta gcgggatttc tgccggattt actcgagtct ttggatcgag ataaggatag ggaatttttg ctttccatct ttcctcgtat tccaaagata gataagaaga cgaaagagat cgttgcatgg gagctaccgg gcgagccaga ggaaggctat ttgttcacag caaacaacct tttccggaat tttcttgaga atccgaaaca tgtgccacga tttatggcag agaggattcc cgaggattgg acgcgtttgc gctcggcccc tgtgtggttt gatgggatgg tgaagcaatg gcagaaggtg gtgaatcagt tggttgaatc tccaggcgcc ctttatcagt tcaatgaaag ttttttgcgt caaagactgc aagcaatgct tacggtctat aagcgggatc tccagactga gaagtttctg aagctgctgg ctgatgtctg tcgtccactc gttgattttt tcggacttgg aggaaatgat attatcttca agtcatgtca ggatccaaga aagcaatggc agactgttat tccactcagt gtcccagcgg atgtttatac agcatgtgaa ggcttggcta ttcgtctccg cgaaactctt ggattcgaat ggaaaaatct gaaaggacac gagcgggaag attttttacg gctgcatcag ttgctgggaa atctgctgtt ctggatcagg gatgcgaaac ttgtcgtgaa gctggaagac tggatgaaca atccttgtgt tcaggagtat gtggaagcac gaaaagccat tgatcttccc ttggagattt tcggatttga ggtgccgatt tttctcaatg gctatctctt ttcggaactg cgccagctgg aattgttgct gaggcgtaag tcggtgatga cgtcttacag cgtcaaaacg acaggctcgc caaataggct cttccagttg gtttacctac ctctaaaccc ttcagatccg gaaaagaaaa attccaacaa ctttcaggag cgcctcgata cacctaccgg tttgtcgcgt cgttttctgg atcttacgct ggatgcattt gctggcaaac tcttgacgga tccggtaact caggaactga agacgatggc cggtttttac gatcatctct ttggcttcaa gttgccgtgt aaactggcgg cgatgagtaa ccatccagga tcctcttcca aaatggtggt tctggcaaaa ccaaagaagg gtgttgctag taacatcggc tttgaaccta ttcccgatcc tgctcatcct gtgttccggg tgagaagttc ctggccggag ttgaagtacc tggaggggtt gttgtatctt cccgaagata caccactgac cattgaactg gcggaaacgt cggtcagttg tcagtctgtg agttcagtcg ctttcgattt gaagaatctg acgactatct tgggtcgtgt tggtgaattc agggtgacgg cagatcaacc tttcaagctg acgcccatta ttcctgagaa agaggaatcc ttcatcggga agacctacct cggtcttgat gctggagagc gatctggcgt tggtttcgcg attgtgacgg ttgacggcga tgggtatgag gtgcagaggt tgggtgtgca tgaagatact cagcttatgg cgcttcagca agtcgccagc aagtctctta aggagccggt tttccagcca ctccgtaagg gcacatttcg tcagcaggag cgcattcgca aaagcctccg cggttgctac tggaatttct atcatgcatt gatgatcaag taccgagcta aagttgtgca tgaggaatcg gtgggttcat ccggtctggt ggggcagtgg ctgcgtgcat ttcagaagga tctcaaaaag gctgatgttc tgcccaagaa gggtggaaaa aatggtgtag acaaaaaaaa gagagaaagc agcgctcagg ataccttatg gggaggagct ttctcgaaga aggaagagca gcagatagcc tttgaggttc aggcagctgg atcaagccag ttttgtctga agtgtggttg gtggtttcag ttggggatgc gggaagtaaa tcgtgtgcag gagagtggcg tggtgctgga ctggaaccgg tccattgtaa ccttcctcat cgaatcctca ggagaaaagg tatatggttt cagtcctcag caactggaaa aaggctttcg tcctgacatc gaaacgttca aaaaaatggt aagggatttt atgagacccc ccatgtttga tcgcaaaggt cggccggccg cggcgtatga aagattcgta ctgggacgtc gtcaccgtcg ttatcgcttt gataaagttt ttgaagagag atttggtcgc agtgctcttt tcatctgccc gcgggtcggg tgtgggaatt tcgatcactc cagtgagcag tcagccgttg tccttgccct tattggttac attgctgata aggaagggat gagtggtaag aagcttgttt atgtgaggct ggctgaactt atggctgagt ggaagctgaa gaaactggag agatcaaggg tggaagaaca gagctcggca caataa

[000117] Any of the gene editor effectors herein can also be tagged with Tev or any other suitable homing protein domains. According to Wolfs, et al. (Proc Natl Acad Sci USA. 2016 Dec 27; 113(52): 14988-14993. doi: 10.1073/pnas.l616343114. Epub 2016 Dec 12), 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.

