Skip to main content
. 2018 Nov 24;8(4):38. doi: 10.3390/jpm8040038

Table 2.

Summary of studies that have used CRISPR/Cas9 approaches for the treatment of DMD.

Cas Enzyme Strategy Target Gene Region(s) Model(s) Delivery Study Highlights Reference
SpCas9 NHEJ reframing, HDR exon correction Dmd exon 23 mdx mice 1-cell embryo injection Dystrophin restoration observed by IHC (up to 100%) and WB; 17% Dmd HDR correction resulted in 47–60% dystrophin-positive fibers in skeletal muscles and the heart 2014 Long et al. [43]
SpCas9 NHEJ reframing, exon skipping, HDR exon knock-in DMD intron 44/exon 45 DMD hiPSCs, hiPSC-derived skeletal muscle cells (ex44 del.) Electroporation Dystrophin restoration in derived skeletal muscle cells observed by WB and IHC for all strategies; CRISPR was as effective as using TALEN 2015 Li et al. [41]
SpCas9 NHEJ reframing, single/multiple exon deletion DMD exons 45–55 (for reframing each exon), introns 50 and 51 (ex51 del.), introns 44 and 55 (ex45–55 del.) immortalized DMD patient muscle cells (ex48–50 del.), immunodeficient NSG mice Electroporation Generated targeted deletions of exon/s in vitro, particularly of the large exon 45–55 region which led to dystrophin rescue by WB; mice transplanted with treated myoblasts (exon 51-deleted) showed dystrophin-positive fibers by IHC 2015 Ousterout et al. [21]
dSpCas9-VP16 Utrophin upregulation UTRN A/B promoter immortalized DMD patient muscle cells (ex45–52 del.) Electroporation 1.7–6.9-fold upregulation of utrophin achieved; restored β-dystroglycan expression observed by WB with as little as 1.7-fold upregulation 2016 Wojtal et al. [44]
SpCas9 Duplicated exons removal DMD intron 27 primary DMD patient fibroblasts (ex18–30 dup.) LV transduction, with Adeno-MyoD 4.42% full-length dystrophin production achieved post-treatment, accompanied with α-dystroglycan restoration 2016 Wojtal et al. [44]
SpCas9 Single exon deletion Dmd exon 23, introns 22 and 23 (ex23 del.) mdx mice AAV9 delivery (i.m., i.p., i.v.) All modes of injection led to appearance of dystrophin-positive fibers as evaluated by IHC: ~25.5% 6 wks post-i.m., ~4.6% and ~9.6% in skeletal and cardiac muscles respectively 12 wks post-i.v., ~1.8% and ~3.2% in skeletal and cardiac muscles respectively 8 wks post-i.p. 2016 Long et al. [45]
SaCas9 Single exon deletion Dmd introns 22 and 23 (ex23 del.) mdx mice AAV8 delivery (i.m., i.p., i.v.) Intramuscular injections led to ~59% of transcripts with exon 23 deleted, which restored about 8% dystrophin of healthy levels by WB, proper relocalization of DGC proteins, and muscle function improvement; systemic injections restored dystrophin production in the heart and skeletal muscles 2016 Nelson et al. [46]
SpCas9, SaCas9 Single exon deletion Dmd introns 22 and 23 (ex23 del.) mdx mice, mdx satellite cells AAV9 delivery (i.m., i.p., i.v.) Dual-vector (Cas9 and gRNAs on separate constructs) had higher cutting efficiency than a single-vector system (Cas9 and gRNAs on the same construct) in vitro; dystrophin restoration >10% observed in the heart and skeletal muscles upon systemic treatment; correction also possible in satellite cells 2016 Tabebordbar et al. [47]
SpCas9 Hybrid exon formation via internal exon deletion DMD exons 50 and 54 immortalized DMD patient muscle cells (ex51–53 del.), hDMD/mdx mice Lipotransfection (in vitro)/ electroporation (in vivo) Dystrophin restoration successful in vitro by WB, not shown in vivo; hybrid exon formation thought to preserve dystrophin rod domain structure better 2016 Iyombe-Engembe et al. [48]
