You are on page 1of 8

Gene Therapy (2017) 24, 377–384

© 2017 Macmillan Publishers Limited, part of Springer Nature. All rights reserved 0969-7128/17
www.nature.com/gt

REVIEW
Current application of CRISPR/Cas9 gene-editing technique
to eradication of HIV/AIDS
Z Huang, A Tomitaka, A Raymond and M Nair

Human immunodeficiency virus (HIV)/acquired immunodeficiency syndrome (AIDS) remains a major health hazard despite
significant advances in prevention and treatment of HIV infection. The major reason for the persistence of HIV/AIDS is the inability
of existing treatments to clear or eradicate the multiple HIV reservoirs that exist in the human body. To suppress the virus
replication and rebound, HIV/AIDS patients must take life-long antiviral medications. The clustered regularly interspaced
palindromic repeats (CRISPR)/CRISPR-associated nuclease 9 (Cas9) system is an emerging gene-editing technique with the potential
to eliminate or disrupt HIV-integrated genomes or HIV-infected cells from multiple HIV reservoirs, which could result in the
complete cure of HIV/AIDS. Encouraging progress has already been reported for the application of the CRISPR/Cas9 technique to
HIV/AIDS treatment and prevention, both in vitro in human patient cells and in vivo in animal model experiments. In this review, we
will summarize the most recent progress in the application of the CRISPR/Cas9 gene-editing technique to HIV/AIDS therapy and
elimination. Future directions and trends of such applications are also discussed.

Gene Therapy (2017) 24, 377–384; doi:10.1038/gt.2017.35

INTRODUCTION human brain. Neurons are believed to be resistant to HIV


Human immunodeficiency virus (HIV)/acquired immunodefi- infection; however, the neurotoxic products released from HIV-
ciency syndrome (AIDS) is one of the most severe pandemic infected brain cells significantly dysregulate neuronal function
diseases in modern history. In 2015, according to the World and homeostasis.26
Health Organization (WHO), about 36.7 million people world- The ultimate cure for HIV/AIDS will be the removal or disruption
wide were living with HIV, and roughly 2.2 million people died of of integrated HIV provirus from latently infected cells or the
AIDS, while more than 2.1 million new patients were diagnosed elimination of these latent cells completely. However, until
with HIV infection. Of all people diagnosed with HIV infections, it recently, gene therapy for HIV/AIDS has progressed very slowly.
is estimated that only 17 million (46%) are on antiretroviral The breakthrough of gene-editing technology raises the hope for
therapy (WHO). With breakthroughs in HIV/AIDS prevention, eradication of HIV as the provirus can be removed from the host
diagnosis, and treatment, the morbidity and mortality of HIV/ cell genome through the newly developed technique of clustered
AIDS has decreased significantly. However, HIV/AIDS remains an regularly interspaced short palindromic repeat (CRISPR)/CRISPR-
incurable, chronic disease due to the multiple HIV latent associated protein 9 (Cas9) gene-editing.27–30 Furthermore,
reservoirs in patients’ bodies. These reservoirs primarily contain CRISPR/Cas9 system can be utilized to induce apoptosis in
very small numbers (for example, about 1 per million memory reservoir cells.31,32 Originally, an adaptive immune response
CD4+ T cells) of latently infected cells1–4 which are located in the mechanism in bacteria,33,34 the CRISPR/Cas9 system has emerged
brain,5–7 peripheral blood,2,3,8,9 lymphoid tissue,1,10,11 gastro- as a popular gene-editing system with proven efficiency in
intestinal tract,10,11 and other locations.12,13 Complicated mammalian cells. As early as 2013, soon after the CRISPR/Cas9
mechanisms underlie the persistence of HIV reservoirs, including technique emerged, its potential applications for treating
the stability of reservoir cells;4 homeostatic proliferation of HIV/AIDS were considered.28 This mini-review addresses the use
reservoir cells;14 replenishment of reservoir cells;15 and low of the CRISPR/Cas9 system as a powerful gene-editing tool to
levels of antiviral drug penetration.16–18 Moreover, even under delete or disrupt HIV provirus from latently infected cell genomes
antiretroviral therapy (ART), about 30–50% of AIDS patients or to knock out CXCR4 and CCR5 receptors for HIV infection
eventually develop HIV-associated neurological disorders treatment and prevention. The elimination or disruption of HIV
(HAND),19,20 which are cognitive, motor and/or behavioral provirus in reservoir cells or the elimination of reservoir cells may
impairments caused by HIV infection in the brain.6,20–23 HAND result in a cure for HIV/AIDS.
can further be grouped into asymptomatic neurocognitive
impairment (ANI), minor neurocognitive disorder (MND) and
the most severe, HIV-associated dementia (HAD).21 The mechan- OVERVIEW OF HIV PRODUCTIVE AND LATENT INFECTION
ism of HAND remains to be elucidated, but it is generally HIV is a retrovirus that belongs to the lentivirus genus. Its DNA
accepted that HAND is tightly correlated with HIV infection of genome has two long terminal repeat (LTR) structures flanking the
astrocytes,22,24–26 microglial cells5,26 and macrophages5,6 in the whole virus genome, which consists of nine overlapping genes:

Department of Immunology, Institute of NeuroImmune Pharmacology, Center for Personalized Nanomedicine, Herbert Wertheim College of Medicine, Florida International
University, Miami, FL, USA. Correspondence: Professor M Nair or Dr Z Huang, Department of Immunology, Institute of NeuroImmune Pharmacology, Herbert Wertheim College of
Medicine, Florida International University, 11200 SW 8th Street, Miami, FL 33199, USA.
E-mail: nairm@fiu.edu or zahuan@fiu.edu
Received 25 August 2016; revised 25 January 2017; accepted 1 February 2017; accepted article preview online 4 May 2017; advance online publication, 1 June 2017
Application of CRISPR/Cas9 gene-editing technique to eradication of HIV
Z Huang et al
378
gag, pol, env, tat, rev, nef, vif, vpr, vpu (Figure 1a). HIV is the HIV targets CD4+ human immune cells, including T cells,
causative agent of AIDS in human patients, and it can be dendritic cells and monocytes, as well as cells within the central
transmitted through contact with infected blood or other body nervous system (CNS), such as perivascular macrophages, astro-
fluids through broken skin or mucous membranes. The two most cytes, and microglial cells. The life cycle of HIV is quite complex
common ways to spread HIV/AIDS are unprotected sex and needle (Figures 1b and c). HIV establishes two types of infection in target
sharing for intravenous drug injection. cells—active infection (Figure 1b) or latent infection (Figure 1c).

