ArticlePDF AvailableLiterature Review

Abstract and Figures

Muscular dystrophies (MDs) comprise a diverse group of inherited disorders characterized by progressive muscle loss and weakness. Given the genetic etiology underlying MDs, researchers have explored the potential of CRISPR/Cas9 genome editing as a therapeutic intervention, resulting in significant advances. Here we review recent progress on the use of CRISPR/Cas9 genome editing as a potential therapy for MDs. Significant strides have been made in this realm, made possible through innovative techniques such as precision genetic editing by modified forms of CRISPR/Cas9. These approaches have shown varying degrees of success in animal models of MD, including Duchenne muscular dystrophy (DMD), congenital muscular dystrophy type 1A (MDC1A), and myotonic dystrophy type 1 (DM1). Even so, there are several challenges facing the development of CRISPR/Cas9-based MD therapies, including the targeting of satellite cells, improved editing efficiency in skeletal and cardiac muscle tissue, delivery vehicle enhancements, and the host immunogenic response. While more work is needed to advance CRISPR/Cas9 genome editing past the preclinical stages, its therapeutic potential for MD is extremely promising and justifies concentrated efforts to move into clinical trials.
Content may be subject to copyright.
Open camera or QR reader and
scan code to access this article
and other resources online.
Advances in CRISPR/Cas9 Genome Editing for the Treatment
of Muscular Dystrophies
Sina Fatehi,
1,2
Ryan M. Marks,
1,2
Matthew J. Rok,
1,2
Lucie Perillat,
1,2
Evgueni A. Ivakine,
1,3,{
and Ronald D. Cohn
1,2,4,5,
*
,{
1
Program in Genetics and Genome Biology, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children Research Institute, Toronto, Ontario,
Canada; Departments of
2
Molecular Genetics and
3
Physiology, University of Toronto, Toronto, Ontario, Canada;
4
Department of Pediatrics, The Hospital for Sick
Children, Toronto, Ontario, Canada; and
5
Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada.
{
Co-senior authors.
Muscular dystrophies (MDs) comprise a diverse group of inherited disorders characterized by progressive muscle loss
and weakness. Given the genetic etiology underlying MDs, researchers have explored the potential of clustered regularly
interspaced short palindromic repeats (CRISPR)/CRISPR-associated protein 9 (Cas9) genome editing as a therapeutic
intervention, resulting in significant advances. Here, we review recent progress on the use of CRISPR/Cas9 genome
editing as a potential therapy for MDs. Significant strides have been made in this realm, made possible through
innovative techniques such as precision genetic editing by modified forms of CRISPR/Cas9. These approaches have
shown varying degrees of success in animal models of MD, including Duchenne MD, congenital muscular dystrophy
type 1A, and myotonic dystrophy type 1. Even so, there are several challenges facing the development of CRISPR/Cas9-
based MD therapies, including the targeting of satellite cells, improved editing efficiency in skeletal and cardiac muscle
tissue, delivery vehicle enhancements, and the host immunogenic response. Although more work is needed to advance
CRISPR/Cas9 genome editing past the preclinical stages, its therapeutic potential for MD is extremely promising and
justifies concentrated efforts to move into clinical trials.
Keywords: muscular dystrophy, Duchenne muscular dystrophy, genome editing, CRISPR/Cas9, adeno-associated
viruses
INTRODUCTION
Muscular dystrophy
MUSCULAR DYSTROPHIES (MDs) are a collection of clini-
cally and genetically heterogeneous disorders character-
ized by progressive muscle weakness and loss.
1,2
The
disorders vary according to the affected muscles, age of
onset, severity, and rate of progression. Currently, most
medical interventions are limited to symptom manage-
ment and delaying disease progression.
1,3
Here, we provide a comprehensive overview of the
current state of genome editing strategies using clustered
regularly interspaced short palindromic repeats (CRISPR)
and CRISPR-associated protein 9 (Cas9) in models of MD.
This work focuses on the utilization of CRISPR/Cas9 as an
in vivo therapeutic strategy to treat Duchenne muscular
dystrophy (DMD), congenital muscular dystrophy type 1A
(MDC1A), and myotonic dystrophy type 1 (DM1). In
addition, we discuss current in vivo delivery options, fo-
cusing on adeno-associated viral (AAV) vectors, recent
advances made, challenges that lie ahead for the field, and
what issues must be resolved for CRISPR/Cas9 genome
editing to become a viable therapy.
DMD is the most prevalent pediatric neuromuscular
disorder due to its X-linked recessive inheritance.
4
It is a
*Correspondence: Dr. Ronald D. Cohn, Program in Genetics and Genome Biology, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children Research
Institute, 686 Bay Street, Toronto, ON M5G 0A4, Canada. E-mail: ronald.cohn@sickkids.ca
388 jHUMAN GENE THERAPY, VOLUME 34, NUMBERS 9 and 10 DOI: 10.1089/hum.2023.059
ª2023 by Mary Ann Liebert, Inc.
Downloaded by McMaster University from www.liebertpub.com at 05/24/23. For personal use only.
life-limiting condition caused by mutations in the DMD
gene, which encodes for dystrophin.
5,6
Dystrophin is a
subsarcolemmal protein integral to the dystrophin-
associated protein complex, which prevents contraction-
induced muscle damage.
7
Most commonly, mutations that
cause DMD generate out-of-frame transcripts, eliminating
dystrophin expression.
8,9
MDC1A is an autosomal recessive neuromuscular
disorder characterized by hypotonia, muscle weakness,
and muscle wasting that begins in infancy.
10
MDC1A is
caused by mutations in the LAMA2 gene, which encodes
laminin-2.
11
Laminins are proteins of the extracellular
matrix that are essential components of the basement
membrane.
11,12
DM1 is the most prevalent adult-onset form of MD.
13
Patients may also develop intellectual impairment, respi-
ratory insufficiency, and cardiac conduction abnormali-
ties, in addition to muscle weakness and stiffness.
14,15
DM1 is caused by a microsatellite expansion of cytosine,
thymine, and guanine (CTG) triplet repeats in the 3¢un-
translated region (UTR) of the dystrophia myotonic pro-
tein kinase (DMPK) gene.
13,15
As a result, these mutated
DMPK transcripts generate nuclear foci, which cause ab-
errant splicing across a broad spectrum of pre-mRNAs.
14
Various treatments for MDs are currently approved or
are in clinical testing. Included in this category are anti-
sense oligonucleotides (ASOs), gene therapies, and stop
codon read-through drugs.
1
Although effective in treating
other neuromuscular disorders such as spinal muscular
atrophy (SMA), unfortunately, with regard to MDs, their
relatively poor performance and transient nature limit their
clinical applications.
1
An effective treatment must target
and, ideally, permanently repair the genetic cause of MDs.
Interventions based on CRISPR/Cas9 are extremely
promising because of their unrivalled utility and accuracy
in performing targeted genome editing.
CRISPR/CAS9 GENOME EDITING STRATEGIES
TO TREAT MDS
Exon skipping and reframing
Exon skipping using CRISPR/Cas9 endonucleases can
restore the open reading frame (ORF) of an out-of-frame
gene and lead to the restoration of a functional protein.
Exon skipping has been achieved in DMD patients with the
use of ASOs, which omit the out-of-frame exon(s) from the
final transcript.
16
Eteplirsen, Golodirsen, and Viltolarsen
are examples of such drugs that use ASOs to target out-of-
frame exons in the DMD gene, excluding them from the
final transcript and restoring the reading frame.
17–21
Un-
fortunately, ASOs typically result in low uptake in cardiac
tissue, limiting their clinical relevance regarding the treat-
ment of MDs with a cardiac pathology.
20,21
As a result, CRISPR/Cas9 is currently under investi-
gation as an alternative to mediating exon skipping and
reframing. CRISPR/Cas9-mediated exon-skipping would
be a one-time therapeutic option that results in permanent
correction and restoration of a functional gene product (a
summary of such studies is listed in Table 1). Three main
exon-skipping and reframing approaches (Fig. 1A) have
been tested in preclinical studies and are described next.
Exon removal. This approach aims at restoring the
ORF of a gene by excising the out-of-frame exon(s), re-
sulting in the expression of a truncated protein. This
strategy has been assessed pre-clinically in several DMD
models, including mice, pigs, and dogs.
22–25
Studies in
mice focused on several exons of the Dmd gene, including
exons 23 and 52–53.
23,25
These studies show restoration of
a functional dystrophin protein and improved disease
phenotypes. Studies in pigs and dogs provided proof-of-
principle that CRISPR/Cas9-mediated exon removal is a
viable therapeutic option in larger animal models, paving
the way for its translation to the clinical trials.
22,24
Exon skipping and inclusion through splice site
manipulation. The next strategy designed to mediate
exon skipping relies on manipulation of splice sites using
CRISPR/Cas9 systems. Here, a single-guide RNA
(sgRNA) is used to disrupt splice sites through the error-
prone non-homologous end-joining (NHEJ) repair path-
way, which subsequently allows for the skipping of the
desired exon(s).
26
The NHEJ-mediated introduction of
random deletions and insertions (indels) renders the splice
site no longer identifiable by the spliceosome.
27,28
The disruption results in the exclusion of the exon from
the final transcript, restoring the ORF and expressing a
truncated gene product. This strategy has been tested pre-
clinically in mouse and canine DMD models and resulted
in restoration of a functional dystrophin protein in skeletal
(including the diaphragm) and cardiac tissues.
22,26
In an MDC1A mouse model, Kemaladewi et al. used a
pair of sgRNAs to remove a mutated splice site in the
LAMA2 gene and allowed NHEJ to restore a functional
splice donor site. As a result, full-length laminin-2 was
restored in muscles and the sciatic nerve. Treated mice
exhibited reduced fibrosis, increased muscle fiber size, and
functional improvements comparable to wild-type mice.
29
Indel-mediated reframing. An alternative refram-
ing approach consists of restoring the ORF by introducing
small indels in the coding sequence of an out-of-frame
exon. The Olson group has tested this strategy by using
the engineered KKH variant of SaCas9 in patient-derived
induced pluripotent stem cells (iPSCs) to induce a single
cut that results in the introduction of indels that are able
to reframe exon 51 of the DMD gene.
30
This approach
resulted in a high frequency of indels introducing a two-
nucleotide deletion and, thus, the restoration of the
reading frame.
CRISPR/CAS9 TREATMENT OF MUSCULAR DYSTROPHIES 389
Downloaded by McMaster University from www.liebertpub.com at 05/24/23. For personal use only.
Table 1. Summary of in vivo CRISPR studies for the treatment of muscular dystrophies
Reference Model Cas System Strategy Age Vector Route of Delivery Highlights
Exon skipping and reframing
Amoasii et al.
22
Beagle DmdD50 SpCas9 nuclease Exon 51 skipping
through splice site
manipulation and
reframing
4 weeks AAV9 Intramuscular (cranial tibialis)
and systemic (intravenous)
Systemic injection of a high viral dose restored dystrophin expression
between 3% and 92% across all analyzed muscles.
Nelson et al.
23
mdx mouse SaCas9 nuclease Removal of exon 23
using two sgRNAs
P2, 6 weeks AAV8 Intramuscular (tibialis anterior)
and systemic (intraperitoneal)
Intramuscular and systemic injections led to dystrophin expression in
myofibers, along with localization of DAPC proteins and nNOS activity
at the sarcolemma, increasing muscle function.
Amoasii et al.
26
Mouse DmdD50 SpCas9 nuclease Exon 51 skipping
through splice site
manipulation and
reframing
P4, P12 AAV9 Intramuscular (tibialis anterior)
and systemic (intraperitoneal)
Systemic application restored dystrophin protein expression in all
examined muscles. Four weeks after injection, treated mice had
enhanced grasp strength.
Kemaladewi
et al.
29
dy
2J
/dy
2J
mouse SaCas9 nuclease Restoration of splice
donor site
P2, 3 weeks AAV9 Intramuscular (tibialis anterior)
and systemic (intraperitoneal
and temporal vein)
Treated mice exhibited reduced fibrosis, increased muscle fiber size, and
improved function.
Muscle and the sciatic nerve were effectively modified by the systemic
application.
Zhang et al.
30
Mouse DmdD50 SaCas9 (KKH)
nuclease
Exon 51 reframing P4 AAV9 Systemic (intraperitoneal) Systemic delivery of the KKH system restored 30–55% of wildtype levels
of dystrophin in skeletal and cardiac muscles.
Min et al.
31
Mouse DmdD43,
DmdD45 and
DmdD52
SpCas9 nuclease Exon skipping through
splice site
manipulation and
reframing
P12 scAAV9
and ssAAV9
Intramuscular (tibialis anterior) All three mutations were successfully corrected with the single-cut
strategy and resulted in systemic expression of dystrophin between
14% and 23% of wild-type levels.
Hakim et al.
119
GRMD, WCMD, and
LRMD
SaCas9 and
SpCas9 nuclease
Exon removal and exon
skipping through
splice site
manipulation
Newborns,
2–6 weeks
AAV8 Intramuscular (biceps femoris,
cranial tibialis,
semitendinosus, lateral
gastrocnemius, deltoid, and
extensor carpi ulnaris) and
systemic injection (cephalic
vein)
Evaluated intramuscular and intravenous delivery of the Cas9 editing
system using AAV8s in three canine models of DMD. Demonstrated
that AAV-mediated expression of CRISPR reagents induced a humoral
and B and T cell-based immune response to Cas9, which reduced the
extent and duration of dystrophin restoration.
Min et al.
127
Mouse DmdD44 SpCas9 nuclease Exon 43 and 45 skipping
through splice site
manipulation and
reframing
P12 AAV9 Intramuscular (tibialis anterior)
and systemic (intraperitoneal)
Cas9 editing is dependent on the quantity of sgRNA present; therefore,
sgRNA is the limiting factor for in vivo editing efficacy. Systemic
administration of 1:10 Cas9:sgRNA led to 90–95% of wild-type
dystrophin expression in cardiomyocytes and myofibers.
