References
1- Qi LS et al. Repurposing CRISPR as an RNA-cuided platform for
sequencespecific control of gene expression. Cell 2013;152:1173–83.
2- Standage-Beier K, Zhang Q, Wang X. Targeted Large-Scale Deletion of
Bacterial Genomes Using CRISPR-Nickases. ACS Synth. Biol.
2015;4:1217–25.
3- Tanenbaum ME, Gilbert LA, Qi LS, Weissman JS, Vale RD. A
Protein-Tagging System for Signal Amplification in Gene Expression and
Fluorescence Imaging. Cell 2014;159:635–46.
4- Gaudelli NM et al. Programmable base editing of A T to G C in genomic
DNA without DNA cleavage. Nature 2017;551:464–71.
5- Komor AC, Kim YB, Packer MS, Zuris JA, Liu DR. Programmable editing
of a target base in genomic DNA without double-stranded DNA cleavage.
Nature 2016;533:420–4.
6- Anzalone AV et al. Search-and-replace genome editing without
double-strand breaks or donor DNA. Nature 2019;576:149–57.
7- Liu Z et al. Efficient and high-fidelity base editor with expanded
PAM compatibility for cytidine dinucleotide. Sci. China Life Sci. 2021;
64:1355–67.
8- Strecker J et al. RNA-guided DNA insertion with CRISPR-associated
transposases. Science (80-.) 2019;365:48–53.
9- Liu Z et al. Efficient and high-fidelity base editor with expanded
PAM compatibility for cytidine dinucleotide. Sci. China Life Sci.
2021;64:1355–67.
10- Burstein D et al. New CRISPR-Cas systems from uncultivated
microbes.Nature 2017;542:237–41.
11- Mout R, Ray M, Lee YW, Scaletti F & Rotello VM In Vivo Delivery of
CRISPR/Cas9 for Therapeutic Gene Editing: Progress and Challenges.
Bioconjugate Chem 28, 880–884 (2017).
12- Ho BX, Loh SJH, Chan WK & Soh BS In Vivo Genome Editing as a
Therapeutic Approach. Int J Mol Sci 19, 2721 (2018).
13- Naldini L Ex vivo gene transfer and correction for cell-based
therapies. Nat Rev Genet 12, 301–315 (2011).
14- Wang HX et al. CRISPR/Cas9-Based Genome Editing for Disease Modeling
and Therapy: Challenges and Opportunities for Nonviral Delivery.
Chemical reviews 117, 9874–9906 (2017).
15- Glass Z, Lee M, Li YM & Xu QB Engineering the Delivery System for
CRISPR-Based Genome Editing. Trends in biotechnology 36, 173–185
(2018).
16- Xu L, Wang J, Liu Y, Xie L, Su B, Mou D, et al. CRISPR-edited stem
cells in a patient with HIV and acute lymphocytic leukemia. N Engl J Med
2019;381:1240–1247.
17- Frangoul H, Altshuler D, Cappellini MD, Chen YS, Domm J, Eustace BK,
et al. CRISPR-Cas9 gene editing for sickle cell disease and
β-thalassemia. N Engl J Med 2021;384:252–260.
18-Stadtmauer EA, Fraietta JA, Davis MM, Cohen AD, Weber KL, Lancaster
E, et al. CRISPR-engineered T cells in patients with refractory cancer.
Science 2020;367:eaba7365
19-Lu Y, Xue J, Deng T, Zhou X, Yu K, Deng L, et al. Safety and
feasibility of CRISPR-edited T cells in patients with refractory
non-small-cell lung cancer. Nat Med 2020;26:732–740.
20-Jing Z, Zhang N, Ding L, Wang X, Hua Y, Jiang M, et al. Safety and
activity of programmed cell death-1 gene knockout engineered t cells in
patients with previously treated advanced esophageal squamous cell
carcinoma: an open-label, single-arm phase I study; 2018 ASCO Annual
Meeting I; American Society of Clinical Oncology; 2018. pp. 3054.