Figure 1. HIV life cycle. (a) HIV DNA genome HIV-1 DNA genome structure is illustrated above and each gene is drawn in ratio. The viral
genome is ~ 10 kb long and contains nine viral genes. All the viral genes’ locations are identified. In addition, tat and rev have two exons. TAT
protein is expressed in a minor form (72 amino acids) and a major forms (86 amino acids), while REV protein is expressed as one form of 116
amino acids. Genes of gag and env encode structural proteins; gene of pol encodes enzymes and all other genes encode regulatory proteins.
(b). HIV active infection: In this illustration, HIV life cycle is divided into seven stages: Binding, fusion and entry (1 and 2): HIV binds to immune
cell surface receptors, including CD4 and CXCR4 or CD4 and CCR5. The binding causes conformation changes and results in the membrane
fusion between HIV and cell membrane. HIV enters the cell, disintegrates and releases the viral RNA and enzymes. Reverse transcription (3):
HIV viral reverse transcriptase converting viral RNA into doubled strand viral DNA. Integration (4): HIV DNA enters host cell nucleus and
integrates into host cell genome with the help of viral integrase. Replication (5): Integrated HIV DNA uses host cell machinery to translate viral
proteins and transcribe viral RNA. Assembly (6): the components of HIV, including the HIV RNA and proteins move to the cell membrane to
form an immature viral particle. Budding (7): The new immature viral particles are released from cell membrane and become mature and
infectious after viral protease action. (c). HIV latent infection: In HIV latent infection, HIV life cycle stops at stage 4 after HIV DNA genome
integration. However, under certain conditions, the integrated HIV genome can be activated and the whole life cycle described in 1B is
accomplished.

Gene Therapy (2017) 377 – 384 © 2017 Macmillan Publishers Limited, part of Springer Nature.
Application of CRISPR/Cas9 gene-editing technique to eradication of HIV
Z Huang et al
379
Active infection occurs in most cells, while latent infection occurs (Figure 2a). Each domain can cleave a DNA strand directed by a
in much fewer cells1,2 and at very early stages of HIV infection.9,35 sgRNA complementary to the target DNA sequence (generally 20
In active infection, HIV provirus is active and HIV virus particles are nucleotides long). The prerequisite to be a target sequence is the
actively replicated; and the infected cells continuously release viral presence of a NGG sequence (N: any nucleotides; G: guanine
progeny; while in latent infection, HIV provirus is transcriptionally nucleotide, also called protospacer adjacent motifs (PAMs))
silenced and no viral progeny is produced. However, under certain downstream (3′ end) of the target site.59,64–66 In the nucleus,
conditions, such as normal immunologic responses to various Cas9, sgRNA and target DNA form a complex, then HNH and RuvC
recall antigens or routine vaccination,36 or the induction of domains each cleave a DNA strand59,64–66 (Figure 2b). The double-
cytokines,2,35 latently infected cells are reactivated to produce strand DNA breaks are consequently repaired by two different
virus;2 subsequently, a pool of new latent cells are generated by approaches. One is non-homologous end-joining (NHEJ) when
these reactivated latent cells.35 These reservoirs ultimately hinder there is no template and the other is homology-directed repair
HIV/AIDS eradication. The mechanisms of how latent HIV infection (HDR) when there is a homologous nucleotide template present
occurs remain elusive, but evidence suggests that the establish- such as ssDNA or dsDNA. This template must possess homology
ment of an HIV reservoir is related to the following factors: HIV sequences (arms) to the two regions that flank the Cas9 excision
provirus integration site;37–39 chromatin environment;40,41 acces- site. If the two homologous arms flank a correct sequence, which
sibility of transcription factors41–43 and RNA interference.44–46 can be used as a template to ‘repair’ the genomic sequences.59,67
Latently infected cells are long-lived and mainly reside in reservoir NHEJ usually results in an insertion or deletion in a DNA sequence,
sites where antiviral drugs may not penetrate, such as the brain,5–7 in total called indel, while HDR results in correct repair as directed
lymphoid tissue1,10,11 and gastrointestinal tract.10,11 Latently by the template.59,67 CRISPR/Cas9 system can also be used in
infected cell populations include memory CD4+ T cells,1–4,10 biological functions other than gene-editing. For example,
macrophages,5,6,11 microglial cells5,26 and astrocytes.22,24–26 The catalytically inactive Cas9 (dCas9) has no active nuclease domains
establishment of an HIV reservoir occurs soon after infection, and and is often applied to be a site-specific DNA-binding factor.68 The
early application of antiviral drugs typically only diminishes rather fusion protein of dCas9 and transcription activator or repressor
than eliminates the reservoir.9,35 domain are often used to regulate gene expressions.59,68 In
To eliminate the viral reservoirs, current ‘shock and kill’ efforts summary, CRISPR/Cas9 system is a full developed multifunction
seek to reactivate the latent reservoirs (the shock) and destroy the gene-editing and regulating system which has unlimited
reactivated cells (the kill) through an intensifying combination of potential.59,67
antiretroviral therapy (cART) and host immune responses.47
Research efforts have shown that using SAHA, a histone
deacetylase inhibitor, in conjunction with cART reactivates latent APPLICATION OF CRISPR/CAS9 SYSTEM TO HIV/AIDS
HIV in infected CD4+ T cells, making these cells susceptible to PREVENTION AND TREATMENTS
specific CD8 T-cell killing in vitro.48,49 However, none of the clinical The first attempt to apply CRISPR/Cas9 system to HIV/AIDS
studies demonstrated a significant therapeutic effect as measured eradication was conducted in Japan in 2013. Ebina et al.28 tried
by HIV DNA or quantitative viral outgrowth assay, which means to eliminate HIV provirus from T cells by targeting the LTR region
they are ineffective.48,49 Now ‘shock and kill’ efforts have of HIV provirus. CRISPR/Cas9 system was applied to target TAR
expanded to include testing and optimizing an increasing number sequence of the R region and NF-κB-binding sequence in the U3
of latency-reversing compounds combined with host immuno- region. The indel formation by CRISPR/Cas9 cleavage of TAR
modulatory strategies.47 The setbacks of ‘shock and kill’ have region is more effective to disrupt provirus transcription and
encouraged researchers to look for alternative approaches to replication.28 The indel formation by CRISPR/Cas9 cleavage of LTR
eradicate HIV reservoirs. One approach to complete HIV eradica- resulted in inhibition of active provirus expression and decreased
tion would involve the actual removal or disruption of the provirus reactivation of latent provirus.28
from HIV reservoir cells. This type of gene therapy approach has For HIV prevention, many investigators focus on applying the
recently been investigated using CRISPR/Cas9 gene-editing CRISPR/Cas9 technique to the deletion or disruption of the HIV
system with promising results.27–30 receptors’ expression on human cells. Since CD4 is the essential
primary receptor in human T cells and is indispensable for normal
cell functions, HIV secondary receptors of CXCR4 and CCR5 are
OVERVIEW OF CRISPR/CAS9 TECHNOLOGY often candidates for the deletion and disruption by CRISPR/Cas9
The ultimate goal of gene-editing is gene repair. Several gene- technique. T-cell genome engineering holds great promise for
editing technologies recently demonstrated strong potential for cell-based therapies for HIV eradication due to the rapid
therapeutic uses. These technologies include zinc-finger nucleases development of gene-editing. Hou et al.69 efficiently disrupted
(ZFNs),50–52 transcription activator–like effector nucleases the CXCR4 gene with CRISPR/Cas9 technology in human primary
(TALENs),53–55 and CRISPR/Cas9 system.34,56–59 In principle, ZFNs CD4+ T cells. Ten sites within the CXCR4 gene were targeted and
and TALENs fuse DNA nuclease and DNA-binding domains CXCR4 expression decreased by ~ 30% as confirmed by western
together to break DNA at specific loci, while Cas9 nucleases work blot.69 The engineered T cells showed resistance to HIV infection
with guide RNA to break DNA at specific loci. Both ZFNs and and p24 production reduced over 60%.69 More importantly, this
TALENs techniques have significant applications and studies in technology indicated high specificity and no off-target effects,
HIV/AIDS prevention and therapy.60–63 However, this review will while cell division and propagation remained normal.69 Schumann
be focusing solely on the CRISPR/Cas9 system due to the et al.70 used Cas9/sgRNA ribonucleoproteins (Cas9 RNPs) to ablate
limited scope. CXCR4 gene expression in human T cells, and about 40% of cells
Compared to the initial CRISPR/Cas9 system in bacteria, the lost surface expression of CXCR4. When paired with repair
current CRISPR/Cas9 system has been simplified, modified and template, Cas9 RNPs produced a desired genome modification
adapted for mammalian genome editing and bioengineered for in human primary T cells.70 Deep sequencing confirmed that Cas9
better nucleus localization and mammalian cell expression.57,59 RNPs achieved about 20% efficiency of knockin genome
The current CRISPR/Cas9 system includes a single guide RNA modifications.70 Because of the success of the Berlin Patient case,
(sgRNA) consisting of crRNA and tracRNA57,59 and Cas9, which Ye et al.71 tried to produce CCR5Δ32-induced pluripotent stem
excises the target DNA.57,59 Cas9 is a nuclease of 1368 amino acids cells (iPSCs) using CRISPR/Cas9 system. Close to 100% efficiency
with two nuclease activity domains named HNH (residues 775– was achieved for homologous recombination of a single allele and
908) and RuvC (residues 1–59, 718–769 and 909–1098)59,64–66 up to 33% of double alleles.71 As expected, monocytes and

