You are on page 1of 9

Review

Genome-Editing Technologies: Concept, Pros, and


Cons of Various Genome-Editing Techniques and
Bioethical Concerns for Clinical Application
Sikandar Hayat Khan1
1Department of Pathology, PNS HAFEEZ Hospital, Pathology E-8, Islamabad, Islamabad 44400, Pakistan

The traditional healthcare system is at the doorstep for entering mon to these methods are three basic steps, including mechanisms
into the arena of molecular medicine. The enormous knowl- for genetic tool entry into the cell and later nucleus; altering gene tran-
edge and ongoing research have now been able to demonstrate scription and onward processing function; and, finally, the end-
methodologies that can alter DNA coding. The techniques used output in the shape of a suppressed, overexpressed, or simply an
to edit or change the genome evolved from the earlier attempts altered protein product.6,7 From a holistic point of view, the tech-
like nuclease technologies, homing endonucleases, and certain niques involve an apparently simplistic concept involving multiple re-
chemical methods. Molecular techniques like meganuclease, ceptor-ligand interactions; varying cell entry modes like lipofection,
transcription activator-like effector nucleases (TALENs), and sonification, and transfection; and further downstream pathway ef-
zinc-finger nucleases (ZFNs) initially emerged as genome-edit- fects. Furthermore, these technologies are variable in terms of their
ing technologies. These initial technologies suffer from lower specificity and sensitivity, off-target effects, finances, and technique
specificity due to their off-targets side effects. Moreover, from expertise. The body’s immune response to accept the foreign genetic
biotechnology’s perspective, the main obstacle was to develop elements within the cells can lead to the rejection of foreign tissues.
simple but effective delivery methods for host cell entry. Later,
small RNAs, including microRNA (miRNA) and small inter- Moreover, molecular knowledge, in terms of methodology differences,
fering RNA (siRNA), have been widely adopted in the research defining targetable diseases, innovative nanotechnology tools for gene
laboratories to replace lab animals and cell lines. The latest dis- editing, and ethical aspects, also needs to be understood. The plat-
covery of CRISPR/Cas9 technology seems more encouraging by forms for these technologies are improving every day, with a plethora
providing better efficiency, feasibility, and multi-role clinical of new data appearing due to technology miniaturization and automa-
application. This later biotechnology seem to take genome- tion and newer discoveries to improve the yield and specificity of
engineering techniques to the next level of molecular engineer- an edited product. Alongside the developmental improvement in
ing. This review generally discusses the various gene-editing genome-wide engineering the regulatory work-up, standardization
technologies in terms of the mechanisms of action, advantages, protocols need to be devised to reduce inter and intra-method impre-
and side effects. cision, defining the indications and contraindications of every tech-
nique to help improve the concept of personalized medicine.
Over the last half century after post-DNA helical structure discovery,
This review briefly explains the available technologies, provides com-
the world has seen a continuous staircase outburst of various molec-
parison and contrast between different genome-editing methods,
ular technologies, which are now heading forward toward transla-
and identifies some newer versions of genome editing with possible
tion into clinical and laboratory practice.1 Given the availability of
bioethical concerns.
sequencing platforms, acquired wisdom about the micro-mechanics
at work within the genetic apparatus, and the introduction of user-
friendly nanotechnologies, it was possible for next-generation scien- Review Methodology
tists to manipulate the genetic codes at various levels.2 Over the last PubMed searches with the keywords genome-editing techniques or
two decades we saw a plethora of molecular techniques, which al- gene-editing techniques in the last 10 and 5 years yielded a total of
lowed us to edit genes or their alter pathways, allowing humans for 4,466 and 4,054 references, except some historical and related refer-
the first time to micro-edit the DNA codes and further to alter the ences. Specific searches for articles dealing with specific genome-edit-
mRNA fate through post-transcriptional modifications.3 ing methods included conventional genome-editing systems (n = 100),

Principally, genome-wide editing techniques can be interpreted as


https://doi.org/10.1016/j.omtn.2019.02.027.
methods where DNA sequences are changed by deletions, mRNA
Correspondence: Sikandar Hayat Khan, Department of Pathology, PNS HAFEEZ
processing, and post-transcriptional modifications to result in altered Hospital, Pathology E-8, Islamabad, Islamabad 44400, Pakistan.
gene expression, leading to functional behavior of proteins.4,5 Com- E-mail: sik_cpsp@yahoo.com

326 Molecular Therapy: Nucleic Acids Vol. 16 June 2019 ª 2019 The Author(s).
This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/).
www.moleculartherapy.org

