Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
CRISPR-ASSOCIATED (CAS) PROTEINS WITH REDUCED IMMUNOGENICITY
Document Type and Number:
WIPO Patent Application WO/2017/081288
Kind Code:
A1
Abstract:
The invention relates to methods of reducing the immunogenicity of CRISPR-associated (Cas) proteins and the modified Cas proteins produced therefrom. In addition, the invention relates to methods for cell and gene therapy, including any and all genetic modifications and alterations of gene expression (and/or genetic elements) made in-vivo or ex- vivo using Cas proteins with reduced immunogenicity.

Inventors:
PAYNE THOMAS (GB)
ZURDO JESÚS (GB)
SMITH NOEL HILLIER (GB)
Application Number:
PCT/EP2016/077487
Publication Date:
May 18, 2017
Filing Date:
November 11, 2016
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
LONZA AG (CH)
International Classes:
C12N9/22; C12N15/52
Domestic Patent References:
WO2014204725A12014-12-24
Foreign References:
US20140068797A12014-03-06
US20140273037A12014-09-18
US20140273226A12014-09-18
US20140295556A12014-10-02
US20140295557A12014-10-02
US20140349405A12014-11-27
US20150045546A12015-02-12
US20150071898A12015-03-12
US20150071899A12015-03-12
US20150071906A12015-03-12
US20130280797A12013-10-24
US20120077429A12012-03-29
US20110280797A12011-11-17
US20090305626A12009-12-10
US8298054B22012-10-30
US7629167B22009-12-08
US5656491A1997-08-12
US7702465B22010-04-20
EP1516275A22005-03-23
Other References:
DAN WANG ET AL: "Adenovirus-Mediated Somatic Genome Editing of Pten by CRISPR/Cas9 in Mouse Liver in Spite of Cas9-Specific Immune Responses", HUMAN GENE THERAPY, vol. 26, no. 7, 18 June 2015 (2015-06-18), US, pages 432 - 442, XP055340813, ISSN: 1043-0342, DOI: 10.1089/hum.2015.087
NISHIMASU HIROSHI ET AL: "Crystal Structure of Cas9 in Complex with Guide RNA and Target DNA", CELL, CELL PRESS, US, vol. 156, no. 5, 13 February 2014 (2014-02-13), pages 935 - 949, XP028667665, ISSN: 0092-8674, DOI: 10.1016/J.CELL.2014.02.001
BENJAMIN P. KLEINSTIVER ET AL: "Engineered CRISPR-Cas9 nucleases with altered PAM specificities", NATURE, vol. 523, no. 7561, 22 June 2015 (2015-06-22), United Kingdom, pages 481 - 485, XP055293257, ISSN: 0028-0836, DOI: 10.1038/nature14592
TANGRI SHABNAM ET AL: "Rationally engineered therapeutic proteins with reduced immunogenicity", THE JOURNAL OF IMMUNOLOGY, THE AMERICAN ASSOCIATION OF IMMUNOLOGISTS, US, vol. 174, no. 6, 15 March 2005 (2005-03-15), pages 3187 - 3196, XP002417364, ISSN: 0022-1767
WEI LEONG CHEW ET AL: "A multifunctional AAV-CRISPR-Cas9 and its host response", HHS PUBLIC ACCESS AUTHOR MANUSCRIPT, vol. 13, no. 10, 5 September 2016 (2016-09-05), GB, pages 868 - 874, XP055339896, ISSN: 1548-7091, DOI: 10.1038/nmeth.3993
GRISWOLD KARL E ET AL: "Design and engineering of deimmunized biotherapeutics", CURRENT OPINION IN STRUCTURAL BIOLOGY, ELSEVIER LTD, GB, vol. 39, 17 June 2016 (2016-06-17), pages 79 - 88, XP029764641, ISSN: 0959-440X, DOI: 10.1016/J.SBI.2016.06.003
SOREK ET AL.: "CRISPR - a widespread system that provides acquired resistance against phages in bacteria and archaea", NAT. REV. MICROBIOL., vol. 6, no. 3, 2008, pages 181 - 6
BARRANGOU, R.; MARRAFFINI, L.A.: "CRISPR-Cas systems: prokaryotes upgrade to adaptive immunity", MOL. CELL, vol. 54, no. 2, 2014, pages 234 - 44
JINEK ET AL.: "A programmable dual-RNA-guided DNA endonuclease in adaptive bacterial immunity", SCIENCE, vol. 337, no. 6096, 2012, pages 816 - 21
CONG ET AL.: "Multiplex genome engineering using CRISPR/Cas systems", SCIENCE, vol. 339, no. 6121, 2013, pages 819 - 23
MALI ET AL.: "RNA-guided human genome engineering via Cas9", SCIENCE, vol. 339, no. 6121, pages 823 - 26
FU ET AL.: "Improving CRISPR-Cas nucleases using truncated guide RNAs", NATURE BIOTECHNOLOGY, vol. 32, no. 3, 2014, pages 279 - 84
SANDER; JOUNG: "CRISPR-Cas sytems for editing, regulating and targeting genomes", NATURE BIOTECHNOLOGY, vol. 32, no. 4, 2014, pages 347 - 55
SANDER, J.D.; JOUNG, J.K.: "CRISPR-Cas systems for editing, regulating and targeting genomes", NATURE BIOTECH., vol. 32, no. 4, 2014, pages 347 - 55
MAUS ET AL.: "Adoptive immunotherapy for cancer or viruses", ANNUAL REV. IMMUNOL., vol. 32, 2014, pages 189 - 225
COX ET AL.: "Therapeutic genome editing: prospects and challenges", NATURE MEDICINE, vol. 21, no. 2, 2015, pages 121 - 31
WANG ET AL., HUMAN GENE THERAPY, vol. 26, no. 7, 2015, pages 432 - 442
ZETSCHE ET AL.: "Cpfl is a Single RNA-Guided Endonuclease of a Class 2 CRISPR-Cas System", CELL, vol. 163, 2015, pages 759 - 771
KARLIN; ALTSCHUL, PROC. NATL. ACAD. SCI. USA, vol. 87, 1990, pages 2264 - 68
ALTSCHUL, PROC. NATL. ACAD. SCI. USA, vol. 90, 1993, pages 5873 - 77
ALTSCHUL ET AL., J. MOL. BIOL., vol. 215, 1990, pages 403 - 10
ALTSCHUL E, NUCLEIC ACIDS RES., vol. 25, no. 17, 1997, pages 3389 - 3402
MALI ET AL.: "Cas9 as a versatile tool for engineering biology", NATURE METHODS, vol. 10, 2013, pages 957 - 63
CHYLINSKI ET AL.: "Classification and evolution of type II CRISPR-Cas sytems", NUCLEIC ACIDS RESEARCH, vol. 42, no. 10, 2014, pages 6091 - 105
RAN ET AL.: "In vivo genome editing using Staphylococcus aureus Cas9", NATURE, vol. 520, no. 7546, 2015, pages 186 - 91
ESVELT ET AL.: "Orthogonal Cas9 proteins for RNA-guided gene regulation and editing", NATURE METHODS, vol. 10, no. 11, 2013, pages 1116 - 21
GRANTHAM ET AL., SCIENCE, vol. 185, 1974, pages 862 - 864
KARPLUS ET AL., NATURWISSENSCHAFTEN, vol. 72, 1985, pages 212 - 213
KOLASKAR ET AL., FEBS LETT., vol. 276, 1990, pages 172 - 174
PARKER ET AL., BIOCHEMISTRY, vol. 25, 1986, pages 5425 - 5432
PONNUSWAMY ET AL., BIOCHIM. BIOPHYS. ACTA, vol. 623, 1980, pages 301 - 326
ATCHLEY ET AL., PROC. NATL. ACAD. SCI. U.S.A., vol. 102, 2005, pages 6395 - 6400
LAUPEZE ET AL.: "Differential expression of major histocompatibility complex class la, lb, and II molecules on monocytes-derived dendritic and macrophagic cells", HUM. IMMUNOL., vol. 60, no. 7, 1999, pages 591 - 7
GANSBACHER; ZIER: "Regulation of HLA-DR, DP, and DQ expression in activated T cells", CELL IMMUNOL., vol. 117, no. L, 1988, pages 22 - 34
BERDOZ ET AL.: "Constitutive and induced expression of the individual HLA-DR beta and alpha chain loci in different cell types", J. IMMUNOL., vol. 139, no. 4, 1987, pages 1336 - 41
STUNZ ET AL.: "HLA-DRB1 and -DRB4 genes are differentially regulated at the transcriptional level", J. IMMUNOL., vol. 143, no. 9, 1989, pages 3081 - 6
VADER ET AL.: "The HLA-DQ2 gene dose effect in celiac disease is directly related to the magnitude and breadth of gluten-specific T cell responses", PNAS, vol. 100, no. 21, 2003, pages 12390 - 5
CASTELLI ET AL.: "HLA-DP4, the most frequent HLA II molecule, defines new supertype of peptide-binding specificity", J. IMMUNOL., vol. 169, no. 12, 2002, pages 6928 - 34
KIRSCHMANN ET AL.: "Naturally processed peptides from rheumatoid arthritis associated and non-associated HLA-DR alleles", J. IMMUNOL., vol. 155, no. 12, 1995, pages 5655 - 62
VERRECK ET AL.: "Natural peptides isolated from Gly86/Val86-containing variants of HLA-DR1, -DR11, -DR13, and -DR52", IMMUNOGENETICS, vol. 43, no. 6, 1996, pages 392 - 7
ROHN ET AL., NATURE IMMUNOL., vol. 5, 2004, pages 909 - 918
PENNA ET AL., J. IMMUNOL., vol. 7, no. 57, 2001, pages 1862 - 1866
KROPSHOFER; SPINDELDREHER: "Antigen Presenting Cells: From Mechanisms to Drug Development", 2005, WILEY-VCH, pages: 159 - 98
AGUSTIN-PAVON ET AL.: "Synthetic biology and therapeutic strategies for the degenerating brain", BIOESSAYS, vol. 36, no. 10, 2014, pages 979 - 990
NISHIMASU ET AL.: "Crystal structure of Cas9 in complex with guide RNA and target DNA", CELL, vol. 156, no. 5, 2014, pages 935 - 49
Attorney, Agent or Firm:
REDL LIFE SCIENCE PATENT ATTORNEYS (AT)
Download PDF:
Claims:
CLAIMS

What is claimed is:

1. A method for reducing the immunogenicity of a CRISPR-associated (Cas) protein, the method comprising introducing one or more amino acid substitutions into one or more residues corresponding to one or more major histocompatibility (MHC) Class I and/or Class II binding sites of the Cas protein to form a recombinant Cas protein.

2. The method of claim 1 , wherein the position of the one or more amino acid substitutions is selected through epitope mapping.

3. The method of claim 2, wherein the epitope mapping is in silico epitope mapping.

4. The method of claim 2, wherein the epitope mapping comprises incubating an antigen presenting cell (APC) in the presence of a Cas protein, and identifying one or more peptides derived from the Cas protein bound to a major histocompatibility (MHC) Class I and/or Class II protein of the APC.

5. The method of claim 4, further comprising isolating at least a portion of the MHC Class I and/or MHC Class II proteins from the APC.

6. The method of claim 5, wherein the MHC Class I and/or MHC Class II proteins are

isolated by immunoprecipitation.

7. The method of any one of claims 4-6, wherein at least one or more Cas peptides bound to the MHC Class I and/or MHC Class II proteins is identified by mass spectrometry.

8. The method of any one of claims 1-7, further comprising assaying the immunogenicity of the recombinant Cas protein by exposing the recombinant Cas protein to a population of T-cells and comparing the level of T-cell activation to the level of T-cell activation induced by a corresponding wild-type Cas protein.

9. The method of claim 8, wherein the T-cell activation is determined by measuring T-cell proliferation.

10. The method of claim 8, wherein the T-cell proliferation is measured by flow cytometry.

1 1. The method of any one of claims 8-10, wherein the T-cells are CD4+ T-cells.

12. The method of any one of claims 8-10, wherein the T-cells are CD8+ T-cells.

13. The method of any one of claims 1-12 wherein the MHC Class I and MHC Class II

proteins are HLA Class I and HLA Class II proteins, respectively.

14. The method of any one of claims 1-13, wherein the recombinant Cas protein is derived from a prokaryote.

15. The method of claim 14, wherein the prokaryote is a bacterium.

16. The method of claim 15, wherein the bacterium is from the genera Streptococcus ,

Staphylococcus, or Neisseria.

17. The method of claim 16, wherein the bacterium is Streptococcus pyogenes, Streptococcus thermophilus , Staphylococcus aureus, or Neisseria meningtidis .

18. The method of any one of claims 1-17, wherein the recombinant Cas protein is derived from Cas3, Cas9, or Cas 10.

19. The method of claim 18, wherein the recombinant Cas protein is derived from Cas9.

20. The method of claim 19, wherein the one or more residues corresponding to the one or more major histocompatibility (MHC) Class I and/or Class II binding sites is selected from the bolded residues designated in Figure 9.

21. The method of claim 19, wherein the one or more residues corresponding to the one or more major histocompatibility (MHC) Class I and/or Class II binding sites is selected from residues 16, 18, 31, 71, 81-82, 86, 97, 98, 105, 118, 126, 128, 132, 135, 136, 138- 139, 155, 158, 164, 168, 185, 188, 192, 196, 216, 244, 271 , 275, 278-279, 290, 300, 321 , 333, 337, 351 -352, 359, 362, 373, 375, 383, 395, 418, 419, 422-423, 430, 432-433, 462, 511-517, 553, 575, 578, 580, 583, 591, 594, 598, 626-627, 653-655, 694, 702, 704, 741, 746-748, 772, 775, 778, 780, 796, 799-800, 812, 814-816, 823, 950, 1001, 1008, 1013, 1015-1016, 1018, 1023, 1028-29, 1033-34, 1036, 1038-1039, 1043, 1045-1046, 1050- 1051, 1067, 1072-1074, 1080, 1086-1089, 1094, 1137-1144, 1146, 1 174, 1179, 1181, 1195-1196, 1198, 1200-1202, 1204, 1213-1215, 1217-1218, 1220, 1235, 1237, 1242, 1294, 1297-98, 1309, 1313, 1318, 1348, 1352, 1355-1356, 1362, 5, 7, 9-11 , 25-27, 31 , 33, 43, 47, 51 , 85, 90, 97-98, 1 18-119, 128, 136, 168-170, 181 , 184, 186, 188, 196, 21 1, 222, 229, 248, 252, 256, 264, 266-267, 278, 292, 300, 302, 305-306, 308, 312, 321 , 322, 324, 334, 335, 351, 352, 359, 362, 363, 372, 375, 377, 380, 383, 390-391, 399, 403, 405, 409, 410, 432, 450-451, 455-456, 462, 469, 471, 473, 476, 488, 491 -492, 529-530, 534, 539, 548, 553, 557-559, 561, 564, 578, 594-595, 598, 600-601, 606, 615, 631 -632, 636, 639, 643, 651 -653, 655-656, 661 , 666, 679-680, 683, 704, 720, 724, 727, 733, 735, 738, 741, 747-751, 753, 758-761, 763, 765-766, 788, 791 , 793, 795-796, 830-834, 836, 838, 841, 845-847, 867, 869-870, 872, 891 , 914, 916-917, 919, 922, 925, 927, 931, 934-935, 943, 953-954, 970, 972-973, 988, 1008-1009, 1013, 1015, 1019, 1021, 1029, 1036-1039, 1042, 1045-1046, 1060, 1092, 1096, 1 100, 1105, 1 110, 1131, 1141, 1 143, 1145-1146, 1149, 1157-1160, 1 163-1164, 1181-1182, 1187, 1 190, 1210, 1212, 1238, 1265, 1266, 1273, 1276, 1280, 1298, 1302, 1310, 1313, 1315, 1326-1327, 1335-1336, 1342, 1347- 1348, 1352, 1355, and/or 1360 of Streptococcus pyogenes Cas9, or corresponding residues of Streptococcus thermophilus Cas9, Staphylococcus aureus Cas9, or Neisseria meningitidis Cas9.

22. The method of claim 19, wherein the one or more residues corresponding to the one or more major histocompatibility (MHC) Class I and/or Class II binding sites is selected from residues 8, 50, 53-54, 76, 77, 79, 82, 89, 92-93, 135, 147, 169, 176-180, 183-184, 203, 208, 21 1, 240-241, 248, 251-252, 266, 268, 271-273, 278, 280, 305, 307-308, 347- 348, 350, 353, 356, 372, 375-377, 379, 381 , 412, 467, 595, 607, 628, 631, 643, 649, 676, 688-690, 702, 718, 724-726, 728, 735, 754-756, 764, 788, 792, 795-796, 799, 828, 835, 864-865, 871-873, 875-876, 932, 934-935, 990, 1024, 1030, 1055, 1069, 1071-1072, 1097, 5, 6, 8, 10, 12, 15, 19, 20, 23, 28, 33-35, 37-38, 40, 43-47, 50, 69-70, 75-76, 79, 82, 93, 118, 120-121, 135-139, 159, 162, 165,169, 183-184, 186, 189, 196, 207, 211, 234, 238, 241, 251 -252, 254, 256, 273, 298, 304-307, 309, 314, 347, 350, 356-357, 359, 362, 364, 367-368, 372, 375, 377-378, 381, 384, 386, 388, 396, 400, 401 , 427, 429, 431 -432, 455, 457, 467, 478, 506-51 1, 523, 528-529, 538-539, 545, 554-556, 567-568, 577, 588, 597, 600, 607, 631, 641, 643, 670, 673, 675-676, 678-700, 684-685, 689, 692, 701-704, 707, 715, 718, 722, 725-726, 728, 747, 753, 755, 761, 764, 769-770, 772, 778, 792, 795- 796, 798-799, 804, 810, 818, 822, 861, 865, 882, 884-885, 889-890, 895-896, 898-899, 919-920, 932, 935, 937-938, 942, 946, 965, 970-971, 999, 1004, 1019, 1021 , 1023-1024, 1031 , 1042-1043, 1066, 1069, 1096, 1099, and/or 1104 of Streptococcus thermophilus Cas9, or corresponding residues of Streptococcus pyogenes Cas9, Staphylococcus aureus Cas9, or Neisseria meningitidis Cas9.

23. The method of claim 19, wherein the one or more residues corresponding to the one or more major histocompatibility (MHC) Class I and/or Class II binding sites is selected from residues 18, 48, 51, 55, 70, 72, 86, 89, 176, 197, 211-212, 219-221, 223, 233, 235- 236, 239-241, 256, 277, 345, 347, 351, 358, 389, 407-408, 415, 420-421, 444, 468, 500, 519, 565, 568-569, 573, 604, 609, 625, 651 , 657, 664, 666, 669, 675, 683, 688, 698-703, 705, 707-708, 713, 744, 754, 760, 769-771, 773-774, 808-809, 837, 855, 857-858, 897, 904-905, 912, 914, 918, 931, 939, 942, 947, 962, 966-967, 971, 974, 996, 1001 , 1004, 5, 18, 21, 23, 26, 29, 34, 35, 51 , 66-67, 70-72, 74, 76, 1 10, 118, 124, 132, 140, 145, 165, 170-171, 176, 190, 193, 197-198, 201 -202, 204, 212, 216, 223, 229-230, 232, 234, 240, 244-245, 247, 249, 251, 262, 269, 277-278, 281, 283-284, 303, 305, 313, 329-330, 332, 335, 347-348, 351, 354-355, 358, 362, 364, 366, 368, 383, 386-387, 404, 407-408, 420, 423, 430, 444, 464, 470, 474-476, 478-480, 508-510, 519, 523-527, 529, 532, 535-536, 538, 541, 543, 555, 558, 565, 601, 615, 625, 627, 630, 632, 635, 637-638, 641 , 644, 646, 680, 687-690, 694, 713-715, 718, 720-721, 732, 745, 771, 809, 816, 827-828, 847, 857, 864-865, 868, 877, 897, 912, 914, 916-917, 947, 974, 982, 989, 992, 1001, 1004, 1015- 1016, 1019, 1030, 1032, 1038, 1041, 1046, and/or 1048 of Staphylococcus aureus Cas9, or corresponding residues of Streptococcus pyogenes Cas9, Streptococcus thermophilus Cas9, or Neisseria meningitidis Cas9.

24. The method of claim 19, wherein the one or more residues corresponding to the one or more major histocompatibility (MHC) Class I and/or Class II binding sites is selected from residues 24-26, 74-75, 77-78, 97, 106, 111 , 113, 1 16, 118, 127, 136-138, 144, 226, 231, 272, 279-280, 287-288, 297, 304-305, 310, 317, 331-332, 335, 337, 342, 417, 423, 425, 427-430, 435, 441, 467, 472, 487, 516, 547, 554, 590, 592, 595-596, 599-600, 623, 632, 635-637, 639, 643-644, 646, 671 -672, 675-676, 680, 686, 690, 692-694, 707, 710, 744, 746-748, 750, 752-753, 778-779, 806-807, 836, 844, 858-860, 864, 876, 878, 880- 881, 883, 885, 888-892, 894-895, 936, 938, 948, 950, 952-953, 960-961, 965, 967, 988, 991, 1006, 1009, 1019-1020, 1033, 1035, 1057-1058, 1066, 12, 17, 41 , 43-46, 49, 68, 95, 97, 120, 122, 125, 130, 158, 174, 185, 196, 199, 203, 205, 208, 212-213, 227, 235, 238- 239, 254-255, 257, 260, 262-263, 283, 287, 289, 304, 311 , 329-332, 337, 360, 365, 372, 374, 386, 388-391, 407-408, 410-412, 434, 439, 448, 458, 472, 478, 481 , 487-488, 491 - 495, 497, 51 1, 513-515, 519, 537, 541, 545, 553-554, 556, 558-559, 568, 573, 577, 596, 604, 611, 632, 635, 643-644, 651 , 662, 680-681 , 685, 694, 702, 705-707, 709, 712, 716, 723, 725, 729, 747, 766, 791, 797, 821 , 824, 828-830, 832, 838, 844, 852, 857, 859-862, 867, 875, 885, 891, 903, 907, 910, 919-920, 923-925, 928, 935, 941, 953, 959, 961-962, 964-965, 967, 973, 985, 988, 995, 999, 1001, 1010, 1016-1017, 1019-1020, 1028, 1030, 1058, 1001 , and/or 1063 ofNeisseria meningitidis Cas9, or corresponding residues of Streptococcus pyogenes Cas9, Streptococcus thermophilus Cas9, or Staphylococcus aureus Cas9.

25. The method of any one of claims 1-24, wherein the MHC Class I and/or Class II binding sites of the Cas protein comprises a peptide comprising 9 (9mer) or 10 (lOmer) amino acids.

26. The method of any one of claims 1-25, wherein the introduced amino acid substitution is a naturally occurring amino acid.

27. A recombinant CRISPR-associated (Cas) protein comprising one or more amino acid substitutions in one or more residues corresponding to one or more MHC Class I and/or MHC Class II binding sites in a wild-type Cas protein, wherein the recombinant Cas protein has reduced immunogenicity compared to a wild-type Cas protein.

28. The recombinant Cas protein of claim 27, wherein the MHC Class I and/or MHC Class II binding sites are HLA Class I and HLA Class II binding sites, respectively.

29. The recombinant Cas protein of claim 27 or 28, wherein the recombinant Cas protein is derived from a prokaryote.

30. The recombinant Cas protein of claim 29, wherein the prokaryote is a bacterium.

31. The recombinant Cas protein of claim 30, wherein the bacterium is from the genera

Streptococcus, Staphylococcus, or Neisseria.

32. The recombinant Cas protein of claim 31 , wherein the bacterium is Streptococcus

pyogenes, Streptococcus thermophilus , Staphylococcus aureus, or Neisseria meningtidis .

33. The recombinant Cas protein of claim 31 or 32, wherein the recombinant Cas protein is derived from Cas3, Cas9, or CaslO.

34. The recombinant Cas protein of claim 33, wherein the recombinant Cas protein is derived from Cas9.

35. The recombinant Cas protein of claim 34, wherein the one or more residues

corresponding to the one or more major histocompatibility (MHC) Class I and/or Class II binding sites is selected from the bolded residues designated in Figure 9.

