Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
METHODS AND PRODUCTS FOR GENETIC ENGINEERING
Document Type and Number:
WIPO Patent Application WO/2017/068077
Kind Code:
A1
Abstract:
The present invention relates to a virus-derived particle comprising one or more Cas protein(s), as well as to kits and methods using the same for altering a target nucleic acid.

Inventors:
OHLMANN THÉOPHILE (FR)
MANGEOT PHILIPPE (FR)
RICCI EMILIANO (FR)
Application Number:
PCT/EP2016/075289
Publication Date:
April 27, 2017
Filing Date:
October 20, 2016
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
INST NAT DE LA SANTE ET DE LA RECH MEDICALE (INSERM) (FR)
CENTRE NAT DE LA RECH SCIENT (CNRS) (FR)
ECOLE NORMALE SUPERIEURE LYON (FR)
UNIV CLAUDE BERNARD LYON (FR)
International Classes:
C12N7/00; C07K14/15; C12N5/10; C12N9/22; C12N15/10; C12N15/11; C12N15/62
Domestic Patent References:
WO1992012237A11992-07-23
WO2012000618A22012-01-05
WO2013163628A22013-10-31
WO2014093595A12014-06-19
WO2015089247A12015-06-18
WO2015089486A22015-06-18
WO1995022617A11995-08-24
Foreign References:
US6060064A2000-05-09
US6204059B12001-03-20
US10968253B22021-04-06
US10968253B22021-04-06
Other References:
S. RAMAKRISHNA ET AL: "Gene disruption by cell-penetrating peptide-mediated delivery of Cas9 protein and guide RNA", GENOME RESEARCH, vol. 24, no. 6, 2 April 2014 (2014-04-02), pages 1020 - 1027, XP055128944, ISSN: 1088-9051, DOI: 10.1101/gr.171264.113
WUJIN SUN ET AL: "Self-Assembled DNA Nanoclews for the Efficient Delivery of CRISPR-Cas9 for Genome Editing", ANGEWANDTE CHEMIE INTERNATIONAL EDITION, vol. 54, no. 41, 5 October 2015 (2015-10-05), DE, pages 12029 - 12033, XP055268872, ISSN: 1433-7851, DOI: 10.1002/anie.201506030
LAFOUNTAINE JUSTIN S ET AL: "Delivery and therapeutic applications of gene editing technologies ZFNs, TALENs, and CRISPR/Cas9", INTERNATIONAL JOURNAL OF PHARMACEUTICS, vol. 494, no. 1, 13 August 2015 (2015-08-13), pages 180 - 194, XP029276502, ISSN: 0378-5173, DOI: 10.1016/J.IJPHARM.2015.08.029
SHEFAH QAZI ET AL: "Programmed Self-Assembly of an Active P22-Cas9 Nanocarrier System", MOLECULAR PHARMACEUTICS, vol. 13, no. 3, 7 March 2016 (2016-03-07), US, pages 1191 - 1196, XP055268807, ISSN: 1543-8384, DOI: 10.1021/acs.molpharmaceut.5b00822
J G CHOI ET AL: "Lentivirus pre-packed with Cas9 protein for safer gene editing", GENE THERAPY, vol. 23, no. 7, 1 July 2016 (2016-07-01), GB, pages 627 - 633, XP055333406, ISSN: 0969-7128, DOI: 10.1038/gt.2016.27
JINEK ET AL., SCIENCE, vol. 337, 2012, pages 816 - 821
OVERBALLE-PETERSEN ET AL., PROC NATL ACAD SCI USA, vol. 110, 2013, pages 19860 - 19865
GONG ET AL., NAT. STRUCT MOL BIOL, vol. 12, 2005, pages 304 - 312
CONG ET AL., SCIENCE, vol. 339, 2013, pages 819 - 823
QI ET AL., CELL, vol. 152, 2013, pages 1173 - 1183
RAN ET AL., CELL, vol. 154, 2013, pages 1380 - 1389
MALI ET AL., SCIENCE, vol. 339, 2013, pages 823 - 826
FABRE ET AL., PLOS NEGL. TROP. DIS., vol. 8, 2014, pages E2671
HWANG ET AL., PLOS ONE, vol. 8, 2013, pages E68708
HAI ET AL., CELL RES., 2014
GUO ET AL., DEVELOPMENT, vol. 141, 2014, pages 707 - 714
DICARLO ET AL., NUCLEIC ACIDS RES., vol. 41, 2013, pages 4336 - 4343
GRATZ ET AL., GENETICS, 2014
NIU ET AL., CELL, vol. 156, 2014, pages 836 - 843
YANG ET AL., J. MOL. CELL BIOL., vol. 6, 2014, pages 97 - 99
MA ET AL., CELL RES., vol. 24, 2014, pages 122 - 125
MASHIKO ET AL., DEV. GROWTH DIFFER., vol. 56, 2014, pages 122 - 129
JINEK ET AL., ELIFE, vol. 2, 2013, pages E00471
ABE ET AL., J VIROL, 1998
O'CONNELL ET AL., NATURE, vol. 516, 2014, pages 263 - 266
ABE ET AL., J VIROL, vol. 72, 1998, pages 6356 - 6361
MANGEOT ET AL., MOL THER J AM SOC GENE THER, vol. 19, 2011, pages 1956 - 1666
KUSHNIR ET AL., VACCINE, vol. 31, 2012, pages 58 - 83
ZELTONS, MOL BIOTECHNOL, vol. 53, 2013, pages 92 - 107
LUDWIG ET AL., CURR OPIN BIOTECHNOL, vol. 18, no. 6, 2007, pages 537 - 55
MAETZIG ET AL., CURRENT GENE THERAPY, vol. 12, 2012, pages 389 - 409
KACZMARCZYK ET AL., PROC NATL ACAD SCI USA, vol. 108, no. 41, 2011, pages 16998 - 17003
YEE ET AL., PROCNATL ACAD SCI, USA, vol. 91, 1994, pages 9564 - 9568
CRONIN ET AL., CURR GENE THER, vol. 5, no. 4, 2005, pages 387 - 398
MANGEOT ET AL., MOLECULAR THERAPY, vol. 19, no. 9, 2011, pages 1656 - 1666
SHARMA ET AL., PROC NATL ACAD SCI USA, vol. 94, 1997, pages 10803 - 10808
GUIBINGUA ET AL., MOLECULAR THERAPY, vol. 5, no. 5, 2002, pages 538 - 546
RASMUSSEN ET AL., VIROLOGY, vol. 178, no. 2, 1990, pages 435 - 451
MANGEOT ET AL., JOURNAL OF VIROLOGY, vol. 71, no. 18, 2000, pages 8307 - 8315
NEGRE ET AL., GENE THERAPY, vol. 7, 2000, pages 1613 - 1623
MANGEOT ET AL., NUCLEIC ACIDS RESEARCH, vol. 32, no. 12, 2004, pages EL02
SAENZ ET AL., COLD SPRING HARB PROTOC, 2012, pages 71 - 76
COLD SPRING HARB PROTOC, 2012, pages 124 - 125
COLD SPRING HARB PROTOC, pages 118 - 123
JALAGUIER ET AL., PLOSONE, vol. 6, no. LL, 2011, pages E28314
CERVERA ET AL., J BIOTECHNOL, vol. 166, no. 4, pages 152 - 165
TANG ET AL., JOURNAL OF VIROLOGY, vol. 86, no. 14, 2012, pages 7662 - 7676
OLSEN, GENE THER, vol. 5, no. L 1, 1998, pages 1481 - 1487
MSELLI-LAKHAL, J VIROL METHODS, vol. 136, no. L-2, 2006, pages 177 - 184
GIRARD-GAGNEPAIN ET AL., BLOOD, vol. 124, no. 8, 2014, pages 1221 - 1231
KANG ET AL., VIRUSES, vol. 7, 2015, pages 1134 - 1152
FONTANA ET AL., VACCINE, vol. 32, no. 24, 2014, pages 2799 - 27804
QUAN ET AL., VIROLOGY, vol. 430, 2012, pages 127 - 135
LATHAM ET AL., JOURNAL OF VIROLOGY, vol. 75, no. 13, 2001, pages 6154 - 6155
TOME-AMAT ET AL., MICROBIAL CELL FACTORIES, vol. 13, 2014, pages 134 - 142
WALPITA ET AL., PLOSONE, 2015
LI ET AL., JOURNAL OF VIROLOGY, vol. 71, no. 10, 1997, pages 7207 - 7213
MURAWSKI ET AL., JOURNAL OF VIROLOGY, vol. 84, no. 2, 2010, pages 1110 - 1123
HERBST-KRALOVETZ ET AL., EXPERT REV VACCINES, vol. 9, no. 3, 2010, pages 299 - 307
OGASAWARA ET AL., IN VIVO, vol. 20, 2006, pages 319 - 324
WANG ET AL., EXPERT REV VACCINES, vol. 12, no. 2, 2013
PEIFANG ET AL., CLIN EXP IMMUNOL, vol. 97, no. 3, 1994, pages 361 - 366
BROWN ET AL., BIOCHEMISTRY, vol. 48, no. 47, 2009, pages 11155 - 11157
CHYLINSKI ET AL., NUCLEIC ACIDS RESEARCH, vol. 42, no. 10, 2014, pages 6091 - 6105
SINKUNAS ET AL., THE EMBO JOURNAL, vol. 30, no. 7, 2011, pages 1335 - 1342
ALIYARI ET AL., IMMUNOLOGICAL REVIEWS, vol. 227, no. L, 2009, pages 176 - 188
CASS ET AL., BIOSCI REP
MAKAROVA ET AL., BIOLOGY DIRECT, vol. 6, 2011, pages 38
GASIUNAS ET AL., PROC NATLACAD SCI USA, vol. 109, no. 39, 2012, pages E2579 - E2586
HELER ET AL., NATURE, vol. 519, no. 7542, 2015, pages 199 - 202
ESVELT ET AL., NAT METHODS, vol. 10, no. LL, 2013
CHYLINSKI ET AL., BIOLOGY, vol. 10, no. 5, 2013, pages 726 - 737
ZEISCHE ET AL., CELL, 2015, Retrieved from the Internet
SANJANA ET AL., NAT METHODS, vol. LL, no. 18, 2014, pages 783 - 784
VOELKEL ET AL., PROC NATL ACAD SCI USA, vol. 107, no. 17, 2010, pages 7805 - 7810
MAIL ET AL., SCIENCE, vol. 339, 2013, pages 823 - 826
WANG ET AL., CELL, vol. 153, 2013, pages 910 - 918
JAO ET AL., PROC NATL ACAD SIC USA, vol. 110, 2013, pages 13904 - 13909
SHALEM ET AL., SCIENCE, vol. 343, 2014, pages 84 - 87
MAEDER ET AL., NAT METHODS, vol. 10, 2013, pages 977 - 979
FARBOUD ET AL., GENETICS, 2015
MA ET AL., BIOMED RESEARCH INTERNATIONAL, vol. 2013, 2013
ABE ET AL., JOURNAL OF VIROLOGY, vol. 72, no. 8, 1998, pages 6356 - 6361
LIN ET AL., ELIFE, vol. 3, 2014, pages E04766
MARKOWITZ ET AL., J. VIROL., vol. 62, 1988, pages 1120
VOELKEL ET AL., PROC NATL ACAD SCI USA, vol. 107, 2010, pages 7805 - 7810
NEGRE ET AL., GENE THER, vol. 7, 2000, pages 1613 - 1623
YEE ET AL., METHIDS CELL BIOL, vol. 43, 1994, pages 99 - 112
KIEUSSEIAN ET AL., BLOOD, vol. 107, 2006, pages 492 - 500
"Methods in Molecular Biology", 1995, HUMANA PRESS
O'REILLY ET AL.: "Baculovirus Expression Vectors: A Laboratory Manual", 1994, OXFORD UNIV. PRESS
SAMULSKI ET AL., J. VIR., vol. 63, 1989, pages 3822 - 8
KAJIGAYA ET AL., PROC. NAT'L. ACAD. SCI. USA, vol. 88, 1991, pages 4646 - 50
RUFFING ET AL., J. VIR., vol. 66, 1992, pages 6922 - 30
KIMBAUER ET AL., VIR., vol. 219, 1996, pages 37 - 44
ZHAO ET AL., VIR, vol. 272, 2000, pages 382 - 93
TAKAHARA ET AL., JOURNAL OF VIROLOGY, vol. 66, no. 6, June 1992 (1992-06-01), pages 3725 - 32
F. WAGNER ET AL., EMBO JOURNAL, vol. 4, no. 3, 1985, pages 663 - 666
MANGEOT ET AL., THER J AM SOC GENE THER, vol. 19, 2011, pages 1656 - 1666
ZURIS ET AL., NAT BIOTECHNOL, vol. 33, no. L, 2015, pages 73 - 80
JOHNSTON ET AL., GENE THER, vol. 21, no. 12, 2014, pages 1008 - 1020
FENARD ET AL., MOLECULAR THERAPY NUCLEIC ACIDS, vol. 2, 2013, pages E90
ANASTASOV ET AL.: "Lentiviral vectors and exosomes as gene and protein delivery tools", METHODS IN MOLECULAR BIOLOGY, vol. 1448, 2016, pages 49 - 61
C. G. A THOMAS, MEDICAL MICROBIOLOGY, 1983
YEE ET AL., METHODS CELL BIOL, vol. 43, 1994, pages 99 - 112
ZHU ET AL., PLOS ONE, vol. 9, 2014, pages E108424
DALE ET AL., METHODS SAN DIEGO CALIF, vol. 53, 2011, pages 20 - 26
GARRONE ET AL., SCI TRANSL MED, vol. 3, 2011, pages 94RA71
MANGEOT ET AL., MOL THER J AM SOC GENE THER, vol. 19, 2011, pages 1656 - 1666
LOMBARDO ET AL., J IMMUNOL BALTIM MD, vol. 178, 1950, pages 3731 - 3739
OKIMOTO ET AL., MOL THER J AM SOC GENE THER, vol. 4, 2001, pages 232 - 238
GIRARD-GAGNEPAIN A ET AL., BLOOD, vol. 124, no. 8, 20 June 2014 (2014-06-20), pages 1221 - 31
CAI YUJIA; ZETSCHE BERND; ZHANG FENG; MIKKELSEN JACOB GIEHM: "Targeted genome editing by lentiviral protein transduction of ZFN and Cas9 proteins", HUMAN GENE THERAPY, vol. 25, no. 11, 1 November 2014 (2014-11-01), pages A31, XP008182536
YUJIA CAI; RASMUS O BAK; JACOB GIEHM MIKKELSEN: "Targeted genome editing by lentiviral protein transduction of zinc-finger and TAL-effector nucleases", ELIFE,, 24 April 2014 (2014-04-24), pages 1 - 19, XP002772303
KRISTIAN ALSBJERG SKIPPER, JACOB GIEHM MIKKELSEN: "Delivering the Goods for genome Engineering and Editing", HUMAN GENE THERAPY, vol. 26, no. 8, 2015, pages 486 - 496, XP055847858
HOCHSTRASSER M L; TAYLOR D W; BHAT P; GUEGLER C K; STERNBERG S H; NOGALES E; DOUDNA J A: "CasA mediates Cas3-catalyzed target degradation during CRISPR RNA-guided interference", PNAS, vol. 111, no. 18, 2014, pages 6618 - 6623, XP055752880
ZURIS JOHN A. ET AL: "Cationic lipid-mediated delivery of proteins enables efficient protein-based genome editing in vitro and in vivo", NATURE BIOTECHNOLOGY, NATURE PUBLISHING GROUP US, NEW YORK, vol. 33, no. 1, 1 January 2015 (2015-01-01), New York, pages 73 - 80, XP093037310, ISSN: 1087-0156, DOI: 10.1038/nbt.3081
KRZYSZTOF CHYLINSKI ET AL: "Classification and evolution of type II CRISPR-Cas systems", NUCLEIC ACIDS RESEARCH, OXFORD UNIVERSITY PRESS, GB, vol. 42, no. 10, 2 June 2014 (2014-06-02), GB , pages 6091 - 6105, XP055484452, ISSN: 0305-1048, DOI: 10.1093/nar/gku241
BERND ZETSCHE ET AL: "Cpf1 Is a Single RNA-Guided Endonuclease of a Class 2 CRISPR-Cas System", CELL, ELSEVIER, AMSTERDAM NL, vol. 163, no. 3, 1 October 2015 (2015-10-01), Amsterdam NL , pages 759 - 771, XP055267511, ISSN: 0092-8674, DOI: 10.1016/j.cell.2015.09.038
T AOKI ET AL., PROTEIN TRANSDUCTION BY PSEUDOTYPED LENTIVIRUS-LIKE NANOPARTICLES, 31 March 2011 (2011-03-31)
CAI YUJIA; ZETSCHE BERND; ZHANG FENG; MIKKELSEN JACOB GIEHM: "Targeted genome editing by lentiviral protein transduction of ZFN and Cas9 proteins", HUMAN GENE THERAPY, MARY ANN LIEBERT, INC. PUBLISHERS, GB, vol. 25, no. 11, 1 November 2014 (2014-11-01), GB , pages A31, XP008182536, ISSN: 1043-0342
JOHN A. ZURIS ET AL.: "Efficient Delivery of Genome-Editing Proteins In Vitro and In Vivo", NAT BIOTECHNOL., vol. 33, no. 1, January 2015 (2015-01-01), pages 73 - 80, XP055562063, DOI: 10.1038/nbt.3081
SINKUNAS TOMAS ET AL: "Cas3 is a single-stranded DNA nuclease and ATP-dependent helicase in the CRISPR/Cas immune system", EMBO (EUROPEAN MOLECULAR BIOLOGY ORGANIZATION) JOURNAL, vol. 30, no. 7, 1 April 2011 (2011-04-01), pages 1335 - 1342, XP002765626, DOI: 10.1038/emboj.2011.41
ROGHIYH ALIYARI; SHOU‐WEI DING: "RNA‐based viral immunity initiated by the Dicer family of host immune receptors", IMMUNOLOGICAL REVIEWS, WILEY-BLACKWELL PUBLISHING, INC., US, vol. 227, no. 1, 19 December 2008 (2008-12-19), US , pages 176 - 188, XP071455353, ISSN: 0105-2896, DOI: 10.1111/j.1600-065X.2008.00722.x
G. GASIUNAS ET AL: "Cas9-crRNA ribonucleoprotein complex mediates specific DNA cleavage for adaptive immunity in bacteria", PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES, NATIONAL ACADEMY OF SCIENCES, vol. 109, no. 39, 25 September 2012 (2012-09-25), pages E2579 - E2586, XP055068588, ISSN: 0027-8424, DOI: 10.1073/pnas.1208507109
ROBERT HELER ET AL: "Cas9 specifies functional viral targets during CRISPR–Cas adaptation", NATURE, NATURE PUBLISHING GROUP UK, LONDON, vol. 519, no. 7542, 1 March 2015 (2015-03-01), London, pages 199 - 202, XP055703544, ISSN: 0028-0836, DOI: 10.1038/nature14245
KEVIN M ESVELT ET AL: "Orthogonal Cas9 proteins for RNA-guided gene regulation and editing", NATURE METHODS, NATURE PUB. GROUP, vol. 10, no. 11, 29 September 2013 (2013-09-29), pages 1116 - 1121, XP055128928, ISSN: 15487091, DOI: 10.1038/nmeth.2681
KRZYSZTOF CHYLINSKI ET AL: "The tracrRNA and Cas9 families of type II CRISPR-Cas immunity systems", RNA BIOLOGY, LANDES BIOSCIENCE, vol. 10, no. 5, 1 May 2013 (2013-05-01), pages 726 - 737, XP055116068, ISSN: 15476286, DOI: 10.4161/rna.24321
ZEISCHE ET AL., CELL, 2015, Retrieved from the Internet
AOKI T ET AL: "Protein transduction by pseudotyped lentivirus-like nanoparticles", GENE THERAPY, NATURE PUBLISHING GROUP, LONDON, GB, vol. 18, no. 9, 31 March 2011 (2011-03-31), GB , pages 936 - 941, XP037772530, ISSN: 0969-7128, DOI: 10.1038/gt.2011.38
YUJIA CAI ET AL: "Targeted genome editing by lentiviral protein transduction of zinc-finger and TAL-effector nucleases.", ELIFE, ELIFE SCIENCES PUBLICATIONS LTD., GB, vol. 3, 24 April 2014 (2014-04-24), GB , pages 1 - 19, XP002772303, ISSN: 2050-084X, DOI: 10.7554/eLife.01911
Attorney, Agent or Firm:
CABINET NONY (FR)
Download PDF:
Claims:
CLAIMS

1. A virus-derived particle comprising one or more Cas protein(s). 2. The virus-derived particle according to claim 1, wherein the said Cas protein is Cas9 or an homolog or a derivative thereof.

3. The virus-derived particle according to any one of claims 1 and 2, further comprising one or more CRISPR-Cas system guide RNA(s).

4. The virus-derived particle according to any one of claims 1 and 2, further being complexed with one or more CRISPR-Cas system guide RNA(s).

5. The virus-derived particle according to any one of claims 1 to 4, further comprising, or further being complexed with, two CRISPR-Cas system guide RNAs that both hybridize with a target nucleic acid, respectively:

- a first CRISPR-Cas system guide RNA that hybridizes with a first target sequence of the said target nucleic acid, and

- a second CRISPR-Cas system guide RNA that hybridizes with a second target sequence of the said target nucleic acid.

6. The virus-derived particle according to any one of claims 1 to 5, which is further complexed with a targeting nucleic acid comprising at least:

- a first sequence that hybridizes with a first target sequence of a selected target nucleic acid, and

- a second sequence that hybridizes with a second target sequence of the said selected target nucleic acid.

7. The virus-derived particle according to any one of claims 1 to 6, which is a retrovirus- derived particle.

8. The virus-derived particle according to claim 7, which is a lentivirus-derived particle.

9. The virus-derived particle according to claim 1, which is selected in a group comprising Moloney murine leukemia virus-derived vector particles, Bovine immunodeficiency virus-derived particles, Simian immunodeficiency virus-derived vector particles, Feline immunodeficiency virus-derived vector particles, Human immunodeficiency virus-derived vector particles, Equine infection anemia virus-derived vector particles, Caprine arthritis encephalitis virus-derived vector particle, Baboon endogenous virus-derived vector particles, Rabies virus- derived vector particles, Influenza virus-derived vector particles, Norovirus-derived vector particles, Respiratory syncytial virus-derived vector particles, Hepatitis A virus-derived vector particles, Hepatitis B virus-derived vector particles, Hepatitis E virus-derived vector particles, Newcastle disease virus-derived vector particles, Norwalk virus-derived vector particles, Parvovirus-derived vector particles, Papillomavirus-derived vector particles, Yeast retrotransposon-derived vector particles, Measles virus-derived vector particles, and bacteriophage-derived vector particles. 10. A composition for altering a target nucleic acid in a eukaryotic cell, which composition comprises at least one virus-derived particle as defined in claim 1.

11. The composition according to claim 10, further comprising one or more CRISPR- Cas system guide RNA(s).

12. The composition according to claim 11, wherein the said one or more CRISPR-Cas system guide RNA(s) is(are) comprised in virus-derived particles.

13. The composition according to claim 10, further comprising one or more transduction helper compounds.

14. A kit for preparing virus-derived particles for altering a target nucleic acid in a eukaryotic cell comprising:

- a nucleic acid comprising an expression cassette encoding a GAG-Cas fusion protein, and - a nucleic acid comprising one or more expression cassette(s) encoding virus-like assembly protein(s),

15. The kit according to claim 14, further comprising a nucleic acid comprising an expression cassette encoding a pseudotyping viral envelope protein.

16. The kit according to any one of claims 9 and 10, wherein the virus-derived assembly protein is a virus-derived GAG protein.

17. The kit according to claim 16, wherein GAG is encoded by an expression cassette selected in a group comprising an expression cassette encoding a GAG-PRO-POL polyprotein and an expression cassette encoding a GAG protein.

18. The kit according to claim 14, further comprising one or more nucleic acid(s) encoding a CRISPR-Cas system guide RNA

19. The kit according to any one of claims 14 to 18, wherein the said nucleic acids are localized in an eukaryotic cell as a result of its transfection into the said eukaryotic cell.

20. The kit according to claim 19, wherein the said eukaryotic cell consists of a packaging cell line.

21. A cell line for producing a viral particle according to claim 1 , comprising:

- one or more nucleic acids encoding the proteins required for forming the said virus- derived particle, and

- a nucleic acid comprising an expression cassette encoding a GAG-Cas fusion protein

22. An in vitro or ex vivo method for altering a target nucleic acid comprising at least a target sequence in an eukaryotic cell, comprising the steps of:

a) bringing into contact the said eukaryotic cell with virus-derived particles as defined in any one of claims 1 to 9 or with a composition as defined in any one of claims 10 to 13, and b) collecting the said eukaryotic cell having an altered target nucleic acid.

23. A fusion protein comprising (i) a viral protein that self assembles for generating a virus-derived particle, the said viral protein being fused to (ii) a Cas protein.

24. The fusion protein according to claim 23, further comprising a cleavable site located between the said viral protein and the said Cas protein.

25. The fusion protein according to any one of claims 23 and 24 consisting of a GAG- Cas fusion protein, preferably a GAG-Cas9 fusion protein.

26. A nucleic acid encoding a fusion protein according to any one of claims 23 to 25.

Description:
TITLE

METHODS AND PRODUCTS FOR GENETIC ENGINEERING

FIELD OF THE INVENTION

The present invention relates to the field of gene targeting by methods using viral-derived vector systems related to Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR) and components thereof.

BACKGROUND OF THE INVENTION

Genome editing using targetable nucleases is an emerging technology for the precise genome modification of organisms ranging from bacteria to plants and animals, including humans. Its attraction is that it can be used for almost all organisms in which targeted genome modification has not been possible with other kinds of methods. Improving protocols for expressing exogenous proteins within human cells is of major interest for research and medical purposes. In spite of the constant evolution of trans fection methods and performances of viral vectors, the efficiency of these approaches can vary dramatically, especially in primary cells that are highly sensitive to modifications of their environment and may be altered in response to transfection agents/vectors. Moreover, delivering genetic information through the transfer of a coding integrative/non-integrative DNA may be responsible for adverse effects like the induction of unwanted stress signals or the unexpected insertion of an exogenous gene within the cellular genome, which is a serious issue for therapeutic applications, particularly in stem cells. Recent approaches to targeted genome modification - zinc-finger nucleases (ZFNs) and transcription-activator like effector nucleases (TALENs) - have enabled researchers to generate permanent mutations by introducing double-stranded breaks to activate repair pathways. The capacity of designed nucleases, like ZFN and TALENs, to generate DNA double-stranded breaks at desired positions in the genome has created optimism for therapeutic translation of locus- directed genome engineering. However, these approaches are costly and time-consuming to engineer, limiting their widespread use, particularly for large scale, high- throughput studies.

More recently, a new tool based on a totally distinct and specific system, namely bacterial CRISPR-associated protein-9 nuclease (Cas9) from Streptococcus pyogenes has generated considerable interest.

To achieve site-specific DNA recognition and cleavage, Cas9 must be complexed with both a crRNA and a separate trans-activating crRNA (tracrRNA or trRNA), that is partially complementary to the crRNA (11). The tracrRNA is required for crRNA maturation from a primary transcript encoding multiple pre-crRNAs.

During the cleavage of target DNA, the HNH and RuvC-like nuclease domains cut both DNA strands, generating double-stranded breaks (DSBs) at sites defined by a 20-nucleotide guide sequence within an associated crRNA transcript that base pairs with the target DNA sequence. The HNH domain cleaves the target DNA strand that is complementary to the guide RNA, while the RuvC domain cleaves the non-complementary strand. The double-stranded endonuclease activity of Cas9 also requires that a short conserved sequence, (2-5 nts) known as protospacer- associated motif (P AM), follows immediately 3'- of the crRNA complementary sequence. The simplicity of the type II CRISPR nuclease, with only three required components (Cas9 along with the crRNA and trRNA) made this system amenable to adaptation for genome editing. This potential was realized in 2012 by the Doudna and Charpentier laboratories (Jinek et al., 2012, Science, Vol.337: 816-821). Based on the type II CRISPR system described previously, a simplified two-component system was developed by combining trRNA and crRNA into a single synthetic single guide RNA (sgRNA). The sgRNA-programmed Cas9 was shown to be as effective as Cas9 programmed with separate trRNA and crRNA in guiding targeted gene alterations.

