Review Article

杜氏肌肉营养不良症基因疗法

卷 24, 期 1, 2024

发表于: 06 January, 2023

页: [17 - 28] 页: 12

弟呕挨: 10.2174/1566523223666221118160932

价格: $65

摘要

杜氏型和贝克型肌营养不良是影响骨骼肌和心肌的等位基因X连锁隐性神经肌肉疾病。因此,由于其单一的X染色体,受影响的男孩会从其不知情的携带者母亲那里获得致病基因突变。目前的药物是姑息性的,可以解决疾病的症状,而不是嵌入肌营养不良蛋白基因DNA序列中的遗传原因。因此,从根本上解决疾病遗传原因的基因药物等替代疗法至关重要,包括基因转移/植入、外显子跳跃和基因编辑。目前,通过基因重编程,可以设计AAV载体,将某些治疗货物专门输送到肌肉或其他器官,无论其血清型如何。类似地,可以引导外泌体的生物发生,将基因编辑成分或某些治疗货物携带到特定的组织或细胞类型,如大脑和肌肉。虽然自体外泌体在免疫上是惰性的,但有可能伪装AAV衣壳和脂质纳米颗粒以逃避免疫系统的识别。在这篇综述中,我们强调了目前Duchenne肌营养不良基因治疗的机会,迄今为止,Duchenne肌肉营养不良是一种无法治愈的遗传病。本文是罕见遗传病基因治疗专题的一部分。

关键词: 杜氏肌肉营养不良症、微营养素、药效团、腺相关病毒、外显子跳跃、CRISPR/Cas基因编辑、基因治疗、基因药物、遗传药物、基因组药物。

图形摘要
[1]
Koenig M, Hoffman EP, Bertelson CJ, Monaco AP, Feener C, Kunkel LM. Complete cloning of the duchenne muscular dystrophy (DMD) cDNA and preliminary genomic organization of the DMD gene in normal and affected individuals. Cell 1987; 50(3): 509-17.
[http://dx.doi.org/10.1016/0092-8674(87)90504-6] [PMID: 3607877]
[2]
Gherardi S, Bovolenta M, Passarelli C, et al. Transcriptional and epigenetic analyses of the DMD locus reveal novel cis acting DNA elements that govern muscle dystrophin expression. Biochim Biophys Acta Gene Regul Mech 2017; 1860(11): 1138-47.
[http://dx.doi.org/10.1016/j.bbagrm.2017.08.010] [PMID: 28867298]
[3]
Young CS, Mokhonova E, Quinonez M, Pyle AD, Spencer MJ. Creation of a novel humanized dystrophic mouse model of duchenne muscular dystrophy and application of a CRISPR/Cas9 gene editing therapy. J Neuromuscul Dis 2017; 4(2): 139-45.
[http://dx.doi.org/10.3233/JND-170218] [PMID: 28505980]
[4]
Saad FA. Novel insights into the complex architecture of osteoporosis molecular genetics. Ann N Y Acad Sci 2020; 1462(1): 37-52.
[http://dx.doi.org/10.1111/nyas.14231] [PMID: 31556133]
[5]
Rodino-Klapac LR, Haidet AM, Kota J, Handy C, Kaspar BK, Mendell JR. Inhibition of myostatin with emphasis on follistatin as a therapy for muscle disease. Muscle Nerve 2009; 39(3): 283-96.
[http://dx.doi.org/10.1002/mus.21244] [PMID: 19208403]
[6]
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.
[http://dx.doi.org/10.1002/humu.22758] [PMID: 25604253]
[7]
Koenig M, Beggs AH, Moyer M, et al. The molecular basis for Duchenne versus Becker muscular dystrophy: Correlation of severity with type of deletion. Am J Hum Genet 1989; 45(4): 498-506.
[PMID: 2491009]
[8]
Love DR, Flint TJ, Genet SA, Middleton-Price HR, Davies KE. Becker muscular dystrophy patient with a large intragenic dystrophin deletion: Implications for functional minigenes and gene therapy. J Med Genet 1991; 28(12): 860-4.
[http://dx.doi.org/10.1136/jmg.28.12.860] [PMID: 1757963]
[9]
Tuffery-Giraud S, Béroud C, Leturcq F, et al. Genotype-phenotype analysis in 2,405 patients with a dystrophinopathy using the UMD-DMD database: A model of nationwide knowledgebase. Hum Mutat 2009; 30(6): 934-45.
[http://dx.doi.org/10.1002/humu.20976] [PMID: 19367636]
[10]
Mostacciuolo ML, Miorin M, Pegoraro E, et al. Reappraisal of the incidence rate of Duchenne and Becker muscular dystrophies on the basis of molecular diagnosis. Neuroepidemiology 1993; 12(6): 326-30.
[http://dx.doi.org/10.1159/000110334] [PMID: 8309507]
[11]
Galvagni F, Saad FA, Danieli GA, et al. A study on duplications of the dystrophin gene: Evidence of a geographical difference in the distribution of breakpoints by intron. Hum Genet 1994; 94(1): 83-7.
[http://dx.doi.org/10.1007/BF02272848] [PMID: 8034300]
[12]
Roberts RG, Bobrow M, Bentley DR. Point mutations in the dystrophin gene. Proc Natl Acad Sci USA 1992; 89(6): 2331-5.
