Advance in Genome Editing of HIV-1 through CRISPR Technique
Jiaqi Liu
a
Davidson School of Chemical Engineering, University of Purdue, West Lafayette, IN 47907, U.S.A.
Keywords: Human Immunodeficiency Virus, Clustered Regularly Interspaced Short Palindromic Repeat (CRISPR)
/CRISPR-Associated Nuclease 9 (Cas9) System, Life Cycle, Genomic Editing, Integrated Viral Genes.
Abstract: Although tons of great efforts and improvement have been made to treat HIV-1 patients, HIV-1/AIDS remain
a big threat to global health. Although antiretroviral treatment (AT) yielded outstanding results in terms of its
impressive effect in the suppression of HIV-1 replication and expression, latent viral reservoirs in HIV-1
patients still exist and potentially activate to disrupt human health in the future. Recent advance in the
clustered regularly interspaced short palindromic repeat (CRISPR)/CRISPR-associated nuclease 9 (Cas9)
system has been well-engineered with finely designed single-guide RNA (sgRNA) to effectively target
cellular co-receptors CCR5 and CXCR4 or HIV-1 genome so that this genomic editing system can reduce
HIV-1 reduction and eradicate integrated provirus. Such a versatile genome editing technology has been
applied to several types of human cells or animal models to testify its value in the prevention of HIV-1. Here,
the progress of the CRISPR/Cas9 -based approach in recent decades will be covered and discussed. In
addition, its potential drawbacks are also analyzed for future perspectives of its full application in the HIV-1
treatment regimen.
1 INTRODUCTION
1
For centuries, human beings have been struggling
against troubles from the human immunodeficiency
virus (HIV), which causes acquired
immunodeficiency syndrome (AIDS). At present, it
has become one of the world’s most severe public
health issues in terms of its high incidence and
prevalence. HIV is normally spread through wounds,
injured skin, or blood containing HIV and sexual
intercourse, etc., which makes it omnipresent in daily
life. Indeed, its nearly ubiquitous presence has caused
a lot of death and fear around the world. Such a
depressing impact continues inflicting both
psychological and physiological pains on everyone
else in recent years, especially in the developing
countries in Africa and the Asia Pacific. In 2018 an
estimated 37.9 million people were living with HIV
(including 1.7 million children), with a global HIV
prevalence of 18% among adults. Roughly 21% of
those people do not realize that they carry the virus
(Global HIV and AIDS Statistics 2020). Statistically,
an estimated 74.9 million people have become
infected with HIV and 32 million people have died of
a
https://orcid.org/0000-0002-7206-5327
HIV-related illnesses since the start of the epidemic
(Global HIV and AIDS Statistics 2020). According
to the data gathered by Kaiser Family Foundation
(KFF), which focused on adults ages 15-49, global
prevalence has leveled since 2001 and was 0.7% in
2019 (Global HIV and AIDS Statistics 2020). A lot
of countries are still suffering from the pains it has
brought. Of those countries, Eastern and Southern
African countries are populated by most people living
with HIV, which is about 20.7 million, accounting for
54% of the total global amount. Although Asia and
the Pacific, Western and Central Europe, and North
America do not have the same amount of infected
population as those African countries, the situation is
far from being good or optimistic since they
collectively account for 21% of the infected global
population (Global HIV and AIDS Statistics 2020).
As stated above, the situation in developed
countries like the U.S. is not optimistic as well. In the
U.S., roughly 1.2 million people are living with HIV
today. About 14 percent of those people are not aware
of it and need testing (U.S. Statistics 2021). It is
obvious that HIV is hard to be tested in the early stage
and can lay dormant for several months or a couple
Liu, J.
Advance in Genome Editing of HIV-1 through CRISPR Technique.