21- Stadtmauer EA, Fraietta JA, Davis MM, Cohen AD, Weber KL, Lancaster
E, et al. CRISPR-engineered T cells in patients with refractory cancer.
Science 2020;367:eaba7365
22- Lu Y, Xue J, Deng T, Zhou X, Yu K, Deng L, et al. Safety and
feasibility of CRISPR-edited T cells in patients with refractory
non-small-cell lung cancer. Nat Med 2020;26:732–740.
23- Gillmore JD, Gane E, Taubel J, Kao J, Fontana M, Maitland ML, et al.
CRISPR-Cas9 in vivo gene editing for transthyretin amyloidosis. N Engl J
Med 2021;385:493–502
24- Maeder ML, Stefanidakis M, Wilson CJ, Baral R, Barrera LA, Bounoutas
GS, et al. Development of a gene-editing approach to restore vision loss
in Leber congenital amaurosis type 10. Nat Med 2019;25:229–233.
25- Jo DH, Song DW, Cho CS, Kim UG, Lee KJ, Lee K, et al.
CRISPR-Cas9-mediated therapeutic editing of Rpe65 ameliorates the
disease phenotypes in a mouse model of Leber congenital amaurosis. Sci
Adv 2019;5:eaax1210
26- Jang HK, Jo DH, Lee SN, Cho CS, Jeong YK, Jung Y, et al. High-purity
production and precise editing of DNA base editing ribonucleoproteins.
Sci Adv 2021;7:eabg2661
27- Jang H, Jo DH, Cho CS, Shin JH, Seo JH, Yu G, et al. Application of
prime editing to the correction of mutations and phenotypes in adult
mice with liver and eye diseases. Nat Biomed Eng. 2021 Aug
26;[Epub]. Available at:
https://doi.org/10.1038/s41551-021-00788-9.
28- Smalley E. First AAV gene therapy poised for landmark approval. Nat
Biotechnol 2017;35:998–999.
29- Yu W, Mookherjee S, Chaitankar V, Hiriyanna S, Kim JW, Brooks M, et
al. Nrl knockdown by AAV-delivered CRISPR/Cas9 prevents retinal
degeneration in mice. Nat Commun 2017;8:14716
30-Moreno AM, Fu X, Zhu J, Katrekar D, Shih YV, Marlett J, et al. In
situ gene therapy via AAV-CRISPR-Cas9-mediated targeted gene regulation.
Mol Ther 2018;26:1818–1827.
31-Goossens R, van den Boogaard ML, Lemmers RJLF, Balog J, van der Vliet
PJ, Willemsen IM, et al. Intronic SMCHD1 variants in FSHD: testing the
potential for CRISPR-Cas9 genome editing. J Med Genet 2019;56:828–837.
32-Vagni P, Perlini LE, Chenais NAL, Marchetti T, Parrini M,
Contestabile A, et al. Gene editing preserves visual functions in a
mouse model of retinal degeneration. Front Neurosci 2019;13:945
33-Latella MC, Di Salvo MT, Cocchiarella F, Benati D, Grisendi G,
Comitato A, et al. In vivo editing of the human mutant rhodopsin gene by
electroporation of plasmid-based CRISPR/Cas9 in the mouse retina. Mol
Ther Nucleic Acids 2016;5:e389
34-Bakondi B, Lv W, Lu B, Jones MK, Tsai Y, Kim KJ, et al. In vivo
CRISPR/Cas9 gene editing corrects retinal dystrophy in the S334ter-3 rat
model of autosomal dominant retinitis pigmentosa. Mol Ther
2016;24:556–563.
35-Hu S, Du J, Chen N, Jia R, Zhang J, Liu X, et al. In vivo
CRISPR/Cas9-mediated genome editing mitigates photoreceptor degeneration
in a mouse model of X-linked retinitis pigmentosa. Invest Ophthalmol Vis
Sci 2020;61:31
36- Yin H, Xue W, Chen S, Bogorad RL, Benedetti E, Grompe M, et al.