[000118] The present invention provides for a composition for treating a lysogenic virus (budding virus) including a vector encoding two or more cloaked CRISPR-associated nucleases such as cloaked Cas9, cloaked Cpfl, cloaked C2cl, cloaked C2c3, cloaked TevCas9, cloaked Archaea Cas9, cloaked CasY.l - CasY.6, and cloaked CasX gRNAs, cloaked Argonaute endonuclease gDNAs and other cloaked gene editors that target viral DNA, and RNA editors such as cloaked C2c2, or any other composition that targets RNA such as siRNA/miRNA/shRNAs/RNAi. Preferably, the composition includes isolated nucleic acid encoding a cloaked CRISPR-associated endonuclease (cloaked 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. Most preferably, 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.

[000119] The present invention also provides for a composition for treating a lytic virus, including a vector encoding two or more cloaked CRISPR-associated nucleases such as cloaked Cas9, cloaked Cpfl, cloaked C2cl, cloaked C2c3, cloaked TevCas9, cloaked Archaea Cas9, cloaked CasY.l - CasY.6, and cloaked CasX gRNAs, cloaked Argonaute endonuclease gDNAs and other cloaked 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) cloaked CRISPR-associated nucleases such as cloaked Cas9, cloaked Cpfl, cloaked C2cl, cloaked C2c3, cloaked TevCas9, cloaked Archaea Cas9, cloaked CasY.l - CasY.6, and cloaked CasX gRNAs, cloaked Argonaute endonuclease gDNAs and other cloaked gene editors that target RNAs (viral mRNA), such as cloaked C2c2, that translate (non coding or coding) viral proteins involved with the formation of virions. Preferably, the composition includes isolated nucleic acid encoding a cloaked CRISPR-associated endonuclease (cloaked Cas9), two or more gRNAs that are complementary to a target DNA sequence in a virus, and either the siRNA/miRNA/shRNAs/RNAi or cloaked CRISPR-associated nucleases such as cloaked Cas9, cloaked Cpfl, cloaked C2cl, cloaked C2c3, cloaked TevCas9, cloaked Archaea Cas9, cloaked CasY.l - CasY.6, and cloaked CasX gRNAs, cloaked Argonaute endonuclease gDNAs and other cloaked 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. As shown in FIGURE 1, lytic and lysogenic viruses need to be treated in different ways. While 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. For example, Nelles, et al. (Cell, Volume 165, Issue 2, p. 488-496, April 7, 2016) 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.

[000120] The siRNA and cloaked 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 siRNAs 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. For example, 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. Preferably, 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). Alternatively, 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. For example, 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.

[000121] The present invention also provides for a composition for treating both lysogenic and lytic viruses, including a vector encoding two or more cloaked CRISPR-associated nucleases such as cloaked Cas9, cloaked Cpfl, cloaked C2cl, cloaked C2c3, cloaked TevCas9, cloaked Archaea Cas9, cloaked CasY.l - CasY.6, and cloaked CasX gRNAs, cloaked Argonaute endonuclease gDNAs, cloaked C2c2, cloaked C2cl, and other cloaked gene editors that target viral RNA. Preferably, the composition includes isolated nucleic acid encoding a cloaked CRISPR-associated endonuclease (cloaked 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 cloaked 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.

[000122] The present invention provides for a composition for treating lytic viruses, including a vector encoding two or more cloaked CRISPR-associated nucleases such as cloaked Cas9, cloaked Cpfl, cloaked C2cl, cloaked C2c3, cloaked TevCas9, cloaked Archaea Cas9, cloaked CasY.l - CasY.6, and cloaked CasX gRNAs, cloaked 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. Preferably, the composition includes isolated nucleic acid encoding a cloaked CRISPR-associated endonuclease (cloaked 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.

[000123] Various viruses 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. [000124] TABLE 2 lists viruses in the picornaviridae/hepeviridae/flaviviridae families and their method of replication.