DOI: 10.5220/0011378700003443
In Proceedings of the 4th International Conference on Biomedical Engineering and Bioinformatics (ICBEB 2022), pages 1131-1137
ISBN: 978-989-758-595-1
Copyright
c
2022 by SCITEPRESS Science and Technology Publications, Lda. All rights reserved
1131
of years. This is indeed the case; most people are
living with HIV before being diagnosed, and others
are diagnosed right after HIV infection. According to
the latest CDC data, in 2018, 37,968 people received
an HIV diagnosis in the United States and dependent
areas. From 2014 through 2018, the annual number
and rate of diagnoses of HIV infection in the United
States decreased. However, trends are mostly
disparate due to different groups of people (U.S.
Statistics 2021). In 2018, the gay or bisexual
population are the most vulnerable groups since they
accounted for 69% of new HIV diagnoses.
Heterosexual people accounted for 24% and the
remaining part all comes from people who inject
drugs (U.S. Statistics 2021). By race, the Blacks or
African American population is 13% of the U.S.
population in 2018, but they sadly contributed to 42%
of new HIV diagnoses. When it comes to age, it
reveals that people aged 25-44 held the highest
number of new HIV diagnoses (U.S. Statistics 2021).
As stated above, it is not quite hard to realize that HIV
prefers certain groups of people. It reminds people to
be careful of their choices on occupations, daily
habits, or ethnicity. Furthermore, there were 15,820
deaths among adults and adolescents with diagnosed
HIV in the U.S. in 2018 (U.S. Statistics 2021).
Given those discouraging statistical data,
scientists and pharmacists have been doing tons of
tests and related research to search for a
pharmacologically or medically efficacious regime to
tackle the problems it causes. To date, there is still no
efficacious cure for HIV-related diseases like AIDS.
Antiretroviral therapy (ART) is being incorporated
into the treatment of AIDS, along with other daily
medications. This treatment regimen yields
outstanding results due to its observable effect in
declining HIV-related deaths and simultaneously
protecting CD4 cells. It normally suppresses the viral
load to an undetectable level in the peripheral blood
of HIV patients. However, ART cannot eradicate the
integrated HIV-1 genome from latently infected cells.
This means that ART is not a desirable cure for HIV
infection and necessitates a lifelong dependence on
this therapy (Prevention 2021, HIV treatment
overview 2021). Accordingly, a promising gene-
editing tool, which is CRISPR/CAS9, has been tested
and investigated both in vivo and in vitro to resolve
those problems in the ART treatment regimen.
2 THE CRISPR/CAS9 ADVANCE
IN HIV-1 TREATMENT
2.1 Life Cycle of HIV
Based on the previous findings, two types of HIV
viruses are identified, which are HIV-1 and HIV-2.
They both can give rise to AIDS in human bodies, but
they do have a lot of distinctions regarding
pervasiveness, geographical distribution, and
lethality. Specifically, HIV-1 is the most common
HIV seen in clinical practices, and it is more fatal and
spreading more quickly than HIV-2. Therefore, HIV-
1 has been regarded as a focal point for the treatment
of AIDS. Here, all the materials and discussion
centers around it. Below is the shared mechanism by
which both HIV-1 and HIV-2 make their invasion
come true.
Here, as shown in Figure 1, In the first stage, HIV
normally enters the human body, and it strives to flow
around CD4 T lymphocytes (CD4 + cells) and
prepares for an invasion by binding its glycoprotein
120 (gp120) to the CD4 receptor on the surface of
CD4 cells, and subsequently to the co-receptors C-C
chemokine receptor type 5 (CCR5) or C-X-C
chemokine receptor type 4 (CXCR4). In the second
stage, the binding from the first stage normally gives
rise to a fusion between HIV and cell membrane,
which provides easy access for HIV to enter and
release its viral RNA. In the third stage, HIV viral
RNA was converted into double-stranded DNA
(dsDNA) with the help of reverse transcriptase. This
stage is also named after reverse transcription. The
conversion of HIV RNA to HIV DNA makes HIV
genetic material penetrate easily into the nucleus of
CD4 cells, and subsequently, HIV DNA goes through
the fourth stage where it employs integrase to embed
its viral DNA into the genome of CD4 cells. Then,
new HIV RNA, which is derived from proviral HIV
DNA, can serve as genomic RNA to manufacture
viral proteins. In the fifth stage, those viral proteins
will aggregate and fuse with HIV viral RNA, and
then they come to the cell surface to generate
immature (noninfectious) HIV particles. Then, it
comes to the last stage of its life cycle, which is
budding. During budding, immature HIV particles
are released out of CD4 cells, and simultaneously it
releases a type of HIV protease, which will generate
several mature (infectious) viruses by breaking the
long protein chains of those immature HIV particles.