Mainly, three different variants of the Cas9 nuclease have been adopted in genome-editing protocols. The first is wild-type Cas9, which can site-specifically cleave double-stranded DNA, resulting in the activation of the double-strand break (DSB) repair machinery. DSBs can be repaired by the cellular Non-Homologous End Joining (NHEJ) pathway (Overballe-Petersen et al., 2013, Proc Natl Acad Sci USA, Vol. 110: 19860-19865), resulting in insertions and/or deletions (indels) which disrupt the targeted locus. Alternatively, if a donor template with homology to the targeted locus is supplied, the DSB may be repaired by the homology-directed repair (HDR) pathway allowing for precise replacement mutations to be made (Overballe- Petersen et al., 2013, Proc Natl Acad Sci USA, Vol. 110: 19860-19865; Gong et al., 2005, Nat. Struct Mol Biol, Vol. 12: 304-312). Cong and colleagues (Cong et al., 2013, Science, Vol. 339: 819-823) took the Cas9 system a step further towards increased precision by developing a mutant form, known as Cas9D 1 OA, with only nickase activity. This means that Cas9D10A cleaves only one DNA strand, and does not activate NHEJ. Instead, when provided with a homologous repair template, DNA repairs are conducted via the high-fidelity HDR pathway only, resulting in reduced indel mutations (Cong et al., 2013, Science, Vol. 339: 819-823; Jinek et al., 2012, Science, Vol.337: 816-821; Qi et al., 2013 Cell, Vol. 152: 1173-1183). Cas9D10A is even more appealing in terms of target specificity when loci are targeted by paired Cas9 complexes designed to generate adjacent DNA nicks (Ran et al., 2013, Cell, Vol. 154: 1380-1389).

The third variant is a nuclease-deficient Cas9 (Qi et al., 2013 Cell, Vol. 152: 1173-1183). Mutations H840A in the HNH domain and D10A in the RuvC domain inactivate cleavage activity, but do not prevent DNA binding. Therefore, this variant can be used to target in a sequence-specific manner any region of the genome without cleavage. Instead, by fusing with various effector domains, dCas9 can be used either as a gene silencing or activation tools. Furthermore, it can be used as a visualization tool by coupling the guide RNA or the Cas9 protein to a fluorophore or a fluorescent protein.

Following its initial demonstration in 2012 (9), the CRISPR/Cas9 system has been widely adopted by the scientific community. It has already been successfully used to target important genes in many cell lines and organisms, including human (Mali et al., 2013, Science, Vol. 339: 823-826), bacteria (Fabre et al., 2014, PLoS Negl. Trop. Dis., Vol. 8:e2671.), zebrafish (Hwang et al., 2013, PLoS One, Vol. 8:e68708.), C. elegans (Hai et al., 2014 Cell Res. doi: 10.1038/cr.2014.11.), bacteria (Fabre et al., 2014, PLoS Negl. Trop. Dis., Vol. 8:e2671.), plants (Mali et al., 2013, Science, Vol. 339: 823-826), Xenopus tropicalis (Guo et al., 2014, Development, Vol. 141 : 707- 714.), yeast (DiCarlo et al., 2013, Nucleic Acids Res., Vol. 41 : 4336-4343.), Drosophila (Gratz et al., 2014 Genetics, doi: 10.1534/genetics.H3.160713), monkeys (Niu et al., 2014, Cell, Vol. 156: 836-843.), rabbits (Yang et al., 2014, J. Mol. Cell Biol., Vol. 6: 97-99.), pigs (Hai et al., 2014, Cell Res. doi: 10.1038/cr.2014.11.), rats (Ma et al., 2014, Cell Res., Vol. 24: 122-125.) and mice (Mashiko et al., 2014, Dev. Growth Differ. Vol. 56: 122-129.). Several groups have now taken advantage of this method to introduce single point mutations (deletions or insertions) in a particular target gene, via a single gRNA. Using a pair of gRNA-directed Cas9 nucleases instead, it is also possible to induce large deletions or genomic rearrangements, such as inversions or translocations. A recent exciting development is the use of the dCas9 version of the CRISPR/Cas9 system to target protein domains for transcriptional regulation, epigenetic modification, and microscopic visualization of specific genome loci.

The CRISPR/Cas9 system requires only the redesign of the crRNA to change target specificity. This contrasts with other genome editing tools, including zinc finger and TALENs, where redesign of the protein-DNA interface is required. Furthermore, CRISPR/Cas9 enables rapid genome-wide interrogation of gene function by generating large gRNA libraries for genomic screening. Thus, the CRISPR/Cas9 technology can be easily adapted to any gene of interest and may offer unchallenged possibilities to alter genes (knock-out, knock- in, introduction of precise mutations). Its spread in the scientific community is amazingly rapid and has triggered a recent burst of scientific communications using it. CRISPR's delivery is commonly performed by DNA transfection or through the use of viral vectors encoding Cas9, both methods being convenient but limited to certain cell types as well as being rather intrusive. Furthermore, maintenance of Cas9 expression for a long period is possibly toxic and at best not necessary, since Cas9-mediated cleavage occurs rapidly ( Jinek et al., 2013, eLife, Vol. 2, e00471) and could even be toxic on long term. Other approaches have succeeded in exploiting recombinant Cas9 and synthetic RNAs to transfer the RNPc by Proteo transfection or by physical microinjection but these CRISPRs systems remain limited to target fragile primary cells.

There is a need in the art for improved tools and methods for gene editing by using CRISPR/Cas technology.

SUMMARY OF THE INVENTION

The present invention relates to products and methods for generating alterations in genomic nucleic acids; which alterations encompass mutations by introduction of nucleic acid insertion and nucleic acid deletion, which include knock-in and knock-out genomic alterations.

More precisely, this invention relates to products aimed at generating nucleic acid alteration events caused by CRISPR-Cas complexes, and especially caused by CRISPR-Cas9 complexes, as well as to methods using the same. This invention relates to a virus-derived particle comprising one or more Cas protein(s), and especially Cas9 protein.

In some embodiments, the said virus-derived particle further comprises, or is further complexed with, one or more CRISPR-Cas system guide RNA(s).

In some embodiments, the said virus-derived particle further comprises, or is further complexed with a targeting nucleic acid.

In some embodiments, the said virus-derived particle is a retrovirus-derived particle, e.g. a lentivirus-derived vector particle.

This invention further pertains to a composition for altering a target nucleic acid in a eukaryotic cell, which composition comprises a virus-derived particle comprising one or more Cas protein(s), and especially Cas9 protein.

In some embodiments, the said composition further comprises, or alternatively is further complexed with, one or more CRISPR-Cas system guide RNA(s).

In some embodiments, the said composition further comprises a targeting nucleic acid. This invention also concerns a kit comprising the required substances for preparing a virus-derived particle or a composition as defined above.

It also relates to genetically modified cells producing virus-derived particles as defined herein, especially cells which are under the form of stable cell lines.

This invention further relates to a fusion protein comprising (i) a viral protein that self assembles for generating a virus-derived particle, the said viral protein being fused to (ii) a Cas protein. In some embodiments, the said fusion protein comprises a cleavable site located between the said viral protein and the said Cas protein, and especially a cleavable site located between a Gag protein and a Cas9 protein.

It also pertains to nucleic acids and vector encoding the said fusion protein.

BRIEF DESCRIPTION OF THE DRAWINGS

Figurel: Molecular Basis of Cas9-VLP assembly and transfer of CRISPRs- components into recipient cells.

(A) Schematic representation of Cas9-VLPs assembly from HEK293T cells. 6 Steps are depicted:

(1) GAG-CAS9, GAG ProPol and a viral envelope protein are transfected in HEK293T cells in association with a construct encoding a guide RNA. GAG and the viral envelope tend to localize at the membrane where the assembly of a virus-derived particle (which may be also termed "Virus-Like Particle" or "VLP" herein) takes place (2). As concentration of GAG increases, mechanical forces induce the formation of a particle (3) that will bud from the producer cell after having incorporated all the actors of the CRISPR machinery (4). These particles can be concentrated and are stable at 4°C more than 15 days. Due to the maturation process, the viral protease may have released most Cas9 proteins from the GAG platform within the particle (5). Exposed to target cells, VLPs will be able to bind and fuse the with cell membrane through an envelope/receptor interaction that thus depends on the envelope used to pseudotype particles and the considered target cell. After fusion with the cellular membrane, VLPs transfer their cargo within recipient cells which may include Cas9/gRNAs ribonucleocomplexes, free gRNAs or Cas9 possibly associated with non-protease GAG (not yet clarified). Fully active CRISPRs RNPc are nevertheless delivered into the nucleus of recipient cells (possibly due to a re-association of Cas9 and free gRNAs within the target cell) and mediate the cleavage of genomic-DNA in the very position specified by the gRNA (6).

(B) Molecular Design of the GAG-Cas9 coding construct. hCMVp (human cytomegalovirus early promoter) drives the expression of a mRNA incorporating an intron (rBG) and a poly adenylation (rBGpA) signal both deriving from the rabbit beta globin gene sequence. The construct consists of the fusion of the MLV-GAG polyprotein with the codon-optimized Cas9 sequence from Streptococcus pyogenes. Both moieties are separated by a MLV protease cleavage site (ps) and a flag-tag sequence fused to the Cas9 sequence. Figure 2: Molecular validation of genetic cleavage by YFP-CRISPRs-VLPs

(A) Localization of gRNAs recognition sites on the YFP gene, primers used and principle of the surveyor assay. L929 murine cells were lentitransduced with a YFP coding vector at low Multiplicity Of Infection (MOI). After 72h, cells were next treated with VLPs loaded with gRNAs targeting the YFP gene at two different positions as indicated. To ascertain the cleavage of YFP, cells treated by Cas9-VLPs were lysed and genomic DNAs were extracted and analyzed by the S 1 nuclease-based surveyor assay. The test detects heteroduplexes that are formed if two closely related ssDNA molecules hybridizes: SI nuclease digestion is thus a proof that DNA was cleaved by Cas9.

(B) Surveyor assay on L929 cells treated by VLPs loaded with gRNAs2, 1, or a combination of both of them. Formation of heteroduplexes is detected for each rRNAs condition, revealing truncated versions of YFP whose size depends on the position of the gRNA on the YFP sequence.

Figure 3: CRISPR-mediated disruption of YFP in L929 murine cells by Cas9-VLPs. (A) A guide RNA was raised to target YFP downstream the initiation codon ATG. After cleavage cells could repair this gap by NHEJ, a mechanism that could create small deletions and indels, a scar that could alter the YFP frame in some cells. 3 different versions of repaired YFP could thus be generated by the natural reparation machinery, only one of them restauring an in- frame YFP and a maintenance of the YFP phenotype. Accordingly, cleavage should induce a detectable but incomplete decrease of YFP in target cells.

(B) VLPs loaded with Cas9 and a guide RNAs targeting YFP were produced and directly introduced into the medium of YFP-L929 cells, a murine fibroblastic cell line# where YFP was stably integrated into the genome by lentivector transduction. Envelope-less VLPs were also produced and used as a negative control. 72h after treatment cells were analyzed by flow cytometry (FACS) and global Mean Fluorescence Intensity (MFI) was monitored revealing the strong effect of YFP-breaking Cas9-VLPs on YFP L929 target cells. A 7-fold decrease of MFI was measured for cells treated by enveloped VLPs as compared with non treated YFP-cells or treated by env less VLPs*. We show here that Cas9-VLPs loaded with a specific guide RNA can be used without further concentration/purification process as CRISPR delivery agents.

Figure 4: Deletion of the Myd88 locus using conventional Cas9 delivery methods vs Cas9 VLPs

(A) Schematic representation of the Myd88 genomic DNA and localization of the different tools used for the Myd88 cleaving assay. Two different CRISPRs sequences were designed against the human Myd88 gene as represented in purple and green. Grey boxes correspond to the regions where the two PCR primers hybridize. They have been optimized for the amplification of gMyd88 which generate an amplicon size of 420 nts. Should the CRISPRs system be active in target cells, a deletion of 160 nts occurs in some cells and can be repaired by NHEJ which gives rises to a 260nts-in size truncated version of the gene.

(B) PCR-amplification of gMyd88 in wt-HEK or after trans fection with both Myd88 CRISPRs and a Cas9 encoding plasmid. After extraction of genomic DNA, a PCR-assay was performed using the Myd88 primer set. In the population treated with CRISPRs components, two amplicons are generated corresponding to the uncleaved form of Myd88 (or a version which has been cleaved by a single CRISPR) and a double-cut version after cleavage of the gene at the two targeted positions. We can note that the Myd88 deletion does not affect all treated cells, indicating that the cleavage mediated by the trans fected CRISPR components system is not complete.

(C) We also attempted to deliver both Myd88 guide RNAs by Cas9 loaded VLPs. For this VLPs were produced following the procedure described in figure 1 and different experimental procedures were explored, varying for the ratio of plasmid used and the nature of the envelope (R1-R4). After collecting and concentrating them, VLPs were introduced into the medium of HEK or Hela cells and efficiency of the Myd88 cleavage was next assessed by PCR.

(D) Myd88 amplification using the Myd88 primer set and the genomic DNA extracted from cells treated by VLPs. While all preparations were equally efficient in cleaving Myd88 in HEK cells (left panel), some differences can be appreciated in Hela cells, reflecting the importance of the envelope/ratio used. A particular protocol seems optimal (Rl) for both cell types. Figure 5: Dose dependent cleavage of the Myd88 gene induced by Cas9-VLPs

Cas9-VLPs loaded with two gRNAs targeting the Myd88 gene were produced, concentrated and stored 15 days at 4°C. Increasing amount of VLPs were next added to the medium of HEK293T target cells plated in a 12-w plate (150000 cells/w). 20h after VLP treatment, cells were lysed and genomic DNA was purified to analyze the genetic cleavage of the Myd88 locus. The signal revealing the deletion of Myd88 (260 nts) increases with the amount of VLPs introduced in the medium.

This shows that Cas9-VLPs are active in target cells less than 24h after their introduction. We also noted that the VLP preparation can be stable at least 15 days at 4°C. Figure 6: Cleavage of the Myd88 gene in HEK targets by Cas9-VLPs is potentiated by polybrene.

Suboptimal doses of Cas9-VLPs loaded with two gRNAs targeting the Myd88 gene were introduced in the medium of 3xl0e6 HEK target cells grown in complete medium supplemented or not with Hexadimethrine bromide (polybrene), a polycation favoring the contact of particles with target cells. 48h after VLP treatment, cells were lysed and genomic DNA was purified to analyze the genetic cleavage of the Myd88 locus. In this condition where under-saturating amounts of VLPs were used, the Myd-88 cleavage is undetectable in cells cultivated in a standard medium but is strongly potentiated by polybrene addition. Figure 7: Cleavage of the Myd88 gene induced by Cas9-VLPs in human monocytes- derived macrophages

Genotype

(A) Cas9-VLPs loaded with two gRNAs targeting the Myd88 gene were concentrated and introduced in the medium of human monocytes-derived macrophages after 6 days of differentiation with Granulocyte-Macrophage Colony Stimulating Factor (GMCSF) (100000 cells per well i a 48-w plate). 48h after treatment with VLPs, cells were lysed and genomic DNA was purified to analyze the genetic cleavage of the Myd88 locus. Under this condition, the Myd88 gene is cleaved with such high efficiency after treatment with VLPs that the wt sequence cannot even be detected by conventional PCR thus suggesting an almost complete cleavage mediated by Myd88-VLPs in primary non dividing cells.

Phenotype

(B) According to Lombardo et al*, human macrophages massively die by apopotosis when cultivated without GMCSF, unless they are stimulated by a TLR-4 agonist like LPS. This resistance to apoptosis is LPS- and Myd88-dependent. To check wether VLPs treated cells lost their Myd88 function, we cultivated them without GMCSF (in RPMI medium) and stimulated them with LPS during 72h. Upon this treatment WT macrophages resisted to GMCSF deprivation (compare condition 2 to condition 3) while macrophages treated with Myd88-cleaving Cas9-VLPs died massively (condition 4). This strongly suggests that VLPs treatment inactivated the Myd88 at the functional level.

Figure 8: Cleavage of the Myd88 gene induced by Cas9-VLPs in freshly-purified primary human lymphocytes.

Human purified lymphocytes (Ficoll/percoll) were plated at 40x10e6 cells/ml in 200ul in a 48w plate. Cells were next treated with a fresh preparation of Cas9-VLPs targeting Myd88 and an older one kept 20 days at 4°C, in a transduction medium supplemented with polybrene (4ug/ml). After 2h, 500μ1 of fresh medium was added to the transduction medium and cells where maintained in culture for 40h before their lysis and genomic DNA extraction. Cleavage of Myd88 was next investigated by PCR revealing the wt or the cleaved form of Myd88 . In both VLP conditions, the Myd88 gene was cleaved. 1 million of quiescent lymphocytes were genetically modified in less than 48h with a single treatment of VLPs without any apparent toxicity.

Figure 9: Cleavage of the Myd88 gene induced by Cas9-VLPs in murine bone marrow-derived macrophages

Macrophages were differentiated from bone marrow cells flushed-out from mouse femurs incubated during 8 days in MCSF containing medium. Cells were next treated with a fresh preparation of Cas9-VLPs targeting mMyd88 in a medium supplemented with polybrene (4ug/ml). It is noteworthy that two new gRNAs were designed for this experiment that specifically targeted the murine gene. Cells were cultivated 48h before lysis and genomic DNA extraction. Cleavage of Myd88 was next investigated by PCR revealing the wt or the cleaved form of the murine Myd88 gene based on the design of the hMyd88 assay. A very efficient cleavage was detected by PCR in VLP- treated cells while bands corresponding to the complete or partially cleaved mMyd88 (by only one gRNA) appear faint.

Figure 10: Targeted insertion of a flag-tag sequence in the endogenous DDX3 genomic locus mediated by 'all in one' Cas9-VLPs

(A) schematic representation of the human DDX3 genomic locus and the different tools used in the experiment. The purple arrow represents the locus cleaved by the DDX3 CRISPR and the grey region an intronic region of DDX3. The FLAG IN rep primer (ssODN) is represented as a single strand DNA exhibiting 40nt homology-repair arms, flanking the Flag-tag sequence, that are homologous to the DDX3 locus. Primers used in the PCR assay are represented. (B) Principle of the 'all in one' VLPs delivering the Cas9 protein, the gRNAs and the repair primer. VLPs targeting DDX3 were produced, centrifuged and stored at 4°C. VLPs were then combined with increasing doses of ssODNs and complexes added on HEK target cells cultivated in classic growing medium. Target cells were next lysed 72 h later for preparation of protein extracts and genomic DNA (C) Western-Blot analysis of cells treated with 'all in one' DDX3 VLPs. A Western-blot signal revealed by the flag antibody and corresponding to the DDX3 expected molecular weight (86KDa) can be detected at the highest concentrations of ssODNs. This indicates that the Flag sequence was successfully inserted at the DDX3 locus of VLP -treated cells. This was further confirmed by PCR on the genomic DNA extracted from VLP -treated cells. Primers used (depicted in A) should amplify a DNA segment only if the Flag sequence is inserted in the DDX3 gene since the forward primer hybridizes to the Flag sequence and the reverse primer hybridizes to the intron of DDX3). As shown in lower panels, the genetic modification is obvious for higher concentration of primers and decreases with the dose but remains detectable -at the DNA level- for a concentration as low as 0,01nmol/ml (lane 4). (D) Introduction of the Flag sequence upstream the endogenous locus of DDX3 in human monocyte-derived dendritic cells (Mo-derived DCs). VLPs and ssODN (5nmol/ul final) were complexed and the mix used to treat Mo-derived DCs in a transduction medium containing polybrene (4ug/ml). Genomic DNA analysis indicates that the flag sequence was succesfully engrafted into the endogenous locus of DDX3 in human primary DCs by a single treatment of VLPs.

Figure 11: Panel of possibilities using Nanoblades and their association with 'helper' VLPs We have shown that 'all in one' VLPs incorporating Cas9, gRNAs and possibly combined with a reparation ssDNA can be generated and deliver the complete package in recipient cells. This agent depicted in (a) is highly versatile in itself. Since VLPs can incorporate several gRNAs and may certainly be complexed after production with different reparation primers, many possibilities are offered to a scientist/company to create customized tools at low cost. As referenced in (Abe et al. J Virol 1998), VLPs of different nature may be complexed outside the cell after production and may complement each other, one helping the other to enter into the cell for example. Given this property of VLPs, we may imagine other ways to prepare an active agent to transfer the actors of the CRISPRs system by mixing particles, each of them being dedicated to a particular cargo. In (b) is proposed a system where could be mixed gRNA-Cas9-VLP and particles simply complexed by the reparation primer: the mixture of both types of particles would be in this case, the final active agent. To go further, gRNAs could also be packaged in a particular type of particle and combined after production with unloaded Cas9-VLPs to create a particle mixture able to deliver all the components. This system is depicted in (c). Considering the theoretical possibility to incorporate several gRNAs in VLPs, to choose different viral envelope to pseudotype each type of VLPs, and to associate them with different types of ssDNA, we may even imagine more complex agents, (d). While this segregation of CRISPRs components in different types of particles may certainly affect the global efficiency of the final agent, it may offer to a company producing nanoblades a vast panel of possibilities to improve the nanoblade service and render it costless. Should the (c) system be efficient enough, it only requires the preparation of a large well-titered batch of generic Cas9-VLPs to be associated with gRNAs particles customized specifically for each application. This system appears highly valuable from the industrial point of view, since a very precise molecular service is offered, which only necessitate the rapid costless preparation of the gRNAs- VLPs.

Figure 12: Western blot analysis and characterization of CAS9 Virus-derived particles separated on a discontinuous sucrose gradient.

Figure 12A illustrates a Western blot gel electrophoresis. Lanes from left to right: (i) incubation with anti-Flag antibodies; (ii) incubation with anti-VSV-G antibodies; (iii) incubation with anti- CAS9 antibodies; (iv) incubation with anti-GAGmlv antibodies.

Figure 12B illustrates a dot blots of fractions 1 to 24 collected after performing the separation of CAS9 virus-derived particles on a discontinuous sucrose gradient. Columns from left to right: fractions n° 1 to n°24. Lanes from the upper part to the lower part of Figure 12B: (i) incubation with anti-VSV-G antibodies; (ii) incubation with anti-CAS9 antibodies, (iii) incubation with anti- GAGmlv antibodies.

Figure 13 Gag/Cas9 fusion actively loads guideRNAs within Virus-like particles (VLPs).

Northern blot directed against the conserved region of the guideRNA using total RNA extracted from producer cells (lanes 2 to 4) or the corresponding purified VLPs (lanes 5 to 7).

Figure 14

Figure 14 A: Schematic representation of the coding cassettes designed for the production of MLV-based VLPs or HIV-1- based VLPs. Both cassettes were incorporated in an eucaryotic expression vector equiped with the early hCMV promoter, the rabbit-Bglobin intron and the rabbit pA signal. Both systems were optimized by exploration and test of diverse proteolytic sites separating the GAG cassette from the Cas9 gene. MLV based VLPs were produced as described elsewhere while HIV-1 based VLPs were produced similarly except that an HIV-1 helper construct encoding GAG POL Tat Rev proteins was transfected instead of the MLV GAG POL plasmid. Production of HIV-1 VLPs follows the same procedure as compared with MLV-based VLPs. Figure 14B: Concentrated VLPs engineered to incorporate a guide RNA targeting the GFP gene were used to transduce 30000 HEK293T cells expressing GFP. HIV-1 and

MLV-based particles were produced with the same loaded gRNA (target sequence: CGAGGAGCTGTTCACCGGGG - SEQ ID NO. 38). Recipient cells were plated the day before in a 96-w plate. Transduction medium was supplemented with polybrene (4ug/ml). 72 hours after treatment with 3 increasing doses of each VLP-batch, fluorescence intensities were measured by a Fluorometer (Excitation 488, Emission 535). Fluorescence decrease was evident in VLPs-treated cells as compared with control non-treated cells (C), revealing the cleavage of the GFP gene within recipient cells. Results indicate that HIV-1 based VLPs are efficient in delivering the CRISPR/CAS9 system to a level slighly less effcient than MLV-based VLPs in these recipient cells (1,5-2 fold less efficient).

Figure 14C: Cleavage of the WASP gene in primary human T cells stimulated with IL7. For this experiment, two guide RNAs targeting the human WASP gene were incorporated within HIV- 1 or MLV-based VLPs before treatment of freshly purifed T-cells stimulated with IL7. 500000 cells were plated in a 24w plate in 400ul of RPMI medium supplemented with polybrene (4ug/ml) and IL-7. Concentrated HIV-1 or MLV VLPs (lOul of VLP dosed at luM CAS9) were added in the culture medium. WASP deletion by CRISPR-CAS9 was next measured by PCR in recipient cells 24 hours after treatment. Primer used for amplification of genomic WASP gene were: forward: 5'- ATTGCGGAAGTTCCTCTTCTTACCCTG (SEQ ID NO. 36) reverse: 5'- TTCCTGGGAAGGGTGGATTATGACGGG (SEQ ID NO. 37).

PCR conditions are: 95°C 5min followed by 25 cycles of (95° 30sec-57° 30sec-72° 30 sec) followed by 5min at 72°C.

Amplicons were next loaded on a gel to reveal the state of WASP into VLP recipient T-cells: wt or cleaved. Gel analysis performed using the ImageJ software allowed a quantification of double- cutting efficiencies for MLV-based VLPs (32%) and HIV-1 -based VLPs (6%).

Figure 15: CRISPR delivery into Thyl-GFP mouse embryos by Cas9-containing virus-derived particles Figure 15A illustrates the injection of CAS-containing virus-derived particles in the zona pellucida of Thyl-GFP mouse embryos.

Figure 15B shows the results of cleaving the Thyl-GFP allele in the adult mice (F0) originating from the mouse embryos injected with CAS9-containing virus-derived particles

Figure 15C shows the alteration of the Thyl-GFP allele in the Fl mice originating from the F0 mice depicted in Figure 15B.

Figures 15 D, E, F and G: Percent of GFP alteration in mice #78, #79, #21 and #22, respectively, as calculated from chromatograms wherein the results are compared with a non-treated Thy-GFP control mouse. In abscissa: percent of GFP alteration in Fl mice.

* % is never complete due to the fact that the chosen Thyl-GFP line carries several copies of GFP/allele (6 to 10). Results should be reproduced in a mouse line bearing one single constitutive GFP copy per allele, which is under preparation

DETAILED DESCRIPTION OF THE INVENTION The present invention relates to the use of virus-derived particles to deliver CRISPR/Cas protein to target cells for generating targeted alteration(s) in the genome of an eukaryotic organism, preferably of a mammal, and especially of a human organism. Surprisingly, the inventors have shown that the generation of a site-directed genome alteration, e.g. a site directed genome deletion or a site-directed genome insertion, may be successfully performed by delivering a Cas protein to the target cells through the use of viral vector particles wherein the said Cas protein has been packaged. The present inventors have conceived a powerful method to transfer the CRISPRs active machinery within human and other mammalian cells, including primary cell types, by using versatile virus-derived particles (which are also termed "Virus Like Particles" or "VLPs" herein).

The inventors have shown that these VLPs ensure a transient and dose-dependent delivery of the CRISPR-RNPc (also termed "CRISPR-RiboNucleoProtein complex") into target cells and induce a robust and rapid cleavage of the desired targeted gene. As illustrated in the examples, when taking the Myd-88 gene as readout, the inventors have observed a complete cleavage of the latter gene in less than 6 hours in human cells, thus with a striking rapidity that may be attributed to the high efficiency of the virus-derived particles system described herein, which system comprises delivering directly a Cas protein, and most preferably a Cas9 protein, as well as CRISPR guide RNAs (also termed "gRNAs" herein) instead of performing a nucleic acid transfer of polynucleotides encoding Cas protein as it is the case for most already known CRISPRs delivery systems. As described in the examples herein, CRISPR guide RNAs are efficiently encapsulated in the CAS-containing VLPs. As it is also described in the examples, encapsulation of the CRISPR guide RNAs is highly subjected to the presence of the CAS protein in the VLPs.

The inventors have also shown that the CAS-containing VLPs may be prepared from a variety of virus-derived particles, and especially with virus-derived particles wherein the GAG protein contained therein may originate from a variety of viruses. Notably, it is described in the examples CAS-containing virus-derived particles comprising a MLV-derived GAG protein, as well as CAS- containing virus-derived particles comprising HIV 1 -derived GAG protein. It is shown herein that both kinds of CAS-containing virus-derived particles efficiently engineer a targeted gene, e.g., efficiently cleave a targeted gene. Further, the inventors have shown that the GAG-containing virus-derived particles efficiently alter desired target sequences in vivo. Illustratively, it is shown in the examples that the GAG- containing virus-derived particles may be used to induce desired genomic alterations (e.g. induce a cleavage at a desired location in the genome) in living embryos. It is also shown herein that the genomic alterations performed in the living embryos are present in the resulting adult mammal and are then transferred to the subsequent generations.

The inventors findings are of a particular importance when considering that major gene expression processes (such as transcription and translation) are less active in some primary cells subsets that may be major targets for CRISPRs strategies and could therefore decrease the efficiency of conventional delivery methods like DNA transfection and conventional lentiviral vectors.

In this regard, the Cas9-virus-derived particles technology that has been conceived by the inventors appears as a tool of choice for genome editing, especially for genome editing in non- activated, non-dividing primary cells like lymphocytes, which poorly support transfection/ transduction procedures, and display a low metabolism prior activation.