Genome editing with Cas9 in adult mice corrects a disease mutation and
phenotype. Nat Biotechnol 2014;32:551–553.
37- Ibraheim R, Song CQ, Mir A, Amrani N, Xue W, Sontheimer EJ.
All-in-one adeno-associated virus delivery and genome editing by
Neisseria meningitidis Cas9 in vivo. Genome Biol 2018;19:137
38-Agudelo D, Carter S, Velimirovic M, Duringer A, Rivest JF, Levesque
S, et al. Versatile and robust genome editing with Streptococcus
thermophilus CRISPR1-Cas9. Genome Res 2020;30:107–117.
39- Song CQ, Jiang T, Richter M, Rhym LH, Koblan LW, Zafra MP, et al.
Adenine base editing in an adult mouse model of tyrosinaemia. Nat Biomed
Eng 2020;4:125–130.
40-Yang L, Wang L, Huo Y, Chen X, Yin S, Hu Y, et al. Amelioration of an
inherited metabolic liver disease through creation of a de novo start
codon by cytidine base editing. Mol Ther 2020;28:1673–1683.
41- Jang H, Jo DH, Cho CS, Shin JH, Seo JH, Yu G, et al. Application of
prime editing to the correction of mutations and phenotypes in adult
mice with liver and eye diseases. Nat Biomed Eng. 2021 Aug 26;
https://doi.org/10.1038/s41551-021-00788-9.
42- Villiger L, Grisch-Chan HM, Lindsay H, Ringnalda F, Pogliano CB,
Allegri G, et al. Treatment of a metabolic liver disease by in vivo
genome base editing in adult mice. Nat Med 2018;24:1519–1525.
43- Richards DY, Winn SR, Dudley S, Nygaard S, Mighell TL, Grompe M, et
al. AAV-mediated CRISPR/Cas9 gene editing in murine phenylketonuria. Mol
Ther Methods Clin Dev 2020;17:234–245.
44-Yin S, Ma L, Shao T, Zhang M, Guan Y, Wang L, et al. Enhanced genome
editing to ameliorate a genetic metabolic liver disease through
co-delivery of adeno-associated virus receptor. Sci China Life Sci. 2020
Aug 17;
https://doi.org/10.1007/s11427-020-1744-6.
45- Yang Y, Wang L, Bell P, McMenamin D, He Z, White J, et al. A dual
AAV system enables the Cas9-mediated correction of a metabolic liver
disease in newborn mice. Nat Biotechnol 2016;34:334–338.
46- Long C, McAnally JR, Shelton JM, Mireault AA, Bassel-Duby R, Olson
EN. Prevention of muscular dystrophy in mice by CRISPR/Cas9-mediated
editing of germline DNA. Science 2014;345:1184–1188.
47- Nelson CE, Hakim CH, Ousterout DG, Thakore PI, Moreb EA, Castellanos
Rivera RM, et al. In vivo genome editing improves muscle function in a
mouse model of Duchenne muscular dystrophy. Science 2016;351:403–407.
48-Tabebordbar M, Zhu K, Cheng JKW, Chew WL, Widrick JJ, Yan WX, et al.
In vivo gene editing in dystrophic mouse muscle and muscle stem cells.
Science 2016;351:407–411.
49-Bengtsson NE, Hall JK, Odom GL, Phelps MP, Andrus CR, Hawkins RD, et
al. Muscle-specific CRISPR/Cas9 dystrophin gene editing ameliorates
pathophysiology in a mouse model for Duchenne muscular dystrophy. Nat
Commun 2017;8:14454
50- Amoasii L, Hildyard JCW, Li H, Sanchez-Ortiz E, Mireault A,
Caballero D, et al. Gene editing restores dystrophin expression in a
canine model of Duchenne muscular dystrophy. Science 2018;362:86–91.