© 2017 Macmillan Publishers Limited, part of Springer Nature. Gene Therapy (2017) 377 – 384
Application of CRISPR/Cas9 gene-editing technique to eradication of HIV
Z Huang et al
380

Figure 2. CRISPR/Cas9 protein structure and working mechanism. (a) CRISP/Cas9 protein domain structure: CRISPR/Cas9 protein structure is
illustrated above. The number of amino acids are labeled above the protein schema, and the domain names are listed below the protein
schema. HNH nuclease domain is labeled in green, and RuvC nuclease domain consists of three sub-domains, which are labeled in red.
(b) CRISPR/Cas9 working mechanism: In detail, Cas9 has two nuclease domains (HNH and RuvC) represented by two grey oval shapes in the
figure. Each nuclease domain can cleave a DNA strand directed by a sgRNA (In this figure, a sgRNA is in a black box), which is complementary
to the target DNA sequence. The prerequisite to be a target sequence is the presence of a NGG (In this figure, it is CGG) sequence downstream
of the target site (3’ end). The length of the target sequence is generally 20 nucleotides (In this figure the target sequence is in green). In cell
nucleus, Cas9, sgRNA and double-strand target DNA form a complex. Each nuclease domain cleave a DNA strand at the third nucleotide from
the NGG (indicated by a thin red line). The double-strand breaks are consequently repaired mainly by either non-homologous end-joining
(NHEJ) or homology-directed repair (HDR) when there is a homologous repair template (not shown). NHEJ is error prone and often results in
deletions and insertions while HDR is a high-fidelity repair according to the homologous repair template. In this figure, an insertion NHEJ is
shown in underlined bold black and a faithful HDR is shown in underlined bold green.

macrophages derived from such engineered iPSCs were found to 50% of the analyzed patients; and (4) HAART therapy might
be resistant to HIV infection.71 Wang et al.72 applied CRISPR/Cas9 reduce the required sgRNA scale to eradicate provirus. For the first
technology to block CCR5 expression in human CD4+ cells. time, the study demonstrated the feasibility of HIV provirus
Lentiviral vectors were used to target three sites of the CCR5 elimination with CRISPR/Cas9 technology in a single patient.27 Zhu
gene.72 The disruption ratio of CCR5 was high, and those cells et al.74 targeted 10 sites of the HIV provirus to eradicate HIV DNA
were resistant to R5 tropic HIV with negligible off-target effect.72 from Jukat cell genome. These sites include three within LTR
However, attempts to disrupt CCR5 gene in human primary T cells region, five within pol gene, and two within rev gene.74 The
with the same approach failed for unknown reasons.72 In another second exon of rev ranked as the best targeting site, and the
study, Li et al.73 applied adenoviral vectors to deliver CRISPR/Cas9 targeting at this site caused a 20-fold reduction in HIV
system to target multiple sites of the fourth exon of CCR5 gene to production.74 To eradicate the latent HIV reservoirs and achieve
block CCR5 expression. First, the TZM-b1 cell line was tested and a complete cure of HIV/AIDS, the ‘shock and kill’ strategy is often
two important target sites, which knockout over 60% of the gene applied to reactivate the latent HIV reservoirs and then kill those
were identified. Secondly, the results were confirmed in a CHO activated reservoir cells by ART.47–49 However, the efficiency,
and a human T-cell lines. Thirdly, CRISPR/Cas9 technique specificity and toxicity of current drugs turned out to be
conferred TZM-B1 cell the resistance to HIV infection. Finally, a disappointing.47–49 Now CRISPR/Cas9 system is utilized to execute
chimeric Ad5/F35 adenoviral vector was used to deliver the the ‘shock and kill’ strategy, in detail, catalytically deficient Cas9
CRISPR/Cas9 system to human primary CD4+ T cells to disrupt the (dCas9)-transcription activator fusion protein/sgRNA is applied to
CCR5 gene, which provided the primary T-cells the resistance to specifically activate HIV latent reservoirs.31,32 With this technique,
HIV infection.73 Zhang et al.32 targeted 20 sites within the LTR U3 region of HIV
Besides the deletion or disruption of HIV receptors on human provirus and found that the two best activation sites are at or near
cells, many scientists have applied the CRISPR/Cas9 technique to the NF-κB-binding site. This targeted activation potently reacti-
remove or disrupt HIV provirus in latently infected human cells vated several HIV cell lines, including HIV-1 latent TZM-bI
(Figure 3). The Drexel Medicine CNS AIDS Research and epithelial, Jurkat T cells and CHME5 microglial cells.32 Moreover,
Eradication Study (CARES) cohort studies27 demonstrated the the reactivation resulted in cell death of Jurkat T cells and CHME5
following: (1) The proviral LTR sequences within the peripheral microglial cells due to the accumulation of viral proteins of TAT,
blood mononuclear cells (PBMC) decreased variations every year NEF, REV and so on.32 Almost at the same time, similarly, Saayman
with ART; (2) HIV LTR sequences continued genetic changes for a et al.31 targeted 23 sites in the LTR U3 region of the HIV provirus
minimum of 6 years even with effective suppressive ART; (3) Next- and identified the best activation site as at near the NF-κB-binding
generation sequencing (NGS) analysis found that a regimen of sites. The targeting system worked in many different in vitro latent
sgRNAs could be designed to target all known HIV quasispecies in T-cell models to reactivate provirus, and these reactivations were