[http://dx.doi.org/10.1073/pnas.89.6.2331] [PMID: 1549596]
[13]
Kilimann M, Pizzuti A, Grompe M, Caskey CT. Point mutations and polymorphisms in the human dystrophin gene identified in genomic DNA sequences amplified by multiplex PCR. Hum Genet 1992; 89(3): 253-8.
[http://dx.doi.org/10.1007/BF00220535] [PMID: 1601417]
[14]
Saad FA, Vitiello L, Merlini L, Mostacciuolo ML, Oliviero S, Danieli GAA. 3′ consensus splice mutation in the human dystrophin gene detected by a screening for intra-exonic deletions. Hum Mol Genet 1992; 1(5): 345-6.
[http://dx.doi.org/10.1093/hmg/1.5.345] [PMID: 1303213]
[15]
Lenk U, Hanke R, Thiele H, Speer A. Point mutations at the carboxy terminus of the human dystrophin gene: Implications for an association with mental retardation in DMD patients. Hum Mol Genet 1993; 2(11): 1877-81.
[http://dx.doi.org/10.1093/hmg/2.11.1877] [PMID: 8281150]
[16]
Prior TW, Papp AC, Snyder PJ, et al. Identification of two point mutations and a one base deletion in exon 19 of the dystrophin gene by heteroduplex formation. Hum Mol Genet 1993; 2(3): 311-3.
[http://dx.doi.org/10.1093/hmg/2.3.311] [PMID: 8499922]
[17]
Saad FA, Vita G, Mora M, et al. A novel nonsense mutation in the human dystrophin gene. Hum Mutat 1993; 2(4): 314-6.
[http://dx.doi.org/10.1002/humu.1380020413] [PMID: 8401539]
[18]
Wilton SD, Johnsen RD, Pedretti JR, Laing NG. Two distinct mutations in a single dystrophin gene: Identification of an altered splice-site as the primary becker muscular dystrophy mutation. Am J Med Genet 1993; 46(5): 563-9.
[http://dx.doi.org/10.1002/ajmg.1320460521] [PMID: 8322822]
[19]
Saad FA, Vita G, Toffolatti L, Danieli GA. A possible missense mutation detected in the dystrophin gene by double strand conformation analysis (DSCA). Neuromuscul Disord 1994; 4(4): 335-41.
[http://dx.doi.org/10.1016/0960-8966(94)90069-8] [PMID: 7981590]
[20]
Tuffery S, Bareil C, Demaille J, Claustres M. Four novel dystrophin point mutations: Detection by protein truncation test and transcript analysis in lymphocytes from Duchenne muscular dystrophy patients. Eur J Hum Genet 1996; 4(3): 143-52.
[http://dx.doi.org/10.1159/000472188] [PMID: 8840114]
[21]
Lasa A, Gallano P, Baiget M. Three novel point mutations in the dystrophin gene in DMD patients. Hum Mutat 1997; 9(5): 473-4.
[http://dx.doi.org/10.1002/(SICI)1098-1004(1997)9:5<473:AID-HUMU16>3.0.CO;2-#] [PMID: 9143930]
[22]
Saad FA, Mostacciuolo ML, Trevisan CP, et al. Novel mutations and polymorphisms in the human dystrophin gene detected by double-strand conformation analysis. Hum Mutat 1997; 9(2): 188-90.
[http://dx.doi.org/10.1002/(SICI)1098-1004(1997)9:2<188:AID-HUMU15>3.0.CO;2-Z] [PMID: 9067763]
[23]
Sitnik R, Campiotto S, Vainzof M, et al. Novel point mutations in the dystrophin gene. Hum Mutat 1997; 10(3): 217-22.
[http://dx.doi.org/10.1002/(SICI)1098-1004(1997)10:3<217:AID-HUMU7>3.0.CO;2-F] [PMID: 9298822]
[24]
Cau M, Cao A, Loi D, et al. Two novel mutations (10410 T→G; 10296 del C) at carboxy-terminus of the dystrophin gene associated with mental retardation. Hum Mutat 1998; 12(1): 70.
[http://dx.doi.org/10.1002/(SICI)1098-1004(1998)12:1<70:AID-HUMU13>3.0.CO;2-G] [PMID: 10627134]
[25]
Saad FA, Merlini L, Mostacciuolo ML, Danieli GA. Double missense mutation in exon 41 of the human dystrophin gene detected by double strand conformation analysis. Am J Med Genet 1998; 80(2): 99-102.
[http://dx.doi.org/10.1002/(SICI)1096-8628(19981102)80:2<99:AID-AJMG1>3.0.CO;2-L] [PMID: 9805122]
[26]
Ikezawa M, Nishino I, Goto Y, Miike T, Nonaka I. Newly recognized exons induced by a splicing abnormality from an intronic mutation of the dystrophin gene resulting in Duchenne muscular dystrophy. Hum Mutat 1999; 13(2): 170.
[http://dx.doi.org/10.1002/(SICI)1098-1004(1999)13:2<170:AID-HUMU12>3.0.CO;2-7] [PMID: 10094556]
[27]
Ginjaar HB, van der Kooi AJ, Ceelie H, et al. Sarcoglycanopathies in Dutch patients with autosomal recessive limb girdle muscular dystrophy. J Neurol 2000; 247(7): 524-9.