51- Moretti A, Fonteyne L, Giesert F, Hoppmann P, Meier AB, Bozoglu T,
et al. Somatic gene editing ameliorates skeletal and cardiac muscle
failure in pig and human models of Duchenne muscular dystrophy. Nat Med
2020;26:207–214.
52- Ryu SM, Koo T, Kim K, Lim K, Baek G, Kim ST, et al. Adenine base
editing in mouse embryos and an adult mouse model of Duchenne muscular
dystrophy. Nat Biotechnol 2018;36:536–539.
53- Gaj T, Ojala DS, Ekman FK, Byrne LC, Limsirichai P, Schaffer DV. In
vivo genome editing improves motor function and extends survival in a
mouse model of ALS. Sci Adv 2017;3:eaar3952
54- Arnaoutova I, Zhang L, Chen HD, Mansfield BC, Chou JY. Correction of
metabolic abnormalities in a mouse model of glycogen storage disease
type Ia by CRISPR/Cas9-based gene editing. Mol Ther 2021;29:1602–1610.
55- Koblan LW, Erdos MR, Wilson C, Cabral WA, Levy JM, Xiong ZM, et al.
In vivo base editing rescues Hutchinson-Gilford progeria syndrome in
mice. Nature 2021;589:608–614.
56- Wilbie D, Walther J, Mastrobattista E. Delivery aspects of
CRISPR/Cas for in vivo genome editing. Acc Chem Res. 2019;52:1555–64.
57-Kay MA, Glorioso JC, Naldini L. Viral vectors for gene therapy: the
art of turning infectious agents into vehicles of therapeutics. Nat Med.
2001;7:33–40.
58-Bessis N, GarciaCozar FJ, Boissier MC. Immune responses to gene
therapy vectors: influence on vector function and effector mechanisms.
Gene Ther. 2004;11(Suppl 1):S10–7.
59-Samulski RJ, Zhu X, Xiao X, Brook JD, Housman DE, Epstein N, et al.
Targeted integration of adeno-associated virus (AAV) into human
chromosome 19. EMBO J. 1991;10:3941–50.
60-Yan J, Kang DD, Turnbull G, Dong Y. Delivery of CRISPR-Cas9 system
for screening and editing RNA binding proteins in cancer. Adv Drug Deliv
Rev. 2021;180:114042.
61-Maggio I, Zittersteijn HA, Wang Q, Liu J, Janssen JM, Ojeda IT, et
al. Integrating gene delivery and gene-editing technologies by
adenoviral vector transfer of optimized CRISPR-Cas9 components. Gene
Ther. 2020;27:209–25.
62-Palmer D, Ng P. Improved system for helper-dependent adenoviral
vector production. Mol Ther. 2003;8:846–52.
63-Jager L, Ehrhardt A. Persistence of high-capacity adenoviral vectors
as replication-defective monomeric genomes in vitro and in murine liver.
Hum Gene Ther. 2009;20:883–96.
64-Byrnes AP, Rusby JE, Wood MJ, Charlton HM. Adenovirus gene transfer
causes inflammation in the brain. Neuroscience. 1995;66:1015–24.
65-Yang Y, Greenough K, Wilson JM. Transient immune blockade prevents
formation of neutralizing antibody to recombinant adenovirus and allows
repeated gene transfer to mouse liver. Gene Ther. 1996;3:412–20.
66-Beer SJ, Matthews CB, Stein CS, Ross BD, Hilfinger JM, Davidson BL.
Poly (lactic-glycolic) acid copolymer encapsulation of recombinant
adenovirus reduces immunogenicity in vivo. Gene Ther. 1998;5:740–6.
67-Tuveson DA, Jacks T. Technologically advanced cancer modeling in
mice. Curr Opin Genet Dev. 2002;12:105–10.
68-Maddalo D, Manchado E, Concepcion CP, Bonetti C, Vidigal JA, Han YC,
et al. In vivo engineering of oncogenic chromosomal rearrangements with
the CRISPR/Cas9 system. Nature. 2014;516:423–7.