TABLE 2

[000125] It should be noted that Hepatitis D propagates only in the presence of Hepatitis B, therefore, 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 cloaked Cas9, cloaked Cpfl, cloaked C2cl, cloaked C2c3, cloaked TevCas9, cloaked Archaea Cas9, cloaked CasY.l - CasY.6, and cloaked CasX gRNAs, cloaked Argonaute endonuclease gDNAs and other cloaked gene editors to treat the lysogenic virus and siRNAs/miRNAs/shRNAs/RNAi to treat the lytic virus.

[000126] TABLE 3 lists viruses in the herpesviridae family and their method of replication.

TABLE 3

[000127] TABLE 4 lists viruses in the orthomyxoviridae family and their method of replication.

TABLE 4

-ssRNA viral genome

[000128] TABLE 5 lists viruses in the retroviridae family and their method of replication.

TABLE 5

[000129] TABLE 6 lists viruses in the papillomaviridae family and their method of replication.

TABLE 6

HPV family dsDNA viral genome Budding from desquamating cells (semi-lysogenic) [000130] TABLE 7 lists viruses in the flaviviridae family and their method of replication.

TABLE 7

[000131] TABLE 8 lists viruses in the reoviridae family and their method of replication.

TABLE 8

[000132] TABLE 9 lists viruses in the rhabdoviridae family and their method of replication.

TABLE 9

[000133] TABLE 10 lists viruses in the bunyanviridae family and their method of replication.

TABLE 10

[000134] TABLE 11 lists viruses in the arenaviridae family and their method of replication.

TABLE 11

[000135] TABLE 12 lists viruses in the filoviridae family and their method of replication.

TABLE 12

[000136] TABLE 13 lists viruses in the polyomaviridae family and their method of replication. TABLE IB

[000137] The 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.

[000138] In the present invention, when any of the compositions are contained within an expression vector, the CRISPR endonuclease 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.

[000139] 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. Generally, 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. Numerous vectors and expression systems are commercially available from such corporations as Novagen (Madison, Wl), Clontech (Palo Alto, CA), Stratagene (La Jolla, CA), and Invitrogen/Life Technologies (Carlsbad, CA).

[000140] 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. For example, a marker can confer biocide resistance, such as resistance to an antibiotic (e.g., kanamycin, G418, bleomycin, or hygromycin). As noted above, 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 Flag™ tag (Kodak, New Haven, CT) sequences typically are expressed as a fusion with the encoded polypeptide. Such tags can be inserted anywhere within the polypeptide, including at either the carboxyl or amino terminus.

[000141] 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. coli plasmids col El, pCRl, pBR322, pMal-C2, pET, pGEX, pMB9 and their derivatives, plasmids such as RP4; phage DNAs, e.g., the numerous derivatives of phage 1, e.g., NM989, and other phage DNA, e.g., M13 and filamentous single stranded phage DNA; yeast plasmids such as the 2m plasmid or derivatives thereof, vectors useful in eukaryotic cells, such as vectors useful in insect or mammalian cells; vectors derived from combinations of plasmids and phage DNAs, such as plasmids that have been modified to employ phage DNA or other expression control sequences.

[000142] Yeast expression systems can also be used. For example, 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.

[000143] The vector can also include a regulatory region. The term "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.

[000144] As used herein, the term "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. For example, to bring a coding sequence under the control of a promoter, 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). 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.

[000145] 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. 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. As described and illustrated in more detail below, 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.

[000146] 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). Modified viral vectors in which a polynucleotide to be delivered is carried on the outside of the viral particle have also been described (see, e.g., Curiel, D T, et al. PNAS 88: 8850-8854, 1991).

[000147] 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. In such cases, the viral vector comprises a strong eukaryotic promoter operably linked to the polynucleotide e.g., a cytomegalovirus (CMV) promoter. The recombinant viral vector can include one or more of the polynucleotides therein, preferably about one polynucleotide. In some embodiments, 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. In embodiments in which the polynucleotide is to be administered with a non-viral vector, 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. [000148] Additional vectors include viral vectors, fusion proteins and chemical conjugates. 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. Press, Oxford England) (1995); Geller, A.l. et al., Proc Natl. Acad. Sci. : U.S.A.:90 7603 (1993); Geller, A.I., et al., Proc Natl. Acad. Sci USA: 87:1149 (1990)], Adenovirus Vectors [LeGal LaSalle et al., Science, 259:988 (1993); Davidson, et al., Nat. Genet. 3: 219 (1993); Yang, et al., J. Virol. 69: 2004 (1995)] and Adeno-associated Virus Vectors [Kaplitt, M.G., et al., Nat. Genet. 8:148 (1994)].