[http://dx.doi.org/10.1007/s004150070151] [PMID: 10993494]
[28]
Percesepe A, Ferrari M, Coviello D, et al. Detection of a novel dystrophin gene mutation through carrier analysis performed during prenatal diagnosis in a case with intragenic recombination. Prenat Diagn 2005; 25(11): 1011-4.
[http://dx.doi.org/10.1002/pd.1238] [PMID: 16231306]
[29]
Baskin B, Banwell B, Khater RA, Hawkins C, Ray PN. Becker muscular dystrophy caused by an intronic mutation reducing the efficiency of the splice donor site of intron 26 of the dystrophin gene. Neuromuscul Disord 2009; 19(3): 189-92.
[http://dx.doi.org/10.1016/j.nmd.2008.11.003] [PMID: 19230662]
[30]
Zhu JF, Liu HH, Zhou T, Tian L. Novel mutation in exon 56 of the dystrophin gene in a child with Duchenne muscular dystrophy. Int J Mol Med 2013; 32(5): 1166-70.
[http://dx.doi.org/10.3892/ijmm.2013.1498] [PMID: 24065205]
[31]
Todeschini A, Gualandi F, Trabanelli C, et al. Becker muscular dystrophy due to an intronic splicing mutation inducing a dual dystrophin transcript. Neuromuscul Disord 2016; 26(10): 662-5.
[http://dx.doi.org/10.1016/j.nmd.2016.08.007] [PMID: 27616544]
[32]
Jiang J, Jiang T, Xu J, Shen J, Gao F. Novel mutation of the dystrophin gene in a child with duchenne muscular dystrophy. Fetal Pediatr Pathol 2018; 37(1): 1-6.
[http://dx.doi.org/10.1080/15513815.2017.1369201] [PMID: 29336709]
[33]
Wang Y, Chen Y, Wang SM, Liu X, Gu YN, Feng Z. Prenatal diagnosis of Duchenne muscular dystrophy revealed a novel mosaic mutation in Dystrophin gene: A case report. BMC Med Genet 2020; 21(1): 222.
[http://dx.doi.org/10.1186/s12881-020-01157-0]
[34]
Zimowski JG, Purzycka J, Pawelec M, Ozdarska K, Zaremba J. Small mutations in Duchenne/Becker muscular dystrophy in 164 unrelated Polish patients. J Appl Genet 2021; 62(2): 289-95.
[http://dx.doi.org/10.1007/s13353-020-00605-0] [PMID: 33420945]
[35]
Monaco AP, Bertelson CJ, Liechti-Gallati S, Moser H, Kunkel LM. An explanation for the phenotypic differences between patients bearing partial deletions of the DMD locus. Genomics 1988; 2(1): 90-5.
[http://dx.doi.org/10.1016/0888-7543(88)90113-9] [PMID: 3384440]
[36]
Aartsma-Rus A, Kaman WE, Weij R, den Dunnen JT, van Ommen GJB, van Deutekom JCT. Exploring the frontiers of therapeutic exon skipping for Duchenne muscular dystrophy by double targeting within one or multiple exons. Mol Ther 2006; 14(3): 401-7.
[http://dx.doi.org/10.1016/j.ymthe.2006.02.022] [PMID: 16753346]
[37]
Echigoya Y, Lim KRQ, Nakamura A, Yokota T. Multiple exon skipping in the duchenne muscular dystrophy hot spots: Prospects and challenges. J Pers Med 2018; 8(4): 41.
[http://dx.doi.org/10.3390/jpm8040041] [PMID: 30544634]
[38]
Millay DP, Sargent MA, Osinska H, et al. Genetic and pharmacologic inhibition of mitochondrial-dependent necrosis attenuates muscular dystrophy. Nat Med 2008; 14(4): 442-7.
[http://dx.doi.org/10.1038/nm1736] [PMID: 18345011]
[39]
Wissing ER, Millay DP, Vuagniaux G, Molkentin JD. Debio-025 is more effective than prednisone in reducing muscular pathology in mdx mice. Neuromuscul Disord 2010; 20(11): 753-60.
[http://dx.doi.org/10.1016/j.nmd.2010.06.016] [PMID: 20637615]
[40]
Schiavone M, Zulian A, Menazza S, et al. Alisporivir rescues defective mitochondrial respiration in Duchenne muscular dystrophy. Pharmacol Res 2017; 125(Pt B): 122-31.
[http://dx.doi.org/10.1016/j.phrs.2017.09.001]
[41]
Pellegrini C, Zulian A, Gualandi F, et al. Melanocytes-A novel tool to study mitochondrial dysfunction in Duchenne muscular dystrophy. J Cell Physiol 2013; 228(6): 1323-31.
[http://dx.doi.org/10.1002/jcp.24290] [PMID: 23169061]
[42]
Zulian A, Tagliavini F, Rizzo E, et al. Melanocytes from patients affected by ullrich congenital muscular dystrophy and bethlem myopathy have dysfunctional mitochondria that can be rescued with cyclophilin inhibitors. Front Aging Neurosci 2014; 6: 324.
[http://dx.doi.org/10.3389/fnagi.2014.00324] [PMID: 25477819]
[43]
Tiepolo T, Angelin A, Palma E, et al. The cyclophilin inhibitor Debio 025 normalizes mitochondrial function, muscle apoptosis and ultrastructural defects in Col6a1−/− myopathic mice. Br J Pharmacol 2009; 157(6): 1045-52.