ICBEB 2022 - The International Conference on Biomedical Engineering and Bioinformatics
1132
Figure 1: Life Cycle of HIV.
2.2
CRISPR/Cas9 Technology
On earth, to defend against the outside attack from
bacterial and archaeal viruses, prokaryotes have
developed a new defensive immune system clustered
regularly interspaced short palindromic repeats &
CRISPR-associated proteins (CRISPR-Cas). Given
its viral genomic editing ability, such a defense
system caught the attention of the scientific
community, especially the pharmaceutical and
medical field. Its biological value is highly evaluated
not only because of its adaptive nature, but also its
therapeutic potential. In medical applications, it
serves as a gene-editing tool by recognizing and
cleaving foreign RNA or DNA in a targeted
sequencing manner. This defense system consists of
three stages, which are adaptation, crRNA biogenesis,
and target interference, respectively.
Figure 2: CRISPR/Cas9 Technology in Genomic Editing of HIV-1.
Advance in Genome Editing of HIV-1 through CRISPR Technique
1133
As shown in Figure 2, there are three essential
stages CRISPR systems must go through, which are
spacer integration (adaptation), expression, and
interference. In the first stage, the bacteriophage
invades prokaryotic cells and releases its viral gene.
To defend against such an attack, prokaryotic cells
give that bacteriophage permission to get access to its
DNA sequence, and simultaneously cell itself
integrates such an invading nucleic acid into its DNA
sequence. Such a foreign viral gene is normally
derived from a phage of plasmid DNA, which is
called a spacer. By structure, CRISPR is comprised
of arrays of short repeating units interspaced by
spacers. The acquisition of foreign spacer paves a
way for prokaryotic cells in further defense against
invasion by phages or plasmids carrying similar
sequences.
In the expression stage, the CRISPR arrays,
which are generated from the spacer acquisition stage,
turn themselves into long primary transcripts via
transcription. Those transcripts are precursor crRNAs
(pre-crRNAs), which play a vital role in subsequent
stages. Subsequently, Pre-crRNA matures into
crRNA by virtue of Cas9 proteins, tracrRNA, and
RNase III. TracrRNA is a trans-encoded small RNA
base-paired with the repeating region located on the
Pre-crRNA.
Then, in the interference stage, an effector
complex is formed by binding of Cas9 proteins into a
single guide RNA (sgRNA), which is formed by the
linkage between tracrRNA and targeting crRNA. The
formation of this effector complex can directly
initiate and control target DNA cleavage in the
Protospacer adjacent motifs (PAMs). Such cleavage
is directed by two essential domains within Cas9
nuclease, which are the histidine-asparagine-
histidine (HNH) domain, and RuvC domain,
respectively. Target DNA strand, which matches the
base pairs with sgRNA, is normally cleaved by the
HNH domain, while the non-target DNA strand is
broken apart by the RuvC domain. Such a cleavage
will give rise to double-stranded DNA break (DSB),
which is subsequently going to be repaired by non-
homologous end-joining (NHEJ) without any
homologous template or homology-directed repair
(HDR).