[000149] Pox viral vectors introduce the gene into the cell's 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. Other 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 which is active in lymphoid cells, mouse mammary tumor virus control region which is active in testicular, breast, lymphoid and mast cells, albumin gene control region which is active in liver, alpha- fetoprotein gene control region which is active in liver, alpha 1-antitrypsin gene control region which is active in the liver, beta-globin gene control region which is active in myeloid cells, myelin basic protein gene control region which is active in oligodendrocyte cells in the brain, myosin light chain-2 gene control region which is active in skeletal muscle, and gonadotropic releasing hormone gene control region which is active in the hypothalamus. Certain proteins can be 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.

[000150] If desired, the polynucleotides of the invention can also be used with a microdelivery vehicle such as cationic liposomes and adenoviral vectors. For a review of the procedures for liposome preparation, targeting and delivery of contents, see Mannino and Gould-Fogerite, BioTechniques, 6:682 (1988). See also, Feigner and Holm, Bethesda Res. Lab. Focus, 11(2):21 (1989) and Maurer, R.A., Bethesda Res. Lab. Focus, 11(2):25 (1989).

[000151] Replication-defective recombinant adenoviral vectors, can be produced in accordance with known techniques. See, Quantin, et al., Proc. Natl. 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).

[000152] 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.

[000153] As described above, the 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. For example, 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. When employed as pharmaceuticals, any of the nucleic acids and vectors can be administered in the form of pharmaceutical compositions. These 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. 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. Pharmaceutical 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.

[000154] This invention also includes pharmaceutical compositions which contain, as the active ingredient, nucleic acids and vectors described herein in combination with one or more pharmaceutically acceptable carriers. The terms "pharmaceutically acceptable" (or "pharmacologically 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. The term "pharmaceutically acceptable carrier," as used herein, 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. In making the compositions of the invention, 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. When the excipient 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. Thus, 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. As is known in the art, the type of diluent can vary depending upon the intended route of administration. The resulting compositions can include additional agents, such as preservatives. In some embodiments, the carrier can be, or can include, a lipid-based or polymer-based colloid. In some embodiments, the carrier material can be a colloid formulated as a liposome, a hydrogel, a microparticle, a nanoparticle, or a block copolymer micelle. As noted, the carrier material can form a capsule, and that material may be a polymer-based colloid.

[000155] 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. For example, PLGA (poly-lacto-co-glycolide) microparticles approximately 1-10 pm in diameter can be used. 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. Alternatively, one can prepare 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. In the relevant polynucleotides (e.g., expression vectors) the nucleic acid sequence encoding 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.

[000156] In some embodiments, the 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.

[000157] 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). 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).

[000158] The present invention provides for a method of preventing antibody neutralizing effects with gene editors in humans, by cloaking a gene editor, and administering the cloaked gene editor to a human without generating antibodies to the cloaked gene editor. The gene editor can be any of those described above. Cloaking can occur by changing any suitable proteins, DNA, or RNA of the gene editor while still maintaining the gene editor's effectivity. For example, as described above, chemical changes can be introduced to the gene editor such as introducing chemical changes to the gene editor chosen from the group consisting of introducing glycosylation, eliminating oxidative sites, changing proteins that generate non-natural amino acids, and combinations thereof. By administering a cloaked form of the gene editor, antibodies do not form against the gene editor in the body or any cells that the gene editor has edited, preventing it from providing a therapeutic effect.

[000159] The present invention provides for a method of treating a lysogenic virus, by administering a composition including two or more cloaked CRISPR-associated nucleases such as cloaked Cas9, cloaked Cpfl, cloaked C2cl, and cloaked TevCas9 gRNAs, cloaked Argonaute endonuclease gDNAs and other cloaked gene editors that target viral DNA to an individual having a lysogenic virus 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.

[000160] In any of the methods described herein, 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 a viral infection and placed in culture, and then treated with the composition). Useful vector systems and formulations are described above. In some embodiments 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. In any of the methods described herein, 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.