Review

Figure 1. A Consolidated Overview of Genome-Editing Techniques

chemical methods (n = 252), meganucleases (n = 83), zinc-finger in vogue or lab use today, the technique is based on physiological
nucleases (ZFNs) (n = 890), transcription activator-like effector nucle- processes involving a double-stranded repair system. However,
ases (TALENs) (n = 1,136), homing endonucleases (n = 265), and some recent data have shown RAD52 protein to be important
CRISPR (n = 11,421). The search was therefore limited to reviews in mediating homologous recombination, and this protein there-
showing conceptual information of common techniques and compar- fore has been considered as a therapy target in certain cancers like
ative information about ZFNs, TALENSs, and CRISPR technologies. BRCA 1 and 2 repair pathways.6,7 However, the technique as of
Finally, the literature was searched for learning newer and advanced now could not gain widespread introduction due to the emer-
gene-editing methods and bioethical concerns associated with genome gence of newer techniques.
biotechnologies. (2) Chemical modalities of genome editing. Komiyama8 utilized
non-restriction enzyme methodology termed artificial restriction
Genome-Editing Techniques DNA cutter (ARCUT). This method uses pseudo-complemen-
The recent expansion and advancements in the field of biotechnology tary peptide nucleic acid (pcPNA), whose job is to specify the
provided us with information and insight into the biochemical and cleavage site within the chromosome or the telomeric region.
molecular mechanisms to edit DNA and, thus, modify downstream Once pcPNA specifies the site, excision here is carried out by
pathways. To date, multiple biotechnologies have shown promise cerium (CE) and EDTA (chemical mixture), which performs
for clinical use, but the field of genome-editing technologies is rapidly the splicing function.8 Furthermore, the technology uses a
evolving and improving. The new techniques seem promising, but the DNA ligase that can later attach any desirable DNA within the
earlier ones have also been updated and improved. For simplicity and spliced site. The advantage of this particular technique is that it
consolidation, an overview of genome-editing techniques is presented can be used in high salt concentrations. Upon initial introduc-
in Figure 1. tion, the technique looked quite appealing to the clinical market;
however, later issues like increased turnaround time and specif-
Representative genome-editing techniques are discussed below. ically the manufacturing of site-specific pcPNA became huge
hurdles (Figure 2).8,9
(1) Conventional genome-editing technique. In the true sense, the (3) Homing endonuclease systems. Homing endocucleases (HEs)
technique may not relate with evolving genome-editing tech- with this word “homing” practically is interpreted as lateral trans-
niques. As highlighted in Figure 1, it includes homologous mission of a genome DNA sequence. The general concept involves
recombination related with gene intervention. While not much a DNA segment where a site is removed by the endocnulceases,

Molecular Therapy: Nucleic Acids Vol. 16 June 2019 327


www.moleculartherapy.org

Review

Figure 2. Excision of Selective Site of dsDNA by Utilizing Artificial Restriction DNA Cutter

which thus results in the formation of 2 segments of DNA frag- off-target effects like reducing site specificity, less DNA integra-
ment.10 So what are these HEs? They are nucleases that occur tion, and possible host genome mutations.14
naturally, with a size almost equivalent to 14 bp, and they are (4) Protein-based nuclease systems. These systems incorporate
capable of splicing slightly larger DNA sequences.11 Recently, nuclease proteins for DNA sequence editing. The common tech-
the introduction of recombinant adeno-associated viruses niques are described below.
(rAAVs) have allowed them as efficient vehicles for transporting Meganucleases. Also termed molecular DNA scissors, these are
genetic tools of genome engineering into the cell, as depicted in large base pair structures that are sometimes found in the genome.
Figure 3.12 Issues pertaining to this technology include engineer- Their potential to excise large pieces of DNA sequences was
ing difficulties in the preparation of these nucleases as well as recently recognized as a genetic tool to modify DNA. This genetic
developing vectors for their entry into cells.13 Another issue potential has been manipulated in labs by modifying the recogni-
with rAAV, though improving with better biotechnology, was tion sites to create nicks, as required for DNA sequence change.

Figure 3. Schematic Showing rAAV Entry,


Movement within Cytoplasm, Attachment with
DNA, and Integration with DNA Segment for
Possible Genome Modification
The steps include the following: (1) entry of rAAV into cell,
(2) uptake by exosome and transport within cytoplasm, (3)
release of rAAV for entry into nucleus, (4) rAAV delivery of
homing endocnulease (HE) and desirable DNA segment,
(5) HE cut of the non-desirable DNA code, and (6) rAAV-
delivered desirable DNA code replacement of the DNA.