Hakim et al.
128
mdx mouse SaCas9 nuclease Removal of exon 23
using two sgRNAs
6 weeks AAV9 Intramuscular (tibialis anterior)
and systemic injection (tail
vein)
Evaluated long-term AAV-mediated CRISPR editing in mdx mice and
observed significant dystrophin restoration in skeletal and cardiac
muscles. Demonstrated disproportional sgRNA vector depletion.
Increasing the sgRNA vector ameliorated the issue, enhancing muscle
histology and hemodynamics.
Young et al.
129
hDMD/mdx and
hDMDD52/mdxD2
SpCas9 nuclease Removal of exon 45–55
using two sgRNAs
12–18.5 weeks Plasmids Muscle electro-poration of flexor
digitorum brevis
First investigation using the CRISPR system to target hDMD/mdx model.
(continued)
390 j
Downloaded by McMaster University from www.liebertpub.com at 05/24/23. For personal use only.
Table 1. (Continued)
Reference Model Cas System Strategy Age Vector Route of Delivery Highlights
Duplication removal
Maino et al.
38
Exons 18–30 duplication SaCas9 nuclease Single sgRNA
duplication removal
P1–P2 AAV9 Systemic (temporal vein) Treated mice demonstrated significant dystrophin protein restoration in
various muscle groups, notably the heart, averaging 17% of wildtype
levels. The dystrophic phenotype was significantly improvement based
on histological and functional assays.
Transcriptional modulation of disease modifiers in MDs
Kemaladewi
et al.
56
dy
2J
/dy
2J
mouse SaCas9 nuclease Upregulation of Lama1 P2, 3 weeks AAV9 Intramuscular (tibialis anterior)
and systemic injection
(temporal vein)
Treatment of aged, symptomatic mice improved and effectively reversed
the progression of the disease.
Pinto et al.
71
HSA
LR
mouse carrying a
human skeletal actin
transgene containing
250 CTG repeats
dSpCas9 nuclease Transcriptional
inhibition of
expanded
microsatellite repeats
P2 AAV6
and AAV9
Systemic (temporal vein) First published study inhibiting transcription in a DM1 murine model using
dCas9. 5–15% of fibers exhibited total absence of CUG RNA foci.
Base-editing in MDs
Ryu et al.
84
Dmd exon 20
A-T to G-C point
mutation
ABE7.10-nSpCas9 Remove premature
termination codon
7 weeks tsAAV9 Intramuscular (tibialis anterior) Restoration of dystrophin expression via modifying a premature
termination codon. Restored expression of dystrophin in *15%
myofibers.
Chemello et al.
85
Mouse DmdD51 ABEmax-nSpCas9-NG Exon-skipping via splice-
site disruption
P12 Split-intein
AAV9
Intramuscular (tibialis anterior) Exon-skipping via disruption of exon splicing motifs. Restored expression
of dystrophin in *96.5% myofibers.
Arbab et al.
86
Smn2D7C6T ABE8e-SpMAC
+nusinersen
6TAto6CG via
adenine base editing
P0 Split-intein
AAV9
Intracerebroventricular Demonstrated the utility of combinatorial approaches in genetic medicine
(ASO+base editing) for synergistic therapeutic outcomes. Extended the
therapeutic window for base editing via early pharmacologic
intervention.
CRISPR/Cas9-mediated exogenous DNA knock-in to restore wild-type proteins
Bengtsson
et al.
25
mdx
4cv
mice with non-
sense mutation in
exon 53
SaCas9 and
SpCas9 nucleases
Deletion of exons 52–53
and exogenous DNA
knock-in (HDR)
10–12 weeks AAV6 Intramuscular (tibialis anterior)
and systemic (retro-orbital)
Removed exon 53 while delivering an HDR template to correct the
mutation. Retro-orbital delivery of AAV6s carrying CRISPR/Cas9
nucleases yielded *34% dystrophin expression in the heart as well as
widespread expression in all analyzed muscle tissues. HDR correction
efficiency was measured at 0.18%.
Mata Lo
´pez
et al.
92
GRMD with a mutation
in intron 6 splice
acceptor site
SpCas9 nuclease Exogenous DNA knock-
in (HDR)
3 months–8
years
Plasmid Intramuscular (cranial tibialis,
long digital extensor and
peroneus longus)
Intramuscular injections of the CRISPR/Cas9 treatments resulted in
inclusion of exon 7 in the transcript, dystrophin restoration between at
2% and 16%.
Lee et al.
93
mdx mouse SpCas9 RNP Exogenous DNA knock-
in (HDR)
4 weeks Gold
nano-particles
Intramuscular (tibialis anterior
and gastrocnemius)
Simultaneous treatment with CRISPR-Gold and cardiotoxin led to
widespread dystrophin restoration with HDR efficiency of 5.4%.
Pickar-Olive
et al.
96
hDMDD52/mdx SaCas9 nuclease Exogenous DNA knock-
in (HITI)
P2 neonates AAV9 Intramuscular (tibialis anterior)
and systemic (temporal vein)
First study to devise a DNA knock-in strategy around NHEJ. Systemic
delivery of components using a dual AAV9 system was able to achieve
knock-in rates of 4.67–7.47%. The ‘‘superexon’’ strategy could
potentially correct a large of known DMD mutations.
AAV, adeno-associated virus; ASO, antisense oligonucleotide; Cas9, CRISPR-associated protein 9; CRISPR, clustered regularly interspaced short palindromic repeats; CTG, cytosine, thymine, and guanine; DAPC, dystrophin-
associated protein complex; dCas9, dead Cas9; DM1, dystrophy type 1; DMD, Duchenne muscular dystrophy; GRMD, golden-retriever model of DMD; HDR, homology-directed repair; HITI, homology-independent targeted
integration; LRMD, Labrador retriever muscular dystrophy; MD, muscular dystrophy; NHEJ, non-homologous end-joining; RNP, ribonucleoprotein; sgRNA, single-guide RNA; tsAAV, trans-splicing adeno-associated virus; WCMD,
Welsh corgi muscular dystrophy.
j391
Downloaded by McMaster University from www.liebertpub.com at 05/24/23. For personal use only.
The group subsequently tested this approach in vivo in
mice with a deletion of Dmd exon 50. Results demon-
strated that this single-cut strategy restored dystrophin
expression to 30–50% in skeletal muscle and 50% in the
heart, along with improved muscle integrity and function.
The authors note that using SpCas9 tends to favor NHEJ-
mediated insertion of one base pair, which is not the case
with SaCas9. To address this challenge, the authors
screened for guide RNAs that can induce double-strand
breaks (DSBs) in regions of microhomology, which tend
to produce predictable deletions.
Min et al. also tested single-cut genome editing strate-
gies in mouse and cell models of DMD.
31
They compared
both reframing and exon skipping for three mutations
Figure 1. Summary of in vivo validated CRISPR/Cas9 genome editing strategies for the treatment of MDs. On the left are gene stretches containing a mutated
exon, highlighted in red, as well as respective CRISPR/Cas9 strategies, leading to the desired outcomes on the right.(A) Restoring the ORF through CRISPR/
Cas9-mediated exon deletion, exon skipping, and exon reframing. (B) Removal of a duplicated region using single and dual sgRNA approaches. (C) Modulating
the expression of a disease modifier using CRISPR/dCas9 fused to a transcriptional activator or repressor. (D) Correction of a splice site mutation (A–G) using
an adenine BE. (E) HDR and NHEJ mediated exogenous DNA knock-in of a deleted exon. BE, base editor; Cas9, CRISPR-associated protein 9; CRISPR,
clustered regularly interspaced short palindromic repeats; dCas9, dead Cas9; HDR, homology-directed repair; MD, muscular dystrophy; NHEJ, non-homologous
end-joining; ORF, open reading frame; sgRNA, single-guide RNA.
392 FATEHI ET AL.
Downloaded by McMaster University from www.liebertpub.com at 05/24/23. For personal use only.
using SaCas9. From their results, they concluded that +1
reframing has comparable editing efficiencies to -1 re-
framing depending on a chosen guide RNA. They also
note that the indel type observed in iPSC-derived cardio-
myocytes can be used to predict the levels of dystrophin
restoration in vivo.
Advantages, limitations, and future directions of
CRISPR/Cas9-mediated exon skipping and re-
framing. CRISPR/Cas9-mediated exon skipping and
reframing has several advantages compared with ASOs,
including the ability to potentially achieve a permanent
correction using a single treatment, which significantly
improves the cost-benefit profile of this approach, espe-
cially as the cost of clinical-grade AAV production con-
tinues to decrease.
32
Exon skipping and reframing have
been tested in vitro and in vivo, including in large animal
models, and have led to significant and widespread res-
toration of dystrophin. The major limitation of these
strategies is that exon skipping and reframing typically
lead to the production of a truncated gene product. De-
pending on which exons are skipped or lost, this may lead
to varying clinical improvements.
33
To address this, focus
should be placed on studying the impact of such correction
strategies on patients as we approach clinical trials.
Duplication removal
Large genomic duplications typically encompass one or
more exons and result in reading frame disruption or im-
pairment of protein function through the addition of these
exons to the transcript. In DMD, these duplications rep-
resent 10–15% of patient mutations and occur at low but
appreciable frequencies for several other MDs, such as
MDC1A.
34–37
Therapeutics specific for treating MD du-
plication mutations have been mostly neglected, leading to
a significant unmet need for a sizeable patient population.
Presently, applications of CRISPR/Cas9 toward treating
large duplications have only been published for DMD,
though the principles behind these studies should be
broadly applicable to other MDs.
Duplication removal using single- and dual-sgRNA
approaches. Large duplications can be corrected in
one of two ways using CRISPR/Cas9 (Fig. 1B). The first
employs two sgRNAs that flank the duplicated region and,
through coordinated cleavage, excises the intervening
duplication, with subsequent ligation restoring the wild-
type sequence. The second relies on a solitary sgRNA
targeting within the duplicated sequence. As a result, the
original and duplicated sequences are cut in the same lo-
cation with the intervening sequence being excised, also
restoring the wild-type sequence.
There are several key advantages to the latter approach
that make it the preferred option for correcting most large
duplications. Using one sgRNA rather than two notably
reduces the complexity of delivery. Minimizing the es-
sential CRISPR components is critical, as AAVs will
likely remain the delivery vehicle of choice for clinical
trials for the foreseeable future. The single-sgRNA ap-
proach enables packaging of all components into one
AAV.
38
Using the same sgRNA for both target sites may
improve coordinated cutting at both sites by reducing the
potential for asynchronous cleavage and thus enhance
excision of the duplication. In addition, the entire dupli-
cated region is made available for guide selection, which
enables the assessment of numerous sgRNAs to identify
the most efficacious candidates. It should be noted that to
minimize potential disruption to the coding sequence,
sgRNA design should be limited to introns.
In DMD patients, large duplications typically manifest in
tandem, which enables the use of the single-sgRNA ap-
proach. It is worth noting that non-contiguous DMD dupli-
cations have been reported, whereby a dual sgRNA strategy
may be the only option.
39,40
Several groups have demon-
strated the successful restoration of full-length dystrophin in
DMD patient-derived myoblasts harboring large duplica-
tions using the solitary sgRNA strategy. Wojtal et al. were
the first in 2016, correcting a duplication of exons 18–30.
41
Studies soon followed, correcting DMD patient dupli-
cations of exon 2, exons 55–59, exons 18–25, and exons 3–
16.
42–45
We were unable to identify any published study
utilizing the dual sgRNA strategy for correcting a dupli-
cation mutation, likely owing to its notable disadvantages
and the relatively early reporting of the successful use of a
solitary sgRNA. Unfortunately, in vivo application of
CRISPR/Cas9 for correcting DMD duplications is limited
to a single 2021 study by Maino et al.
There, a novel DMD mouse model harboring a 130 kb
exon 18–30 duplication was generated and subsequently
treated with the single-sgRNA approach using SaCas9 and
delivered systemically by AAV9s. Significant full-length
dystrophin restoration, averaging *17%, was observed in
muscle tissues, most notably in the heart, resulting in sig-
nificant improvements in muscle histology and function.
38
Advantages, limitations, and future directions of
CRISPR/Cas9-mediated duplication removal. In vivo
studies on large duplications are extremely limited owing
to the immense difficulty in generating appropriate animal
models relative to large deletions and small insertions or
duplications. In addition to the exon 18–30 duplication
mouse, only one other model has been described, which
possesses an exon 2 duplication, the most common du-
plication observed in DMD patients.
39,46
However, this
model was obtained by inserting exon 2 and only small
portions of its flanking intronic sequences into a duplica-
tion hotspot in intron 2.
46
As a result, the full genomic architecture of a typical
large tandem duplication is lost, limiting investigations
of duplication removal strategies. There does remain
CRISPR/CAS9 TREATMENT OF MUSCULAR DYSTROPHIES 393
Downloaded by McMaster University from www.liebertpub.com at 05/24/23. For personal use only.
immense utility for this model, as demonstrated by the
successful evaluation of an AAV-packaged ASO, which is
currently under investigation in a clinical trial.
47–49
Most
recently, a family of Labrador retrievers possessing a
tandem 400 kb duplication of exons 2–7 in the DMD gene
has been described and may prove to be an important large
animal model for pre-clinical research into duplication-
specific CRISPR/Cas9 therapies.
50
Although ASOs demonstrate some potential use for
single exon duplications, care must be taken as different
exons respond variably to ASOs, with non-productive
skipping of both exons having been observed.
40,47,51
Multi-exonic duplications magnify the difficulty propor-
tional to their size and remain extremely problematic to
treat with ASO.
51
CRISPR/Cas9 provides a solution for
treating duplications, particularly tandem ones, using
single- and dual-sgRNA approaches. Maino et al. have
importantly demonstrated that generating mouse models
with large multi-exonic duplications is feasible.