Gene Therapy (2017) 377 – 384 © 2017 Macmillan Publishers Limited, part of Springer Nature.
Application of CRISPR/Cas9 gene-editing technique to eradication of HIV
Z Huang et al
381

Figure 3. Application of CRISPR/Cas9 technology to eradicate HIV reservoir cells. The major application of CRISPR/Cas9 technology to
eradicate HIV latent reservoir cells (a) is to eliminate (b) or disrupt the HIV provirus (c) from host reservoir cells genome. In detail, several
sgRNAs, which target two or more sites of HIV provirus are applied to HIV reservoir cells with Cas9. This action results in multiple cuts at the
multi-sites of HIV provirus, thus provirus is removed from host cell DNA (b) or provirus is disrupted (c). The DNA of host HIV reservoir cells is
further repaired by DNA non-homologous end-joining or homology-directed repair when the homologous repair template is available (not
shown).

consistent and more efficient than latency breaking reagents such reagents due to the host cell DNA repair. Yoder et al.78 recently
as prostratin and SAHA.31 More importantly, the activation was reported research results with the similar data and conclusions.
specific to the provirus, while latency breaking reagents caused These negative findings suggest the need for careful design and
widespread side effects. In addition, the reactivation also blocked cautious evaluations of the application of the CRISPR/Cas9
the NF-κB reactivation.31 Hu et al.29 tried using the CRISPR/Cas9 technique to HIV/AIDS eradication. Multiplex gRNAs and several
system to eradicate HIV provirus in microglial cells, pro- excision sites of a provirus may be necessary to forever eliminate
monocytes, and T cells. The LTR U3 region was tested for four or disrupt HIV provirus in reservoir cells.
sites individually or in combination. It turned out that two sites
combined approach achieved the best eradication results.29
Furthermore, no off-target effects were found, and the stable CRISPR/CAS9 SYSTEM DELIVERY APPROACH
expression of Cas9 and sgRNA conferred cells with the ability to The major challenge of curing HIV/AIDS lies in the persistence of
resist new HIV infections.29 Kaminski et al.30 applied a similar multiple reservoirs in several drug- inaccessible parts of the
approach to remove the entire HIV provirus from latently infected human body. Moreover, while current HIV/AIDS patients can take
human CD4 T cells. No off-target effects were found and cell medications to suppress symptoms, the side effects of these drugs
health parameters were not affected.30 The persistent expression may contribute to high morbidity in these patients. The complete
of Cas9 and sgRNA protected the human CD4 T cells from new HIV cure is the elimination or disruption of HIV provirus from the
infection.30 The delivery of Cas9 and sgRNA using lentivirus vector patient’s reservoir cells’ genomes or the elimination of patient
could reduce HIV replication in HIV-infected primary human T cells reservoir cells. As stated above, gene-editing technology makes it
and decrease viral load in ex vivo patient CD4+ T cells.30 Kaminski possible to manipulate DNA at the cell genomic level. Since its
et al.75 has also applied a recombinant adeno-associated virus 9 discovery, CRISPPR/Cas9 system has been a premier tool in gene-
(rAAV9) vector expressing saCas9 (a smaller version of Cas9 editing. As mentioned before, many experimental efforts and
derived from Staphylococcus aureus to fit AAV vectors) and sgRNAs clinical trials are underway for the application of the CRISPR/Cas9
to remove integrated HIV DNA in a mouse transgenic model. A system for eradication of HIV/AIDS. The most substantive progress
978 bp DNA fragment was removed from integrated HIV DNA to date has been reported are ex vivo studies. Future efforts will be
from lymphocytes and cells in multiple organs, including brain, directed towards efficient delivery of CRISPR/Cas9 system
heart, kidney, liver, lung and spleen.75 Similar results were found in components to latently infected cells.
transgenic rat experiments.75 This is the first report of such studies Currently, the delivery methods of CRISPR/Cas9 system are
in transgenic animal models. These encouraging progress divided into viral delivery and non-viral delivery. The CRISPR/Cas9
demonstrated the great promise of CRISPR/Cas9 technique in system is very flexible and can be delivered in the form of DNA,
eradication of HIV/AIDS. But there were scientific findings that RNA or protein/RNA complex. The most popular approach is the
pointed out some pitfalls of CRISPR/Cas9 application to HIV/AIDS DNA form as one or several plasmids. In the viral delivery
eradication. Wang et al.76 demonstrated that Cas9/sgRNA can methods, lentivirus,29,79,80 baculovirus81 and recombinant adeno-
inhibit HIV replication initially, but soon HIV-1 escape variants associated virus (rAAV) have been tried.75,80 Among these virus
were produced due to the NHEJ repair, and the escape variants vectors, rAAV has the advantages of high safety, flexible
contained mutations around the Cas9 cleavage sites. Interestingly, administration routes and low or no immune response, but it
the more conservative the Cas9/gRNA targeting sequences were, has the disadvantage of limited cargo size. The major administra-
the longer it took for the HIV escape variants to appear.76 In tion approaches of rAAV include stereotactic injection, intranasal
another research group, Wang et al.77 found that: CRISPR/Cas9 and intratracheal administration, intravenous injection, intraper-
could derive mutant HIV which could resist Cas9 and sgRNA itoneal Injection and intramuscular Injection. In the non-viral

© 2017 Macmillan Publishers Limited, part of Springer Nature. Gene Therapy (2017) 377 – 384
Application of CRISPR/Cas9 gene-editing technique to eradication of HIV
Z Huang et al
382

Figure 4. Brain targeting delivery by magnetic or ligand-labeled nanoparticles. Drug and reagents loaded nanoparticles are systematically
administered and target the CNS cells in human brain. In magnetic targeting (a), nanoparticles complexes target brain cells and cross the BBB
under the force of magnetic field. In Receptor-mediated transcytosis (b), nanoparticle complexes target brain cells and cross BBB by brain-
specific receptor-mediated transcytosis. After crossing the BBB, drugs are released to conduct the therapeutic procedure.