328 Molecular Therapy: Nucleic Acids Vol. 16 June 2019


www.moleculartherapy.org

Review

Figure 4. Schematic Showing Step by Step Zinc-Finger Nuclease-Induced Genome Editing


The mechanisms include the following: (1) ZFNs containing FokI endonucleases and protein-binding domains are introduced into the cell, (2) FokI and protein-binding
domains are released to enter the nucleus, (3) protein-binding domains attach with DNA fragment to be removed, (3) FokI cuts out the identified DNA segment by creating
double-stranded DNA break, and (4) the desirable DNA segment is inserted and integrated into the DNA sequence.

These meganucleases are sometimes joined by proteins to create more site specific with fewer off-target effects.20 However, the
large variants like DmoCre and E-Drel, which can further provide techniques share many similarities (Figure 5).
nucleotide site-specific cleavage.15 The technique revolves around (5) RNA DNA systems. These systems primarily include the
two basic steps: first is the recognition of a cleavage site, and then different types of CRISPR methods. The concept of CRISPR is
endonucleases splice out the region.16 The positive aspect related
primitive and has been derived from an ancient immunity sys-
to meganucleases is less toxicity, as they are naturally occurring tem, adopted in nature by some prokaryotic cells like Archea
and provide very specific site cleavage. However, there are newer and probably some bacteria.21 CRISPR in itself has two compo-
techniques now in the clinical arena that have not allowed them to nents, including SPR termed sometimes as spacers, which are
flourish more. hallmarked by varying and differing nucleotide sequences, and
ZFNs. ZFNs are purely artificial structures generated by a combi- probably each one of them represents a past exposure to foreign
natorial approach where restriction endonucleases are joined with antigen. The CRI may represent the genetic memory for a bacte-
zinc-finger-binding domain protein. Figure 4 explains their mech- rium and can be re-activated once encountered with a similar
anism of action in detail, where a binding protein domain iden- foreign antigen. CRI has similar nucleotides (repeats) represent-
tifies after reaching the desirable splice site, which is then cut at ing like separators between different CRIs.22 Figure 6 attempts to
a specific codon by special restriction endonucleases called FokI. provide a basic overview of the CRISPR/Cas9 concept.
The biotechnology is restricted in terms of attachment with 3 co- Cas especially Cas9 as depicted in Figure 6 has a nuclease function.
dons on either side of the DNA chain. The technique in recent Whenever CRISPR RNA (crRNA; also termed guide RNA
years has gained widespread popularity due to its simplicity and [gRNA]) guides the Cas9 protein regarding a possible antigenic
specificity, and it is being employed in clinical usage for certain threat, like a bacteriophage, it with the help of gRNA creates dou-
diseases.17,18 ble-stranded DNA (dsDNA) nicks at the guided selected sites,
TALENs. TALENS almost resemble ZFNs in terms of causing a site-specific cleavage and, thus, destruction of the anti-
manufacturing and mode of action. They are made by a similar gen.23 Moreover, the memory from the antigen is stored as spacer
principle where a restriction nuclease is bound to a DNA-binding within CRISPR.24
protein domain called TAL effector.19 The difference between This physiological role of Cas9/CRISPR as explained above had
TALENs and ZFNs is that the former can target 3 nt in one go recently been extensively utilized for multiple clinical condi-
and the latter can only address 1 nt, thus making TALENs slightly tions.25–27 At the time of writing this review, the news broke about

Molecular Therapy: Nucleic Acids Vol. 16 June 2019 329


www.moleculartherapy.org

Review

Figure 5. Diagram Showing Mechanisms of Transcription Activator-like Effector Nucleases


The steps of gene editing include the following: (1) TALENs containing FokI endonucleases and TALE domains are introduced into the cell, (2) FokI and TALE domains are
released to enter the nucleus, (3) TALE recognizes the non-desirable DNA segments and attaches with them, (4) FokI cleaves the non-desirable DNA segments, and (5) after
the non-desirable DNA segments are cleaved, the desirable segment of DNA is incorporated into the DNA.