36. The recombinant Cas protein of claim 34, wherein the one or more residues

corresponding to the one or more major histocompatibility (MHC) Class I and/or Class II binding sites is selected from residues 9-11 , 25-27, 81-82, 97-98, 1 18-119, 138-139, 168- 170, 266-267, 278-279, 305-306, 351-352, 390-391, 422-423, 432-433, 450-451, 455- 456, 491-492, 511-517, 529-530, 557-559, 594-595, 600-601, 626-627, 631 -632, 651- 653, 653-655, 655-656, 679-680, 746-748, 747-751, 758-761, 765-766, 795-796, 799- 800, 814-816, 830-834, 845-847, 869-870, 916-917, 934-935, 953-954, 972-973, 1008- 1009, 1015-1016, 1028-1029, 1033-1034, 1036-1039, 1038-1039, 1045-1046, 1050- 1051 , 1072-1074, 1086-1089, 1137-1144, 1 145-1146, 1157-1160, 1163-1164, 1181- 1182, 1195-1196, 1200-1202, 1213-1215, 1217-1218, 1297-1298, 1326-1327, 1335- 1336, 1347-1348, and/or 1355-1356 of Streptococcus pyogenes Cas9, or corresponding residues of Streptococcus thermophilus Cas9, Staphylococcus aureus Cas9, or Neisseria meningitidis Cas9.

37. The recombinant Cas protein of claim 34, wherein the one or more residues

corresponding to the one or more major histocompatibility (MHC) Class I and/or Class II binding sites is selected from residues 33-35, 37-38, 43-47, 53-54, 69-70, 75-76, 92-93, 120-121, 135-139, 176-180, 183-184, 240-241, 251-252, 271-273, 304-307, 307-308, 347-348, 356-357, 367-368, 375-377, 377-378, 431-432, 506-51 1, 528-529, 538-539, 554-556, 567-568, 675-676, 678-700, 684-685, 688-690, 701-704, 724-726, 725-726, 754-756, 769-770, 795-796, 798-799, 864-865, 871-873, 875-876, 884-885, 889-890, 895-896, 898-899, 919-920, 934-935, 937-938, 970-971, 1023-1024, 1042-1043, and/or 1071-1072 of Streptococcus thermophilus Cas9, or corresponding residues of

Streptococcus pyogenes Cas9, Staphylococcus aureus Cas9, or Neisseria meningitidis Cas9.

38. The recombinant Cas protein of claim 34, wherein the one or more residues

corresponding to the one or more major histocompatibility (MHC) Class I and/or Class II binding sites is selected from residues 66-67, 70-72, 170-171, 197-198, 201 -202, 211 - 212, 219-221, 229-230, 235-236, 239-241, 244-245, 277-278, 283-284, 329-330, 347- 348, 354-355, 386-387, 407-408, 420-421, 474-476, 478-480, 508-510, 523-527, 535- 536, 568-569, 637-638, 687-690, 698-703, 707-708, 713-715, 720-721, 769-771, 773- 774, 808-809, 827-828, 857-858, 864-865, 904-905, 916-917, 966-967, and/or 1015-1016 of Staphylococcus aureus Cas9, or corresponding residues of Streptococcus pyogenes Cas9, Streptococcus thermophilus Cas9, or Neisseria meningitidis Cas9.

39. The recombinant Cas protein of claim 34, wherein the one or more residues

corresponding to the one or more major histocompatibility (MHC) Class I and/or Class II binding sites is selected from residues 24-26, 43-46, 74-75, 77-78, 136-138, 212-213, 238-239, 254-255, 262-263, 279-280, 287-288, 304-305, 329-332, 331-332, 388-391, 407-408, 410-412, 427-430, 487-488, 491 -495, 513-515, 553-554, 558-559, 595-596, 599-600, 635-637, 643-644, 671 -672, 675-676, 680-681, 692-694, 705-707, 746-748, 752-753, 778-779, 806-807, 858-860, 859-862, 880-881, 888-892, 894-895, 919-920, 923-925, 952-953, 960-961, 961 -962, 964-965, 1016-1017, 1019-1020, and/or 1057-1058 of Neisseria meningitidis Cas9, or corresponding residues of Streptococcus pyogenes Cas9, Streptococcus thermophilus Cas9, or Staphylococcus aureus Cas9.

40. The recombinant Cas protein of any one of claims 27 to 39, wherein the MHC Class I and/or Class II binding sites of the Cas protein comprises a peptide comprising 9 (9mer) or 10 (lOmer) amino acids.

41. The recombinant Cas protein of any one of claims 27 to 40, wherein the introduced

amino acid substitution is a naturally occurring amino acid.

42. The recombinant Cas protein of claim 27, wherein the wild-type Cas protein comprises an amino sequence having at least 95% identity to the amino acid sequence of any one of SEQ ID Os: 1 -4.

43. A recombinant CRISPR-associated (Cas) protein made by a process comprising

introducing one or more amino acid substitutions into one or more residues

corresponding to one or more MHC Class I and/or MHC Class II binding sites of a wild- type Cas protein to form the recombinant Cas protein, wherein the recombinant Cas protein has reduced immunogenicity compared to the wild-type Cas protein.

44. The recombinant Cas protein of claim 43, wherein the position of the one or more amino acid substitutions is determined by epitope mapping.

45. The recombinant Cas protein of claim 44, wherein the epitope mapping is performed using in silico methods.

46. The recombinant Cas protein of claim 44, wherein the epitope mapping is performed by a method comprising incubating an antigen presenting cell (APC) in the presence of a Cas protein and identifying peptides derived from the Cas protein bound to major

histocompatibility (MHC) Class I and/or Class II proteins.

47. The recombinant Cas protein of claim 46, wherein at least a portion of the MHC Class I and/or MHC Class II proteins bound to the Cas peptides is isolated.

48. The recombinant Cas protein of claim 47, wherein the MHC Class I and/or MHC Class II proteins bound to the Cas peptides are isolated by immunoprecipitation.

49. The recombinant Cas protein of any one of claims 44-48, wherein the recombinant Cas protein is derived from a prokaryote.

50. The recombinant Cas protein of claim 49, wherein the prokaryote is a bacterium.

51. The recombinant Cas protein of claim 50, wherein the bacterium is from the genera

Streptococcus, Staphylococcus, or Neisseria.

52. The recombinant Cas protein of claim 51 , wherein the bacterium is Streptococcus

pyogenes, Streptococcus thermophilus , Staphylococcus aureus, or Neisseria meningtidis .

53. The recombinant Cas protein of any one of claims 49-52, wherein the recombinant Cas protein is derived from Cas3, Cas9, or Cas 10.

54. The recombinant Cas protein of claim 53, wherein the recombinant Cas protein is derived from Cas9.

55. The recombinant Cas protein of claim 54, wherein the wild-type Cas protein comprises an amino sequence having at least 95% identity to the amino acid sequence of any one of SEQ ID Os: 1 -4.

56. A recombinant CRISPR-associated (Cas) protein comprising one or more substitutions in one or more amino acid residues of a wild-type Cas protein as designated in Figure 9, wherein the recombinant Cas protein has reduced immunogenicity compared to the wild- type Cas protein.

57. An isolated nucleic acid molecule comprising a nucleotide sequence that encodes a

recombinant CRISPR-associated (Cas) protein, wherein the recombinant Cas protein has one or more amino acid substitutions in one or more residues corresponding to one or more MHC Class I and/or MHC Class II binding sites of a wild-type Cas protein, wherein the recombinant Cas protein has reduced immunogenicity compared to a wild-type Cas protein.

58. The isolated nucleic acid molecule of claim 57, wherein the recombinant Cas protein is derived from a prokaryote.

59. The isolated nucleic acid molecule of claim 58, wherein the prokaryote is a bacterium.

60. The isolated nucleic acid molecule of claim 59, wherein the bacterium is from the genera Streptococcus, Staphylococcus, or Neisseria.

61. The isolated nucleic acid molecule of claim 60, wherein the bacterium is Streptococcus pyogenes, Streptococcus thermophilus , Staphylococcus aureus, or Neisseria meningtidis .

62. The isolated nucleic acid molecule of claim 61 , wherein the recombinant Cas protein is derived from Cas3, Cas9, or CaslO.

63. The isolated nucleic acid molecule of claim 62, wherein the recombinant Cas protein is derived from Cas9.

64. A vector comprising the nucleic acid of any one of claims 57-63.

65. The vector of claim 64, wherein the vector is a viral vector.

66. The vector of claim 65, wherein the viral vector is an adeno-associated viral vector or a lentiviral vector.

67. A cell comprising the recombinant Cas protein of any one of claims 27-56.

68. A cell comprising the nucleic acid of any one of claims 57-63.

69. A cell comprising the vector of any one of claims 64-66.

70. The cell of any one of claims 67-69, wherein the cell is a prokaryotic cell.

71. The cell of claim 70, wherein the cell is an E. coli cell.

72. The cell of any one of claims 67-69, wherein the cell is a eukaryotic cell.

73. The cell of claim 72, wherein the cell is selected from the group consisting of a

lymphocytic cell, a myeloid cell, and an induced pluripotent stem cell (iPSC), or a T-cell.

74. A library for identifying a CRISPR-associated (Cas) protein with reduced

immunogenicity, the library comprising at least one recombinant Cas protein comprising one or more amino acid substitutions in one or more residues corresponding to one or more MHC Class I and/or MHC Class II binding sites of a wild-type Cas protein.

75. A method for altering the gene expression and/or DNA sequence at a genomic location containing a target sequence, the method comprising introducing into a cell containing the genetic element a guide R A that hybridizes to the target sequence and a

recombinant CRISPR-associated (Cas) protein comprising one or more amino acid substitutions in one or more residues corresponding to one or more MHC Class I and/or MHC Class II binding sites in a wild-type Cas protein, wherein expression of the one or more genetic elements is altered.

76. The method of claim 75, wherein the MHC Class I and/or MHC Class II binding sites are HLA Class I and HLA Class II binding sites, respectively.

77. The method of claim 75 or 76, wherein the recombinant Cas protein is derived from a prokaryote.

78. The method of claim 77, wherein the prokaryote is a bacterium.

79. The method of claim 78, wherein the bacterium is from the genera Streptococcus ,

Staphylococcus, or Neisseria.

80. The method of claim 79, wherein the bacterium is Streptococcus pyogenes, Streptococcus thermophilus , Staphylococcus aureus, or Neisseria meningtidis .

81. The method of claim 80, wherein the recombinant Cas protein is derived from Cas3, Cas9, or Cas 10.

82. The method of claim 81 , wherein the recombinant Cas protein is derived from Cas9.

83. The method of claim 75 or 76, wherein the cell is a eukaryotic cell.

84. The method of any one of claims 75-83, wherein the guide RNA and/or the recombinant Cas protein are transfected into the cell.

85. The method of any one of claims 75-83, wherein the guide RNA and/or the recombinant Cas protein are located on a vector.

86. The method of claim 85, wherein the guide RNA and the recombinant Cas protein are located on the same vector.

87. The method of claim 85 or 86, wherein the vector is a viral vector.

88. The method of claim 87, wherein the viral vector is an adeno-associated viral vector or a lentiviral vector. A recombinant CRISPR-associated (Cas) protein, wherein said protein comprises one more amino acid variation compared to a while-type Cas protein, wherein the one or more amino acid variation is at one or more positions corresponding to amino acid residues in the polypeptide of SEQ ID NO: 5, wherein the variation is at a position selected from the group consisting of:

i. residues 100 to 120,

ii. residues 250 to 280,

iii. residues 690 to 710,

iv. residues 840 to 860, and

v. residues 1270 to 1295.

The Cas protein of claim 89, wherein the variation is at a position selected from the group consisting of:

i. residues 102 to 120,

ii. residues 253 to 277,

iii. residues 692 to 709,

iv. residues 692 to 709, and

v. residues 1276 to 1292.

91. The Cas protein of claim 89, wherein the variation is at a position selected from residues 102 to 120.

92. The Cas protein of claim 89, wherein the variation is at a position selected from residues 253 to 277.

93. The Cas protein of claim 89, wherein the variation is at a position selected from residues 692 to 709.

94. The Cas protein of claim 89, wherein the variation is at a position selected from residues 692 to 709.

95. The Cas protein of claim 89, wherein the variation is at a position selected from residues 1276 to 1292.

96. The Cas protein of claim 89, wherein said recombinant Cas protein comprises two or more amino acid variations.

97. The Cas protein of claim 89, wherein said recombinant Cas protein comprises three or more amino acid variations.

98. The Cas protein of claim 89, wherein said recombinant Cas protein comprises five or more amino acid variations.

99. The Cas protein of claim 89, wherein said amino acid variation is a substitution selected from the group consisting of one or more of the following:

Position Original Possible substitutions

residue

105 F D,E

106 L D,E,G,K,P,Q,R

107 V D,E,G

258 L D, E, G, K, P

263 K A,D,E,G,N,P,S,T

264 L A,D,E,G,H,K,N,P,Q,Pv,S,T,V

265 Q D,E,G,N,P,T

266 L A,D,E,G,N,P,Q,S,T, V

267 S A,D,E,G,H,P,T

696 L E,G,P

846 F E,W

847 L D,E,F,G,H,K,N,P,Q,S,T,W

852 I D,E,F,G,Y

855 K D,E,G,P,S

1278 K A,D,E,F,G,N,P,Q,S,T,V,W

1279 R D,E,H,K,Q

1280 V A,D,E,G,K,N,P,Q,S,T

1281 I A,D,E,F,G,H,K,N,P,Q,Pv,S,T,W 1282 A,D,E,G,H,N,P,S,T

100. The Cas protein of claim 89, wherein said amino acid variations include the following substitutions: L106D, K263D, L696G, L847D, and 1128 ID.

101. The Cas protein of claim 89, wherein the amino acid variation is determined using in silico epitope mapping.

102. The Cas protein of claim 89, wherein the amino acid variation is a substitution of one or more amino acids with one or more different amino acids.

103. The Cas protein of claim 102, wherein the amino acid variation comprises replacing one or more of aspargine, glutamine, leucine, lysine, methionine, serine, threonine, and valine residues with a different amino acid.

104. The Cas protein of claim 102, wherein the amino acid variation comprises replacing a polar amino acid with a nonpolar amino acid.

105. The Cas protein of claim 102, wherein the amino acid variation comprises replacing a hydrophilic amino acid with a hydrophobic amino acid.

106. The Cas protein of claim 89, wherein the amino acid variation is an insertion of one or more amino acids residues.

107. The Cas protein of claim 89, wherein the amino acid variation is a deletion of one or more amino acid residues.

108. The Cas protein of claim 89, wherein the wild-type Cas protein has a sequence 90%

identical to SEQ ID NO: 5.

109. The Cas protein of claim 89, wherein the wild-type Cas protein is derived from a

prokaryote.

1 10. The Cas protein of claim 109, wherein the prokaryote is a bacterium.

11. The Cas protein of claim 1 10, wherein the bacterium is from the genera Streptococcus, Staphylococcus, or Neisseria. 12. The Cas protein of claim 1 11 , wherein the bacterium is Streptococcus pyogenes,

Streptococcus thermophilus, Staphylococcus aureus, or Neisseria meningtidis . 13. The Cas protein of any one of claims 89 to 112, wherein the recombinant Cas protein is derived from Cas3, Cas9, or Cas 10. 14. The Cas protein of claim 1 13, wherein the recombinant Cas protein is derived from Cas9. 15. A polynucleotide encoding the Cas protein of any one of claims 89 to 114. 16. An expression vector comprising the polynucleotide of claim 115. 17. A host cell comprising the expression vector of claim 1 16.

Description:
CRISPR-ASSOCIATED (Cas) PROTEINS WITH REDUCED IMMUNOGENICITY FIELD OF THE INVENTION

[0001] The field of the invention is cellular and molecular biology and genetic engineering. Specifically, the invention relates to methods of reducing the immunogenicity of CRISPR- associated (Cas) proteins and the modified Cas proteins produced therefrom. In addition, the invention relates to methods for cell and gene therapy, including any and all genetic modifications and alterations of gene expression (and/or genetic elements) made in-vivo or ex- vivo, using Cas proteins with reduced immunogenicity.

BACKGROUND OF THE INVENTION

[0002] The Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR) and associated proteins (Cas proteins), which comprise the CRISPR-Cas system, were first identified in selected bacterial species and form part of a prokaryotic adaptive immune system. See Sorek, et al, "CRISPR - a widespread system that provides acquired resistance against phages in bacteria and archaea," Nat. Rev. Microbiol. 6(3)181-6 (2008), which is incorporated by reference herein in its entirety. CRISPR-Cas systems have been classified into three main types: Type I, Type II, and Type III. The main defining features of the separate Types are the various cas genes, and the respective proteins they encode, that are employed. The casl and cas2 genes appear to be universal across the three main Types, whereas cas3, cas9, and caslO are thought to be specific to the Type I, Type II, and Type III systems, respectively. See, e.g., Barrangou, R. and Marraffini, L.A., "CRISPR-Cas systems: prokaryotes upgrade to adaptive immunity," Mol. Cell. 54(2):234-44 (2014), which is incorporated by reference herein in its entirety.

[0003] In general, the CRISPR-Cas system functions by capturing short regions of invading viral or plasmid DNA and integrating the captured DNA into the host genome to form so-called CRISPR arrays that are interspaced by repeated sequences within the CRISPR locus. This acquisition of DNA into CRISPR arrays is followed by transcription and RNA processing.

[0004] Depending on the bacterial species, CRISPR RNA processing proceeds differently. For example, in the Type II system, originally described in the bacterium Streptococcus pyogenes, the transcribed RNA is paired with a transactivating RNA (tracrRNA) before being cleaved by RNase III to form an individual CRISPR-RNA (crR A). The crR A is further processed after binding by the Cas9 nuclease to produce the mature crRNA. The crRNA Cas9 complex subsequently binds to DNA containing sequences complimentary to the captured regions (termed proto spacers). The Cas9 protein then cleaves both strands of DNA in a site- specific manner, forming a double-strand break (DSB). This provides a DNA-based memory, resulting in rapid degradation of viral or plasmid DNA upon repeat exposure and/or infection. The native CRISPR system has been comprehensively reviewed (see, e.g., Barrangou, R. and Marraffini, L.A., 2014).

[0005] Since its original discovery, multiple groups have done extensive research around potential applications of the CRISPR system in genetic engineering, including gene editing (Jinek et al., "A programmable dual-RNA-guided DNA endonuclease in adaptive bacterial immunity," Science 337(6096):816-21 (2012); Cong et al., "Multiplex genome engineering using CRISPR Cas systems," Science 339(6121):819-23 (2013); and Mali et /.,"RNA-guided human genome engineering via Cas9," Science 339(6121):823-26; each of which is incorporated by reference herein in its entirety). One major development was utilization of a chimeric RNA to target the Cas9 protein, designed around individual units from the CRISPR array fused to the tracrRNA. This creates a single RNA species, called a small guide RNA (gRNA) where modification of the sequence in the protospacer region can target the Cas9 protein site- specifically. Considerable work has been done to understand the nature of the base-pairing interaction between the chimeric RNA and the target site, and its tolerance to mismatches, which is highly relevant in order to predict and assess off-target effects (see, e.g., Fu et al., "Improving CRISPR-Cas nucleases using truncated guide RNAs," Nature Biotechnology 32(3):279-84 (2014), and supporting material, which is incorporated by reference herein in its entirety).

[0006] The CRISPR-Cas9 gene editing system has been used successfully in a wide range of organisms and cell lines, both in order to induce DSB formation using the wild type Cas9 protein or to nick a single DNA strand using a mutant protein termed Cas9n/Cas9 D10A (see, e.g., Mali et al., (2013) and Sander and Joung, "CRISPR-Cas sytems for editing, regulating and targeting genomes," Nature Biotechnology 32(4):347-55 (2014), each of which is incorporated by reference herein in its entirety). While double-strand break (DSB) formation results in creation of small insertions and deletions (indels) that can disrupt gene function, the Cas9n/Cas9 D10A nickase avoids indel creation (the result of repair through non-homologous end-joining) while stimulating the endogenous homologous recombination machinery. Thus, the Cas9n/Cas9 D10A nickase can be used to insert regions of D A into the genome with high-fidelity.

[0007] In addition to genome editing, the CRISPR system has a multitude of other applications, including regulating gene expression, genetic circuit construction, and functional genomics, amongst others. Reviewed in Sander, J.D. and Joung, J.K. "CRISPR-Cas systems for editing, regulating and targeting genomes," Nature Biotech. 32(4):347-55 (2014), which is incorporated by reference herein in its entirety.

[0008] There is wide spread interest in using CRISPR both in vivo and ex vivo for gene and cell therapy applications. Such applications are particularly relevant where single (monogenic) genetic modifications have been associated with specific disease conditions. Furthermore, CRISPR can be used to aid insertion of heterologous DNA sequences to support cell engineering. A good example of this is in the generation of immunotherapies involving genetically engineered T cells, or CAR-T (chimeric antigen receptors) T-cell mediated therapies. See, e.g., Maus et al., "Adoptive immunotherapy for cancer or viruses," Annual Rev. Immunol. 32:189-225 (2014), which is incorporated by reference herein in its entirety. However, the broad applicability of CRISPR, particularly in the generation of new therapeutic treatments, comes with a number of challenges, many of which are described in Cox et al., "Therapeutic genome editing: prospects and challenges," Nature Medicine 21(2):121-31 (2015) ("Cox 2015"), which is incorporated by reference herein in its entirety.

[0009] One key challenge identified by Cox et al., "Therapeutic genome editing: prospects and challenges," Nature Medicine 21(2):121-31 (2015), is the immunogenicity of the Cas9 protein. The use of polypeptides as therapeutics has the associated risk of generating undesirable immune responses in patients, typically defined by the generation of anti-drug antibody (ADA) responses. Such responses can be motivated by the presence of "foreign" epitopes in the molecule and can be exacerbated by extrinsic factors, such as the genomic and disease background of the patient, the dosing and administration regime utilized, the formulation, and the route of administration and impurities, amongst others. These immune responses can have a variety of consequences, from altered pharmacology, to increased drug clearance or neutralization and loss of therapeutic efficacy. In extreme cases, protein therapeutics can cause the development of severe allergic and anaphylactic reactions, with considerable risk to the patient.

[0010] Another well-characterized immune reaction to "foreign" agents is the so-called graft or transplant rejection (also termed host-versus-graft reaction), in which the endogenous immune system reacts against, causing the destruction of, foreign tissue. Tissue rejection can be mediated by humoral and cellular immune responses. In the case of genetically modified cells generated for the purpose of incorporating a missing copy of a gene (gene therapy) or to help the patient eliminating cancerous cells (e.g., CAR-T therapies), there is a risk that some of the "machinery" utilized for the genetic modification of the cells could be "presented" by the modified cells and be recognized by the host as a "foreign" agent. Such recognition would trigger a rejection reaction, which could potentially render ineffective such treatments or, in severe cases, potentially cause auto-immune reactions. For example, depending on the method of delivery for the Cas protein, there are different associated risks in relation to unwanted immune responses. Where the Cas protein is encoded by the cell (e.g. following viral delivery), there is a potential risk that cells engineered to express Cas proteins could present Cas peptides on their HLA (human leukocyte antigen) Class I proteins and trigger an immune reaction. Where Cas9 protein is "transfected" (ex vivo) there is additional potential risk of HLA Class II- mediated humoral or anaphylactic responses.

[0011] Various publications are cited herein, the disclosures of which are incorporated by reference herein in their entireties.

SUMMARY OF THE INVENTION

[0012] The present invention reduces the immunogenicity of Cas proteins, e.g., Cas9 proteins. As Cas9 is a bacterial protein, it is highly likely that it will contain immunogenic epitopes, as has been confirmed by a recent study (Wang et al., Human Gene Therapy 26(7):432- 442 (2015), incorporated herein by reference in its entirety). The role of Cas protein immunogenicity in the use of CRISPR for therapeutic applications will depend on factors such as the method utilized to engineer the therapeutic cells (including the Cas9 delivery method) and the duration for which the Cas9 protein is required to be present in the recipient cell in order for the engineering to be efficacious. Thus, there is a need for methods for producing Cas proteins with reduced immunogenicity and the availability of such Cas proteins for use in genetic engineering, including gene and cell therapies.

[0013] In some embodiments, the invention is directed to a method for reducing the immunogenicity of a CRISPR-associated (Cas) protein, the method comprising introducing one or more amino acid substitutions into one or more residues corresponding to one or more major histocompatibility (MHC) Class I and/or Class II binding sites of the Cas protein to form a recombinant Cas protein.

[0014] In some embodiments, the position of the one or more amino acid substitutions is selected through epitope mapping. In some embodiments, the epitope mapping is in silico epitope mapping. In some embodiments, the epitope mapping comprises incubating an antigen presenting cell (APC) in the presence of a Cas protein, and identifying one or more peptides derived from the Cas protein bound to a major histocompatibility (MHC) Class I and/or Class II protein of the APC.

[0015] In some embodiments, the method of the invention further comprises isolating at least a portion of the MHC Class I and/or MHC Class II proteins from the APC. In some embodiments, the MHC Class I and/or MHC Class II proteins are isolated by immunoprecipitation. In some embodiments, at least one or more Cas peptides bound to the MHC Class I and/or MHC Class II proteins is identified by mass spectrometry.