SpCas9 NHEJ reframing, single/multiple exon deletion DMD exons 51, 53, introns 52 and 53 (ex53 del.), 43 and 54 (ex44–54 del.) immortalized DMD patient muscle cells (ex48–50, or 45–52 del.) Sequential LV then AdV transduction/AdV transduction Study showed the possibility of combining both TALEN and CRISPR approaches in one gene editing strategy; also, comparable editing was obtained with Cas9 and gRNA delivered either together or separately in AdV 2016 Maggio et al. [49]
SpCas9 Multiple exon deletion Dmd introns 20 and 23 (ex21–23 del.) mdx mice Electroporation/AdV transduction Treatment restored proper calcium dynamics in muscle (electroporation), and restored dystrophin to 50% of wild-type levels, as well as dystrophin-associated complex sarcolemmal localization and muscle membrane integrity (transduction) 2016 Xu et al. [50]
SpCas9 Multiple exon deletion DMD introns 44 and 55 (ex45–55 del.) DMD hiPSCs, hiPSC-derived skeletal and cardiac muscle cells (ex46–51 or 46–47 del., ex50 dup.), immunodeficient NSG-mdx mice Nucleofection CRISPR-mediated deletion of the large exon 45–55 region achieved, restored membrane function and dystrophin, β-dystroglycan expression by WB and IHC; mice transplanted with hiPSC-derived skeletal muscle cells showed dystrophin-positive fibers by IHC 2016 Young et al. [42]
SpCas9 NHEJ reframing, single/multiple exon deletion DMD exons 51, 53 (for reframing) introns 52 and 53 (ex53 del.), introns 43 and 54 (ex44–54 del.) immortalized DMD patient muscle cells (ex48–50, or 45–52 del.) AdV transduction AdV with 2gRNA-SpCas9 constructs work as good as those with 1gRNA-SpCas9 constructs in terms of corrective ability and dystrophin restoration 2016 Maggio et al. [51]
SpCas9, SaCas9 Multiple exon deletion, HDR exon correction Dmd exon 53, introns 51 and 53 (ex52–53 del.) mdx4cv mice (nonsense ex53 mutation) AAV6 delivery (i.m., i.v.) Dual vector approach (SpCas9 and gRNA separate) yielded higher correction efficiency than single vector approach (SaCas9 and gRNA together); systemic treatment restored dystrophin expression in the heart (~34% dystrophin-positive fibers) and skeletal muscles (~10–50% dystrophin-positive fibers) 2017 Bengtsson et al. [52]
LbCpf1, AsCpf1 NHEJ reframing, single exon skipping, HDR exon correction DMD exon 51, intron 50 DMD hiPSCs, hiPSC-derived cardiac muscle cells (ex48–50 del.), mdx mice Nucleofection (in vitro)/ 1-cell embryo injection (in vivo) Cpf1 editing successfully restored dystrophin expression and improved mitochondrial function in cardiomyocytes; 5/24 pups (injected at the embryo stage) showed HDR correction and had ameliorated dystrophic phenotypes 2017 Zhang et al. [53]
SpCas9 Duplicated exon removal DMD exon 2, intron 2 immortalized DMD patient muscle cells (ex2 dup.) PEI transfection/LV transduction Use of a single gRNA can delete a duplicated exon, resulting in slight dystrophin rescue by WB and IHC 2017 Lattanzi et al. [54]
SpCas9 HDR exon correction Dmd exon 23 mdx mice, mdx satellite cells Lipotransfection (template, gRNA), AdV transduction (Cas9)/AdV transduction Higher transduction efficiency obtained when AdVs were used for both Cas9 and gRNA-HDR template delivery; mice transplanted with corrected satellite cells showed dystrophin-positive fibers by IHC 2017 Zhu et al. [55]
SpCas9 Multiple exon deletion DMD introns 44 and 55 (ex45–55 del.) humanized mdx mice with DMD exon 45 del. Electroporation Exon 45–55 deletion by CRISPR possible in vivo; first use of the humanized DMD mouse model with exon 45 del. for CRISPR studies 2017 Young et al. [56]
SpCas9 Multiple exon deletion DMD introns 2 and 7 (ex3–9 del.), introns 5 and 7 (ex6–7 del.), introns 6 and 11 (ex7–11 del.) DMD hiPSCs, hiPSC-derived cardiac muscle cells (ex8–9 or ex3–7 del.) Nucleofection Dystrophin with ex7–11 del. showed the least functionality, while those with ex3–9 del. had the highest functionality in terms of assessing iPSC-derived cardiomyocyte calcium cycling 2017 Kyrychenko et al. [57]