[http://dx.doi.org/10.1111/j.1476-5381.2009.00316.x] [PMID: 19519726]
[44]
Angelin A, Tiepolo T, Sabatelli P, et al. Mitochondrial dysfunction in the pathogenesis of Ullrich congenital muscular dystrophy and prospective therapy with cyclosporins. Proc Natl Acad Sci USA 2007; 104(3): 991-6.
[http://dx.doi.org/10.1073/pnas.0610270104] [PMID: 17215366]
[45]
Merlini L, Nishino I. 201st ENMC International Workshop: Autophagy in muscular dystrophies – Translational approach, 1–3 November 2013, Bussum, The Netherlands. Neuromuscul Disord 2014; 24(6): 546-61.
[http://dx.doi.org/10.1016/j.nmd.2014.03.009] [PMID: 24746377]
[46]
Cencic R, Miura H, Malina A, et al. Protospacer adjacent motif (PAM)-distal sequences engage CRISPR Cas9 DNA target cleavage. PLoS One 2014; 9(10): e109213.
[http://dx.doi.org/10.1371/journal.pone.0109213] [PMID: 25275497]
[47]
Gilbert LA, Larson MH, Morsut L, et al. CRISPR-mediated modular RNA-guided regulation of transcription in eukaryotes. Cell 2013; 154(2): 442-51.
[http://dx.doi.org/10.1016/j.cell.2013.06.044] [PMID: 23849981]
[48]
Eid A, Alshareef S, Mahfouz MM. CRISPR base editors: Genome editing without double-stranded breaks. Biochem J 2018; 475(11): 1955.1964.
[49]
Gapinske M, Luu A, Winter J, et al. CRISPR-SKIP: Programmable gene splicing with single base editors. Genome Biol 2018; 19(1): 107.
[http://dx.doi.org/10.1186/s13059-018-1482-5] [PMID: 30107853]
[50]
Anzalone AV, Randolph PB, Davis JR, et al. Search-and-replace genome editing without double-strand breaks or donor DNA. Nature 2019; 576(7785): 149-57.
[http://dx.doi.org/10.1038/s41586-019-1711-4] [PMID: 31634902]
[51]
Chen PJ, Hussmann JA, Yan J, et al. Enhanced prime editing systems by manipulating cellular determinants of editing outcomes. Cell 2021; 184(22): 5635-5652.e29.
[http://dx.doi.org/10.1016/j.cell.2021.09.018]
[52]
Li HL, Fujimoto N, Sasakawa N, et al. Precise correction of the dystrophin gene in duchenne muscular dystrophy patient induced pluripotent stem cells by TALEN and CRISPR-Cas9. Stem Cell Reports 2015; 4(1): 143-54.
[http://dx.doi.org/10.1016/j.stemcr.2014.10.013] [PMID: 25434822]
[53]
Ousterout DG, Kabadi AM, Thakore PI, Majoros WH, Reddy TE, Gersbach CA. Multiplex CRISPR/Cas9-based genome editing for correction of dystrophin mutations that cause Duchenne muscular dystrophy. Nat Commun 2015; 6(1): 6244.
[http://dx.doi.org/10.1038/ncomms7244] [PMID: 25692716]
[54]
Kourakis S, Timpani CA, Campelj DG, et al. Standard of care versus new-wave corticosteroids in the treatment of Duchenne muscular dystrophy: Can we do better? Orphanet J Rare Dis 2021; 16(1): 117.
[http://dx.doi.org/10.1186/s13023-021-01758-9] [PMID: 33663533]
[55]
Angelini C. The role of corticosteroids in muscular dystrophy: A critical appraisal. Muscle Nerve 2007; 36(4): 424-35.
[http://dx.doi.org/10.1002/mus.20812] [PMID: 17541998]
[56]
Shah D. Are corticosteroids effective in Duchenne muscular dystrophy. Indian Pediatr 2008; 45(5): 401-2.
[PMID: 18515930]
[57]
Manzur AY, Kuntzer T, Pike M, Swan A. Glucocorticoid corticosteroids for Duchenne muscular dystrophy. Cochrane Database Syst Rev 2008; 23(1): CD003725.
[PMID: 18254031]
[58]
Matthews E, Brassington R, Kuntzer T, Jichi F, Manzur AY. Corticosteroids for the treatment of Duchenne muscular dystrophy. Cochrane Libr 2016; 2016(6): CD003725.
[http://dx.doi.org/10.1002/14651858.CD003725.pub4] [PMID: 27149418]
[59]
Schreiber A, Brochard S, Rippert P, et al. Corticosteroids in Duchenne muscular dystrophy: Impact on the motor function measure sensitivity to change and implications for clinical trials. Dev Med Child Neurol 2018; 60(2): 185-91.
[http://dx.doi.org/10.1111/dmcn.13590] [PMID: 28990163]
[60]
Buckon C, Sienko S, Bagley A, et al. Can quantitative muscle strength and functional motor ability differentiate the influence of age and corticosteroids in ambulatory boys with duchenne muscular dystrophy? PLoS Curr 2016. 8. ecurrents.md.1ced64dff945f8958221fddcd4ee60b0.