Lulu and Nana being claimed to be the first genetically modified Type 3 CRISPR/Cas system. Like the type 1 system, this category
babies, where the human genome was edited to create resistance uses Cas6 for processing crRNA 30 end trimming. The uniqueness
against HIV infection.28 Specific crRNA/gRNA has been engi- of this technique is its targeting of RNA, which is done by a specific
neered, which can be introduced into cell nuclei and later Cas9 complex called type III Csm/Cmr complex.30
where the non-desirable dsDNA is associated with the Cas9 after
Apart from the aforementioned conventional style classification of
guidance provided by the specific crRNA/gRNA. This comple- CRISPR/Cas classification, the data review provided multiple
mentary binding between gRNA and the non-desirable segment other biotechnologies now being utilized. Some examples include
allows Cas9 to destroy the DNA fragment. In clinical and research photo-activating CRISPR system,31 Intein-inducible split Cas9,32
practice, the created nick can be specifically filled by inserting the and modifications like hybrid crRNA-tracrRNA.33
sequence of choice to change the non-desirable sequence of nucle-
otides.29 (6) Gene-silencing techniques. These methods may not fall truly under
genome editing, but they still are capable of modifying the DNA
Over the last few years, CRISPR/Cas9 technology has gained wide- sequence. These technologies include RNAi, CRISPR interference
spread popularity on account of its simplicity and specificity, with (CRISPRi), and morpholino oligonucleotide techniques.34–36
different versions of the original now under research. Multiple ex-
perimentations and biotechnologies have been re-defining the
Comparative Analysis
CRISPR/Cas technologies into 3 distinct types of CRISPR-Cas
The above provides a gist of the various commonly used genome-edit-
types, based on crRNA processing and further action, including
ing techniques. Though there is enormous development, innovation,
the following:
and design of newer ways to edit the genome, we focus our further dis-
Type 1 CRISPR/Cas system. This version utilized Cas5 or Cas6 for cussion on the comparison of common techniques, including ZFN,
pre-processing of crRNA; further cleavage function needs Cas3, TALEN, and CRISPR methods. Tables 1, 2, and 3 provide a compara-
Cascade, and crRNA for interference. tive assessment among these methods.
Type 2 CRISPR/Cas system. Though Cas9 typically functions un-
der the guidance of crRNA to target DNA, RNase III, trans acti- Advancements in Genome Engineering
vating RNA (tracrRNA), and a yet-to-be-identified protein factor The biotechnology is booming with a lot of newer modalities to edit
are involved in trimming at the 50 end. the genome. Oligodeoxyribonucleotide (ODN) can be utilized with

330 Molecular Therapy: Nucleic Acids Vol. 16 June 2019


www.moleculartherapy.org

Review

Figure 6. Schematic Demonstrating the Concept of CRISPR/Cas9 Interactions Leading to the Destruction of Viral Genome at the Selected Splice Site by the
crRNA/gRNA

double-stranded transcription factor decoy (TFD) to act as a thera- template, and, thus, it is termed footprint-free genome editing.38 The
peutic target for multiple diseases, which can affect the transcription technique seems to have minimal off-target effects and is considered
factor and thus bring in the requisite change in transcription and to be a safer version.
further downstream protein actions.37 Papaioannou et al.38 have uti-
lized single-stranded ODNs to precisely cut genomes for repairing Other novel modalities of genome editing are also appearing in the
very small point mutations, giving a footprint-free genome-editing literature, with slight modifications of existing techniques. Martínez-
modality. This concept involves a drug (doxycycline)-induced Cs9 Gálvez et al.39 used single-stranded DNA (ssDNA) and argonautes
transgene, which is carried into the cell by a specific transposon, in gene editing and helped improved gene editing. Some researchers
providing us with very specific and efficient Cas9-mediated editing have utilized certain enzymes like integrases and in the future may
of the genome. This technique does not need the conventional donor obviate the need for nucleases.40

Table 1. Biotechnology Differences among Prototype Genome-Editing Techniques

Serial No. Parameter ZFN TALEN CRISPER/Cas Reference


slightly complex (identical repeats are
moderate (ZFNs need customized simpler (available versions for 48
1 design simplicity multiple, which creates technical issues
protein for every DNA sequence) crRNA can be easily designed)
of engineering and delivery into cells)
24,49
2 engineering feasibility low higher highest
high-yield multiplexing available
48,50
3 multiplex genome editing few models few models (no need for obtaining embryonic
stem cells)
progress demonstrated (CRISPR
not much progress (need not much progress (need individual 51
4 large-scale library preparation only requires plasmid containing
individual gene tailoring) gene tailoring)
small oligonucleotides)
24
5 specificity low higher highest
a b 24,48,52
6 efficiency normal normal high
53
7 cost low high low
a
Some new versions are more efficient24,48 but CRISPR science is evolving more.
b
Cpf1 protein addition will probably improve cell delivery methods.51,52