[0016] In some embodiments, the method further comprises assaying the immunogenicity of the recombinant Cas protein by exposing the recombinant Cas protein to a population of T-cells and comparing the level of T-cell activation to the level of T-cell activation induced by a corresponding wild-type Cas protein. In some embodiments, the T-cell activation is determined by measuring T-cell proliferation. In some embodiments, the T-cell proliferation is measured by flow cytometry or ELISpot. In some embodiments, the T-cells are CD4+ T-cells. In some embodiments, the T-cells are CD8+ T-cells.

[0017] In some embodiments, the method further comprises assaying the immunogenicity of the recombinant Cas protein by exposing the recombinant Cas protein to a population of B-cells and comparing the level of B-cell activation to the level of B-cell activation induced by a corresponding wild-type Cas protein.

[0018] In some embodiments, the MHC Class I and MHC Class II proteins are HLA Class I and HLA Class II proteins, respectively.

[0019] In some embodiments, the recombinant Cas protein is derived from a prokaryote. In some embodiments, the prokaryote is a bacterium. In some embodiments, the bacterium is from the genera Streptococcus, Staphylococcus, or Neisseria. In some embodiments, the bacterium is Streptococcus pyogenes, Streptococcus thermophilus, Staphylococcus aureus, or Neisseria meningtidis . In some embodiments, the recombinant Cas protein is derived from Cas3, Cas9, or CaslO. In some embodiments, the recombinant Cas protein is derived from a Cas9-like protein, such as Cpfl , disclosed in Zetsche et al., "Cpfl is a Single RNA-Guided Endonuclease of a Class 2 CRISPR-Cas System," Cell 163:759-771 (2015), incorporated herein by reference in its entirety. In some embodiments, the recombinant Cas protein is derived from Cas9.

[0020] In some embodiments, residues selected for substitution, which are contained within the one or more major histocompatibility (MHC) Class I and/or Class II bound epitopes are selected from the residues designated in Figure 9. In some embodiments, the residues for substitution are selected from residues 16, 18, 31 , 71 , 81 -82, 86, 97, 98, 105, 1 18, 126, 128, 132, 135, 136, 138-139, 155, 158, 164, 168, 185, 188, 192, 196, 216, 244, 271 , 275, 278-279, 290, 300, 321 , 333, 337, 351 -352, 359, 362, 373, 375, 383, 395, 418, 419, 422-423, 430, 432-433, 462, 511-517, 553, 575, 578, 580, 583, 591 , 594, 598, 626-627, 653-655, 694, 702, 704, 741 , 746-748, 772, 775, 778, 780, 796, 799-800, 812, 814-816, 823, 950, 1001 , 1008, 1013, 1015- 1016, 1018, 1023, 1028-29, 1033-34, 1036, 1038-1039, 1043, 1045-1046, 1050-1051 , 1067, 1072-1074, 1080, 1086-1089, 1094, 1137-1144, 1146, 1174, 1179, 1181 , 1195-1196, 1198, 1200-1202, 1204, 1213-1215, 1217-1218, 1220, 1235, 1237, 1242, 1294, 1297-98, 1309, 1313, 1318, 1348, 1352, 1355-1356, and/or 1362 of Streptococcus pyogenes Cas9, or corresponding residues of Streptococcus thermophilus Cas9, Staphylococcus aureus Cas9, or Neisseria meningitidis Cas9.

[0021] In some embodiments, the residues for substitution are selected from residues 5, 7, 9- 11 , 25-27, 31 , 33, 43, 47, 51, 85, 90, 97-98, 1 18-1 19, 128, 136, 168-170, 181, 184, 186, 188, 196, 211 , 222, 229, 248, 252, 256, 264, 266-267, 278, 292, 300, 302, 305-306, 308, 312, 321, 322, 324, 334, 335, 351 , 352, 359, 362, 363, 372, 375, 377, 380, 383, 390-391, 399, 403, 405, 409, 410, 432, 450-451 , 455-456, 462, 469, 471 , 473, 476, 488, 491 -492, 529-530, 534, 539, 548, 553, 557-559, 561 , 564, 578, 594-595, 598, 600-601 , 606, 615, 631-632, 636, 639, 643, 651-653, 655-656, 661 , 666, 679-680, 683, 704, 720, 724, 727, 733, 735, 738, 741, 747-751 , 753, 758-761, 763, 765-766, 788, 791 , 793, 795-796, 830-834, 836, 838, 841, 845-847, 867, 869- 870, 872, 891 , 914, 916-917, 919, 922, 925, 927, 931, 934-935, 943, 953-954, 970, 972-973, 988, 1008-1009, 1013, 1015, 1019, 1021, 1029, 1036-1039, 1042, 1045-1046, 1060, 1092, 1096, 1100, 1105, 11 10, 1131 , 1141 , 1143, 1145-1146, 1149, 1157-1160, 1163-1164, 1181-1182, 1187, 1190, 1210, 1212, 1238, 1265, 1266, 1273, 1276, 1280, 1298, 1302, 1310, 1313, 1315, 1326-1327, 1335-1336, 1342, 1347-1348, 1352, 1355, and/or 1360 of Streptococcus pyogenes Cas9, or corresponding residues of Streptococcus thermophilus Cas9, Staphylococcus aureus Cas9, or Neisseria meningitidis Cas9.

[0022] In some embodiments, the residues for substitution are selected from residues 8, 50, 53-54, 76, 77, 79, 82, 89, 92-93, 135, 147, 169, 176-180, 183-184, 203, 208, 211 , 240-241 , 248, 251-252, 266, 268, 271 -273, 278, 280, 305, 307-308, 347-348, 350, 353, 356, 372, 375-377, 379, 381 , 412, 467, 595, 607, 628, 631 , 643, 649, 676, 688-690, 702, 718, 724-726, 728, 735, 754- 756, 764, 788, 792, 795-796, 799, 828, 835, 864-865, 871-873, 875-876, 932, 934-935, 990, 1024, 1030, 1055, 1069, 1071-1072, and/or 1097 of Streptococcus thermophilus Cas9, or corresponding residues of Streptococcus pyogenes Cas9, Staphylococcus aureus Cas9, or Neisseria meningitidis Cas9.

[0023] In some embodiments, the residues for substitution are selected from residues 5, 6, 8, 10, 12, 15, 19, 20, 23, 28, 33-35, 37-38, 40, 43-47, 50, 69-70, 75-76, 79, 82, 93, 118, 120-121, 135-139, 159, 162, 165,169, 183-184, 186, 189, 196, 207, 21 1, 234, 238, 241, 251-252, 254, 256, 273, 298, 304-307, 309, 314, 347, 350, 356-357, 359, 362, 364, 367-368, 372, 375, 377-378, 381 , 384, 386, 388, 396, 400, 401 , 427, 429, 431-432, 455, 457, 467, 478, 506-511 , 523, 528- 529, 538-539, 545, 554-556, 567-568, 577, 588, 597, 600, 607, 631, 641, 643, 670, 673, 675- 676, 678-700, 684-685, 689, 692, 701-704, 707, 715, 718, 722, 725-726, 728, 747, 753, 755, 761 , 764, 769-770, 772, 778, 792, 795-796, 798-799, 804, 810, 818, 822, 861 , 865, 882, 884- 885, 889-890, 895-896, 898-899, 919-920, 932, 935, 937-938, 942, 946, 965, 970-971 , 999, 1004, 1019, 1021 , 1023-1024, 1031 , 1042-1043, 1066, 1069, 1096, 1099, and/or 1104 of Streptococcus thermophilus Cas9, or corresponding residues of Streptococcus pyogenes Cas9, Staphylococcus aureus Cas9, or Neisseria meningitidis Cas9.

[0024] In some embodiments, the residues for substitution are selected from residues 18, 48, 51 , 55, 70, 72, 86, 89, 176, 197, 211-212, 219-221 , 223, 233, 235-236, 239-241, 256, 277, 345, 347, 351 , 358, 389, 407-408, 415, 420-421 , 444, 468, 500, 519, 565, 568-569, 573, 604, 609, 625, 651 , 657, 664, 666, 669, 675, 683, 688, 698-703, 705, 707-708, 713, 744, 754, 760, 769- 771 , 773-774, 808-809, 837, 855, 857-858, 897, 904-905, 912, 914, 918, 931 , 939, 942, 947, 962, 966-967, 971, 974, 996, 1001 and/or 1004 of Staphylococcus aureus Cas9, or corresponding residues of Streptococcus pyogenes Cas9, Streptococcus thermophilus Cas9, or Neisseria meningitidis Cas9.

[0025] In some embodiments, the residues for substitution are selected from residues 5, 18, 21, 23, 26, 29, 34, 35, 51 , 66-67, 70-72, 74, 76, 110, 118, 124, 132, 140, 145, 165, 170-171 , 176, 190, 193, 197-198, 201-202, 204, 212, 216, 223, 229-230, 232, 234, 240, 244-245, 247, 249, 251, 262, 269, 277-278, 281 , 283-284, 303, 305, 313, 329-330, 332, 335, 347-348, 351 , 354-355, 358, 362, 364, 366, 368, 383, 386-387, 404, 407-408, 420, 423, 430, 444, 464, 470, 474-476, 478-480, 508-510, 519, 523-527, 529, 532, 535-536, 538, 541 , 543, 555, 558, 565, 601 , 615, 625, 627, 630, 632, 635, 637-638, 641 , 644, 646, 680, 687-690, 694, 713-715, 718, 720-721 , 732, 745, 771 , 809, 816, 827-828, 847, 857, 864-865, 868, 877, 897, 912, 914, 916-917, 947, 974, 982, 989, 992, 1001 , 1004, 1015-1016, 1019, 1030, 1032, 1038, 1041, 1046, and/or 1048 of Staphylococcus aureus Cas9, or corresponding residues of Streptococcus pyogenes Cas9, Streptococcus thermophilus Cas9, or Neisseria meningitidis Cas9.

[0026] In some embodiments, the one or more residues corresponding to the one or more major histocompatibility (MHC) Class I and/or Class II binding sites is selected from residues 24-26, 74-75, 77-78, 97, 106, 1 11 , 1 13, 116, 118, 127, 136-138, 144, 226, 231 , 272, 279-280, 287-288, 297, 304-305, 310, 317, 331-332, 335, 337, 342, 417, 423, 425, 427-430, 435, 441 , 467, 472, 487, 516, 547, 554, 590, 592, 595-596, 599-600, 623, 632, 635-637, 639, 643-644, 646, 671-672, 675-676, 680, 686, 690, 692-694, 707, 710, 744, 746-748, 750, 752-753, 778-779, 806-807, 836, 844, 858-860, 864, 876, 878, 880-881, 883, 885, 888-892, 894-895, 936, 938, 948, 950, 952-953, 960-961 , 965, 967, 988, 991 , 1006, 1009, 1019-1020, 1033, 1035, 1057-1058, and/or 1066 of Neisseria meningitidis Cas9, or corresponding residues of Streptococcus pyogenes Cas9, Streptococcus thermophilus Cas9, or Staphylococcus aureus Cas9.

[0027] In some embodiments, the residues for substitution are selected from residues 12, 17, 41 , 43-46, 49, 68, 95, 97, 120, 122, 125, 130, 158, 174, 185, 196, 199, 203, 205, 208, 212-213, 227, 235, 238-239, 254-255, 257, 260, 262-263, 283, 287, 289, 304, 311 , 329-332, 337, 360, 365, 372, 374, 386, 388-391 , 407-408, 410-412, 434, 439, 448, 458, 472, 478, 481, 487-488, 491-495, 497, 511 , 513-515, 519, 537, 541 , 545, 553-554, 556, 558-559, 568, 573, 577, 596, 604, 611 , 632, 635, 643-644, 651 , 662, 680-681 , 685, 694, 702, 705-707, 709, 712, 716, 723, 725, 729, 747, 766, 791, 797, 821 , 824, 828-830, 832, 838, 844, 852, 857, 859-862, 867, 875, 885, 891 , 903, 907, 910, 919-920, 923-925, 928, 935, 941 , 953, 959, 961-962, 964-965, 967, 973, 985, 988, 995, 999, 1001 , 1010, 1016-1017, 1019-1020, 1028, 1030, 1058, 1001, and/or 1063 of Neisseria meningitidis Cas9, or corresponding residues of Streptococcus pyogenes Cas9, Streptococcus thermophilus Cas9, or Staphylococcus aureus Cas9.

[0028] In some embodiments, the MHC Class I and/or Class II binding sites of the Cas protein comprises a peptide comprising 9 (9mer) or 10 (lOmer) amino acids. In some embodiments, the introduced amino acid substitution is a naturally occurring amino acid.

[0029] In some embodiments, the invention is directed to a recombinant CRISPR-associated (Cas) protein comprising one or more amino acid substitutions in one or more residues corresponding to one or more MHC Class I and/or MHC Class II binding sites in a wild-type Cas protein, wherein the recombinant Cas protein has reduced immunogenicity compared to a wild- type Cas protein. In some embodiments, wherein the MHC Class I and/or Class II binding sites of the Cas protein comprises a peptide comprising 9 (9mer) or 10 (lOmer) amino acids. In some embodiments, the introduced amino acid substitution is a naturally occurring amino acid.

[0030] In some embodiments, the wild-type Cas protein comprises an amino sequence having at least 95% identity to the amino acid sequence of any one of SEQ ID NOs: 1-4.

[0031] In some embodiments, the invention is directed to a recombinant CRISPR-associated (Cas) protein made by a process comprising introducing one or more amino acid substitutions into one or more residues corresponding to one or more MHC Class I and/or MHC Class II binding sites of a wild-type Cas protein to form the recombinant Cas protein, wherein the recombinant Cas protein has reduced immunogenicity compared to the wild-type Cas protein. In some embodiments, the position of the one or more amino acid substitutions is determined by epitope mapping. In some embodiments, the epitope mapping is performed using in silico methods. In some embodiments, the epitope mapping is performed by a method comprising incubating an antigen presenting cell (APC) in the presence of a Cas protein and identifying peptides derived from the Cas protein bound to major histocompatibility (MHC) Class I and/or Class II proteins. In some embodiments, at least a portion of the MHC Class I and/or MHC Class II proteins bound to the Cas peptides is isolated. In some embodiments, the MHC Class I and/or MHC Class II proteins bound to the Cas peptides are isolated by immunoprecipitation.

[0032] In some embodiments, the invention is directed to a recombinant CRISPR-associated (Cas) protein comprising one or more substitutions in one or more amino acid residues of a wild- type Cas protein as designated in Figure 9, wherein the recombinant Cas protein has reduced immunogenicity compared to the wild-type Cas protein.

[0033] In some embodiments, the invention is directed to an isolated nucleic acid molecule comprising a nucleotide sequence that encodes a recombinant CRISPR-associated (Cas) protein, wherein the recombinant Cas protein has one or more amino acid substitutions in one or more residues corresponding to one or more MHC Class I and/or MHC Class II binding sites of a wild-type Cas protein, wherein the recombinant Cas protein has reduced immunogenicity compared to a wild-type Cas protein.

[0034] In some embodiments, the invention is directed to a vector comprising the nucleic acid as described herein. In some embodiments, the vector is a viral vector. In some embodiments, the viral vector is an adeno-associated viral vector or a lentiviral vector.

[0035] In some embodiments, the invention is directed to a cell comprising the recombinant Cas protein as described herein. In some embodiments, the invention is directed to a cell comprising the nucleic acid as described herein. In some embodiments, the invention is directed to a cell comprising a vector as described herein. In some embodiments, the cell is a prokaryotic cell. In some embodiments, the cell is an E. coli cell. In some embodiments, the cell is a eukaryotic cell. In embodiments, the cell is a mammalian cell. In some embodiments, the cell is a lymphocytic cell, a myeloid cell, an induced pluripotent stem cell (iPSC), or a T cell, although all mammalian cell types are envisaged as being used in the invention.

[0036] In some embodiments, the invention is directed to a library for identifying a CRISPR- associated (Cas) protein with reduced immunogenicity, the library comprising at least one recombinant Cas protein comprising one or more amino acid substitutions in one or more residues corresponding to one or more MHC Class I and/or MHC Class II binding sites of a wild-type Cas protein.

[0037] In some embodiments, the invention is directed to a method for altering the DNA sequence and/or gene expression at a genomic location containing a target sequence, the method comprising introducing into a cell containing the genetic element a guide RNA that hybridizes to the target sequence and a recombinant CRISPR-associated (Cas) protein comprising one or more amino acid substitutions in one or more residues corresponding to one or more MHC Class I and/or MHC Class II binding sites in a wild-type Cas protein, wherein expression of the one or more genetic elements is altered. In some embodiments, the guide RNA and/or the recombinant Cas protein are transfected into the cell. In some embodiments, the guide RNA and/or the recombinant Cas protein are located on a vector. In some embodiments, the guide RNA and the recombinant Cas protein are located on the same vector. In some embodiments, the vector is a viral vector. In some embodiments, the viral vector is an adeno-associated viral vector or a lentiviral vector.

[0038] In some embodiments, the disclosure is directed to a recombinant CRISPR-associated (Cas) protein, wherein said protein comprises one or more amino acid variation compared to a while-type Cas protein, wherein the one or more amino acid variation is at one or more positions corresponding to amino acid residues in the polypeptide of SEQ ID NO: 5, wherein the variation is at a position selected from the group consisting of (i) residues 100 to 120, (ii) residues 250 to 280, (iii) residues 690 to 710, (iv) residues 840 to 860, and (v) residues 1270 to 1295.

[0039] In some embodiments, the variation is at a position selected from the group consisting of (i) residues 102 to 120, (ii) residues 253 to 277, (iii) residues 692 to 709, (iv) residues 692 to 709, and (v) residues 1276 to 1292. [0040] In some embodiments, the variation is at a position selected from residues 102 to 120. In some embodiments, the variation is at a position selected from residues 253 to 277. In some embodiments, the variation is at a position selected from residues 692 to 709. In some embodiments, the variation is at a position selected from residues 692 to 709. In some embodiments, the variation is at a position selected from residues 1276 to 1292.

[0041] In some embodiments, the recombinant Cas protein comprises two or more amino acid variations. In some embodiments, the recombinant Cas protein comprises three or more amino acid variations. In some embodiments, the recombinant Cas protein comprises five or more amino acid variations. In some embodiments, the amino acid variation is a substitution of SEQ ID NO: 5 selected from the group consisting of one or more of the following:

[0042] In some embodiments, the amino acid variation is a substitution of SEQ ID NO: 5, wherein said amino acid substitutions include the following: L106D, K263D, L696G, L847D, and II 28 ID.

[0043] In some embodiments, the amino acid variation is determined using in silico epitope mapping.

[0044] In some embodiments, the amino acid variation is a substitution of one or more amino acids with one or more different amino acids.

[0045] In some embodiments, the amino acid variation comprises replacing one or more of aspargine, glutamine, leucine, lysine, methionine, serine, threonine, and valine residues with a different amino acid.

[0046] In some embodiments, the amino acid variation comprises replacing a polar amino acid with a nonpolar amino acid.

[0047] In some embodiments, the amino acid variation comprises replacing a hydrophilic amino acid with a hydrophobic amino acid.

[0048] In some embodiments, the amino acid variation is an insertion of one or more amino acids residues. In some embodiments, the amino acid variation is a deletion of one or more amino acid residues.

[0049] In some embodiments, the wild-type Cas protein has a sequence 90% identical to SEQ ID NO: 5. In some embodiments, the wild-type Cas protein is derived from a prokaryote. In some embodiments, the prokaryote is a bacterium. In some embodiments, the bacterium is from the genera Streptococcus, Staphylococcus, or Neisseria. In some embodiments, the bacterium is Streptococcus pyogenes, Streptococcus thermophilus, Staphylococcus aureus, or Neisseria meningtidis.

[0050] In some embodiments, the recombinant Cas protein is derived from Cas3, Cas9, or CaslO. In some embodiments, the recombinant Cas protein is derived from Cas9.

[0051] In some embodiments, the disclosure is directed to a polynucleotide encoding any Cas protein as described herein. In some embodiments, the disclosure is directed to an expression vector comprising any polynucleotide as described herein. In some embodiments, the disclosure is directed to a host cell comprising any expression vector as described herein.

BRIEF DESCRIPTION OF THE DRAWINGS

[0052] Figure 1 is a map of strong (S) and medium (M) peptide binding sites, epitopes, derived from Streptococcus pyogenes Cas9 for HLA Class I allotypes. Epitopes in each category for 9-mer and 10-mer sequences are presented. Data are presented for the first 125 amino acids of the Cas9 sequence. Values for different allotypes are represented separately, with allotype references restricted to two digits.

[0053] Figures 2A, 2B, and 2C are maps of strong (S) and medium (M) peptide binding sites, epitopes, derived from Streptococcus pyogenes Cas9 for HLA Class II allotypes. Epitopes in each category for 10-mer sequences are presented. Data are presented for the first 125 amino acids of the Cas9 sequence. Values for different allotypes are represented separately, with allotype references restricted to two digits.

[0054] Figure 3 is a map of strong (S) and medium (M) peptide binding sites, epitopes, derived from Streptococcus pyogenes Cas9 for HLA Class I allotypes. Epitopes in each category for 9-mer and 10-mer sequences are presented. Data are only presented for amino acids 1026- 1 150 of the Cas9 sequence. Values for different allotypes are represented separately, with allotype references restricted to two digits.

[0055] Figures 4A 4B and 4C are maps of strong (S) and medium (M) peptide binding sites, epitopes, derived from Streptococcus pyogenes Cas9 for HLA Class II allotypes. Epitopes in each category for 10-mer sequences are presented. Data are only presented for amino acids 1026- allotype references restricted to two digits.

[0056] Figures 5A, 5B and 5C depict the randomization of amino acids 1026-1 150 from Streptococcus pyogenes Cas9 and assessment of impact on 9mer immunogenicity. Data are presented for HLA-A and HLA-B Class I allotypes only. The left hand panel indicates the effect of amino acid substitution on the number of allotypes binding the specific epitope ("binders"). The right hand panel indicates the sum of the number of binders for each allotype multiplied by the allotype frequency in the global population (presented separately for A and B allotypes).

[0057] Figures 6A, 6B, 6C, and 6D depict the randomization of amino acids 1026-1150 from Streptococcus pyogenes Cas9 and assessment of impact on lOmer and global Cas9 immunogenicity. Data are presented for HLA-A and HLA-B Class I allotypes, and the DRB1 Class II allotypes only. The left hand panel indicates the effect of amino acid substitution on the number of allotypes binding the specific epitope ("binders") from the Class I and II allotypes above. The right hand panel indicates the sum of the number of binders for each allotype multiplied by the allotype frequency in the global population (presented separately for HLA-A, HLA-B and DRBl allotypes).

[0058] Figure 7 depicts a calculated protein sequence alignment between Streptococcus pyogenes Cas9 protein (SEQ ID NO: 5), Streptococcus thermophilus LMD-9 Cas9 protein (SEQ ID NO: 6), Staphylococcus aureus subsp. aureus Cas9 protein (SEQ ID NO: 7), and Neisseria meningitidis Cas9 protein (SEQ ID NO: 8).

[0059] Figure 8A depicts the amino acid substitution deimmunisation score for

Streptococcus pyogenes Cas9 protein (SEQ ID NO: 5) for 9mers (HLA Class I only). The amino acid substitution deimmunisation score is the sum of the number of HLA allotypes binding the given epitope when all possible substitutions are assessed for a specific position. The score therefore indicates the likelihood that for any given position a substitution can be identified that will deimmunise the corresponding epitope. A positive score indicates that substitutions at that position will give a greater chance of increasing epitope immunogenicity, where a negative score indicates that substitutions will give a greater chance in decreasing epitope immunogenicity. [0060] Figure 8B depicts the amino acid substitution deimmunisation score for

Streptococcus pyogenes Cas9 protein (SEQ ID NO: 5) for 9mers (HLA Class I only) where a cut-off of <-20 is applied. Amino acid positions in this category are viewed as targets for substitution in order to reduce Cas9 immunogenicity.

[0061] Figure 8C depicts the amino acid substitution deimmunisation score for

Streptococcus pyogenes Cas9 protein (SEQ ID NO: 5) for lOmers (HLA Class I and II). The amino acid substitution deimmunisation score is the sum of the number of HLA allotypes binding the given epitope when all possible substitutions are assessed for a specific position. The score therefore indicates the likelihood that for any given position a substitution can be identified that will deimmunise the corresponding epitope. A positive score indicates that substitutions at that position will give a greater chance of increasing epitope immunogenicity, where a negative score indicates that substitutions will give a greater chance in decreasing epitope immunogenicity.

[0062] Figure 8D depicts the amino acid substitution deimmunisation score for Streptococcus pyogenes Cas9 protein (SEQ ID NO: 5) for lOmers (HLA Class I and II) where a cut-off of <-20 is applied. Amino acid positions in this category are viewed as targets for substitution in order to reduce Cas9 immunogenicity.