delivery approach, vectors such as cationic polymer polyethyle- polymer nanoparticles,95 gold nanoparticles91 and iron oxide
neimine (PEI),82 liposomes,58,80 lipid nanoparticles,83,84 virus-like nanoparticles.94,96
particles derived from bacteriophage,85 and self-assembled DNA Magnetic targeting is another approach to improve BBB
nanoparticles86 have been applied for the delivery of the CRISPR/ transmigration of nanoparticles (Figure 4). Nair et al.92,97–100 have
Cas9 system. Since viral and non-viral approaches have relative made important progress in brain delivery using magnetic
advantages and disadvantages, some investigators started trying nanoparticles and magneto-electric nanoparticles. Magneto-
the combined approach. For example, lipid nanoparticle-mediated electric nanoparticles have been used as carrier molecules, which
delivery of Cas9 mRNA has been combined with adeno-associated could cross the BBB and mediate controlled release of the loaded
viruses encoding a sgRNA and a repair template.87 This delivery drugs. These nanoparticles were able to cross the BBB using
strategy successfully repaired a disease gene in an adult mouse
magnetic force induced on the nanoparticles by the magnetic
model.87
field gradient.97,98 To date, these nanoparticles have not been
No matter which delivery strategy is being used, the emphases
are biocompatible and targetable carriers, controllable reagents applied to the delivery of the CRISPR/Cas9 system nor have they
release, and low-invasive administration. As for HIV/AIDS eradica- been fully tested in animal models. However, the potential for
tion, in order to establish more efficient evaluation methods, HIV/ these nanoparticles to deliver CRISPR/Cas9 system and target HIV
AIDS primate models will be the center of the next phase of study. reservoir in brain seems high due to the nanoparticles’ capacities
Drug brain delivery is the key for the eradication of the HIV of specific targeting, crossing the BBB, constant or controlled
reservoir in patients’ CNS systems. The obstacle for drug brain release and non-invasive application.
delivery of the CRISPR/Cas9 system is the existence of the blood–
brain barrier (BBB). The tight junction between capillary endothe-
lial cells block transportation of large molecules and only limited CONCLUSION AND FUTURE DIRECTIONS
molecules with molecular weight smaller than 400 Da and
For the future of applying CRISPR/Cas9 technique to eradicate HIV
appropriate lipophilicity can cross. All large molecules and most
of the small molecules cannot cross the BBB. Great efforts have reservoirs, safety, efficiency and specificity will be the main
been made to improve BBB permeability for therapeutic agents emphases. Research efforts using the CRISPR/Cas9 technique will
for the treatment of various brain diseases and HAND. However, now be focused on primate models and phase I clinical trials. From
currently available procedures are mainly invasive, including the point of this review, the CRISPR/Cas9 technique holds great
intracerebroventricular (ICV) infusion, intra-cerebral injection. promise for the eradication of HIV/AIDS. Since the FDA has already
Although these approaches have shown successful disruption of approved use of CRISPR/Cas9 technique in several clinical trials, it
the BBB, the invasiveness of the technique and the risk of damage is surely not long before the CRISPR/Cas9 system can be applied
to the brain remain significant concerns. To overcome these for clinical therapeutics and prevention on HIV/AIDS.
problems, the use of nanomaterials as drug delivery carriers to the
brain has been gaining appreciation.
The most popular approach for brain targeting using nanoma-
CONFLICT OF INTEREST
terials is receptor-mediated transcytosis (Figure 4). The endothelial
The authors declare no conflict of interest.
cells that form the BBB are known to express receptors, including
the transferrin receptor and insulin receptor.88–90 Ligand functio-
nalized nanoparticles can promote efficient brain delivery through
the binding of ligands to the receptors on endothelial cells. ACKNOWLEDGEMENTS
Various nanoparticles have been developed for brain targeting This work was supported by grants (R01DA037838 and R01DA034547) from National
using this receptor-mediated transcytosis such as transferrin,91,92 Institutes of Health, US. We also thank Maureen Pelham and Courtney Myhr for their
lactoferrin93,94 or monoclonal antibody-conjugated liposomes,93 help in English editing.