Molecular Therapy: Nucleic Acids Vol. 16 June 2019 331


www.moleculartherapy.org

Review

Table 2. Side Effect Profiles for Genome-Editing Methods Table 3. Clinical and Research Applications across Important Genome-
Editing Techniques
Serial No. Parameter ZFN TALEN CRISPER/Cas Reference
Serial No. Parameter ZFN TALEN CRISPER/Cas Reference
off-target effect
1 – – – 54
60
incidence 1 diagnostic utility + + +++
61
homologous 2 clinical trial use ++ + +++
a recombination + + + –
utility as epigenetic 62
rate frequency 3 ++ +++ ++++
marker
non-homologous
++ (only with 55,56 making gene-knockout 63
b end joining (NHEJ) + + 4 no no yes (CRISPRi)
earlier versions) models for research
mutation rates
capacity for modification 64
immune reaction 57,58 5 no no probable
c less less more of mitochondrial DNA
susceptibility
genetic editing in 65
RNA-guided 6 no no yes
human babies
endonuclease
66
d (RGEN)-induced   ++ 59 7 RNA editing no no yes
off-target
mutatagenesis
2 cytotoxicity chances ++ + + – Conclusions
This review discussed multiples aspects of genome-editing technolo-
gies, including a classification; some basic explanatory concepts on
The most interesting part of the genome-editing technique, which
mechanisms; and comparison between methods, newer advancements,
may be the game changer in genome editing, is the whole genome
and bioethical concerns. It seems that CRISPR/Cas technologies are
engineering by synthesis that in fact would re-create the genome
probably superseding ZFNs and TALENS. However, the CRIPSR/
from scratch as per the given designed DNA code. This probably
Cas methods are also being improvised, and newer additions have
will become the synthetic genomics of the future.41 Though research
further enhanced its functional capabilities with reduced off-target
work in this domain stands preliminary, over time it is anticipated
effects. Furthermore, the process of engineering better gene modifica-
that this technology may overtake the concept of genome editing.
tion technologies is evolving and can one day replace even CRISPR/
Cas, possibly shifting to synthetic genomics. Among all these revolu-
Bioethical Issues and Genome-Editing Techniques
tionary developments, bioethical concerns need serious attention.
Genome-editing tools are powerful in terms of their potential to not
only bring biotechnological revolution in the field of crop develop-
ment and human pathology but also, in the wrong hands, lead to REFERENCES
1. Friedrich, M. (1871). Ueber die chemische Zusammensetzung der Eiterzellen [On the
abuse and misuse in multiple ways, including manipulation of germ- chemical composition of pus cells]. Medicinisch-chemische Untersuchungen 4,
line genetics. Genuine bioethical concerns have been raised by many 441–460.
experts.42 While time will be the actual judge of these technologies as 2. Watson, J.D., and Crick, F.H.C. (1953). Molecular structure of nucleic acids; a struc-
boon or bane, still the methods can impact the human race probably ture for deoxyribose nucleic acid. Nature 171, 737–738.
in the most nuclear ways, and our incoming human race may be 3. Sanger, F., and Coulson, A.R. (1975). A rapid method for determining sequences in
victimized in ways we do not yet understand.43 Principal concerns DNA by primed synthesis with DNA polymerase. J. Mol. Biol. 94, 441–448.

apart from illegal germline mutation include the morality, the eu- 4. Mullis, K.B. (2016). https://www.karymullis.com/.
genics helping the fittest to survive, ongoing clinical debates about 5. Pavletich, N.P., and Pabo, C.O. (1991). Zinc finger-DNA recognition: crystal struc-
informed consent, religious debate, the possible rise of clones, ture of a Zif268-DNA complex at 2.1 A. Science 252, 809–817.

designer babies, and possibly superhumans.44–46 Moreover, the cur- 6. Claussin, C., and Chang, M. (2016). Multiple Rad52-Mediated Homology-Directed
Repair Mechanisms Are Required to Prevent Telomere Attrition-Induced
rent literature also rules in the possibility of genome editing as a future Senescence in Saccharomyces cerevisiae. PLoS Genet. 12, e1006176.
weapon of war.47
7. Arai, N., and Kagawa, W. (2014). [Molecular mechanisms of homologous recombi-
nation promoted by budding yeast Rad52]. Seikagaku 86, 693–697.
While the quest for a healthy baby and right of best possible treatment 8. Komiyama, M. (2014). Chemical modifications of artificial restriction DNA cutter
choice have been acknowledged in many societies, the approaching (ARCUT) to promote its in vivo and in vitro applications. Artif. DNA PNA XNA
biotechnological revolution seems imminent and undeniable. The 5, e1112457.
pressing need, therefore demands a harmonious and regulated 9. Ito, K., and Komiyama, M. (2014). Site-selective scission of human genome using
translation of needed aspects of genome-editing-related technologies PNA-based artificial restriction DNA cutter. Methods Mol. Biol. 1050, 111–120.