[000161] Any composition 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. In terms of 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. In a further example, an aerosol preparation of a composition can be given to a host by inhalation.

[000162] 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.

[000163] 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). For example, 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. It is also noted that the frequency of treatment can be variable. For example, the present compounds can be administered once (or twice, three times, etc.) daily, weekly, monthly, or yearly.

[000164] 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. In addition, 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.

[000165] The present invention also provides for a method for treating a lytic virus, including administering a vector encoding two or more cloaked CRISPR-associated nucleases such as cloaked Cas9, cloaked Cpfl, cloaked C2cl, cloaked C2c3, cloaked TevCas9, cloaked Archaea Cas9, cloaked CasY.l - CasY.6, and cloaked CasX gRNAs, cloaked Argonaute endonuclease gDNAs and other cloaked gene editors that target viral DNA and a composition chosen from siRNAs/miRNAs/shRNAs/RNAi and cloaked CRISPR-associated nucleases such as cloaked Cas9, cloaked Cpfl, cloaked C2cl, cloaked C2c3, cloaked TevCas9, cloaked Archaea Cas9, cloaked CasY.l - CasY.6, and cloaked CasX gRNAs, cloaked Argonaute endonuclease gDNAs and other cloaked gene editors that target viral RNA to an individual having a lytic virus, and inactivating the lytic virus. 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.

[000166] 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 cloaked CRISPR- associated nucleases such as cloaked Cas9, cloaked Cpfl, cloaked C2cl, cloaked C2c3, cloaked TevCas9, cloaked Archaea Cas9, cloaked CasY.l - CasY.6, and cloaked CasX gRNAs, cloaked Argonaute endonuclease gDNAs and other cloaked gene editors that target viral RNA to an individual having a lysogenic virus and lytic virus, and inactivating the lysogenic virus and lytic virus. 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.

[000167] At the point of infection or when the virus has entered the cytoplasm, it can contain an RNA-based genome that is non-integrating (not converted to DNA) yet contributes to lysogenic type replication cycle. At this upstream point, the viral genome can be eliminated. On the other hand, the approach can be utilized to also target viral mRNA which occurs downstream (as the genome is translated). Although Argonaute is cited throughout the art, to this date it has not been modified to recognize RNA molecules.

[000168] The present invention provides for a method for treating lytic viruses, by administering a composition including a vector encoding two or more cloaked CRISPR-associated nucleases such as cloaked Cas9, cloaked Cpfl, cloaked C2cl, cloaked C2c3, cloaked TevCas9, cloaked Archaea Cas9, cloaked CasY.l - CasY.6, and cloaked CasX gRNAs, cloaked Argonaute endonuclease gDNAs and other cloaked gene editors that target viral RNA and siRNA/miRNAs/shRNAs/RNAi that target viral RNA to an individual having a lytic virus, and inactivating the lytic virus. 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.

[000169] The invention is further described in detail by reference to the following experimental examples. These examples are provided for the purpose of illustration only and are not intended to be limiting unless otherwise specified. Thus, the invention should in no way be construed as being limited to the following examples, but rather, should be construed to encompass any and all variations which become evident as a result of the teaching provided herein.

[000170] EXAMPLE 1

[000171 ] Expression of the bacterium Cas9 gene in human cells may activate an immune response leading to the destruction of the Cas9 protein before it has a chance to perform its function or cause cellular toxicity and cell lysis. Therefore, it is needed to identify potential epitopes within Cas9 that cause an immune response when expressed in human cells. Optimization of Cas9 function is the first step for increasing the efficiency of CRISPR-based editing specific sites within the human genome. A modified Cas9 protein will also be an essential tool in understanding the relationship between Cas9 protein half-life and off-target effects as well as in determining the utility of CRISPRi (interference) as a form of genetic regulation in human cells.

[000172] Aim 1: Identification of potential antigen epitopes on SaCas9 that react with human IgG and/or IgM immunoglobulins. In order to increase the efficiency of the CRISPR-Cas9 editing function, the level of expression and protein half-life of Cas9 is optimized in human host cells. The goal of this aim is to identify any potential epitopes within SaCas9 that are reactive against human serum. This protocol is to outline a functional approach to identifying potential epitope sites within SaCas9 that react with immunoglobulins in human serum. Identification and modification of SaCas9-epitope(s) will aid in the engineering of more stable and efficient Cas9 molecules for expression in human cells.