Further, according to the inventors results, the effect of the CRISPR RNPc is transient in the recipient target cell and is expected to exert its biological activity for at most a few hours after introduction of the RNPc through the contact of the virus-derived particles described herein with the target cells.

The transient delivery of CRIPSR components into target cells and the fact that this technology does not introduce plasmidic DNA in the target cells is expected to reduce the potential toxicity and also to reduce the risk of off-target cleavages.

Further, the fact that this technology does not introduce plasmidic DNA in the target cells allows avoiding the potential incorporation of exogenous-DNA in the target genome.

Illustratively, as it is shown in the examples, treatment of fragile human stem-CD34 + cells or human lymphocytes with the virus-derived particles described herein has not induced detectable cell toxicity and has not led to cell-death, even after a massive input of the said virus-derived particles. Notably, the present inventors have engineered a chimeric Cas9-protein upon fusion with the structural GAG protein of the murine leukemia virus to have the Cas9 protein packaged into MLV-derived VLPs or to HIV- 1 -derived VLPs.

This concept has thus been easily extended to other viral structural proteins like the GAG polyprotein from HIV-1 or the GAG polyprotein from Rous Sarcoma Virus (RSV) with success.

The virus-derived particles produced as described herein, are shown to efficiently transfer the CRISPR-RNPc into the desired target cells. Exploiting the technology of virus-derived particles described herein offers a large panel of viral envelopes that can be selected to pseudotype the said virus-derived particles, thus conferring particular properties to the preparation (tropism, complement resistance, robustness). Insertion of the coding sequences of a Cas protein and one or more CRISPR gRNAs in expression cassettes, and especially the coding sequences of Cas9 and a specifically designed gRNA in expression cassettes, may also be performed in the backbone of recombinant viruses like Measles or certain Influenza strains, permissive to incorporation of foreign sequences. This allows an extensive diffusion of the active CRISPR RNPc in specific cells/tissues/organs permissive to the considered virus.

Beyond the exploitation of the Cas9 Streptococcus pyogenes endonuclease, the technology described herein is easily extended to Cas proteins from other organisms that can be alternatively fused with a structural viral protein. The growing cohort of Cas9-derivatives may also be delivered by the virus-derived particles described herein so as to achieve a large variety of genome alterations, such as cleaving only one DNA-strand, activating transcription and labelling precise genomic loci. The technology described herein also allows performing a Cas-based CRISPR strategy, especially a Cas9-based CRISPR strategy, for targeting intracellular mRNAs and induce their cleavage, as described by O'Connell et al. (2014, Nature, Vol. 516: 263-266), which is a technique involving small DNA sequences (PAMmers) provided in trans. The virus- derived particles technology described herein may be adapted to this RNA targeting approach by a simple combination of particles with ssDNA PAMmers on the model of the flaging-DDX3 strategy described in the examples (See also FigurelO herein). The possibility to combine the virus-derived particles described herein, after their production, with ssDNA or even dsDNA offers vast possibilities in terms of industrial developments and rapid and costless customizations for various nucleic acid engineering purposes. Moreover, it is to note that viral-derived nanoparticles differing for their envelope or their proteic/nucleic cargo can trans-complement when combined as a mixture, as it was described in another technical context by Abe et al. (1998, J Virol, Vol. 72: 6356-6361).

A plurality of ways of combining the virus-derived particles of the invention and to transfer the CRISPRs effect into target cells are described elsewhere in the present specification. Some of these various embodiments are depicted in Figure 11 herein. Preliminary data indicate that the virus-derived particles comprising a Cas protein, especially a Cas9 protein, may be combined with vesicles incorporating one or more CRISPR gRNA(s) when these virus-derived particles and vesicles are prepared and used as taught in another technical context by Mangeot et al. (2011 , Mol Ther J Am Soc Gene Ther, Vol. 19: 1956-1666), and that the CRISPRs action is efficient in cells treated by the resulting mixture of the Cas-containing virus-derived particles and the gRNA(s)- containing vesicles. The opportunity to segregate the CRISPRs components in different types of particles may be of high interest from an industrial point of view and offers versatile technical solutions for generating the desired nucleic acid alteration(s).

The Cas-containing virus-derived particles described herein, especially the Cas9-containing virus- derived particles, may be easily produced in large amounts in the absence of gRNAs, to obtain VLP batches that are carefully dosed and quality-controlled. These Cas9-VLPs can later be combined in a custom-dependent manner with gRNA(s)-containing vesicles and/or targeting nucleic acid- containing vesicles, so as to complement the system by specific gRNAs or specific reparation template, or both.

As it is fully illustrated in the present specification, the Cas-containing virus-derived particles technology that is described herein offers new possibilities to the CRISPR community and notably upgrade the available toolbox to target challenging cell-types and explore innovative therapeutic CRISPR-based approaches for in/ex vivo gene therapy.

Thus, the inventors have successfully packaged a Cas protein into virus-derived particles by conceiving packaging cells expressing a cleavable fusion protein between (i) a viral structural protein and (ii) the said Cas protein. Thus, the present invention relates to a virus-derived particle comprising one or more Cas protein(s). As used herein, a virus-derived particle means a particle formed from the assembly of viral structural proteins which are associated so as to form the particle core that will be later enveloped with a membrane, (which virus-derived particle does not contain any nucleic acid encoding a nucleic acid or a protein of interest). Thus, in contrast to most of the virus-derived particles known in the art, which are designed for delivering expression nucleic acids in the transduced cells, a virus-derived particle as described herein is designed for delivering proteins, and optionally non- coding nucleic acids in the transduced cells, i.e. at least a Cas protein. As it is described in detail in the present specification, a virus-derived particle according to the invention may also contain one or more non-coding nucleic acids, which non-coding nucleic acids encompass CRISPR-Cas system guide RNA(s) and targeting nucleic acids. For the sake of clarity, it may arise that a virus- derived particle as described herein may contain traces of coding nucleic acids originating from the cells that are used for producing them, such as traces of mRNAs or plasmidic DNA originating from the said producing cells. The small amount of coding nucleic acids that may in some occasions be present within the virus-derived particles are generally passively encapsulated. However, it shall be clearly understood that the virus-derived particles described herein are not at all dedicated to transport any coding nucleic acid of interest but, as described in detail throughout the entire specification, these virus-derived particles are in contrast only dedicated to transport proteins, mainly one or more proteins having a Cas endonuclease activity, and in some embodiments also non-coding nucleic acids of interest, namely (i) one or more CRISPR-guide RNA(s) and/or one or more targeting nucleic acid(s).

As shown in the examples herein, the said cleavable fusion protein between (i) a viral structural protein and (ii) the said Cas protein is successfully incorporated in the virus-derived particles that are produced by the packaging cells and the resulting virus-derived particles successfully deliver the Cas protein to the target cells for altering the target cells genome through site-directed genomic DNA cleavage and, in some embodiments, also nucleic acid insertion by homologous recombination. As shown in the examples herein, the said fusion protein contributes to the formation of the virus-derived particles wherein it is associated with the viral structural proteins.

The inventors have notably shown that successful genomic alteration is obtained by using these virus-derived particles in combination with one or more CRIPSR-Cas system guide RNA(s), and in particular by using virus-derived particles further containing the said one or more CRIPSR-Cas system guide RNA(s) inside the said particles. As shown in the examples, the virus-derived particles described herein have been successfully used for disrupting or deleting various genes, both in vitro and in vivo, so as to generate organisms wherein the said various genes have been knocked-out.

As it is also shown in the examples, the virus-derived particles described herein have been successfully used for the targeted insertion of nucleic acids of interest in the genome of target cells, so as to generate knock-in organisms. As experimentally illustrated herein, the inventors have fused a Cas9 protein with the GAG protein of Murine Leukemia Virus and have used this construct to produce functional Cas9-loaded virus-derived particles delivering the Cas9 activity into recipient cells.

As further experimentally illustrated herein, the inventors have fused a Cas9 protein with the GAG protein of HIV- 1 and have used this construct to produce functional Cas9-loaded virus- derived particles delivering the Cas9 activity into recipient cells.

Moreover it is shown in the examples herein that the guide RNAs can also be incorporated successfully in virus-derived particles, creating a fully active CRISPR-RNPc within viral like particles that can be transmitted into recipient cells. The experimental results of the inventors illustrate the high efficiency of these Cas-containing virus-derived particles. These virus-derived particles are fully able to deliver CRISPRs in different cells types, including primary cells, without apparent toxicity. Cleavage efficiency of the genomic target nucleic acid is remarkably close to 100% in human naive lymphocytes simply treated by the said virus-derived particles cleaving the human (hMyd88) gene.

The present invention relates to a virus-derived particle comprising one or more Cas protein(s). Various embodiments of the virus-derived particles described in the present specification are illustrated in Figure 11.

Virus-derived particles

As used herein, a virus-derived particle consists of a virus-like particle formed by one or more virus-derived protein(s), which virus-derived particle is substantially devoid of any nucleic acid encoding a nucleic acid or a protein of interest, or alternatively is devoid of any nucleic acid encoding a nucleic acid or a protein of interest. Notably, a virus-derived particle according to the invention is substantially devoid of any nucleic acid encoding a viral nucleic acid or a viral protein of interest, or alternatively is devoid of any nucleic acid encoding a viral nucleic acid or a viral protein of interest. A virus-derived particle according to the invention is replication- incompetent.

Virus-derived particles

Any virus suitable for gene therapy may be used, including but not limited to adeno-associated virus ("AAV"); adenovirus; herpes virus; lentivirus and retrovirus. Adeno-associated virus ("AAV") may be selected in a group comprising AAV1, AAV6, AAV7, AAV8, AAV9 or rhlO, which AAV are particularly suitable for use in human subjects.

The general methods that are known in the art for producing viral vector particles, which generally contain coding nucleic acids of interest, may also be used for producing the virus- derived particles according to the present invention, which do not contain coding nucleic acids of interest.

Conventional viral vector particles encompass retroviral, lentiviral, adenoviral and adeno- associated viral vector particles that are well known in the art. For a review of various viral vector particles that may be used, the one skilled in the art may notably refer to Kushnir et al. (2012, Vaccine, Vol. 31 : 58-83), Zeltons (2013, Mol Biotechnol, Vol. 53: 92-107), Ludwig et al. (2007, Curr Opin Biotechnol, Vol. 18(n°6): 537-55) and Naskalaska et al. (2015, Vol. 64 (n°l): 3-13). Further, references to various methods using virus-derived particles for delivering proteins to cells are found by the one skilled in the art in the article of Maetzig et al. (2012, Current Gene therapy, Vol. 12: 389-409) as well as the article of Kaczmarczyk et al. (2011, Proc Natl Acad Sci USA, Vol. 108 (n° 41): 16998-17003).

Generally, a virus-derived particle that is used according to the invention, which virus-derived particle may also be termed "Virus-Like Particle" or "VLP", is formed by one or more virus- derived structural protein(s) and/or one more virus-derived envelope protein.

A virus-derived particle that is used according to the present invention is replication incompetent in a host cell wherein it has entered. In preferred embodiments, a virus-derived particle is formed by one or more retrovirus-derived structural protein(s) and optionally one or more virus-derived envelope protein(s).

In preferred embodiments, the virus-derived structural protein is a retroviral gag protein or a peptide fragment thereof. As it is known in the art, Gag and Gag/pol precursors are expressed from full length genomic RNA as polyproteins, which require proteolytic cleavage, mediated by the retroviral protease (PR), to acquire a functional conformation. Further, Gag, which is structurally conserved among the retroviruses, is composed of at least three protein units: matrix protein (MA), capsid protein (CA) and nucleocapsid protein (NC), whereas Pol consists of the retroviral protease, (PR), the retrotranscriptase (RT) and the integrase (IN).

In some embodiments, a virus-derived particle comprises a retroviral Gag protein but does not comprise a Pol protein. As it is known in the art, the host range of retroviral vector, including lentiviral vectors, may be expanded or altered by a process known as pseudotyping. Pseudotyped lentiviral vectors consist of viral vector particles bearing glycoproteins derived from other enveloped viruses. Such pseudotyped viral vector particles possess the tropism of the virus from which the glycoprotein is derived.

In some embodiments, a virus-derived particle is a pseudotyped virus-derived particle comprising one or more viral structural protein(s) or viral envelope protein(s) imparting a tropism to the said virus-derived particle for certain eukaryotic cells. A pseudotyped virus-derived particle as described herein may comprise, as the viral protein used for pseudotyping, a viral envelope protein selected in a group comprising VSV-G protein, Measles virus HA protein, Measles virus F protein, Influenza virus HA protein, Moloney virus MLV-A protein, Moloney virus MLV-E protein, Baboon Endogenous retrovirus (BAEV) envelope protein, Ebola virus glycoprotein and foamy virus envelope protein, or a combination of two or more of these viral envelope proteins. A well-known illustration of pseudotyping viral vector particles consists of the pseudotyping of viral vector particles with the vesicular stomatitis virus glycoprotein (VSV-G). For the pseudotyping of viral vector particles, the one skilled in the art may notably refer to Yee et al. (1994, ProcNatl Acad Sci, USA, Vol. 91 : 9564-9568) Cronin et al. (2005, Curr Gene Ther, Vol. 5(n°4): 387-398). For producing virus-derived particles, and more precisely VSV-G pseudotypes virus-derived particles, for delivering protein(s) of interest into target cells, the one skilled in the art may refer to Mangeot et al. (2011, Molecular Therapy, Vol. 19 (n°9): 1656-1666).

In some preferred embodiments, the VSV-G protein which is used for pseudotyping a virus- derived particle of the invention has the amino acid sequence of SEQ ID NO. 23, that may be encoded by a nucleic acid comprising the sequence of SEQ ID NO. 28. In some preferred embodiments, the BAEV-G (BAEV) protein which is used for pseudotyping a virus-derived particle of the invention has the amino acid sequence of SEQ ID NO. 25, that may be encoded by a nucleic acid comprising the sequence of SEQ ID NO. 27.

Thus, in some embodiments, a virus-derived particle further comprises a viral envelope protein, wherein either (i) the said viral envelope protein originates from the same virus as the viral structural protein, e.g. originates from the same virus as the viral Gag protein, or (ii) the said viral envelope protein originates from a virus distinct from the virus from which originates the viral structural protein, e.g. originates from a virus distinct from the virus from which originates the viral Gag protein.

As it is readily understood by the one skilled in the art, a virus-derived particle that is used according to the invention may be selected in a group comprising Moloney murine leukemia virus-derived vector particles, Bovine immunodeficiency virus-derived particles, Simian immunodeficiency virus-derived vector particles, Feline immunodeficiency virus-derived vector particles, Human immunodeficiency virus-derived vector particles, Equine infection anemia virus- derived vector particles, Caprine arthritis encephalitis virus-derived vector particle, Baboon endogenous virus-derived vector particles, Rabies virus-derived vector particles, Influenza virus- derived vector particles, Norovirus-derived vector particles, Respiratory syncytial virus-derived vector particles, Hepatitis A virus-derived vector particles, Hepatitis B virus-derived vector particles, Hepatitis E virus-derived vector particles, Newcastle disease virus-derived vector particles, Norwalk virus-derived vector particles, Parvovirus-derived vector particles, Papillomavirus-derived vector particles, Yeast retrotransposon-derived vector particles, Measles virus-derived vector particles, and bacteriophage-derived vector particles. In particular, a virus-derived particle that is used according to the invention is a retrovirus-derived particle. Such retrovirus may be selected among Moloney murine leukemia virus, Bovine immunodeficiency virus, Simian immunodeficiency virus, Feline immunodeficiency virus, Human immunodeficiency virus, Equine infection anemia virus, and Caprine arthritis encephalitis virus.

In another embodiment, a virus-derived particle that is used according to the invention is a lentivirus-derived particle. Lentiviruses belong to the retroviruses family, and have the unique ability of being able to infect non-dividing cells.

Such lentivirus may be selected among Bovine immunodeficiency virus, Simian immunodeficiency virus, Feline immunodeficiency virus, Human immunodeficiency virus, Equine infection anemia virus, and Caprine arthritis encephalitis virus. For preparing Moloney murine leukemia virus-derived vector particles, the one skilled in the art may notably refer to the methods disclosed by Sharma et al. (1997, Proc Natl Acad Sci USA, Vol. 94: 10803+-10808), Guibingua et al. (2002, Molecular Therapy, Vol. 5(n°5): 538-546). Moloney murine leukemia virus-derived (MLV-derived) vector particles may be selected in a group comprising MLV-A-derived vector particles and MLV-E-derived vector particles.

For preparing Bovine Immunodeficiency virus-derived vector particles, the one skilled in the art may notably refer to the methods disclosed by Rasmussen et al. (1990, Virology, Vol. 178(n°2): 435-451) For preparing Simian immunodeficiency virus-derived vector particles, including VSV-G pseudotyped SIV virus-derived particles, the one skilled in the art may notably refer to the methods disclosed by Mangeot et al. (2000, Journal of Virology, Vol. 71(n°18): 8307-8315), Negre et al. (2000, Gene Therapy, Vol. 7: 1613-1623) Mangeot et al. (2004, Nucleic Acids Research, Vol. 32 (n° 12), el 02)

For preparing Feline Immunodeficiency virus-derived vector particles, the one skilled in the art may notably refer to the methods disclosed by Saenz et al. (2012, Cold Spring Harb Protoc, (1): 71-76; 2012, Cold Spring Harb Protoc, (1): 124-125; 2012, Cold Spring Harb Protoc, (1): 118- For preparing Human immunodeficiency virus-derived vector particles, the one skilled in the art may notably refer to the methods disclosed by Jalaguier et al. (2011, PlosOne, Vol. 6(n°l l), e28314), Cervera et al. (J Biotechnol, Vol. 166(n°4): 152-165), Tang et al. (2012, Journal of Virology, Vol. 86(n°14): 7662-7676)

For preparing Equine infection anemia virus-derived vector particles, the one skilled in the art may notably refer to the methods disclosed by Olsen (1998, Gene Ther, Vol. 5(n°l 1): 1481-1487). For preparing Caprine arthritis encephalitis virus-derived vector particles, the one skilled in the art may notably refer to the methods disclosed by Mselli-Lakhal ety al. (2006, J Virol Methods, Vol. 136(n°l-2): 177-184).

For preparing Baboon endogenous virus-derived vector particles, the one skilled in the art may notably refer to the methods disclosed by Girard-Gagnepain et al. (2014, Blood, Vol. 124(n°8): 1221-1231)

For preparing Rabies virus-derived vector particles, the one skilled in the art may notably refer to the methods disclosed by Kang et al. (2015, Viruses, Vol. 7: 1134-1152, doi: 10.3390/v7031134), Fontana et al. (2014, Vaccine, Vol. 32(n°24): 2799-27804) or to the PCT application published under n° WO 2012/0618.

For preparing Influenza virus-derived vector particles, the one skilled in the art may notably refer to the methods disclosed by Quan et al. (2012, Virology, Vol. 430: 127-135) and to Latham et al. (2001, Journal of Virology, Vol. 75(n°13),: 6154-6155).

For preparing Norovirus-derived vector particles, the one skilled in the art may notably refer to the methods disclosed by Tome-Amat et al., (2014, Microbial Cell Factories, Vol. 13: 134-142). For preparing Respiratory syncytial virus-derived vector particles, the one skilled in the art may notably refer to the methods disclosed by Walpita et al. (2015, PlosOne, DOI: 10.1371/journal.pone.0130755) For preparing Hepatitis B virus-derived vector particles, the one skilled in the art may notably refer to the methods disclosed by Hong et al. (2013, Vol. 87(n°12): 6615-6624).

For preparing Hepatitis E virus-derived vector particles, the one skilled in the art may notably refer to the methods disclosed by Li et al. (1997, Journal of Virology, Vol. 71(n°10): 7207-7213).

For preparing Newcastle disease virus-derived vector particles, the one skilled in tha art may notably refer to the methods disclosed by Murawski et al. (2010, Journal of Virology, Vol. 84(n°2): 1110-1123)

For preparing Norwalk virus-derived vector particles, the one skilled in the art may notably refer to the methods disclosed by Herbst-Kralovetz et al. (2010, Expert Rev Vaccines, Vol. 9(n°3): 299-307). For preparing Parvovirus-derived vector particles, the one skilled in the art may notably refer to the methods disclosed by Ogasawara et al. (2006, In Vivo, Vol. 20: 319-324)

For preparing Papillomavirus-derived vector particles, the one skilled in the art may notably refer to the methods disclosed by Wang et al. (2013, Expert Rev Vaccines, Vol. 12(n°2): doi: 10.1586/erv.l2.151)

For preparing Yeast retrotransposon-derived vector particles, the one skilled in the art may refer to the methods disclosed by Peifang et al. (1994, Clin Exp Immunol, Vol. 97(n°3): 361-366) or to the US patent n° US 6,060,064

For preparing Measles virus-derived vector particles, the one skilled in the art may notably refer to the methods disclosed by Brandler et al. (2008, Vol. 31(n°2-3): 271-291).

For preparing bacteriophage-derived vector particles, and in particular Q-beta virus-like particles, the one skilled in the art may notably refer to the methods disclosed by Brown et al. (2009, Biochemistry, Vol. 48(n°47): 11155-11157).

A virus-derived particle that is used herein comprises a Gag protein, and most preferably a Gag protein originating from a virus selected in a group comprising Rous Sarcoma Virus (RSV) Feline Immunodeficiency Virus (FIV), Simian Immunodeficiency Virus (SIV), Moloney Leukemia Virus (MLV) and Human Immunodeficiency Viruses (HIV-1 and HIV-2) especially Human Immunodeficiency Virus of type 1 (HIV-1).

In some embodiments, a virus-derived particle may also comprise one or more viral envelope protein(s). The presence of one or more viral envelope protein(s) may impart to the said virus- derived particle a more specific tropism for the cells which are targeted, as it is known in the art. The one or more viral envelope protein(s) may be selected in a group comprising envelope proteins from retroviruses, envelope proteins from non-retroviral viruses, and chimeras of these viral envelope proteins with other peptides or proteins. An example of a non-lentiviral envelope glycoprotein of interest is the lymphocytic choriomeningitis virus (LCMV) strain WE54 envelope glycoprotein. These envelope glycoproteins increase the range of cells that can be transduced with retroviral derived vectors.

In some preferred embodiments, the virus-derived particle comprises a Gag protein originating from a virus selected in a group comprising Rous Sarcoma Virus (RSV) and Moloney Leukemia Virus (MLV).

In some preferred embodiments, a virus-derived particle that is used herein, further comprises a pseudotyping viral envelope protein, and most preferably a VSV-G protein.

Cas protein

A virus-derived particle comprises a Cas protein. The said Cas protein may be selected in a group comprising a type I Cas protein, a type II Cas protein and a type III Cas protein. For using a type I, type II or type III Cas protein, the one skilled in the art may refer to Chylinski et al. (2014, Nucleic Acids Research, Vol. 42(n°10): 6091-6105), Sinkunas et al. (2011, The EMBO Journal, Vol. 30(n°7): 1335-1342), Aliyari et al. (2009, Immunological Reviews, Vol. 227(n°l): 176-188), Cass et al. (Biosci Rep, doi: 10.1042/BSR20150043), Makarova et al. (201 1, Biology Direct, Vol. 6: 38), Gasiunas et al. (2012, Proc NatlAcad Sci USA, Vol. 109(n°39): E2579-E2586), Heler et al. (2015, Nature, Vol. 519(n°7542): 199-202), Esvelt et al. (2013, Nat Methods, Vol. 10(n°l l): doi : 10.138/nmeth.2681) or Chylinski et al. (2013, Biology, Vol. 10(n°5): 726-737). In some embodiments, the Cas protein may consist of the Type II Cas protein named Cpfl which is disclosed by Zeische et al. (2015, Cell, http://dx.doi.Org/10.1016/j.cell.2015.09.038, in Press).

Preferably, a virus-derived protein comprises a type II Cas protein. A type II Cas protein is most preferably a Cas9 protein.

Most preferably, the Cas protein which is contained inside a virus-derived particle as described herein is a Cas9 protein, or an homolog or a derivative thereof. The Cas9 protein may be selected in a group comprising a Cas9 protein originating from Streptococcus thermophilus and a Cas9 protein originating from Streptococcus pyogenes, or an homolog thereof or a derivative thereof. Cas9 protein originating from Streptococcus thermophilus is described notably by Gasiunas et al. (2012, Proc NatlAcad Sci USA, Vol. 109(n°39): E2579-E2586). Cas9 protein originating from Streptococcus pyogenes is described notably by Heler et al. (2015, Nature, Vol. 519(n°7542): 199-202) and Sanjana et al. (2014, Nat Methods, Vol. l l(n°18): 783-784).

A Cas9 protein that may be used according to the present invention encompasses proteins which are homologs, variants or derivatives of the naturally occurring Cas9 proteins, such as the Cas9 proteins described by Cong et al. (2013, Science, Vol. 339: 819-823). A Cas9 protein as well as vectors encoding a Cas9 protein are commercially available from Sigma- Aldrich Company. Cas9 protein and variants thereof that may be used in virus-derived particles described herein are also described in the PCT applications published under n° WO 2013/163628, WO 2014/093595, WO 2015/089247 and WO 2015/089486. In some embodiments, Cas9 protein is produced so as to be incorporated in the virus-derived particles during their formation. Illustratively, Cas9 may be encoded by a nucleic acid sequence inserted in an expression vector contained in the virus-derived particles producing cells. In preferred embodiments, the Cas9-encoding nucleic acid is placed under the control of regulatory sequences allowing its over-expression in the producing cells. In some embodiments, the Cas9 protein consists of the protein of SEQ ID NO. 31, which is encoded by the nucleic acid sequence of SEQ ID NO. 32.

In some embodiments of a virus-derived particle described herein, the Cas protein is produced and integrated within the said virus-derived particle as a fusion protein between (i) a viral structural protein and (ii) the said Cas protein. In some of these embodiments, the Cas protein is produced and integrated within the said virus-derived particle as a GAG-Cas9 fusion protein. As it has been ascertained by the present inventors, such a fusion protein is successfully integrated within the resulting virus-derived particle and the Cas moiety is fully active, i.e. the Cas moiety possesses its endonuclease activity. According to those embodiments, the embedded Cas protein is released inside the target cells following the entering of the virus-derived particles.

In other embodiments, a Cas protein comprised in a virus-derived particle is initially produced as a cleavable fusion protein between (i) a viral structural protein and (ii) a Cas protein. An illustration of such a cleavable fusion protein is the cleavable GAG-Cas9 protein that is described in the examples herein. According to these other embodiments, the said cleavable fusion protein is integrated within the resulting virus-derived particle at the time of its production by the producing cells. Then, part or all of the said fusion proteins may be cleaved in the final virus-derived particles, leading to a population of virus-derived particles comprising (i) a part of the virus- derived particles wherein none the said cleavable fusion protein has been cleaved, (ii) a part of the virus-derived particles wherein at least a part of the said cleavable fusion proteins have been cleaved, leading the release of Cas protein moiety inside the virus-derived particles and (iii) a part of the virus-derived particles wherein all or almost all of the said cleavable fusion proteins have been cleaved, leading the release of all or almost all of the Cas protein moieties inside the virus- derived particles. In some preferred embodiments, a cleavable GAG-Cas9 protein is the GAG- Cas9 protein having the amino acid sequence of SEQ ID NO. 22, that may be encoded by a sequence of SEQ ID NO. 26. In other embodiments, it may be used a cleavable GAG-Cas9 protein encoded by the nucleic acid sequence of SEQ ID NO. 34 (that may be termed "KLAP229" herein).

Thus, in a virus-derived particle that may be used according to the invention, the Cas protein, typically the Cas9 protein, may be present either as (i) a non-cleavable fusion protein, typically a non-cleavable Gag-Cas9 fusion protein, as (ii) a cleavable fusion protein, typically a cleavable Gag-Cas9 fusion protein, as (iii) a Cas protein, typically a Cas9 protein, resulting from the proteolytic cleavage of the said fusion protein, or (iv) both the fusion protein and the Cas protein. It shall be understood that a virus-derived particle as used herein is produced in packaging cells that notably express a protein between (i) a viral structural protein and (ii) a Cas protein, typically a cleavable Gag-Cas9 fusion protein, which encompasses the cleavable fusion protein between (i) a viral structural protein and (ii) a Cas protein, typically the cleavable Gag-Cas9 fusion protein. The cleavable fusion protein is incorporated as such in the virus-derived particle and is then at least partly cleaved in the virus-derived particle so as to release the Cas protein, which is functional in the virus-derived particle as it is shown in the examples herein. However, because the Cas protein is initially incorporated in the virus-derived particles under the form of the said cleavable fusion protein, there are a number of intermediate states wherein the Cas protein is partly present under the form of the cleavable fusion protein and partly present as a free Cas protein resulting from the cleavage of the cleavable fusion protein.