2.3 CRISPR/CAS9 Technique in
Genome Editing of HIV-1
Currently, ART is still being used as a major
treatment regimen for HIV-1/AIDS patients in the
clinic. However, it cannot easily eradicate latent viral
reservoirs in HIV-1 patients. To achieve this, more
and more attention was paid to recently popular new
genome editing technology, which is the CRISPR
system. Such a functional system, along with
nuclease 9 (Cas9), is engineered to target certain
genomic regions in the HIV genome or cellular co-
factors, which can give rise to a big reduction in HIV
infection and an elimination of internal viral genes.
Based on the life cycle of HIV-1, there are two
pathways to reach our goal. The first one is to directly
target the HIV-1 genome to inactivate and eliminate
the HIV provirus. The second one is to disrupt co-
receptors CCR5 and CXCR4 by CRISPR/Cas9
technology. Tests or assays have been conducted to
verify the feasibility and effectiveness of
CRISPR/Cas9 in the suppression of HIV-1
expression and elimination of provirus within the
host cell. The CRISPR/Cas9-based approach was
first tested in HIV-1/AIDS treatment in 2013 (Ebina
et al. 2013). Their test results revealed that the
CRISPR/Cas9 system successfully targets HIV long
terminal repeat (LTR) and then suppresses the
expression of HIV genes. The target sites in their tests
were the NV-kB binding cassettes and TAE
sequences, which were situated in the U3 region of
LTR and R region, respectively. By targeting those
sites, HIV-1 provirus transcription and replication
were effectively restrained (Ebina et al. 2013). Most
importantly, the ability of CRISPR/Cas9 to eliminate
internal integrated provirus from the host cell genome
was proved. Such a desired ability is not something
we cannot see from ART treatment. The HIV-1
patients treated with ART mostly have a risk of HIV-
1 rebound since it cannot eradicate the latent provirus
inside the host cells. That is why such a functional
elimination of internal viral genes is really valued in
the clinic. Soon afterwards, Wenhui Hu and his
colleagues conducted another research to apply
CRISPR/Cas9 based approach to excise genome of
HIV-1, and they succeeded in getting some good
results showing that inactivating viral gene
expression and suppression of viral replication in a
HIV-1 latently infected T cell line, microglial cell
line with minor genotoxicity, pro-monocytic cell line,
and no detectable level of off target genomic editing,
was achieved (Hu et al. 2014). In their research, they
designed four different LTR Guide RNA (gRNA) to
separately test out their specificity and efficacy in
editing HIV-target genome. It turned out all those
gRNA worked well with different cell lines during
the assay, but not all of them were functional, which
indicates their individual specificity in genomic
editing.
Later, Liao Hsin-Kai and his group members also
selected several potential gRNA target sites located
ICBEB 2022 - The International Conference on Biomedical Engineering and Bioinformatics
1134
inside the genome of HIV-1, and they identified
optimal target sites that can provide the human body
with long-term and effective protection against HIV-
1. The host cells they used here were primary T cells
and human pluripotent stem cells (hPSC). By
analyzing mean fluorescence intensity (MFI) of
enhanced green fluorescent protein (EGFP) via
FACS analysis and the percentage of green
fluorescence protein (GFP) cells, it indicated
targeting LTR sequences in the R region was more
efficient than the other gRNA target sites (Liao et al.
2015). Then, they experimentally concluded that
different targeting/disruption strategies and
efficiencies in different stages (pre-integration and
provirus) and different targeting sites as well (coding
and non-coding regions) (Liao et al. 2015).
When double-stranded breaks (DSBs) happen,
exonucleases inside host cells are capable to degrade
the viral genome near the DSBs, and the Coding
region in the genome of HIV-1 was targeted and its
viral genes were disrupted via non-homologous end
joining (NHEJ), which includes replacement,
insertion, and deletion. On the contrary, the structural
disruption can be triggered in a pre-integration stage
when targeting non-coding regions like LTR regions
7
.