[61]
Merlini L, Cecconi I, Parmeggiani A, Cordelli DM, Dormi A. Quadriceps muscle strength in Duchenne muscular dystrophy and effect of corticosteroid treatment. Acta Myol 2020; 39(4): 200-6.
[PMID: 33458575]
[62]
Pereira RC, Delany AM, Canalis E. Effects of cortisol and bone morphogenetic protein-2 on stromal cell differentiation: Correlation with CCAAT-enhancer binding protein expression. Bone 2002; 30(5): 685-91.
[http://dx.doi.org/10.1016/S8756-3282(02)00687-7] [PMID: 11996905]
[63]
Weinstein RS, Jilka RL, Parfitt AM, Manolagas SC. Inhibition of osteoblastogenesis and promotion of apoptosis of osteoblasts and osteocytes by glucocorticoids. Potential mechanisms of their deleterious effects on bone. J Clin Invest 1998; 102(2): 274-82.
[http://dx.doi.org/10.1172/JCI2799] [PMID: 9664068]
[64]
Dallas SL, Prideaux M, Bonewald LF. The osteocyte: An endocrine cell... and more. Endocr Rev 2013; 34(5): 658-90.
[http://dx.doi.org/10.1210/er.2012-1026] [PMID: 23612223]
[65]
Mirza F, Canalis E. Management of endocrine disease: Secondary osteoporosis: Pathophysiology and management. Eur J Endocrinol 2015; 173(3): R131-51.
[http://dx.doi.org/10.1530/EJE-15-0118] [PMID: 25971649]
[66]
Hofbauer LC, Gori F, Riggs BL, et al. Stimulation of osteoprotegerin ligand and inhibition of osteoprotegerin production by glucocorticoids in human osteoblastic lineage cells: Potential paracrine mechanisms of glucocorticoid-induced osteoporosis. Endocrinology 1999; 140(10): 4382-9.
[http://dx.doi.org/10.1210/endo.140.10.7034] [PMID: 10499489]
[67]
Van Staa TP, Leufkens HGM, Abenhaim L, Zhang B, Cooper C. Use of oral corticosteroids and risk of fractures. J Bone Miner Res 2000; 15(6): 993-1000.
[http://dx.doi.org/10.1359/jbmr.2000.15.6.993] [PMID: 10841167]
[68]
Heier CR, Yu Q, Fiorillo AA, et al. Vamorolone targets dual nuclear receptors to treat inflammation and dystrophic cardiomyopathy. Life Sci Alliance 2019; 2(1): e201800186.
[http://dx.doi.org/10.26508/lsa.201800186] [PMID: 30745312]
[69]
Donovan JM, Zimmer M, Offman E, Grant T, Jirousek M. A novel NF‐κB inhibitor, Edasalonexent (CAT‐1004), in development as a disease‐Modifying treatment for patients with duchenne muscular dystrophy: Phase 1 safety, pharmacokinetics, and pharmacodynamics in adult subjects. J Clin Pharmacol 2017; 57(5): 627-39.
[http://dx.doi.org/10.1002/jcph.842] [PMID: 28074489]
[70]
Finkel RS, Finanger E, Vandenborne K, et al. Disease-modifying effects of edasalonexent, an NF-κB inhibitor, in young boys with Duchenne muscular dystrophy: Results of the MoveDMD phase 2 and open label extension trial. Neuromuscul Disord 2021; 31(5): 385-96.
[http://dx.doi.org/10.1016/j.nmd.2021.02.001] [PMID: 33678513]
[71]
Guiraud S, Davies KE. Pharmacological advances for treatment in Duchenne muscular dystrophy. Curr Opin Pharmacol 2017; 34: 36-48.
[http://dx.doi.org/10.1016/j.coph.2017.04.002] [PMID: 28486179]
[72]
Zhang Y, Li H, Min YL, et al. Enhanced CRISPR-Cas9 correction of Duchenne muscular dystrophy in mice by a self-complementary AAV delivery system. Sci Adv 2020; 6(8): eaay6812.
[http://dx.doi.org/10.1126/sciadv.aay6812] [PMID: 32128412]
[73]
Yang Y, Wang L, Bell P, et al. A dual AAV system enables the Cas9-mediated correction of a metabolic liver disease in newborn mice. Nat Biotechnol 2016; 34(3): 334-8.
[http://dx.doi.org/10.1038/nbt.3469] [PMID: 26829317]
[74]
Chemello F, Chai AC, Li H, et al. Precise correction of Duchenne muscular dystrophy exon deletion mutations by base and prime editing. Sci Adv 2021; 7(18): eabg4910.
[http://dx.doi.org/10.1126/sciadv.abg4910] [PMID: 33931459]
[75]
Lau CH, Suh Y. In vivo genome editing in animals using AAV-CRISPR system: Applications to translational research of human disease. F1000 Res 2017; 6: 2153-73.
[http://dx.doi.org/10.12688/f1000research.11243.1] [PMID: 29333255]
[76]
Wang JZ, Wu P, Shi ZM, Xu YL, Liu ZJ. The AAV-mediated and RNA-guided CRISPR/Cas9 system for gene therapy of DMD and BMD. Brain Dev 2017; 39(7): 547-56.