[000173] Expression of SaCas9 in human extracts:

[000174] SaCas9 will be expressed and translated via a 1-step in vitro translation system (ThermoFisher, Cat#:88881). The extent of expression will be determined by SDS-PAGE stained with coommassie blue. If the level of translation is low (as defined by being less than 5x more abundant than the most intense extract protein band), then the SaCas9 will be modified to include an epitope tag to facilitate purification.

[000175] A procedure to isolate Cas9 nuclease from whole cell bacterial extracts:

[000176] In the event that in vitro translation does not provide enough starting material, Cas9 enzymes will be isolated from bacterial extracts. Microbial strains will be purchased from ATCC

(commercially available), grow under the proper conditions, stressed with bacteriophage infection and processed to produce whole cell extracts. Bacteriophage infection has been demonstrated to increase the expression of CRISPR regions, along with the linked Editor, within bacterial genomes. The whole cell extracts will be pre-cleared for non-guide RNA and non-specific bead binding by incubation of the cell extracts with agarose beads. The flowthrough will be incubated with agarose beads conjugated to the human IgGs.

[000177] SoCos9 Proteose Cleavage:

[000178] In vitro translated SaCas9 is cleaved to completion (in parallel) with proteases (tentatively, Asp-N and Glutamyl Endopeptidase, but open to others). Under these conditions, the proteases should self cleave and become inactive. A small sample amount (whole extract) of the Cas9 extract will be removed to analyze by HPLC (214nm). The cleaved Cas9 peptide extracts will be incubated with human IgG or human IgM associated agarose beads (or the agarose beads alone to determine "sticky" proteins that associate with only the beads) to allow any potential epitopes to associate with agarose conjugated Ig's. After a suitable incubation time, an analogous amount (as compared to the whole extract) of the flow-through will be set aside and later analyzed by HPLC. The beads will be washed multiple times with the same volume of buffer as the initial whole extract. Analogous amounts (as compared to the whole extract) of the wash flow-through will be set aside for analysis by HPLC. With each wash step, the intensity of the 214nm chromatogram will decrease. When this intensity is below the level of accurate detectability (based on the HPLC data), then the bound peptides will be eluted from the column with a low pH solution (<3). The eluted peptide solution will then be neutralized and submitted for mass spectrometry. A table with the theoretical masses after protease cleavage is given below (TABLE 14). A positive result will be the multiple detection of peptides of the proper predicted masses. A comparison of all positive peptides will narrow the number of amino acids comprising the epitope. The epitope can be narrowed further using this protocol and other specific proteases. This protocol can also determine whether the epitope interacts with the human IgG and the human IgM immunoglobulins. TABLE 14

[000179] SaCas9 epitope necessity:

[000180] Upon the identification of an SaCas9 epitope(s), epitope deleted constructs of SaCas9 are constructed (in the human in vitro translation system vectors). After in vitro expression, these deleted constructs are separated by SDS-PAGE, transferred to nitrocellulose and probed with human serum and compared to the full length SaCas9 in vitro translated protein. A negative result, i.e., the inability to detect a protein band from a deletion construct as compared to detection of a WT SaCas9 protein band, will be considered a candidate epitope and genetic engineering will be performed on the epitope sequence in an attempt to limit its antigenicity.

[000181 ] Aim 2: Identification of amino acid residues of SaCas9 that affect antigen-epitope recognition but have no affect on SaCas9 gRNA binding/DNA association and/or DNA nuclease activity. This protocol will allow the identification of amino acid residues of SaCas9 that affect antigen- epitope recognition but have no affect on SaCas9 gRNA binding/DNA association and/or DNA nuclease activity. The goal of Aim 2 is to optimize the protein half-life of SaCas9 in human cells by the screening of Cas9 alleles and point mutations identified in Aim 1 using a yeast in vivo system. To this end, random and site-directed mutagenesis of SaCas9 and assay for Cas9 function in the yeast Saccharomyces cerevisiae are performed. The assay is able to discern mutations that reduce or eliminate DNA nuclease activity with those that affect gRNA-dependent DNA association by Cas9. In this system, a yeast colony color sectoring assay can differentiate between Cas9 alleles that possess nuclease activity and those alleles that lack nuclease activity but still interfere with gene expression.