Most importantly, they found out multiplexed
CRISPR/Cas9 systems, which involve different
gRNAs, can increase the level of disruption and
excision of the pre-integrated proviral genome.
Additionally, targeting multiple conserved sites
instead of single one can effectively disrupt the
genome of HIV-1 while clearing the concern about
HIV-1 variants forming a resistance to singly guided
CRISPR/Cas9 (Liao et al. 2015).
Besides directly targeting the genome of the HIV-
1 virus, CRISPR/Cas9 can also serve as a useful tool
to prevent HIV-1 from invading primary T cells or
other cell lines by editing of co-receptor, which are
CCR5 and CXCR4. As stated above, HIV-1 makes
its entry into CD4 T lymphocytes by binding to the
CD4 receptor, as well as CCR5 and CXCR4. If either
CCR5 or CXCR4 was genetically modified or
engineered, a negative impact on entry of HIV-1 will
be expected. In the research of Liu et al, they
designed two types of sgRNAs, which served as a
tool to target CXCR4 and CCR5 at the same time.
What they were trying to do was apply CRISPR-
sgRNAs-Cas9 to trigger the genomic editing of
CXCR4 and CCR5 in different cell lines including
CD4 T cells (Liu et al. 2017). Then, by off-target and
apoptosis assays to figure out if such editing can
result in any non-specific editing or cytotoxic effect
on cell viability. The final result turned out to be very
impressive because both CXCR4 modified and
CCR5 modified cells exhibited a selective advantage
over unmodified cells during HIV-1 infection. Most
importantly, there weren’t any nonspecific editing or
cytotoxic symptoms, which concluded that
CRISPR/Cas9 based approach in editing both co-
receptors are quite safe and effective to suppress X4-
or/and R5-tropic HIV-1 infection (Liu et al. 2017).
Overall, the CRISPR/Cas9 systems have an
apparent advantage over the other gene-editing
techniques, such as ZFN and TALEN. CRISPR/Cas9
can change the cleavage target by simply changing
the gRNA sequence. ZFN and TALEN systems need
a redesigned protein binding domain to make
changing of cleavage target sequences occur (Gaj et
al. 2012, Khalili et al. 2017, Ousterout et al. 2016).
Such an advantage makes CRISPR/CAS9 impressive
from a design perspective. Besides, Yin Chaoran et al
demonstrated the effective excision of HIV-1
proviral DNA from the host-host genome in pre-
clinical animal models using saCas9 and multiplex
sgRNAs by AAV-DJ/8 vector (Yin et al. 2017). Their
research further supports the application of
CRISPR/CAS9 in HIV treatment by animal model
validation, which really lowers the sounds of those
skeptics.
Despite the promising aspects of genomic editing
of CRISPR/CAS9, challenges still exist. At present,
one major concern is probably the off-target effect,
which may give rise to genetic mutation or chromosol
translocation. If CRISPR/Cas9 enters the clinic field,
the reduction of the off-target effect would be
necessary and urgent. Some researchers have already
proved that the off-target effect found in
CRISPR/Cas9 system was very limited compared
with other editing techniques such as TALENs, ZFNs,
or homing endonucleases by ChIP-seq (Duan et al.
2014). However, improvement is still needed for
CRISPR/Cas9 to eliminate mismatches as many as
possible.
Another challenge would be what kind of vectors
should be utilized to deliver CRISPR/Cas9. As we
know, CRISPR/Cas9 system is normally introduced
into host cells through transfection. Normally, a
successful transfection does need an appropriate
delivering vector. The main vector being used
consists of lentiviral, adenoviral, and adeno-
associated viral vectors. Of those vectors, lentiviral
vectors are widely utilized to deliver CRISPR
systems with high efficiency and stable expression,
but simultaneously the chances of off-target effect
can be even higher (Wang et al. 2014). It is apparent
that finding a vector with less off-target would be a
big concern we should always keep in mind.