[0063] Figure 9 represents target amino acid positions for substitution. Positions are identified from applying <-20 threshold to the amino acid substitution epitope deimmunisation score. Inclusion here indicates a high likelihood that a substitution can reduce epitope immunogenicity, however the specific substitutions made can be considered based on individual epitope binding scores and other related criteria such as amino acid characteristics and functional and structural localization (as discussed elsewhere).

[0064] Figure 10A represents the CD4+ T cell response induced by the whole Cas9 protein {Streptococcus pyogenes) in the DC:CD4 activation assay. Each dot represents a single healthy donor with a stimulation index > 2 deemed a positive T cell response. KLH is a highly immunogenic protein and induces a CD4+ T cell response in all 9 healthy donors. Cas9 induced a CD4+ T cell response in 5 of the 9 (56%) healthy donors tested suggesting that Cas9 contains CD4+ T cell epitopes and is capable of raising a helper T cell response in human cells. This assay can be used to evaluate deimmunised versions of the Cas9 protein and determine which have a reduced immunogenicity compared to the wild-type Cas9 protein.

[0065] Figure 10B represents the CD8+ T cell response induced by the whole Cas9 protein {Streptococcus pyogenes) in the DC:CD8 activation assay. Each dot represents a single healthy donor with a stimulation index > 2 deemed a positive T cell response. CEF is a highly immunogenic peptide pool and induces a CD4+ T cell response in all 9 healthy donors. Cas9 induced a CD8+ T cell response in 6 of the 9 (67%) healthy donors tested suggesting that Cas9 contains CD8+ T cell epitopes and is capable of raising a cytotoxic T cell response in human cells.

[0066] Figure 11 highlights the HLA-binding peptides eluted from HLA-DR molecules on the dendritic cells from a healthy donor after treatment with the whole Cas9 protein {Streptococcus pyogenes; SEQ ID NO: 5) using the MAPPs (MHC-associated peptide proteomics) assay. The Cas9 amino acid sequence is shown with the eluted HLA-binding peptides shown in bars underneath the sequences. The areas of the Cas9 protein highlighted by these bars represent areas of the Cas9 protein likely to be responsible for driving a CD4+ T cell response in this healthy donor. In conjunction with the in silico analysis, MAPPs analysis can help identify the area within the Cas9 protein responsible for driving the immune response and help guide protein engineering to remove these T cell epitopes.

DETAILED DESCRIPTION OF THE INVENTION

[0067] The invention provided herein uses components of the CRISPR-Cas system, which can be utilized to accomplish genomic engineering, including gene editing and altering expression of a gene and/or genetic element. Such genomic engineering can be used in various therapeutic strategies, including the treatment of genetic diseases. In addition, described herein are methods for creating Cas proteins with reduced immunogenicity that can be used in conjunction with other CRISPR components. In certain embodiments, described herein are recombinant Cas proteins with reduced immunogenicity, isolated nucleic acids that encode such recombinant Cas proteins, vectors comprising these nucleic acids, and cells comprising the nucleic acids and/or vectors. The creation of Cas proteins with reduced immunogenicity may allow for more stable, efficient, and efficacious use of the CRISPR-Cas system in a host organism, including humans.

Definitions

[0068] The use of the word "a" or "an" when used in conjunction with the term "comprising" in the claims and/or the specification may mean "one," but it is also consistent with the meaning of "one or more," "at least one," and "one or more than one."

[0069] Throughout this application, the term "about" is used to indicate that a value includes the inherent variation of error for the method/device being employed to determine the value, or the variation that exists among the study subjects. Typically the term is meant to encompass approximately or less than 1%, 2%, 3%, 4%, 5%, 6%, 7%, 8%, 9%, 10%, 1 1%, 12%, 13%, 14%, 15%, 16%, 17%, 18%, 19% or 20% variability depending on the situation.

[0070] The use of the term "or" in the claims is used to mean "and/or" unless explicitly indicated to refer only to altematives or the alternatives are mutually exclusive, although the disclosure supports a definition that refers to only alternatives and "and/or."

[0071] As used in this specification and claim(s), the words "comprising" (and any form of comprising, such as "comprise" and "comprises"), 'Tiaving" (and any form of having, such as "have" and '¾as"), "including" (and any form of including, such as "includes" and "include") or "containing" (and any form of containing, such as "contains" and "contain") are inclusive or open-ended and do not exclude additional, unrecited, elements or method steps. It is contemplated that any embodiment discussed in this specification can be implemented with respect to any method, system, host cells, expression vectors, and/or composition of the invention. Furthermore, compositions, systems, host cells, and/or vectors of the invention can be used to achieve methods and proteins of the invention.

[0072] The use of the term "for example" and its corresponding abbreviation "e.g." (whether italicized or not) means that the specific terms recited are representative examples and embodiments of the invention that are not intended to be limited to the specific examples referenced or cited unless explicitly stated otherwise. [0073] A "nucleic acid," "nucleic acid molecule," "oligonucleotide" or "polynucleotide" means a polymeric compound comprising covalently linked nucleotides. The term "nucleic acid" includes polyribonucleic acid (RNA) and polydeoxyribonucleic acid (DNA), both of which may be single- or double-stranded. DNA includes, but is not limited to, complimentary DNA (cDNA), genomic DNA, plasmid or vector DNA, and synthetic DNA. In some embodiments, the invention is directed to a polynucleotide encoding any one of the polypeptides disclosed herein, e.g., is directed to a polynucleotide encoding a Cas protein or variant thereof. In some embodiments, the invention is directed to a polynucleotide encoding Cas3, Cas9, CaslO or variants thereof.

[0074] A "gene" refers to an assembly of nucleotides that encode a polypeptide, and includes cDNA and genomic DNA nucleic acid molecules. "Gene" also refers to a nucleic acid fragment that can act as a regulatory sequences preceding (5' non-coding sequences) and following (3' non-coding sequences) the coding sequence.

[0075] The terms "peptide," "polypeptide," and "protein" are used interchangeably herein, and refer to a polymeric form of amino acids of any length, which can include coded and non- coded amino acids, chemically or biochemically modified or derivatized amino acids, and polypeptides having modified peptide backbones.

[0076] An "amino acid" as used herein refers to a compound containing both a carboxyl (- COOH) and amino (-NH 2 ) group. "Amino acid" refers to both natural and unnatural, i.e., synthetic, amino acids. Natural amino acids, with their three-letter and single letter abbreviations, include Alanine (Ala; A); Arginine (Arg, R); Asparagine (Asn; N); Aspartic acid (Asp; D); Cysteine (Cys; C); Glutamine (Gin; Q); Glutamic acid (Glu; E ); Glycine (Gly; G); Histidine (His; H); Isoleucine (He; I); Leucine (Leu; L); Lysine (Lys; K); Methionine (Met; M); Phenylalanine (Phe; F); Proline (Pro; P); Serine (Ser; S); Threonine (Thr; T); Tryptophan (Trp; W); Tyrosine (Tyr; Y); and Valine (Val; V).

[0077] An "amino acid substitution" refers to a polypeptide or protein comprising one or more substitutions of a wild-type or naturally occurring amino acid with a different amino acid relative to the wild-type or naturally occurring amino acid at that amino acid residue. The substituted amino acid of the invention may be a synthetic or naturally occurring amino acid. In certain embodiments, the substituted amino acid is a naturally occurring amino acid selected from the group consisting of: A, R, N, D, C, Q, E, G, H, I, L, K, M, F, P, S, T, W, Y, and V. Substitution mutants may be described using an abbreviated system. For example, a substitution mutation in which the fifth (5 th ) amino acid residue is substituted may be abbreviated as "X5Y," wherein "X" is the wild-type or naturally occurring amino acid to be replaced, "5" is the amino acid residue within the protein or polypeptide, and "Y" is the substituted, or non-wild-type or non-naturally occurring, amino acid.

[0078] The term "recombinant" when used in reference to a nucleic acid molecule, peptide, polypeptide, or protein means of, or resulting from, a new combination of genetic material that is not known to exist in nature. A recombinant molecule can be produced by any of the well-known techniques available in the field of recombinant technology, including, but not limited to, polymerase chain reaction (PCR), gene splicing (e.g., using restriction endonucleases), and solid state synthesis of nucleic acid molecules, peptides, or proteins.

[0079] An "isolated" polypeptide, protein, peptide, or nucleic acid is a molecule that has been removed from its natural environment. It is also to be understood that "isolated" polypeptides, proteins, peptides, or nucleic acids may be formulated with excipients such as diluents or adjuvants and still be considered isolated.

[0080] The terms "sequence identity" or "% identity" in the context of nucleic acid sequences or amino acid sequences refers to the percentage of residues in the compared sequences that are the same when the sequences are aligned over a specified comparison window. A comparison window can be a segment of at least 10 to over 1000 residues in which the sequences can be aligned and compared. Methods of alignment for determination of sequence identity are well- known can be performed using publically available databases such as BLAST (blast.ncbi.nlm.nih.gov/Blast.cgi.). "Percent identity" or "% identity" when referring to amino acid sequences can be determined by methods known in the art. For example, in some embodiments, "percent identity" of two amino acid sequences is determined using the algorithm of Karlin and Altschul, Proc. Natl. Acad. Sci. USA 57:2264-68, 1990, modified as in Karlin and Altschul Proc. Natl. Acad. Sci. USA 90:5873-77, 1993. Such an algorithm is incorporated into the NBLAST and XBLAST programs (version 2.0) of Altschul, et al, J. Mol. Biol. 275:403-10 (1990). BLAST protein searches can be performed with the XBLAST program, score=50, wordlength=3 to obtain amino acid sequences homologous to the protein molecules of the invention. Where gaps exist between two sequences, Gapped BLAST can be utilized as described in Altschul et al, Nucleic Acids Res. 25(17):3389-3402, 1997. When utilizing BLAST and Gapped BLAST programs, the default parameters of the respective programs (e.g., XBLAST and NBLAST) can be used.

[0081] In certain embodiments of the invention, polypeptides or nucleic acid molecules have 70%, at least 70%, 75%, at least 75%, 80%, at least 80%, 85%, at least 85%, 90%, at least 90%, 95%, at least 95%, 97%, at least 97%, 98%, at least 98%, 99%, or at least 99% or 100% sequence identity with a reference polypeptide or nucleic acid molecule, respectively (or a fragment of the reference polypeptide or nucleic acid molecule). In some embodiments, polypeptides or nucleic acid molecules have about 70%, at least about 70%, about 75%, at least about 75%, about 80%, at least about 80%, about 85%, at least about 85%, about 90%, at least about 90%, about 95%, at least about 95%, about 97%, at least about 97%, about 98%, at least about 98%, about 99%, at least about 99% or about 100% sequence identity with a reference polypeptide or nucleic acid molecule, respectively (or a fragment of the reference polypeptide or nucleic acid molecule).

[0082] The RNA molecule that binds to CRISPR-Cas components and targets them to a specific location within the target DNA is referred to herein as "guide RNA," "gRNA," or "small guide RNA" and may also be referred to herein as a "DNA-targeting RNA." A guide RNA comprises at least two nucleotide segments: at least one "DNA-binding segment" and at least one "polypeptide -binding segment." By "segment" is meant a part, section, or region of a molecule, e.g., a contiguous stretch of nucleotides of an RNA molecule. The definition of "segment," unless otherwise specifically defined, is not limited to a specific number of total base pairs.

[0083] The guide RNA can be introduced into the target cell as an isolated RNA molecule, or is introduced into the cell using an expression vector containing DNA encoding the guide RNA.

[0084] The "DNA-binding segment" (or "DNA-targeting sequence") of the guide RNA comprises a nucleotide sequence that is complementary to a specific sequence within a target DNA. [0085] The guide R A of the current disclosure can include one or more polypeptide - binding sequences/segments. The polypeptide -binding segment (or "protein-binding sequence") of the guide RNA interacts with the RNA-binding domain of a Cas protein of the current disclosure. Such polypeptide -binding segments or sequences are known to those of skill in the art, e.g., those disclosed in U.S. patent application publications 2014/0068797, 2014/0273037, 2014/0273226, 2014/0295556, 2014/0295557, 2014/0349405, 2015/0045546, 2015/0071898, 2015/0071899, and 2015/0071906, the disclosures of which are incorporated herein in their entireties.

[0086] "T cell" or "T-cell" are used interchangeably and refer to a type of lymphocytic cell that plays a central role in cell-mediated immunity and expresses a T-cell receptor (TCR) on its surface. T-cells include CD4+ and CD8+ T-cells but are distinguished from other lymphocytes, such as B cells and natural killer cells (NK cells).

[0087] A "major histocompatibility complex" or "MHC" protein as used herein refers to a set of cell surface molecules encoded by a large gene family that play a significant role in the immune system of vertebrates. A key function of these proteins is to bind peptide fragments derived from endogenous or exogenous (foreign) proteins and display them on the cell surface for recognition by the appropriate T-cells of the host organism. The MHC gene family is divided into three subgroups: Class I, Class II, and Class III. The human MHC Class I and Class II genes are also referred to as human leukocyte antigen (HLA) - HLA Class I and HLA Class II, respectively. Some of the most studied HLA genes in humans are the nine MHC genes: HLA-A, HLA-B, HLA-C, HLA-DPAl, HLA-DPBl, HLA-DQAl, HLA-DQBl, HLA-DRA, HLA-DRBl and HLA-DRB345.

[0088] "Binding" or "interaction" as used herein refers to a non-covalent interaction between macromolecules (e.g., between DNA and RNA, or between a polypeptide and a polynucleotide). "Binding" may also be referred to as "associated with" or "interacting." "Binding" as used herein means that the binding partners are capable of binding to each other (e.g., will not necessarily bind to each other). Some portions of a binding interaction may be sequence-specific, but not all components of a binding interaction need be sequence-specific (e.g., contacts with phosphate residues in a DNA backbone). Binding interactions are generally characterized by a dissociation constant (Kd), e.g., less than 1 mM, less than 100 uM, less than 10 uM, less than 1 uM, less than 100 nM, less than 10 nM. "Affinity" refers to the strength of binding, increased binding affinity being correlated with a lower Kd.

[0089] As used herein, "promoter," "promoter sequence," or "promoter region" refers to a DNA regulatory region/sequence capable of binding RNA polymerase and involved in initiating transcription of a downstream coding or non-coding sequence. In some examples of the present disclosure, the promoter sequence includes the transcription initiation site and extends upstream to include the minimum number of bases or elements necessary to initiate transcription at levels detectable above background. In some embodiments, the promoter sequence includes a transcription initiation site, as well as protein binding domains responsible for the binding of RNA polymerase. Eukaryotic promoters will often, but not always, contain "TATA" boxes and "CAT" boxes. Various promoters, including inducible promoters, may be used to drive the various vectors of the present invention.

[0090] A "vector" or "expression vector" is a replicon, such as a plasmid, phage, virus, or cosmid, to which another DNA segment may be attached to bring about the replication and/or expression of the attached DNA segment in a cell. "Vector" includes episomal (e.g., plasmids) and non episomal vectors. In some embodiments of the present disclosure the vector is an episomal vector, which is removed/lost from a population of cells after a number of cellular generations, e.g., by asymmetric partitioning. The term "vector" includes both viral and nonviral means for introducing a nucleic acid molecule into a cell in vitro, in vivo, or ex vivo. Vectors may be introduced into the desired host cells by well-known methods, including, but not limited to, transfection, transduction, cell fusion, and lipofection. Vectors can comprise various regulatory elements including promoters. In some embodiments, vector designs can be based on constructs designed by Mali et al. "Cas9 as a versatile tool for engineering biology," Nature Methods 10:957-63 (2013). In some embodiments, the invention is directed to an expression vector comprising any of the polynucleotides described herein, e.g., an expression vector comprising polynucleotides encoding a Cas protein or variant thereof. In some embodiments, the invention is directed to an expression vector comprising polynucleotides encoding a Cas3, Cas9, or Cas 10 protein or variant thereof. [0091] "Transfection" as used herein means the introduction of an exogenous nucleic acid molecule, including a vector, into a cell. A "transfected" cell comprises an exogenous nucleic acid molecule inside the cell and a "transformed" cell is one in which the exogenous nucleic acid molecule within the cell induces a phenotypic change in the cell. The transfected nucleic acid molecule can be integrated into the host cell's genomic DNA and/or can be maintained by the cell, temporarily or for a prolonged period of time, extra-chromosomally. Host cells or organisms that express exogenous nucleic acid molecules or fragments are referred to as "recombinant," "transformed," or "transgenic" organisms. In some embodiments, the invention is directed to a host cell comprising any of the expression vectors described herein, e.g., an expression vector comprising a polynucleotide encoding a Cas protein or variant thereof. In some embodiments, the invention is directed to a host cell comprising an expression vector comprising a polynucleotide encoding a Cas3, Cas 9 or Cas 10 protein or variant thereof.

[0092] The term "in silico" as used herein refers to a process or analysis preformed on a computer, including computer modeling and computer simulation. In some embodiments, the term "in silico" refers to the EPIBASE ® epitope prediction method.

Recombinant CRISPR-associated (Cas) proteins

[0093] As described above, Cas proteins are a critical component in the CRISPR-Cas system, which can be used for, inter alia, genome editing, gene regulation, genetic circuit construction, and functional genomics. While the Casl and Cas2 proteins appear to be universal to all the presently identified CRISPR systems, the Cas3, Cas9, and Cas 10 proteins are thought to be specific to the Type I, Type II, and Type III CRISPR systems, respectively. In certain embodiments of the present invention, the recombinant Cas protein with reduced immunogenicity, including methods of deriving such proteins, is derived from a Cas3, Cas9, or CaslO protein. In some embodiments, the recombinant Cas protein with reduced immunogenicity, including methods of deriving such a protein, is derived from a Cas9 protein.

[0094] Following initial publications around the CRISPR-Cas9 system (Type II system), Cas9 variants have been identified in a range of bacterial species and a number have been functionally characterized. See, e.g., Chylinski, et al., "Classification and evolution of type II CRISPR-Cas sytems," Nucleic Acids Research 42(10):6091-105 (2014), Ran, et al, "In vivo genome editing using Staphylococcus aureus Cas9," Nature 520(7546): 186-91 (2015) and Esvelt, et al, "Orthogonal Cas9 proteins for RNA-guided gene regulation and editing," Nature Methods 10(1 1):1 116-21 (2013), each of which is incorporated by reference herein in its entirety.

[0095] Cas9 variants from Streptococcus pyogenes, Streptococcus thermophilus, staphylococcus aureus, and Neisseria meningitidis, in particular, have been well-characterized. In some embodiments, the recombinant Cas protein with reduced immunogenicity, including methods of deriving such a protein, is derived from a Cas9 protein from a bacterium of the genera, Streptococcus, Staphylococcus, or Neisseria. In certain embodiments, the recombinant Cas9 protein is derived from a Cas9 protein from Streptococcus pyogenes, Streptococcus thermophilus, Staphylococcus aureus, or Neisseria meningitidis. In some embodiments, the recombinant Cas9 protein is derived from a Streptococcus pyogenes Cas9 protein. In other embodiments, the recombinant Cas9 protein is derived from a Staphylococcus aureus Cas9 protein.

[0096] Following is an exemplary nucleotide sequence (SEQ ID NO: 1) that encodes the Streptococcus pyogenes Cas9 protein:

ATGGACAAGAAGTACTCCATTGGGCTCGATATCGGCACAAACAGCGTC

GGCTGGGCCGTCATTACGGACGAGTACAAGGTGCCGAGCAAAAAATT

CAAAGTTCTGGGCAATACCGATCGCCACAGCATAAAGAAGAACCTCA

TTGGCGCCCTCCTGTTCGACTCCGGGGAGACGGCCGAAGCCACGCGGC

TCAAAAGAACAGCACGGCGCAGATATACCCGCAGAAAGAATCGGATC

TGCTACCTGCAGGAGATCTTTAGTAATGAGATGGCTAAGGTGGATGAC

TCTTTCTTCCATAGGCTGGAGGAGTCCTTTTTGGTGGAGGAGGATAAA

AAGCACGAGCGCCACCCAATCTTTGGCAATATCGTGGACGAGGTGGC

GTACCATGAAAAGTACCCAACCATATATCATCTGAGGAAGAAGCTTGT

AGACAGTACTGATAAGGCTGACTTGCGGTTGATCTATCTCGCGCTGGC

GCATATGATCAAATTTCGGGGACACTTCCTCATCGAGGGGGACCTGAA

CCCAGACAACAGCGATGTCGACAAACTCTTTATCCAACTGGTTCAGAC

TTACAATCAGCTTTTCGAAGAGAACCCGATCAACGCATCCGGAGTTGA

CGCCAAAGCAATCCTGAGCGCTAGGCTGTCCAAATCCCGGCGGCTCGA AAACCTCATCGCACAGCTCCCTGGGGAGAAGAAGAACGGCCTGTTTG

GTAATCTTATCGCCCTGTCACTCGGGCTGACCCCCAACTTTAAATCTAA

CTTCGACCTGGCCGAAGATGCCAAGCTTCAACTGAGCAAAGACACCTA

CGATGATGATCTCGACAATCTGCTGGCCCAGATCGGCGACCAGTACGC

AGACCTTTTTTTGGCGGCAAAGAACCTGTCAGACGCCATTCTGCTGAG

TGATATTCTGCGAGTGAACACGGAGATCACCAAAGCTCCGCTGAGCGC

TAGTATGATCAAGCGCTATGATGAGCACCACCAAGACTTGACTTTGCT

GAAGGCCCTTGTCAGACAGCAACTGCCTGAGAAGTACAAGGAAATTTT

CTTCGATCAGTCTAAAAATGGCTACGCCGGATACATTGACGGCGGAGC

AAGCCAGGAGGAATTTTACAAATTTATTAAGCCCATCTTGGAAAAAAT

GGACGGCACCGAGGAGCTGCTGGTAAAGCTTAACAGAGAAGATCTGT

TGCGCAAACAGCGCACTTTCGACAATGGAAGCATCCCCCACCAGATTC

ACCTGGGCGAACTGCACGCTATCCTCAGGCGGCAAGAGGATTTCTACC

CCTTTTTGAAAGATAACAGGGAAAAGATTGAGAAAATCCTCACATTTC

GGATACCCTACTATGTAGGCCCCCTCGCCCGGGGAAATTCCAGATTCG

CGTGGAT GACTCGC AAAT CAGAAGAGACC ATC ACTCCCTGGAACTTCG

AGGAAGTCGTGGATAAGGGGGCCTCTGCCCAGTCCTTCATCGAAAGG

ATGACTAACTTTGATAAAAATCTGCCTAACGAAAAGGTGCTTCCTAAA

CACTCTCTGCTGTACGAGTACTTCACAGTTTATAACGAGCTCACCAAG

GTCAAATACGTCACAGAAGGGATGAGAAAGCCAGCATTCCTGTCTGG

AGAGCAGAAGAAAGCTATCGTGGACCTCCTCTTCAAGACGAACCGGA

AAGTTACCGTGAAACAGCTCAAAGAAGACTATTTCAAAAAGATTGAA

TGTTTCGACTCTGTTGAAATCAGCGGAGTGGAGGATCGCTTCAACGCA

TCCCTGGGAACGTATCACGATCTCCTGAAAATCATTAAAGACAAGGAC

TTCCTGGACAATGAGGAGAACGAGGACATTCTTGAGGACATTGTCCTC

ACCCTTACGTTGTTTGAAGATAGGGAGATGATTGAAGAACGCTTGAAA

ACTTACGCTCATCTCTTCGACGACAAAGTCATGAAACAGCTCAAGAGG

CGCCGATATACAGGATGGGGGCGGCTGTCAAGAAAACTGATCAATGG

GATCCGAGACAAGCAGAGTGGAAAGACAATCCTGGATTTTCTTAAGTC

CGATGGATTTGCCAACCGGAACTTCATGCAGTTGATCCATGATGACTC

TCTCACCTTTAAGGAGGACATCCAGAAAGCACAAGTTTCTGGCCAGGG GGACAGTCTTCACGAGCACATCGCTAATCTTGCAGGTAGCCCAGCTAT

CAAAAAGGGAATACTGCAGACCGTTAAGGTCGTGGATGAACTCGTCA

AAGTAATGGGAAGGCATAAGCCCGAGAATATCGTTATCGAGATGGCC

CGAGAGAACCAAACTACCCAGAAGGGACAGAAGAACAGTAGGGAAA

GGATGAAGAGGATTGAAGAGGGTATAAAAGAACTGGGGTCCCAAATC

CTTAAGGAACACCCAGTTGAAAACACCCAGCTTCAGAATGAGAAGCT

CTACCTGTACTACCTGCAGAACGGCAGGGACATGTACGTGGATCAGGA

ACTGGACATCAATCGGCTCTCCGACTACGACGTGGATCATATCGTGCC

CCAGTCTTTTCTCAAAGATGATTCTATTGATAATAAAGTGTTGACAAG

ATCCGATAAAAATAGAGGGAAGAGTGATAACGTCCCCTCAGAAGAAG

TTGTCAAGAAAATGAAAAATTATTGGCGGCAGCTGCTGAACGCCAAA

CTGATCACACAACGGAAGTTCGATAATCTGACTAAGGCTGAACGAGGT

GGCCTGTCTGAGTTGGATAAAGCCGGCTTCATCAAAAGGCAGCTTGTT

GAGACACGCCAGATCACCAAGCACGTGGCCCAAATTCTCGATTCACGC

ATGAACACCAAGTACGATGAAAATGACAAACTGATTCGAGAGGTGAA

AGTTATTACTCTGAAGTCTAAGCTGGTCTCAGATTTCAGAAAGGACTT

TCAGTTTTATAAGGTGAGAGAGATCAACAATTACCACCATGCGCATGA

TGCCTACCTGAATGCAGTGGTAGGCACTGCACTTATCAAAAAATATCC

CAAGCTTGAATCTGAATTTGTTTACGGAGACTATAAAGTGTACGATGT

TAGGAAAATGATCGCAAAGTCTGAGCAGGAAATAGGCAAGGCCACCG

CTAAGTACTTCTTTTACAGCAATATTATGAATTTTTTCAAGACCGAGAT

TACACTGGCCAATGGAGAGATTCGGAAGCGACCACTTATCGAAACAA

ACGGAGAAACAGGAGAAATCGTGTGGGACAAGGGTAGGGATTTCGCG

ACAGTCCGGAAGGTCCTGTCCATGCCGCAGGTGAACATCGTTAAAAAG

ACCGAAGTACAGACCGGAGGCTTCTCCAAGGAAAGTATCCTCCCGAA

AAGGAACAGCGACAAGCTGATCGCACGCAAAAAAGATTGGGACCCCA

AGAAATACGGCGGATTCGATTCTCCTACAGTCGCTTACAGTGTACTGG

TTGTGGCCAAAGTGGAGAAAGGGAAGTCTAAAAAACTCAAAAGCGTC

AAGGAACTGCTGGGCATCACAATCATGGAGCGATCAAGCTTCGAAAA

AAACCCCATCGACTTTCTCGAGGCGAAAGGATATAAAGAGGTCAAAA

AAGACCTCATCATTAAGCTTCCCAAGTACTCTCTCTTTGAGCTTGAAAA CGGCCGGAAACGAATGCTCGCTAGTGCGGGCGAGCTGCAGAAAGGTA

ACGAGCTGGCACTGCCCTCTAAATACGTTAATTTCTTGTATCTGGCCAG

CCACTATGAAAAGCTCAAAGGGTCTCCCGAAGATAATGAGCAGAAGC

AGCTGTTCGTGGAACAACACAAACACTACCTTGATGAGATCATCGAGC

AAATAAGCGAATTCTCCAAAAGAGTGATCCTCGCCGACGCTAACCTCG

ATAAGGTGCTTTCTGCTTACAATAAGCACAGGGATAAGCCCATCAGGG

AGCAGGCAGAAAACATTATCCACTTGTTTACTCTGACCAACTTGGGCG

CGCCTGCAGCCTTCAAGTACTTCGACACCACCATAGACAGAAAGCGGT

ACACCTCTACAAAGGAGGTCCTGGACGCCACACTGATTCATCAGTCAA

TTACGGGGCTCTATGAAACAAGAATCGACCTCTCTCAGCTCGGTGGAG

ACTGA

Following is an exemplary nucleotide sequence (SEQ ID NO: 2) that enco ococcus thermophilus Cas9 protein:

ATGAGCGACCTGGTGCTGGGCCTGGACATCGGCATCGGCAGCGTGGG

CGTGGGCATCCTGAACAAGGTGACCGGCGAGATCATCCACAAGAACA

GTCGCATCTTCCCTGCTGCTCAGGCTGAGAACAACCTGGTGCGCCGCA

CCAACCGCCAGGGTCGCCGGCTTGCTCGCCGCAAGAAGCACCGGCGC

GTGCGCCTGAACCGCCTGTTCGAGGAGAGCGGCCTGATCACCGACTTC

ACCAAGATCAGCATCAACCTGAACCCCTACCAGCTGCGCGTGAAGGG

CCTGACCGACGAGCTGAGCAACGAGGAGCTGTTCATCGCCCTGAAGA

ACATGGTGAAGCACCGCGGCATCAGCTACCTGGACGACGCCAGCGAC

GACGGCAACAGCAGCGTGGGCGACTACGCCCAGATCGTGAAGGAGAA

CAGCAAGCAGCTGGAGACCAAGACCCCCGGCCAGATCCAGCTGGAGC

GCTACCAGACCTACGGCCAGCTGCGCGGCGACTTCACCGTGGAGAAG

GACGGCAAGAAGCACCGCCTGATCAACGTGTTCCCCACCAGCGCCTAC

CGCAGCGAGGCCCTGCGCATCCTGCAGACCCAGCAGGAGTTCAACCC

CCAGATCACCGACGAGTTCATCAACCGCTACCTGGAGATCCTGACCGG

CAAGCGCAAGTACTACCACGGCCCCGGCAACGAGAAGAGCCGCACCG

ACTACGGCCGCTACCGCACCAGCGGCGAGACCCTGGACAACATCTTCG

GCATCCTGATCGGCAAGTGCACCTTCTACCCCGACGAGTTCCGCGCCG CCAAGGCCAGCTACACCGCCCAGGAGTTCAACCTGCTGAACGACCTG

AACAACCTGACCGTGCCCACCGAGACCAAGAAGCTGAGCAAGGAGCA

GAAGAACCAGATCATCAACTACGTGAAGAACGAGAAGGCCATGGGCC

CCGCCAAGCTGTTCAAGTACATCGCCAAGCTGCTGAGCTGCGACGTGG

CCGACATCAAGGGCTACCGCATCGACAAGAGCGGCAAGGCCGAGATC

CACACCTTCGAGGCCTACCGCAAGATGAAGACCCTGGAGACCCTGGA

CATCGAGCAGATGGACCGCGAGACCCTGGACAAGCTGGCCTACGTGC

TGACCCTGAACACCGAGCGCGAGGGCATCCAGGAGGCCCTGGAGCAC

GAGTTCGCCGACGGCAGCTTCAGCCAGAAGCAGGTGGACGAGCTGGT

GCAGTTCCGCAAGGCCAACAGCAGCATCTTCGGCAAGGGCTGGCACA

ACTTCAGCGTGAAGCTGATGATGGAGCTGATCCCCGAGCTGTACGAGA

CCAGCGAGGAGCAGATGACCATCCTGACCCGCCTGGGCAAGCAGAAG

ACCACCAGCAGCAGCAACAAGACCAAGTACATCGACGAGAAGCTGCT

GACCGAGGAGATCTACAACCCCGTGGTGGCCAAGAGCGTGCGCCAGG

CCATCAAGATCGTGAACGCCGCCATCAAGGAGTACGGCGACTTCGAC

AACATCGTGATCGAGATGGCCCGCGAGACCAACGAGGACGACGAGAA

GAAGGCCATCCAGAAGATCCAGAAGGCCAACAAGGACGAGAAGGAC

GCCGCCATGCTGAAGGCCGCCAACCAGTACAACGGCAAGGCCGAGCT

GCCCCACAGCGTGTTCCACGGCCACAAGCAGCTGGCCACCAAGATCC

GCCTGTGGCACCAGCAGGGCGAGCGCTGCCTGTACACCGGCAAGACC

ATCAGCATCCACGACCTGATCAACAACAGCAACCAGTTCGAGGTGGA

CCACATCCTGCCCCTGAGCATCACCTTCGACGACAGCCTGGCCAACAA

GGTGCTGGTGTACGCCACCGCCAACCAGGAGAAGGGCCAGCGCACCC

CCTACCAGGCCCTGGACAGCATGGACGACGCCTGGAGCTTCCGCGAG

CTGAAGGCCTTCGTGCGCGAGAGCAAGACCCTGAGCAACAAGAAGAA

GGAGTACCTGCTGACCGAGGAGGACATCAGCAAGTTCGACGTGCGCA

AGAAGTTCATCGAGCGCAACCTGGTGGACACCCGCTACGCCAGCCGC

GTGGTGCTGAACGCCCTGCAGGAGCACTTCCGCGCCCACAAGATCGAC

ACCAAGGTGAGCGTGGTGCGCGGCCAGTTCACCAGCCAGCTGCGCCG

CCACTGGGGCATCGAGAAGACCCGCGACACCTACCACCACCACGCCG

TGGACGCCCTGATCATTGCGGCTTCTAGCCAGCTGAACCTGTGGAAGA AGCAGAAGAACACCCTGGTGAGCTACAGCGAGGACCAGCTGCTGGAC

ATCGAGACCGGCGAGCTGATCAGCGACGACGAGTACAAGGAGAGCGT

GTTCAAGGCCCCCTACCAGCACTTCGTGGACACCCTGAAGAGCAAGG

AGTTCGAGGACAGCATCCTGTTCAGCTACCAGGTGGACAGCAAGTTCA

ACCGCAAGATCAGCGACGCCACCATCTACGCCACCCGCCAGGCCAAG

GTGGGCAAGGACAAGGCCGACGAGACCTACGTGCTGGGCAAGATCAA

GGACATCTACACCCAGGACGGCTACGACGCCTTCATGAAGATCTACAA

GAAGGACAAGAGCAAGTTCCTGATGTACCGCCACGACCCCCAGACCT

TCGAGAAGGTGATCGAGCCCATCCTGGAGAACTACCCCAACAAGCAG

ATCAACGATAAAGGCAAGGAGGTGCCCTGCAACCCCTTCCTGAAGTA

CAAGGAGGAGCACGGCTACATCCGCAAGTACAGCAAGAAGGGCAACG

GCCCCGAGATCAAGAGCCTGAAGTACTACGACAGCAAGCTGGGCAAC

CACATCGACATCACCCCCAAGGACAGCAACAACAAGGTGGTGCTGCA

GAGCGTGAGCCCCTGGCGCGCCGACGTGTACTTCAACAAGACCACCG

GCAAGTACGAGATCCTGGGCCTGAAGTACGCCGACCTGCAGTTTGATA

AGGGCACCGGCACCTACAAGATCAGCCAGGAGAAGTACAACGACATC

AAGAAGAAGGAGGGCGTGGACAGCGACAGCGAGTTCAAGTTCACCCT

GTACAAGAACGACCTTCTGCTGGTGAAGGACACCGAGACCAAGGAGC

AACAGCTGTTCCGCTTCCTGAGCCGCACCATGCCCAAGCAGAAGCACT

ACGTGGAGCTGAAGCCCTACGACAAGCAGAAGTTCGAGGGCGGCGAG

GCCCTGATCAAGGTGCTGGGCAACGTGGCCAACAGCGGCCAGTGCAA

GAAGGGCCT GGGCAAGAGCAACAT CAGCATCT ACAAGGTGCGC ACCG

ACGTGCTGGGCAACCAGCACATCATCAAGAACGAGGGCGACAAGCCC

AAGCTGGACTTCTGA

[0098] Following is an exemplary nucleotide sequence (SEQ ID NO: 3) that enco Staphylococcus aureus Cas9 protein:

ATGAAGCGGAACTACATCCTGGGCCTGGACATCGGCATCACCAGCGT GGGCTACGGCATCATCGACTACGAGACACGGGACGTGATCGATGCCG GCGTGCGGCTGTTCAAAGAGGCCAACGTGGAAAACAACGAGGGCAGG CGGAGCAAGAGAGGCGCCAGAAGGCTGAAGCGGCGGAGGCGGCATA GAATCCAGAGAGTGAAGAAGCTGCTGTTCGACTACAACCTGCTGACCG

ACCACAGCGAGCTGAGCGGCATCAACCCCTACGAGGCCAGAGTGAAG

GGCCTGAGCCAGAAGCTGAGCGAGGAAGAGTTCTCTGCCGCCCTGCT

GCACCT GGCC AAGAGAAGAGGCGT GCAC AACGTGAACGAGGTGGAAG

AGGACACCGGCAACGAGCTGTCCACCAAAGAGCAGATCAGCCGGAAC

AGCAAGGCCCTGGAAGAGAAATACGTGGCCGAACTGCAGCTGGAACG

GCTGAAGAAAGACGGCGAAGTGCGGGGCAGCATCAACAGATTCAAGA

CCAGCGACTACGTGAAAGAAGCCAAACAGCTGCTGAAGGTGCAGAAG

GCCTACCACCAGCTGGACCAGAGCTTCATCGACACCTACATCGACCTG

CTGGAAACCCGGCGGACCTACTATGAGGGACCTGGCGAGGGCAGCCC

CTTCGGCTGGAAGGACATCAAAGAATGGTACGAGATGCTGATGGGCC

ACTGCACCTACTTCCCCGAGGAACTGCGGAGCGTGAAGTACGCCTACA

ACGCCGACCTGTACAACGCCCTGAACGACCTGAACAATCTCGTGATCA

CCAGGGACGAGAACGAGAAGCTGGAATATTACGAGAAGTTCCAGATC

ATCGAGAACGTGTTCAAGCAGAAGAAGAAGCCCACCCTGAAGCAGAT

CGCCAAAGAAATCCTCGTGAACGAAGAGGATATTAAGGGCTACAGAG

TGACCAGCACCGGCAAGCCCGAGTTCACCAACCTGAAGGTGTACCAC

GACATCAAGGACATTACCGCCCGGAAAGAGATTATTGAGAACGCCGA

GCTGCTGGATCAGATT GCCAAGATCCT GACC ATCTACCAGAGC AGCGA

GGACATCCAGGAAGAACTGACCAATCTGAACTCCGAGCTGACCCAGG

AAGAGATCGAGCAGATCTCTAATCTGAAGGGCTATACCGGCACCCAC

AACCTGAGCCTGAAGGCCATCAACCTGATCCTGGACGAGCTGTGGCAC

ACCAACGACAACCAGATCGCTATCTTCAACCGGCTGAAGCTGGTGCCC

AAGAAGGTGGACCTGTCCCAGCAGAAAGAGATCCCCACCACCCTGGT

GG ACG ACTTC ATCCT GAGCCCCGTCGT GAAGAGAAGCTTCATCCAGAG

CATCAAAGTGATCAACGCCATCATCAAGAAGTACGGCCTGCCCAACG

ACATCATTATCGAGCTGGCCCGCGAGAAGAACTCCAAGGACGCCCAG

AAAATGATCAACGAGATGCAGAAGCGGAACCGGCAGACCAACGAGC

GGATCGAGGAAATCATCCGGACCACCGGCAAAGAGAACGCCAAGTAC

CTGATCGAGAAGATCAAGCTGCACGACATGCAGGAAGGCAAGTGCCT

GTACAGCCTGGAAGCCATCCCTCTGGAAGATCTGCTGAACAACCCCTT CAACTATGAGGTGGACCACATCATCCCCAGAAGCGTGTCCTTCGACAA

CAGCTTCAACAACAAGGTGCTCGTGAAGCAGGAAGAAAACAGCAAGA

AGGGCAACCGGACCCCATTCCAGTACCTGAGCAGCAGCGACAGCAAG

ATCAGCTACGAAACCTTCAAGAAGCACATCCTGAATCTGGCCAAGGG

CAAGGGCAGAATCAGCAAGACCAAGAAAGAGTATCTGCTGGAAGAAC

GGGACATCAACAGGTTCTCCGTGCAGAAAGACTTCATCAACCGGAAC

CTGGTGGATACCAGATACGCCACCAGAGGCCTGATGAACCTGCTGCG

GAGCTACTTCAGAGTGAACAACCTGGACGTGAAAGTGAAGTCCATCA

ATGGCGGCTTCACCAGCTTTCTGCGGCGGAAGTGGAAGTTTAAGAAAG

AGCGGAACAAGGGGTACAAGCACCACGCCGAGGACGCCCTGATCATT

GCCAACGCCGATTTCATCTTCAAAGAGTGGAAGAAACTGGACAAGGC

CAAAAAAGTGATGGAAAACCAGATGTTCGAGGAAAAGCAGGCCGAGA

GCATGCCCGAGATCGAAACCGAGCAGGAGTACAAAGAGATCTTCATC

ACCCCCCACCAGATCAAGCACATTAAGGACTTCAAGGACTACAAGTA

CAGCCACCGGGTGGACAAGAAGCCTAATAGAGAGCTGATTAACGACA

CCCTGTACTCCACCCGGAAGGACGACAAGGGCAACACCCTGATCGTG

AACAATCTGAACGGCCTGTACGACAAGGACAATGACAAGCTGAAAAA

GCTGATCAACAAGAGCCCCGAAAAGCTGCTGATGTACCACCACGACC

CCCAGACCTACCAGAAACTGAAGCTGATTATGGAACAGTACGGCGAC

GAGAAGAATCCCCTGTACAAGTACTACGAGGAAACCGGGAACTACCT

GACCAAGTACTCCAAAAAGGACAACGGCCCCGTGATCAAGAAGATTA

AGTATTACGGCAACAAACTGAACGCCCATCTGGACATCACCGACGACT

ACCCCAACAGCAGAAACAAGGTCGTGAAGCTGTCCCTGAAGCCCTAC

AGATTCGACGTGTACCTGGACAATGGCGTGTACAAGTTCGTGACCGTG

AAGAATCTGGATGTGATCAAAAAAGAAAACTACTACGAAGTGAATAG

CAAGTGCTATGAGGAAGCTAAGAAGCTGAAGAAGATCAGCAACCAGG

CCGAGTTTATCGCCTCCTTCTACAACAACGATCTGATCAAGATCAACG

GCGAGCTGTATAGAGTGATCGGCGTGAACAACGACCTGCTGAACCGG

ATCGAAGTGAACATGATCGACATCACCTACCGCGAGTACCTGGAAAA

CATGAACGACAAGAGGCCCCCCAGGATCATTAAGACAATCGCCTCCA

AGACCCAGAGCATTAAGAAGTACAGCACAGACATTCTGGGCAACCTG TATGAAGTGAAATCTAAGAAGCACCCTCAGATCATCAAAAAGGGCTA A

[0099] Following is an exemplary nucleotide sequence (SEQ ID NO: 4) that enco Neiserria meningitidis Cas9 protein:

ATGGCCGCCTTCAAGCCCAACCCCATCAACTACATCCTGGGCCTGGAC

ATCGGCATCGCCAGCGTGGGCTGGGCCATGGTGGAGATCGACGAGGA

CG AGAACCCC ATCTGCCTGATCGACCT GGGTGTGCGCGT GTTCGAGCG

CGCTGAGGTGCCCAAGACTGGTGACAGTCTGGCTATGGCTCGCCGGCT

TGCTCGCTCTGTTCGGCGCCTTACTCGCCGGCGCGCTCACCGCCTTCTG

CGCGCTCGCCGCCTGCTGAAGCGCGAGGGTGTGCTGCAGGCTGCCGAC

TTCGACGAGAACGGCCTGATCAAGAGCCTGCCCAACACTCCTTGGCAG

CTGCGCGCTGCCGCTCTGGACCGCAAGCTGACTCCTCTGGAGTGGAGC

GCCGTGCTGCTGCACCTGATCAAGCACCGCGGCTACCTGAGCCAGCGC

AAGAACGAGGGCGAGACCGCCGACAAGGAGCTGGGTGCTCTGCTGAA

GGGCGTGGCCGACAACGCCCACGCCCTGCAGACTGGTGACTTCCGCAC

TCCTGCTGAGCTGGCCCTGAACAAGTTCGAGAAGGAGAGCGGCCACA

TCCGCAACCAGCGCGGCGACTACAGCCACACCTTCAGCCGCAAGGAC

CTGCAGGCCGAGCTGATCCTGCTGTTCGAGAAGCAGAAGGAGTTCGGC

AACCCCCACGTGAGCGGCGGCCTGAAGGAGGGCATCGAGACCCTGCT

GATGACCCAGCGCCCCGCCCTGAGCGGCGACGCCGTGCAGAAGATGC

TGGGCCACTGCACCTTCGAGCCAGCCGAGCCCAAGGCCGCCAAGAAC

ACCTACACCGCCGAGCGCTTCATCTGGCTGACCAAGCTGAACAACCTG

CGCATCCTGGAGCAGGGCAGCGAGCGCCCCCTGACCGACACCGAGCG

CGCCACCCTGATGGACGAGCCCTACCGCAAGAGCAAGCTGACCTACG

CCCAGGCCCGCAAGCTGCTGGGTCTGGAGGACACCGCCTTCTTCAAGG

GCCTGCGCTACGGCAAGGACAACGCCGAGGCCAGCACCCTGATGGAG

ATGAAGGCCTACCACGCCATCAGCCGCGCCCTGGAGAAGGAGGGCCT

GAAGGACAAGAAGAGTCCTCTGAACCTGAGCCCCGAGCTGCAGGACG

AGATCGGCACCGCCTTCAGCCTGTTCAAGACCGACGAGGACATCACCG

GCCGCCTGAAGGACCGCATCCAGCCCGAGATCCTGGAGGCCCTGCTG AAGCACATCAGCTTCGACAAGTTCGTGCAGATCAGCCTGAAGGCCCTG

CGCCGCATCGTGCCCCTGATGGAGCAGGGCAAGCGCTACGACGAGGC

CTGCGCCGAGATCTACGGCGACCACTACGGCAAGAAGAACACCGAGG

AGAAGATCTACCTGCCTCCTATCCCCGCCGACGAGATCCGCAACCCCG

TGGTGCTGCGCGCCCTGAGCCAGGCCCGCAAGGTGATCAACGGCGTG

GTGCGCCGCTACGGCAGCCCCGCCCGCATCCACATCGAGACCGCCCGC

GAGGTGGGCAAGAGCTTCAAGGACCGCAAGGAGATCGAGAAGCGCCA

GGAGGAGAACCGCAAGGACCGCGAGAAGGCCGCCGCCAAGTTCCGCG

AGTACTTCCCCAACTTCGTGGGCGAGCCCAAGAGCAAGGACATCCTGA

AGCTGCGCCTGTACGAGCAGCAGCACGGCAAGTGCCTGTACAGCGGC

AAGGAGATCAACCTGGGCCGCCTGAACGAGAAGGGCTACGTGGAGAT

CGACCACGCCCTGCCCTTCAGCCGCACCTGGGACGACAGCTTCAACAA

CAAGGTGCTGGTGCTGGGCAGCGAGAACCAGAACAAGGGCAACCAGA

CCCCCTACGAGTACTTCAACGGCAAGGACAACAGCCGCGAGTGGCAG

GAGTTCAAGGCCCGCGTGGAGACCAGCCGCTTCCCCCGCAGCAAGAA

GCAGCGCATCCTGCTGCAGAAGTTCGACGAGGACGGCTTCAAGGAGC

GCAACCT GAACGACACCCGCT ACGTGAACCGCTTCCTGT GCC AGTTCG

TGGCCGACCGCATGCGCCTGACCGGCAAGGGCAAGAAGCGCGTGTTC

GCCAGCAACGGCCAGATCACCAACCTGCTGCGCGGCTTCTGGGGCCTG

CGCAAGGTGCGCGCCGAGAACGACCGCCACCACGCCCTGGACGCCGT

GGTGGTGGCCTGCAGCACCGTGGCCATGCAGCAGAAGATCACCCGCTT

CGTGCGCTACAAGGAGATGAACGCCTTCGACGGTAAAACCATCGACA

AGGAGACCGGCGAGGTGCTGCACCAGAAGACCCACTTCCCCCAGCCC

TGGGAGTTCTTCGCCCAGGAGGTGATGATCCGCGTGTTCGGCAAGCCC

GACGGCAAGCCCGAGTTCGAGGAGGCCGACACCCCCGAGAAGCTGCG

CACCCTGCTGGCCGAGAAGCTGAGCAGCCGCCCTGAGGCCGTGCACG

AGTACGTGACTCCTCTGTTCGTGAGCCGCGCCCCCAACCGCAAGATGA

GCGGTCAGGGTCACATGGAGACCGTGAAGAGCGCCAAGCGCCTGGAC

GAGGGCGTGAGCGTGCTGCGCGTGCCCCTGACCCAGCTGAAGCTGAA

GGACCTGGAGAAGATGGTGAACCGCGAGCGCGAGCCCAAGCTGTACG

AGGCCCT GAAGGCCCGCCT GGAGGCCCACAAGGACGACCCCGCCAAG GCCTTCGCCGAGCCCTTCTACAAGTACGACAAGGCCGGCAACCGCACC

CAGCAGGTGAAGGCCGTGCGCGTGGAGCAGGTGCAGAAGACCGGCGT

GTGGGTGCGCAACCACAACGGCATCGCCGACAACGCCACCATGGTGC

GCGTGGACGTGTTCGAGAAGGGCGACAAGTACTACCTGGTGCCCATCT

ACAGCTGGCAGGTGGCCAAGGGCATCCTGCCCGACCGCGCCGTGGTG

CAGGGCAAGGACGAGGAGGACTGGCAGCTGATCGACGACAGCTTCAA

CTTCAAGTTCAGCCTGCACCCCAACGACCTGGTGGAGGTGATCACCAA

GAAGGCCCGCATGTTCGGCTACTTCGCCAGCTGCCACCGCGGCACCGG

CAACATCAACATCCGCATCCACGACCTGGACCACAAGATCGGCAAGA

ACGGCAT CCTGGAGGGCAT CGGCGTGAAGACCGCCCTGAGCTTCCAG

AAGTACCAGATCGACGAGCTGGGCAAGGAGATCCGCCCCTGCCGCCT

GAAG AAGCGCCCTCCTGT GCGCTGA

[00100] In some embodiments of the present invention, the recombinant Cas protein is derived from a Cas protein encoded by a nucleic acid molecule comprising a nucleotide sequence having at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identity with the nucleotide sequence of SEQ ID NO: 1.