SpCas9 HDR correction Dmd exon 23 mdx primary muscle cells, mdx mice CRISPR-Gold nanoparticles (i.m.) 5.4% HDR correction of the Dmd mutation in mdx was observed after CRISPR treatment and cardiotoxin injection, dystrophin-positive fibers found by IHC; 0.8% HDR correction observed without cardiotoxin co-injection, which led to significantly improved hanging test performance 2017 Lee et al. [58]
SpCas9 NHEJ reframing, single exon skipping Dmd exon 51 mice with Dmd exon 50 del. AAV9 delivery (i.m., i.p.) Successful dystrophin restoration in the heart and skeletal muscles; systemic injections led to improved muscle function; first application of CRISPR in the ex50 del. mouse model 2017 Amoasii et al. [59]
SpCas9 Single exon deletion Dmd introns 50 and 51 (ex51 del.) primary human skeletal muscle cells HCAdV delivery Up to 93.3% exon 51 deletion observed in vitro upon delivery of CRISPR agents by HCAdV 2017 Ehrke-Schulz et al. [60]
SpCas9 NHEJ reframing, exon skipping DMD exon 51, introns 47, 50, 54 DMD hiPSCs, hiPSC-derived cardiac muscle cells (ex48–50 del., pseudo-ex47, ex55–59 dup.) Nucleofection All strategies corrected the respective patient mutations and restored dystrophin production in iPSC-derived cardiomyocytes; 3D-engineered heart muscle produced from treated iPSC-derived cardiomyocytes showed improved contractile force 2018 Long et al. [61]
CjCas9 NHEJ reframing Dmd exon 23 mice with deletions in Dmd exon 23 AAV9 delivery (i.m.) CjCas9 displayed higher targeting specificity than SpCas9; use of CjCas9-based CRISPR can lead to successful dystrophin restoration and improvement in muscle function as well 2018 Koo et al. [62]
SaCas9 Hybrid exon formation via multiple exon deletion DMD exons 47 and 58 DMD skeletal muscle cells (ex51–53 del., ex49–50 del., ex51–56 del., ex50–52 del.), humanized mdx mice with DMD ex52 del. LV transduction (in vitro)/AAV9 delivery (in vivo; i.v.) gRNAs designed to produce exon deletions that best preserved dystrophin protein structure were able to show dystrophin restoration in vitro and in vivo (slight rescue in the heart) 2018 Duchêne et al. [63]
SpCas9 NHEJ reframing, exon skipping Dystrophin exon 51 deltaE50-MD canine model (ex50 del.) AAV9 delivery (i.m., i.v.) First published study on dystrophin gene correction in a dog model; ~3–70% dystrophin restoration of healthy levels in skeletal muscles and ~92% in the heart found by WB 2018 Amoasii et al. [64]
nSpCas9-ABE7.10 Base editing to correct a nonsense mutation Dmd exon 20 mice with a nonsense mutation in Dmd exon 20 trans-splicing AAV2/9 delivery (i.m.) ~3.3% base editing frequency achieved 8 weeks post-treatment with no detectable off-target effects; ~17% dystrophin-positive fibers and restored localization of nNOS observed by IHC 2018 Ryu et al. [65]
dSa/SpCas9-TAM Base editing to induce exon skipping DMD intron 50 5′ splice site DMD hiPSCs, hiPSC-derived cardiac muscle cells (ex51 del.) Lipotransfection ~100% base editing efficiency achieved; corrected iPSC-derived cardiomyocytes had restored dystrophin protein, low CK and miR-31 levels, and restoration of β-dystroglycan expression 2018 Yuan et al. [66]

Abbreviations: NHEJ, non-homologous end joining; HDR, homology-directed repair; ex, exon; NSG, NOD scid IL2R gamma; hiPSC, human induced pluripotent stem cells; LV, lentivirus; AAV, adeno-associated virus; AdV, adenovirus; HCAdV, high-capacity adenoviral vector; PEI, polyethyleneimine; i.m., intramuscular; i.p., intraperitoneal; i.v., intravenous; WB, Western blot; IHC, immunohistochemistry; TALEN, transcription activator-like effector nuclease; nNOS, neuronal nitric oxide synthase; CK, creatine kinase; gRNA, guide RNA.