38
Looking ahead, focus should be shifted toward devel-
oping novel duplication animal models of various MDs
that faithfully reflect patient genomic architecture, in-
cluding humanized mouse models. These will be essential
for enabling translational CRISPR/Cas9 preclinical re-
search. Although studies on the topic remain sparse, there
is a growing appreciation for large duplications in MD and
how CRISPR/Cas9 can be applied to correct them.
Transcriptional modulation of disease
modifiers in MDs
CRISPR/Cas9 technology can be harnessed to modulate
the expression of target genes of interest (Fig. 1C). In this
approach, it is typical to target an inactivated or ‘dead’
Cas9 (dCas9) protein, fused to a transcriptional activator or
repressor, to the promoter region of a target gene.
52
CRISPR activation (CRISPRa) enables overexpression of
target genes via recruitment of transcriptional machinery or
relaxation of surrounding chromatin.
53
CRISPR interference (CRISPRi) is a similar technique
that enables the suppression of target gene expression by
interfering with transcriptional machinery to transiently
inhibit gene expression.
54
The technique works by tar-
geting a dCas9 fused to a transcriptional repressor, to the
promoter region of the gene of interest. Alternatively,
dCas9 can be fused to a variety of epigenetic enzymes that
perform biochemical modifications to condense local
chromatin structure and/or methylate promoters, masking
the gene of interest from transcriptional machinery. For a
more detailed description of CRISPR/Cas9-mediated
transcriptional regulation systems, please refer to the fol-
lowing review.
55
CRISPR/Cas9-mediated transcriptional regulation
of MD disease modifiers. Disease modifier genes are
those, separate from the mutated gene, that can affect the
severity and progression of a disorder. Restoring full-
length dystrophin via gene therapy remains challenging
due to its large size.
1
Expression of alternative homologs
capable of performing the functions of dystrophin would
be an ideal mutation-independent approach, applicable to
all patients.
56,57
Utrophin is a cytoskeletal protein, highly
similar in structure and function to dystrophin, encoded by
the autosomal UTRN gene.
58–61
Utrophin is primarily expressed in fetal skeletal muscle
and is largely replaced by dystrophin in adult tissues.
58
Interestingly, patients with DMD have endogenously in-
creased levels of utrophin distributed in damaged muscle,
and several early studies demonstrated that utrophin
overexpression could rescue the DMD phenotype in
mice.
62,63
In fact, double knockout mice of Dmd and Utrn
possess a far more severe phenotype than Dmd null
mice.
64,65
Although several small molecule-based ap-
proaches are in development to promote utrophin ex-
pression and/or decrease the rate of UTRN transcript
downregulation,
59,66,67
CRISPRa has been demonstrated to be effective as a
highly specific and efficacious method for upregulation of
utrophin. Wojtal et al. demonstrated that a dCas9-VP160
fusion targeting either the UTRN A or Bpromoters could
effectively upregulate utrophin protein expression in
patient-derived myoblasts.
41
Although these results were
recently validated in DMD patient-derived stem cells,
57
this approach requires assessment of efficacy, longevity of
utrophin expression, and characterization of its effects on
the dystrophic phenotype in vivo.
A similar CRISPRa approach has been used to treat
MDC1A via disease modifier upregulation. There are over
350 known pathogenic nonsense, missense, splice site, and
deletion mutations in LAMA2; an efficacious disease
modifier would provide a therapeutic alternative to gene-
editing that is applicable to all MDC1A patients.
68
LAMA1, which encodes the structurally similar laminin-a1
protein, was demonstrated via overexpression studies to be
a suitable candidate to substitute the function of laminin-
a2 in vivo.
Kemaladewi et al. were able to upregulate Lama1 in
skeletal muscle via systemic delivery of a dSaCas9-VP64
protein in mice harboring a splice site mutation in the
Lama2 gene.
56
This strategy led to significant expression
of the laminin-a1 protein and was correlated with im-
provements in the histological hallmarks of MDC1A, in-
cluding significantly reduced fibrosis. Importantly,
systemic treatment of MDC1A mice led to a remarkable
rescue of the advanced hind-limb paralysis phenotype,
with significant improvements in ambulation as well as
increases in specific tetanic force and conduction velocity
in the sciatic nerve.
It is predicted that this approach may also provide new
opportunities for treatment of DMD, as overexpression of
Lama1 has been shown to stabilize the sarcolemma in
394 FATEHI ET AL.
Downloaded by McMaster University from www.liebertpub.com at 05/24/23. For personal use only.
dystrophic myofibers from mice with dystrophinopathy,
further expanding the utility of this approach.
69,70
Ulti-
mately, these results demonstrate the utility of disease
modifiers as legitimate therapeutic strategies for the
treatment of MDs.
The modular nature of the CRISPR/dCas9 system
means it can be used not only to upregulate but also to
downregulate the expression of a target gene. Pinto et al.
treated the transgenic HSA
LR
mouse, which carries a
fragment of the human skeletal actin (HSA) gene with 250
CTG repeats in the 3¢UTR, with dCas9 targeting the CTG
repeats and preventing their inclusion in HSA tran-
scripts.
71
This resulted in a significant decrease in repeat
transcription and an improvement in phenotype. Appli-
cations involving a combinatorial approach with concur-
rent upregulation of protective disease-modifier genes and
downregulation of detrimental genes could represent a
new mutation-independent approach for ameliorating
disease phenotype.
Advantages, limitations, and future directions of
CRISPR/Cas9-mediated transcriptional regulation of
disease modifiers. Modulation of gene expression by
CRISPR has many advantages over traditional nuclease-
based approaches. First, as CRISPRa/CRISPRi is based on
fusions of effector domains linked to a catalytically in-
activated Cas9, there is negligible risk for the introduction
of double-stranded DNA breaks at the target site and the
induction of severe chromosomal damage.
72,73
Although it is still possible for a dCas9-fusion to lo-
calize effector modules to an off-target location in the
genome, it is unlikely that this region is proximal to the
promoter or enhancer motifs, further reducing the chance
of modulating the transcriptional output of an off-target
gene.
74
Transcriptomic studies from the dCas9-VP160-
mediated Lama1 upregulation study largely suggest this
concern is negligible.
56
Second, modulation of disease
modifiers via CRISPRa/CRISPRi offers a mutation-
independent approach to treating genetic diseases.
Combining CRISPR transcriptional modulation with
traditional CRISPR-mediated gene editing strategies can
generate novel therapeutic strategies to treat MDs. These
would involve using CRISPRa to increase the expression of
a CRISPR/Cas9-corrected gene. We believe the primary
advantage of this combinatorial strategy may be especially
relevant to diseases where a small number of corrected
nuclei can rescue a phenotype by supplying sufficient gene
product for the entire tissue. MDs can also benefitfrom such
a strategy since in many neuromuscular disorders, a single
edited nucleus can complement the entire myofiber syn-
cytium with a functional gene product.
75
Base-editing in MDs
The introduction of base-editing was the first iteration
of true gene-editors capable of producing targeted modi-
fications to specified regions of DNA (Fig. 1D).
76
Base-
editing directly generates precise point mutations in
genomic DNA without double-stranded breaks, a DNA
donor template, or cellular homology-directed repair
(HDR).
76–78
These editors comprised a fusion between a
catalytically impaired Cas9 (D10A) nickase and a base-
modifying enzyme that operates on transient single-
stranded DNA intermediates.
76,79
Two classes of DNA base editors have been described
to date: cytosine base editors (CBEs), which convert a C-G
base pair into a T-A base pair, and adenine base editors
(ABEs), which convert an A-T base pair into a G-C base
pair.
76,80
Collectively, CBEs and ABEs can mediate
all four possible transition mutations (C-to-T, A-to-G,
T-to-C, and G-to-A). More information can be found on
the development of base editors in this review by Rees
and Liu.
81
Use of base-editors in the therapeutic context for
MDs. Therapeutic applications of base editing for
neuromuscular disease are a promising strategy due to its
highly efficient nature and precise editing of pathogenic
point mutations in post-mitotic cells, including neurons
and myofibers.
82
Specifically, base editors have the po-
tential to be used as therapeutic interventions in *16% of
DMD patients who harbor a pathogenic single nucleotide
polymorphism that could be corrected by this technology
alone.
83
Base editors have been utilized as such in two
publications, which highlight their potential for correcting
nonsense mutations and manipulating splice sites in exon-
skipping strategies in vivo.
Ryu et al. used a trans-splicing adeno-associated virus
vector system to deliver an ABE7.10 base editor that was
programmed to target a point mutation in Dmd exon 20, a
causative mutation that introduces a premature termina-
tion codon and subsequently produces a DMD pheno-
type.
84
Intramuscular delivery of the ABE7.10 via AAV9s
to the tibialis anterior (TA) was able to successfully per-
form single-nucleotide substitution via A-T to G-C de-
amination to restructure the premature termination codon
at a frequency of 3.3%, 8 weeks post-injection.
Notably, this genomic correction was accompanied by a
17% restoration of dystrophin-positive myofibers com-
pared to control. Base editors can also be used as a mod-
ified approach for exon-skipping in DMD, as described
earlier. Chemello et al. utilized a dual AAV9 split-intein
trans-splicing ABEmax-SpCas9-NG system to accomplish
exon-skipping via A-T to G-C deamination of the splice
donor site to interrupt it and exclude exon 50 from the
mature Dmd transcript in a Dmd exon 51 deletion mouse.
85
Intramuscular injection into the TA achieved an on-
target genomic editing efficiency of 35%, and Sanger se-
quencing of transcripts illustrated scarless splicing of Dmd
exon 49 to exon 52 as expected. Immunoblotting for full-
length dystrophin demonstrated 54% protein restoration
CRISPR/CAS9 TREATMENT OF MUSCULAR DYSTROPHIES 395
Downloaded by McMaster University from www.liebertpub.com at 05/24/23. For personal use only.
compared with wild-type control. Histological findings
suggested a substantial reduction in fibrosis and 96.5%
restoration of dystrophin-positive fibers in treated animals.
However, the authors note that the titer of intramuscular
AAV9s delivered in this study would extrapolate to a
systemic intravenous dosage corresponding to 1.5E16 vi-
ral genomes per milliliter, significantly higher than an
acceptable threshold for therapeutic usage.
In a recent study, the Liu lab used an ABE to treat an
SMA mouse model (D7SMA).
86
Conveniently, SMN1 has
a paralogous gene in humans known as SMN2, which
mainly differs from SMN1 by a C-G-to-T-A transition in
exon 7, resulting in the skipping of exon 7 in most SMN2
transcripts and the production of only low levels of SMN.
Following intracerebroventricular injection of a split-
intein ABE-Cas9 packaged in an AAV9 vector in neonatal
mice, the authors could detect *40% A-to-G editing on
average in bulk cortical tissue. The treatment also in-
creased the lifespan of the D7SMA mice by *33%, al-
though the authors note that the short lifespan of these
animals makes phenotypic analysis challenging.
Therapeutic advantages, limitations, and future
directions of base-editing in MDs. Although the
technological framework has been rapidly developed to
date, there exist several key challenges to the translation of
base-editing into the clinic, some of which are currently
being resolved. First, the original base-editing platforms
are too large for packaging into traditional AAV vectors
in clinical use today.
87
Currently, most preclinical
base-editing work is being performed using a dual-vector
system, whereby the base editor is assembled post-
translationally in a co-transduced cell via a split-intein
mechanism.
85,88
The nature of dual-vector systems necessitates in-
creased titers of viral vehicles, which possess immuno-
logical and toxicological concerns. In addition, dual
vector systems are more expensive to produce. None-
theless, the Liu lab has recently developed an optimized
suite of ABEs that can be constructed into a single, all-in-
one vector with the added benefit of increased efficiency
of transgene expression, suggesting potentially lower
titer requirements.
89
This was achieved by fusing a single evolved TadA
deaminase domain to smaller nickase variants of Cas9
(SaCas9, SaKKH Cas9, SauriCas9, and NmeCas9), which
has the additional benefit of creating a toolbox of base
editor enzymes with alternative or expanded protospacer
adjacent motif or PAM, theoretically permitting base-
editing at *80% of sites within the genome by the com-
bination of these four smaller nickases. In addition, the
group has utilized directed evolution to generate highly
active cytosine deaminase enzymes from the previously
evolved and smaller adenosine deaminase, as well as hy-
brid deaminase enzymes capable of both adenosine and
cytosine deamination. These novel base editor variants
should lead to the development of CBEs that are small
enough for single AAV packaging in the near future.
CRISPR/Cas9-mediated exogenous DNA
knock-in to restore wild-type proteins
To permanently correct MDs, restoration of the wild-
type protein at physiologically relevant levels is required.
Multi-exonic deletions constitute the majority of DMD
patient mutations (*60%). For precise CRISPR/Cas9
correction of deletions, two DNA knock-in strategies have
been pursued: HDR and NHEJ (Fig. 1E).
HDR-mediated exogenous DNA knock-in. Al-
though HDR is not an efficient pathway in muscle cells
due to their post-mitotic nature,
90,91
several investigations
have attempted to correct a variety of mutations using this
strategy in vivo. In a golden-retriever model of DMD,
Mata Lo
´pez et al. demonstrated correction of a splice ac-
ceptor mutation by delivering plasmids carrying the Cas9
endonuclease and an HDR donor template to the tibio-
tarsal flexor muscle, restoring 2–16% of wild-type dys-
trophin levels.
92
In another study, local delivery of AAV9s carrying
CRISPR/Cas9 and an HDR template to the TA success-
fully replaced exon 53 with an efficiency of 0.18%.
25
Fi-
nally, Lee et al. obtained an HDR frequency of 0.8% in
mdx mice via local delivery using gold nanoparticles
(CRISPR-Gold).
93
HDR efficiency was raised to 5.4%
when mdx mice were simultaneously treated with cardio-
toxin, which activates the proliferation of muscle stem
cells.