69-Wang D, Mou H, Li S, Li Y, Hough S, Tran K, et al.
Adenovirus-mediated somatic genome editing of Pten by CRISPR/Cas9 in
mouse liver in spite of Cas9-specific immune responses. Hum Gene Ther.
2015;26:432–42.
70-Huang J, Chen M, Whitley MJ, Kuo HC, Xu ES, Walens A, et al.
Generation and comparison of CRISPR-Cas9 and Cre-mediated genetically
engineered mouse models of sarcoma. Nat Commun. 2017;8:15999.
71-Wang D, Tai PWL, Gao G. Adeno-associated virus vector as a platform
for gene therapy delivery. Nat Rev Drug Discov. 2019;18:358–78.
72-Luo J, Luo Y, Sun J, Zhou Y, Zhang Y, Yang X. Adeno-associated
virus-mediated cancer gene therapy: current status. Cancer Lett.
2015;356:347–56.
73-Nelson CE, Gersbach CA. Engineering delivery vehicles for genome
editing. Annu Rev Chem Biomol Eng. 2016;7:637–62.
74-Dong JY, Fan PD, Frizzell RA. Quantitative analysis of the packaging
capacity of recombinant adeno-associated virus. Hum Gene Ther.
1996;7:2101–12.
75-Senís E, Fatouros C, Große S, Wiedtke E, Niopek D, Mueller AK, et al.
CRISPR/Cas9-mediated genome engineering: an adeno-associated viral (AAV)
vector toolbox. Biotechnol J. 2014;9:1402–12.
76-Yang Y, Wang L, Bell P, McMenamin D, He Z, White J, et al. A dual AAV
system enables the Cas9-mediated correction of a metabolic liver disease
in newborn mice. Nat Biotechnol. 2016;34:334–8.
77-Sun JY, Anand-Jawa V, Chatterjee S, Wong KK. Immune responses to
adeno-associated virus and its recombinant vectors. Gene Ther.
2003;10:964–76.
78-Huttner NA, Girod A, Perabo L, Edbauer D, Kleinschmidt JA, Büning H,
et al. Genetic modifications of the adeno-associated virus type 2 capsid
reduce the affinity and the neutralizing effects of human serum
antibodies. Gene Ther. 2003;10:2139–47.
79-Vakulskas CA, Behlke MA. Evaluation and reduction of CRISPR
off-target cleavage events. Nucleic Acid Ther. 2019;29:167–74.
80-Swiech L, Heidenreich M, Banerjee A, Habib N, Li Y, Trombetta J, et
al. In vivo interrogation of gene function in the mammalian brain using
CRISPR-Cas9. Nat Biotechnol. 2015;33:102–6.
81-Winters IP, Chiou SH, Paulk NK, McFarland CD, Lalgudi PV, Ma RK, et
al. Multiplexed in vivo homology-directed repair and tumor barcoding
enables parallel quantification of Kras variant oncogenicity. Nat
Commun. 2017;8:2053.
82-Chow RD, Guzman CD, Wang G, Schmidt F, Youngblood MW, Ye L, et al.
AAV-mediated direct in vivo CRISPR screen identifies functional
suppressors in glioblastoma. Nat Neurosci. 2017;20:1329–41.
83-Naldini L. Lentiviruses as gene transfer agents for delivery to
non-dividing cells. Curr Opin Biotechnol. 1998;9:457–63.
84-Kantor B, Bailey RM, Wimberly K, Kalburgi SN, Gray SJ. Methods for
gene transfer to the central nervous system. Adv Genet. 2014;87:125–97.
85-Ling S, Yang S, Hu X, Yin D, Dai Y, Qian X, et al. Lentiviral
delivery of co-packaged Cas9 mRNA and a Vegfa-targeting guide RNA
prevents wet age-related macular degeneration in mice. Nat Biomed Eng.
2021;5:144–56.
86-Wanisch K, Yáñez-Muñoz RJ. Integration-deficient lentiviral vectors:
a slow coming of age. Mol Ther. 2009;17:1316–32.