for molecular medicine and other non-clinical crop and food indus- 10. Schultz, B.R., and Chamberlain, J.S. (2008). Recombinant adeno-associated virus
transduction and integration. Mol. Ther. 16, 1189–1199.
tries. This will need consensus in public opinion, debates among
11. Smith, J., Grizot, S., Arnould, S., Duclert, A., Epinat, J.C., Chames, P., Prieto, J.,
experts, involvement of biotechnologists, opinions of bioethical ex-
Redondo, P., Blanco, F.J., Bravo, J., et al. (2006). A combinatorial approach to create
perts, regulatory frameworks within legislatures, and final guidelines artificial homing endonucleases cleaving chosen sequences. Nucleic Acids Res. 34,
and oversight for the finally allowed limited application. e149.

332 Molecular Therapy: Nucleic Acids Vol. 16 June 2019


www.moleculartherapy.org

Review

12. Yoon, Y., Wang, D., Tai, P.W.L., Riley, J., Gao, G., and Rivera-Pérez, J.A. (2018). 34. Fischer, S.E. (2015). RNA Interference and MicroRNA-Mediated Silencing. Curr.
Streamlined ex vivo and in vivo genome editing in mouse embryos using recombinant Protoc. Mol. Biol. 112, 26.1.1–5.
adeno-associated viruses. Nat. Commun. 9, 412.
35. Yoshida, M., Yokota, E., Sakuma, T., Yamatsuji, T., Takigawa, N., Ushijima, T.,
13. Rey-Rico, A., and Cucchiarini, M. (2016). Controlled release strategies for rAAV- Yamamoto, T., Fukazawa, T., and Naomoto, Y. (2018). Development of an integrated
mediated gene delivery. Acta Biomater. 29, 1–10. CRISPRi targeting DNp63 for treatment of squamous cell carcinoma. Oncotarget 9,
29220–29232.
14. Jin, L., Lange, W., Kempmann, A., Maybeck, V., Günther, A., Gruteser, N., Baumann,
A., and Offenhäusser, A. (2016). High-efficiency transduction and specific expression 36. Paul, S., and Caruthers, M.H. (2016). Synthesis of Phosphorodiamidate Morpholino
of ChR2opt for optogenetic manipulation of primary cortical neurons mediated by Oligonucleotides and Their Chimeras Using Phosphoramidite Chemistry. J. Am.
recombinant adeno-associated viruses. J. Biotechnol. 233, 171–180. Chem. Soc. 138, 15663–15672.
15. Zaslavskiy, M., Bertonati, C., Duchateau, P., Duclert, A., and Silva, G.H. (2014). 37. Hecker, M., and Wagner, A.H. (2017). Transcription factor decoy technology: A ther-
Efficient design of meganucleases using a machine learning approach. BMC apeutic update. Biochem. Pharmacol. 144, 29–34.
Bioinformatics 15, 191.
38. Papaioannou, I., Simons, J.P., and Owen, J.S. (2012). Oligonucleotide-directed gene-
16. McMurrough, T.A., Brown, C.M., Zhang, K., Hausner, G., Junop, M.S., Gloor, G.B., editing technology: mechanisms and future prospects. Expert Opin. Biol. Ther. 12,
and Edgell, D.R. (2018). Active site residue identity regulates cleavage preference of 329–342.
LAGLIDADG homing endonucleases. Nucleic Acids Res. 46, 11990–12007.
39. Martínez-Gálvez, G., Ata, H., Campbell, J.M., and Ekker, S.C. (2016). ssDNA and the
17. Ochiai, H., and Yamamoto, T. (2017). Construction and Evaluation of Zinc Finger Argonautes: The Quest for the Next Golden Editor. Hum. Gene Ther. 27, 419–422.
Nucleases. Methods Mol. Biol. 1630, 1–24.
40. Ledford, H. (2016). Beyond CRISPR: A guide to the many other ways to edit a
18. Ji, H., Lu, P., Liu, B., Qu, X., Wang, Y., Jiang, Z., Yang, X., Zhong, Y., Yang, H., Pan, H., genome. Nature 536, 136–137.
et al. (2018). Zinc-Finger Nucleases Induced by HIV-1 Tat Excise HIV-1 from the
Host Genome in Infected and Latently Infected Cells. Mol. Ther. Nucleic Acids 12, 41. Luo, Z., Yang, Q., Geng, B., Jiang, S., Yang, S., Li, X., Cai, Y., and Dai, J. (2018). Whole
67–74. genome engineering by synthesis. Sci. China Life Sci. 61, 1515–1527.