[http://dx.doi.org/10.1016/j.braindev.2017.03.024] [PMID: 28390761]
[77]
Mendell JR, Al-Zaidy S, Shell R, et al. Single-dose gene-replacement therapy for spinal muscular atrophy. N Engl J Med 2017; 377(18): 1713-22.
[http://dx.doi.org/10.1056/NEJMoa1706198] [PMID: 29091557]
[78]
Cotten M, Wagner E. Non-viral approaches to gene therapy. Curr Opin Biotechnol 1993; 4(6): 705-10.
[http://dx.doi.org/10.1016/0958-1669(93)90053-Y] [PMID: 7764468]
[79]
Rando TA. Non-viral gene therapy for Duchenne muscular dystrophy: Progress and challenges. Biochim Biophys Acta Mol Basis Dis 2007; 1772(2): 263-71.
[http://dx.doi.org/10.1016/j.bbadis.2006.07.009] [PMID: 17005381]
[80]
Yang J, Liu X, Yu J, et al. A non-viral vector for potential DMD gene therapy study by targeting a minidystrophin-GFP fusion gene into the hrDNA locus. Acta Biochim Biophys Sin (Shanghai) 2009; 41(12): 1053-60.
[http://dx.doi.org/10.1093/abbs/gmp080] [PMID: 20011980]
[81]
Loperfido M, Jarmin S, Dastidar S, et al. piggyBac transposons expressing full-length human dystrophin enable genetic correction of dystrophic mesoangioblasts. Nucleic Acids Res 2016; 44(2): 744-60.
[http://dx.doi.org/10.1093/nar/gkv1464] [PMID: 26682797]
[82]
Xu L, Park KH, Zhao L, et al. CRISPR-mediated genome editing restores dystrophin expression and function in mdx mice. Mol Ther 2016; 24(3): 564-9.
[http://dx.doi.org/10.1038/mt.2015.192] [PMID: 26449883]
[83]
Zuris JA, Thompson DB, Shu Y, et al. Cationic lipid-mediated delivery of proteins enables efficient protein-based genome editing in vitro and in vivo. Nat Biotechnol 2015; 33(1): 73-80.
[http://dx.doi.org/10.1038/nbt.3081] [PMID: 25357182]
[84]
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(11): 889-901.
[http://dx.doi.org/10.1038/s41551-017-0137-2] [PMID: 29805845]
[85]
Aartsma-Rus A, Fokkema I, Verschuuren J, et al. Theoretic applicability of antisense-mediated exon skipping for Duchenne muscular dystrophy mutations. Hum Mutat 2009; 30(3): 293-9.
[http://dx.doi.org/10.1002/humu.20918] [PMID: 19156838]
[86]
Mizobe Y, Miyatake S, Takizawa H, et al. In vivo Evaluation of single-exon and multiexon skipping in mdx52 mice. Methods Mol Biol 2018; 1828: 275-92.
[http://dx.doi.org/10.1007/978-1-4939-8651-4_17] [PMID: 30171548]
[87]
Min YL, Bassel-Duby R, Olson EN. CRISPR correction of duchenne muscular dystrophy. Annu Rev Med 2019; 70(1): 239-55.
[http://dx.doi.org/10.1146/annurev-med-081117-010451] [PMID: 30379597]
[88]
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.
[http://dx.doi.org/10.1126/scitranslmed.aan8081] [PMID: 29187645]
[89]
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.
[http://dx.doi.org/10.1126/science.aau1549] [PMID: 30166439]
[90]
Incitti T, De Angelis FG, Cazzella V, et al. Exon skipping and duchenne muscular dystrophy therapy: Selection of the most active U1 snRNA antisense able to induce dystrophin exon 51 skipping. Mol Ther 2010; 18(9): 1675-82.
[http://dx.doi.org/10.1038/mt.2010.123] [PMID: 20551908]
[91]
Cirak S, Feng L, Anthony K, et al. Restoration of the dystrophin-associated glycoprotein complex after exon skipping therapy in Duchenne muscular dystrophy. Mol Ther 2012; 20(2): 462-7.
[http://dx.doi.org/10.1038/mt.2011.248] [PMID: 22086232]
[92]
Novak JS, Spathis R, Dang UJ, et al. Interrogation of dystrophin and dystroglycan complex protein turnover after exon skipping therapy. J Neuromuscul Dis 2021; 8(s2): S383-402.
[http://dx.doi.org/10.3233/JND-210696] [PMID: 34569969]
[93]
Flanigan KM, Dunn DM, von Niederhausern A, et al. Mutational spectrum of DMD mutations in dystrophinopathy patients: Application of modern diagnostic techniques to a large cohort. Hum Mutat 2009; 30(12): 1657-66.
[http://dx.doi.org/10.1002/humu.21114] [PMID: 19937601]
[94]
Kemaladewi DU, Hoogaars WMH, van Heiningen SH, et al. Dual exon skipping in myostatin and dystrophin for Duchenne muscular dystrophy. BMC Med Genomics 2011; 4(1): 36.
[http://dx.doi.org/10.1186/1755-8794-4-36] [PMID: 21507246]
[95]
Malerba A, Kang JK, McClorey G, et al. Dual myostatin and dystrophin exon skipping by morpholino nucleic acid oligomers conjugated to a cell-penetrating peptide is a promising therapeutic strategy for the treatment of duchenne muscular dystrophy. Mol Ther Nucleic Acids 2012; 1(12): e62.