[00101] In certain embodiments, the recombinant Cas protein is derived from a Cas protein encoded by a nucleic acid molecule comprising a nucleotide sequence having at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identity with the nucleotide sequence of SEQ ID NO: 2.

[00102] In some embodiments, the recombinant Cas protein is derived from a Cas protein encoded by a nucleic acid molecule comprising a nucleotide sequence having at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identity with the nucleotide sequence of SEQ ID NO: 3.

[00103] In certain embodiments, the recombinant Cas protein is derived from a Cas protein encoded by a nucleic acid molecule comprising a nucleotide sequence having at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identity with the nucleotide sequence of SEQ ID NO: 4. [00104] Following is the amino acid sequence of the Streptococcus pyogenes Cas9 protein (SEQ ID NO: 5) (see Esvelt, et al., "Orthogonal Cas9 proteins for RNA-guided gene regulation and editing," Nature Methods 10(1 1): 11 16-21 (2013), which is incorporated by reference herein in its entirety):

MDKKYSIGLDIGTNSVGWAVITDEYKVPSKKFKVLGNTDRHSIKKNLIGA

LLFD S GET AE ATRLKRT ARRRYTRRKNRICYLQEIF SNEM AKVDD SFFHR

LEESFLVEEDKKHERHPIFGNIVDEVAYHEKYPTIYHLRKKLVDSTDKADL

RLIYLALAHMIKFRGHFLIEGDLNPDNSDVDKLFIQLVQTYNQLFEENPIN

ASGVDAKAILSARLSKSRRLENLIAQLPGEKKNGLFGNLIALSLGLTPNFK

SNFDLAEDAKLQLSKDTYDDDLDNLLAQIGDQYADLFLAAKNLSDAILLS

DILRVNTEITKAPLSASMIKRYDEHHQDLTLLKALVRQQLPEKYKEIFFDQ

SKNGYAGYIDGGASQEEFYKFIKPILEKMDGTEELLVKLNREDLLRKQRT

FDNGSIPHQIHLGELHAILRRQEDFYPFLKDNREKIEKILTFRIPYYVGPLAR

GNSRFAWMTRKSEETrrPWNFEEWDKGASAQSFIERMTNFDKNLPNEK

VLPKHSLLYEYFTVYNELTKVKYVTEGMRKPAFLSGEQKKAIVDLLFKTN

RKVTVKQLKEDYFKKIECFDSVEISGVEDRFNASLGTYHDLLKIIKDKDFL

DNEENEDILEDIVLTLTLFEDREMIEERLKTYAHLFDDKVMKQLKRRRYT

GWGRLSRKLINGIRDKQSGKTILDFLKSDGFANRNFMQLIHDDSLTFKEDI

QKAQVSGQGDSLHEHIANLAGSPAIKKGILQTVKWDELVKVMGRHKPE

NIVIEMARENQTTQKGQKNSRERMKRIEEGIKELGSQILKEHPVENTQLQN

EKLYLYYLQNGRDMYVDQELDINRLSDYDVDHIVPQSFLKDDSIDNKVL

TRSDKNRGKSDNVPSEEWKKMKNYWRQLLNAKLrrQRKFDNLTKAER

GGLSELDKAGFIKRQLVETRQrTKHVAQILDSRMNTKYDENDKLIREVKVI

TLKSKLVSDFRKDFQFYKVREINNYHHAHDAYLNAWGTALIKKYPKLE

SEFWGDYKWDVRKMIAKSEQEIGKATAKYFFYSNIMNFFKTEITLANG

EIRKRPLIETNGETGEIVWDKGRDFATVRKVLSMPQWIVKKTEVQTGGF

SKESILPKRNSDKLIARKKDWDPKKYGGFDSPTVAYSVLWAKVEKGKS

KKLKSVKELLGITIMERSSFEKNPIDFLEAKGYKEVKKDLIIKLPKYSLFEL

ENGRKRMLASAGELQKGNELALPSKYVNFLYLASHYEKLKGSPEDNEQK

QLFVEQHKHYLDEIIEQISEFSKRVILADANLDKVLSAYNKHRDKPIREQA ENIIHLFTLTNLGAPAAFKYFDTTIDRKRYTSTKEVLDATLIHQSITGLYET

RIDLSQLGGD

[00105] Following is the amino acid sequence of the Streptococcus thermophilus LMD-9 Cas9 protein (SEQ ID NO: 6) (see Esvelt, et al., "Orthogonal Cas9 proteins for RNA-guided gene regulation and editing," Nature Methods 10(1 1):1 1 16-21 (2013), which is incorporated by reference herein in its entirety):

MSDLVLGLDIGIGSVGVGILNKVTGEIIHKNSRIFPAAQAENNLVRRTNRQ

GRRLARRKKHRRVRLNRLFEESGLrTDFTKISINLNPYQLRVKGLTDELSN

EELFIALKNMVKHRGISYLDDASDDGNSSVGDYAQIVKENSKQLETKTPG

QIQLERYQTYGQLRGDFTVEKDGKKHRLINVFPTSAYRSEALRILQTQQEF

NPQITDEFINRYLEILTGKRKYYHGPGNEKSRTDYGRYRTSGETLDNIFGIL

IGKCTFYPDEFRAAKASYTAQEFNLLNDLNNLTVPTETKKLSKEQKNQIIN

Y VKJSiEKAMGP AKLFKYI AKLLS CD VADIKGYRIDKS GKAE IHTFE AYRK

MKTLETLDIEQMDRETLDKLAYVLTLNTEREGIQEALEHEFADGSFSQKQ

VDELVQFRKANSSIFGKGWHNFSVKLMMELIPELYETSEEQMTILTRLGK

QKTTSSSNKTKYIDEKLLTEEIYNPWAKSVRQAIKIVNAAIKEYGDFDNI

VIEMARETNEDDEKKAIQKIQKANKDEKDAAMLKAANQYNGKAELPHS

VFHGHKQLATKIRLWHQQGERCLYTGKTISIHDLINNSNQFEVDHILPLSIT

FDDSLANKVLWATANQEKGQRTPYQALDSMDDAWSFRELKAFVRESK

TLSNKKKEYLLTEEDISKFDVRKKFIERNLVDTRYASRWLNALQEHFRA

HKIDTKVSWRGQFTSQLRRHWGIEKTRDTYHHHAVDALIIAASSQLNLW

KKQKNTLVSYSEDQLLDIETGELISDDEYKESVFKAPYQHFVDTLKSKEFE

DSILFSYQVDSKFNRKISDATIYATRQAKVGKDKADETYVLGKIKDIYTQD

GYDAFMKIYKKDKSKFLMYRHDPQTFEKVIEPILENYPNKQINDKGKEVP

CNPFLKYKEEHGYIRKYSKKGNGPEIKSLKYYDSKLGNHIDITPKDSNNK

WLQSVSPWRADWFNKTTGKYEILGLKYADLQFDKGTGTYKISQEKYN

DIKKKEGVDSDSEFKFTLYKNDLLLVKDTETKEQQLFRFLSRTMPKQKHY

VELKPYDKQKFEGGEALIKVLGNVANSGQCKKGLGKSNISIYKVRTDVLG

NQHIIKNEGDKPKLDF [00106] Following is the amino acid sequence of the Staphylococcus aureus subsp. aureus Cas9 protein (SEQ ID NO: 7) (see Ran, et al, "In vivo genome editing using Staphylococcus aureus Cas9," Nature 520(7546): 186-91 (2015), which is incorporated by reference herein in its entirety):

MKRNYILGLDIGITSVGYGIIDYETRDVIDAGVRLFKEANVENNEGRRSKRG

ARRLKRRRRHRIQRVKKLLFDYNLLTDHSELSGINPYEARVKGLSQKLSEE

EFSAALLHLAKRRGVHNVNEVEEDTGNELSTKEQISRNSKALEEKYVAELQ

LERLKKDGEVRGSINRFKTSDYVKEAKQLLKVQKAYHQLDQSFIDTYIDLL

ETRRTYYEGPGEGSPFGWKDIKEWYEMLMGHCTYFPEELRSVKYAYNAD

LYNALNDLNNLVrTRDENEKLEYYEKFQIIENVFKQKKKPTLKQIAKEILVN

EEDIKGYRVTSTGKPEFTNLKWHDIKDrrARKEIIENAELLDQIAKILTIYQS

SEDIQEELTNLNSELTQEEIEQISNLKGYTGTHNLSLKAINLILDELWHTNDN

QIAIFNRLKLVPKKVDLSQQKEIPTTLVDDFILSPWKRSFIQSIKVINAIIKKY

GLPNDIIIELAREKNSKDAQKMINEMQKRNRQTNERIEEIIRTTGKENAKYLI

EKIKLHDMQEGKCLYSLEAIPLEDLLNNPFNYEVDHIIPRSVSFDNSFNNKV

LVKQEENSKKGNRTPFQYLSSSDSKISYETFKKHILNLAKGKGRISKTKKEY

LLEERDINRFSVQKDFINRNLVDTRYATRGLMNLLRSYFRVNNLDVKVKSI

NGGFTSFLRRKWKFKKERNKGYKHHAEDALIIANADFIFKEWKKLDKAKK

VMENQMFEEKQAESMPEIETEQEYKEIFrrPHQIKHIKDFKDYKYSHRVDK

KPNRELINDTLYSTRKDDKGNTLIVNNLNGLYDKDNDKLKKLINKSPEKLL

MYHHDPQTYQKLKLIMEQYGDEKNPLYKYYEETGNYLTKYSKKDNGPVI

KKIKYYGNKLNAHLDITDDYPNSRNKWKLSLKPYRFDWLDNGWKFVT

VKNLDVIKKENYYEVNSKCYEEAKKLKKISNQAEFIASFYNNDLIKINGEL

YRVIG /NNDLLNRIEWMIDITYREYLENMNDKRPPRIIKTIASKTQSIKKYS

TDILGNLYEVKSKKHPQIIKKG

[00107] Following is the amino acid sequence of the Neisseria meningitidis Cas9 protein (SEQ ID NO: 8) (see Esvelt, et al., "Orthogonal Cas9 proteins for RNA-guided gene regulation and editing," Nature Methods 10(1 1): 11 16-21 (2013), which is incorporated by reference herein in its entirety): MAAFKPNPINYILGLDIGIASVGWAMVEIDEDENPICLIDLGVRVFERAEV

PKTGDSLAMARRLARSVRRLTRRRAHRLLRARRLLKREGVLQAADFDEN

GLIKSLPNTPWQLRAAALDRKLTPLEWSAVLLHLIKHRGYLSQRK EGET

ADKELGALLKGVADNAHALQTGDFRTPAELALNKFEKESGHIRNQRGDY

SHTFSRKDLQAELILLFEKQKEFGNPHVSGGLKEGIETLLMTQRPALSGDA

VQKMLGHCTFEPAEPKAAKNTYTAERFIWLTKLNNLRILEQGSERPLTDT

ERATLMDEPYRKSKLTYAQARKLLGLEDTAFFKGLRYGKDNAEASTLME

MKAYHAISRALEKEGLKDKKSPLNLSPELQDEIGTAFSLFKTDEDITGRLK

DRIQPEILEALLKHISFDKFVQISLKALRRIVPLMEQGKRYDEACAEIYGDH

YGKK TEEKIYLPPIPADEIR PWLRALSQARKVINGWRRYGSPARIHIE

TAREVGKSFKDRKEIEKRQEENRKDREKAAAKFREYFPNFVGEPKSKDIL

KLRLYEQQHGKCLYSGKEINLGRLNEKGYVEIDHALPFSRTWDDSFNNK

VLVLGSENQNKGNQTPYEYFNGKDNSREWQEFKARVETSRFPRSKKQRI

LLQKFDEDGFKERNLNDTRYV RFLCQFVADRMRLTGKGKKRVFASNG

QITNLLRGFWGLRKVRAENDRHHALDAVVVACSTVAMQQKITRFVRYK

EMNAFDGKTIDKETGEVLHQKTHFPQPWEFFAQEVMIRVFGKPDGKPEFE

E ADTPEKLRTLL AEKLS SRPE AVHE Y VTPLF VSRAPNRKM S GQ GHMET V

KSAKRLDEGVSVLRVPLTQLKLKDLEKMV REREPKLYEALKARLEAHK

DDPAKAFAEPFYKYDKAGNRTQQVKAVRVEQVQKTGVWVRNHNGIAD

NATMVRVDVFEKGDKYYLVPIYSWQVAKGILPDRAWQGKDEEDWQLI

DDSFNFKFSLHPNDLVEVITKKARMFGYFASCHRGTGNINIRIHDLDHKIG

K GILEGIGVKTALSFQKYQIDELGKEIRPCRLKKRPPVR

[00108] In some embodiments of the present invention, the recombinant Cas protein is derived from a Cas protein comprising an amino acid sequence having at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identity with the amino acid sequence of SEQ ID NO: 5.

[00109] In certain embodiments, the recombinant Cas protein is derived from a Cas protein comprising an amino acid sequence having at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identity with the amino acid sequence of SEQ ID NO: 6. [00110] In some embodiments, the recombinant Cas protein is derived from a Cas protein comprising an amino acid sequence having at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identity with the amino acid sequence of SEQ ID NO: 7.

[00111] In certain embodiments, the recombinant Cas protein is derived from a Cas protein comprising an amino acid sequence having at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identity with the amino acid sequence of SEQ ID NO: 8.

[00112] The present invention is also directed to recombinant Cas proteins comprising one or more amino acid substitutions in one or more residues corresponding to one or more MHC Class I and/or MHC Class II binding sites in a wild-type Cas protein. In certain embodiments, the recombinant Cas protein is derived from the wild-type protein containing the corresponding MHC Class I and/or Class II binding sites. In some embodiments, the recombinant Cas protein has reduced immunogenicity compared to the wild-type Cas protein from which it is derived. In some embodiments, the recombinant Cas protein is isolated and in other embodiments it is located within a cell.

[00113] In some embodiments, the recombinant Cas protein is derived from a Cas9-like protein, such as Cpfl , disclosed in Zetsche et al., "Cpfl is a Single RNA-Guided Endonuclease of a Class 2 CRISPR-Cas System," Cell 163:759-771 (2015), incorporated herein by reference in its entirety.

[00114] In some embodiments, the disclosure is directed to a recombinant CRISPR-associated (Cas) protein, wherein said protein comprises one or more amino acid variation compared to a while-type Cas protein, wherein the one or more amino acid variation is at one or more positions corresponding to amino acid residues in the polypeptide of SEQ ID NO: 5, wherein the variation is at a position selected from the group consisting of (i) residues 100 to 120, (ii) residues 250 to 280, (iii) residues 690 to 710, (iv) residues 840 to 860, and (v) residues 1270 to 1295. In some embodiments, the variation is at a position selected from the group consisting of (i) residues 102 to 120, (ii) residues 253 to 277, (iii) residues 692 to 709, (iv) residues 692 to 709, and (v) residues 1276 to 1292. [00115] In some embodiments, the variation is at a position selected from residues 102 to 120. In some embodiments, the variation is at a position selected from residues 253 to 277. In some embodiments, the variation is at a position selected from residues 692 to 709. In some embodiments, the variation is at a position selected from residues 692 to 709. In some embodiments, the variation is at a position selected from residues 1276 to 1292.

[00116] In some embodiments, the recombinant Cas protein comprises two or more amino acid variations. In some embodiments, the recombinant Cas protein comprises three or more amino acid variations. In some embodiments, the amino acid variation is determined using in silico epitope mapping.

[00117] The term "variation," when referring to a variation at a given polypeptide residue position, refers to any modifications at those residues. For example, the term variation can refer to substituting (i.e., replacing) an amino acid (i.e., residue) at a given position with a different amino acid. In some embodiments, the amino acid can be substituted with a different amino acid having a different property. Various predictive algorithms for determining the immunogenic properties of a polypeptide are known in the art, and can include (1) the ratio between the frequencies of aspargine, glutamine, leucine, lysine, methionine, serine, threonine, and valine residues in the stretch and the remaining antigen sequence; (2) Grantham polarity scale (Grantham et al., Science. 185: 862-864 (1974)); (3) Karplus and Schulz flexibility scale (Karplus et al., Naturwissenschaften 72: 212-213 (1985)); (4) Kolaskar and Tongaonkar antigenicity scale (Kolaskar et al, FEBS Lett. 276: 172-174 (1990)); (5) Parker hydrophilicity scale (Parker et al., Biochemistry 25: 5425-5432 (1986)); (6) Ponnuswamy polarity scale (Ponnuswamy et al., Biochim. Biophys. Acta. 623: 301-326 (1980)); and, (7) Atchley et al. factorl scale (Atchley et al, Proc. Natl. Acad. Sci. U.S.A. 102: 6395-6400 (2005)). In some embodiments, EPIBASE® can be used to determine what amino acid can be used to reduce immunogenicity of the polypeptide.

[00118] For example, in some embodiments, one or more of aspargine, glutamine, leucine, lysine, methionine, serine, threonine, and valine residues can be replaced with a different amino acid. In some embodiments, the amino acid variation comprises replacing a polar amino acid with a nonpolar amino acid. In some embodiments, the amino acid variation comprises replacing a hydrophilic amino acid with a hydrophobic amino acid. In some embodiments, and amino acid with a large side chain can be replaced with an amino acid with a smaller side chain.

[00119] In some embodiments, the term variation can refer to deleting one or more amino acids residues into the Cas protein. For example, in some embodiments, 2, 3, 4, 5, 6, 7, 8, 9, 10, 12, 14, 16, 18 or 20 amino acids can be deleted in one epitope. In some embodiments, one or more amino acids can be deleted from two or more epitopes on the same protein. In some embodiments, the deletions are not contiguous on the polypeptide, e.g., one deletion can occur on one region of the polypeptide, and a separate deletion can occur on a separate part of the polypeptide.

[00120] In some embodiments, the term variation can refer to inserting one or more amino acid residues into the Cas protein. For example, in some embodiments, 2, 3, 4, 5, 6, 7, 8, 9, 10, 12, 14, 16, 18 or 20 amino acids can be inserted in one epitope. In some embodiments, one or more amino acids can be inserted into two or more epitopes on the same protein. In some embodiments, the insertions are not contiguous on the polypeptide, e.g., one insertion can occur on one region of the polypeptide, and a separate insertion can occur on a separate part of the polypeptide.

[00121] In some embodiments, the Cas protein can have various types of variations. For example, in some embodiments, the Cas protein can have one or more amino acid substitutions, one or more amino acid deletions, and one or more amino acid insertions.

[00122] In some embodiments, the wild-type Cas protein has a sequence 90% identical to SEQ ID NO: 5. In some embodiments, the wild-type Cas protein has a sequence 90% identical to SEQ ID NO: 6. In some embodiments, the wild-type Cas protein has a sequence 90% identical to SEQ ID NO: 7. In some embodiments, the wild-type Cas protein has a sequence 90% identical to SEQ ID NO: 8. In some embodiments, the wild-type Cas protein has a sequence 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% identical to SEQ ID NO: 5. In some embodiments, the wild-type Cas protein has a sequence 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% identical to SEQ ID NO: 6. In some embodiments, the wild-type Cas protein has a sequence 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% identical to SEQ ID NO: 7. In some embodiments, the wild-type Cas protein has a sequence 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% identical to SEQ ID NO: 8.

[00123] In some embodiments, the wild-type Cas protein is derived from a prokaryote. In some embodiments, the prokaryote is a bacterium. In some embodiments, the bacterium is from the genera Streptococcus, Staphylococcus, or Neisseria. In some embodiments, the bacterium is Streptococcus pyogenes, Streptococcus thermophilus, Staphylococcus aureus, or Neisseria meningtidis.

[00124] In some embodiments, the recombinant Cas protein is derived from Cas3, Cas9, or CaslO. In some embodiments, the recombinant Cas protein is derived from Cas9.

Polynucleotides, Expression Vectors, and Host Cells

[00125] In some embodiments, the polynucleotides or expression vectors encoding Cas proteins (or variants thereof) are placed in a host cell, e.g, a prokaryote or eukaryote cell. The devices, facilities and methods described herein are suitable for culturing any desired cell line including prokaryotic and/or eukaryotic cell lines. Further, in some embodiments, the devices, facilities and methods are suitable for culturing suspension cells or anchorage-dependent (adherent) cells and are suitable for production operations configured for production of pharmaceutical and biopharmaceutical products— such as polypeptide products, nucleic acid products (for example DNA or RNA).

[00126] In embodiments, the host cells express or produce a product, such as a Cas protein or variant thereof, or expression vectors encoding the Cas protein or variant thereof.

[00127] In some embodiments, devices, facilities and methods allow for the production of host eukaryotic cells comprising the described expression vectors, e.g., mammalian cells or lower eukaryotic cells such as for example yeast cells or filamentous fungi cells, or prokaryotic cells such as Gram-positive or Gram-negative cells, synthesised by the host cells in a large-scale manner. Unless stated otherwise herein, the devices, facilities, and methods can include any desired volume or production capacity including but not limited to bench-scale, pilot-scale, and full production scale capacities. [00128] Moreover and unless stated otherwise herein, the host cells can be generated using devices, facilities, and methods including any suitable reactor(s) including but not limited to stirred tank, airlift, fiber, micro fiber, hollow fiber, ceramic matrix, fluidized bed, fixed bed, and/or spouted bed bioreactors. As used herein, "reactor" can include a fermentor or fermentation unit, or any other reaction vessel and the term "reactor" is used interchangeably with "fermentor." For example, in some aspects, an example bioreactor unit can perform one or more, or all, of the following: feeding of nutrients and/or carbon sources, injection of suitable gas (e.g., oxygen), inlet and outlet flow of fermentation or cell culture medium, separation of gas and liquid phases, maintenance of temperature, maintenance of oxygen and C02 levels, maintenance of pH level, agitation (e.g., stirring), and/or cleaning/sterilizing. Example reactor units, such as a fermentation unit, may contain multiple reactors within the unit, for example the unit can have 1 , 2, 3, 4, 5, 10, 15, 20, 25, 30, 35, 40, 45, 50, 60, 70, 80, 90, or 100, or more bioreactors in each unit and/or a facility may contain multiple units having a single or multiple reactors within the facility. In various embodiments, the bioreactor can be suitable for batch, semi fed-batch, fed- batch, perfusion, and/or a continuous fermentation processes. Any suitable reactor diameter can be used. In embodiments, the bioreactor can have a volume between about 100 mL and about 50,000 L. Non-limiting examples include a volume of 100 mL, 250 mL, 500 mL, 750 mL, 1 liter, 2 liters, 3 liters, 4 liters, 5 liters, 6 liters, 7 liters, 8 liters, 9 liters, 10 liters, 15 liters, 20 liters, 25 liters, 30 liters, 40 liters, 50 liters, 60 liters, 70 liters, 80 liters, 90 liters, 100 liters, 150 liters, 200 liters, 250 liters, 300 liters, 350 liters, 400 liters, 450 liters, 500 liters, 550 liters, 600 liters, 650 liters, 700 liters, 750 liters, 800 liters, 850 liters, 900 liters, 950 liters, 1000 liters, 1500 liters, 2000 liters, 2500 liters, 3000 liters, 3500 liters, 4000 liters, 4500 liters, 5000 liters, 6000 liters, 7000 liters, 8000 liters, 9000 liters, 10,000 liters, 15,000 liters, 20,000 liters, and/or 50,000 liters. Additionally, suitable reactors can be multi-use, single-use, disposable, or non-disposable and can be formed of any suitable material including metal alloys such as stainless steel (e.g., 316L or any other suitable stainless steel) and Inconel, plastics, and/or glass.