Gene Therapy (2017) 377 – 384 © 2017 Macmillan Publishers Limited, part of Springer Nature.
Application of CRISPR/Cas9 gene-editing technique to eradication of HIV
Z Huang et al
383
REFERENCES 23 Ellis RJ, Rosario D, Clifford DB, McArthur JC, Simpson D, Alexander T et al.
1 Chun TW, Carruth L, Finzi D, Shen X, DiGiuseppe JA, Taylor H et al. Quantification Continued high prevalence and adverse clinical impact of human immunode-
of latent tissue reservoirs and total body viral load in HIV-1 infection. Nature ficiency virus-associated sensory neuropathy in the era of combination anti-
1997; 387: 183–188. retroviral therapy: the CHARTER Study. Arch Neurol 2010; 67: 552–558.
2 Chun TW, Engel D, Mizell SB, Ehler LA, Fauci AS. Induction of HIV-1 replication in 24 Churchill MJ, Gorry PR, Cowley D, Lal L, Sonza S, Purcell DF et al. Use of laser
latently infected CD4+ T cells using a combination of cytokines. J Exp Med 1998; capture microdissection to detect integrated HIV-1 DNA in macrophages and
188: 83–91. astrocytes from autopsy brain tissues. J Neurovirol 2006; 12: 146–152.
3 Finzi D, Hermankova M, Pierson T, Carruth LM, Buck C, Chaisson RE et al. Iden- 25 Narasipura SD, Kim S, Al-Harthi L. Epigenetic regulation of HIV-1 latency in
tification of a reservoir for HIV-1 in patients on highly active antiretroviral astrocytes. J Virol 2014; 88: 3031–3038.
therapy. Science 1997; 278: 1295–1300. 26 Nath A. Eradication of human immunodeficiency virus from brain reservoirs.
4 Siliciano JD, Kajdas J, Finzi D, Quinn TC, Chadwick K, Margolick JB et al. Long- J Neurovirol 2015; 21: 227–234.
term follow-up studies confirm the stability of the latent reservoir for HIV-1 in 27 Dampier W, Nonnemacher MR, Sullivan NT, Jacobson JM, Wigdahl B. HIV excision
resting CD4+ T cells. Nature Medicine 2003; 9: 727–728. utilizing crispr/cas9 technology: attacking the proviral quasispecies in reservoirs
5 Bagasra O, Lavi E, Bobroski L, Khalili K, Pestaner JP, Tawadros R et al. Cellular to achieve a cure. MOJ Immunol 2014; 1: pii:00022.
reservoirs of HIV-1 in the central nervous system of infected individuals: iden- 28 Ebina H, Misawa N, Kanemura Y, Koyanagi Y. Harnessing the CRISPR/Cas9 system
tification by the combination of in situ polymerase chain reaction and immu- to disrupt latent HIV-1 provirus. Sci Rep 2013; 3: 2510.
nohistochemistry. AIDS 1996; 10: 573–585. 29 Hu W, Kaminski R, Yang F, Zhang Y, Cosentino L, Li F et al. RNA-directed gene
6 Fischer-Smith T, Croul S, Sverstiuk AE, Capini C, L'Heureux D, Regulier EG et al. editing specifically eradicates latent and prevents new HIV-1 infection. Proc Natl
CNS invasion by CD14+/CD16+ peripheral blood-derived monocytes in HIV Acad Sci USA 2014; 111: 11461–11466.
dementia: perivascular accumulation and reservoir of HIV infection. J Neurovirol 30 Kaminski R, Chen Y, Fischer T, Tedaldi E, Napoli A, Zhang Y et al. Elimination of
2001; 7: 528–541. HIV-1 genomes from human T-lymphoid cells by CRISPR/Cas9 gene editing. Sci
7 Petito CK, Chen H, Mastri AR, Torres-Munoz J, Roberts B, Wood C. HIV infection of Rep 2016; 6: 22555.
choroid plexus in AIDS and asymptomatic HIV-infected patients suggests that 31 Saayman SM, Lazar DC, Scott TA, Hart JR, Takahashi M, Burnett JC et al. Potent
the choroid plexus may be a reservoir of productive infection. J Neurovirol 1999; and targeted activation of latent HIV-1 using the CRISPR/dCas9 activator com-
5: 670–677. plex. Mol Ther 2016; 24: 488–498.
8 McElrath MJ, Steinman RM, Cohn ZA. Latent HIV-1 infection in enriched popu- 32 Zhang Y, Yin C, Zhang T, Li F, Yang W, Kaminski R et al. CRISPR/gRNA-directed
lations of blood monocytes and T cells from seropositive patients. J Clin Invest synergistic activation mediator (SAM) induces specific, persistent and robust
1991; 87: 27–30. reactivation of the HIV-1 latent reservoirs. Sci Rep 2015; 5: 16277.
9 Chun TW, Engel D, Berrey MM, Shea T, Corey L, Fauci AS. Early establishment of a 33 Chylinski K, Le Rhun A, Charpentier E. The tracrRNA and Cas9 families of type II
pool of latently infected, resting CD4(+) T cells during primary HIV-1 infection. CRISPR-Cas immunity systems. RNA Biol 2013; 10: 726–737.
Proc Natl Acad Sci USA 1998; 95: 8869–8873. 34 Jinek M, Chylinski K, Fonfara I, Hauer M, Doudna JA, Charpentier E. A pro-
10 Chun TW, Nickle DC, Justement JS, Meyers JH, Roby G, Hallahan CW et al. Per- grammable dual-RNA-guided DNA endonuclease in adaptive bacterial immunity.
sistence of HIV in gut-associated lymphoid tissue despite long-term antiretroviral Science 2012; 337: 816–821.
therapy. J Infect Dis 2008; 197: 714–720. 35 Chavez L, Calvanese V, Verdin E. HIV latency is established directly and early in
11 Smith PD, Meng G, Salazar-Gonzalez JF, Shaw GM. Macrophage HIV-1 infection both resting and activated primary CD4 T cells. PLoS Pathog 2015; 11: e1004955.
and the gastrointestinal tract reservoir. J Leuko Biol 2003; 74: 642–649. 36 Stanley SK, Ostrowski MA, Justement JS, Gantt K, Hedayati S, Mannix M et al.
12 Lambert-Niclot S, Peytavin G, Duvivier C, Poirot C, Algarte-Genin M, Effect of immunization with a common recall antigen on viral expression in
Pakianather S et al. Low frequency of intermittent HIV-1 semen excretion in patients infected with human immunodeficiency virus type 1. N Engl J Med 1996;
patients treated with darunavir-ritonavir at 600/100 milligrams twice a day plus 334: 1222–1230.
two nucleoside reverse transcriptase inhibitors or monotherapy. Antimicrob 37 Brady T, Agosto LM, Malani N, Berry CC, O'Doherty U, Bushman F. HIV integration
Agents Chemother 2010; 54: 4910–4913. site distributions in resting and activated CD4+ T cells infected in culture. AIDS
13 Cu-Uvin S, DeLong AK, Venkatesh KK, Hogan JW, Ingersoll J, Kurpewski J et al. 2009; 23: 1461–1471.
Genital tract HIV-1 RNA shedding among women with below detectable plasma 38 Schneider M, Tigges B, Meggendorfer M, Helfer M, Ziegenhain C, Brack-Werner R.
viral load. AIDS 2010; 24: 2489–2497. A new model for post-integration latency in macroglial cells to study HIV-1
14 Chomont N, El-Far M, Ancuta P, Trautmann L, Procopio FA, Yassine-Diab B et al. reservoirs of the brain. AIDS 2015; 29: 1147–1159.
HIV reservoir size and persistence are driven by T cell survival and homeostatic 39 Sunshine S, Kirchner R, Amr SS, Mansur L, Shakhbatyan R, Kim M et al. HIV
proliferation. Nat Med 2009; 15: 893–900. Integration site analysis of cellular models of HIV latency with a probe-enriched
15 Chun TW, Nickle DC, Justement JS, Large D, Semerjian A, Curlin ME et al. next-generation sequencing assay. J Virol 2016; 90: 4511–4519.
HIV-infected individuals receiving effective antiviral therapy for extended 40 du Chene I, Basyuk E, Lin YL, Triboulet R, Knezevich A, Chable-Bessia C et al.
periods of time continually replenish their viral reservoir. J Clin Invest 2005; 115: Suv39H1 and HP1gamma are responsible for chromatin-mediated HIV-1 tran-
3250–3255. scriptional silencing and post-integration latency. EMBO J 2007; 26: 424–435.
16 Fletcher CV, Staskus K, Wietgrefe SW, Rothenberger M, Reilly C, Chipman JG et al. 41 Gallastegui E, Millan-Zambrano G, Terme JM, Chavez S, Jordan A. Chromatin
Persistent HIV-1 replication is associated with lower antiretroviral drug con- reassembly factors are involved in transcriptional interference promoting HIV
centrations in lymphatic tissues. Proc Natl Acad Sci USA 2014; 111: 2307–2312. latency. J Virol 2011; 85: 3187–3202.
17 Letendre S, Marquie-Beck J, Capparelli E, Best B, Clifford D, Collier AC et al. 42 Barboric M, Nissen RM, Kanazawa S, Jabrane-Ferrat N, Peterlin BM. NF-kappaB
Validation of the CNS Penetration-Effectiveness rank for quantifying anti- binds P-TEFb to stimulate transcriptional elongation by RNA polymerase II. Mol
retroviral penetration into the central nervous system. Arch Neurol 2008; 65: Cell 2001; 8: 327–337.
65–70. 43 Lenasi T, Contreras X, Peterlin BM. Transcriptional interference antagonizes
18 Solas C, Lafeuillade A, Halfon P, Chadapaud S, Hittinger G, Lacarelle B. proviral gene expression to promote HIV latency. Cell Host Microbe 2008; 4:
Discrepancies between protease inhibitor concentrations and viral load in 123–133.
reservoirs and sanctuary sites in human immunodeficiency virus-infected 44 Huang J, Wang F, Argyris E, Chen K, Liang Z, Tian H et al. Cellular microRNAs
patients. Antimicrob Agents Chemother 2003; 47: 238–243. contribute to HIV-1 latency in resting primary CD4+ T lymphocytes. Nat Med
19 Heaton RK, Clifford DB, Franklin Jr DR, Woods SP, Ake C, Vaida F et al. HIV- 2007; 13: 1241–1247.
associated neurocognitive disorders persist in the era of potent antiretroviral 45 Patel P, Ansari MY, Bapat S, Thakar M, Gangakhedkar R, Jameel S. The microRNA
therapy: CHARTER Study. Neurology 2010; 75: 2087–2096. miR-29a is associated with human immunodeficiency virus latency. Retrovirology
20 Heaton RK, Franklin Jr DR, Deutsch R, Letendre S, Ellis RJ, Casaletto K et al. 2014; 11: 108.
Neurocognitive change in the era of HIV combination antiretroviral therapy: the 46 Ruelas DS, Chan JK, Oh E, Heidersbach AJ, Hebbeler AM, Chavez L et al.
longitudinal CHARTER study. Clin Infect Dis 2015; 60: 473–480. MicroRNA-155 Reinforces HIV Latency. J Biol Chem 2015; 290: 13736–13748.
21 Antinori A, Arendt G, Becker JT, Brew BJ, Byrd DA, Cherner M et al. Updated 47 Delagreverie HM, Delaugerre C, Lewin SR, Deeks SG, Li JZ. Ongoing clinical trials
research nosology for HIV-associated neurocognitive disorders. Neurology 2007; of human immunodeficiency virus latency-reversing and immunomodulatory
69: 1789–1799. agents. open forum. Infect Dis 2016; 3: ofw189.
22 Churchill MJ, Wesselingh SL, Cowley D, Pardo CA, McArthur JC, Brew BJ et al. 48 Archin NM, Liberty AL, Kashuba AD, Choudhary SK, Kuruc JD, Crooks AM et al.
Extensive astrocyte infection is prominent in human immunodeficiency virus- Administration of vorinostat disrupts HIV-1 latency in patients on antiretroviral
associated dementia. Ann Neurol 2009; 66: 253–258. therapy. Nature 2012; 487: 482–485.