93,94
Although several potential HDR-based strate-
gies have been developed to repair mutations in other
forms of MDs, no in vivo studies have been conducted
to date.
95
The inability of HDR-based techniques to restore
clinically significant quantities of protein in muscle cells
prevents their use as a treatment for MD. These findings
demonstrate the limitations of HDR and the need for al-
ternative strategies to improve its effectiveness in muscle.
NHEJ-mediated exogenous DNA knock-in. NHEJ
is the predominant DNA repair pathway in muscle cells
that corrects DSBs. Unlike HDR, NHEJ does not require a
homologous repair template. Homology-independent tar-
geted integration (HITI) relies on CRISPR/Cas9 and
NHEJ to achieve precise DNA knock-ins. Pickar-Oliver
et al. used HITI to knock-in exon 52 of DMD in a hu-
manized mouse model (hDMDD52/mdx) both locally and
systemically.
96
Localized delivery to the TA using AAV9s
resulted in knock-in rates of 0.24–0.67%. In addition to
reintroducing a single exon, a novel strategy using a
‘‘superexon’ donor was investigated.
This knock-in template includes DMD exons 52–79 and
a synthetic poly-A signal, which has the potential to
396 FATEHI ET AL.
Downloaded by McMaster University from www.liebertpub.com at 05/24/23. For personal use only.
correct many DMD mutations located downstream of exon
51. Delivery of the superexon to TAs resulted in knock-in
rates of 0.17%. This treatment was also administered
systemically and resulted in knock-in efficiencies of 0.15–
3.05% across multiple tissues, including both skeletal and
cardiac muscles. Although generally more efficient at in-
troducing exogenous DNA sequences to the genome of
muscle cells, NHEJ-based knock-in strategies are still too
inefficient to warrant translation to clinical trials. Further
investigation is required to improve the efficiency of this
approach in skeletal and cardiac tissue.
Advantages, limitations, and future directions of
CRISPR/Cas9-mediated exogenous DNA knock-
Ins. Low editing efficiencies in muscle are a major
hurdle slowing the development of DNA knock-in strat-
egies to treat MDs. Effective integration of exogenous
DNA would enable a range of new CRISPR applications,
including those unrelated to MDs. Due to the post-mitotic
nature of myotubes and satellite cells, alternative strate-
gies to HDR must be developed to allow for efficient
editing in these cells.
97,98
HITI, homology-mediated end
joining (HMEJ), and prime editing are three emerging
techniques that could address this challenge.
96,99,100
It is proposed that HMEJ occurs via single-strand an-
nealing using a modified DNA donor template that con-
tains sgRNA target sites for its excision from a delivery
vector. AAV9 delivery into the mouse cortex resulted in a
transduced neuron population with a knock-in rate of 50%.
An alternative in vitro study on human-derived IPSCs and
myoblasts demonstrated knock-in rates of 6–30% when
inserting the full DMD transcript.
101
HMEJ is a promising
strategy for the correction of a multitude of MD mutations,
although further investigation is required regarding its
efficacy in vivo.
Cas9 nickase and reverse transcriptase proteins are
fused to create prime editors.
100
Utilizing a 3¢-extended
pegRNA encoding the required template, comparable
in vitro knock-in rates to HDR have been achieved. Che-
mello et al. rescued a deletion of DMD exon 51 by de-
livering a prime editor to patient-derived IPSCs.
85
This
strategy aimed at restoring the expression of the gene by
reframing exon 52 through a +2 insertion and achieved a
correction rate of 20.2%. Prime editing, however, is highly
inefficient at introducing larger sequences of exogenous
DNA, such as those the size of exons.
100,102
Novel strategies such as twin-PE allow for the intro-
duction of large sequences into the genome using prime
editors and integrases together.
102
This creates the possi-
bility of designing systems that can integrate entire gene
transcripts, including engineered promoter regions to ex-
press proteins of interest at physiologically relevant levels.
For in vivo MD applications, however, downsizing of the
technology and validation in muscle cells are still required.
It, nevertheless, presents a promising avenue to pursue.
IN VIVO CRISPR/CAS9 DELIVERY SYSTEMS
Progress and current challenges with
adeno-associated viral vectors
Adeno-associated vectors have emerged as a promising
tool for gene therapy in MDs.
103,104
One of the most
promising AAV serotypes for MD gene therapy is AAV9,
which has shown remarkable efficacy in preclinical stud-
ies.
105–108
AAV9 has a broad tropism for various cell
types, including muscle cells, and can efficiently transduce
both skeletal and cardiac tissues.
109
However, AAVs also
have limitations and disadvantages that must be consid-
ered. One of the main disadvantages is their limited cargo
capacity.
109
In addition, AAVs can elicit an immune response,
particularly on repeated administration or due to pre-
existing immunity in some patients.
110–113
Further, the
production and purification of AAV vectors is currently
complex and expensive, leading to high manufacturing
and treatment costs for patients.
32
Despite these chal-
lenges, the use of AAVs for gene therapy is a promising
approach, and ongoing research is focused on overcoming
these limitations and further advancing the clinical ap-
plication of AAV-mediated gene therapies for MDs.
In 2020, the Grimm group described a novel AAV
capsid variant termed AAVMYO, which consistently
outperformed AAV9s in skeletal and cardiac tissue when
analyzing the level of transcribed cargo.
114
Another recent
advance is the generation of a novel muscle-specific se-
rotype termed MyoAAV1.
115
With a novel engineered
capsid, MyoAAV1 promises higher specificity for muscle
tissue and reduced tropism for the liver. Increasing the
efficiency of cargo delivery to skeletal and cardiac cells
while reducing the potential for side effects is associated
with off-target transduction. Further research is needed to
fully evaluate the safety and efficacy of AAVMYO and
MyoAAV1 for the treatment of MDs.
DISCUSSION
Long-term considerations for the use
of in vivo CRISPR gene editing
It is essential to understand the long-term consequences
and effects of utilizing CRISPR-mediated editing as a
therapeutic strategy. The stability of genomic manipula-
tion in muscle cells is still poorly understood within the
context of MD patients, with a key concern being the rate
of muscle turnover. It is our opinion that without the
correction of satellite cell populations, it is unlikely that
the life-long benefits of a single, virally mediated treat-
ment will be realized. This issue is especially pertinent for
MDs, where the optimal therapeutic window is early
childhood, before the progression of more severe symp-
toms.
1,86
Rapid muscle growth at this age may quickly
render CRISPR therapies that target non-satellite cells
ineffective through the loss or dilution of corrected nuclei.
CRISPR/CAS9 TREATMENT OF MUSCULAR DYSTROPHIES 397
Downloaded by McMaster University from www.liebertpub.com at 05/24/23. For personal use only.
Although AAVs are considered to be a relatively effi-
cient vector for use in humans, adverse effects have been
documented, which may limit their clinical utility. Im-
munogenicity against the virus and the Cas9 machinery, as
well as integration of the AAV into the genome, further
exacerbate immunological and genotoxic con-
cerns.
113,116,117
In a study by the Gersbach group, a hu-
moral and cellular immune response was observed in adult
mdx mice when treated by AAV8/9s carrying an SaCas9,
which was mostly avoided when the same treatment was
given to P2 neonates.
117
AAV-based delivery is the current gold standard for
FDA-approved systemic treatments, and the dose-
dependent adverse effects of AAV, such as temporary
thrombocytopenia and transaminitis, can be effectively
managed.
112,118
The Duan group evaluated intramuscular
and intravenous Cas9 and sgRNA delivery in three
distinct canine models of DMD to measure the effect of
pre-existing Cas9 immunity.
119
They discovered that
AAV-mediated expression of CRISPR reagents not only
promoted editing of the DMD gene but also induced a
B and T cell-based immune response to Cas9, which re-
duced the extent and duration of dystrophin restoration.
Expression of a novel protein that was absent during the
removal of self-reactive lymphocytes can stimulate im-
munological responses.
113
Anti-dystrophin antibodies and
immunological rejections have been observed in DMD
animals who have received remedial treatments.
120
Sev-
eral studies have also demonstrated significantly higher
rates of AAV integration into the genome than previously
thought.
113,116,117
AAV genomes can spontaneously inte-
grate into the AAVS1 safe harbor site on chromosome 19
and were previously thought to rarely integrate into DSBs
created in the genome by Cas9.
121
Across two studies, the Gersbach group reports be-
tween *1and*5% of editing outcomes, resulting in
AAV genome integration in both skeletal and cardiac
tissue when Cas9 nucleases were delivered systemically
to mice.
96,117
AAV integration events can pose serious
immunogenic concerns when paired with pre-existing
Cas9 immunity. Hence, along with pharmacological op-
tions such as immunosuppressants to counter immune
Figure 2. Current challenges with in vivo CRISPR/Cas9 genome editing for the treatment of MDs. An overview of the technical difficulties associated with the
use of AAVs for MD therapies, delivery of CRISPR/Cas9 components to satellite cells, immune responses to targeted or edited cells, and CRISPR/Cas9 editing
efficiencies in muscle. AAVs, adeno-associated viruses.
398 FATEHI ET AL.
Downloaded by McMaster University from www.liebertpub.com at 05/24/23. For personal use only.
reactions, extensive safety monitoring will be required
during clinical trials to reduce immune response in
patients.
Conclusions and future directions for the MD
gene editing field
Current innovations in the application of CRISPR/Cas9
have created various new treatment prospects for MDs.
There is tangible excitement in this field, but there con-
tinues to be a lot of necessary work ahead as we begin to
consider clinical trials for these therapeutic avenues. Here,
we lay out four areas that require the most focus: efficient
targeting of satellite cells, improved editing efficiency in
muscle tissue, vector enhancements, and immunogenic
response (Fig. 2).
An ideal MD therapy would target satellite cells, and so
it is imperative to determine if currently available serotypes
can target these cells efficiently. If not, further work needs
to be done to design or evolve a novel serotype that can
efficiently deliver gene editing components to these targets.
Other systems, such as virus-like particles or nanoparticles,
are also potential candidates for this role but require further
in vivo testing regarding efficacy and safety.
CRISPR/Cas9 editing efficiencies are lower than desired
when targeting muscle tissue. Although less than ideal rates
of correction in muscle are partly due to the challenges
associated with delivery of editing machinery to cells, there
is a need to further develop more efficient systems and
strategies. Tools such as base and prime editors are prom-
ising but require further development and in vivo validation.
Knock-in strategies allow for restoration of the wild-type
proteins; however, current approaches such as HDR are too
inefficient. Development of systems reliant on HMEJ and
integrases could open new opportunities to develop MD
therapies based on DNA knock-ins.
AAV9s will likely be used in the first generation of
CRISPR therapeutics for MDs, given their track record in
in vivo investigations and clinical trials. AAV9s may be
the best option available, but future therapies will certainly
require an improved delivery mechanism. It may be in the
form of nanoparticles, as they are highly modular and can
carry a range of transiently expressed cargo.
122,123
By re-
administering nanoparticles to counteract the conse-
quences of muscle turnover, musculoskeletal dystrophies
could be cured.
The scarcity of published studies reveals that systemic
muscle delivery by nanoparticles remains a formidable
obstacle, most likely due to the large quantity of muscle in
the human body and the necessity to target muscles within
the thoracic cavity. Future research should prioritize the
development of an effective, muscle-specific nanoparticle
that can be delivered through the circulatory system.
The immune system poses a significant, multi-
dimensional challenge for gene therapy strategies. Pre-
existing immunity toward AAVs and Cas9 can pose a
health risk to patients. Clearing of edited cells by the im-
mune system due to the expression of Cas9 or the thera-
peutically relevant protein can reduce the effectiveness of
any strategy. The use of immunosuppressants is a potential
strategy to address the immune response elicited by
CRISPR gene therapies, particularly in the case of repeated
administrations
.124–126
However, immunosuppressants in-
crease the risk of infections, cancer, and potential organ
damage. Further work is required to investigate innate and
humoral responses to CRISPR/Cas9 gene editing strategies
in vivo and potential solutions to these challenges.
Although there are challenges to bringing therapeutic
genome editing for MDs closer to the clinic, many exciting
advances have been made to date in the field of gene-
editing in MDs. Ongoing research focusing on improving
delivery vehicles and new formulations of CRISPR com-
ponents such as mRNA and ribonucleoproteins for tran-
sient and tissue-specific delivery will undoubtedly bring
this technology to the forefront of clinical translation.
ACKNOWLEDGMENTS
The Cohn and Ivakine lab members are gratefully ac-
knowledged for their input in this work.
AUTHORS’ CONTRIBUTIONS
S.F.: Conceptualization, Data curation, Writing—
Original draft, Writing—Review and editing, Visualization.
R.M.M.: Conceptualization, Data curation, Writing—
Review and editing, Visualization.
M.J.R.: Conceptualization, Data curation, Writing—
Review and editing.
L.P.: Conceptualization, Data curation, Writing—
Review and editing.
E.A.I.: Conceptualization, Writing—Review and edit-
ing, Supervision.
R.D.C.: Conceptualization, Writing—Review and
editing, Supervision.
AUTHOR DISCLOSURE
No competing financial interests exist.
FUNDING INFORMATION
This work was funded by the Canadian Institutes of
Health Research (Ronald D. Cohn and Evgueni A. Iva-
kine), the McArthur family (Ronald D. Cohn), Jesse’s
Journey (Ronald D. Cohn), and the Michael Hyatt Foun-
dation (Ronald D. Cohn).
CRISPR/CAS9 TREATMENT OF MUSCULAR DYSTROPHIES 399
Downloaded by McMaster University from www.liebertpub.com at 05/24/23. For personal use only.