87-Kim W, Lee S, Kim HS, Song M, Cha YH, Kim YH, et al. Targeting mutant
KRAS with CRISPR-Cas9 controls tumor growth. Genome Res.
2018;28:374–82.
88-Chen SH, Hsieh YY, Tzeng HE, Lin CY, Hsu KW, Chiang YS, et al. ABL
genomic editing sufficiently abolishes oncogenesis of human chronic
myeloid leukemia cells in vitro and in vivo. Cancers (Basel).
2020;12(6):1399.
89-High KA. Turning genes into medicines-what have we learned from gene
therapy drug development in the past decade? Nat Commun. 2020;11:5821.
90-Baum C, Kustikova O, Modlich U, Li Z, Fehse B. Mutagenesis and
oncogenesis by chromosomal insertion of gene transfer vectors. Hum Gene
Ther. 2006;17:253–63.
91-Waehler R, Russell SJ, Curiel DT. Engineering targeted viral vectors
for gene therapy. Nat Rev Genet. 2007;8:573–87.
92-Pack DW, Hoffman AS, Pun S, Stayton PS. Design and development of
polymers for gene delivery. Nat Rev Drug Discov. 2005;4:581–93.
93-Pahle J, Walther W. Vectors and strategies for nonviral cancer gene
therapy. Expert Opin Biol Ther. 2016;16:443–61.
94-Witzigmann D, Kulkarni JA, Leung J, Chen S, Cullis PR, van der Meel
R. Lipid nanoparticle technology for therapeutic gene regulation in the
liver. Adv Drug Deliv Rev. 2020;159:344–63.
95-Sercombe L, Veerati T, Moheimani F, Wu SY, Sood AK, Hua S. Advances
and challenges of liposome assisted drug delivery. Front Pharmacol.
2015;6:286.
96-Mehnert W, Mäder K. Solid lipid nanoparticles: production,
characterization and applications. Adv Drug Deliv Rev. 2001;47:165–96.
97-Pardi N, Hogan MJ, Pelc RS, Muramatsu H, Andersen H, DeMaso CR, et
al. Zika virus protection by a single low-dose nucleoside-modified mRNA
vaccination. Nature. 2017;543:248–51.
98-Finn JD, Smith AR, Patel MC, Shaw L, Youniss MR, van Heteren J, et
al. A single administration of CRISPR/Cas9 lipid nanoparticles achieves
robust and persistent in vivo genome editing. Cell Rep.
2018;22:2227–35.
99-Cheng Q, Wei T, Farbiak L, Johnson LT, Dilliard SA, Siegwart DJ.
Selective organ targeting (SORT) nanoparticles for tissue-specific mRNA
delivery and CRISPR-Cas gene editing. Nat Nanotechnol. 2020;15:313–20.
100-Song R, Murphy M, Li C, Ting K, Soo C, Zheng Z. Current development
of biodegradable polymeric materials for biomedical applications. Drug
Des Devel Ther. 2018;12:3117–45.
101-Yan M, Du J, Gu Z, Liang M, Hu Y, Zhang W, et al. A novel
intracellular protein delivery platform based on single-protein
nanocapsules. Nat Nanotechnol. 2010;5:48–53.
102-Chen G, Abdeen AA, Wang Y, Shahi PK, Robertson S, Xie R, et al. A
biodegradable nanocapsule delivers a Cas9 ribonucleoprotein complex for
in vivo genome editing. Nat Nanotechnol. 2019;14:974–80.
103-Wan T, Chen Y, Pan Q, Xu X, Kang Y, Gao X, et al. Genome editing of
mutant KRAS through supramolecular polymer-mediated delivery of Cas9
ribonucleoprotein for colorectal cancer therapy. J Control Release.
2020;322:236–47.
104-Guo P, Yang J, Huang J, Auguste DT, Moses MA. Therapeutic genome
editing of triple-negative breast tumors using a noncationic and
deformable nanolipogel. Proc Natl Acad Sci U S A.