Advance in Genome Editing of HIV-1 through CRISPR Technique
1135
Also, HIV-genetic variation is highly likely to
badly influence the efficacy of a CRISPR/Cas9 based
treatment regimen. To resolve this problem,
researchers designed dual-gRNA/Cas9 strategy, and
their results demonstrated that although such a
strategy can cure T cells infected by distinct HIV-1
isolates, the efficacy of this strategy is really
compromised by sequence variation of the target sites
in HIV-1 (Darcis et al. 2014). Later on, a quadruplex
sgRNAs/saCas9 strategy was applied and tested. The
results demonstrated maximization of the possibility
of multiple indel mutations on six on-target sites and
fragmental deletions among these sites Furthermore,
this strategy can offer additional advantages, such as
reducing the potential of HIV-1 escape (Wang et al.
2016), the high possibility of HIV-1 excision despite
the continuous proviral mutation in the clinical HIV-
1 patient population, a reliable loss-of-function
achievement due to removal of a substantial portion
of the target gene or genome (Bauer et al. 2015), and
optimal efficiency of excision (Yin et al. 2016).
3 CONCLUSIONS
Currently, ART is still being used as the major
treatment regimen for HIV-1/AIDS patients in the
clinic. By the long-term effect of ART, there would
be an obvious decrease in HIV-1 expression and
symptoms can be alleviated to an undetectable level.
However, meanwhile, a lot of side effects, such as
appetite loss, or lipodystrophy, become a big concern.
Although there has been a lot of improvement in ART
to minimize its possible side effects, some severe side
effects are still out there. Additionally, ART does
require a long-term intake regularly. If patients forget
or skip one of the doses of the day or the week, the
HIV-1 virus would take full advantage of this chance
to replicate and copy itself in their bodies again. At
worst, it could result in drug-resistant, and then there
would be few feasible HIV-1 treatment regimens for
HIV patients to choose from. Hence, genomic editing
technologies start to get more and more attention due
to their potential in the suppression of HIV-1 viral
gene and elimination of integrated HIV-1 viral gene
within CD4 lymphocytes. CRISPR/Cas9 is one of
them and the one with no cytotoxic effect and less
off-target effect. However, there is still a long way
ahead until this technique can be fully applied to the
clinic field. Off-target and delivery vectors need
more time and effort to deal with and eventually this
technique is going to be more mature and feasible to
be clinically qualified enough in the near future.
REFERENCES
Bauer D. E., Canver M. C., & Orkin S. H., Generation of
genomic deletions in mammalian cell lines via
CRISPR/Cas9. J Vis Exp, e52118, doi:10.3791/52118
(2015).
Duan J., Lu G., Xie Z., Lou M., Luo J., Guo L., & Zhang
Y., Genome-wide identification of CRISPR/Cas9 off-
targets in human genome. Cell Res 24, 1009-1012,
doi:10.1038/cr.2014.87 (2014).
Darcis G., Binda C. S., Klaver B., Herrera-Carrillo E.,
Berkhout B., & Das A. T., The Impact of HIV-1
Genetic Diversity on CRISPR-Cas9 Antiviral Activity
and Viral Escape. Viruses 11, doi:10.3390/v11030255
(2019).
Ebina H, Misawa N., Kanemura Y., & Koyanagi Y.,
Harnessing the CRISPR/Cas9 system to disrupt latent
HIV-1 provirus. Sci Rep 3, 2510,
doi:10.1038/srep02510 (2013).
Global HIV and AIDS Statistics,
<https://www.avert.org/global-hiv-and-aids-statistics>
(2020).
Gaj, T., Guo, J., Kato, Y., Sirk, S. J. & Barbas, C. F., 3rd.
Targeted gene knockout by direct delivery of zinc-
finger nuclease proteins. Nat Methods 9, 805-807,
doi:10.1038/nmeth.2030 (2012).