[000182] Strategy:

[000183] Yeast colony analysis can be a powerful tool as a screening device. To this end, a diploid S. cerevisiae strain (MATa/a ura3-52/ura3-52 Ieu2-3,112/leu2-3,112) is engineered to express the bacterial CSY4 gene (fused with a nuclear localization sequence, NLS, to give CSY4-NLS) from a constitutive tet promotor. The CSY4 protein recognizes RNA hairpins and cleaves the transcript 28nts from the hairpin. Using this approach, several gRNAs, each with its own hairpin, can be sub-cloned as a single cistronic message in one plasmid. The CSY4 protein cleaves this long message into individual gRNAs suitable to use with Cas9 for genome editing. The plasmid carrying all the gRNAs will be marked with the LEU2 auxotrophic marker gene. The tet-driven CSY4-NLS gene can be inserted into the yeast genome at the HIS3 locus.

[000184] To reduce the number of false positives, a diploid yeast strain can be used ensuring that both copies of the color selection markers, in this case, the ADE2 gene, have to be edited in order for there to be a color change. In addition, gRNAs representing other auxotrophic markers (MET15 and LYS2) will also be expressed. Finally, the plasmid will also express gRNAs representing the CAN1 gene. Mutation of CAN1 is a negative selection that makes the yeast cell resistant to L-canavanine. With this entire set-up, Cas9 alleles expressed from a plasmid marked with the URA3 gene will monitored for editing activity by first selecting transformed colonies that are resistant to L-canavanine. These colonies can then be examined for the ability to grow on medium lacking methionine and lysine. Candidates can then be assayed on rich medium for the ability to turn red in color (for this assay, the number of cells tested has to be low to allow single isolates to grow). Colonies that are red in color will then be grown on medium containing 5'-Fluoroorodic Acid (5'-FOA) which acts as a negative selection for the URA3 gene, thereby selecting for cells that no longer possess the plasmid expressing the Cas9 allele. Cells that grow on the medium containing 5'-FOA can then be retested on rich medium.

[000185] There are three possible scenarios: [000186] 1. After the first examination on rich medium, the colonies do not turn red, i.e., the colonies are white. These will be discarded.

[000187] 2. After the first examination on rich medium, the colonies are red. These are candidate alleles for Cas9 and will be grown on medium containing 5'-FOA.

[000188] 3. After the first examination on rich medium, the colonies are sectored, i.e., containing wedges of red and white. These are also Cas9 allele candidates and will be grown on medium containing 5'-FOA to select for cells that no longer contain the URA3-based plasmid.

[000189] After growth on 5'-FOA, the cells will again be grown on rich medium leading to two possible outcomes:

[000190] 1. The colonies are white. This demonstrates that the Cas9 allele was interfering with ADE2 expression and not necessarily editing the DNA. These colonies can be re-examined on medium lacking methionine and lysine to determine if these auxotrophic requirements were also reverted.

[000191] 2. The colonies are red. This denotes that the ADE2 was indeed edited (mutated).

[000192] At this point, the plasmids expressing the Cas9 alleles will be rescued and the Cas9 sequence determined. As an alternative, known alleles of Cas9 can be examined using this screen, however, after the screening steps, the plasmids expressing the alleles should be rescued and sequenced to ensure there are no other mutations which gave rise to the phenotypes.

[000193] Conclusion:

[000194] Although the aims of this proposal are related, the establishment of the yeast screening protocol will be useful for future studies where the activity and function of a Cas9 allele is in question. Taken together, this protocol will identify potentially antigenic domains of Cas9 expressed in human cells. Changing amino acids in these domains will abrogate the antigenicity and should also increase the half-life of the Cas9 allele making that particular allele more powerful as a gene editing tool.

[000195] Throughout this application, various publications, including United States patents, are referenced by author and year and patents by number. Full citations for the publications are listed below. The disclosures of these publications and patents in their entireties are hereby incorporated by reference into this application in order to more fully describe the state of the art to which this invention pertains.

[000196] The invention has been described in an illustrative manner, and it is to be understood that the terminology, which has been used is intended to be in the nature of words of description rather than of limitation.

[000197] Obviously, many modifications and variations of the present invention are possible in light of the above teachings. It is, therefore, to be understood that within the scope of the appended claims, the invention can be practiced otherwise than as specifically described.