REFERENCES
1. Duan D, Goemans N, Takeda S, et al. Duchenne
muscular dystrophy. Nat Rev Dis Primers 2021;
7(1):13; doi: 10.1038/s41572-021-00248-3
2. Kustermann M, Rok MJ, Cohn RD, et al. In vivo
genome engineering for the treatment of mus-
cular dystrophies. Curr Stem Cell Rep 2020;6(3):
52–66; doi: 10.1007/s40778-020-00173-3
3. Crispi V, Matsakas A. Duchenne muscular dys-
trophy: Genome editing gives new hope for
treatment. Postgrad Med J 2018;94(1111):296–
304; doi: 10.1136/postgradmedj-2017-135377
4. Emery AEH. Population frequencies of inherited
neuromuscular diseases—A world survey. Neu-
romuscul Disord 1991;1(1):19–29; doi: 10.1016/
0960-8966(91)90039-U
5. Passamano L, Taglia A, Palladino A, et al. Im-
provement of survival in Duchenne Muscular
Dystrophy: Retrospective analysis of 835 pa-
tients. Acta Myol 2012;31(2):121–125.
6. Broomfield J, Hill M, Guglieri M, et al. Life ex-
pectancy in Duchenne muscular dystrophy.
Neurology 2021;97(23):e2304–e2314; doi:
10.1212/WNL.0000000000012910
7. Lynch GS. Role of contraction-induced injury in the
mechanisms of muscle damage in muscular dys-
trophy. Clin Exp Pharmacol Physiol 2004;31(8):
557–561; doi: 10.1111/j.1440-1681.2004.04026.x
8. Tayeb MT. Deletion mutations in Duchenne
muscular dystrophy (DMD) in Western Saudi
children. Saudi J Biol Sci 2010;17(3):237–240;
doi: 10.1016/j.sjbs.2010.04.008
9. Flanigan KM, Dunn DM, von Niederhausern A,
et al. Mutational spectrum of DMD mutations in
dystrophinopathy patients: Application of mod-
ern diagnostic techniques to a large cohort. Hum
Mutat 2009;30(12):1657–1666; doi: 10.1002/
humu.21114
10. He Z, Luo X, Liang L, et al. Merosin-deficient
congenital muscular dystrophy type 1A: A case
report. Exp Ther Med 2013;6(5):1233–1236; doi:
10.3892/etm.2013.1271
11. Gawlik KI, Durbeej M. Skeletal muscle laminin
and MDC1A: Pathogenesis and treatment strat-
egies. Skelet Muscle 2011;1(1):9; doi: 10.1186/
2044-5040-1-9
12. Accorsi A, Cramer ML, Girgenrath M. Fibrogenesis
in LAMA2-related muscular dystrophy is a central
tenet of disease etiology. Front Mol Neurosci
2020;13:3; doi: 10.3389/fnmol.2020.00003
13. Meola G, Cardani R. Myotonic dystrophies: An
update on clinical aspects, genetic, pathology,
and molecular pathomechanisms. Biochim Bio-
phys Acta 2015;1852(4):594–606; doi: 10.1016/
j.bbadis.2014.05.019
14. Bird TD. Myotonic Dystrophy Type 1. University
of Washington: Seattle; 2021.
15. Ozimski LL, Sabater-Arcis M, Bargiela A, et al.
The hallmarks of myotonic dystrophy type 1
muscle dysfunction. Biol Rev Camb Philos Soc
2021;96(2):716–730; doi: 10.1111/brv.12674
16. Rodrigues M, Yokota T. An overview of recent
advances and clinical applications of exon skip-
ping and splice modulation for muscular dys-
trophy and various genetic diseases. Methods
Mol Biol 2018;1828:31–55; doi: 10.1007/978-1-
4939-8651-4_2
17. Lim KRQ, Maruyama R, Yokota T. Eteplirsen in
the treatment of Duchenne muscular dystrophy.
Drug Des Devel Ther 2017;11:533–545; doi:
10.2147/DDDT.S97635
18. Frank DE, Schnell FJ, Akana C, et al. Increased
dystrophin production with golodirsen in patients
with Duchenne muscular dystrophy. Neurology
2020;94(21):e2270–e2282; doi: 10.1212/WNL.00
00000000009233
19. Roshmi RR, Yokota T. Viltolarsen for the treat-
ment of Duchenne muscular dystrophy. Drugs
Today 2019;55(10):627–639; doi: 10.1358/dot
.2019.55.10.3045038
20. Alfano LN, Charleston JS, Connolly AM, et al.
Long-term treatment with eteplirsen in non-
ambulatory patients with Duchenne muscular
dystrophy. Medicine 2019;98(26):e15858; doi:
10.1097/MD.0000000000015858
21. Mendell JR, Goemans N, Lowes LP, et al.
Longitudinal effect of eteplirsen versus historical
control on ambulation in Duchenne muscular
dystrophy. Ann Neurol 2016;79(2):257–271; doi:
10.1002/ana.24555
22. Amoasii L, Hildyard JCW, Li H, et al. Gene editing
restores dystrophin expression in a canine model
of Duchenne muscular dystrophy. Science 2018;
362(6410):86–91; doi: 10.1126/science.aau1549
23. Nelson CE, Hakim CH, Ousterout DG, et al.
In vivo genome editing improves muscle function
in a mouse model of Duchenne muscular dys-
trophy. Science 2016;351(6271):403–407; doi:
10.1126/science.aad5143
24. Moretti A, Fonteyne L, Giesert F, et al. Somatic
gene editing ameliorates skeletal and cardiac
muscle failure in pig and human models of Du-
chenne muscular dystrophy. Nat Med 2020;26(2):
207–214; doi: 10.1038/s41591-019-0738-2
25. Bengtsson NE, Hall JK, Odom GL, et al. Muscle-
specific CRISPR/Cas9 dystrophin gene editing
ameliorates pathophysiology in a mouse model
for Duchenne muscular dystrophy. Nat Commun
2017;8(1):1–10; doi: 10.1038/ncomms14454
26. Amoasii L, Long C, Li H, et al. Single-cut genome
editing restores dystrophin expression in a new
mouse model of muscular dystrophy. Sci Transl
Med 2017;9(418):eaan8081; doi: 10.1126/
scitranslmed.aan8081
27. Song B, Yang S, Hwang G-H, et al. Analysis of
NHEJ-based DNA repair after CRISPR-mediated
DNA cleavage. Int J Mol Sci 2021;22(12):6397;
doi: 10.3390/ijms22126397
28. Kluesner MG, Lahr WS, Lonetree C-L, et al.
CRISPR-Cas9 cytidine and adenosine base edit-
ing of splice-sites mediates highly-efficient dis-
ruption of proteins in primary and immortalized
cells. Nat Commun 2021;12(1):1–12; doi:
10.1038/s41467-021-22009-2
29. Kemaladewi DU, Maino E, Hyatt E, et al. Cor-
rection of a splicing defect in a mouse model of
congenital muscular dystrophy type 1A using a
homology-directed-repair-independent mecha-
nism. Nat Med 2017;23(8):984–989; doi:
10.1038/nm.4367
30. Zhang Y, Nishiyama T, Li H, et al. A consolidated
AAV system for single-cut CRISPR correction of a
common Duchenne muscular dystrophy mutation.
Mol Ther Methods Clin Dev 2021;22:122–132;
doi: 10.1016/j.omtm.2021.05.014
31. Min Y-L, Chemello F, Li H, et al. Correction of
three prominent mutations in mouse and human
models of Duchenne muscular dystrophy by
single-cut genome editing. Mol Ther 2020;28(9):
2044–2055; doi: 10.1016/j.ymthe.2020.05.024
32. Rodrı
´guez Pinhao Miessner D. Production of AAV
Vectors for Gene Therapy: A Cost-Effectiveness
and Risk Assessment. Massachusetts Institute of
Technology: Cambridge, MA; 2016.
33. Echigoya Y, Lim KRQ, Nakamura A, et al. Multiple
exon skipping in the Duchenne muscular dystro-
phy hot spots: Prospects and challenges. J Pers
Med 2018;8(4):41; doi: 10.3390/jpm8040041
34. Bladen CL, Salgado D, Monges S, et al. The
TREAT-NMD DMD Global Database: Analysis of
more than 7,000 Duchenne muscular dystrophy
mutations. Hum Mutat 2015;36(4):395–402; doi:
10.1002/humu.22758
35. Oliveira J, Gruber A, Cardoso M, et al. LAMA2
gene mutation update: Toward a more compre-
hensive picture of the laminin-a2 variome and its
related phenotypes. Hum Mutat 2018;39(10):
1314–1337; doi: 10.1002/humu.23599
36. Oliveira J, Gonc¸alves A, Oliveira ME, et al. Re-
viewing large LAMA2 deletions and duplications
in congenital muscular dystrophy patients. J
Neuromuscul Dis 2014;1(2):169–179.
37. Ge L, Liu A, Gao K, et al. Deletion of exon 4 in
LAMA2 is the most frequent mutation in Chinese
patients with laminin a2-related muscular dys-
trophy. Sci Rep 2018;8(1):14989; doi: 10.1038/
s41598-018-33098-3
38. Maino E, Wojtal D, Evagelou SL, et al. Targeted
genome editing in vivo corrects a DMD dupli-
cation restoring wild-type dystrophin expression.
EMBO Mol Med 2021;13(5):e13228; doi:
10.15252/emmm.202013228
39. White SJ, Aartsma-Rus A, Flanigan KM, et al.
Duplications in the DMD gene. Hum Mutat 2006;
27(9):938–945; doi: 10.1002/humu.20367
40. Aartsma-Rus A, Janson AAM, van Ommen G-JB,
et al. Antisense-induced exon skipping for duplica-
400 FATEHI ET AL.
Downloaded by McMaster University from www.liebertpub.com at 05/24/23. For personal use only.
tions in Duchenne muscular dystrophy. BMC Med
Genet 2007;8:43; doi: 10.1186/1471-2350-8-43
41. Wojtal D, Kemaladewi DU, Malam Z, et al. Spell
checking nature: Versatility of CRISPR/Cas9 for
developing treatments for inherited disorders.
Am J Hum Genet 2016;98(1):90–101; doi:
10.1016/j.ajhg.2015.11.012
42. Lattanzi A, Duguez S, Moiani A, et al. Correction
of the Exon 2 duplication in DMD myoblasts by a
single CRISPR/Cas9 system. Mol Ther Nucleic
Acids 2017;7:11–19; doi: 10.1016/j.omtn
.2017.02.004
43. Long C, Li H, Tiburcy M, et al. Correction of
diverse muscular dystrophy mutations in human
engineered heart muscle by single-site genome
editing. Sci Adv 2018;4(1):eaap9004; doi:
10.1126/sciadv.aap9004
44. Wang D-N, Wang Z-Q, Jin M, et al. CRISPR/
Cas9-based genome editing for the modification
of multiple duplications that cause Duchenne
muscular dystrophy. Gene Ther 2022;29(12):730–
737; doi: 10.1038/s41434-022-00336-3
45. Pini V, Mariot V, Dumonceaux J, et al. Transiently
expressed CRISPR/Cas9 induces wild-type dys-
trophin in vitro in DMD patient myoblasts carrying
duplications. Sci Rep 2022;12(1):3756; doi:
10.1038/s41598-022-07671-w
46. Vulin A, Wein N, Simmons TR, et al. The first
exon duplication mouse model of Duchenne
muscular dystrophy: A tool for therapeutic de-
velopment. Neuromuscul Disord 2015;25(11):
827–834; doi: 10.1016/j.nmd.2015.08.005
47. Wein N, Vetter TA, Vulin A, et al. Systemic
delivery of an AAV9 exon-skipping vector sig-
nificantly improves or prevents features of Du-
chenne muscular dystrophy in the Dup2 mouse.
Mol Ther Methods Clin Dev 2022;26:279–293;
doi: 10.1016/j.omtm.2022.07.005
48. Wein N, Dunn DM, Waldrop MA, et al. Absence
of significant off-target splicing variation with a
U7snRNA vector targeting DMD exon 2 dupli-
cations. Hum Gene Ther 2021;32(21–22):1346–
1359; doi: 10.1089/hum.2020.315
49. Anonymous. AAV9 U7snRNA Gene Therapy to
Treat Boys With DMD Exon 2 Duplications.
Available from: https://clinicaltrials.gov/ct2/
show/NCT04240314 [Last accessed: April 11,
2023].
50. Shelton GD, Minor KM, Vieira NM, et al. Tan-
dem duplication within the DMD gene in Lab-
rador retrievers with a mild clinical phenotype.
Neuromuscul Disord 2022;32(10):836–841; doi:
10.1016/j.nmd.2022.08.001
51. Greer KL, Lochmu
¨ller H, Flanigan K, et al. Tar-
geted exon skipping to correct exon duplications
in the dystrophin gene. Mol Ther Nucleic Acids
2014;3(3):e155; doi: 10.1038/mtna.2014.8
52. Ca
´mara E, Lenitz I, Nyga
˚rd Y. A CRISPR activa-
tion and interference toolkit for industrial Sac-
charomyces cerevisiae strain KE6–12. Sci Rep
2020;10(1):14605; doi: 10.1038/s41598-020-
71648-w
53. Heidersbach AJ, Dorighi KM, Gomez JA, et al. A
versatile, high-efficiency platform for CRISPR-
based gene activation. Nat Commun 2023;14(1):
1–10; doi: 10.1038/s41467-023-36452-w
54. Larson MH, Gilbert LA, Wang X, et al. CRISPR
interference (CRISPRi) for sequence-specific
control of gene expression. Nat Protoc 2013;
8(11):2180–2196; doi: 10.1038/nprot.2013.132
55. Adli M. The CRISPR tool kit for genome editing
and beyond. Nat Commun 2018;9(1):1–13; doi:
10.1038/s41467-018-04252-2
56. Kemaladewi DU, Bassi PS, Erwood S, et al. A
mutation-independent approach for muscular
dystrophy via upregulation of a modifier gene.