HIV.gov. HIV treatment overview. (2021)
Hu W., Kaminski R., Yang F., Zhang Y., Cosentino L., Li
F., Luo B., Alvares-Caarbonell D., Garcia-Mesa Y.,
Karn J., Mo X., & Khalili K., RNA-directed gene
editing specifically eradicates latent and prevents new
HIV-1 infection. Proc Natl Acad Sci U S A 111, 11461-
11466, doi:10.1073/pnas.1405186111 (2014).
Khalili K., White M. K., & Jacobson J. M., Novel AIDS
therapies based on gene editing. Cell Mol Life Sci 74,
2439-2450, doi:10.1007/s00018-017-2479-z (2017).
Liao H. K., Gu Y., Diaz A., Marlett J., Takahashi Y., Li M.,
Suzuki K., Xu R., Hishida T., Chang C.J., Esteban
C.R., Young J., & Izpisua Belmonte J.C., Use of the
CRISPR/Cas9 system as an intracellular defense
against HIV-1 infection in human cells. Nat Commun
6, 6413, doi:10.1038/ncomms7413 (2015).
Liu Z., Chen S., Jin X., Wang Q., Yang K., Li C., Xiao Q.,
Hou P., Liu S., Wu S., Hou W., Xiong Y., Kong C.,
Zhao X., Wu L., Li C., Sun G., & Guo D., Genome
editing of the HIV co-receptors CCR5 and CXCR4 by
CRISPR-Cas9 protects CD4(+) T cells from HIV-1
infection. Cell Biosci 7, 47, doi:10.1186/s13578-017-
0174-2 (2017).
Ousterout D. G., & Gersbach C. A., The Development of
TALE Nucleases for Biotechnology. Methods Mol
Biol 1338, 27-42, doi:10.1007/978-1-4939-2932-0_3
(2016).
Prevention, C. f. D. C. a. Living with HIV (2021)
U.S. Statistics, <https://www.hiv.gov/hiv-
basics/overview/data-and-trends/statistics> (2021)
Wang W., Ye C., Liu J., Zhang D., Kimata J.T., & Zhou P.,
CCR5 gene disruption via lentiviral vectors expressing
Cas9 and single guided RNA renders cells resistant to
ICBEB 2022 - The International Conference on Biomedical Engineering and Bioinformatics
1136
HIV-1 infection. PLoS One 9, e115987,
doi:10.1371/journal.pone.0115987 (2014).
Wang Z., Pan Q., Gendron P., Zhu W., Guo F., Cen S.,
Wainberg M. A., & Liang C., CRISPR/Cas9-Derived
Mutations Both Inhibit HIV-1 Replication and
Accelerate Viral Escape. Cell Rep 15, 481-489,
doi:10.1016/j.celrep.2016.03.042 (2016).
Wang G., Zhao N., Berkhout B. & Das A. T., CRISPR-
Cas9 Can Inhibit HIV-1 Replication but NHEJ Repair
Facilitates Virus Escape. Mol Ther 24, 522-526,
doi:10.1038/mt.2016.24 (2016).
Yin C., Zhang T., Qu X., Zhang Y., Putataunda R., Xiao
X., Li F., Xiao W., Zhao H., Dai S., Qin X., Mo X.,
Young W.B., Khalili K., & Hu W., In Vivo Excision of
HIV-1 Provirus by saCas9 and Multiplex Single-Guide
RNAs in Animal Models. Mol Ther 25, 1168-1186,
doi:10.1016/j.ymthe.2017.03.012 (2017).
Yin C., Zhang T., Li F., Yang F., Putatunda R., Young W.
B., Khalili K., Hu W., & Zhang Y. Functional
screening of guide RNAs targeting the regulatory and
structural HIV-1 viral genome for a cure of AIDS.
AIDS 30, 1163-1174,
doi:10.1097/QAD.0000000000001079 (2016).
Advance in Genome Editing of HIV-1 through CRISPR Technique
1137