105-Deng H, Tan S, Gao X, Zou C, Xu C, Tu K, et al. Cdk5 knocking out
mediated by CRISPR-Cas9 genome editing for PD-L1 attenuation and
enhanced antitumor immunity. Acta Pharm Sin B. 2020;10:358–73.
106-Gindy ME, Prud’homme RK. Multifunctional nanoparticles for imaging,
delivery and targeting in cancer therapy. Expert Opin Drug Deliv.
2009;6:865–78.
107-Bowman MC, Ballard TE, Ackerson CJ, Feldheim DL, Margolis DM,
Melander C. Inhibition of HIV fusion with multivalent gold
nanoparticles. J Am Chem Soc. 2008;130:6896–7.
108-Chompoosor A, Saha K, Ghosh PS, Macarthy DJ, Miranda OR, Zhu ZJ, et
al. The role of surface functionality on acute cytotoxicity, ROS
generation and DNA damage by cationic gold nanoparticles. Small.
2010;6:2246–9.
109-Ahmad S, Zamry AA, Tan HT, Wong KK, Lim J, Mohamud R. Targeting
dendritic cells through gold nanoparticles: a review on the cellular
uptake and subsequent immunological properties. Mol Immunol.
2017;91:123–33.
110-Lino CA, Harper JC, Carney JP, Timlin JA. Delivering CRISPR: a
review of the challenges and approaches. Drug Deliv. 2018;25:1234–57.
111-Lee K, Conboy M, Park HM, Jiang F, Kim HJ, Dewitt MA, et al.
Nanoparticle delivery of Cas9 ribonucleoprotein and donor DNA in vivo
induces homology-directed DNA repair. Nat Biomed Eng. 2017;1:889–901.
112-Yen HJ, Hsu SH, Tsai CL. Cytotoxicity and immunological response of
gold and silver nanoparticles of different sizes. Small.
2009;5:1553–61.
113-Bastús NG, Sánchez-Tilló E, Pujals S, Farrera C, López C, Giralt E,
et al. Homogeneous conjugation of peptides onto gold nanoparticles
enhances macrophage response. ACS Nano. 2009;3:1335–44.
114-Lee JY, Park W, Yi DK. Immunostimulatory effects of gold nanorod and
silica-coated gold nanorod on RAW 264.7 mouse macrophages. Toxicol Lett.
2012;209:51–7.
115-Mout R, Ray M, Lee YW, Scaletti F, Rotello VM. In Vivo Delivery of
CRISPR/ Cas9 for Therapeutic Gene Editing: Progress and Challenges.
Bioconjug. Chem. 2017;28:880–4.
116- Hansen-Bruhn M et al. Active Intracellular Delivery of a Cas9/sgRNA
Complex Using Ultrasound-Propelled Nanomotors. Angew. Chemie
2018;130:2687–91.
117- Hur J, Chung AJ. Microfluidic and Nanofluidic Intracellular
Delivery. Adv. Sci. 2021;8.
118- Sessions JW et al. CRISPR-Cas9 directed knock-out of a
constitutively expressed gene using lance array nanoinjection.
Springerplus 2016;5:1521.
119– Song X et al. Delivery of CRISPR/Cas systems for cancer gene
therapy and immunotherapy. Adv. Drug Deliv. Rev. 2021;168:158–80.
120-Yin H et al. Genome editing with Cas9 in adult mice corrects a
disease mutation and phenotype. Nat. Biotechnol. 2014;32:551–3.
121- Samulski, R. J. & Muzyczka, N. AAV-Mediated Gene Therapy for
Research and Therapeutic Purposes.
doi:10.1146/annurev-virology-031413-085355.
122-Jiang, S. et al. CRISPR/Cas9-Mediated Genome Editing in Epstein-Barr
Virus- Transformed Lymphoblastoid B-Cell Lines. Curr. Protoc. Mol. Biol.