Nature 2019;572(7767):125–130; doi: 10.1038/
s41586-019-1430-x
57. Sengupta K, Mishra MK, Loro E, et al. Genome
editing-mediated utrophin upregulation in Du-
chenne muscular dystrophy stem cells. Mol Ther
Nucleic Acids 2020;22:500–509; doi: 10.1016/
j.omtn.2020.08.031
58. Blake DJ, Tinsley JM, Davies KE. Utrophin: A
structural and functional comparison to dystro-
phin. Brain Pathol 1996;6(1):37–47; doi: 10.1111/
j.1750-3639.1996.tb00781.x
59. Soblechero-Martı
´nP,Lo
´pez-Martı
´nez A, de la
Puente-Ovejero L, et al. Utrophin modulator
drugs as potential therapies for Duchenne and
Becker muscular dystrophies. Neuropathol Appl
Neurobiol 2021;47(6):711–723; doi: 10.1111/
nan.12735
60. Duan D. Micro-utrophin therapy for Duchenne
muscular dystrophy. Mol Ther 2019;27(11):1872–
1874; doi: 10.1016/j.ymthe.2019.10.011
61. Song Y, Morales L, Malik AS, et al. Non-
immunogenic utrophin gene therapy for the
treatment of muscular dystrophy animal models.
Nat Med 2019;25(10):1505–1511; doi: 10.1038/
s41591-019-0594-0
62. Janghra N, Morgan JE, Sewry CA, et al. Corre-
lation of utrophin levels with the dystrophin
protein complex and muscle fibre regeneration in
Duchenne and Becker muscular dystrophy mus-
cle biopsies. PLoS One 2016;11(3):e0150818; doi:
10.1371/journal.pone.0150818
63. Galvagni F, Cantini M, Oliviero S. The utrophin
gene is transcriptionally up-regulated in re-
generating muscle. J Biol Chem 2002;277(21):
19106–19113; doi: 10.1074/jbc.M109642200
64. Deconinck AE, Rafael JA, Skinner JA, et al.
Utrophin-dystrophin-deficient mice as a model
for Duchenne muscular dystrophy. Cell 1997;
90(4):717–727; doi: 10.1016/s0092-8674(00)
80532-2
65. Isaac C, Wright A, Usas A, et al. Dystrophin and
utrophin ‘double knockout’ dystrophic mice ex-
hibit a spectrum of degenerative musculoskeletal
abnormalities. J Orthop Res 2013;31(3):343–349;
doi: 10.1002/jor.22236
66. Muntoni F, Tejura B, Spinty S, et al. A phase 1b
trial to assess the pharmacokinetics of ezutromid
in pediatric Duchenne muscular dystrophy pa-
tients on a balanced diet. Clin Pharmacol Drug
Dev 2019;8(7):922–933; doi: 10.1002/cpdd.642
67. Loro E, Sengupta K, Bogdanovich S, et al. High-
throughput identification of post-transcriptional
utrophin up-regulators for Duchenne muscle
dystrophy (DMD) therapy. Sci Rep 2020;10(1):
2132; doi: 10.1038/s41598-020-58737-6
68. Oliveira J, Freixo JP, Santos M, et al. LAMA2
Muscular Dystrophy. University of Washington:
Seattle; 2020.
69. Barraza-Flores P, Bates CR, Oliveira-Santos A,
et al. Laminin and integrin in LAMA2-related
congenital muscular dystrophy: From disease to
therapeutics. Front Mol Neurosci 2020;13:1; doi:
10.3389/fnmol.2020.00001
70. Gupta VA, Kawahara G, Myers JA, et al. A splice
site mutation in laminin-a2 results in a severe
muscular dystrophy and growth abnormalities in
zebrafish. PLoS One 2012;7(8):e43794; doi:
10.1371/journal.pone.0043794
71. Pinto BS, Saxena T, Oliveira R, et al. Impeding
transcription of expanded microsatellite repeats
by deactivated Cas9. Mol Cell 2017;68(3):479–
490.e5; doi: 10.1016/j.molcel.2017.09.033
72. Whinn KS, Kaur G, Lewis JS, et al. Nuclease
dead Cas9 is a programmable roadblock for DNA
replication. Sci Rep 2019;9(1):13292; doi:
10.1038/s41598-019-49837-z
73. Brezgin S, Kostyusheva A, Kostyushev D, et al.
Dead Cas systems: Types, principles, and appli-
cations. Int J Mol Sci 2019;20(23):6041; doi:
10.3390/ijms20236041
74. National Research Council (US) Committee on
Mapping. Sequencing the Human Genome. In-
troduction. National Academies Press (US):
Washington, DC; 1988.
75. Frair PM, Peterson AC. The nuclear-cytoplasmic
relationship in ‘‘mosaic’ skeletal muscle fibers
from mouse chimaeras. Exp Cell Res 1983;145(1):
167–178; doi: 10.1016/s0014-4827(83)80018-4
76. Komor AC, Kim YB, Packer MS, et al. Program-
mable editing of a target base in genomic DNA
without double-stranded DNA cleavage. Nature
2016;533(7603):420–424; doi: 10.1038/nature
17946
77. Yu S-Y, Birkenshaw A, Thomson T, et al. In-
creasing the targeting scope of CRISPR base
editing system beyond NGG. CRISPR J 2022;5(2):
187–202; doi: 10.1089/crispr.2021.0109
78. Porto EM, Komor AC, Slaymaker IM, et al. Base
editing: Advances and therapeutic opportunities.
Nat Rev Drug Discov 2020;19(12):839–859; doi:
10.1038/s41573-020-0084-6
79. Shen B, Zhang W, Zhang J, et al. Efficient ge-
nome modification by CRISPR-Cas9 nickase with
minimal off-target effects. Nat Methods 2014;
11(4):399–402; doi: 10.1038/nmeth.2857
80. Gaudelli NM, Komor AC, Rees HA, et al. Pro-
grammable base editing of ATtoGCin
genomic DNA without DNA cleavage. Nature
CRISPR/CAS9 TREATMENT OF MUSCULAR DYSTROPHIES 401
Downloaded by McMaster University from www.liebertpub.com at 05/24/23. For personal use only.
2017;551(7681):464–471; doi: 10.1038/
nature24644
81. Rees HA, Liu DR. Base editing: precision chem-
istry on the genome and transcriptome of living
cells. Nat Rev Genet 2018;19(12):770–788; doi:
10.1038/s41576-018-0059-1
82. Yeh W-H, Chiang H, Rees HA, et al. In vivo base
editing of post-mitotic sensory cells. Nat Commun
2018;9(1):2184; doi: 10.1038/s41467-018-04580-3
83. Triana-Fonseca P, Parada-Ma
´rquez JF, Silva-
Aldana CT, et al. Genetic profile of the dystro-
phin gene reveals new mutations in Colombian
patients affected with muscular dystrophino-
pathy. Appl Clin Genet 2021;14:399–408; doi:
10.2147/TACG.S317721
84. Ryu S-M, Koo T, Kim K, et al. Adenine base editing
in mouse embryos and an adult mouse model of
Duchenne muscular dystrophy. Nat Biotechnol
2018;36(6):536–539; doi: 10.1038/nbt.4148
85. Chemello F, Chai AC, Li H, et al. Precise cor-
rection of Duchenne muscular dystrophy exon
deletion mutations by base and prime editing.
Sci Adv 2021;7(18):eabg4910; doi: 10.1126/
sciadv.abg4910
86. Arbab M, Matuszek Z, Kray KM, et al. Base
editing rescue of spinal muscular atrophy in cells
and in mice. Science 2023;380(6642):eadg6518;
doi: 10.1126/science.adg6518
87. Wang D, Tai PWL, Gao G. Adeno-associated
virus vector as a platform for gene therapy de-
livery. Nat Rev Drug Discov 2019;18(5):358–378;
doi: 10.1038/s41573-019-0012-9
88. Truong D-JJ, Ku
¨hner K, Ku
¨hn R, et al. Develop-
ment of an intein-mediated split-Cas9 system for
gene therapy. Nucleic Acids Res 2015;43(13):
6450–6458; doi: 10.1093/nar/gkv601
89. Davis JR, Wang X, Witte IP, et al. Efficient
in vivo base editing via single adeno-associated
viruses with size-optimized genomes encoding
compact adenine base editors. Nat Biomed Eng
2022;6(11):1272–1283; doi: 10.1038/s41551-022-
00911-4
90. Merlo D, Di Stasi AMM, Bonini P, et al. DNA
repair in post-mitotic neurons: A gene-trapping
strategy. Cell Death Differ 2005;12(3):307–309;
doi: 10.1038/sj.cdd.4401572
91. Zhao X, Wei C, Li J, et al. Cell cycle-dependent
control of homologous recombination. Acta Bio-
chim Biophys Sin 2017;49(8):655–668; doi:
10.1093/abbs/gmx055
92. Mata Lo
´pez S, Balog-Alvarez C, Vitha S, et al.
Challenges associated with homologous directed
repair using CRISPR-Cas9 and TALEN to edit the
DMD genetic mutation in canine Duchenne
muscular dystrophy. PLoS One 2020;15(1):
e0228072; doi: 10.1371/journal.pone.0228072
93. Lee K, Conboy M, Park HM, et al. Nanoparticle
delivery of Cas9 ribonucleoprotein and donor
DNA in vivo induces homology-directed DNA
repair. Nat Biomed Eng 2017;1:889–901; doi:
10.1038/s41551-017-0137-2
94. Garry GA, Antony ML, Garry DJ. Cardiotoxin in-
duced injury and skeletal muscle regeneration.
Methods Mol Biol 2016;1460:61–71; doi:
10.1007/978-1-4939-3810-0_6
95. Turan S, Farruggio AP, Srifa W, et al. Precise
correction of disease mutations in induced plu-
ripotent stem cells derived from patients with
limb girdle muscular dystrophy. Mol Ther 2016;
24(4):685–696; doi: 10.1038/mt.2016.40
96. Pickar-Oliver A, Gough V, Bohning JD, et al. Full-
length dystrophin restoration via targeted exon
integration by AAV-CRISPR in a humanized
mouse model of Duchenne muscular dystrophy.
Mol Ther 2021;29(11):3243–3257; doi: 10.1016/
j.ymthe.2021.09.003
97. Yin H, Price F, Rudnicki MA. Satellite cells and
the muscle stem cell niche. Physiol Rev 2013;
93(1):23–67; doi: 10.1152/physrev.00043.2011
98. Partridge TA. Cells that participate in regenera-
tion of skeletal muscle. Gene Ther 2002;9(11):
752–753; doi: 10.1038/sj.gt.3301764
99. Yao X, Wang X, Hu X, et al. Homology-mediated
end joining-based targeted integration using
CRISPR/Cas9. Cell Res 2017;27(6):801–814; doi:
10.1038/cr.2017.76
100. Anzalone AV, Randolph PB, Davis JR, et al. Search-
and-replace genome editing without double-strand
breaks or donor DNA. Nature 2019;576(7785):149–
157; doi: 10.1038/s41586-019-1711-4
101. Tasca F, Brescia M, Wang Q, et al. Large-scale
genome editing based on high-capacity adeno-
vectors and CRISPR-Cas9 nucleases rescues full-
length dystrophin synthesis in DMD muscle cells.
Nucleic Acids Res 2022;50(13):7761–7782; doi:
10.1093/nar/gkac567
102. Anzalone AV, Gao XD, Podracky CJ, et al. Pro-
grammable deletion, replacement, integration
and inversion of large DNA sequences with twin
prime editing. Nat Biotechnol 2022;40(5):731–
740; doi: 10.1038/s41587-021-01133-w
103. Sharma G, Sharma AR, Bhattacharya M, et al.
CRISPR-Cas9: A preclinical and clinical perspective
for the treatment of human diseases. Mol Ther 2021;
29(2):571–586; doi: 10.1016/j.ymthe.2020.09.028
104. Xu CL, Ruan MZC, Mahajan VB, et al. Viral de-
livery systems for CRISPR. Viruses 2019;11(1):28;
doi: 10.3390/v11010028
105. Manini A, Abati E, Nuredini A, et al. Adeno-
associated virus (AAV)-mediated gene therapy
for Duchenne muscular dystrophy: The issue of
transgene persistence. Front Neurol 2021;12:
814174; doi: 10.3389/fneur.2021.814174
106. Birch SM, Lawlor MW, Conlon TJ, et al. As-
sessment of systemic AAV-microdystrophin gene
therapy in the GRMD model of Duchenne mus-
cular dystrophy. Sci Transl Med 2023;15(677):
eabo1815; doi: 10.1126/scitranslmed.abo1815
107. Naso MF, Tomkowicz B, Perry WL 3rd, et al.
Adeno-associated virus (AAV) as a vector for
gene therapy. BioDrugs 2017;31(4):317–334; doi:
10.1007/s40259-017-0234-5
108. Challis RC, Ravindra Kumar S, Chen X, et al.
Adeno-associated virus toolkit to target diverse
brain cells. Annu Rev Neurosci 2022;45:447–469;
doi: 10.1146/annurev-neuro-111020-100834
109. Forsayeth JR, Bankiewicz KS. AAV9: Over the
fence and into the woods. Mol Ther 2011;19(6):
1006–1007; doi: 10.1038/mt.2011.95
110. Shirley JL, de Jong YP, Terhorst C, et al. Immune
responses to viral gene therapy vectors. Mol
Ther 2020;28(3):709–722; doi: 10.1016/j.ymthe
.2020.01.001
111. Whitehead M, Osborne A, Yu-Wai-Man P, et al.
Humoral immune responses to AAV gene therapy
in the ocular compartment. Biol Rev Camb Philos
Soc 2021;96(4):1616–1644; doi: 10.1111/brv.12718
112. Mingozzi F, High KA. Immune responses to AAV
vectors: Overcoming barriers to successful gene
therapy. Blood 2013;122(1):23–36; doi: 10.1182/
blood-2013-01-306647
113. Dowling JJ. CRISPR editing as a therapeutic
strategy for Duchenne muscular dystrophy-anti-
Cas9 immune response casts its shadow over
safety and efficacy. Gene Ther 2022;29(10–11):
575–577; doi: 10.1038/s41434-022-00323-8
114. Weinmann J, Weis S, Sippel J, et al. Identifi-
cation of a myotropic AAV by massively parallel
in vivo evaluation of barcoded capsid variants.