[00129] In some embodiments and unless stated otherwise herein, the methods described herein can also include any suitable unit operation and/or equipment not otherwise mentioned, such as operations and/or equipment for separation, purification, and isolation of Cas proteins or variants thereof, or expression vectors expressing the Cas proteins or variants thereof. Any suitable facility and environment can be used, such as traditional stick-built facilities, modular, mobile and temporary facilities, or any other suitable construction, facility, and/or layout. For example, in some embodiments modular clean-rooms can be used. Additionally and unless otherwise stated, the devices, systems, and methods described herein can be housed and/or performed in a single location or facility or alternatively be housed and/or performed at separate or multiple locations and/or facilities.

[00130] By way of non-limiting examples and without limitation, U.S. Publication Nos. 2013/0280797; 2012/0077429; 2011/0280797; 2009/0305626; and U.S. Patent Nos. 8,298,054; 7,629,167; and 5,656,491 , which are hereby incorporated by reference in their entirety, describe example facilities, equipment, and/or systems that may be suitable.

[00131] In some embodiments, the host cells are eukaryotic cells, e.g., mammalian cells. The mammalian cells can be for example human or rodent or bovine cell lines or cell strains. Examples of such cells, cell lines or cell strains are e.g. mouse myeloma (NSO)-cell lines, Chinese hamster ovary (CHO)-cell lines, HT1080, H9, HepG2, MCF7, MDBK Jurkat, NHBT3, PC12, BHK (baby hamster kidney cell), VERO, SP2/0, YB2/0, Y0, C127, L cell, COS, e.g., COS1 and COS7, QCl -3,HEK-293, VERO, PER.C6, HeLA, EB1, EB2, EB3, oncolytic or hybridoma-cell lines. Preferably the mammalian cells are CHO-cell lines. In one embodiment, the cell is a CHO cell. In one embodiment, the cell is a CHO-K1 cell, a CHO-K1 SV cell, a DG44 CHO cell, a DUXBl l CHO cell, a CHOS, a CHO GS knock-out cell, a CHO FUT8 GS knock-out cell, a CHOZN, or a CHO-derived cell. The CHO GS knock-out cell (e.g., GSKO cell) is, for example, a CHO-K1 SV GS knockout cell. The CHO FUT8 knockout cell is, for example, the Potelligent® CHOK1 SV (Lonza Biologies, Inc.). Eukaryotic cells can also be avian cells, cell lines or cell strains, such as for example, EBx® cells, EB14, EB24, EB26, EB66, or EBvl3.

[00132] In some embodiments, the host cells are stem cells. The stem cells can be, for example, pluripotent stem cells, including embryonic stem cells (ESCs), adult stem cells, induced pluripotent stem cells (iPSCs), tissue specific stem cells (e.g., hematopoietic stem cells) and mesenchymal stem cells (MSCs). [00133] In some embodiments, the host cell is a differentiated form of any of the cells described herein. In one embodiment, the host cell is a cell derived from any primary cell in culture.

[00134] In some embodiments, the host cell is a hepatocyte such as a human hepatocyte, animal hepatocyte, or a non-parenchymal cell. For example, the host cell can be a plateable metabolism qualified human hepatocyte, a plateable induction qualified human hepatocyte, plateable Qualyst Transporter Certified™ human hepatocyte, suspension qualified human hepatocyte (including 10-donor and 20-donor pooled hepatocytes), human hepatic kupffer cells, human hepatic stellate cells, dog hepatocytes (including single and pooled Beagle hepatocytes), mouse hepatocytes (including CD-I and C57BI/6 hepatocytes), rat hepatocytes (including Sprague-Dawley, Wistar Han, and Wistar hepatocytes), monkey hepatocytes (including Cynomolgus or Rhesus monkey hepatocytes), cat hepatocytes (including Domestic Shorthair hepatocytes), and rabbit hepatocytes (including New Zealand White hepatocytes). Example hepatocytes are commercially available from Triangle Research Labs, LLC, 6 Davis Drive Research Triangle Park, North Carolina, USA 27709.

[00135] In one embodiment, the host cell is a lower eukaryotic cell such as e.g. a yeast cell (e.g., Pichia genus (e.g. Pichia pastoris, Pichia methanolica, Pichia kluyveri, and Pichia angusta), Komagataella genus (e.g. Komagataella pastoris, Komagataella pseudopastoris or Komagataella phaffii), Saccharomyces genus (e.g. Saccharomyces cerevisae, cerevisiae, Saccharomyces kluyveri, Saccharomyces uvarum), Kluyveromyces genus (e.g. Kluyveromyces lactis, Kluyveromyces marxianus), the Candida genus (e.g. Candida utilis, Candida cacaoi, Candida boidinii,), the Geotrichum genus (e.g. Geotrichum fermentans), Hansenula polymorpha, Yarrowia lipolytica, or Schizosaccharomyces pombe. Preferred is the species Pichia pastoris. Examples for Pichia pastoris strains are X33, GS115, KM71, KM71H; and CBS7435.

[00136] In some embodiments, the host cell is a fungal cell (e.g. Aspergillus (such as A. niger, A. fumigatus, A. orzyae, A. nidula), Acremonium (such as A. thermophilum), Chaetomium (such as C. thermophilum), Chrysosporium (such as C. thermophile), Cordyceps (such as C. militaris), Corynascus, Ctenomyces, Fusarium (such as F. oxysporum), Glomerella (such as G. graminicola), Hypocrea (such as H. jecorina), Magnaporthe (such as M. orzyae), Myceliophthora (such as M. thermophile), Nectria (such as N. heamatococca), Neurospora (such as N. crassa), Penicillium, Sporotrichum (such as S. thermophile), Thielavia (such as T. terrestris, T. heterothallica), Trichoderma (such as T. reesei), or Verticillium (such as V. dahlia)).

[00137] In some embodiments, the host cell is an insect cell (e.g., Sf9, Mimic™ Sf9, S£21 , High Five™ (BT1-TN-5B1-4), or BT1 -Ea88 cells), an algae cell (e.g., of the genus Amphora, Bacillariophyceae, Dunaliella, Chlorella, Chlamydomonas, Cyanophyta (cyanobacteria), Nannochloropsis, Spirulina,or Ochromonas), or a plant cell (e.g., cells from monocotyledonous plants (e.g., maize, rice, wheat, or Setaria), or from a dicotyledonous plants (e.g., cassava, potato, soybean, tomato, tobacco, alfalfa, Physcomitrella patens or Arabidopsis).

[00138] In some embodiments, the host cell is a bacterial or prokaryotic cell.

[00139] In some embodiments, the host cell is a Gram-positive cells such as Bacillus, Streptomyces Streptococcus, Staphylococcus or Lactobacillus. Bacillus that can be used is, e.g. the B.subtilis, B.amyloliquefaciens, B.licheniformis, B.natto, or B.megaterium. In embodiments, the cell is B.subtilis, such as B.subtilis 3NA and B.subtilis 168. Bacillus is obtainable from, e.g., the Bacillus Genetic Stock Center , Biological Sciences 556, 484 West 12 m Avenue, Columbus OH 43210-1214.

[00140] In some embodiments, the host cell is a Gram-negative cell, such as Salmonella spp. or Escherichia coli, such as e.g., TGI , TG2, W3110, DH1 , DHB4, DH5a, HMS 174, HMS174 (DE3), NM533, C600, HB101, JM109, MC4100, XLl-Blue and Origami, as well as those derived from E.coli B-strains, such as for example BL-21 or BL21 (DE3), all of which are commercially available.

[00141] Suitable host cells are commercially available, for example, from culture collections such as the DSMZ (Deutsche Sammlung von Mikroorganismen and Zellkulturen GmbH, Braunschweig, Germany) or the American Type Culture Collection (ATCC).

Immunogenicity and T-cell Responses in Humans

[00142] In humans, a cytotoxic T lymphocyte (CTL) response is initiated when a T-cell's unique T-cell receptor (TCR) recognizes a peptide bound to the HLA Class I molecules, displayed on the surface of most nucleated cells. Only those peptides with sufficient affinity for the HLA Class I receptor can be presented on the cell's surface, and potentially trigger a CTL response. Knowing which peptides within a protein have a strong affinity for HLA Class I receptors is therefore an important step towards determining the immunogenic regions of the protein.

[00143] The HLA Class I molecule is made of up two polypeptide chains: the a chain and β-2 microglobulin, each of which is derived from a different gene. Individuals express multiple genes encoding the a-chain: A, B, C, E, F, G, K and L. The dominant a-chain genes, A-C, are highly polymorphic. Different HLA Class I molecules are described as HLA allotypes.

[00144] For a complete analysis of the immunogenicity of a protein or peptide, one should therefore test the affinity/immunogenicity of each peptide for each possible HLA Class I allotype. Fortunately, however, some allotypes have a higher prevalence in a given population than others. As a result, the analysis for any population can be done in most cases by focusing on a limited number of allotypes. In general, an individual's T-cell population has been selected not to contain cells that recognize "self-peptides" (peptides derived from endogenous proteins) presented on HLA Class I receptors. Therefore, peptides from an exogenous protein that correspond to (known) self-peptides will normally not induce a CTL response, even if they have a high affinity for HLA Class I receptors. It is not always clear which endogenous proteins are presented and as such give rise to self-peptides.

[00145] A T (T -helper) response is sparked when a T Cell's unique T-Cell receptor (TCR) recognizes a peptide bound to the HLA Class II molecules displayed on antigen presenting cells (APCs). These peptides are generated from proteins internalized by an antigen presenting cell, and then cleaved through its endosomal cleavage pathway. Only those peptides with sufficient affinity for the HLA Class II receptor can be presented on the cells surface and potentially trigger a T h response. Knowing which peptides within a protein have a strong affinity for HLA Class II receptors is therefore an important step towards determining the protein's immunogenic regions.

[00146] The picture, however, can be complicated by the fact that there are several HLA Class II genes, almost all of which are highly polymorphic. Each HLA Class II molecule consists of an a and β chain, each derived from a different gene, which further increases the number of possible HLA Class II molecules. Specifically, every human individual expresses the following genes: DRA/DRB, DQA DQB and DPA DPB. Of these, only DRA is non-polymorphic. In addition, a "second" DRB gene (DRB3, DRB4 or DRB5) may also be present, whose product also associates with the DRA chain.

[00147] For a complete analysis of the immunogenicity of a protein or peptide, one should therefore test the affinity/immunogenicity of each peptide for each possible HLA Class II allotype. Fortunately however, some allotypes have a higher prevalence in a given population than others. As a result, the analysis for any population can be performed in most cases by focusing on a limited number of allotypes.

[00148] Furthermore, the expression levels of receptors of the DQ and DP gene families are known to be significantly lower than those of DRB, making the latter the primary focus of immunogenicity profiling. See Laupeze et al., "Differential expression of major histocompatibility complex class la, lb, and II molecules on monocytes-derived dendritic and macrophagic cells," Hum. Immunol. 60(7):591-7 (1999); Gansbacher and Zier, "Regulation of HLA-DR, DP, and DQ expression in activated T cells," Cell Immunol. 1 17(l):22-34 (1988); Berdoz, et al., "Constitutive and induced expression of the individual HLA-DR beta and alpha chain loci in different cell types," J. Immunol. 139(4): 1336-41 (1987); and Stunz, et al., "HLA- DRB1 and -DRB4 genes are differentially regulated at the transcriptional level," J. Immunol. 143(9):3081-6 (1989), each of which is incorporated by reference herein in its entirety.

[00149] Differences of expression exist between presenting cells, e.g., dendritic cells vs. macrophages. See Laupeze, et al. Also, differences in HLA expression levels have been correlated with the magnitude of the T-cell response. Vader, et al., "The HLA-DQ2 gene dose effect in celiac disease is directly related to the magnitude and breadth of gluten-specific T cell responses," PNAS 100(21): 12390-5 (2003), which is incorporated by reference herein in its entirety. Cases of DQ- and DP-restricted T-cell response have been documented (see e.g., Castelli, et al., "HLA-DP4, the most frequent HLA II molecule, defines new supertype of peptide -binding specificity," J. Immunol. 169(12):6928-34 (2002), which is incorporated by reference herein in its entirety), thus it is recommended that strong DQ and DP binders are not ignored when analyzing the immunogenicity of a protein. [00150] In general, an individual's T-cell population has been selected not to contain cells that recognize "self-peptides" presented on HLA Class II receptors. These are peptides derived from endogenous proteins after internalization by an antigen presenting cell. Therefore, peptides from an exogenous protein which correspond to (known) self-peptides will normally not induce a T response, even if they have a high affinity for HLA Class II receptors. It is not clear which endogenous proteins are internalized and as such give rise to self-peptides. See Kirschmann, et al., "Naturally processed peptides from rheumatoid arthritis associated and non-associated HLA- DR alleles," J. Immunol. 155(12):5655-62 (1995) and Verreck, et al., "Natural peptides isolated from Gly86/Val86-containing variants of HLA-DR1 , -DR11 , -DR13, and -DR52," Immunogenetics 43(6):392-7 (1996), each of which is incorporated by reference herein in its entirety.

[00151] In addition to epitopes bound to HLA molecules, as described above, it is also important to consider presence of epitopes bound by surface IgG molecules (B cell receptors) found on B cells. B cell receptors are responsible for selective uptake of antigens and presentation on B cells via HLA Class II, for subsequent interaction with T cell receptors found on T helper cells. In contrast to peptides bound by HLA molecules which have been processed such that they are considered only in linear terms, B cell epitopes can also be conformational, as they are found in the native context of the protein.

[00152] The conformational aspect of B cell epitopes makes in silico prediction problematic relative to T cell epitopes (HLA binding). In vitro assays, however, can be used to identify B cell activation in response to either full length proteins (conformational and linear epitopes) or derived peptides (linear epitopes) covering the relevant regions.

Methods for Reducing Immuno genicity

[00153] Certain embodiments of the present invention are directed to methods for making a recombinant Cas protein having reduced immunogenicity, i.e., with a reduced ability to elicit a CTL or T response in a host, such as a human. In some embodiments, the method comprises introducing one or more amino acid residues corresponding to one or more histocompatibility (MHC) Class I and/or Class II binding sites of the Cas protein to form a recombinant Cas protein, wherein the recombinant Cas protein has reduced immunogenicity compared to the Cas protein from which it was derived. In some embodiments of the invention, the position of the amino acid substitution(s) is determined through epitope mapping. In certain embodiments, the position of the amino acid substitution(s) is determined using in silico epitope mapping and in some embodiments the position of the amino acid substitution(s) is determined using cell-based epitope mapping.

Methods for Epitope Mapping

[00154] In certain embodiments of the invention, the position of one or more amino acid substitutions for reducing the immunogenicity of a Cas protein is selected through in silico epitope mapping. Methods for in silico prediction of immunogenicity that can be used in the present invention are available from academic and commercial sources. Examples include those provided by Lonza (EPIBASE ® ; see, e.g., U.S. Patent No. 7,702,465 and EP1516275, each of which is incorporated by reference herein in its entirety) and the Centre of Biological Sequence Analysis at the Technical University of Denmark (NetMHC; cbs.dtu.dk/services/NetMHC/). Such tools, on occasion, can be over-predictive of immunogenic epitopes and are therefore frequently combined with in vitro or ex vivo assays, such as those provided herein, in order to further refine results. A description of such in silico methodologies in combination with cell- based assays, as well as its utilization to reduce the impact of immunogenicity risks in therapeutics, has been reviewed elsewhere (see e.g., Zurdo 2013 and Zurdo et al. 2015, each of which is incorporated by reference herein in its entirety).

[00155] In some embodiments of the present invention, epitope mapping of Cas proteins is performed using cell-based methods. For example, in certain embodiments, antigen presenting cells (APCs) are incubated in the presence of Cas proteins. Subsequently, MHC Class I and Class II proteins {e.g., HLA Class I and Class II proteins) are isolated from the APCs and the Cas9-derived peptides bound the MHC Class I and/or Class II proteins are identified. In certain embodiments, the MHC Class I and Class II proteins are isolated by immunoprecipitation. In some embodiments, the Cas9 derived peptides bound the MHC Class I and Class II proteins are identified by mass spectrometry (MS).

[00156] In other embodiments of the invention, short synthetic peptides representing putative T-cell epitopes from the Cas proteins are incubated with cells (e.g., dendritic cells (DCs)). The cells are co-cultured with CD4+ and/or CD8+ T-cells and T-cell activation is determined. The amount of T-cell activation is indicative of the immunogenicity of the putative epitope where higher T-cell activation indicates a more immunogenic peptide or epitope. In certain embodiments, T-cell activation is measured by flow cytometry or FluoroSpot.

Assaying Immunogenicity of Cas-derived Proteins

[00157] In certain embodiments of the invention, the immunogenicity of recombinant Cas proteins made by the methods described herein is determined by measuring the level of T-cell activation induced by such recombinant proteins. In some embodiments, the T-cells comprise CD4+ and/or CD8+ T-cells. In certain embodiments, the level of activation of T-cells is measured using methods well-known in the art, including, but not limited to, flow cytometry and/or FluoroSpot. In some embodiments, the level of T-cell activation induced by the recombinant Cas protein is compared to the level of T-cell activation induced by the wild-type Cas protein from which the recombinant Cas protein was derived. If the level of T-cell activation induced by the recombinant Cas protein is lower than that induced by the wild-type Cas protein from which the recombinant Cas protein was derived, then the recombinant Cas protein is considered to have reduced immunogenicity.

MHC-associated Peptide Proteomics (MAPPs) Assay

[00158] In some embodiments, MHC-associated Peptide Proteomics (MAPPs) assays are used to identify prominent epitopes from the Cas protein. MAPPs technology is a high-throughput approach yielding hundreds of peptide sequences from a small amount of starting material, allowing sequence analysis of self-peptide repertoires from low abundance cell types. See, e.g., Rohn, et al, Nature Immunol. 5:909-918 (2004), and Penna et al, J. Immunol. 7(57:1862-1866 (2001). The MHC-associated Peptide Proteomics (MAPPs) assay involves in vitro identification of HLA class II or class I associated peptides, which are processed by professional antigen presenting cells (APCs) such as dendritic cells, and used to identify specific peptides that are bound to either Class II or Class I antigens. Antigen uptake, processing and presentation processes are taken into account. The naturally processed HLA class II-associated peptides or class I-associated peptides are identified by liquid chromatography-mass spectrometry (Kropshofer and Spindeldreher (2005) in Antigen Presenting Cells: From Mechanisms to Drug Development, eds. Kropshofer and Vogt, Wiley- VCH, Weinheim, 159-98).

Recombinant Cas proteins Made by the Methods Described Herein

[00159] The present invention is also directed to recombinant Cas proteins made by the methods described above. For example, in certain embodiments, the invention is directed to a recombinant Cas protein made by a process comprising introducing one or more amino acid substitutions into one or more residues corresponding to one or more MHC Class I and/or Class II binding sites of a wild-type Cas protein to form the recombinant protein. In some embodiments, the recombinant Cas protein made by the methods disclosed herein has reduced immunogenicity compared to a wild-type Cas protein.

Vectors of the Invention

[00160] Viral transduction with adeno -associated virus (AAV) and lentiviral vectors (where administration can be local, targeted or systemic) have been used as delivery methods for in-vivo gene therapy. In certain embodiments of the present invention, the Cas protein can be expressed intracellularly by transduced cells. Immunogenicity responses to the Cas would thus be largely mediated by MHC Class I receptors (e.g., HLA Class I receptors), which are present in all nucleated cells, thereby activating CD8+ T cells.

[00161] For many therapeutic strategies, included those envisaged by the present invention, Cas protein expression may only be required transiently. As a result, in certain embodiments of the invention, delivery of the Cas protein into cells can be achieved using non-integrative viral vectors. In addition, there are certain embodiments where the expression of CRISPR-Cas system components is required for extended periods - for example, when used in gene circuits which are permanently integrated into the genome of target cells. Such applications have been discussed by Agustin-Pavon, et al., "Synthetic biology and therapeutic strategies for the degenerating brain," Bioessays 36(10):979-990 (2014), which is incorporated by reference herein in its entirety. In some embodiments of the present invention, immunogenicity can have elevated relevance in relation to potential selective clearance of cells stably expressing Cas protein by the immune system. [00162] In certain embodiments, the Cas proteins and methods of the present invention can be used in ex vivo gene editing, such as CAR-T type therapies. These embodiments may involve modification of cells from human donors. In these instances, viral vectors can be also used; however, there is the additional option to directly transfect the Cas protein (along with in-vitro transcribed guide RNA and donor DNA) into cultured cells. In this case both MHC Class I and Class II (e.g., HLA Class 1 and HLA Class 2) receptors may be involved. Furthermore, a phenomenon known as "cross-presentation" can result in presentation of peptides derived from extracellular proteins by HLA Class I receptors, adding additional weight to considering immunogenicity of Cas proteins in relation to both Class I and Class II HLA presentation.

Cell-types of the Invention

[00163] The present invention is also directed to a cell comprising the recombinant Cas protein of the invention, a nucleic acid expressing the recombinant protein of the invention, and/or a vector comprising a nucleic acid of the invention. In certain embodiments, the cell is a prokaryotic cell, for example an E. coli cell. In some embodiments, the cell is a mammalian cell. In some embodiments, the cell is a eukaryotic cell, for example, a lymphocytic cell, a myeloid cell, an induced pluripotent stem cell (iPSC), or a T cell, although all mammalian cell types are envisage as being used in the invention.

Library for Identifying Cas proteins with reduced immunogenicity

[00164] The present invention also encompasses a library comprising at least one recombinant Cas protein comprising one or more amino acid substitutions in one or more residues corresponding to one or more MHC Class I and/or MHC Class II binding sites of a wild-type Cas protein. In certain embodiments, the library can be used to screen for functional recombinant Cas proteins, including recombinant Cas proteins with reduced immunogenicity. This is important as it is highly likely that many amino acid substitutions that reduce immunogenicity may also reduce Cas functionality that is essential to its application. The immunogenicity of functional recombinant Cas proteins in the library can be assessed using well-known techniques, including those described herein.

Methods for altering gene expression using the recombinant Cas proteins [00165] The invention is also directed to methods for altering the DNA sequence at defined genomic locations containing a target sequence comprising introducing into a cell containing the target genetic element a guide R A that hybridizes to the target sequence and a recombinant Cas protein of the invention.

EXAMPLES

[00166] Described below are experimental procedures for reducing the immunogenicity of Cas proteins, in particular Cas9 variants from S. pyogenes and S. aureus. S. pyogenes Cas9 has been more extensively characterized for in vitro gene-editing studies, whereas S. aureus Cas9 has also been utilized for in vivo gene editing, in part due to its smaller size facilitating delivery via adeno-associated virus (AAV) vectors.

Example 1: In-silico prediction of Cas9 immunogenic epitopes for Cas9 variants and cell based immunogenicity assays for wildtype Cas9 (S. pyogenes)

Example 1.1 : In silico prediction of HLA Class I and Class II binding for Cas9 proteins

[00167] This process involves creating a profile sequence for HLA Class I & Class II T-cell epitopes using in silico methods such as those described above, for Cas9 sequences from Streptococcus pyogenes, Streptococcus thermophilus, staphylococcus aureus, and Neisseria meningitidis

[00168] A randomizer algorithm was applied to assess effects of amino acid substitutions on immunogenicity of all 9 and lOmer (containing 9 or 10 amino acid residues, respectively) sequences within the Cas9 proteins. This indicates whether potential substitutions can increase or decrease the immunogenicity of the associated 9mer or 1 Omer.

HLA Class I Mapping

[00169] HLA Class I epitopes were mapped for the four Cas9 variant sequences provided above (SEQ ID NOs: 1-4), but are exemplified for Streptococcus pyogenes Cas9 (SEQ ID NO: 5) below. In short, the platform analyzes the HLA binding specificities of all 9 and 10-mer peptides derived from a target sequence (for more details refer to paragraphs below). Profiling is performed at the allotype level for a total of 28 HLA Class I receptors. [00170] EPIBASE ® calculates binding affinity of a peptide for each of the 28 HLA Class I receptors. Based on this, peptides are classified as strong (S), medium (M), or non (N) binders.

[00171] The allotypes predicted were selected based on frequencies in a variety of populations (Caucasian, Indo-European, Oriental, North Oriental, West African, East African, Mestizo and Austronesian).

[00172] Compiled data are presented in Table 1 , indicating the number of Streptococcus pyogenes Cas9 epitopes specific to each HLA allotype. Peptides in the two binding classes (S and M) are counted separately.

Table 1: Strong and medium binding Streptococcus pyogenes Cas9 epitopes for HLA-A and HLA-B. Total numbers of epitopes in each category for 9-mer and 10-mer sequences are presented (values for different allotypes are represented separately, with allotype references restricted to two digits).