© 2017 Macmillan Publishers Limited, part of Springer Nature. Gene Therapy (2017) 377 – 384
Application of CRISPR/Cas9 gene-editing technique to eradication of HIV
Z Huang et al
384
49 Rasmussen TA, Tolstrup M, Brinkmann CR, Olesen R, Erikstrup C, Solomon A et al. 77 Wang Z, Pan Q, Gendron P, Zhu W, Guo F, Cen S et al. CRISPR/Cas9-derived
Panobinostat, a histone deacetylase inhibitor, for latent-virus reactivation in mutations both inhibit HIV-1 replication and accelerate viral escape. Cell Rep
HIV-infected patients on suppressive antiretroviral therapy: a phase 1/2, single 2016; 15: 481–489.
group, clinical trial. Lancet HIV 2014; 1: e13–e21. 78 Yoder KE, Bundschuh R. Host double strand break repair generates hiv-1 strains
50 Porteus MH, Baltimore D. Chimeric nucleases stimulate gene targeting in resistant to CRISPR/Cas9. Sci Rep 2016; 6: 29530.
human cells. Science 2003; 300: 763. 79 Heckl D, Kowalczyk MS, Yudovich D, Belizaire R, Puram RV, McConkey ME et al.
51 Miller JC, Holmes MC, Wang J, Guschin DY, Lee YL, Rupniewski I et al. An Generation of mouse models of myeloid malignancy with combinatorial genetic
improved zinc-finger nuclease architecture for highly specific genome editing. lesions using CRISPR-Cas9 genome editing. Nat Biotechnol 2014; 32: 941–946.
Nat Biotechnol 2007; 25: 778–785. 80 Platt RJ, Chen S, Zhou Y, Yim MJ, Swiech L, Kempton HR et al. CRISPR-Cas9
52 Wood AJ, Lo TW, Zeitler B, Pickle CS, Ralston EJ, Lee AH et al. Targeted genome knockin mice for genome editing and cancer modeling. Cell 2014; 159: 440–455.
editing across species using ZFNs and TALENs. Science 2011; 333: 307. 81 Mansouri M, Bellon-Echeverria I, Rizk A, Ehsaei Z, Cianciolo Cosentino C, Silva CS
53 Miller JC, Tan S, Qiao G, Barlow KA, Wang J, Xia DF et al. A TALE nuclease et al. Highly efficient baculovirus-mediated multigene delivery in primary cells.
architecture for efficient genome editing. Nat Biotechnol 2011; 29: 143–148. Nat Commun 2016; 7: 11529.
54 Zhang F, Cong L, Lodato S, Kosuri S, Church GM, Arlotta P. Efficient construction 82 Zuckermann M, Hovestadt V, Knobbe-Thomsen CB, Zapatka M, Northcott PA,
of sequence-specific TAL effectors for modulating mammalian transcription. Nat Schramm K et al. Somatic CRISPR/Cas9-mediated tumour suppressor disruption
Biotechnol 2011; 29: 149–153. enables versatile brain tumour modelling. Nat Commun 2015; 6: 7391.
55 Reyon D, Tsai SQ, Khayter C, Foden JA, Sander JD, Joung JK. FLASH assembly of 83 Wang L, Hao Y, Li H, Zhao Y, Meng D, Li D et al. Co-delivery of doxorubicin and
TALENs for high-throughput genome editing. Nat Biotechnol 2012; 30: 460–465. siRNA for glioma therapy by a brain targeting system: angiopep-2-modified poly
56 Barrangou R, Fremaux C, Deveau H, Richards M, Boyaval P, Moineau S et al. (lactic-co-glycolic acid) nanoparticles. J Drug Target 2015; 23: 832–846.
CRISPR provides acquired resistance against viruses in prokaryotes. Science 2007; 84 Wang M, Zuris JA, Meng F, Rees H, Sun S, Deng P et al. Efficient delivery of
315: 1709–1712. genome-editing proteins using bioreducible lipid nanoparticles. Proc Natl Acad
57 Cong L, Ran FA, Cox D, Lin S, Barretto R, Habib N et al. Multiplex genome Sci USA 2016; 113: 2868–2873.
engineering using CRISPR/Cas systems. Science 2013; 339: 819–823. 85 Qazi S, Miettinen HM, Wilkinson RA, McCoy K, Douglas T, Wiedenheft B.
58 Mali P, Yang L, Esvelt KM, Aach J, Guell M, DiCarlo JE et al. RNA-guided human Programmed self-assembly of an active P22-Cas9 nanocarrier system. Mol Pharm
genome engineering via Cas9. Science 2013; 339: 823–826. 2016; 13: 1191–1196.
59 Hsu PD, Lander ES, Zhang F. Development and applications of CRISPR-Cas9 for 86 Sun W, Ji W, Hall JM, Hu Q, Wang C, Beisel CL et al. Self-assembled DNA
genome engineering. Cell 2014; 157: 1262–1278. nanoclews for the efficient delivery of CRISPR-Cas9 for genome editing. Angew
60 Shi B, Li J, Shi X, Jia W, Wen Y, Hu X et al. TALEN-mediated knockout of CCR5 Chem Int Ed Engl 2015; 54: 12029–12033.
confers protection against infection of human immunodeficiency virus. J Acquir 87 Yin H, Song CQ, Dorkin JR, Zhu LJ, Li Y, Wu Q et al. Therapeutic genome editing
by combined viral and non-viral delivery of CRISPR system components in vivo.
Immune Defic Syndr 2016.
Nat Biotechnol 2016; 34: 328–333.
61 Tebas P, Stein D, Tang WW, Frank I, Wang SQ, Lee G et al. Gene editing of CCR5
88 Bickel U, Kang YS, Yoshikawa T, Pardridge WM. In vivo demonstration of
in autologous CD4 T cells of persons infected with HIV. N Engl J Med 2014; 370:
subcellular localization of anti-transferrin receptor monoclonal antibody-
901–910.
colloidal gold conjugate in brain capillary endothelium. J Histochem Cytochem
62 Maier DA, Brennan AL, Jiang S, Binder-Scholl GK, Lee G, Plesa G et al. Efficient
1994; 42: 1493–1497.
clinical scale gene modification via zinc finger nuclease-targeted disruption of
89 Yoshikawa T, Pardridge WM. Biotin delivery to brain with a covalent conjugate of
the HIV co-receptor CCR5. Hum Gene Ther 2013; 24: 245–258.