Nat Commun 2020;11(1):5432; doi: 10.1038/
s41467-020-19230-w
115. Tabebordbar M, Lagerborg KA, Stanton A, et al.
Directed evolution of a family of AAV capsid
variants enabling potent muscle-directed gene
delivery across species. Cell 2021;184(19):4919–
4938.e22; doi: 10.1016/j.cell.2021.08.028
116. Hanlon KS, Kleinstiver BP, Garcia SP, et al. High
levels of AAV vector integration into CRISPR-
induced DNA breaks. Nat Commun 2019;10(1):
4439; doi: 10.1038/s41467-019-12449-2
117. Nelson CE, Wu Y, Gemberling MP, et al. Long-
term evaluation of AAV-CRISPR genome editing
for Duchenne muscular dystrophy. Nat Med 2019;
25(3):427–432; doi: 10.1038/s41591-019-0344-3
118. Kishimoto TK, Samulski RJ. Addressing high dose
AAV toxicity—‘One and done or ‘slower and
lower’? Expert Opin Biol Ther 2022;22(9):1067–
1071; doi: 10.1080/14712598.2022.2060737
119. Hakim CH, Kumar SRP, Pe
´rez-Lo
´pez DO, et al.
Cas9-specific immune responses compromise local
and systemic AAV CRISPR therapy in multiple
dystrophic canine models. Nat Commun 2021;
12(1):1–12; doi: 10.1038/s41467-021-26830-7
120. Tripodi L, Villa C, Molinaro D, et al. The immune
system in Duchenne muscular dystrophy patho-
genesis. Biomedicines 2021;9(10):1447; doi:
10.3390/biomedicines9101447
121. Hayashi H, Kubo Y, Izumida M, et al. Efficient
viral delivery of Cas9 into human safe harbor. Sci
Rep 2020;10(1):21474; doi: 10.1038/s41598-020-
78450-8
122. Nie D, Guo T, Yue M, et al. Research progress on
nanoparticles-based CRISPR/Cas9 system for
402 FATEHI ET AL.
Downloaded by McMaster University from www.liebertpub.com at 05/24/23. For personal use only.
targeted therapy of tumors. Biomolecules 2022;
12(9):1239; doi: 10.3390/biom12091239
123. Kazemian P, Yu S-Y, Thomson SB, et al. Lipid-
nanoparticle-based delivery of CRISPR/Cas9
genome-editing components. Mol Pharm 2022;
19(6):1669–1686; doi: 10.1021/acs.molpharma
ceut.1c00916
124. Prasad S, Dimmock DP, Greenberg B, et al. Im-
mune responses and immunosuppressive strat-
egies for adeno-associated virus-based gene
therapy for treatment of central nervous system
disorders: Current knowledge and approaches.
Hum Gene Ther 2022;33(23–24):1228–1245; doi:
10.1089/hum.2022.138
125. Lowenstein PR. The case for immunosuppression
in clinical gene transfer. Mol Ther 2005;12(2):
185–186; doi: 10.1016/j.ymthe.2005.06.439
126. Arruda VR, Favaro P, Finn JD. Strategies to
modulate immune responses: A new frontier for
gene therapy. Mol Ther 2009;17(9):1492–1503;
doi: 10.1038/mt.2009.150
127. Min Y-L, Li H, Rodriguez-Caycedo C, et al. CRISPR-
Cas9 corrects Duchenne muscular dystrophy exon 44
deletion mutations in mice and human cells. Sci Adv
2019;5(3):eaav4324; doi: 10.1126/sciadv.aav4324
128. Hakim CH, Wasala NB, Nelson CE, et al. AAV
CRISPR editing rescues cardiac and muscle func-
tion for 18 months in dystrophic mice. JCI Insight
2018;3(23):124297; doi: 10.1172/jci.insight.124297
129. Young CS, Mokhonova E, Quinonez M, et al.
Creation of a novel humanized dystrophic mouse
model of Duchenne muscular dystrophy and ap-
plication of a CRISPR/Cas9 gene editing therapy.
J Neuromuscul Dis 2017;4(2):139–145; doi:
10.3233/JND-170218
Received for publication April 13, 2023;
accepted after revision April 24, 2023.
Published online: April 28, 2023.
CRISPR/CAS9 TREATMENT OF MUSCULAR DYSTROPHIES 403
Downloaded by McMaster University from www.liebertpub.com at 05/24/23. For personal use only.
... Unlike earlier gene editing methods, CRISPR-Cas9 allows highly accurate targeting of particular DNA sequences, significantly reducing the occurrence of off-target effects. Moreover, this technology can be employed to modify genes in a wide range of organisms, such as plants, animals, and humans, making it a versatile tool for present and future scientific research and potential therapeutic use [11,27,28]. Due to its exceptional precision and versatility in handling diverse samples, CRISPR-Cas9 boasts an impressive efficiency rate. ...
... RT uses pegRNA as a guide to precisely insert, delete, or replace genetic information in the target DNA strand. In the last stage, DNA repair and finalization are performed by sealing the DNA pieces, resulting in the incorporation of edited genetic information into the genome [28,45]. ...
Article
Full-text available
The increasing emergence of zoonotic diseases originating from vectors, rodents, mammals, and others has increased the potential for outbreaks of pandemics in the Indonesian territory. Although control and prevention have been implemented, these efforts have not yet revealed a bright spot; therefore, elaboration with advanced CRISPR-Cas9 technology is a way to accelerate efforts to control zoonotic diseases in Indonesia. However, there is limited literature on this topic. This review aims to comprehensively describe, identify, and summarize the application of CRISPR-Cas9 genome-editing technology in zoonotic disease control in Indonesia. Our findings show that CRISPR-Cas9 genome editing technology offers an innovative approach for zoonotic disease control by targeting disease vectors, modifying animal reservoirs, improving disease surveillance, enhancing vaccine development, and exploring traditional medicine candidates and immunotherapy. The high level of precision, efficiency, and versatility in targeting genomes capable of disrupting, damaging, and disrupting the disease transmission cycle in pathogens makes CRISPR-Cas9 highly effective. However, challenges such as off-target impacts, regulatory complexity, and ethical considerations must be overcome with inter-and multidisciplinary collaboration to promote transparency, equity, and public engagement throughout the process of implementing this technology in the field, especially in Indonesia.
... While there is no cure, ongoing pre-clinical research seeks to restore the normal genetic sequence through emerging gene editing technologies such as CRISPR or produce truncated dystrophin transcripts with exon skipping and microdystrophin gene therapies [10][11][12]. Exon skipping therapy was recently approved by the FDA in the USA for certain mutations, but the technology must be adapted for each of the numerous mutations. Their ability to partially improve muscle dysfunction underscores the importance of maintaining glucocorticoid therapy as a major standard of care, given that systemic inflammation is a major contributor to muscle dysfunction [13][14][15] for virtually all persons with DMD. ...
Article
Full-text available
Duchenne muscular dystrophy (DMD) is caused by genetic mutations in the cytoskeletal-sarcolemmal anchor protein dystrophin. Repeated cycles of sarcolemmal tearing and repair lead to a variety of secondary cellular and physiological stressors that are thought to contribute to weakness, atrophy, and fibrosis. Collectively, these stressors can contribute to a pro-inflammatory milieu in locomotor, cardiac, and respiratory muscles. Given the many unwanted side effects that accompany current anti-inflammatory steroid-based approaches for treating DMD (e.g., glucocorticoids), there is a need to develop new therapies that address inflammation and other cellular dysfunctions. Adiponectin receptor (AdipoR) agonists, which stimulate AdipoR1 and R2 isoforms on various cell types, have emerged as therapeutic candidates for DMD due to their anti-inflammatory, anti-fibrotic, and pro-myogenic properties in pre-clinical human and rodent DMD models. Although these molecules represent a new direction for therapeutic intervention, the mechanisms through which they elicit their beneficial effects are not yet fully understood, and DMD-specific data is limited. The overarching goal of this review is to investigate how adiponectin signaling may ameliorate pathology associated with dystrophin deficiency through inflammatory-dependent and -independent mechanisms and to determine if current data supports their future progression to clinical trials.
... While there is no cure, ongoing pre-clinical research seeks to restore the normal genetic sequence through emerging gene editing technologies such as CRISPR or produce truncated dystrophin transcripts with exon skipping and microdystrophin gene therapies [10][11][12]. Exon skipping therapy was recently approved by the FDA in the USA for certain mutations, but the technology must be adapted for each of the numerous mutations. Their ability to partially improve muscle dysfunction underscores the importance of maintaining glucocorticoid therapy as a major standard of care given systemic inflammation is a major contributor to muscle dysfunction [13][14][15] for virtually all persons with DMD. ...
Preprint
Full-text available
Duchenne muscular dystrophy (DMD) is caused by genetic mutations in the cytoskeletal-sarcolemmal anchor protein dystrophin. Repeated cycles of sarcolemmal tearing and repair lead to a variety of secondary cellular and physiological stressors that are thought to contribute to weakness, atrophy, and fibrosis. Collectively, these stressors can contribute to a pro-inflammatory milieu in locomotor, cardiac, and respiratory muscles. Given the many unwanted side effects that accompany current anti-inflammatory steroid-based approaches for treating DMD (e.g., glucocorticoids), there is a need to develop new therapies that address inflammation and other cellular dysfunctions. Adiponectin receptor (AdipoR) agonists, which stimulate AdipoR1 and R2 isoforms on various cell types, have emerged as therapeutic candidates for DMD due to their anti-inflammatory, anti-fibrotic, and pro-myogenic properties in pre-clinical human and rodent DMD models. Although these molecules represent a new direction for therapeutic intervention, the mechanisms through which they elicit their beneficial effects are not yet fully understood, and DMD-specific data is limited. The overarching goal of this review is to investigate how adiponectin signaling may ameliorate pathology associated with dystrophin deficiency through inflammatory-dependent and -independent mechanisms, and to determine if current data supports their future progression to clinical trials.
... In the medical field, for instance, the use of CRISPRGE in Casgevy therapy yielded remarkable results in the treatments of β-thalassemia and sickle cell disease [71], earning approval from the UK government and the US Food and Drug Administration. Further trials to explore its potential in treating muscular dystrophy [72] and cancer immunotherapy [73] are also ongoing. While it is evident that GE could revolutionize the treatment of genetic and terminal diseases, its implications on society, individuals, and the environment could as well be profound in the realm of food and agriculture [19,74]. ...
... The consequences of these neuromuscular disorders (NMDs) often include skeletal muscle wasting, muscle degeneration, loss of muscle function, lack of mobility, impaired breathing, cardiomyopathy and diaphragm abnormalities (Emery, 1991;Dubowitz, 1978;Monaco et al., 1986;Hoffman et al., 1987;Morrison, 2016;Mary et al., 2018). Recent approaches, including gene editing, gene replacement therapy, cell therapy and RNAbased technologies such as antisense oligonucleotides, have made progress toward clinical trials that could be beneficial in ameliorating certain NMDs (Findlay and Weihl, 2022;Lim and Yokota, 2023;Fatehi et al., 2023). However, these approaches have significant challenges, such as mutation-specific targeting and attaining efficient and body-wide delivery. ...
Article
Full-text available
Skeletal muscular diseases predominantly affect skeletal and cardiac muscle, resulting in muscle weakness, impaired respiratory function and decreased lifespan. These harmful outcomes lead to poor health-related quality of life and carry a high healthcare economic burden. The absence of promising treatments and new therapies for muscular disorders requires new methods for candidate drug identification and advancement in animal models. Consequently, the rapid screening of drug compounds in an animal model that mimics features of human muscle disease is warranted. Zebrafish are a versatile model in preclinical studies that support developmental biology and drug discovery programs for novel chemical entities and repurposing of established drugs. Due to several advantages, there is an increasing number of applications of the zebrafish model for high-throughput drug screening for human disorders and developmental studies. Consequently, standardization of key drug screening parameters, such as animal husbandry protocols, drug compound administration and outcome measures, is paramount for the continued advancement of the model and field. Here, we seek to summarize and explore critical drug treatment and drug screening parameters in the zebrafish-based modeling of human muscle diseases. Through improved standardization and harmonization of drug screening parameters and protocols, we aim to promote more effective drug discovery programs.
... Recently, delandistrogene moxeparvovec, which is an adeno-associated virus (AAV) vector-based gene therapy designed to deliver a gene encoding a micro-dystrophin protein, has been approved in the United States for the treatment of ambulatory DMD pediatric patients and a defined variant in DMD.54 The ongoing preclinical trials in canine models are supporting the treatment developments and options[55][56][57] and substantial advances have been made in a variety of treatments.[58][59][60][61][62][63][64] Defining the feline DMD variants supports the use of cats as a biomedical model in preclinical trials, like that of canine models. ...