In preferred embodiments, the fusion protein comprises a proteolysis cleavage site located between the viral structural protein moiety and the Cas protein moiety, typically between the Gag protein moiety and the Cas9 protein moiety. Proteolytic sites, which may also be termed protease sites, are well known form the one skilled in the art. A protease site that may be contained in the cleavable fusion protein may be a site that is cleavable by a protease selected in a group comprising trypsin (EC 3.4.21.4), chymotrypsin (EC 3.4.21.1), endoproteinase Glu C (EC 3.4.21.19), endoproteinase Lys-C (EC 3.4.21.50), pepsin (EC 3.4.23.1), elastase (EC 3.4.21.36) abd carboxypeptidase (EC 3.4.17.1).

In some embodiments, the protease cleavage site is selected in a group comprising the amino acid sequences SSLYPALTP (SEQ ID N° 29), that may be encoded by a sequence comprising SEQ ID NO. 30.

Protease cleavable fusion protein between Gag and a protein of interest, as well as vectors for expressing such fusion proteins are notably described by Voelkel et al. (2010, Proc Natl Acad Sci USA, Vol. 107(n°17): 7805-7810), to which the one skilled in the art may refer.

As described in the examples herein, some embodiments of a virus-derived particles are formed in packaging cells expressing a Gag-Pro-Pol viral protein. Without wishing to be bound by any particular theory, the inventors believe that in these embodiments, the Pro protein (i.e. the viral protease) is released in the virus-derived particles and cleaves the fusion protein, typically the Gag Cas fusion protein, especially the Gag-Cas9 fusion protein, so as to generate the free Cas protein, especially the free Cas9 protein. In some preferred embodiments, the Gag-Pro-Pol protein has the amino acid sequence of SEQ ID NO. 24. However, as it is also illustrated in the examples herein, A functional Cas protein, typically a functional Cas9 protein, is released in the target cells in the embodiments wherein the virus- derived particles are devoid of any viral protease, e.g.; when the virus-derived particles are formed in packaging cells that express a viral structural protein (e.g. Gag) and optionally one or more viral envelope protein (e.g. VSV-G and/or BAEV-G).

Guide RNAs

For generating a site-directed alteration in a target nucleic acid, when using a virus-derived particle as described herein, one or more CRISPR-Cas guide RNAs are required.

The number of CRISPR-Cas guide RNAs, which may also be termed "guide RNAs" or "gRNAs", may vary depending of the kind of alteration(s) to the target nucleic acids which is(are) sought. A single guide RNA may be used in combination with a virus-derived particle for generating a single DNA cleavage event in the target nucleic acid. Two or more guide RNAs may be used in combination with a virus-derived particle for generating two or more cleavage events in the target nucleic acids, or alternatively to generate cleavage event(s) in a plurality of target nucleic acids.

Methods for designing guide RNAs that, when combined with a Cas protein, generate the cleavage of a target nucleic acid, are well known from the one skilled in the art. As it is well known in the art, a guide RNA is a polynucleotide having sufficient complementarity with a target nucleic acid to hybridize with the said target nucleic acid and direct sequence-specific binding of a CRISPR complex to the said target nucleic acid.

Various tools are readily available to the one skilled in the art for designing guide RNAs, which include the tool marketed under the name GenCRISPR™ gRNA constructs by the Company GenScript (United States). The GenCRISPR™ gRNA constructs collection comprise about six guide RNAs to specifically target each of about 20,000 genes in the human genome. Guide RNAs may also be designed according to the teachings of Ran et al. (2013, Cell, Vol. 154: 1380-1389), Mail et al. (2013, Science, Vol. 339: 823-826), Wang et al. (2013, Cell, Vol. 153: 910-918), Jao et al. (2013, Proc Natl Acad Sic USA, Vol. 110: 13904-13909), Cong et al. (2013, Science, Vol. 339: 819-823), Shalem et al. (2014, Science, Vol. 343: 84-87), Maeder et al. (2013, Nat Methods; Vol. 10: 977-979), Qi et al. (2013, Cell, Vol. 152: 1173-1183), Farboud et al. (2015, Genetics, doi 10.1534/genetics.115.175166) or Ma et al. (2013, BioMed research International, Vol. 2013, Article ID 270805, doi.org/10.1155/2013/270805). In some embodiments, a virus-derived particle as described herein further comprises one or more CRISPR-Cas guide RNA(s). Each guide RNA hybridizes with a specific target sequence comprised in a target nucleic acid.

In some embodiments, a virus-derived particle as described herein comprises a single guide RNA. Such embodiments of a virus-derived particle allow generating a single cleavage at a desired location of the target nucleic acid.

In some other embodiments, a virus-derived particle as described herein comprises two distinct guide RNAs, each guide RNA hybridizing with a specific target sequence comprised in the same target nucleic acid, so as to generate two cleavage events at the sites recognized by the respective two distinct guide RNAs. Such embodiments allow introducing a deletion of the polynucleotide framed by the two cleavage sites within the target nucleic acid. When a template nucleic acid of interest is further added, such embodiments allow the insertion of a desired exogenous nucleic acid of interest in the nucleic acid target, between these two cleavage sites.

In some preferred embodiments, the one or more guide RNAs are comprised inside the virus- derived particle. Typically, the virus-derived particles are produced by packaging cells expressing (i) the required viral structural protein(s) (e.g. Gag), (ii) the one or more viral envelope protein(s) (e.g. VSV-G and/or BAEV-G), (iii) the Cas fusion protein (e.g. a Gag-Cas9 fusion protein) and (iv) the one or more CRISPR-Cas guide RNAs. According to these embodiments, the one or more guide RNAs are incorporated within the virus-derived particles while these are produced by the packaging cells. In these embodiments wherein the virus-derived particles comprise a Cas protein, especially a Cas9 protein, and one or more guide RNA(s), the said virus-derived particles comprise CRISPR-Cas ribonucleoprotein complexes which are complexes of the Cas protein with a guide RNA.

According to some of these embodiments, the said virus-derived particles comprise one or more kinds of complexes of a Cas protein and a guide RNA, wherein each CRISPR-Cas complex comprise a single Cas protein complexed with a single guide RNA. In some of these embodiments wherein a plurality of cleavages of a target nucleic acid is sought, the said virus-derived particles comprise the same number of kinds of CRISPR-Cas complexes, each kind of CRIPSR-Cas complex being specific for generating a DNA cleavage at a desired location of a target nucleic acid to which the corresponding guide RNA hybridize.

In further preferred embodiments, the one or more guide RNAs are initially produced by specific packaging cells expressing the said one or more guide RNAs and also expressing the viral protein(s) which are required for producing other viral particles or other viral vesicles (or other Virus-Like Particles or VLPs). Then, the guide RNA(s)-containing viral particles are brought into contact with a virus-derived particle comprising a Cas protein, so as to generate, by complementation, the final virus-derived particles comprising both a Cas protein and the one or more guide RNAs that were initially contained in the said other viral particles, for obtaining these virus-derived particles by complementation, the one skilled in the art may notably refer to Abe et al. (1998, Journal of Virology, Vol. 72(n°8): 6356-6361). Illustratively, Gag-based Virus-derived particles comprising a Cas protein which are described herein may be brought into contact with VSV-G-based viral particles comprising one or more CRSIPS-Cas guide RNAs, so as to obtain final virus-derived particles comprising the said Cas protein and the said one or more CRISPR- Cas guide RNAs and wherein the said final virus-derived particles consist of VSV-G pseudotyped Gag-based VLPs.

In some other embodiments part of all of the said one or more guide RNAs are not comprised inside the virus-derived particles but are instead complexed with these virus-derived particles. According to these other embodiments, the guide RNAs which are complexed with the virus- derived particles also enter into the target cells with the virus-derived particles to which these guide RNAs are complexed. Targeting nucleic acids

For the purpose of altering a target nucleic acid by using virus-derived particles as described herein, especially when an alteration of the target nucleic acid by homologous recombination is sought, it is further made use of a targeting nucleic acid in combination with these virus-derived particles.

Methods for targeting nucleic acids for the purpose of altering their sequence by homologous recombination are well known from the one skilled in the art. Typically, a homologous repair donor nucleic acid comprises (i) a first sequence that is homologous to a first locus of the targeted genomic sequence and (ii) a second sequence that is homologous to a second locus of the genomic sequence. Generally, for the purpose of altering a target nucleic acid by homologous recombination, the said first sequence (i) and the said second sequence (ii) are located at each side of the cleavage site created by the CRISPR-Cas/guide RNA(s) complex. Methods for performing alterations in a target nucleic acid through homologous recombination by using a CRISPR-Cas system are well known in the art. The one skilled in the art may notably refer to Jinek et al. (2013, eLife, Vol.2: e00471, doi: 10.754/eLife.00471) and Lin et al. (2014, eLife, Vol. 3: e04766, DOI: 10.7554/eLife.04766). Typically, an Homologous Recombination template nucleic acid, which may also be termed a template nucleic acid herein, comprises an exogenous sequence of variable length, flanked at its 5' and 3' ends, respectively, by sequences that hybridizes to the target nucleic acid. If the exogenous sequence that will be inserted in the genome is below 50nt long, the flanking hybridizing sequences, also called homology recombination arms, should range from 20 to 50 nucleotides in length. If the exogenous sequence to insert is longer than 1 OOnt, the homology recombination arms should be considerably longer (around 800bp).

The targeting nucleic acid, or template nucleic acid, may have any suitable length, such as about 10, 15, 20, 25, 50, 75, 100, 150, 200, 500, 1000 or more nucleotides in length. When optimally aligned, a targeting nucleic acid might overlap with one or more nucleotides of a target sequence, e.g. about or more than about 1, 5, 10, 15, 20 or more nucleotides.

Based on the general knowledge from the one skilled in the art, practically the sole requirement for designing a targeting nucleic acid for the purpose of homologous recombination is the prior knowledge of the nucleic sequence of the target nucleic acid.

In some embodiments, a targeting nucleic acid is comprised inside a virus-derived particle as described herein. According to these embodiments, the virus-derived particle comprising a Cas protein, one or more guide RNAs and one or more targeting nucleic acids are preferably produced by packaging cells that express the said Cas protein, the required viral proteins, the required guide RNAs and the requited targeting nucleic acid(s).

In some other embodiments, a targeting nucleic acid is not comprised inside the virus-derived particle but is complexed to the virus-derived particles. Nucleic acid expression vectors

As already stated elsewhere in the present specification, a virus-derived particle as described herein is produced in cells, also named packaging cells herein, which express the required proteins, i.e. at least a fusion viral structural protein/Cas protein and one or more viral proteins required for forming the viral particles, which may also be termed Virus-Like Particles or VLPs. In preferred embodiments, the packaging cells also express one or more CRISPR-Cas guide RNAs and, when necessary, also a targeting nucleic acid (also termed template nucleic acid).

The term "expression vector" as used herein refers to a recombinant DNA molecule containing a desired coding sequence and appropriate nucleic acid sequences necessary for the expression of the operably linked coding sequence in a particular host organism. Nucleic acid sequences necessary for expression in eukaryotic cells generally comprise promoters, enhancers, and termination and polyadenylation signals. In some embodiments, "expression vectors" are used in order to permit pseudotyping of the viral envelope proteins.

Generally, vectors for expressing the required proteins or nucleic acids are vector suitable for expressing nucleic sequences within the desired host cells that are used as packaging cells. Preferably, the packaging cells are mammalian cells. Notably, vectors for expressing the required proteins or nucleic acids comprise an open reading frame which is placed under the control of regulatory elements that are functional in the packaging cell wherein their expression is sought. Notably, these vectors comprise, for each protein or nucleic acid to be expressed, an open reading frame which is placed under the control of a suitable promoter sequence, as well as a polyadenylation sequence.

The packaging cell line provides the viral proteins required for particle assembly (Markowitz et al., 1988, J. Virol., Vol. 62 :1120). As it is well known in the art, a nucleic acid vector is introduced into the packaging cell by any of a variety of techniques (e.g., calcium phosphate co-precipitation, lipofection, electroporation). The viral proteins produced by the packaging cell mediate the insertion of the viral protein(s) and of the Cas protein into virus-derived particles, which are then released into the culture supernatant. The nucleic acid vectors used may be derived from a retrovirus (e.g., a lentivirus). Retrovirus vectors suitable for producing the virus-derived particles described herein allow (1) transfection of the packaging vectors and envelope vectors into the host cell to form a packaging cell line that produces the virus-derived particles essentially free from packaging vector RNA, and (2) the packaging of the Cas protein and optionally also of the CRISPR guide RNA(s) and eventually of a targeting nucleic acid into the virus-derived particles.

Vectors and packaging cells for use according to the present invention are illustrated in the examples herein.

Illustratively, a vector for expressing the viral structural protein/Cas protein, e.g. a Gag-Cas9 protein, may be prepared by the one skilled in the art as taught by Voelkel et al. (2010, Proc Natl Acad Sci USA, Vol. 107: 7805-7810).

Illustratively, a vector for expressing the viral structural protein, e.g. a Gag protein or a Gag-Pro- Pol fusion protein, and optionally also a viral envelope protein, e.g. a VSV-G protein or a BAEV- G protein, may be prepared by the one skilled in the art according to the teachings of Negre et al. (2000, Gene Ther, Vol. 7: 1613-1623) and of Yee et al. (1994, Methids Cell Biol, Vol. 43 PtA: 99-112).

Illustratively, a vector for expressing a CRISPR guide RNA may be prepared as taught by Kieusseian et al. (2006, Blood, Vol. 107: 492-500).

Packaging cells

The host cell is a cell into which a vector of interest may be introduced and wherein it may be replicated, and, in the case of an expression vector, in which one or more vector-based genes may be expressed.

Any suitable permissive or packaging cell known in the art may be employed in the production of the virus-derived particles described herein. Mammalian cells or insect cells are preferred. Examples of cells useful for the production of the virus-derived particles in the practice of the invention include, for example, human cell lines, such as VERO, WI38, MRC5, A549, HEK293, HEK293T, B-50 or any other HeLa cells, HepG2, Saos-2, HuH7, and HT1080 cell lines. Illustrative cell lines for use as packaging cells are insect cell lines. Any insect cell which allows for replication of AAV and which can be maintained in culture can be used in accordance with the present invention. Examples include Spodoptera frugiperda, such as the Sf9 or Sf21 cell lines, Drosophila spp. cell lines, or mosquito cell lines, e.g., Aedes albopictus derived cell lines. A preferred insect cell line is the Spodoptera frugiperda Sf9 cell line. The following references are incorporated herein for their teachings concerning use of insect cells for expression of heterologous polypeptides, methods of introducing nucleic acids into such cells, and methods of maintaining such cells in culture: Methods in Molecular Biology, ed. Richard, Humana Press, NJ (1995 ); O'Reilly et al., Baculovirus Expression Vectors: A Laboratory Manual, Oxford Univ. Press (1994 ); Samulski et al., J. Vir. 63:3822-8 (1989 ); Kajigaya et al., Proc. Nat'l. Acad. Sci. USA 88: 4646-50 (1991 ); Ruffing et al., J. Vir. 66:6922-30 (1992 ); Kimbauer et al., Vir. 219:37- 44 (1996 ); Zhao et al., Vir. 272:382-93 (2000 ); and Samulski et al., U.S. Pat. No. 6,204,059 .

The cells may be supplied with any one or more of the stated functions already incorporated, e.g., a cell line with one or more vector functions incorporated extra-chromosomally or integrated into the cell's chromosomal DNA, a cell line with one or more packaging functions incorporated extra- chromosomally or integrated into the cell's chromosomal DNA, or a cell line with helper functions incorporated extra-chromosomally or integrated into the cell's chromosomal DNA. A packaging cell line is a suitable host cell transfected by one or more nucleic acid vectors that, under achievable conditions, produces virus-derived particles comprising a Cas protein and, in some embodiments, also one or more CRIPSR guide RNA(s) and eventually also a targeting nucleic acid.

As used herein, the term "packaging cell lines" is typically used in reference to cell lines that express viral structural proteins (e.g., gag, pol and env), but do not contain a packaging signal. For example, a cell line has been genetically engineered to carry at one chromosomal site within its genome, a 5'-LTR-gag-pol-3'-LTR fragment that lacks a functional psi+ sequence (designated as Δ-psi), and a 5'-LTR-env-3'-LTR fragment that is also Δ-psi located at another chromosomal site. A number of cell types can be used, which encompasses:

a) NIH-3T3 murine cells which are currently widely used as packaging cells producing recombinant retroviruses in clinical use (Takahara et al., Journal of Virology, (June 1992), 66 (6) 3725-32). b) TK " cell lines have already been described, including NIH-3T3 TK cells (F. Wagner et al., EMBO Journal (1985), Vol. 4 (n°3): 663-666); these cells can be killed when they are cultivated in selective culture media such as HAT. If they are complemented for the kinase thymidine function, for example those from the HSV1-TK virus, they can grow in a selective medium; such lines thus offer the possibility of using the HSV1-TK gene as a selection gene. The gene coding for the thymidine kinase of HSV1 or one of its functional derivatives is also widely used as a transgene as a pro-drug transforming ganciclovir or acyclovir into a drug which is cytotoxic for the cell, and it can thus be applied to selective cell destruction, for example of cancerous cells (see, for example, International patent application WO 95/22617).

Illustratively, the packaging cells may be the well-known HEK293T cell line, as shown in the examples herein.

The present invention also relates to a cell line for producing a virus-derived particle as described herein, comprising:

- one or more nucleic acids encoding the proteins required for forming the said virus- derived particle, and

- a nucleic acid comprising an expression cassette encoding a viral structural protein-Cas fusion protein.

In some embodiments, a nucleic acid encoding a protein required for forming the said virus- derived particle encompasses a nucleic acid encoding a viral structural protein, such as a Gag protein. In some embodiments, the said cell line also comprises a nucleic acid encoding a viral envelope protein, such as a viral envelope protein selected in a group comprising a VSV-G protein and a BAEV-G protein.

In some embodiments, the said cell line further comprises nucleic acid(s) encoding one or more CRISPR guide RNA(s).

In some embodiments, the said cell line further comprises nucleic acid(s) encoding one or more targeting nucleic acid(s). Compositions and kits

The present invention provides virus-derived particles compositions and kits suitable for use in therapy (in vivo or ex vivo) that are described herein. In some embodiments, the said compositions comprise virus-derived particles comprising a Cas protein, especially a Cas9 protein, and is devoid of a guide RNA and of a targeting nucleic acid. In these embodiments, the gRNA(s) or the targeting nucleic acid are absent from the virus-derived particles, either as nucleic acids located inside the said virus-derived particles or as nucleic acids complexed with the said virus-derived particles.

The present invention relates to a composition for altering a target nucleic acid in a eukaryotic cell, which composition comprises at least one virus-derived particle as described in the present specification. In some embodiments, the said composition further comprises one or more CRISPR-Cas system guide RNA(s).

In some of these embodiments, the said one or more CRISPR-Cas system guide RNA(s) is(are) comprised in virus-derived particles.

In some other embodiments, the said one or more CRISPR-Cas system guide RNA(s) is(are) complexed with the said virus-derived particles.

In some embodiments of the compositions, the said compositions comprise (i) Cas-containing virus-derived particles in combination with (ii) vesicles comprising gRNA(s) and /or targeting nucleic acid(s). According to some of these embodiments, each gRNA present in the composition is comprised in a specific kind vesicles. According to some other of these embodiments, more than one gRNA, which includes all gRNA(s), are comprised in a specific kind of vesicles. In some of these embodiments, a targeting nucleic acid is comprised in a specific kind of vesicles. In some other of these embodiments, when more than one targeting nucleic acid is present in the composition, all the targeting nucleic acids are all comprised in a specific kind of vesicles. In still further embodiments, the whole gRNA(s) and targeting nucleic acid(s) present in the composition are all comprised in the same vesicles. A "specific kind" of vesicle, as used herein is defined uniquely as regards its specific content in gRNA(s) and/or targeting nucleic acid(s), irrespective of the structural features of the vesicle itself. Most preferably, the said vesicles are comprised of viral proteins. In some embodiments, the said vesicles have the same structural features of viral proteins as the virus-derived particles containing a Cas protein that are described in the present specification. In some other embodiments, the said vesicles are mainly or fully composed of viral envelope proteins, such as, for example, VSV-G or BAEV-G.

When present in a composition according to the invention, the Cas-containing virus-derived particles and the gRNA(s)- and/or targeting nucleic acid- containing vesicles trans-complement so as to efficiently generate the desired nucleic acid alteration(s) in the target cells. Such a trans- complementation in another technical context is taught by Mangeot et al. (2011, Ther J am Soc Gene Ther, Vol. 19: 1656-1666).

Compositions as described herein encompass pharmaceutical compositions that are used for the purpose of performing a method of gene therapy in mammals in need thereof, which includes non- human mammals and human individuals in need thereof.

Compositions of the invention may be formulated for delivery to animals for veterinary purposes (e.g., livestock such as cattle, pigs, etc), and other non-human mammalian subjects, as well as to human subjects. The virus-derived particles may be formulated with a physiologically acceptable carrier for use in gene transfer and gene therapy applications.

In some embodiments, the said composition further comprises one or more transduction helper compounds. The transduction helper compounds are preferably selected in a group comprising cationic polymers, as described notably by Zuris et al. (2015, Nat Biotechnol, Vol. 33(n°l): 73- 80). The transduction helper compound may be selected in a group comprising polybrene (that may be also termed hexadimethrine bromide), protamine sulfate, 12-myristate 13-acetate (also termed phorbol myristate acetate or PMA, as described by Johnston et al., 2014, Gene Ther, Vol. 21(12): 1008-1020), vectofusin (as described by Fenard et al., 2013, Molecular Therapy Nucleic Acids, Vol. 2: e90), poloxamer P338 (as described by Anastasov et al., 2016, Lentiviral vectors and exosomes as gene and protein delivery tools, in Methods in Molecular Biology, Vol. 1448: 49-61), RetroNectin® Reagent (commercialized by Clontech Laboratories Inc.), Viral Plus ® transduction enhancer (commercialized by Applied Biological Materials Inc.), TransPlus® Virus Transduction Enhancer(commercialized by Clinisciences), Lentiboost® (commercialized by Sirion Biotech), or ExpressMag® Transduction System (commercialized by Sigma-Aldrich). As shown in the examples herein, the said cationic transduction helper compound may consist of polybrene.

The virus-derived particles may be formulated in a conventional manner using one or more physiologically acceptable carriers or excipients. The virus-derived particles may be formulated for parenteral administration by injection, e.g., by bolus injection or continuous infusion. Formulations for injection may be presented in unit dosage form, e.g., in ampoules or in multi- dose containers, with an added preservative. The virus-derived particles compositions may take such forms as suspensions, solutions or emulsions in oily or aqueous vehicles, and may contain formulation agents such as suspending, stabilizing and/or dispersing agents. Liquid preparations of the virus-derived particles compositions may be prepared by conventional means with pharmaceutically acceptable additives such as suspending agents (e.g., sorbitol syrup, cellulose derivatives or hydrogenated edible fats); emulsifying agents (e.g., lecithin or acacia); non-aqueous vehicles (e.g., almond oil, oily esters, ethyl alcohol or fractionated vegetable oils); and preservatives (e.g., methyl or propyl-p-hydroxybenzoates or sorbic acid). The preparations may also contain buffer salts. Alternatively, the compositions may be in powder form for constitution with a suitable vehicle, e.g., sterile pyrogen-free water, before use.

The virus-derived particles compositions of the invention may be administered to a subject at therapeutically effective doses to generate the desired genome alteration in a target nucleic acid contained in a target cell, in a target tissue or organ or in a target organism, particularly a target mammal, which encompasses a target non-human mammal and a human individual. A therapeutically effective dose refers to an amount of the pharmaceutical composition sufficient to result in amelioration of symptoms caused by the occurrence of the desired genome alteration event in the target nucleic acid.

In an embodiment, an amount of virus-derived particles composition of the invention is administered at a dose unit that is in the range of about 0.1-5 micrograms ^g)/kilogram (kg). To this end, a virus-derived particles composition of the invention may be formulated in doses in the range of about 7 mg to about 350 mg to treat to treat an average subject of 70 kg in body weight. The amount of virus-derived particles composition of the invention that may be administered may be selected in a group comprising 0.1 mg/kg, 0.2 mg/kg, 0.3 mg/kg, 0.4 mg/kg, 0.5 mg/kg, 0.6 mg/kg, 0.7 mg/kg, 0.8 mg/kg, 0.9 mg/kg, 1.0 mg/kg, 1.5 mg/kg, 2.0 mg/kg, 2.5 mg/kg, 3.0 mg/kg, 3.5 mg/kg, 4.0 mg/kg, 4.5 mg/kg or 5.0 mg/kg. The dose of virus-derived particles in a unit dosage of the composition may be selected in a group comprising 7 mg, 8 mg, 9 mg, 10 mg, 20 mg, 25 mg, 30 mg, 35 mg, 40 mg, 45 mg, 50 mg, 55 mg, 60 mg, 65 mg, 70 mg, 75 mg, 80 mg, 85 mg 90 mg, 95 mg, 100 mg, 125 mg, 150 mg, 175 mg, 200 mg, 225 mg, 250 mg, 275 mg, 300 mg, 325 mg, 350 mg, 375 mg, 400 mg, 425 mg, 450 mg, 475 mg, 500 mg, 525 mg, 550 mg, 575 mg, 600 mg, 625 mg, 650 mg, 675 mg, 700 mg, 725 mg, or 750 mg, especially for treating an average subject of 70 kg in body weight. These doses can be given once or repeatedly, such as daily, every other day, weekly, biweekly, or monthly. In some embodiments, a virus-derived particles composition may be administered to a subject in one dose, or in two doses, or in three doses, or in four doses, or in five doses, or in six doses or more. The interval between dosages may be determined based the practitioner's determination that there is a need thereof.

The virus-derived particles compositions may, if desired, be presented in a pack or dispenser device that may contain one or more unit dosage forms containing the active ingredient. The pack may for example comprise metal or plastic foil, such as a blister pack. The pack or dispenser device may be accompanied by instructions for administration.

A virus-derived particles composition may be in liquid or solid (e.g. lyophilized) form. Kits

The present invention further relates to kits for preparing the virus-derived particles described in the present specification.

This invention concerns a kit for preparing virus-derived particles for altering a target nucleic acid in a eukaryotic cell comprising:

- a nucleic acid comprising an expression cassette encoding a GAG-Cas fusion protein, and

- a nucleic acid comprising one or more expression cassette(s) encoding virus-like assembly protein(s),

In some embodiments, the said kit further comprises a nucleic acid comprising an expression cassette encoding a pseudotyping viral envelope protein. In some embodiments of the said kit, the virus-derived assembly protein is a virus-derived Gag protein. In some embodiments, the said Gag protein is encoded by an expression cassette selected in a group comprising an expression cassette encoding a GAG-PRO-POL polyprotein and an expression cassette encoding a GAG protein.

In some embodiments, the said kit further comprises one or more nucleic acid(s) encoding a CRISPR-Cas system guide RNA

In certain embodiments of the said kit, the said nucleic acids are localized in an eukaryotic cell as a result of its transfection into the said eukaryotic cell. In some of these embodiments, the said nucleic acids are under the form of nucleic acid vectors in the said eukaryotic cells, which cells may also be termed packaging cells herein. In some other of these embodiments, part or all of these nucleic acids are integrated in the genome of these eukaryotic cells, which cells may also be termed packaging cells herein.

Thus, in some embodiments of a kit according to the invention, the said eukaryotic cell consists of a packaging cell line.

The kits of the present invention may optionally comprise different containers (e.g., vial, ampoule, test tube, flask or bottle) for each individual composition or element comprised therein. The kit may contain additional reagents, such as buffers, diluents and the like, for formulation the individual components. Each component will generally be suitable as aliquoted in its respective container or provided in a concentrated form.

Instructions for using the kit according to the methods described herein may be included. The instructional material may comprise a publication, a recording, a diagram, or any other medium of expression which can be used to communicate the usefulness of the method of the invention in the kit for assessment of oocyte quality. A package insert may comprise text housed in any physical medium, e.g., paper, cardboard, film, or may be housed in an electronic medium such as a diskette, chip, memory stick or other electronic storage form. The instructional material of the kit of the invention may, for example, be affixed to a container which contains other contents of the kit, or be shipped together with a container which contains the kit. Alternatively, the instructional material may be shipped separately from the container with the intention that the instructional material and the contents of the kit be used cooperatively by the recipient. Methods for altering a target nucleic acid

The virus-derived particles as well as the compositions comprising them may be used for gene therapy.

A further aspect of the invention is a method of treating subjects with the virus-derived particles according to the invention or with compositions comprising them.

Administration of the virus-derived particles to a human subject or an animal in need thereof can be by any means known in the art for administering virus vectors. Exemplary modes of administration include rectal, transmucosal, topical, transdermal, inhalation, parenteral (e.g., intravenous, subcutaneous, intradermal, intramuscular, and intraarticular) administration, and the like, as well as direct tissue or organ injection, alternatively, intrathecal, direct intramuscular, intraventricular, intravenous, intraperitoneal, intranasal, or intraocular injections. Injectables can be prepared in conventional forms, either as liquid solutions or suspensions, solid forms suitable for solution or suspension in liquid prior to injection, or as emulsions. Alternatively, one may administer the virus in a local rather than systemic manner, for example, in a depot or sustained-release formulation.