19. Hensel, G., and Kumlehn, J. (2019). Genome Engineering Using TALENs. Methods 42. Krishan, K., Kanchan, T., and Singh, B. (2016). Human Genome Editing and Ethical
Mol. Biol. 1900, 195–215. Considerations. Sci. Eng. Ethics 22, 597–599.

20. Chandrasegaran, S., and Carroll, D. (2016). Origins of Programmable Nucleases for 43. Krishan, K., Kanchan, T., Singh, B., Baryah, N., and Puri, S. (2018). Germline Editing:
Genome Engineering. J. Mol. Biol. 428 (5 Pt B), 963–989. Editors Cautionary. Clin. Ter. 169, e58–e59.

21. Fischer, S., Maier, L.K., Stoll, B., Brendel, J., Fischer, E., Pfeiffer, F., Dyall-Smith, M., 44. Shinwari, Z.K., Tanveer, F., and Khalil, A.T. (2018). Ethical Issues Regarding CRISPR
and Marchfelder, A. (2012). An archaeal immune system can detect multiple proto- Mediated Genome Editing. Curr. Issues Mol. Biol. 26, 103–110.
spacer adjacent motifs (PAMs) to target invader DNA. J. Biol. Chem. 287, 33351– 45. Liao, S.M. (2019). Designing humans: A human rights approach. Bioethics 33,
33363. 98–104.
22. Eid, A., and Mahfouz, M.M. (2016). Genome editing: the road of CRISPR/Cas9 from 46. Hofmann, B. (2018). The gene-editing of super-ego. Med. Health Care Philos. 21,
bench to clinic. Exp. Mol. Med. 48, e265. 295–302.
23. Zhang, F., Wen, Y., and Guo, X. (2014). CRISPR/Cas9 for genome editing: progress, 47. Fraser, C.M., and Dando, M.R. (2001). Genomics and future biological weapons: the
implications and challenges. Hum. Mol. Genet. 23 (R1), R40–R46. need for preventive action by the biomedical community. Nat. Genet. 29, 253–256.
24. Zhang, J.H., Adikaram, P., Pandey, M., Genis, A., and Simonds, W.F. (2016). 48. Wang, H., La Russa, M., and Qi, L.S. (2016). CRISPR/Cas9 in Genome Editing and
Optimization of genome editing through CRISPR-Cas9 engineering. Bioengineered Beyond. Annu. Rev. Biochem. 85, 227–264.
7, 166–174.
49. Salsman, J., and Dellaire, G. (2017). Precision genome editing in the CRISPR era.
25. Hussain, W., Mahmood, T., Hussain, J., Ali, N., Shah, T., Qayyum, S., and Khan, I. Biochem. Cell Biol. 95, 187–201.
(2019). CRISPR/Cas system: A game changing genome editing technology, to treat
human genetic diseases. Gene 685, 70–75. 50. Sakuma, T., Nishikawa, A., Kume, S., Chayama, K., and Yamamoto, T. (2014).
Multiplex genome engineering in human cells using all-in-one CRISPR/Cas9 vector
26. Wang, L., Zheng, W., Liu, S., Li, B., and Jiang, X. (2019). Delivery of CRISPR/Cas9 by system. Sci. Rep. 4, 5400.
novel strategies for gene therapy. ChemBioChem 20, 634–643.
51. Poirier, J.T. (2017). CRISPR Libraries and Screening. Prog. Mol. Biol. Transl. Sci. 152,
27. Cai, B., Sun, S., Li, Z., Zhang, X., Ke, Y., Yang, J., and Li, X. (2018). Application of 69–82.
CRISPR/Cas9 technologies combined with iPSCs in the study and treatment of retinal
degenerative diseases. Hum. Genet. 137, 679–688. 52. Sun, B., Yang, J., Yang, S., Ye, R.D., Chen, D., and Jiang, Y. (2018). A CRISPR-Cpf1-
Assisted Non-Homologous End Joining Genome Editing System of Mycobacterium
28. Regalado, A. (2018). Exclusive: Chinese scientists are creating CRISPR babies. A
smegmatis. Biotechnol. J. 13, e1700588.
daring effort is under way to create the first children whose DNA has been tailored
using gene editing. MIT Technology Review. https://www.technologyreview.com/s/ 53. Qiu, X.Y., Zhu, L.Y., Zhu, C.S., Ma, J.X., Hou, T., Wu, X.M., Xie, S.S., Min, L., Tan,
612458/exclusive-chinese-scientists-are-creating-crispr-babies/. D.A., Zhang, D.Y., and Zhu, L. (2018). Highly Effective and Low-Cost MicroRNA
Detection with CRISPR-Cas9. ACS Synth. Biol. 7, 807–813.
29. Damian, M., and Porteus, M.H. (2013). A crisper look at genome editing: RNA-
guided genome modification. Mol. Ther. 21, 720–722. 54. Koo, T., Lee, J., and Kim, J.S. (2015). Measuring and Reducing Off-Target Activities of
Programmable Nucleases Including CRISPR-Cas9. Mol. Cells 38, 475–481.
30. Rath, D., Amlinger, L., Rath, A., and Lundgren, M. (2015). The CRISPR-Cas immune
system: biology, mechanisms and applications. Biochimie 117, 119–128. 55. Symington, L.S., and Gautier, J. (2011). Double-strand break end resection and repair
pathway choice. Annu. Rev. Genet. 45, 247–271.
31. Polstein, L.R., and Gersbach, C.A. (2015). A light-inducible CRISPR-Cas9 system for
control of endogenous gene activation. Nat. Chem. Biol. 11, 198–200. 56. Yin, H., Xue, W., Chen, S., Bogorad, R.L., Benedetti, E., Grompe, M., Koteliansky, V.,
Sharp, P.A., Jacks, T., and Anderson, D.G. (2014). Genome editing with Cas9 in adult
32. Zetsche, B., Volz, S.E., and Zhang, F. (2015). A split-Cas9 architecture for inducible
mice corrects a disease mutation and phenotype. Nat. Biotechnol. 32, 551–553.
genome editing and transcription modulation. Nat. Biotechnol. 33, 139–142.
57. Jordan, B. (2016). Les débuts de CRISPR en thérapie génique - Chroniques
33. Mali, P., Aach, J., Stranges, P.B., Esvelt, K.M., Moosburner, M., Kosuri, S., Yang, L.,
génomiques [First use of CRISPR for gene therapy]. Med. Sci. (Paris) 32, 1035–1037.
and Church, G.M. (2013). CAS9 transcriptional activators for target specificity
screening and paired nickases for cooperative genome engineering. Nat. Biotechnol. 58. Chew, W.L. (2018). Immunity to CRISPR Cas9 and Cas12a therapeutics. Wiley
31, 833–838. Interdiscip. Rev. Syst. Biol. Med. 10, https://doi.org/10.1002/wsbm.1408.