121, 31.12.1-31.12.23 (2018).
123-Park A et al. Sendai virus, an RNA virus with no risk of genomic
integration, delivers CRISPR/Cas9 for efficient gene editing. Mol. Ther.
- Methods Clin. Dev. 2016;3:16057.
124- Mansouri M, Ehsaei Z, Taylor V, Berger P. Baculovirus-based genome
editing in primary cells. Plasmid 2017;90:5–9
125 - Zuris JA et al. Cationic lipid-mediated delivery of proteins
enables efficient protein-based genome editing in vitro and in vivo.
Nat. Biotechnol. 2015;33:73–80.
126-Hu Z et al. Disruption of HPV16-E7 by CRISPR/Cas System Induces
Apoptosis and Growth Inhibition in HPV16 Positive Human Cervical Cancer
Cells. Biomed Res. Int. 2014;2014:1–9.
127-Lao Y-H et al. HPV Oncogene Manipulation Using Nonvirally Delivered
CRISPR/Cas9 or Natronobacterium gregoryi Argonaute. Adv. Sci.
2018;5:1700540.
128-Zhang, L. et al. Lipid nanoparticle-mediated efficient delivery of
CRISPR/Cas9 for tumor therapy. NPG Asia Mater. 9, e441–e441 (2017).
129-Sun W et al. Self-Assembled DNA Nanoclews for the Efficient Delivery
of CRISPR-Cas9 for Genome Editing. Angew. Chemie Int. Ed.
2015;54:12029–33.
130-Zhou W, Cui H, Ying L, Yu X-F. Enhanced Cytosolic Delivery and
Release of CRISPR/Cas9 by Black Phosphorus Nanosheets for Genome
Editing. Angew. Chemie Int. Ed. 2018;57:10268–72.
131-He Z-Y et al. In Vivo Ovarian Cancer Gene Therapy Using CRISPR-Cas9.
Hum. Gene Ther. 2018;29:223–33
132-Kim SM et al. Simple in Vivo Gene Editing via Direct Self-Assembly
of Cas9 Ribonucleoprotein Complexes for Cancer Treatment. ACS Nano
2018;12:7750–60.
133-Wang H-X et al. Nonviral gene editing via CRISPR/Cas9 delivery by
membrane-disruptive and endosomolytic helical polypeptide. Proc. Natl.
Acad. Sci. 2018;115:4903–8.
134-Li M et al. Knockdown of hypoxia-inducible factor-1 alpha by tumor
targeted delivery of CRISPR/Cas9 system suppressed the metastasis of
pancreatic cancer. J. Control. Release 2019;304:204–15.
135-Pan Y et al. Near-infrared upconversion–activated CRISPR-Cas9
system: A remote-controlled gene editing platform. Sci. Adv. 2019;5.
136-Yin H et al. Therapeutic genome editing by combined viral and
non-viral delivery of CRISPR system components in vivo. Nat. Biotechnol.
2016;34:328–33.
137-Lyu P, Wang L, Lu B. Virus-like particle mediated crispr/cas9
delivery for efficient and safe genome editing. Life 2020;10:1–16.
138-Kleinstiver BP et al. High-fidelity CRISPR–Cas9 nucleases with no
detectable genome-wide off-target effects. Nature 2016;529:490–5.
139- Slaymaker IM et al. Rationally engineered Cas9 nucleases with
improved specificity. Science 2016;351:84–8.
140- Charlesworth CT et al. Identification of preexisting adaptive
immunity to Cas9 proteins in humans. Nat. Med. 2019;25:249–54.
141- Li C, Brant E, Budak H, Zhang B. CRISPR/Cas: a Nobel Prize
award-winning precise genome editing technology for gene therapy and
crop improvement. J. Zhejiang Univ. Sci. B 2021;22:253–84.
142- Pickar-Oliver A, Gersbach CA. The next generation of CRISPR–Cas
technologies and applications. Nat. Rev. Mol. Cell Biol.
2019;20:490–507.