This invention also relates to a method for altering a target nucleic acid comprising at least a target sequence in an eukaryotic cell, comprising the steps of:

a) bringing into contact the said eukaryotic cell with virus-derived particles as described herein, or with a composition as described in the present specification, and

b) collecting the said eukaryotic cell having an altered target nucleic acid.

In some embodiments, the virus-derived particles, or compositions comprising them, are administered directly to the subject, in vivo. In some other embodiments, subject's cells are provided, and then the said cells are transduced in vitro with the virus-derived particles, or with a composition comprising them. In a further method step, the transduced subject's cells are administered back to the body of the subject. In some embodiments, said method is performed in vitro or ex vivo.

The present invention also relates to a composition as described in the present specification, for its use for preventing or treating any disease or disorder that is amenable to gene therapy.

The present invention provides for methods for preventing or treating any disease or disorder that is amenable to gene therapy. In one embodiment, "treatment" or "treating" refers to an amelioration of a disease or disorder, or at least one discernible symptom thereof. In another embodiment, "treatment" or "treating" refers to an amelioration of at least one measurable physical parameter associated with a disease or disorder, not necessarily discernible by the subject. In yet another embodiment, "treatment" or "treating" refers to inhibiting the progression of a disease or disorder, either physically, e.g., stabilization of a discernible symptom, physiologically, e.g., stabilization of a physical parameter, or both. Other conditions, including cancer, immune disorders, and veterinary conditions, may also be treated.

Types of diseases and disorders that can be treated by methods of the present invention include, but are not limited to, age-related macular degeneration; diabetic retinopathy; infectious diseases e.g., HIV pandemic flu, category 1 and 2 agents of biowarfare, or any new emerging viral infection; autoimmune diseases; cancer; multiple myeloma; diabetes; systemic lupus erythematosus (SLE); hepatitis C; multiple sclerosis; Alzheimer's disease; parkinson's disease; amyotrophic lateral sclerosis (ALS), huntington's disease; epilepsy; chronic obstructive pulmonary disease (COPD); joint inflammation, arthritis; myocardial infarction (MI); congestive heart failure (CHF); hemophilia A; or hemophilia B. Infectious diseases that can be treated or prevented by the methods of the present invention are caused by infectious agents including, but not limited to, viruses, bacteria, fungi, protozoa, helminths, and parasites. The invention is not limited to treating or preventing infectious diseases caused by intracellular pathogens. Many medically relevant microorganisms have been described extensively in the literature, e.g., see C. G. A Thomas, Medical Microbiology, Bailliere Tindall, Great Britain 1983, the entire contents of which are hereby incorporated herein by reference.

Types of cancers that can be treated or prevented by the methods of the present invention include, but are not limited to human sarcomas and carcinomas, e.g., fibrosarcoma, myxosarcoma, liposarcoma, chondrosarcoma, osteogenic sarcoma, chordoma, angiosarcoma, endotheliosarcoma, lymphangiosarcoma, lymphangioendotheliosarcoma, synovioma, mesothelioma, Ewing's tumor, leiomyosarcoma, rhabdomyosarcoma, colon carcinoma, pancreatic cancer, breast cancer, ovarian cancer, prostate cancer, squamous cell carcinoma, basal cell carcinoma, adenocarcinoma, sweat gland carcinoma, sebaceous gland carcinoma, papillary carcinoma, papillary adenocarcinomas, cystadenocarcinoma, medullary carcinoma, bronchogenic carcinoma, renal cell carcinoma, hepatoma, bile duct carcinoma, choriocarcinoma, seminoma, embryonal carcinoma, Wilms' tumor, cervical cancer, testicular tumor, lung carcinoma, small cell lung carcinoma, bladder carcinoma, epithelial carcinoma, glioma, astrocytoma, medulloblastoma, craniopharyngioma, ependymoma, pinealoma, hemangioblastoma, acoustic neuroma, oligodendroglioma, meningioma, melanoma, neuroblastoma, retinoblastoma; leukemias, e.g., acute lymphocytic leukemia and acute myelocytic leukemia (myeloblastic, promyelocytic, myelomonocytic, monocytic and erythroleukemia); chronic leukemia (chronic myelocytic (granulocytic) leukemia and chronic lymphocytic leukemia); and polycythemia vera, lymphoma (Hodgkin's disease and non-Hodgkin's disease), multiple myeloma, Waldenstrom's macroglobulinemia, and heavy chain disease.

The present invention is further illustrated, without being in any way limited to, the examples below. EXAMPLES

A. Materials and Methods

A.l . Constructs

The GAG-Cas9 coding plasmid was designed as described in (Voelkel et al., 2010). The codon- optimized sequence of flag-Cas9 from Streptococcus pyogenes was PCR-amplified using the lentiCRISPR plasmid (Addgene plasmid 4953) as a template. This last construct was a gift from F. Zhang laboratory (Shalem et al., 2014, Science, Vol. 343: 84-87). Flag-Cas9 was next inserted downstream the Murine Leukemia Virus GAG sequence (MA-CA-NC). Frames were harmonized to generate a polyprotein. Both moieties were separated by a MLV-protease cleavage site that releases flag-Cas9 from GAG during the viral maturation process. This chimeric protein was expressed under control of the hCMV promoter and is equiped with an intron and a poly A signal both derived from the rabbit beta-Globin mRNA. The expression plasmid encoding the GagProPol polyprotein of MLV (Negre et al., 2000, Gene Ther, Vol. 7: 1613-1623) and the VSVG coding plasmid (Yee et al., 1994, Methods Cell Biol, Vol. 43 PtA: 99-112) were described elsewhere.

The gRNA coding plasmids termed as « CRIZI » derives from a previously described lentiviral construct (Kieusseian et al., 2006, Blood, Vol. 107: 492-500) in which was inserted the U6 cassette from the lentiCRISPR plasmid. Cloning of CRISPR gRNA sequences in CRIZI was performed between BsmBI sites upstream the U6 promoter following the procedure described by the authors. Sequences of gRNAs used in this study were designed using the Crispseek software (potential off-targets> 3 mismatches) (Zhu et al., 2014, PloS One, Vol. 9: el08424). Primer sequences are given for each gRNA:

YFP

YFPgRNAlf caccgCGAGGAGCTGTTCACCGGGG (SEQ ID NO. 1)

YFPgRNA 1 r aaacCCCCGGTGAACAGCTCCTCGc (SEQ ID NO. 2)

YFPgRNA2f caccgTCACCATACCGGTAGCCAGC (SEQ ID NO. 3)

YFPgRNA2r aaacGCTGGCTaccggtATGGTGAc (SEQ ID NO. 4)

Myd88

hMyd88gRNAl f caccgGAGACCTCAAGGGTAGAGGT (SEQ ID NO. 5)

hMyd88gRNAlr aaacACCTCTACCCTTGAGGTCTCc (SEQ ID NO. 6)

hMyd88gRNA2f caccgGCAGCCATGGCGGGCGGTCC (SEQ ID NO. 7)

hMyd88gRNA2r aaacGGACCGCCCGCCATGGCTGCc (SEQ ID NO. 8)

mMyd88gRNAlf caccggagcgtactggacggcaccg (SEQ ID NO. 9)

mMyd88gRNAlr aaaccggtgccgtccagtacgctcc (SEQ ID NO. 10)

mMyd88gRNA2f caccggcccatctcctccgccagca (SEQ ID NO. 11)

mMyd88gRNA2r aaactgctggcggaggagatgggcc (SEQ ID NO. 12)

DDX3

DDX3gRNAlf CACCGAGGGATGAGTCATGTGGCAG (SEQ ID NO. 13)

DDX3gRNAlr AAACCTGCCACATGACTCATCCCTC (SEQ ID NO 14)

A.2. Production of VLPs Cas9-VLP refered as a preparation of VLPs incorporating one of several gRNAs targeting a specific gene. Preparation of VLPs requires a cotransfection of several plasmids. VLPs were produced upon transfection of Lenti-X™ 293T (Clontech) using JetPei (Polyplus) following the manufacturer instructions. The JetPrime transfection agent (Polyplus) or the Calcium phosphate method (CalPhos mammalian kit, Clontech) may be used as well.

Classical ratio of the plasmids mixed in the JetPei transfection recipe are: GAG-Cas9 (20%), GagProPol (20%), VSV-G or other envelope (20%) and gRNAs encoding constructs (40%). For Cas9Myd88VLPs, two different gRNA were introduced in the recipe to achieve copackaging of both RNA-species in the nascent VLPs. HEK293T cells plated at 3xl0e6 cells/ lOcm-plate 24h prior transfection were trans fected with a mix containing:

- Rl : 4ug of GAG-Cas9, 4ug of GagProPol, 2ug of VSVG, 2ug of BaEV envelope (Girard- Gagnepain et al., 2014), 2ug of Myd88-gRNAl coding plasmid, 2ug of Myd88-gRNA2 coding plasmid.

- R2: 6ug of GAG-Cas9, 6ug of GagProPol, 4ug of VSVG, 4ug of Myd88-gRNAl coding plasmid, 4ug of Myd88-gRNA2 coding plasmid.

- R3: 4ug of GAG-Cas9, 8ug of GagProPol, 2ug of VSVG, 2ug of BaEV envelope, 2ug of Myd88-gRNAl coding plasmid, 2ug of Myd88-gRNA2 coding plasmid.

- R4: 4-6ug of GAG-Cas9, 2ug of VSVG, 2ug of BaEV envelope, 5ug of Myd88-gRNAl coding plasmid, 5ug of Myd88-gRNA2 coding plasmid.

40 hours after transfection, VLP-containing supernatants were collected and clarified by a short centrifugation (2000g, 3 min). Clarified Cas9-YFPVLPs were used directly to transduce L929 cells. Cas9-h-h/mMyd88VLPs and Cas9-DDX3VLPs were pelleted by ultracentrifugation lh at 35000 rpm in a SW41 -rotor and re-suspended in ice-cold PBS by overnight gentle agitation.10ml of supernatant were concentrated to produce lOOul of concentrated VLPs in ice-cold PBS (concentration fold: lOOx). Concentrated VLPs batches were stored at -80°C and were shown to be stable at least 2 weeks at 4°C after thawing. Filtration of VLP-containing supernatant can be performed with a 0,45um-in-size pore filter before centrifugation/transduction.

A.3. ProteoTransduction procedure using Cas9-VLPs 3xl0e5 L929-YFP cells were transduced with Cas9-YFPVLPs by addition of 400ul of clarified VLP-containing supernatant in 400ul of medium in a 6well plate. After two hours, medium was supplemented with 2ml of DMEM 10%FCS. Transduction of primary cells was classically performed in 48-well plates upon addition of 5-20ul of lOOx VLPs in 300ul of medium. After two hours, this transduction medium was supplemented by 0,5ml of fresh culture medium. Polybrene addition was shown to potentialize proteotransduction when used at a final concentration of 4ug/ml in the transduction medium.

A.4. PCR-based genotyping assays

Genomic-DNA extraction of cells treated by VLPs was achieved using the Nucleospin Tissue Kit (Machery Nagel) according to the manufacturer instructions. DNA preparations were performed 24-48h after VLP -treatment but additional experiments indicate that cleavage was complete in HEK293T recipient cells as soon as 6h after exposure with VLPs.

PCPv amplifications of Myd88 were performed in 50ul using the GOTAQ polymerase (Promega). lOOng of cellular genomic DNA was used as a template for a PCR reaction as follow: 94°C 5 min, 3 cycles (94°C 30 sec, 68°C 30sec), 3 cycles (94°C 30 sec, 64°C 30 sec, 72°C, 30 sec), 27 cycles (94°C 30 sec, 57°C 30 sec, 72°C 30 sec), 72°C 5min, 12°C. PCR amplicons were analysed in Ethidium Bromide stained 2,5% agarose gels.

A.5. Primers used for genotyping analysis (5'-ΝΝΝ-3'):

YFP (surveyor assay)

YFPf tcTAATACGACTCACTATAGGGAGAGGTCTATATAAGCAGAGCTCGTTTAG

(SEQ ID NO. 15)

YFPr GGCCATGATATAGACGTTGTGGCTG (SEQ ID NO. 16)

Myd88 hMyd88f2 TTACGCCCCCCACATCACCCGCC (SEQ ID NO. 17)

hMyd88rl GTCTCCAGTTGCCGGATCTCCAAG (SEQ ID NO. 18) mMyd88f2 ggaaactccacaggcgagcgtac (SEQ ID NO. 19) mMyd88r2 ggcagtcctcctcgatgcgcgacttc (SEQ ID NO. 20)

A.6. Combination of Cas9-DDX3 VLPs with ssDNA

15ul of concentrated Cas9-DDX3VLPs were mixed in lOul of PBS containing 8ug/ml of polybrene. This mixture was next supplemented with 5ul of each dilutions of the Flag-DDX3 primer, best results being obtained with the higher concentration (5ul of primer at lOOpmol/ul). This 'all in one' complex was incubated 15min at 4°C and the 30ul were added in the medium (400ul+polybrene 4ug/ml) of HEK293T cultivated in a 12-well-plate (200000 cells plated the day before). After two hours, the transduction medium was supplemented with 1ml of DMEM 10% FCS. 40 hours after VLP -treatment, cells were splitted for amplification and analysis of the genetic insertion of the flag sequence upstream the DDX3 gene and WB analysis were performed 72 hours latter.

A.7. Sequence of the Flag-DDX3 primer (HPLC-purified):

5 '-ACTCGCTTAGCAGCGGAAGACTCCGAGTTCTCGGTACTCTTCAGGGATGGA

CTACAAGGACGACGATGACAAGAGTCATGTGGCAGTGGAAAATGCGCTCGGGCTGG ACCAGCAGGTGA-3 ' (SEQ ID NO. 21)

Example 1: Cleavage of the YFP gene by Cas9-YFPVLPs

Molecular engineering of viral structures allows the generation of viruses/VLPs that can incorporate a protein of interest. Amongst numerous examples can be cited the design of an HIV- 1 clone incorporating a fluorescent gene allowing an easy monitoring of infection (Dale et al., 201 1 , Methods San Diego Calif, Vol. 53 : 20-26), VLPs harboring viral epitopes useful for vaccination purposes in animal (Garrone et al., 201 1 , Sci Transl Med, Vol. 3 : 94ra71), or VLPs used to deliver their proteic functionnal cargo in recipient cells (Voelkel et al., 2010, Proc Natl Acad Sci USA, Vol. 107: 7805-7810), (Mangeot et al., 201 1 , Mol Ther J Am Soc Gene Ther, Vol. 19: 1656-1666). To achieve the production of efficient Cas9-VLPs, Cas9 was fused to the structural GAG protein of murine Leukemia Virus (MLV) as previously described (Voelkel et al., 2010, Supra). Basically, expression of this chimeric protein with a viral protease (Pro) and an envelope in HEK293T cells is expected to produce VLPs incorporating a Cas9 moiety in their viral core, Cas9 being cleaved from the GAG platform by the viral protease.

Considering the affinity of Cas9 for gRNAs, we further assumed that expression of gRNAs in VLP -producing cells could be sufficient to allow their incorporation within particles which then would be able to vehicle all the components of the CRISPR machinery. To check these hypothesis, we designed gRNAs expressing plasmids to target the YFP gene and attempted to incorporate the gRNAs into Cas9-VLPs produced from HEK293T cells. A schema recapitulating this approach and the constructs used is depicted in Figure 1. VLP-containing supernatants were next introduced in the medium of murine L929 cells expressing a stable version of YFP and cleavage of the fluorescent gene was next investigated by a surveyor assay. Results shown in Figure 2 indicates that Cas9-VLPs delivered the CRISPR machinery and allowed the cleavage of the YFP gene at the expected position defined by the incorporated gRNA. This disruption of the gene was associated with a dramatic and irreversible loss of fluorescence in the treated population resulting from the expected rupture of the YFP-reading frame (Figure 3). These observations validated the use of Cas9- VLPs as a potent delivery agent of the CRISPR components.

Example 2: Disruption of the Myd88 gene in HEK293T and Hela recipient cells

We further explored the capacity of VLPs to incorporate several gRNAs in order to mediate the deletion of the hMyd88 gene. Myd88 is a crucial adapter protein transmitting the signal of most TLRs to activate the transcription of nuclear factors and is notably implicated in the survival of macrophages in certain conditions (Lombardo et al., 2007, J Immunol Baltim Md 1950, Vol. 178: 3731-3739). Two gRNAs were designed to mediate two distinct cleavages in the human Myd88 gene resulting in a deletion of the endogenous gene (Figure 4A-B). Cas9-Myd88VLPs were produced upon transfection of different combinations of plasmids (described in Material and method). Released particles were next concentrated and used to alter the Myd88 gene in different recipient cells including Hela and HEK293T. Results shown Figure 4 C and D indicate that all Cas9-VLPs types loaded with Myd88 gRNAs were efficient in deleting the expected portion of the Myd88 gene in HEK293T cells. However, Hela cells seemed more reluctant and were poorly modified by particular VLP species. Notably, we noted that VLPs devoid of protease (R4) were inefficient in Hela cells while remaining fully active in HEK293T target cells. These data suggest that the VLP recipe should be optimized for each targeted cell type. Additional experiments show that the effect of Cas9-Myd88VLPs was dose dependent (Figure 5) and is potentiated by polybrene addition (Figure 6).

Example 3: VLP-mediated disruption of the Myd88 gene in primary cells of human and murine origin. A more challenging issue is to deliver CRISPRs components into primary cells which are hardly permissive to conventional methods of transfection and may be difficult to transduce by viral vectors. Efficacy of Cas9-Myd88VLPs was thus monitored in different cell types freshly isolated from living organisms including human macrophages derived from human monocytes. Genotype analyses of treated cells reveals an obvious and very efficient cleavage of the Myd88 gene by a single administration of Cas9-Myd88VLPs in cultured macrophages (Figure 7A). Beyond the genotype PCR-based assay which ascertains the cleavage of the gene, Myd88 disruption was responsible for a strong phenotype as revealed in Figure 7B, confirming the inactivation of the Myd88 function. The amazing efficiency of VLPs to deliver CRISPRs in primary cells was further verified in human non activated-lymphocytes typically reluctant to most existing gene- modification techniques (Figure 8). To generalize our observations, we designed another couple of gRNAs targeting the murine Myd88 gene and prepared a VLP batch dedicated to murine cells. This new VLP batch was used to deliver the CRISPRs RNPc in macrophages of murine origin with a high efficacy as indicated in Figure 9. Altogether these results validate Cas9-VLPs as efficient agents to introduce the functional CRISPRs machinery in primary cells.

Example 4: Cas9-VLP can mediate the transfer of a reparation template: Generation of 'all in one' VLPs complexes.

Previous works related the ability of MLV-derived VLPs and other VSV-G-induced particles to mediate the delivery of plasmids into human cells and to serve as viral-derived transfection agent. (Okimoto et al., 2001, Mol Ther J Am Soc Gene Ther, Vol. 4: 232-238). Since particles can be combined with dsDNA molecules, we reasoned that MLV-derived Cas9-VLPs could support a combination with ssDNA and mediate their delivery into cells. We took advantage of this knowledge and tried to combine Cas9-VLPs with a reparation primer composed of ssDNA. By this approach we propose to use VLPs to cleave an endogenous gene and to have it repaired in the cell by a homologous recombination-like mechanism (HR) using the provided reparation template. This was investigated using the DDX3 human gene as a model and a reparation primer designed to insert the FLAG sequence upstream of the ATG codon of the endogenous DDX3 gene. Figure 10 depicts the principle of this 'All in one' VLP strategy that we have directly investigated both in transformed cells and primary cells. Cas9-DDX3VLPs combined with the Flag reparation primer successfully cleaved the DDX3 gene (not shown) and allowed the genetic targeted insertion of the flag sequence at the correct predicted site in the 5 '-sequence of DDX3. This was checked by a PCR-based genotyping assay and the detection of the flagged DDX3 protein by Western Blot (Figure IOC). This result was also verified at the genetic level in primary human dendritic cells exposed to a single treatment of Cas9-DDX3VLPs.

Example 5: Characterization of CAS9 virus-derived particles.

CAS9 VLPS were produced as disclosed in the Materials and Methods section and concentrated by a first Ultracentrifugation on a 20%sucrose cushion. Resulting pellet was next resuspended in PBS and recentrifuged on two sucrose cushions: a 50% sucrose cushion at the bottom of the tube and a 20%-sucrose cushion separating the 50% cushion from the sample. After 2h of centrifugation the interface separating the 50% and the 20% was harvested and recentrifuged to obtain highly pure CAS9- VLPs resuspended in PBS. 10 ug of VLPs were lysed in Laemlli buffer and heat at 95°C during 5 min before western blot analysis.

Western blot analysis is represented in Figure 12 A (lOug per lane). Antibodies used were directed againt GAGmlv (ABCAM R187), against VSVG (ABCAM P5D4), against CAS9 (7A9- 3A3 clone Cell Signaling), against the Flag sequence engrafted on Cas9 (Sigma). Different forms of mlv GAG are revealed, corresponding to the cleaved products processed by the viral Protease. VSVG is clearly detected in the particle preparation at its expected size. CAS9 antibody reveals a higher product above 200KDa which corresponds to the GAG-CAS9 fusion (225 KDA expected), a protein that is also detected by the Flag antibody (left panel). Both CAS9 and FLAG antibodies reveals smaller CAS9 products (ranging from 160 to 200 KDa) that might corresponds to the free CAS9 protein or cleaved CAS9 products, released from GAG after protease processing.

As it is illustrated in Figure 12B, 30 ug of total highly pure VLPs were loaded on a discontinuous sucrose gradient (10%-60% sucrose in PBS) in a total volume of 12ml. After 16h of centrifugation (25000 rpm SW41), 500ul fractions were collected from the top of the tube and named 1-24. 2ul of each fraction were next spotted onto a nitrocellulose membrane immediately blocked by milk addition (TBST 5% low-fat Milk). Similar antibodies described above were next used to detect VSVG CAS9 and GAG MLV for each fractions. Results indicate that CAS9 VLPs sedimented at a density between 1 ,14 and 1 ,21 with a peak at 1 ,17. Example 6: Loading of guide RNAs in the CAS9 virus-derived particles

Example 6 shows that the CAS9 virus-derived particles efficiently integrate guide RNAs.

Northern blot directed against the conserved region of the guideRNA using total RNA extracted from producer cells (lanes 2 to 4) or the corresponding purified VLPs (lanes 5 to 7). Lane 1. Control sample corresponding to total RNA of cells that do not produce VLPs. Lane 2. Total RNA from cells expressing the Gag/Cas9 fusion, viral envelope and guideRNA. Lane 3. Total RNA from cells expressing the Gag/Cas9 fusion, viral envelope and a modified guideRNA with a longer stem structure. Lane4. Total RNA from cells expressing wild-type Cas9 and the guideRNA in absence of Gag. Lanes 5, 6 and 7. Total RNA extracted from the supernatant of the corresponding producer cells (Lane 5 corresponds to the supernatant of cells from lane 2 and so on) after clearing cellular debris and filtrating on a 0.8μηι filter. Lane7 shows that when the Gag/Cas9 fusion is not expressed, the guideRNA is not efficiently incorporated within particles. Interestingly, the modified guideRNA with a longer stem structure (lanes 3 and 6) does not appear to be incorporated more efficiently into VLPs than the wild- type guideRNA (lanes 2 and 5).

Example 7: Comparison of MLV-based virus-derived particles with HIV-based virus- derived particles

Figure 14A illustrates a schematic representation of the coding cassettes designed for the production of MLV-based VLPs or HIV-1- based VLPs. Both cassettes were incorporated in an eucaryotic expression vector equiped with the early hCMV promoter, the rabbit-Bglobin intron and the rabbit pA signal. Both systems were optimized by exploration and test of diverse proteolytic sites separating the GAG cassette from the Cas9 gene. MLV based VLPs were produced as described in the Materials and Methods section while HIV-1 based VLPs were produced similarly except that an HIV-1 helper construct (construct of SEQ ID NO. 33) encoding GAG POL Tat Rev proteins was transfected instead of the MLV GAG POL plasmid. Production of HIV-1 VLPs follows the same procedure as compared with MLV-based VLPs. Figure 14B illustrates the test of concentrated VLPs engineered to incorporate a guide RNA targeting the GFP gene were used to transduce 30000 HEK293T cells expressing GFP. HIV-1 and MLV-based particles were produced with the same loaded gRNA (target sequence: CGAGGAGCTGTTCACCGGGG - SEQ ID NO. 35). Recipient cells were plated the day before in a 96-w plate. Transduction medium was supplemented with polybrene (4ug/ml). 72 hours after treatment with 3 increasing doses of each VLP -batch, fluorescence intensities were measured by a Fluorometer (Excitation 488, Emission 535). Fluorescence decrease was evident in VLPs-treated cells as compared with control non-treated cells (C), revealing the cleavage of the GFP gene within recipient cells. Results indicate that HIV-1 based VLPs are efficient in delivering the CRISPR/CAS9 system to a level slighly less efficient than MLV-based VLPs in these recipient cells (1,5-2 fold less efficient).

Figure 14C illustrates the cleavage of the WASP gene in primary human T cells stimulated with IL7. For this experiment, two guide RNAs targeting the human WASP gene were incorporated within HIV- 1 or MLV-based VLPs before treatment of freshly purified T-cells stimulated with IL7. WASP deletion by CRISPR-CAS9 was next measured by PCR in recipient cells 24 hours after treatment. Gel analysis performed using the ImageJ software allowed a quantification of double-cutting efficiencies for MLV-based VLPs (32%) and HIV- 1 -based VLPs (6%).

Example 8: CRISPR delivery into Thyl-GFP mouse embryos by Cas9-containing virus- derived particles.

Cas9 VLPs incorporating a guide RNA targeting the GFP gene were produced and highly purified before injection into the zona pellucida of mouse embryos (stage 1-cell). Heterozygous embryos were all carrying the Thyl-GFP allele responsible for GFP expression in motoneurons. The aim of the study is to evaluate the capacity of VLPs to cleave GFP within embryos and to generate animals altered in their Thyl-GFP cassette after reimplantation of VLP -treated embryos into female mice. Few nanoliters of a preparation (6,5 uM Cas9) were used for two rounds of injections performed without perforing the cell membrane as depicted in A. No embryo died upon this injection protocol. After reimplantion we obtained a total of 20 animals (F0). Genomic DNAs from new borns-fingers were extracted and analysed by a T7-endonuclease assay revealing the cleavage of the GFP cassette. As shown in B, 6 animals amongst 20 were positive for the assay (arrow) and 4/9 for the first injection experiment (left panel): animals 5,7,8,12 and animal 40 and 45 (weak) for the second injection. Animals 7 8 and 12 were next crossed with wt-C57B6 animals to evaluate the transmission of the cleaved GFP allele to descendants. Roughly half of the Fl- descendance was noted to be heterozygous for the Thyl-GFP allele (for all 3 founders) as expected. The state of the Thyl-GFP allele in heterozygous Fl mice was next measured by a T7- endonuclease assay shown in C*.GFP was shown to be altered in all Fl heterozygous descendants of mice 7 and mice 12 and 33% of descendants of mice 8. Sequencing of the Thyl-GFP allele was next performed on the allele of animals #78 #79 #21 and #22 and chromatograms were compared to a sequence obtained for a Thyl-GFP non treated animal. TIDE software was used for this purpose and provided histograms describing the nature of indels for each animal and the % of sequence alteration**. Results given in D E F and G indicate the % of GFP alteration in Fl- mice***. Altogether these date show that Cas9-VLPs can assist animal transgenesis and be used as CRISPR-delivering agents to alter genes into mammal embryos without transfer of genetic material nor harming the egg-cell.

Protocol of T7-endonuclease assay: Mouse genomic DNAs were extracted from mouse fingers using the Nucleospin Tissue Kit (Macherey Nagel). 3ul of DNA template were next used in a 50ul-PCR reaction (PCR conditions are: 95°C 5min followed by 3 cycles of (95° 30sec-64° 30 sec-72° 30sec) and 25 cycles of (95° 30sec-57° 30sec-72° 30 sec) followed by 5min at 72°C using primers: Forward: 5'- TCTGAGTGGCAAAGGACCTTAGG (Thyl primer - SEQ ID NO. 39)

Reverse: 5'- GAAGTCGTGCTGCTTCATGTGGTCGG (GFP primer - SEQ ID NO. 40)

Thyl-GFP Amplicons were next submitted to the T7endonuclease assay (NEB) as described by the manufacturer in a 40ul reaction tube.

(https://www.neb.eom/protocols/2014/08/l l/determining-genome-targeting-efficiency-using-t7- endonuclease-i) Digestions were finally loaded on a 2,5%-agarose gel.