Molecular Therapy: Nucleic Acids Vol. 16 June 2019 333


www.moleculartherapy.org

Review

59. Cho, S.W., Kim, S., Kim, Y., Kweon, J., Kim, H.S., Bae, S., and Kim, J.S. (2014). 63. Zhang, B., Yang, Q., Chen, J., Wu, L., Yao, T., Wu, Y., Xu, H., Zhang, L., Xia, Q., and
Analysis of off-target effects of CRISPR/Cas-derived RNA-guided endonucleases Zhou, D. (2016). CRISPRi-Manipulation of Genetic Code Expansion via RF1 for
and nickases. Genome Res. 24, 132–141. Reassignment of Amber Codon in Bacteria. Sci. Rep. 6, 20000.
60. Shang, W., Wang, F., Fan, G., and Wang, H. (2017). Key elements for designing and 64. Fogleman, S., Santana, C., Bishop, C., Miller, A., and Capco, D.G. (2016). CRISPR/
performing a CRISPR/Cas9-based genetic screen. J. Genet. Genomics 44, 439–449. Cas9 and mitochondrial gene replacement therapy: promising techniques and ethical
61. Wassef, M., Luscan, A., Battistella, A., Le Corre, S., Li, H., Wallace, M.R., Vidaud, M., considerations. Am. J. Stem Cells 5, 39–52.
and Margueron, R. (2017). Versatile and precise gene-targeting strategies for func- 65. Callaway, E. (2016). Gene-editing research in human embryos gains momentum.
tional studies in mammalian cell lines. Methods 121-122, 45–54. Nature 532, 289–290.
62. Vojta, A., Dobrinic, P., Tadic, V., Bockor, L., Korac, P., Julg, B., Klasic, M., and Zoldos, 66. Zimmer, C. (2016). Scientists Find Form of Crispr Gene Editing With New Capabilities.
V. (2016). Repurposing the CRISPR-Cas9 system for targeted DNA methylation. The New York Times, June 3, 2016. https://www.nytimes.com/2016/06/04/science/
Nucleic Acids Res. 44, 5615–5628. rna-c2c2-gene-editing-dna-crispr.html.

334 Molecular Therapy: Nucleic Acids Vol. 16 June 2019

You might also like