[http://dx.doi.org/10.1038/mtna.2012.54] [PMID: 23250360]
[96]
Aoki Y, Yokota T, Wood MJA. Development of multiexon skipping antisense oligonucleotide therapy for Duchenne muscular dystrophy. BioMed Res Int 2013; 2013: 1-8.
[http://dx.doi.org/10.1155/2013/402369] [PMID: 23984357]
[97]
Béroud C, Tuffery-Giraud S, Matsuo M, et al. Multiexon skipping leading to an artificial DMD protein lacking amino acids from exons 45 through 55 could rescue up to 63% of patients with Duchenne muscular dystrophy. Hum Mutat 2007; 28(2): 196-202.
[http://dx.doi.org/10.1002/humu.20428] [PMID: 17041910]
[98]
Yazaki M, Yoshida K, Nakamura A, et al. Clinical characteristics of aged Becker muscular dystrophy patients with onset after 30 years. Eur Neurol 1999; 42(3): 145-9.
[http://dx.doi.org/10.1159/000008089] [PMID: 10529540]
[99]
Nakamura A, Yoshida K, Fukushima K, et al. Follow-up of three patients with a large in-frame deletion of exons 45-55 in the Duchenne muscular dystrophy (DMD) gene. J Clin Neurosci 2008; 15(7): 757-63.
[http://dx.doi.org/10.1016/j.jocn.2006.12.012] [PMID: 18261911]
[100]
Nakamura A, Shiba N, Miyazaki D, et al. Comparison of the phenotypes of patients harboring in-frame deletions starting at exon 45 in the Duchenne muscular dystrophy gene indicates potential for the development of exon skipping therapy. J Hum Genet 2017; 62(4): 459-63.
[http://dx.doi.org/10.1038/jhg.2016.152] [PMID: 27974813]
[101]
Heald A, Anderson LVB, Bushby KMD, Shaw PJ. Becker muscular dystrophy with onset after 60 years. Neurology 1994; 44(12): 2388-90.
[http://dx.doi.org/10.1212/WNL.44.12.2388] [PMID: 7991131]
[102]
Ferreiro V, Giliberto F, Muñiz GMN, et al. Asymptomatic Becker muscular dystrophy in a family with a multiexon deletion. Muscle Nerve 2009; 39(2): 239-43.
[http://dx.doi.org/10.1002/mus.21193] [PMID: 19012301]
[103]
Anthony K, Cirak S, Torelli S, et al. Dystrophin quantification and clinical correlations in Becker muscular dystrophy: Implications for clinical trials. Brain 2011; 134(12): 3547-59.
[http://dx.doi.org/10.1093/brain/awr291] [PMID: 22102647]
[104]
van den Bergen JC, Schade van Westrum SM, Dekker L, et al. Clinical characterisation of Becker muscular dystrophy patients predicts favourable outcome in exon-skipping therapy. J Neurol Neurosurg Psychiatry 2014; 85(1): 92-8.
[http://dx.doi.org/10.1136/jnnp-2012-304729] [PMID: 23667215]
[105]
Taglia A, Petillo R, D’Ambrosio P, et al. Clinical features of patients with dystrophinopathy sharing the 45-55 exon deletion of DMD gene. Acta Myol 2015; 34(1): 9-13.
[PMID: 26155064]
[106]
Nakamura A, Fueki N, Shiba N, et al. Deletion of exons 3−9 encompassing a mutational hot spot in the DMD gene presents an asymptomatic phenotype, indicating a target region for multiexon skipping therapy. J Hum Genet 2016; 61(7): 663-7.
[http://dx.doi.org/10.1038/jhg.2016.28] [PMID: 27009627]
[107]
Mori-Yoshimura M, Mitsuhashi S, Nakamura H, et al. Characteristics of japanese patients with becker muscular dystrophy and intermediate muscular dystrophy in a japanese national registry of muscular dystrophy (Remudy): Heterogeneity and clinical variation. J Neuromuscul Dis 2018; 5(2): 193-203.
[http://dx.doi.org/10.3233/JND-170225] [PMID: 29614690]
[108]
Tanihata J, Nagata T, Ito N, et al. Truncated dystrophin ameliorates the dystrophic phenotype of mdx mice by reducing sarcolipin-mediated SERCA inhibition. Biochem Biophys Res Commun 2018; 505(1): 51-9.
[http://dx.doi.org/10.1016/j.bbrc.2018.09.039] [PMID: 30236982]
[109]
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.
[http://dx.doi.org/10.1126/sciadv.aap9004] [PMID: 29404407]
[110]
Jinek M, Chylinski K, Fonfara I, Hauer M, Doudna JA, Charpentier E. A programmable dual-RNA-guided DNA endonuclease in adaptive bacterial immunity. Science 2012; 337(6096): 816-21.
[http://dx.doi.org/10.1126/science.1225829] [PMID: 22745249]
[111]
Cong L, Ran FA, Cox D, et al. Multiplex genome engineering using CRISPR/Cas systems. Science 2013; 339(6121): 819-23.
[http://dx.doi.org/10.1126/science.1231143] [PMID: 23287718]
[112]
Mali P, Yang L, Esvelt KM, et al. RNA-guided human genome engineering via Cas9. Science 2013; 339(6121): 823-6.