Article
Full-text available
Background Muscular dystrophies (MDs) are a large, heterogeneous group of degenerative muscle diseases. X‐linked dystrophin‐deficient MD in cats is the first genetically characterized cat model for a human disease and a few novel forms have been identified. Hypothesis/Objectives Muscular dystrophy was suspected in a young male domestic shorthair cat. Clinical, molecular, and genetic techniques could provide a definitive diagnosis. Animals A 1‐year‐old male domestic shorthair cat presented for progressive difficulty walking, macroglossia and dysphagia beginning at 6 months of age. The tongue was thickened, protruded with constant ptyalism, and thickening and rigidity of the neck and shoulders were observed. Methods A complete neurological examination, baseline laboratory evaluation and biopsies of the trapezius muscle were performed with owner consent. Indirect immunofluorescence staining of muscle cryosections was performed using several monoclonal and polyclonal antibodies against dystrophy‐associated proteins. DNA was isolated for genomic analyses by whole genome sequencing and comparison to DNA variants in the 99 Lives Cat Genome Sequencing dataset. Results and Clinical Importance Aspartate aminotransferase (687 IU/L) and creatine kinase (24 830 IU/L) activities were increased and mild hypokalemia (3.7 mmol/L) was present. Biopsy samples from the trapezius muscle confirmed a degenerative and regenerative myopathy and protein alterations identified by immunohistochemistry resulted in a diagnosis of a in dystrophin‐deficient form of X‐linked MD. A stop gain variant (c.4849C>T; p.Gln1617Ter) dystrophin was identified by genome sequencing. Precision/genomic medicine efforts for the domestic cat and in veterinary medicine support disease variant and animal model discovery and provide opportunities for targeted treatments for companion animals.
... Most of the CRISPR Cas experiments are still at the preclinical primary stage of their development [98][99][100]. ...
Article
Introduction: Duchenne muscular dystrophy (DMD) is one of the most severe and devastating neuromuscular hereditary diseases with a male newborn incidence of 20 000 cases each year. The disease caused by mutations (exon deletions, nonsense mutations, intra-exonic insertions or deletions, exon duplications, splice site defects, and deep intronic mutations) in the DMD gene, progressively leads to muscle wasting and loss of ambulation. This situation is painful for both patients and their families, calling for an emergent need for effective treatments. Areas covered: In this review, the authors describe the state of the gene therapy approach in clinical trials for DMD. This therapeutics included gene replacement, gene substitution, RNA-based therapeutics, readthrough mutation, and the CRISPR approach. Expert opinion: Only a few drug candidates have yet been granted conditional approval for the treatment of DMD. Most of these therapies have only a modest capability to restore the dystrophin or improve muscle function, suggesting an important unmet need in the development of DMD therapeutics. Complementary genes and cellular therapeutics need to be explored to both restore dystrophin, improve muscle function, and efficiently reconstitute the muscle fibers in the advanced stage of the disease.
Article
Duchenne muscular dystrophy (DMD) was named more than 150 years ago. About four decades ago, the DMD gene was discovered, and the reading frame shift was determined as the genetic underpinning. These pivotal findings significantly changed the landscape of DMD therapy development. Restoration of dystrophin expression with gene therapy became a primary focus. Investment in gene therapy has led to the approval of exon skipping by regulatory agencies, multiple clinical trials of systemic microdystrophin therapy using adeno-associated virus vectors, and revolutionary genome editing therapy using the CRISPR technology. However, many important issues surfaced during the clinical translation of DMD gene therapy (such as low efficiency of exon skipping, immune toxicity-induced serious adverse events, and patient death). In this issue of Human Gene Therapy, several research articles highlighted some of the latest developments in DMD gene therapy. Importantly, a collection of articles from experts in the field reviewed the progress, major challenges, and future directions of DMD gene therapy. These insightful discussions have significant implications for gene therapy of other neuromuscular diseases.
Article
Full-text available
Spinal muscular atrophy (SMA), the leading genetic cause of infant mortality, arises from SMN protein insufficiency following SMN1 loss. Approved therapies circumvent endogenous SMN regulation and require repeated dosing or may wane. We describe genome editing of SMN2, an insufficient copy of SMN1 harboring a C6>T mutation, to permanently restore SMN protein levels and rescue SMA phenotypes. We used nucleases or base editors to modify five SMN2 regulatory regions. Base editing converted SMN2 T6>C, restoring SMN protein levels to wild-type. AAV9-mediated base editor delivery in Δ7SMA mice yielded 87% average T6>C conversion, improved motor function, and extended average lifespan, which was enhanced by one-time base editor+nusinersen co-administration (111 versus 17 days untreated). These findings demonstrate the potential of a one-time base editing treatment for SMA.
Article
Full-text available
CRISPR-mediated transcriptional activation (CRISPRa) is a powerful technology for inducing gene expression from endogenous loci with exciting applications in high throughput gain-of-function genomic screens and the engineering of cell-based models. However, current strategies for generating potent, stable, CRISPRa-competent cell lines present limitations for the broad utility of this approach. Here, we provide a high-efficiency, self-selecting CRISPRa enrichment strategy, which combined with piggyBac transposon technology enables rapid production of CRISPRa-ready cell populations compatible with a variety of downstream assays. We complement this with an optimized guide RNA scaffold that significantly enhances CRISPRa functionality. Finally, we describe a synthetic guide RNA tool set that enables transient, population-wide gene activation when used with the self-selecting CRISPRa system. Taken together, this versatile platform greatly enhances the potential for CRISPRa across a wide variety of cellular contexts. The generation of CRISPR-mediated transcriptional activation (CRISPRa)-competent cell lines pose significant technical challenges. Here the authors report a platform for production of CRISPRa-ready cell populations which they combine with optimised expressed and synthetic gRNA scaffolds to enhance functionality.
Article
Full-text available
Cancer is a genetic mutation disease that seriously endangers the health and life of all human beings. As one of the most amazing academic achievements in the past decade, CRISPR/Cas9 technology has been sought after by many researchers due to its powerful gene editing capability. CRISPR/Cas9 technology shows great potential in oncology, and has become one of the most promising technologies for cancer genome-editing therapeutics. However, its efficiency and the safety issues of in vivo gene editing severely limit its widespread application. Therefore, developing a suitable delivery method for the CRISPR/Cas9 system is an urgent problem to be solved at present. Rapid advances in nanomedicine suggest nanoparticles could be a viable option. In this review, we summarize the latest research on the potential use of nanoparticle-based CRISPR/Cas9 systems in cancer therapeutics, in order to further their clinical application. We hope that this review will provide a novel insight into the CRISPR/Cas9 system and offer guidance for nanocarrier designs that will enable its use in cancer clinical applications.
Article
Full-text available
Adeno-associated viruses (AAVs) are being increasingly used as gene therapy vectors in clinical studies especially targeting central nervous system (CNS) disorders. Correspondingly, host immune responses to the AAV capsid or the transgene-encoded protein have been observed in various clinical and preclinical studies. Such immune responses may adversely impact patients' health, prevent viral transduction, prevent repeated dosing strategies, eliminate transduced cells, and pose a significant barrier to the potential effectiveness of AAV gene therapy. Consequently, multiple immunomodulatory strategies have been employed in attempts to limit immune-mediated responses to the vector, enable readministration of AAV gene therapy, prevent end organ toxicity, and increase the duration of transgene-encoded protein expression. Herein we review the innate and adaptive immune responses that may occur during CNS-targeted AAV gene therapy as well as host- and treatment-specific factors that could impact the immune response. We also summarize the available preclinical and clinical data on immune responses specifically to CNS-targeted AAV gene therapy and discuss potential strategies for incorporating prophylactic immunosuppression regimens to circumvent adverse immune responses.
Article
Full-text available
A form of dystrophinopathy with mild or subclinical neuromuscular signs has been previously reported in a family of Labrador retrievers. Markedly and persistently elevated creatine kinase activity was first noted at 6 months of age. Skeletal muscle biopsies revealed a dystrophic phenotype, with dystrophin non-detectable on western blotting and immunohistochemical staining, and with increased utrophin expression. In this report we demonstrate with western blotting that α-dystroglycan is present at essentially normal levels. Whole genome sequencing has also now revealed an approximately 400kb tandem genomic DNA duplication including exons 2-7 of the DMD gene that was inserted into intron 7 of the wild type gene. Skeletal muscle cDNA from 2 cases contained DMD transcripts as expected from an in-frame properly-spliced exon 2 – 7 tandem insertion. A similar 5’ duplication involving DMD exons 2-7 has been reported in a human family with dilated cardiomyopathy but without skeletal myopathy. This is the 3rd confirmed mutation in the DMD gene in Labrador retrievers.
Article
Full-text available
The viral delivery of base editors has been complicated by their size and by the limited packaging capacity of adeno-associated viruses (AAVs). Typically, dual-AAV approaches based on trans-splicing inteins have been used. Here we show that, compared with dual-AAV systems, AAVs with size-optimized genomes incorporating compact adenine base editors (ABEs) enable efficient editing in mice at similar or lower doses. Single-AAV-encoded ABEs retro-orbitally injected in mice led to editing efficiencies in liver (66%), heart (33%) and muscle (22%) tissues that were up to 2.5-fold those of dual-AAV ABE8e, and to a 93% knockdown (on average) of human PCSK9 and of mouse Pcsk9 and Angptl3 in circulation, concomitant with substantial reductions of plasma cholesterol and triglycerides. Moreover, three size-minimized ABE8e variants, each compatible with single-AAV delivery, collectively offer compatibility with protospacer-adjacent motifs for editing approximately 82% of the adenines in the human genome. ABEs encoded within single AAVs will facilitate research and therapeutic applications of base editing by simplifying AAV production and characterization, and by reducing the dose required for the desired level of editing.
Article
Full-text available
Duchenne muscular dystrophy (DMD) is typically caused by mutations that disrupt the DMD reading frame, but nonsense mutations in the 5’ part of the gene induce utilization of an internal ribosomal entry site (IRES) in exon 5, driving expression of a highly functional N-truncated dystrophin. We have developed an AAV9 vector expressing U7 small nuclear RNAs targeting DMD exon 2 and tested it in a mouse containing a duplication of exon 2, in which skipping of both exon 2 copies induces IRES-driven expression, and skipping of one copy leads to wild-type dystrophin expression. One-time intravascular injection at either P0-P1 or at 2 months results in efficient exon skipping and dystrophin expression, and significant protection from functional and pathologic deficits. Immunofluorescence quantification showed 33-53% average dystrophin intensity and 55-79% average dystrophin-positive fibers in mice treated in adulthood, with partial amelioration of DMD pathology and correction of DMD-associated alterations in gene expression. In mice treated neonatally, dystrophin immunofluorescence reached 49-85% of normal intensity and 76-99% dystrophin-positive fibers, with near-complete correction of dystrophic pathology, and these beneficial effects persisted for at least 6 months. Our results demonstrate the robustness, durability, and safety of exon 2 skipping using scAAV9.U7snRNA.ACCA, supporting its clinical use.
Article
Full-text available
Targeted chromosomal insertion of large genetic payloads in human cells leverages and broadens synthetic biology and genetic therapy efforts. Yet, obtaining large-scale gene knock-ins remains particularly challenging especially in hard-to-transfect stem and progenitor cells. Here, fully viral gene-deleted adenovector particles (AdVPs) are investigated as sources of optimized high-specificity CRISPR-Cas9 nucleases and donor DNA constructs tailored for targeted insertion of full-length dystrophin expression units (up to 14.8-kb) through homologous recombination (HR) or homology-mediated end joining (HMEJ). In muscle progenitor cells, donors prone to HMEJ yielded higher CRISPR-Cas9-dependent genome editing frequencies than HR donors, with values ranging between 6% and 34%. In contrast, AdVP transduction of HR and HMEJ substrates in induced pluripotent stem cells (iPSCs) resulted in similar CRISPR-Cas9-dependent genome editing levels. Notably, when compared to regular iPSCs, in p53 knockdown iPSCs, CRISPR-Cas9-dependent genome editing frequencies increased up to 6.7-fold specifically when transducing HMEJ donor constructs. Finally, single DNA molecule analysis by molecular combing confirmed that AdVP-based genome editing achieves long-term complementation of DMD-causing mutations through the site-specific insertion of full-length dystrophin expression units. In conclusion, AdVPs are a robust and flexible platform for installing large genomic edits in human cells and p53 inhibition fosters HMEJ-based genome editing in iPSCs.
Article
Full-text available
Gene editing mediated by CRISPR/Cas9 systems is due to become a beneficial therapeutic option for treating genetic diseases and some cancers. However, there are challenges in delivering CRISPR components which necessitate sophisticated delivery systems for safe and effective genome editing. Lipid nanoparticles (LNPs) have become an attractive nonviral delivery platform for CRISPR-mediated genome editing due to their low immunogenicity and application flexibility. In this review, we provide a background of CRISPR-mediated gene therapy, as well as LNPs and their applicable characteristics for delivering CRISPR components. We then highlight the challenges of CRISPR delivery, which have driven the significant development of new, safe, and optimized LNP formulations in the past decade. Finally, we discuss considerations for using LNPs to deliver CRISPR and future perspectives on clinical translation of LNP-CRISPR gene editing.
Article
Duchenne muscular dystrophy (DMD) is a progressive muscle wasting disease caused by the absence of dystrophin, a membrane-stabilizing protein encoded by the DMD gene. Although mouse models of DMD provide insight into the potential of a corrective therapy, data from genetically homologous large animals, such as the dystrophin-deficient golden retriever muscular dystrophy (GRMD) model, may more readily translate to humans. To evaluate the clinical translatability of an adeno-associated virus serotype 9 vector (AAV9)-microdystrophin (μDys5) construct, we performed a blinded, placebo-controlled study in which 12 GRMD dogs were divided among four dose groups [control, 1 × 1013 vector genomes per kilogram (vg/kg), 1 × 1014 vg/kg, and 2 × 1014 vg/kg; n = 3 each], treated intravenously at 3 months of age with a canine codon-optimized microdystrophin construct, rAAV9-CK8e-c-μDys5, and followed for 90 days after dosing. All dogs received prednisone (1 milligram/kilogram) for a total of 5 weeks from day -7 through day 28. We observed dose-dependent increases in tissue vector genome copy numbers; μDys5 protein in multiple appendicular muscles, the diaphragm, and heart; limb and respiratory muscle functional improvement; and reduction of histopathologic lesions. As expected, given that a truncated dystrophin protein was generated, phenotypic test results and histopathologic lesions did not fully normalize. All administrations were well tolerated, and adverse events were not seen. These data suggest that systemically administered AAV-microdystrophin may be dosed safely and could provide therapeutic benefit for patients with DMD.