avidin and a monoclonal antibody to the transferrin receptor. J Pharmacol Exp
63 Strong CL, Guerra HP, Mathew KR, Roy N, Simpson LR, Schiller MR. Damaging the
Ther 1992; 263: 897–903.
Integrated HIV Proviral DNA with TALENs. PloS One 2015; 10: e0125652.
90 Zhang Y, Boado RJ, Pardridge WM. In vivo knockdown of gene expression in
64 Anders C, Niewoehner O, Duerst A, Jinek M. Structural basis of PAM-dependent
brain cancer with intravenous RNAi in adult rats. J Gene Med 2003; 5: 1039–1045.
target DNA recognition by the Cas9 endonuclease. Nature 2014; 513: 569–573.
91 Clark AJ, Davis ME. Increased brain uptake of targeted nanoparticles by adding
65 Jinek M, Jiang F, Taylor DW, Sternberg SH, Kaya E, Ma E et al. Structures of Cas9
an acid-cleavable linkage between transferrin and the nanoparticle core. Proc
endonucleases reveal RNA-mediated conformational activation. Science 2014;
Natl Acad Sci USA 2015; 112: 12486–12491.
343: 1247997.
92 Ding H, Sagar V, Agudelo M, Pilakka-Kanthikeel S, Atluri VS, Raymond A et al.
66 Nishimasu H, Ran FA, Hsu PD, Konermann S, Shehata SI, Dohmae N et al. Crystal
Enhanced blood-brain barrier transmigration using a novel transferrin embed-
structure of Cas9 in complex with guide RNA and target DNA. Cell 2014; 156:
ded fluorescent magneto-liposome nanoformulation. Nanotechnology 2014; 25:
935–949. 055101.
67 Ran FA, Hsu PD, Wright J, Agarwala V, Scott DA, Zhang F. Genome engineering 93 De Luca MA, Lai F, Corrias F, Caboni P, Bimpisidis Z, Maccioni E et al. Lactoferrin-
using the CRISPR-Cas9 system. Nat Protoc 2013; 8: 2281–2308. and antitransferrin-modified liposomes for brain targeting of the NK3 receptor
68 Konermann S, Brigham MD, Trevino AE, Joung J, Abudayyeh OO, Barcena C et al. agonist senktide: preparation and in vivo evaluation. Int J Pharm 2015; 479:
Genome-scale transcriptional activation by an engineered CRISPR-Cas9 complex. 129–137.
Nature 2015; 517: 583–588. 94 Tomitaka A, Arami H, Gandhi S, Krishnan KM. Lactoferrin conjugated iron oxide
69 Hou P, Chen S, Wang S, Yu X, Chen Y, Jiang M et al. Genome editing of CXCR4 by nanoparticles for targeting brain glioma cells in magnetic particle imaging.
CRISPR/cas9 confers cells resistant to HIV-1 infection. Sci Rep 2015; 5: 15577. Nanoscale 2015; 7: 16890–16898.
70 Schumann K, Lin S, Boyer E, Simeonov DR, Subramaniam M, Gate RE et al. 95 Fornaguera C, Dols-Perez A, Caldero G, Garcia-Celma MJ, Camarasa J, Solans C.
Generation of knock-in primary human T cells using Cas9 ribonucleoproteins. PLGA nanoparticles prepared by nano-emulsion templating using low-energy
Proc Natl Acad Sci USA 2015; 112: 10437–10442. methods as efficient nanocarriers for drug delivery across the blood-brain bar-
71 Ye L, Wang J, Beyer AI, Teque F, Cradick TJ, Qi Z et al. Seamless modification rier. J Control Release 2015; 211: 134–143.
of wild-type induced pluripotent stem cells to the natural CCR5Delta32 96 Huang Y, Zhang B, Xie S, Yang B, Xu Q, Tan J et al. Modified with Tween 80 pass
mutation confers resistance to HIV infection. Proc Natl Acad Sci USA 2014; 111: through the intact blood-brain barrier in rats under magnetic field. ACS Appl
9591–9596. Mater Interfaces 2016; 8: 11336–11341.
72 Wang W, Ye C, Liu J, Zhang D, Kimata JT, Zhou P. CCR5 gene disruption via 97 Kaushik A, Jayant RD, Nikkhah-Moshaie R, Bhardwaj V, Roy U, Huang Z et al.
lentiviral vectors expressing Cas9 and single guided RNA renders cells resistant Magnetically guided central nervous system delivery and toxicity evaluation of
to HIV-1 infection. PloS One 2014; 9: e115987. magneto-electric nanocarriers. Sci Rep 2016; 6: 25309.
73 Li C, Guan X, Du T, Jin W, Wu B, Liu Y et al. Inhibition of HIV-1 infection of primary 98 Nair M, Guduru R, Liang P, Hong J, Sagar V, Khizroev S. Externally controlled
CD4+ T-cells by gene editing of CCR5 using adenovirus-delivered CRISPR/Cas9. on-demand release of anti-HIV drug using magneto-electric nanoparticles as
J Gen Virol 2015; 96: 2381–2393. carriers. Nat Commun 2013; 4: 1707.
74 Zhu W, Lei R, Le Duff Y, Li J, Guo F, Wainberg MA et al. The CRISPR/Cas9 system 99 Sagar V, Pilakka-Kanthikeel S, Atluri VS, Ding H, Arias AY, Jayant RD et al.
inactivates latent HIV-1 proviral DNA. Retrovirology 2015; 12: 22. Therapeutical neurotargeting via magnetic nanocarrier: implications to opiate-
75 Kaminski R, Bella R, Yin C, Otte J, Ferrante P, Gendelman HE et al. Excision of induced neuropathogenesis and neuroAIDS. J Biomed Nanotechnol 2015; 11:
HIV-1 DNA by gene editing: a proof-of-concept in vivo study. Gene Ther 2016; 23: 1722–1733.
696. 100 Jayant RD, Atluri VS, Agudelo M, Sagar V, Kaushik A, Nair M. Sustained-release
76 Wang G, Zhao N, Berkhout B, Das AT. CRISPR-Cas9 can inhibit HIV-1 replication nanoART formulation for the treatment of neuroAIDS. Int J Nanomed 2015; 10:
but NHEJ repair facilitates virus escape. Mol Ther 2016; 24: 522–526. 1077–1093.

Gene Therapy (2017) 377 – 384 © 2017 Macmillan Publishers Limited, part of Springer Nature.

You might also like