9-mer 10-mer

HLA Strong Medium Strong Medium Allotype

A*01 8 60 6 43

A*02 30 110 37 99

A*02 39 110 43 99

A*02 51 127 53 112

A*02 22 64 18 69

A*03 34 88 29 106

A* l l 43 89 41 84

A*23 32 93 1 14 233

A*24 18 57 51 135

A*26 15 66 22 78

A*29 45 119 61 142

A*30 24 104 58 190

A*31 60 159 50 169

A*33 24 113 74 226

A*68 59 143 234 300

A*68 48 128 107 218

B*07 14 47 6 62

B*08 26 105 15 80

B* 15 16 64 23 73

B* 18 21 84 46 121

B*27 7 100 13 105

B*35 24 110 21 73

B*40 19 49 1 1 57

B*44 0 25 1 28

B*51 0 7 0 11

B*53 14 77 35 132

B*57 0 15 0 25

B*58 15 94 1 1 119

[00173] HLA Class I epitope maps for two regions of Streptococcus pyogenes Cas9 (positions 1-125 and 1026-1150) are shown in Figures 1 and 3 respectively. The latter region was selected as an exemplar region with high S epitope counts. [00174] Information regarding allotype frequencies can be helpful to further assess the global immunogenic response of the protein of interest. It is remarked that allotype frequencies cannot just be added up to estimate total population response: epitope processing, clustering of binders into short sequence fragments, immunodominance, T-cell receptor properties, presence of B-cell epitopes (antigenicity) and Mendelian genetics are all factors that complicate global immunogenicity assessment. As a result, the population response can be different from the sum of allotype frequencies.

HLA Class II Mapping

[00175] HLA Class II epitopes have been mapped for the four Cas9 variants sequences provided above (SEQ ID NOs: 1-4), but are exemplified for Streptococcus pyogenes Cas9 (SEQ ID NO: 5), below. In short, the platform analyzes the HLA binding specificities of all 10-mer peptides derived from a target sequence (for more details refer to the paragraphs below). Profiling is performed at the allotype level for a number of HLA Class II receptors. Table 2 indicates the number of Streptococcus pyogenes Cas9 epitopes specific to each receptor.

[00176] EPIBASE ® calculates the binding affinity of a peptide for each of the available HLA Class II receptors. Based on this, peptides are classified as strong (S), medium (M), or non- binders (N). We refer to Kapoerchan et al., for the successful usage of EPIBASE ® in predicting binders against selected HLA Class II receptors.

Table 2: Strong and medium binding Streptococcus pyogenes Cas9 epitopes for different HLA Class II allotypes. Total numbers of epitopes in each category for 10-mer sequences are presented (values for different allotypes are represented separately, with allotype references restricted to two digits).

10-mer

Allotype Strong Medium

DRB1*01 16 65

DRB1*01 21 81

DRB1*03 7 46 DRB1*03 6 33

DRB1*03 7 46

DRB1*04 8 26

DRB1*04 10 35

DRB1*04 10 35

DRB1*04 16 45

DRB1*04 16 46

DRB1*04 9 32

DRB1*04 19 68

DRB1*04 19 68

DRB1*07 8 39

DRB1*08 4 64

DRB1*08 5 67

DRB1*08 4 64

DRB1*08 7 70

DRB1*09 6 30

DRB1*10 15 67

DRB1*11 21 70

DRB1*11 6 24

DRB1*11 27 86

DRB1*11 27 86

DRB1*11 21 70

DRB1*12 12 92

DRB1*12 12 92

DRB1*13 8 28 DRB1*13 5 29

DRB1*13 26 59

DRB1*13 1 31

DRB1*13 26 59

DRB1*14 27 73

DRB1*14 32 77

DRB1*14 27 73

DRB1*14 27 86

DRB1*14 33 87

DRB1*15 16 6

DRB1*15 13 52

DRB1*15 16 67

DRB1*15 16 67

DRB1*16 16 78

DRB1*16 16 78

DRB3*01 4 44

DRB3*02 0 48

DRB3*02 1 40

DRB3*03 7 67

DRB4*01 9 37

DRB5*01 28 82

DRB5*01 2 40

DRB5*02 5 45

DQA1*01 2 43

DQA1*01 2 43

[00177] HLA Class II epitope maps for two regions of Streptococcus pyogenes Cas9 (positions 1 -125 and 1026-1150) are shown in Figures 2 and 4 respectively. The latter region was selected as an exemplar region with high S epitope counts.

[00178] In the humoral response raised against an antigen, the observed T Cell activation proliferation is generally interpreted in terms of the DRB1 specificity. However, it can also be important to take into account the possible contribution of the DRB3/4/5, DQ and DP genes.

[00179] Given the lower expression levels of the DQ and DP receptors compared to the DRB receptors, the class of medium epitopes for DQ and DP was ignored. Those epitopes that are strong binders (or better) to any allotype or are medium binders for DRB1 or DRB3/4/5 are operationally defined as "critical epitopes."

[00180] Information regarding allotype frequencies can be beneficial for further assessment of the global immunogenicity risk of therapeutic proteins. Allotype frequencies cannot simply be added together to estimate the total population response: epitope processing, clustering of binders into short sequence fragments, immunodominance, T-cell receptor properties, presence of B-cell epitopes (antigenicity) and Mendelian genetics are all factors which contribute to a global immunogenicity assessment. As a result, the population response can be different from the sum of allotype frequencies.

Randomization of Cas9 Sequences for Reduced Immunogenicity [00181] Randomization was carried out in relation to HLA Class I and Class II for all four Cas9 sequences {Streptococcus pyogenes, Streptococcus thermophilus , staphylococcus aureus, and Neisseria meningitides). Data is presented on a single identified immunogenic region from Streptococcus pyogenes Cas9. This can be found between amino acids 1035 and 1038 of the Cas9 sequence (originally identified from the HLA Class I epitope profile). The epitope maps for amino acids 1026-1 150 (i.e., including this region) are shown for HLA Class I and HLA Class II in Figure 3 and Figures 4A-C, respectively. It is interesting to note that this region appears to be strongly immunogenic for both HLA Classes.

[00182] The randomization data and plots are shown in for 9-mers and 10-mers in Figures 5A- C and Figures 6A-D respectively. In these plots, substitutions can be clearly identified, which reduce immunogenicity of the specific epitope for both HLA Class I and Class II.

[00183] Figures 5A and 6A in the randomization plots indicates the effect of each amino acid substitution on the number of HLA allotypes binding the specific epitope. A positive score indicates that the substitution increases the number of binders, where a negative score does the opposite.

[00184] The amino acid substitution deimmunisation scores for 9-mers and 10-mers for Streptococcus pyogenes Cas9 are presented in Figures 8A-D. This indicates the likelihood that for any given position a substitution can be identified that will deimmunise the corresponding epitope. This does not exclude the fact that some epitopes may only be deimmunised by a single substitution. The method used for data collation means that scores for 9-mers are applicable to HLA Class I epitopes and scores for 10-mers are applicable to both HLA Class I and Class II epitopes.

[00185] Amino acid substitution deimmunisation scores (and their corresponding amino acid positions) were subsequently filtered based on those which gave scores lower than a cut-off threshold of -20. These are exemplified for Streptococcus pyogenes Cas9 in Figures 8B and 8D. The positions identified are also presented in table form in Figure 9 for Streptococcus pyogenes, Streptococcus thermophilus, Staphylococcus aureus, and Neisseria meningtidis Cas9 variants.

Example 1.2: Assessment of Cas9 structural and functional regions [00186] A three-dimensional model was generated using the coordinates of a publically available Cas9 complex crystal structure in the protein data-bank (PDB) (see Nishimasu, et ah, "Crystal structure of Cas9 in complex with guide R A and target DNA," Cell 156(5):935-49 (2014), which is incorporated by reference herein in its entirety). This structural model can be used to assess structural criteria for each amino acid residue. This information can be combined with immunogenicity data from Example 1.1 to prioritize sequence modifications of the Cas9 protein which are likely to maintain functionality while reducing immunogenicity.

[00187] Further to the data generated in the preceding paragraph, the proximity of prioritized/re levant HLA Class I and HLA Class II epitopes to key structural or functional motifs of the Cas9 sequence were performed. The Cas9 proteins from S. pyogenes and S. aureus were used for illustrative purposes.

[00188] While the macro -homology across Cas9 variants is limited - see Figure 7 - it is possible to identify relevant epitopes, as well as structural features, that are conserved in multiple Cas9 variants. In order to do this, known three-dimensional structures deposited in the PDB were compared to the target Cas9 proteins. The quality of the comparison (e.g., percent homology on macro- and micro- scale) was determined and only those sequences with a relevant level of sequence homology selected for subsequent analysis.

[00189] Those epitopes that were chosen for further characterization were aligned to corresponding domains of other species. Based, at least in part, on these alignments, three- dimensional molecular models of the target epitopes were generated. Functionally relevant positions/regions in the Cas9 proteins were identified and mapped into the three-dimensional structure of the proteins.

Example 1.3: Prioritisation of Cas9 amino acid substitutions based on Examples 1.1 and 1.2

[00190] The data compiled from Examples 1.1 and 1.2 were used to identify which epitopes could be targeted to both minimize immunogenicity risks while also retaining as much biological activity as possible in the recombinant, i.e., engineered, protein. Such domains have been described by Nishimasu et al. (2014). For example, one criterion utilized was to avoid changing amino acids present within 5 angstroms of the known nuclease domains of Cas9. For those epitopes selected to effectuate reduced immunogenicity, amino acid substitutions were selected to preserve structural features of the Cas9 protein (e.g., alpha helices/beta- sheets) as much as possible.

[00191] A global list of positions/regions that can be randomized/changed for potential reduced immunogenicity can be generated. Assessment of the effect of amino acid substitution of the identified regions can be carried out in multiple ways.

[00192] Limitations exist in the number of Cas9 variants that can be practically screened for immunogenicity in Example 2, below. It therefore highly likely that the final Cas9 variants assessed will contain multiple substitutions. However, there is considerable flexibility in how the Cas9 functionality screens can be carried out. A step-wise approach can be utilized to minimize the potential impact of the deimmunising substitutions in Cas9 function. In this method more variants can be screened (e.g., using a library approach), exhibiting larger numbers of substitutions, individually and in combination, and also to utilize multiple subsequent rounds of screening-selection, where results from primary screens are used to inform library design for subsequent rounds.

Example 1.4: In-vitro immunogenicity assays on purified Cas9 protein

[00193] Assays were carried out, using purified, non-deimmunised ("wildtype"), Cas9 proteins, i.e., SEQ ID NOs: 5-8, in order to assess the starting level of immunogenicity relative to proteins of known immunogenicity (shown in Figure 10A and 10B). A histidine purification tag (his-tag), known to be non-immunogenic, was added in order to facilitate purification.

[00194] Assays were performed using peripheral blood mononuclear cells (PBMCs) from healthy human donors corresponding to different HLA allotypes. Typically PBMCs from at least 10 or 20 different donors were utilized. There was prior evidence that HLA Class I presentation can occur even in cases where cells are exposed "externally" to whole antigens (a phenomenon known as "cross-presentation"). This was used to explore how antigen presentation can influence the activation of CD4+ (helper T-cells) or CD8+ (cytotoxic T-cells) lymphocytes.

[00195] PBMCs were incubated with recombinant Cas9 protein (Cas9 protein with incorporated amino acid substitutions determined from the analysis in Examples 1.1-1.3 above). The recombinant Cas9 protein was purified to eliminate LPS (lipopolysaccharide) contamination (endotoxin can induce T cell activation and hence give false positive results). CD4+ or CD8+ T- cell activation was assessed by measuring the proliferation of the T-cells by flow cytometry or FluoroSpot. Activation of CD4+ T-cells indicated the presence of HLA Class II T-cell epitopes, whereas the activation of CD8+ T-cells indicated that HLA Class I epitopes were present in the Cas9 protein sequence.

Example 1.5: HLA Class I and HLA Class II MAPPS (MHC-associated Peptide Proteomics) assay

[00196] Further to use of the EPIBASE ® platform for in silico prediction of immunogenic epitopes, an in vitro approach was applied to experimentally identify which epitopes are presented by HLA Class I and Class II molecules. Use of EPIBASE ® can be helpful due to the high number of immunogenic epitopes identified in silico.

[00197] This assay involved immunoprecipitation of HLA Class I (HLA-A, B and C) or HLA Class II (HLA-DR, DP and DQ) proteins from antigen presenting cells (APCs) previously incubated in the presence of antigen, i.e., Cas9 protein. Cas9-derived peptides bound to the HLA molecules were acid-eluted and identified by Mass Spectrometry (MS). Identification of Cas9- derived HLA-binding peptides provides data on which peptides are processed and presented preferentially by these APCs. Analysis of this data helped to further define which of the in silico predicted epitopes are actually more relevant in potentially triggering an immune response (an example is shown in Figure 1 1).

[00198] Analysis of HLA Class I bound peptides may additionally be carried out using cells expressing and/or transfected with Cas9.

Example 1.6: Activation of CD4+ and CD8+ T-cells by short peptides from Cas9

[00199] These assays evaluate the relative immunogenicity of different Cas9-derived peptides by incubating dendritic cells (DC) with short synthetic peptides representing the potential T-cell epitopes from Cas9 and co-culturing them with autologous T-cells (CD4+ or CD8+) to evaluate the impact of each peptide on T-cell activation. These assays can be utilized to rank the relative importance of different in sz ' /z ' co-predicted epitopes and prioritize those to be removed to achieve reduced immunogenicity. These assays could also help inform the engineering process coming out of Example 1.3.

Example 1.7: Identification of B-cell epitopes

[00200] In addition to epitopes bound to HLA molecules, as described above, it is also important to consider presence of epitopes bound by surface IgG molecules (B cell receptors) found on B cells. B cell receptors are responsible for selective uptake of antigens and presentation on B cells via HLA Class II, for subsequent interaction with T cell receptors found on T helper cells. In contrast to peptides bound by HLA molecules, which have been processed such that they are considered only in linear terms, B cell epitopes can also be conformational, as they are found in the native context of the protein.

[00201] The conformational aspect of B cell epitopes makes in silico prediction problematic relative to T cell epitopes (HLA binding). In vitro assays, however, can be used to identify B cell activation in response to either full length proteins (conformational and linear epitopes) or derived peptides (linear epitopes) covering the relevant regions.

Example 2: Functional Testing of Cas9 Variants in mammalian cell lines

Example 2.1 : Generation and preparation of mammalian expression vectors for Cas9 variants and gR As

[00202] Based on the data from Example 1 above, transient vectors for mammalian expression of recombinant Cas9 can be generated for a number of Cas9 variants.

[00203] Vector designs can be based on constructs designed by Mali et al. "Cas9 as a versatile tool for engineering biology," Nature Methods 10:957-63 (2013) - see Supplementary Figure SI , which is incorporated by reference herein in its entirety. Coding sequences for different Cas9 variants (either from different bacterial species, or containing modified immunogenic epitopes) can be inserted in place of the S. pyogenes sequence as appropriate. Guide R A and Cas9 regions can be synthesized by a gene synthesis company (e.g., GeneART/DNA2.0) and cloned into a standard E. coli cloning vector. Guide RNA sequences can be appropriate to the Cas9 variant in question, i.e., as described by Esvelt (2013) and Ran et al. (2015). Large preparations of these vectors can be made using standard plasmid preparation procedures (e.g., Qiagen Maxi- prep) in order to produce DNA suitable for transfection.

[00204] Cas9 and guide R A plasmids can be co-precipitated (standard sodium acetate/ethanol precipitation) and re-suspended in TE ready for transfection. Final DNA concentrations are assessed by spectrophotometry (Nanodrop instrument, or equivalent) before being adjusted to compensate for variable precipitation efficiency.

Example 2.2: Testing of z ' «-vzYro_functionality in CHO cells (vs. WT control) using model cell line expressing eGFP.

[00205] A Chinese hamster ovary cell line derived from CHOK1 SV and containing stably integrated copies of eGFP can be used to test functionality of the different Cas9 variants identified above as having potential reduced immunogenicity. In this cell line, expression of eGFP is constitutive and is directed by the cytomegalovirus (CMV) promoter.

[00206] A gRNA vector can be designed to target a region early in the eGFP coding sequence. Co-transfection of the gRNA and Cas9 vector will result in introduction of double-strand breaks (DSB) in the eGFP coding sequence. DSBs are repaired by the cell's endogenous DNA repair machinery but this is largely error prone, resulting in introduction of small insertions and deletions (indels). In the majority of cases, indels result in frame shift mutations to the coding sequence, which would prevent production of functional (fluorescent) eGFP.

[00207] Functionality of Cas9 variants can be assessed by knock-down of eGFP flurourescence 2-4 days after transfection and/or by analysis of genomic DNA-derived PCR products flanking the target site. In all cases, results from Cas9 variants can be compared to the unmodified sequences in order to assess relative functionality.

[00208] PCR products can be treated with an enzyme that cleaves heteroduplex DNA (e.g., T7EI) and analyzed qualitatively by gel electrophoresis. In this case, the percentage of the PCR product that is cleaved by the T7EI enzyme is indicative of the efficiency, i.e., functionality, of the Cas9 enzyme. Alternatively, the PCR products can be sequenced using next generation sequencing techniques, where the high level repeat sequencing of the PCR product gives a quantitative measure of the different modified or unmodified sequences present (again indicating Cas9 functionality).

Alternatives to Examples 2.1 and 2.2

[00209] Multiple different approaches can be employed to generate rationally designed libraries of Cas9 variants, which can be screened to identify those variants that are functional. One approach would be to utilize a cell line with an integrated, substrate dependent suicide gene. Viral transduction of the cell line with DNA encoding the Cas9 library plus a guide RNA targeting the suicide gene would result in introduction of a double strand break (DSB) early in the suicide gene coding sequence where the Cas9 is functional. Addition of the suicide gene substrate would kill cells where Cas9 had failed to cleave the suicide gene sequence. Cas9 sequences could be recovered from surviving cells and analyzed by next-generation sequencing. Limitations imposed by library construction methods and transduction efficiency, for example, would inform the number of variants that could be screened using the selected method. Results from Example 2, in which ever from, will need to be ranked/prioritized in order to provide a practical number of variants for input to the process of Example 3 below.

Example 3: Generation of purified Cas9 proteins for immunogenicity assays using E.coli expression system

Example 3.1 : Expression of S. pyogenes Cas9 protein in E. coli

[00210] Cas9 from S. pyogenes can be expressed in E.coli using methods and expression constructs described by Gagnon et al. (2014), or equivalent. See Gagnon, et al. "Efficient mutagenesis by Cas9 protein-mediated oligonucleotide insertion and large-scale assessment of single-guide RNAs," PLOS One 9(8):el06396 (2014), which is incorporated by reference herein in its entirety. All variants will contain a 6xHis-tag to aid purification.

[00211] Selected variants will feature SV40 (or alternative) nuclear localization sequences to test additional impact on immunogenicity. It is currently unclear whether such sequences would be required for the therapeutic use of Cas9.

Example 3.2: Purification of E. co/z-derived Cas9 protein [00212] Cas9 purification methods can be tested and endotoxin load assessed (aim 5-10mg, <lEU/mg). A commercial nickel column can be used for protein purification (such as the Ni- NTA system from Life Technologies) according to the manufacturer's instructions. This can be followed by a Capto Q (GE Life Sciences) purification step to reduce endotoxin load. Endotoxin levels can be assessed by a commercial assay (such as the Endosafe-PTS system from Charles River), according to the manufacturer's instructions. Proteins can be quantified during purification procedure and in the final eluted product by Bradford assay or A280 reading on suitable spectrophotometer ( anodrop or equivalent).

Example 3.3: Expression and purification of Cas9 variants for immunogenicity testing

[00213] Based on data from Examples 1 and 2 above, a range of different recombinant Cas9 variants can be produced and purified. E. coli expression vectors tested in Example 3.1 can be modified to encode for the preferred recombinant Cas9 variant sequences using standard molecular cloning techniques. Proteins can be expressed and purified as described in Example 3.2 above.

Example 4: Evaluation of Immunogenicity of Cas9 Variants

[00214] Cas9 variants produced and purified in Example 3 above can be assessed again in PBMC samples obtained from the same donors assessed in Example 1. PBMCs can be incubated with different endotoxin-free Cas9 variants (up to 10-12). CD4+ or CD8+ T-cell activation can be assessed by measuring the proliferation of the T-cells by flow cytometry or ELISpot.

[00215] CD4+ or CD8+ T-cell activation can be assessed for each one of the new variants and compared to that of the parental (wild-type) molecule. Analysis of the response will determine whether additional cycles of re-engineering would be required to reduce even further the immunogenicity of the protein.

Example 5: Additional Iterations of the Above Examples

[00216] Optionally, additional re-design processes outlined in the above Examples can be implemented in order to refine the final designs. [00217] Additional iterations of the procedures in the above Examples can be performed at various stages throughout the entire process above. For example, in silico mapping, or in silico re-engineering, can be performed by repeating the process of Examples 1.1-1.3 and incorporating the information obtained from Examples 1.5, 1.6, 2 and 4.

[00218] For example, deimmunising substitutions were determined for Cas9 from Streptococcus pyogenes. Several naturally presented HLA Class II binding peptides from Cas9 protein were identified using MAPPs HLA Class II assay (one donor only). These peptides form five different clusters (see Table 3 and Figure 11).

Table 3. Naturally presented HLA binding regions from Cas9.

[00219] EPIBASE® in silico platform was used to identify residue substitutions in these clusters which are predicted to remove HLA binders from these regions. Table 4 contains a list of residues and possible amino acid substitutions (the list is not exclusive) which reduce/remove HLA binding in the identified clusters. In addition, Table 4 contains a predicted reduction in DRB1 score of the protein if one of the suggested substitutions is made.

[00220] The immunogenic risk of a protein or peptide can be represented by an approximate score, the DRB1 score, calculated taking into account a number of factors including the numbers of critical HLA binders and the population frequencies of the affected HLA allotypes.

Table 4. List of residues and possible amino acid substitutions removing/reducing HLA binding.

Cluster Position Original Possible substitutions Reduction in DRB1 residue score due to substitution cluster 1 105 F D,E -34

cluster 1 106 L D,E,G,K,P,Q,R between -22 and -58 cluster 1 107 V D,E,G between -28 and -40 cluster 2 258 L D, E, G, K, P -24

cluster 2 263 K A,D,E,G,N,P,S,T between -36 and -49 cluster 2 264 L A,D,E,G,H,K,N,P,Q,R,S,T,V between -31 and -61 cluster 2 265 Q D,E,G,N,P,T between -28 and -50 cluster 2 266 L A,D,E,G,N,P,Q,S,T, V between -24 and -50 cluster 2 267 S A,D,E,G,H,P,T between -34 and -50 cluster 3 696 L E,G,P between -26 and -30 cluster 4 846 F E,W between -31 and -33 cluster 4 847 L D,E,F,G,H,K,N,P,Q,S,T,W between -33 and -58 cluster 4 852 I D,E,F,G,Y -20

cluster 4 855 K D,E,G,P,S between -23 and -25 cluster 5 1278 K A,D,E,F,G,N,P,Q,S,T,V,W between -22 and -92 cluster 5 1279 R D,E,H,K,Q between -23 and -1 16 cluster 5 1280 V A,D,E,G,K,N,P,Q,S,T between -31 and -92 cluster 5 1281 I A,D,E,F,G,H,K,N,P,Q,R,S,T,W between -47 and -1 16 cluster 5 1282 L A,D,E,G,H,N,P,S,T between -34 and -102

[00221] Substitutions listed in Table 4 were selected with the objective of removing/reducing HLA binding. Aspects of structural integrity and functionality of Cas9 were not considered at this point. In addition, deimmunising substitutions were selected only in five clusters identified based on data from one donor.

[00222] The following example demonstrates a reduction in predicted immunogenic risk if five substitutions (one in each cluster) are made. Calculations were determined using the following substitutions in the amino acid sequence of Cas9: L106D, K263D, L696G, L847D, I1281D. The deimmunised version of Cas9 protein has a reduced DRBl score, as shown in Table 5, with overall reduction by 295. Looking at the DRBl score restricted to the combined five clusters, the original Cas9 has a DRBl score of 177.6 and the deimmunised Cas9 protein has a DRBl score of 23.3, achieving an 87% reduction. Furthermore, the five substitutions completely removed DRBl epitopes restricted by the HLA allotypes of the donor (DRB1 *03 and DRB1*13).

Table 5. DRBl scores of the Cas9 protein and deimmunised Cas9 protein with five substitutions.

DRBl score for DRBl score due to Number of critical whole protein five clusters * epitopes in five

cluster regions *

Cas9 12693.6 177.6 16

Deimmunised Cas9 12398.6 23.3 9

with 5 substitutions

* Only epitopes/binders fully contained in the ive clusters are includec