** TIDE software is a free online tool: https://tide.nki.nl/. Chromatograms sequence (abi files) were uploaded into the sofware and the TIDE runs performed without modification of default settings. TIDE histograms are given

*** % is never complete due to the fact that the chosen Thyl-GFP line carries several copies of GFP/allele (6 to 10). Results should be reproduced in a mouse line bearing one single constitutive GFP copy per allele, which is under preparation.

Other sequences

GAG-Cas9 Amino acid sequence (SEP ID NO. 22):

MGQAVTTPLSLTLDHWKDVERTAHNLSVEVRKRRWVTFCSAEWPTFNVGWPRDGTFNP

DIITQVKIKVFSPGPHGHPDQVPYIVTWEAIAVDPPPWVRPFVHPKPPLSLPPSAPS LPPEPP

LSTPPQSSLYPALTSPLNTKPRPQVLPDSGGPLIDLLTEDPPPYRDPGPPSPDGNGD SGEVA

PTEGAPDPSPMVSRLRGRKEPPVADSTTSQAFPLRLGGNGQYQYWPFSSSDLYNWKN NN

PSFSEDPAKLTALIESVLLTHQPTWDDCQQLLGTLLTGEEKQRVLLEARKAVRGEDG RPT

QLPNDINDAFPLERPDWDYNTQRGRNHLVHYRQLLLAGLQNAGRSPTNLAKVKGITQ GP

NESPSAFLERLKEAYRRYTPYDPEDPGQETNVAMSFIWQSAPDIGRKLERLEDLKSK TLG

DLVREAEKIFNKRETPEEREERIRRETEEKEERRRAEDVQREKERDRRRHREMSKLL ATV

VSGQRQDRQGGERRRPQLDHDQCAYCKEKGHWARDCPKKPRGPRGPRPQASLLTRSS L

YPALTPTGDYKDDDDKMAPKKKRKVGIHGVPAADKKYSIGLDIGTNSVGWAVITDEY K

VPSKKFKVLGNTDRHSIKKNLIGALLFDSGETAEATRLKRTARRRYTRRKNRICYLQ EIFS

NEMAKVDDSFFHRLEESFLVEEDKKHERHPIFGNIVDEVAYHEKYPTIYHLRKKLVD STD

KADLRLIYLALAHMIKFRGHFLIEGDLNPDNSDVDKLFIQLVQTYNQLFEENPINAS GVDA

KAILSARLSKSRRLENLIAQLPGEKKNGLFGNLIALSLGLTPNFKSNFDLAEDAKLQ LSKD

TYDDDLDNLLAQIGDQYADLFLAAKNLSDAILLSDILRVNTEITKAPLSASMIKRYD EHH

QDLTLLKALVRQQLPEKYKEIFFDQSKNGYAGYIDGGASQEEFYKFIKPILEKMDGT EELL

VKLNREDLLRKQRTFDNGSIPHQIHLGELHAILRRQEDFYPFLKDNREKIEKILTFR IPYYV

GPLARGNSRFAWMTRKSEETITPWNFEEVVDKGASAQSFIERMTNFDKNLPNEKVLP KH

SLLYEYFTVYNELTKVKYVTEGMRKPAFLSGEQKKAIVDLLFKTNRKVTVKQLKEDY FK

KIECFDSVEISGVEDRFNASLGTYHDLLKIIKDKDFLDNEENEDILEDIVLTLTLFE DREMIE

ERLKTYAHLFDDKVMKQLKRRRYTGWGRLSRKLINGIRDKQSGKTILDFLKSDGFAN RN

FMQLIHDDSLTFKEDIQKAQVSGQGDSLHEHIANLAGSPAIKKGILQTVKVVDELVK VMG

RHKPENIVIEMARENQTTQKGQKNSRERMKRIEEGIKELGSQILKEHPVENTQLQNE KLY

LYYLQNGRDMYVDQELDINRLSDYDVDHIVPQSFLKDDSIDNKVLTRSDKNRGKSDN VP

SEEVVKKMKNYWRQLLNAKLITQRKFDNLTKAERGGLSELDKAGFIKRQLVETRQIT KH

VAQILDSRMNTKYDENDKLIREVKVITLKSKLVSDFRKDFQFYKVREINNYHHAHDA YL

NAVVGTALIKKYPKLESEFVYGDYKVYDVRKMIAKSEQEIGKATAKYFFYSNIMNFF KT

EITLANGEIRKRPLIETNGETGEIVWDKGRDFATVRKVLSMPQVNIVKKTEVQTGGF SKES

ILPKRNSDKLIARKKDWDPKKYGGFDSPTVAYSVLWAKVEKGKSKKLKSVKELLGIT I

MERSSFEKNPIDFLEAKGYKEVKKDLIIKLPKYSLFELENGRKRMLASAGELQKGNE LAL

PSKYVNFLYLASHYEKLKGSPEDNEQKQLFVEQHKHYLDEIIEQISEFSKRVILADA NLDK

VLSAYNKHRDKPIREQAENIIHLFTLTNLGAPAAFKYFDTTIDRKRYTSTKEVLDAT LIHQ

SITGLYETRIDLSQLGGDKRPAATKKAGQAKKKKARA

VSV-G Amino acid sequence (Sequence ID NO. 23):

MKCLLYLAFLFIGVNCKFTIVFPHNQKGNWKNVPSNYHYCPSSSDLNWHNDLIGTAIQV

KMPKSHKAIQADGWMCHASKWVTTCDFRWYGPKYITQSIRSFTPSVEQCKESIEQTK QG

TWLNPGFPPQSCGYATVTDAEAVIVQVTPHHVLVDEYTGEWVDSQFINGKCSNYICP TV

HNSTTWHSDYKVKGLCDSNLISMDITFFSEDGELSSLGKEGTGFRSNYFAYETGGKA CK

MQYCKHWGVRLPSGVWFEMADKDLFAAARFPECPEGSSISAPSQTSVDVSLIQDVER ILD YSLCQETWSKIRAGLPISPVDLSYLAPKNPGTGPAFTIINGTLKYFETRYIRVDIAAPIL SRM VGMISGTTTERELWDDWAPYEDVEIGPNGVLRTS SGYKFPLYMIGHGMLDSDLHLS SKA QVFEHPHIQDAASQLPDDESLFFGDTGLSKNPIELVEGWFSSWKSSIASFFFIIGLIIGL FLVL RVGIHLCIKLKHTKKRQIYTDIEMNRLGK

GAG PRO POL Amino acid sequence (Sequence ID NO. 24):

MGQAVTTPLSLTLDHWKDVERTAHNLSVEVRKRRWVTFCSAEWPTFNVGWPRDGTFNP

DIITQVKIKVFSPGPHGHPDQVPYIVTWEAIAVDPPPWVRPFVHPKPPLSLPPSAPS LPPEPP

LSTPPQSSLYPALTSPLNTKPRPQVLPDSGGPLIDLLTEDPPPYRDPGPPSPDGNGD SGEVA

PTEGAPDPSPMVSRLRGRKEPPVADSTTSQAFPLRLGGNGQYQYWPFSSSDLYNWKN NN

PSFSEDPAKLTALIESVLLTHQPTWDDCQQLLGTLLTGEEKQRVLLEARKAVRGEDG RPT

QLPNDINDAFPLERPDWDYNTQRGRNHLVHYRQLLLAGLQNAGRSPTNLAKVKGITQ GP

NESPSAFLERLKEAYRRYTPYDPEDPGQETNVAMSFIWQSAPDIGRKLERLEDLKSK TLG

DLVREAEKIFNKRETPEEREERIRRETEEKEERRRAEDVQREKERDRRRHREMSKLL ATV

VSGQRQDRQGGERRRPQLDHDQCAYCKEKGHWARDCPKKPRGPRGPRPQASLLTLDD T

LDDQGGQGQEPPPEPRITLRVGGQPVTFLVDTGAQHSVLTQNPGPLSDKSAWVQGAT GG

KRYRWTTDRRVHLATGKVTHSFLHVPDCPYPLLGRDLLTKLKAQIHFEGSGAQVVGP M

GQPLQVLTLNIEDEYRLHETSKGPDVPLGSTWLSDFPQAWAETGGMGLAVRQAPLII PLK

ATSTPVSIKQYPMSQEARLGIKPHIQRLLDQGILVPCQSPWNTPLLPVKKPGTNDYR PVQD

LREVNKRVEDIHPTVPNPYNLLSGLPPSHQWYTVLDLKDAFFCLRLHPTSQSLFAFE WRD

PEMGISGQLTWTRLPQGFKNSPTLFDEALHRDLADFRIQHPDLILLQYVDDLLLAAT SELD

CQQGTRALLQTLGDLGYRASAKKAQICQKQVKYLGYLLKEGQRWLTEARKETVMGQP

TPKTPRQLREFLGTAGFCRLWIPGFAEMAAPLYPLTKTGTLFEWGPDQQKAYQEIKQ ALL

TAPALGLPDLTKPFELFVDEKQGYAKGVLTQKLGPWRRPVAYLSKKLDPVAAGWPPC L

RMVAAIAVLTKDAGKLTMGQPLVILAPHAVEALVKQPPDRWLSNARMTHYQALLLDT D

RVQFGPIVALNPATLLPLPEEGLQHDCLDILAEAHGTRPDLTDQPLPDADHTWYTDG SSF

LQEGQRKAGAAVTTETEVVWAKALPAGTSAQRAELIALTQALKMAEGKKLNVYTDSR Y

AFATAHIHGEIYRRRGLLTSEGKEIKNKDEILALLKALFLPKRLSIIHCPGHQKGNR AEARG

NRMADQAAREVATRETPETSTLLIENSAPYTHEHFHYTVTDIKDLTKLGATYDDAKK CW

VYQGKPVMPDQFTFELLDFLHQLTHLSFSKTKALLERNYCPYYMLNRDRTLKDITET CQ

ACAQVNASKSAVKQGTRVRGHRPGTHWEIDFTEVKPGLYGYKYLLVFIDTFSGWVEA FP

TKKETAKVVTKKLLEEIFPRFGMPQVLGTDNGPAFVSKVSQTVADLLGVDWKLHCAY RP

QSSGQVERMNRTIKETLTKLTLATGSRDWVLLLPLALYRARNTPGPHGLTPYEILYG APP

PLVNFPDPDMAKVTHNPSLQAHLQALYLVQHEVWRPLAAAYQEQLDRPWPHPFRVGD

TVWVRRHQTKNLEPRWKGPYTVLLTTPTALKVDGIAAWIHAAHVKAADTRIEPPSES TW

RVQRSQNPLKIRLTRGTS*

BAEV-G Amino acid sequence (SEP ID NO. 25)

MGFTTKIIFLYNLVLVYAGFDDPRKAIELVQKRYGRPCDCSGGQVSEPPSDRVSQVTCSG KTAYLMPDQRWKCKSIPKDTSPSGPLQECPCNSYQSSVHSSCYTSYQQCRSGNKTYYTA TLLKTQTGGTSDVQVLGSTNKLIQSPCNGIKGQSICWSTTAPIHVSDGGGPLDTTRIKSV Q RKLEEIHKALYPELQYHPLAIPKVRDNLMVDAQTLNILNATYNLLLMSNTSLVDDCWLC LKLGPPTPLAIPNFLLSYVTRSSDNISCLIIPPLLVQPMQFSNSSCLFSPSYNSTEEIDL GHVA FSNCTSITNVTGPICAVNGSVFLCG NMAYTYLPTNWTGLCVLATLLPDIDIIPGDEPVPIP AIDHFIYRPKRAIQFIPLLAGLGITAAFTTGATGLGVSVTQYTKLSNQLISDVQILSSTI QDL QDQVDSLAEWLQNRRGLDLLTAEQGGICLALQEKCCFYVNKSGIVRDKIKTLQEELERR RKDLASNPLWTGLQGLLPYLLPFLGPLLTLLLLLTIGPCIFNRLTAFINDKLNIIHAM

GAGmlv-CAS9 sequence (SEQ I D NO. 26):

ATGGGCCAGGCTGTTACCACCCCCTTAAGTTTGACTTTAGACCACTGGAAGGATGTC GAACGGACAGCCCACA ACCTGTCG GTAG AG GTT AG A AA A AG GCG CTG G GTT AC ATTCTG CTCTG C AG AATG G CC AACCTTC AACGTCG G ATGGCCACGAGACGGCACTTTTAACCCAGACATTATTACACAGGTTAAGATCAAGGTCTT CTCACCTGGCCCAC ATGGACATCCGGATCAGGTCCCCTACATCGTGACCTGGGAAGCTATAGCAGTAGACCCCC CTCCCTGGGTCAG ACCCTTCGTGCACCCTAAACCTCCCCTCTCTCTTCCCCCTTCAGCCCCCTCTCTCCCACC TGAACCCCCACTCTCGA CCCCGCCCCAGTCCTCCCTCTATCCGGCTCTCACTTCTCCTTTAAACACCAAACCTAGGC CTCAAGTCCTTCCTGA TAGCGGAGGACCACTCATTGATCTACTCACGGAGGACCCTCCGCCTTACCGGGACCCAGG GCCACCCTCTCCT GACGGGAACGGCGATAGCGGAGAAGTGGCCCCTACAGAAGGAGCCCCTGACCCTTCCCCA ATGGTATCCCGC CTGCGGGGAAGAAAAGAACCCCCCGTGGCGGATTCTACTACCTCTCAGGCGTTCCCCCTT CGCCTGGGAGGGA ATGGACAGTATCAATACTGGCCATTTTCCTCCTCTGACCTCTATAACTGGAAAAATAACA ACCCCTCTTTCTCCG AGGACCCAGCTAAATTGACAGCTTTGATCGAGTCCGTTCTCCTTACTCATCAGCCCACTT GGGATGACTGCCAA CAGCTATTAGGGACCCTGCTGACGGGAGAAGAAAAACAGCGAGTGCTCCTAGAGGCCCGA AAGGCGGTTCG AGGGGAGGACGGACGCCCAACTCAGCTGCCCAATGACATTAATGATGCTTTTCCCTTGGA ACGTCCCGACTGG GACTACAACACCCAACGAGGTAGGAACCACCTAGTCCACTATCGCCAGTTGCTCCTAGCG GGTCTCCAAAACG CGGGCAGAAGCCCCACCAATTTGGCCAAGGTAAAAGGGATAACCCAGGGACCTAATGAGT CTCCCTCAGCCTT TTTAGAGAGACTCAAGGAGGCCTATCGCAGATACACTCCTTATGACCCTGAGGACCCAGG GCAAGAAACCAAT GTGGCCATGTCATTCATCTGGCAGTCCGCCCCgGATATCGGGCGAAAGTTAGAGCGGTTA GAAGATTTGAAGA GTAAGACCTTAGGAGACTTAGTGAGGGAAGCTGAAAAGATCTTTAATAAACGAGAAACCC CGGAAGAAAGAG AGGAACGTATTAGGAGAGAAACAGAGGAAAAGGAAGAACGCCGTAGGGCAGAGGATGTGC AGAGAGAGAA GGAGAGGGACCGCAGAAGACATAGAGAAATGAGTAAGTTGCTGGCTACTGTCGTTAGCGG GCAGAGACAGG ATAGACAGGGAGGAGAGCGAAGGAGGCCCCAACTCGACCACGACCAGTGTGCCTACTGCA AAGAAAAGGGA CATTGGGCTAGAGATTGCCCCAAGAAGCCAAGAGGACCCCGGGGACCACGACCCCAGGCC TCCCTCCTGacgc gtagttccctgtatccagccctcacacctaccggtGATTACAAAGACGATGACGATAAGA TGGCCCCAAAGAAGAAGCGGA AGGTCGGTATCCACGGAGTCCCAGCAGCCGACAAGAAGTACAGCATCGGCCTGGACATCG GCACCAACTCTG TGGGCTGGGCCGTGATCACCGACGAGTACAAGGTGCCCAGCAAGAAATTCAAGGTGCTGG GCAACACCGACC GGCACAGCATCAAGAAGAACCTGATCGGAGCCCTGCTGTTCGACAGCGGCGAAACAGCCG AGGCCACCCGGC TGAAGAGAACCGCCAGAAGAAGATACACCAGACGGAAGAACCGGATCTGCTATCTGCAAG AGATCTTCAGCA ACGAGATGGCCAAGGTGGACGACAGCTTCTTCCACAGACTGGAAGAGTCCTTCCTGGTGG AAGAGGATAAGA AGCACGAGCGGCACCCCATCTTCGGCAACATCGTGGACGAGGTGGCCTACCACGAGAAGT ACCCCACCATCTA CCACCTGAGAAAGAAACTGGTGGACAGCACCGACAAGGCCGACCTGCGGCTGATCTATCT GGCCCTGGCCCA CATGATCAAGTTCCGGGGCCACTTCCTGATCGAGGGCGACCTGAACCCCGACAACAGCGA CGTGGACAAGCT GTTCATCCAGCTGGTGCAGACCTACAACCAGCTGTTCGAGGAAAACCCCATCAACGCCAG CGGCGTGGACGCC AAGGCCATCCTGTCTGCCAGACTGAGCAAGAGCAGACGGCTGGAAAATCTGATCGCCCAG CTGCCCGGCGAG AAGAAGAATGGCCTGTTCGGCAACCTGATTGCCCTGAGCCTGGGCCTGACCCCCAACTTC AAGAGCAACTTCG ACCTGGCCGAGGATGCCAAACTGCAGCTGAGCAAGGACACCTACGACGACGACCTGGACA ACCTGCTGGCCC AGATCGGCGACCAGTACGCCGACCTGTTTCTGGCCGCCAAGAACCTGTCCGACGCCATCC TGCTGAGCGACAT CCTGAGAGTGAACACCGAGATCACCAAGGCCCCCCTGAGCGCCTCTATGATCAAGAGATA CGACGAGCACCAC CAGGACCTGACCCTGCTGAAAGCTCTCGTGCGGCAGCAGCTGCCTGAGAAGTACAAAGAG ATTTTCTTCGACC AGAGCAAGAACGGCTACGCCGGCTACATTGACGGCGGAGCCAGCCAGGAAGAGTTCTACA AGTTCATCAAGC CCATCCTGGAAAAGATGGACGGCACCGAGGAACTGCTCGTGAAGCTGAACAGAGAGGACC TGCTGCGGAAG CAGCGGACCTTCGACAACGGCAGCATCCCCCACCAGATCCACCTGGGAGAGCTGCACGCC ATTCTGCGGCGGC AGGAAGATTTTTACCCATTCCTGAAGGACAACCGGGAAAAGATCGAGAAGATCCTGACCT TCCGCATCCCCTA CTACGTGGGCCCTCTGGCCAGGGGAAACAGCAGATTCGCCTGGATGACCAGAAAGAGCGA GGAAACCATCAC CCCCTGGAACTTCGAGGAAGTGGTGGACAAGGGCGCTTCCGCCCAGAGCTTCATCGAGCG GATGACCAACTTC GATAAGAACCTGCCCAACGAGAAGGTGCTGCCCAAGCACAGCCTGCTGTACGAGTACTTC ACCGTGTATAACG AGCTGACCAAAGTGAAATACGTGACCGAGGGAATGAGAAAGCCCGCCTTCCTGAGCGGCG AGCAGAAAAAG GCCATCGTGGACCTGCTGTTCAAGACCAACCGGAAAGTGACCGTGAAGCAGCTGAAAGAG GACTACTTCAAG AAAATCGAGTGCTTCGACTCCGTGGAAATCTCCGGCGTGGAAGATCGGTTCAACGCCTCC CTGGGCACATACC ACGATCTGCTGAAAATTATCAAGGACAAGGACTTCCTGGACAATGAGGAAAACGAGGACA TTCTGGAAGATA TCGTGCTGACCCTGACACTGTTTGAGGACAGAGAGATGATCGAGGAACGGCTGAAAACCT ATGCCCACCTGTT CGACGACAAAGTGATGAAGCAGCTGAAGCGGCGGAGATACACCGGCTGGGGCAGGCTGAG CCGGAAGCTGA TCAACGGCATCCGGGACAAGCAGTCCGGCAAGACAATCCTGGATTTCCTGAAGTCCGACG GCTTCGCCAACAG AAACTTCATGCAGCTGATCCACGACGACAGCCTGACCTTTAAAGAGGACATCCAGAAAGC CCAGGTGTCCGGC CAGGGCGATAGCCTGCACGAGCACATTGCCAATCTGGCCGGCAGCCCCGCCATTAAGAAG GGCATCCTGCAG ACAGTGAAGGTGGTGGACGAGCTCGTGAAAGTGATGGGCCGGCACAAGCCCGAGAACATC GTGATCGAAAT GGCCAGAGAGAACCAGACCACCCAGAAGGGACAGAAGAACAGCCGCGAGAGAATGAAGCG GATCGAAGAG GGCATCAAAGAGCTGGGCAGCCAGATCCTGAAAGAACACCCCGTGGAAAACACCCAGCTG CAGAACGAGAA GCTGTACCTGTACTACCTGCAGAATGGGCGGGATATGTACGTGGACCAGGAACTGGACAT CAACCGGCTGTCC GACTACGATGTGGACCATATCGTGCCTCAGAGCTTTCTGAAGGACGACTCCATCGACAAC AAGGTGCTGACCA GAAGCGACAAGAACCGGGGCAAGAGCGACAACGTGCCCTCCGAAGAGGTCGTGAAGAAGA TGAAGAACTAC TGGCGGCAGCTGCTGAACGCCAAGCTGATTACCCAGAGAAAGTTCGACAATCTGACCAAG GCCGAGAGAGGC GGCCTGAGCGAACTGGATAAGGCCGGCTTCATCAAGAGACAGCTGGTGGAAACCCGGCAG ATCACAAAGCAC GTGGCACAGATCCTGGACTCCCGGATGAACACTAAGTACGACGAGAATGACAAGCTGATC CGGGAAGTGAAA GTGATCACCCTGAAGTCCAAGCTGGTGTCCGATTTCCGGAAGGATTTCCAGTTTTACAAA GTGCGCGAGATCA ACAACTACCACCACGCCCACGACGCCTACCTGAACGCCGTCGTGGGAACCGCCCTGATCA AAAAGTACCCTAA GCTGGAAAGCGAGTTCGTGTACGGCGACTACAAGGTGTACGACGTGCGGAAGATGATCGC CAAGAGCGAGC AGGAAATCGGCAAGGCTACCGCCAAGTACTTCTTCTACAGCAACATCATGAACTTTTTCA AGACCGAGATTACC CTGGCCAACGGCGAGATCCGGAAGCGGCCTCTGATCGAGACAAACGGCGAAACCGGGGAG ATCGTGTGGGA TAAGGGCCGGGATTTTGCCACCGTGCGGAAAGTGCTGAGCATGCCCCAAGTGAATATCGT GAAAAAGACCGA GGTGCAGACAGGCGGCTTCAGCAAAGAGTCTATCCTGCCCAAGAGGAACAGCGATAAGCT GATCGCCAGAAA GAAGGACTGGGACCCTAAGAAGTACGGCGGCTTCGACAGCCCCACCGTGGCCTATTCTGT GCTGGTGGTGGC CAAAGTGGAAAAGGGCAAGTCCAAGAAACTGAAGAGTGTGAAAGAGCTGCTGGGGATCAC CATCATGGAAA GAAGCAGCTTCGAGAAGAATCCCATCGACTTTCTGGAAGCCAAGGGCTACAAAGAAGTGA AAAAGGACCTGA TCATCAAGCTGCCTAAGTACTCCCTGTTCGAGCTGGAAAACGGCCGGAAGAGAATGCTGG CCTCTGCCGGCGA ACTGCAGAAGGGAAACGAACTGGCCCTGCCCTCCAAATATGTGAACTTCCTGTACCTGGC CAGCCACTATGAG AAGCTGAAGGGCTCCCCCGAGGATAATGAGCAGAAACAGCTGTTTGTGGAACAGCACAAG CACTACCTGGAC GAGATCATCGAGCAGATCAGCGAGTTCTCCAAGAGAGTGATCCTGGCCGACGCTAATCTG GACAAAGTGCTGT CCGCCTACAACAAGCACCGGGATAAGCCCATCAGAGAGCAGGCCGAGAATATCATCCACC TGTTTACCCTGAC CAATCTGGGAGCCCCTGCCGCCTTCAAGTACTTTGACACCACCATCGACCGGAAGAGGTA CACCAGCACCAAA GAGGTGCTGGACGCCACCCTGATCCACCAGAGCATCACCGGCCTGTACGAGACACGGATC GACCTGTCTCAGC TGGGAGGCGACAAGCGTCCTGCTGCTACTAAGAAAGCTGGTCAAGCTAAGAAAAAGAAAG CTAGAGCTTGA

BAEV-G Baboon envelope LESS variant BAEV Iess: (also noted B L in the text and figures) SEQ ID

NO. 27 'referenced in Girard-Gagnepain A et al. 2014. Blood. 2014 Aug 21 ;124(8):1221-31. doi: 10.1182/blood- 2014-02-558163. Epub 2014 Jun 20).

atgggctttaccaccaagattatttttctgtataacctggtgctggtgtatgcgggc tttgatgatccgcgcaaagcgattgaactggtgcagaaac gctatggccgcccgtgcgattgcagcggcggccaggtgagcgaaccgccgagcgatcgcg tgagccaggtgacctgcagcggcaaaaccgcgt atctgatgccggatcagcgctggaaatgcaaaagcattccgaaagataccagcccgagcg gcccgctgcaggaatgcccgtgcaacagctatc agagcagcgtgcatagcagctgctataccagctatcagcagtgccgcagcggcaacaaaa cctattataccgcgaccctgctgaaaacccagac cggcggcaccagcgatgtgcaggtgctgggcagcaccaacaaactgattcagagcccgtg caacggcattaaaggccagagcatttgctggag caccaccgcgccgattcatgtgagcgatggcggcggcccgctggataccacccgcattaa aagcgtgcagcgcaaactggaagaaattcataa agcgctgtatccggaactgcagtatcatccgctggcgattccgaaagtgcgcgataacct gatggtggatgcgcagaccctgaacattctgaacg cgacctataacctgctgctgatgagcaacaccagcctggtggatgattgctggctgtgcc tgaaactgggcccgccgaccccgctggcgattccg aactttctgctgagctatgtgacccgcagcagcgataacattagctgcctgattattccg ccgctgctggtgcagccgatgcagtttagcaacagc agctgcctgtttagcccgagctataacagcaccgaagaaattgatctgggccatgtggcg tttagcaactgcaccagcattaccaacgtgaccgg cccgatttgcgcggtgaacggcagcgtgtttctgtgcggcaacaacatggcgtataccta tctgccgaccaactggaccggcctgtgcgtgctggc gaccctgctgccggatattgatattattccgggcgatgaaccggtgccgattccggcgat tgatcattttatttatcgcccgaaacgcgcgattcag tttattccgctgctggcgggcctgggcattaccgcggcgtttaccaccggcgcgaccggc ctgggcgtgagcgtgacccagtataccaaactgag caaccagctgattagcgatgtgcagattctgagcagcaccattcaggatctgcaggatca ggtggatagcctggcggaagtggtgctgcagaac cgccgcggcctggatctgctgaccgcggaacagggcggcatttgcctggcgctgcaggaa aaatgctgcttttatgtgaacaaaagcggcattgt gcgcgataaaattaaaaccctgcaggaagaactggaacgccgccgcaaagatctggcgag caacccgctgtggaccggcctgcagggcctgct gccgtatctgctgccgtttctgggcccgctgctgaccctgctgctgctgctgaccattgg cccgtgcatttttaaccgcctgaccgcgtttattaacga taaactgaacattattcatgcgatgtaa

VSV-G sequence (SEQ ID NO 28):

ATGAAGTGCCTTTTGTACTTAGCCTTTTTATTCATTGGGGTGAATTGCAAGTTCACC ATAGTTTTTCCACACAAC CAAAAAGGAAACTGGAAAAATGTTCCTTCTAATTACCATTATTGCCCGTCAAGCTCAGAT TTAAATTGGCATAA TGACTTAATAGGCACAGCCTTACAAGTCAAAATGCCCAAGAGTCACAAGGCTATTCAAGC AGACGGTTGGATG TGTCATGCTTCCAAATGGGTCACTACTTGTGATTTCCGCTGGTATGGACCGAAGTATATA ACACATTCCATCCG ATCCTTCACTCCATCTGTAGAACAATGCAAGGAAAGCATTGAACAAACGAAACAAGGAAC TTGGCTGAATCCA GGCTTCCCTCCTCAAAGTTGTGGATATGCAACTGTGACGGATGCCGAAGCAGTGATTGTC CAGGTGACTCCTC ACCATGTGCTGGTTGATGAATACACAGGAGAATGGGTTGATTCACAGTTCATCAACGGAA AATGCAGCAATTA CATATGCCCCACTGTCCATAACTCTACAACCTGGCATTCTGACTATAAGGTCAAAGGGCT ATGTGATTCTAACCT CATTTCCATGGACATCACCTTCTTCTCAGAGGACGGAGAGCTATCATCCCTGGGAAAGGA GGGCACAGGGTTC AGAAGTAACTACTTTGCTTATGAAACTGGAGGCAAGGCCTGCAAAATGCAATACTGCAAG CATTGGGGAGTCA GACTCCCATCAGGTGTCTGGTTCGAGATGGCTGATAAGGATCTCTTTGCTGCAGCCAGAT TCCCTGAATGCCCA GAAGGGTCAAGTATCTCTGCTCCATCTCAGACCTCAGTGGATGTAAGTCTAATTCAGGAC GTTGAGAGGATCTT GGATTATTCCCTCTGCCAAGAAACCTGGAGCAAAATCAGAGCGGGTCTTCCAATCTCTCC AGTGGATCTCAGCT ATCTTGCTCCTAAAAACCCAGGAACCGGTCCTGCTTTCACCATAATCAATGGTACCCTAA AATACTTTGAGACCA GATACATCAGAGTCGATATTGCTGCTCCAATCCTCTCAAGAATGGTCGGAATGATCAGTG GAACTACCACAGA AAGGGAACTGTGGGATGACTGGGCACCATATGAAGACGTGGAAATTGGACCCAATGGAGT TCTGAGGACCA GTTCAGGATATAAGTTTCCTTTATACATGATTGGACATGGTATGTTGGACTCCGATCTTC ATCTTAGCTCAAAGG CTCAGGTGTTCGAACATCCTCACATTCAAGACGCTGCTTCGCAACTTCCTGATGATGAGA GTTTATTTTTTGGTG ATACTGGGCTATCCAAAAATCCAATCGAGCTTGTAGAAGGTTGGTTCAGTAGTTGGAAAA GCTCTATTGCCTCT TTTTTCTTTATC ATAG G GTTA ATC ATTG G ACTATTCTTG GTTCTCCG AGTTG GTATCC ATCTTTG C ATTAA ATTA A AGCACACCAAGAAAAGACAGATTTATACAGACATAGAGATGAACCGACTTGGAAAGTAA

Cleavage site:

Amino acid: SSLYPALTP (SEQ ID NO 29)

Nucleic acid: agttccctgtatccagccctcacacct (SEQ ID NO 30)

Cas9 Amino acid sequence (SEQ ID NO.31) MAPKKKRKVGIHGVPAADKKYSIGLDIGTNSVGWAVITDEYKVPSKKFKVLGNTDRHSIK KNLIGALLFDSGETAEAT RLKRTARRRYTRRKNRICYLQEIFSNEMAKVDDSFFHRLEESFLVEEDKKHERHPIFGNI VDEVAYHEKYPTIYHLRKKL VDSTDKADLRLIYLALAHMIKFRGHFLIEGDLNPDNSDVDKLFIQLVQTYNQLFEENPIN ASGVDAKAILSARLSKSRR LENLIAQLPGEKKNGLFGNLIALSLGLTPNFKSNFDLAEDAKLQLSKDTYDDDLDNLLAQ IGDQYADLFLAAKNLSDAI LLSDILRVNTEITKAPLSASMIKRYDEHHQDLTLLKALVRQQLPEKYKEIFFDQSKNGYA GYIDGGASQEEFYKFIKPILE KMDGTEELLVKLNREDLLRKQRTFDNGSIPHQIHLGELHAILRRQEDFYPFLKDNREKIE KILTFRIPYYVGPLARGNSR FAWMTRKSEETITPWNFEEVVDKGASAQSFIERMTNFDKNLPNEKVLPKHSLLYEYFTVY NELTKVKYVTEGMRKP AFLSGEQKKAIVDLLFKTNRKVTVKQLKEDYFKKIECFDSVEISGVEDRFNASLGTYHDL LKIIKDKDFLDNEENEDILED IVLTLTLFEDREMIEERLKTYAHLFDDKVMKQLKRRRYTGWGRLSRKLINGIRDKQSGKT ILDFLKSDGFANRNFMQL IHDDSLTFKEDIQKAQVSGQGDSLHEHIANLAGSPAIKKGILQTVKVVDELVKVMGRHKP ENIVIEMARENQTTQKG QKNSRERMKRIEEGIKELGSQILKEHPVENTQLQNEKLYLYYLQNGRDMYVDQELDINRL SDYDVDHIVPQSFLKDD SIDNKVLTRSDKNRGKSDNVPSEEVVKKMKNYWRQLLNAKLITQRKFDNLTKAERGGLSE LDKAGFIKRQLVETRQI TKHVAQILDSRMNTKYDENDKLIREVKVITLKSKLVSDFRKDFQFYKVREINNYHHAHDA YLNAVVGTALIKKYPKLE SEFVYGDYKVYDVRKMIAKSEQEIGKATAKYFFYSNIMNFFKTEITLANGEIRKRPLIET NGETGEIVWDKGRDFATVR KVLSMPQVNIVKKTEVQTGGFSKESILPKRNSDKLIARKKDWDPKKYGGFDSPTVAYSVL VVAKVEKGKSKKLKSVK ELLGITIMERSSFEKNPIDFLEAKGYKEVKKDLIIKLPKYSLFELENGRKRMLASAGELQ KGNELALPSKYVNFLYLASHY EKLKGSPEDNEQKQLFVEQHKHYLDEIIEQISEFSKRVILADANLDKVLSAYNKHRDKPI REQAENIIHLFTLTNLGAPA AFKYFDTTIDRKRYTSTKEVLDATLIHQSITGLYETRIDLSQLGGDKRPAATKKAGQAKK KKA

Cas9 Nucleic acid sequence (SEQIDNO.32) ATGGCCCCAAAGAAGAAGCGGAAGGTCGGTATCCACGGAGTCCCAGCAGCCGACAAGAAG TACAGCATCGG CCTGGACATCGGCACCAACTCTGTGGGCTGGGCCGTGATCACCGACGAGTACAAGGTGCC CAGCAAGAAATT CAAGGTGCTGGGCAACACCGACCGGCACAGCATCAAGAAGAACCTGATCGGAGCCCTGCT GTTCGACAGCGG CGAAACAGCCGAGGCCACCCGGCTGAAGAGAACCGCCAGAAGAAGATACACCAGACGGAA GAACCGGATCT GCTATCTGCAAGAGATCTTCAGCAACGAGATGGCCAAGGTGGACGACAGCTTCTTCCACA GACTGGAAGAGTC CTTCCTGGTGGAAGAGGATAAGAAGCACGAGCGGCACCCCATCTTCGGCAACATCGTGGA CGAGGTGGCCTA CCACGAGAAGTACCCCACCATCTACCACCTGAGAAAGAAACTGGTGGACAGCACCGACAA GGCCGACCTGCG GCTGATCTATCTGGCCCTGGCCCACATGATCAAGTTCCGGGGCCACTTCCTGATCGAGGG CGACCTGAACCCC GACAACAGCGACGTGGACAAGCTGTTCATCCAGCTGGTGCAGACCTACAACCAGCTGTTC GAGGAAAACCCCA TCAACGCCAGCGGCGTGGACGCCAAGGCCATCCTGTCTGCCAGACTGAGCAAGAGCAGAC GGCTGGAAAATC TGATCGCCCAGCTGCCCGGCGAGAAGAAGAATGGCCTGTTCGGCAACCTGATTGCCCTGA GCCTGGGCCTGAC CCCCAACTTCAAGAGCAACTTCGACCTGGCCGAGGATGCCAAACTGCAGCTGAGCAAGGA CACCTACGACGAC GACCTGGACAACCTGCTGGCCCAGATCGGCGACCAGTACGCCGACCTGTTTCTGGCCGCC AAGAACCTGTCCG ACGCCATCCTGCTGAGCGACATCCTGAGAGTGAACACCGAGATCACCAAGGCCCCCCTGA GCGCCTCTATGAT CAAGAGATACGACGAGCACCACCAGGACCTGACCCTGCTGAAAGCTCTCGTGCGGCAGCA GCTGCCTGAGAA GTACAAAGAGATTTTCTTCGACCAGAGCAAGAACGGCTACGCCGGCTACATTGACGGCGG AGCCAGCCAGGA AGAGTTCTACAAGTTCATCAAGCCCATCCTGGAAAAGATGGACGGCACCGAGGAACTGCT CGTGAAGCTGAAC AGAGAGGACCTGCTGCGGAAGCAGCGGACCTTCGACAACGGCAGCATCCCCCACCAGATC CACCTGGGAGAG CTGCACGCCATTCTGCGGCGGCAGGAAGATTTTTACCCATTCCTGAAGGACAACCGGGAA AAGATCGAGAAG ATCCTGACCTTCCGCATCCCCTACTACGTGGGCCCTCTGGCCAGGGGAAACAGCAGATTC GCCTGGATGACCA GAAAGAGCGAGGAAACCATCACCCCCTGGAACTTCGAGGAAGTGGTGGACAAGGGCGCTT CCGCCCAGAGCT TCATCGAGCGGATGACCAACTTCGATAAGAACCTGCCCAACGAGAAGGTGCTGCCCAAGC ACAGCCTGCTGTA CGAGTACTTCACCGTGTATAACGAGCTGACCAAAGTGAAATACGTGACCGAGGGAATGAG AAAGCCCGCCTTC CTGAGCGGCGAGCAGAAAAAGGCCATCGTGGACCTGCTGTTCAAGACCAACCGGAAAGTG ACCGTGAAGCAG CTGAAAGAGGACTACTTCAAGAAAATCGAGTGCTTCGACTCCGTGGAAATCTCCGGCGTG GAAGATCGGTTCA ACGCCTCCCTGGGCACATACCACGATCTGCTGAAAATTATCAAGGACAAGGACTTCCTGG ACAATGAGGAAAA CGAGGACATTCTGGAAGATATCGTGCTGACCCTGACACTGTTTGAGGACAGAGAGATGAT CGAGGAACGGCT GAAAACCTATGCCCACCTGTTCGACGACAAAGTGATGAAGCAGCTGAAGCGGCGGAGATA CACCGGCTGGGG CAGGCTGAGCCGGAAGCTGATCAACGGCATCCGGGACAAGCAGTCCGGCAAGACAATCCT GGATTTCCTGAA GTCCGACGGCTTCGCCAACAGAAACTTCATGCAGCTGATCCACGACGACAGCCTGACCTT TAAAGAGGACATC CAGAAAGCCCAGGTGTCCGGCCAGGGCGATAGCCTGCACGAGCACATTGCCAATCTGGCC GGCAGCCCCGCC ATTAAGAAGGGCATCCTGCAGACAGTGAAGGTGGTGGACGAGCTCGTGAAAGTGATGGGC CGGCACAAGCC CGAGAACATCGTGATCGAAATGGCCAGAGAGAACCAGACCACCCAGAAGGGACAGAAGAA CAGCCGCGAGA GAATGAAGCGGATCGAAGAGGGCATCAAAGAGCTGGGCAGCCAGATCCTGAAAGAACACC CCGTGGAAAAC ACCCAGCTGCAGAACGAGAAGCTGTACCTGTACTACCTGCAGAATGGGCGGGATATGTAC GTGGACCAGGAA CTGGACATCAACCGGCTGTCCGACTACGATGTGGACCATATCGTGCCTCAGAGCTTTCTG AAGGACGACTCCAT CGACAACAAGGTGCTGACCAGAAGCGACAAGAACCGGGGCAAGAGCGACAACGTGCCCTC CGAAGAGGTCG TGAAGAAGATGAAGAACTACTGGCGGCAGCTGCTGAACGCCAAGCTGATTACCCAGAGAA AGTTCGACAATC TGACCAAGGCCGAGAGAGGCGGCCTGAGCGAACTGGATAAGGCCGGCTTCATCAAGAGAC AGCTGGTGGAA ACCCGGCAGATCACAAAGCACGTGGCACAGATCCTGGACTCCCGGATGAACACTAAGTAC GACGAGAATGAC AAGCTGATCCGGGAAGTGAAAGTGATCACCCTGAAGTCCAAGCTGGTGTCCGATTTCCGG AAGGATTTCCAGT TTTACAAAGTGCGCGAGATCAACAACTACCACCACGCCCACGACGCCTACCTGAACGCCG TCGTGGGAACCGC CCTGATCAAAAAGTACCCTAAGCTGGAAAGCGAGTTCGTGTACGGCGACTACAAGGTGTA CGACGTGCGGAA GATGATCGCCAAGAGCGAGCAGGAAATCGGCAAGGCTACCGCCAAGTACTTCTTCTACAG CAACATCATGAAC TTTTTCAAGACCGAGATTACCCTGGCCAACGGCGAGATCCGGAAGCGGCCTCTGATCGAG ACAAACGGCGAA ACCGGGGAGATCGTGTGGGATAAGGGCCGGGATTTTGCCACCGTGCGGAAAGTGCTGAGC ATGCCCCAAGTG AATATCGTGAAAAAGACCGAGGTGCAGACAGGCGGCTTCAGCAAAGAGTCTATCCTGCCC AAGAGGAACAGC GATAAGCTGATCGCCAGAAAGAAGGACTGGGACCCTAAGAAGTACGGCGGCTTCGACAGC CCCACCGTGGCC TATTCTGTGCTGGTGGTGGCCAAAGTGGAAAAGGGCAAGTCCAAGAAACTGAAGAGTGTG AAAGAGCTGCTG GGGATCACCATCATGGAAAGAAGCAGCTTCGAGAAGAATCCCATCGACTTTCTGGAAGCC AAGGGCTACAAA GAAGTGAAAAAGGACCTGATCATCAAGCTGCCTAAGTACTCCCTGTTCGAGCTGGAAAAC GGCCGGAAGAGA ATGCTGGCCTCTGCCGGCGAACTGCAGAAGGGAAACGAACTGGCCCTGCCCTCCAAATAT GTGAACTTCCTGT ACCTGGCCAGCCACTATGAGAAGCTGAAGGGCTCCCCCGAGGATAATGAGCAGAAACAGC TGTTTGTGGAAC AGCACAAGCACTACCTGGACGAGATCATCGAGCAGATCAGCGAGTTCTCCAAGAGAGTGA TCCTGGCCGACG CTAATCTGGACAAAGTGCTGTCCGCCTACAACAAGCACCGGGATAAGCCCATCAGAGAGC AGGCCGAGAATA TCATCCACCTGTTTACCCTGACCAATCTGGGAGCCCCTGCCGCCTTCAAGTACTTTGACA CCACCATCGACCGGA AGAGGTACACCAGCACCAAAGAGGTGCTGGACGCCACCCTGATCCACCAGAGCATCACCG GCCTGTACGAGA CACGGATCGACCTGTCTCAGCTGGGAGGCGACAAGCGTCCTGCTGCTACTAAGAAAGCTG GTCAAGCTAAGA AAAAG AAAG CTAG

HIV-1 encapsidation construct "p8.91" (encoding HIV-1 Gag-Pol-Tat-Rev) (SEQIDNO. 33) ttgattattgactagttattaatagtaatcaattacggggtcattagttcatagcccata tatggagttccgcgttacataacttacggtaaatggcc cgcctggctgaccgcccaacgacccccgcccattgacgtcaataatgacgtatgttccca tagtaacgccaatagggactttccattgacgtcaat gggtggagtatttacggtaaactgcccacttggcagtacatcaagtgtatcatatgccaa gtacgccccctattgacgtcaatgacggtaaatggc ccgcctggcattatgcccagtacatgaccttatgggactttcctacttggcagtacatct acgtattagtcatcgctattaccatggtgatgcggtttt ggcagtacatcaatgggcgtggatagcggtttgactcacggggatttccaagtctccacc ccattgacgtcaatgggagtttgttttggcaccaaa atcaacgggactttccaaaatgtcgtaacaactccgccccattgacgcaaatgggcggta ggcgtgtacggtgggaggtctatataagcagagct cgtttagtgaaccgtcagatcgcctggagacgccatccacgctgttttgacctccataga agacaccgggaccgatccagcctccgcggccggga acggtgcattggaacgcggattccccgtgccaagagtgacgtaagtaccgcctatagagt ctataggcccacccccttggcttcttatgcgacgga tcgatcccgtaataagcttcgaggtccgcggccggccgcgttgacgcgcacggcaagagg cgaggggcggcgactggtgagagatgggtgcga gagcgtcagtattaagcgggggagaattagatcgatgggaaaaaattcggttaaggccag ggggaaagaaaaaatataaattaaaacatata gtatgggcaagcagggagctagaacgattcgcagttaatcctggcctgttagaaacatca gaaggctgtagacaaatactgggacagctacaac catcccttcagacaggatcagaagaacttagatcattatataatacagtagcaaccctct attgtgtgcatcaaaggatagagataaaagacacc aaggaagctttagacaagatagaggaagagcaaaacaaaagtaagaaaaaagcacagcaa gcagcagctgacacaggacacagcaatcag gtcagccaaaattaccctatagtgcagaacatccaggggcaaatggtacatcaggccata tcacctagaactttaaatgcatgggtaaaagtagt agaagagaaggctttcagcccagaagtgatacccatgttttcagcattatcagaaggagc caccccacaagatttaaacaccatgctaaacaca gtggggggacatcaagcagccatgcaaatgttaaaagagaccatcaatgaggaagctgca gaatgggatagagtgcatccagtgcatgcagg gcctattgcaccaggccagatgagagaaccaaggggaagtgacatagcaggaactactag tacccttcaggaacaaataggatggatgacaca taatccacctatcccagtaggagaaatctataaaagatggataatcctgggattaaataa aatagtaagaatgtatagccctaccagcattctgg acataagacaaggaccaaaggaaccctttagagactatgtagaccgattctataaaactc taagagccgagcaagcttcacaagaggtaaaaa attggatgacagaaaccttgttggtccaaaatgcgaacccagattgtaagactattttaa aagcattgggaccaggagcgacactagaagaaat gatgacagcatgtcagggagtggggggacccggccataaagcaagagttttggctgaagc aatgagccaagtaacaaatccagctaccataat gatacagaaaggcaattttaggaaccaaagaaagactgttaagtgtttcaattgtggcaa agaagggcacatagccaaaaattgcagggcccct aggaaaaagggctgttggaaatgtggaaaggaaggacaccaaatgaaagattgtactgag agacaggctaattttttagggaagatctggcctt cccacaagggaaggccagggaattttcttcagagcagaccagagccaacagccccaccag aagagagcttcaggtttggggaagagacaaca actccctctcagaagcaggagccgatagacaaggaactgtatcctttagcttccctcaga tcactctttggcagcgacccctcgtcacaataaaga taggggggcaattaaaggaagctctattagatacaggagcagatgatacagtattagaag aaatgaatttgccaggaagatggaaaccaaaaa tgatagggggaattggaggttttatcaaagtaagacagtatgatcagatactcatagaaa tctgcggacataaagctataggtacagtattagta ggacctacacctgtcaacataattggaagaaatctgttgactcagattggctgcacttta aattttcccattagtcctattgagactgtaccagtaa aattaaagccaggaatggatggcccaaaagttaaacaatggccattgacagaagaaaaaa taaaagcattagtagaaatttgtacagaaatgg aaaaggaaggaaaaatttcaaaaattgggcctgaaaatccatacaatactccagtatttg ccataaagaaaaaagacagtactaaatggagaa aattagtagatttcagagaacttaataagagaactcaagatttctgggaagttcaattag gaataccacatcctgcagggttaaaacagaaaaa atcagtaacagtactggatgtgggcgatgcatatttttcagttcccttagataaagactt caggaagtatactgcatttaccatacctagtataaac aatgagacaccagggattagatatcagtacaatgtgcttccacagggatggaaaggatca ccagcaatattccagtgtagcatgacaaaaatct tagagccttttagaaaacaaaatccagacatagtcatctatcaatacatggatgatttgt atgtaggatctgacttagaaatagggcagcataga acaaaaatagaggaactgagacaacatctgttgaggtggggatttaccacaccagacaaa aaacatcagaaagaacctccattcctttggatgg gttatgaactccatcctgataaatggacagtacagcctatagtgctgccagaaaaggaca gctggactgtcaatgacatacagaaattagtggg aaaattgaattgggcaagtcagatttatgcagggattaaagtaaggcaattatgtaaact tcttaggggaaccaaagcactaacagaagtagta ccactaacagaagaagcagagctagaactggcagaaaacagggagattctaaaagaaccg gtacatggagtgtattatgacccatcaaaaga cttaatagcagaaatacagaagcaggggcaaggccaatggacatatcaaatttatcaaga gccatttaaaaatctgaaaacaggaaagtatgc aagaatgaagggtgcccacactaatgatgtgaaacaattaacagaggcagtacaaaaaat agccacagaaagcatagtaatatggggaaaga ctcctaaatttaaattacccatacaaaaggaaacatgggaagcatggtggacagagtatt ggcaagccacctggattcctgagtgggagtttgtc aatacccctcccttagtgaagttatggtaccagttagagaaagaacccataataggagca gaaactttctatgtagatggggcagccaataggg aaactaaattaggaaaagcaggatatgtaactgacagaggaagacaaaaagttgtccccc taacggacacaacaaatcagaagactgagtta caagcaattcatctagctttgcaggattcgggattagaagtaaacatagtgacagactca caatatgcattgggaatcattcaagcacaaccaga taagagtgaatcagagttagtcagtcaaataatagagcagttaataaaaaaggaaaaagt ctacctggcatgggtaccagcacacaaaggaat tggaggaaatgaacaagtagataaattggtcagtgctggaatcaggaaagtactattttt agatggaatagataaggcccaagaagaacatga gaaatatcacagtaattggagagcaatggctagtgattttaacctaccacctgtagtagc aaaagaaatagtagccagctgtgataaatgtcagc taaaaggggaagccatgcatggacaagtagactgtagcccaggaatatggcagctagatt gtacacatttagaaggaaaagttatcttggtagc agttcatgtagccagtggatatatagaagcagaagtaattccagcagagacagggcaaga aacagcatacttcctcttaaaattagcaggaaga tggccagtaaaaacagtacatacagacaatggcagcaatttcaccagtactacagttaag gccgcctgttggtgggcggggatcaagcaggaat ttggcattccctacaatccccaaagtcaaggagtaatagaatctatgaataaagaattaa agaaaattataggacaggtaagagatcaggctga acatcttaagacagcagtacaaatggcagtattcatccacaattttaaaagaaaaggggg gattggggggtacagtgcaggggaaagaatagt agacataatagcaacagacatacaaactaaagaattacaaaaacaaattacaaaaattca aaattttcgggtttattacagggacagcagaga tccagtttggaaaggaccagcaaagctcctctggaaaggtgaaggggcagtagtaataca agataatagtgacataaaagtagtgccaagaag aaaagcaaagatcatcagggattatggaaaacagatggcaggtgatgattgtgtggcaag tagacaggatgaggattaacacatggaattctg caacaactgctgtttatccatttcagaattgggtgtcgacatagcagaataggcgttact cgacagaggagagcaagaaatggagccagtagat cctagactagagccctggaagcatccaggaagtcagcctaaaactgcttgtaccaattgc tattgtaaaaagtgttgctttcattgccaagtttgtt tcatgacaaaagccttaggcatctcctatggcaggaagaagcggagacagcgacgaagag ctcatcagaacagtcagactcatcaagcttctct atcaaagcagtaagtagtacatgtaatgcaacctataatagtagcaatagtagcattagt agtagcaataataatagcaatagttgtgtggtcca tagtaatcatagaatataggaaaatggccgctgatcttcagacctggaggaggagatatg agggacaattggagaagtgaattatataaatata aagtagtaaaaattgaaccattaggagtagcacccaccaaggcaaagagaagagtggtgc agagagaaaaaagagcagtgggaataggagc tttgttccttgggttcttgggagcagcaggaagcactatgggcgcagcgtcaatgacgct gacggtacaggccagacaattattgtctggtatagt gcagcagcagaacaatttgctgagggctattgaggcgcaacagcatctgttgcaactcac agtctggggcatcaagcagctccaggcaagaatc ctggctgtggaaagatacctaaaggatcaacagctcctggggatttggggttgctctgga aaactcatttgcaccactgctgtgccttggaatgct agttggagtaataaatctctggaacagatttggaatcacacgacctggatggagtgggac agagaaattaacaattacacaagcttaatacact ccttaattgaagaatcgcaaaaccagcaagaaaagaatgaacaagaattattggaattag ataaatgggcaagtttgtggaattggtttaacat aacaaattggctgtggtatataaaattattcataatgatagtaggaggcttggtaggttt aagaatagtttttgctgtactttctatagtgaatagag ttaggcagggatattcaccattatcgtttcagacccacctcccaaccccgaggggacccg acaggcccgaaggaatagaagaagaaggtggag agagagacagagacagatccattcgattagtgaacggatccttggcacttatctgggacg atctgcggagcctgtgcctcttcagctaccaccgct tgagagacttactcttgattgtaacgaggattgtggaacttctgggacgcagggggtggg aagccctcaaatattggtggaatctcctacaatatt ggagtcaggagctaaagaatagtgctgttagcttgctcaatgccacagccatagcagtag ctgaggggacagatagggttatagaagtagtaca aggagcttgtagagctattcgccacatacctagaagaataagacagggcttggaaaggat tttgctataagctcgaggccgccccggtgaccttc agaccttggcactggaggtggcccggcagaagcgcggcatcgtggatcagtgctgcacca gcatctgctctctctaccaactggagaactactgc aactaggcccaccactaccctgtccacccctctgcaatgaataaaacctttgaaagagca ctacaagttgtgtgtacatgcgtgcatgtgcatatg tggtgcggggggaacatgagtggggctggctggagtggcgatgataagctgtcaaacatg agaattaattcttgaagacgaaagggcctcgtga tacgcctatttttataggttaatgtcatgataataatggtttcttagtctagaattaatt ccgtgtattctatagtgtcacctaaatcgtatgtgtatga tacataaggttatgtattaattgtagccgcgttctaacgacaatatgtacaagcctaatt gtgtagcatctggcttactgaagcagaccctatcatct ctctcgtaaactgccgtcagagtcggtttggttggacgaaccttctgagtttctggtaac gccgtcccgcacccggaaatggtcagcgaaccaatc agcagggtcatcgctagccagatcctctacgccggacgcatcgtggccggcatcaccggc gccacaggtgcggttgctggcgcctatatcgccga catcaccgatggggaagatcgggctcgccacttcgggctcatgagcgcttgtttcggcgt gggtatggtggcaggccccgtggccgggggactgt tgggcgccatctccttgcatgcaccattccttgcggcggcggtgctcaacggcctcaacc tactactgggctgcttcctaatgcaggagtcgcataa gggagagcgtcgaatggtgcactctcagtacaatctgctctgatgccgcatagttaagcc agccccgacacccgccaacacccgctgacgcgccc tgacgggcttgtctgctcccggcatccgcttacagacaagctgtgaccgtctccgggagc tgcatgtgtcagaggttttcaccgtcatcaccgaaa cgcgcgagacgaaagggcctcgtgatacgcctatttttataggttaatgtcatgataata atggtttcttagacgtcaggtggcacttttcggggaa atgtgcgcggaacccctatttgtttatttttctaaatacattcaaatatgtatccgctca tgagacaataaccctgataaatgcttcaataatattga aaaaggaagagtatgagtattcaacatttccgtgtcgcccttattcccttttttgcggca ttttgccttcctgtttttgctcacccagaaacgctggtg aaagtaaaagatgctgaagatcagttgggtgcacgagtgggttacatcgaactggatctc aacagcggtaagatccttgagagttttcgccccga agaacgttttccaatgatgagcacttttaaagttctgctatgtggcgcggtattatcccg tattgacgccgggcaagagcaactcggtcgccgcata cactattctcagaatgacttggttgagtactcaccagtcacagaaaagcatcttacggat ggcatgacagtaagagaattatgcagtgctgccat aaccatgagtgataacactgcggccaacttacttctgacaacgatcggaggaccgaagga gctaaccgcttttttgcacaacatgggggatcatg taactcgccttgatcgttgggaaccggagctgaatgaagccataccaaacgacgagcgtg acaccacgatgcctgtagcaatggcaacaacgtt gcgcaaactattaactggcgaactacttactctagcttcccggcaacaattaatagactg gatggaggcggataaagttgcaggaccacttctgc gctcggcccttccggctggctggtttattgctgataaatctggagccggtgagcgtgggt ctcgcggtatcattgcagcactggggccagatggta agccctcccgtatcgtagttatctacacgacggggagtcaggcaactatggatgaacgaa atagacagatcgctgagataggtgcctcactgatt aagcattggtaactgtcagaccaagtttactcatatatactttagattgatttaaaactt catttttaatttaaaaggatctaggtgaagatccttttt gataatctcatgaccaaaatcccttaacgtgagttttcgttccactgagcgtcagacccc gtagaaaagatcaaaggatcttcttgagatcctttttt tctgcgcgtaatctgctgcttgcaaacaaaaaaaccaccgctaccagcggtggtttgttt gccggatcaagagctaccaactctttttccgaaggt aactggcttcagcagagcgcagataccaaatactgttcttctagtgtagccgtagttagg ccaccacttcaagaactctgtagcaccgcctacata cctcgctctgctaatcctgttaccagtggctgctgccagtggcgataagtcgtgtcttac cgggttggactcaagacgatagttaccggataaggcg cagcggtcgggctgaacggggggttcgtgcacacagcccagcttggagcgaacgacctac accgaactgagatacctacagcgtgagctatga gaaagcgccacgcttcccgaagggagaaaggcggacaggtatccggtaagcggcagggtc ggaacaggagagcgcacgagggagcttccag ggggaaacgcctggtatctttatagtcctgtcgggtttcgccacctctgacttgagcgtc gatttttgtgatgctcgtcaggggggcggagcctatg gaaaaacgccagcaacgcggcctttttacggttcctggccttttgctggccttttgctca catgttctttcctgcgttatcccctgattctgtggataac cgtattaccgcctttgagtgagctgataccgctcgccgcagccgaacgaccgagcgcagc gagtcagtgagcgaggaagcggaagagcgccca atacgcaaaccgcctctccccgcgcgttggccgattcattaatgcagctgtggaatgtgt gtcagttagggtgtggaaagtccccaggctccccag caggcagaagtatgcaaagcatgcatctcaattagtcagcaaccaggtgtggaaagtccc caggctccccagcaggcagaagtatgcaaagca tgcatctcaattagtcagcaaccatagtcccgcccctaactccgcccatcccgcccctaa ctccgcccagttccgcccattctccgccccatggctg actaattttttttatttatgcagaggccgaggccgcctcggcctctgagctattccagaa gtagtgaggaggcttttttggaggcctaggcttttgca aaaagcttggacacaagacaggcttgcgagatatgtttgagaataccactttatcccgcg tcagggagaggcagtgcgtaaaaagacgcggact catgtgaaatactggtttttagtgcgccagatctctataatctcgcgcaacctattttcc cctcgaacactttttaagccgtagataaacaggctggg acacttcacatgagcgaaaaatacatcgtcacctgggacatgttgcagatccatgcacgt aaactcgcaagccgactgatgccttctgaacaatg gaaaggcattattgccgtaagccgtggcggtctgtaccgggtgcgttactggcgcgtgaa ctgggtattcgtcatgtcgataccgtttgtatttcca gctacgatcacgacaaccagcgcgagcttaaagtgctgaaacgcgcagaaggcgatggcg aaggcttcatcgttattgatgacctggtggatac cggtggtactgcggttgcgattcgtgaaatgtatccaaaagcgcactttgtcaccatctt cgcaaaaccggctggtcgtccgctggttgatgactat gttgttgatatcccgcaagatacctggattgaacagccgtgggatatgggcgtcgtattc gtcccgccaatctccggtcgctaatcttttcaacgcct ggcactgccgggcgttgttctttttaacttcaggcgggttacaatagtttccagtaagta ttctggaggctgcatccatgacacaggcaaacctgag cgaaaccctgttcaaaccccgctttaaacatcctgaaacctcgacgctagtccgccgctt taatcacggcgcacaaccgcctgtgcagtcggccct tgatggtaaaaccatccctcactggtatcgcatgattaaccgtctgatgtggatctggcg cggcattgacccacgcgaaatcctcgacgtccaggc acgtattgtgatgagcgatgccgaacgtaccgacgatgatttatacgatacggtgattgg ctaccgtggcggcaactggatttatgagtgggcccc ggatctttgtgaaggaaccttacttctgtggtgtgacataattggacaaactacctacag agatttaaagctctaaggtaaatataaaatttttaac ccggatctttgtgaaggaaccttacttctgtggtgtgacataattggacaaactacctac agagatttaaagctctaaggtaaatataaaattttta agtgtataatgtgttaaactactgattctaattgtttgtgtattttagattccaacctat ggaactgatgaatgggagcagtggtggaatgcctttaa tgaggaaaacctgttttgctcagaagaaatgccatctagtgatgatgaggctactgctga ctctcaacattctactcctccaaaaaagaagagaa aggtagaagaccccaaggactttccttcagaattgctaagttttttgagtcatgctgtgt ttagtaatagaactcttgcttgctttgctatttacacca caaaggaaaaagctgcactgctatacaagaaaattatggaaaaatattctgtaaccttta taagtaggcataacagttataatcataacatactg ttttttcttactccacacaggcatagagtgtctgctattaataactatgctcaaaaattg tgtacctttagctttttaatttgtaaaggggttaataag gaatatttgatgtatagtgccttgactagagatcataatcagccataccacatttgtaga ggttttacttgctttaaaaaacctcccacacctccccc tgaacctgaaacataaaatgaatgcaattgttgttgttgggctgcaggaattaattcgag ctcgcccgaca

HIV-1 GAG-CAS9-encoding nucleic acid "KLAP229" (SEQ ID NO. 34)

GCGGCCGCTCTAGAGAGCTTGGCCCATTGCATACGTTGTATCCATATCATAATATGTACA TTTATATTGGCTCAT GTCCAACATTACCGCCATGTTGACATTGATTATTGACTAGTTATTAATAGTAATCAATTA CGGGGTCATTAGTTC ATAGCCCATATATGGAGTTCCGCGTTACATAACTTACGGTAAATGGCCCGCCTGGCTGAC CGCCCAACGACCCC CGCCCATTGACGTCAATAATGACGTATGTTCCCATAGTAACGCCAATAGGGACTTTCCAT TGACGTCAATGGGT GGAGTATTTACGGTAAACTGCCCACTTGGCAGTACATCAAGTGTATCATATGCCAAGTAC GCCCCCTATTGACG TCAATGACGGTAAATGGCCCGCCTGGCATTATGCCCAGTACATGACCTTATGGGACTTTC CTACTTGGCAGTAC ATCTACGTATTAGTCATCGCTATTACCATGGTGATGCGGTTTTGGCAGTACATCAATGGG CGTGGATAGCGGTT TGACTCACGGGGATTTCCAAGTCTCCACCCCATTGACGTCAATGGGAGTTTGTTTTGGCA CCAAAATCAACGGG ACTTTCCAAAATGTCGTAACAACTCCGCCCCATTGACGCAAATGGGCGGTAGGCGTGTAC GGTGGGAGGTCTA TATAAGCAGAGCTCGTTTAGTGAACCGTCAGATCGCCTGGAGACGCCATCCACGCTGTTT TGACCTCCATAGAA GACACCGGGACCGATCCAGCCTCCGGTCGACCGATCCTGAGAACTTCAGGGTGAGTTTGG GGACCCTTGATTG TTCTTTCTTTTTCG CTATTGTA AA ATTC ATGTTAT ATG G AG G G G G C A AAGTTTTC AG G GTGTTGTTTAG A ATG G G AAGATGTCCCTTGTATCACCATGGACCCTCATGATAATTTTGTTTCTTTCACTTTCTACT CTGTTGACAACCATTG TCTCCTCTTATTTTCTTTTCATTTTCTTGTAACTTTTTCGTTAAACTTTAGCTTGCATTT GTAACGAATTTTTAAA^ C ACTTTTGTTT ATTTGTC AG ATTGT AAGTACTTTCTCTA ATC ACTTTTTTTTC A AG G C A ATC AG G GTATATTATATT GTACTTCAGCACAGTTTTAGAGAACAATTGTTATAATTAAATGATAAGGTAGAATATTTC TGCATATAAATTCTG GCTGGCGTGGAAATATTCTTATTGGTAGAAACAACTACATCCTGGTCATCATCCTGCCTT TCTCTTTATGGTTAC AATGATATACACTGTTTGAGATGAGGATAAAATACTCTGAGTCCAAACCGGGCCCCTCTG CTAACCATGTTCAT G CCTTCTTCTTTTTCCTAC AG CTCCTGGGCAACGTG CTG GTT ATTGTG CTGTCTC ATC ATTTTG G C AA AG AATTCc tcgagatgggtgcgagagcgtcggtattaagcgggggagaattagataaatgggaaaaaa ttcggttaaggccagggggaaagaaacaatat aaactaaaacatatagtatgggcaagcagggagctagaacgattcgcagttaatcctggc cttttagagacatcagaaggctgtagacaaatac tgggacagctacaaccatcccttcagacaggatcagaagaacttagatcattatataata caatagcagtcctctattgtgtgcatcaaaggata gatgtaaaagacaccaaggaagccttagataagatagaggaagagcaaaacaaaagtaag aaaaaggcacagcaagcagcagctgacaca ggaaacaacagccagGTCAGCCAAAATTACCCTATAGTGCAGAACctccaggggcaaatg gtacatcaggccatatcacctagaa ctttaaatgcatgggtaaaagtagtagaagagaaggctttcagcccagaagtaataccca tgttttcagcattatcagaaggagccaccccacaa gatttaaataccatgctaaacacagtggggggacatcaagcagccatgcaaatgttaaaa gagaccatcaatgaggaagctgcagaatgggat agattgcatccagtgcatgcagggcctattgcaccaggccagatgagagaaccaagggga agtgacatagcaggaactactagtacccttcag gaacaaataggatggatgacacataatccacctatcccagtaggagaaatctataaaaga tggataatcctgggattaaataaaatagtaaga atgtatagccctaccagcattctggacataagacaaggaccaaaggaaccctttagagac tatgtagaccgattctataaaactctaagagccg agcaagcttcacaagaggtaaaaaattggatgacagaaaccttgttggtccaaaatgcga acccagattgtaagactattttaaaagcattggg accaggagcgacactagaagaaatgatgacagcatgtcagggagtggggggacccggcca tAAAGCAAGAGTTTTGGCTGAAGCA ATGAGCcaagtaacaaatccagctaccataatgatacagaaaggcaattttaggaaccaa agaaagactgttaagtgtttcaattgtggcaa agaagggcacatagccaaaaattgcagggcccctaggaaaaagggctgttggaaatgtgg aaaggaaggacaccaaatgaaagattgtactG AGAGACAGGCTAATTTTTTAGGGAAGATCtggccttcccacaagggaAGGCCAGGGAATT TTCTTCAGAGCAGACCA gagccaacagccccaccagaagagagcttcaggtttggggaagagacaacaactccctct cagaagcaggagccgatagacaaggaactgta tcctttagcttccctcagatcactctttggcagcgacccctcgtcacaaCCGGGGACCAC GACCCCAGGcAAAGCAAGAGTTTTG GCTGAAGCAATGAGCaccggtGATTACAAAGACGATGACGATAAGATGGCCCCAAAGAAG AAGCGGAAGGTCG GTATCCACGGAGTCCCAGCAGCCGACAAGAAGTACAGCATCGGCCTGGACATCGGCACCA ACTCTGTGGGCT GGGCCGTGATCACCGACGAGTACAAGGTGCCCAGCAAGAAATTCAAGGTGCTGGGCAACA CCGACCGGCACA GCATCAAGAAGAACCTGATCGGAGCCCTGCTGTTCGACAGCGGCGAAACAGCCGAGGCCA CCCGGCTGAAGA GAACCGCCAGAAGAAGATACACCAGACGGAAGAACCGGATCTGCTATCTGCAAGAGATCT TCAGCAACGAGA TGGCCAAGGTGGACGACAGCTTCTTCCACAGACTGGAAGAGTCCTTCCTGGTGGAAGAGG ATAAGAAGCACG AGCGGCACCCCATCTTCGGCAACATCGTGGACGAGGTGGCCTACCACGAGAAGTACCCCA CCATCTACCACCT GAGAAAGAAACTGGTGGACAGCACCGACAAGGCCGACCTGCGGCTGATCTATCTGGCCCT GGCCCACATGAT CAAGTTCCGGGGCCACTTCCTGATCGAGGGCGACCTGAACCCCGACAACAGCGACGTGGA CAAGCTGTTCATC CAGCTGGTGCAGACCTACAACCAGCTGTTCGAGGAAAACCCCATCAACGCCAGCGGCGTG GACGCCAAGGCC ATCCTGTCTGCCAGACTGAGCAAGAGCAGACGGCTGGAAAATCTGATCGCCCAGCTGCCC GGCGAGAAGAAG AATGGCCTGTTCGGCAACCTGATTGCCCTGAGCCTGGGCCTGACCCCCAACTTCAAGAGC AACTTCGACCTGGC CGAGGATGCCAAACTGCAGCTGAGCAAGGACACCTACGACGACGACCTGGACAACCTGCT GGCCCAGATCGG CGACCAGTACGCCGACCTGTTTCTGGCCGCCAAGAACCTGTCCGACGCCATCCTGCTGAG CGACATCCTGAGA GTGAACACCGAGATCACCAAGGCCCCCCTGAGCGCCTCTATGATCAAGAGATACGACGAG CACCACCAGGACC TGACCCTGCTGAAAGCTCTCGTGCGGCAGCAGCTGCCTGAGAAGTACAAAGAGATTTTCT TCGACCAGAGCAA GAACGGCTACGCCGGCTACATTGACGGCGGAGCCAGCCAGGAAGAGTTCTACAAGTTCAT CAAGCCCATCCTG GAAAAGATGGACGGCACCGAGGAACTGCTCGTGAAGCTGAACAGAGAGGACCTGCTGCGG AAGCAGCGGAC CTTCGACAACGGCAGCATCCCCCACCAGATCCACCTGGGAGAGCTGCACGCCATTCTGCG GCGGCAGGAAGAT TTTTACCCATTCCTGAAGGACAACCGGGAAAAGATCGAGAAGATCCTGACCTTCCGCATC CCCTACTACGTGGG CCCTCTGGCCAGGGGAAACAGCAGATTCGCCTGGATGACCAGAAAGAGCGAGGAAACCAT CACCCCCTGGAA CTTCGAGGAAGTGGTGGACAAGGGCGCTTCCGCCCAGAGCTTCATCGAGCGGATGACCAA CTTCGATAAGAA CCTGCCCAACGAGAAGGTGCTGCCCAAGCACAGCCTGCTGTACGAGTACTTCACCGTGTA TAACGAGCTGACC AAAGTGAAATACGTGACCGAGGGAATGAGAAAGCCCGCCTTCCTGAGCGGCGAGCAGAAA AAGGCCATCGT GGACCTGCTGTTCAAGACCAACCGGAAAGTGACCGTGAAGCAGCTGAAAGAGGACTACTT CAAGAAAATCGA GTGCTTCGACTCCGTGGAAATCTCCGGCGTGGAAGATCGGTTCAACGCCTCCCTGGGCAC ATACCACGATCTG CTGAAAATTATCAAGGACAAGGACTTCCTGGACAATGAGGAAAACGAGGACATTCTGGAA GATATCGTGCTG ACCCTGACACTGTTTGAGGACAGAGAGATGATCGAGGAACGGCTGAAAACCTATGCCCAC CTGTTCGACGACA AAGTGATGAAGCAGCTGAAGCGGCGGAGATACACCGGCTGGGGCAGGCTGAGCCGGAAGC TGATCAACGGC ATCCGGGACAAGCAGTCCGGCAAGACAATCCTGGATTTCCTGAAGTCCGACGGCTTCGCC AACAGAAACTTCA TGCAGCTGATCCACGACGACAGCCTGACCTTTAAAGAGGACATCCAGAAAGCCCAGGTGT CCGGCCAGGGCG ATAGCCTGCACGAGCACATTGCCAATCTGGCCGGCAGCCCCGCCATTAAGAAGGGCATCC TGCAGACAGTGAA GGTGGTGGACGAGCTCGTGAAAGTGATGGGCCGGCACAAGCCCGAGAACATCGTGATCGA AATGGCCAGAG AGAACCAGACCACCCAGAAGGGACAGAAGAACAGCCGCGAGAGAATGAAGCGGATCGAAG AGGGCATCAA AGAGCTGGGCAGCCAGATCCTGAAAGAACACCCCGTGGAAAACACCCAGCTGCAGAACGA GAAGCTGTACCT GTACTACCTGCAGAATGGGCGGGATATGTACGTGGACCAGGAACTGGACATCAACCGGCT GTCCGACTACGA TGTGGACCATATCGTGCCTCAGAGCTTTCTGAAGGACGACTCCATCGACAACAAGGTGCT GACCAGAAGCGAC AAGAACCGGGGCAAGAGCGACAACGTGCCCTCCGAAGAGGTCGTGAAGAAGATGAAGAAC TACTGGCGGCA GCTGCTGAACGCCAAGCTGATTACCCAGAGAAAGTTCGACAATCTGACCAAGGCCGAGAG AGGCGGCCTGAG CGAACTGGATAAGGCCGGCTTCATCAAGAGACAGCTGGTGGAAACCCGGCAGATCACAAA GCACGTGGCACA GATCCTGGACTCCCGGATGAACACTAAGTACGACGAGAATGACAAGCTGATCCGGGAAGT GAAAGTGATCAC CCTGAAGTCCAAGCTGGTGTCCGATTTCCGGAAGGATTTCCAGTTTTACAAAGTGCGCGA GATCAACAACTACC ACCACGCCCACGACGCCTACCTGAACGCCGTCGTGGGAACCGCCCTGATCAAAAAGTACC CTAAGCTGGAAAG CGAGTTCGTGTACGGCGACTACAAGGTGTACGACGTGCGGAAGATGATCGCCAAGAGCGA GCAGGAAATCG GCAAGGCTACCGCCAAGTACTTCTTCTACAGCAACATCATGAACTTTTTCAAGACCGAGA TTACCCTGGCCAAC GGCGAGATCCGGAAGCGGCCTCTGATCGAGACAAACGGCGAAACCGGGGAGATCGTGTGG GATAAGGGCCG GGATTTTGCCACCGTGCGGAAAGTGCTGAGCATGCCCCAAGTGAATATCGTGAAAAAGAC CGAGGTGCAGAC AGGCGGCTTCAGCAAAGAGTCTATCCTGCCCAAGAGGAACAGCGATAAGCTGATCGCCAG AAAGAAGGACTG GGACCCTAAGAAGTACGGCGGCTTCGACAGCCCCACCGTGGCCTATTCTGTGCTGGTGGT GGCCAAAGTGGA AAAGGGCAAGTCCAAGAAACTGAAGAGTGTGAAAGAGCTGCTGGGGATCACCATCATGGA AAGAAGCAGCT TCGAGAAGAATCCCATCGACTTTCTGGAAGCCAAGGGCTACAAAGAAGTGAAAAAGGACC TGATCATCAAGCT GCCTAAGTACTCCCTGTTCGAGCTGGAAAACGGCCGGAAGAGAATGCTGGCCTCTGCCGG CGAACTGCAGAA GGGAAACGAACTGGCCCTGCCCTCCAAATATGTGAACTTCCTGTACCTGGCCAGCCACTA TGAGAAGCTGAAG GGCTCCCCCGAGGATAATGAGCAGAAACAGCTGTTTGTGGAACAGCACAAGCACTACCTG GACGAGATCATC GAGCAGATCAGCGAGTTCTCCAAGAGAGTGATCCTGGCCGACGCTAATCTGGACAAAGTG CTGTCCGCCTACA ACAAGCACCGGGATAAGCCCATCAGAGAGCAGGCCGAGAATATCATCCACCTGTTTACCC TGACCAATCTGGG AGCCCCTGCCGCCTTCAAGTACTTTGACACCACCATCGACCGGAAGAGGTACACCAGCAC CAAAGAGGTGCTG GACGCCACCCTGATCCACCAGAGCATCACCGGCCTGTACGAGACACGGATCGACCTGTCT CAGCTGGGAGGC GACAAGCGTCCTGCTGCTACTAAGAAAGCTGGTCAAGCTAAGAAAAAGAAAGCTAGAGCT TGATATCCTGCAG ACGCGTAGGATCCGTCGAGGAATTCACTCctCAGGTGCAGGCTGCCTATCAGAAGGTGGT GGCTGGTGTGGCC AATGCCCTGGCTCACAAATACCACTGAGATCTTTTTCCCTCTGCCAAAAATTATGGGGAC ATCATGAAGCCCCTT GAGCATCTGACTTCTGGCTAATAAAGGAAATTTATTTTCATTGCAATAGTGTGTTGGAAT TTTTTGTGTCTCTCA CTCGGAAGGACATATGGGAGGGCAAATCATTTAAAACATCAGAATGAGTATTTGGTTTAG AGTTTGGCAACAT ATGCCCATATGCTGGCTGCCATGAACAAAGGTTGGCTATAAAGAGGTCATCAGTATATGA AACAGCCCCCTGC TGTCCATTCCTTATTCCATAGAAAAGCCTTGACTTGAGGTTAGA I I I I I I TTATATTTTGTTTTGTGTTATTTTTTT CTTTAACATCCCTAAAATTTTCCTTACATGTTTTACTAGCCAGATTTTTCCTCCTCTCCT GACTACTCCCAGTCATA GCTGTCCCTCTTCTCTTATGGAGATCCCTCGACGGATCGGCCGCAATTCGTAATCATGTC ATAGCTGTTTCCTGT GTGAAATTGTTATCCGCTCACAATTCCACACAACATACGAGCCGGAAGCATAAAGTGTAA AGCCTGGGGTGCC TAATGAGTGAGCTAACTCACATTAATTGCGTTGCGCTCACTGCCCGCTTTCCAGTCGGGA AACCTGTCGTGCCA GCTGCATTAATGAATCGGCCAACGCGCGGGGAGAGGCGGTTTGCGTATTGGGCGCTCTTC CGCTTCCTCGCTC ACTGACTCGCTGCGCTCGGTCGTTCGGCTGCGGCGAGCGGTATCAGCTCACTCAAAGGCG GTAATACGGTTAT CCACAGAATCAGGGGATAACGCAGGAAAGAACATGTGAGCAAAAGGCCAGCAAAAGGCCA GGAACCGTAAA AAGGCCGCGTTGCTGGCGTTTTTCCATAGGCTCCGCCCCCCTGACGAGCATCACAAAAAT CGACGCTCAAGTCA GAGGTGGCGAAACCCGACAGGACTATAAAGATACCAGGCGTTTCCCCCTGGAAGCTCCCT CGTGCGCTCTCCT GTTCCGACCCTGCCGCTTACCGGATACCTGTCCGCCTTTCTCCCTTCGGGAAGCGTGGCG CTTTCTCATAGCTCA CGCTGTAGGTATCTCAGTTCGGTGTAGGTCGTTCGCTCCAAGCTGGGCTGTGTGCACGAA CCCCCCGTTCAGCC CGACCGCTGCGCCTTATCCGGTAACTATCGTCTTGAGTCCAACCCGGTAAGACACGACTT ATCGCCACTGGCAG CAGCCACTGGTAACAGGATTAGCAGAGCGAGGTATGTAGGCGGTGCTACAGAGTTCTTGA AGTGGTGGCCTA ACTACGGCTACACTAGAAGAACAGTATTTGGTATCTGCGCTCTGCTGAAGCCAGTTACCT TCGGAAAAAGAGT TGGTAGCTCTTGATCCGGCAAACAAACCACCGCTGGTAGCGGTGG I I I I I I I GTTTGCAAGCAGCAGATTACGC GCAGAAAAAAAGGATCTCAAGAAGATCCTTTGATCTTTTCTACGGGGTCTGACGCTCAGT GGAACGAAAACTC ACGTTAAGGGATTTTGGTCATGAGATTATCAAAAAGGATCTTCACCTAGATCCTTTTAAA TTAAAAATGAAGTT TTAAATCAATCTAAAGTATATATGAGTAAACTTGGTCTGACAGTTACCAATGCTTAATCA GTGAGGCACCTATC TCAGCGATCTGTCTATTTCGTTCATCCATAGTTGCCTGACTCCCCGTCGTGTAGATAACT ACGATACGGGAGGG CTTACCATCTGGCCCCAGTGCTGCAATGATACCGCGAGACCCACGCTCACCGGCTCCAGA TTTATCAGCAATAA ACCAGCCAGCCGGAAGGGCCGAGCGCAGAAGTGGTCCTGCAACTTTATCCGCCTCCATCC AGTCTATTAATTG TTGCCGGGAAGCTAGAGTAAGTAGTTCGCCAGTTAATAGTTTGCGCAACGTTGTTGCCAT TGCTACAGGCATC GTGGTGTCACGCTCGTCGTTTGGTATGGCTTCATTCAGCTCCGGTTCCCAACGATCAAGG CGAGTTACATGATC CCCCATGTTGTGCAAAAAAGCGGGTTAGCTCCTTCGGTCCTCCGATCGTTGTCAGAAGTA AGTTGGCCGCAGT GTT ATC ACTC ATG GTTATG G C AG C ACTG C ATA ATTCTCTT ACTGTC ATG CC ATCCGTA AG ATG CTTTTCTGTG AC TGGTGAGTACTCAACCAAGTCATTCTGAGAATAGTGTATGCGGCGACCGAGTTGCTCTTG CCCGGCGTCAATA CGGGATAATACCGCGCCACATAGCAGAACTTTAAAAGTGCTCATCATTGGAAAACGTTCT TCGGGGCGAAAAC TCTCAAGGATCTTACCGCTGTTGAGATCCAGTTCGATGTAACCCACTCGTGCACCCAACT GATCTTCAGCATCTT TTACTTTCACCAGCGTTTCTGGGTGAGCAAAAACAGGAAGGCAAAATGCCGCAAAAAAGG GAATAAGGGCGA CACGGAAATGTTGAATACTCATACTCTTCCTTTTTCAATATTATTGAAGCATTTATCAGG GTTATTGTCTCATGA GCGGATACATATTTGAATGTATTTAGAAAAATAAACAAATAGGGGTTCCGCGCACATTTC CCCGAAAAGTGCC ACCTAAATTGTAAGCGTTAATATTTTGTTAAAATTCGCGTTAAATTTTTGTTAAATCAGC TCATTTTTTAACCAAT AGGCCGAAATCGGCAAAATCCCTTATAAATCAAAAGAATAGACCGAGATAGGGTTGAGTG TTGTTCCAGTTTG GAACAAGAGTCCACTATTAAAGAACGTGGACTCCAACGTCAAAGGGCGAAAAACCGTCTA TCAGGGCGATGG CCCACTACGTGAACCATCACCCTAATCAAGTTTTTTGGGGTCGAGGTGCCGTAAAGCACT AAATCGGAACCCTA AAGGGAGCCCCCGATTTAGAGCTTGACGGGGAAAGCCGGCGAACGTGGCGAGAAAGGAAG GGAAGAAAGC GAAAGGAGCGGGCGCTAGGGCGCTGGCAAGTGTAGCGGTCACGCTGCGCGTAACCACCAC ACCCGCCGCGCT TAATGCGCCGCTACAGGGCGCGTCCCATTCGCCATTCAGGCTGCGCAACTGTTGGGAAGG GCGATCGGTGCG GGCCTCTTCGCTATTACGCCAGCTGGCGAAAGGGGGATGTGCTGCAAGGCGATTAAGTTG GGTAACGCCAGG GTTTTCCCAGTCACGACGTTGTAAAACGACGGCCAGTGAGCGCGCGTAATACGACTCACT ATAGGGCGAATTG GAGCTCCACCGCGGTG

Sequences

SEQ ID NO. Type Description

1 Nucleic acid YFPgRNA If primer

2 Nucleic acid YFPgRNA 1 r primer

3 Nucleic acid YFPgRNA2f primer

4 Nucleic acid YFPgRNA2r primer

5 Nucleic acid hMyd88gRNA 1 f primer

6 Nucleic acid hMyd88gRNAlr primer

7 Nucleic acid hMyd88gRNA2f primer

8 Nucleic acid hMyd88gRNA2r primer

9 Nucleic acid mMyd88 gRNA 1 f primer

10 Nucleic acid mMyd88 gRNA 1 r primer

11 Nucleic acid mMyd88 gRNA2f primer

12 Nucleic acid mMyd88gRNA2r primer

13 Nucleic acid DDX3 gRNA If primer

14 Nucleic acid DDX3gRNAlr primer

15 Nucleic acid YFPf primer

16 Nucleic acid YFPr primer

17 Nucleic acid hMyd88f2 primer

18 Nucleic acid hMyd88rl primer

19 Nucleic acid mMyd88f2 primer

20 Nucleic acid mMyd88r2 primer

21 Nucleic acid Flag-DDX3 primer

22 Amino acid GAG-Cas9 fusion protein

23 Amino acid VSV-G protein SEQ ID NO. Type Description

24 Amino acid GAG PRO POL polyprotein

25 Amino acid BAEV-G protein

26 Nucleic acid encoding Gag-Cas9 fusion protein

27 Nucleic acid encoding BAEV-G

28 Nucleic acid encoding VSV-G protein

29 Amino acid cleavage sequence

30 Nucleic acid encoding the cleavage sequence of SEQ ID NO. 29

31 Amino acid Cas9 protein

32 Nucleic acid encoding Cas9 protein

33 Nucleic acid HIV encapsidation construct (may be termed "p8.91")

34 Nucleic acid encoding HIV-1 GAG-CAS9 polypeptide (may be termed

"KLAP229")

35 Nucleic acid target sequence

36 Nucleic acid primer

37 Nucleic acid primer

38 Nucleic acid target sequence

39 Nucleic acid primer

40 Nucleic acid primer