[http://dx.doi.org/10.1126/science.1232033] [PMID: 23287722]
[113]
Mendell JR, Rodino-Klapac LR. Duchenne muscular dystrophy: CRISPR/Cas9 treatment. Cell Res 2016; 26(5): 513-4.
[http://dx.doi.org/10.1038/cr.2016.28] [PMID: 26926391]
[114]
Hagan M, Ashraf M, Kim I, Weintraub NL, Tang Y. Effective regeneration of dystrophic muscle using autologous iPSC-derived progenitors with CRISPR-Cas9 mediated precise correction. Med Hypotheses 2018; 110: 97-100.
[http://dx.doi.org/10.1016/j.mehy.2017.11.009] [PMID: 29317080]
[115]
Long C, Amoasii L, Mireault AA, et al. Postnatal genome editing partially restores dystrophin expression in a mouse model of muscular dystrophy. Science 2016; 351(6271): 400-3.
[http://dx.doi.org/10.1126/science.aad5725] [PMID: 26721683]
[116]
Nelson CE, Hakim CH, Ousterout DG, et al. In vivo genome editing improves muscle function in a mouse model of Duchenne muscular dystrophy. Science 2016; 351(6271): 403-7.
[http://dx.doi.org/10.1126/science.aad5143] [PMID: 26721684]
[117]
Tabebordbar M, Zhu K, Cheng JKW, et al. In vivo gene editing in dystrophic mouse muscle and muscle stem cells. Science 2016; 351(6271): 407-11.
[http://dx.doi.org/10.1126/science.aad5177] [PMID: 26721686]
[118]
Rees HA, Liu DR. Base editing: Precision chemistry on the genome and transcriptome of living cells. Nat Rev Genet 2018; 19(12): 770-88.
[http://dx.doi.org/10.1038/s41576-018-0059-1] [PMID: 30323312]
[119]
Sakata RC, Ishiguro S, Mori H, et al. Base editors for simultaneous introduction of C-to-T and A-to-G mutations. Nat Biotechnol 2020; 38(7): 865-9.
[http://dx.doi.org/10.1038/s41587-020-0509-0] [PMID: 32483365]
[120]
Ryu SM, 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): 536-9.
[http://dx.doi.org/10.1038/nbt.4148] [PMID: 29702637]
[121]
Hampton T. DNA prime editing: A new CRISPR-based method to correct most disease-causing mutations. JAMA 2020; 323(5): 405-6.
[http://dx.doi.org/10.1001/jama.2019.21827] [PMID: 32016291]
[122]
Li Y, Chen J, Tsai SQ, Cheng Y. Easy-Prime: A machine learning–based prime editor design tool. Genome Biol 2021; 22(1): 235.
[http://dx.doi.org/10.1186/s13059-021-02458-0] [PMID: 34412673]
[123]
England SB, Nicholson LVB, Johnson MA, et al. Very mild muscular dystrophy associated with the deletion of 46% of dystrophin. Nature 1990; 343(6254): 180-2.
[http://dx.doi.org/10.1038/343180a0] [PMID: 2404210]
[124]
Wells DJ, Wells KE, Asante EA, et al. Expression of human full-length and minidystrophin in transgenic mdx mice: Implications for gene therapy of Duchenne muscular dystrophy. Hum Mol Genet 1995; 4(8): 1245-50.
[http://dx.doi.org/10.1093/hmg/4.8.1245] [PMID: 7581360]
[125]
Scott JM, Li S, Harper SQ, et al. Viral vectors for gene transfer of micro-, mini-, or full-length dystrophin. Neuromuscul Disord 2002; 12 (Suppl. 1): S23-9.
[http://dx.doi.org/10.1016/S0960-8966(02)00078-0] [PMID: 12206791]
[126]
Fabb SA, Wells DJ, Serpente P, Dickson G. Adeno-associated virus vector gene transfer and sarcolemmal expression of a 144 kDa micro-dystrophin effectively restores the dystrophin-associated protein complex and inhibits myofibre degeneration in nude/mdx mice. Hum Mol Genet 2002; 11(7): 733-41.
[http://dx.doi.org/10.1093/hmg/11.7.733] [PMID: 11929846]
[127]
Yue Y, Li Z, Harper SQ, Davisson RL, Chamberlain JS, Duan D. Microdystrophin gene therapy of cardiomyopathy restores dystrophin-glycoprotein complex and improves sarcolemma integrity in the mdx mouse heart. Circulation 2003; 108(13): 1626-32.
[http://dx.doi.org/10.1161/01.CIR.0000089371.11664.27] [PMID: 12952841]
[128]
Duchenne. The pathology of paralysis with muscular degeneration (paralysie myosclerotique), or paralysis with apparent hypertrophy. BMJ 1867; 2(363): 541-2.
[http://dx.doi.org/10.1136/bmj.2.363.541] [PMID: 20744949]
[129]
Olson EN. Toward the correction of muscular dystrophy by gene editing. Proc Natl Acad Sci USA 2021; 118(22): e2004840117.
[http://dx.doi.org/10.1073/pnas.2004840117] [PMID: 34074727]
[130]
Wilton-Clark H, Yokota T. Antisense and gene therapy options for duchenne muscular dystrophy arising from mutations in the N-terminal hotspot. Genes (Basel) 2022; 13(2): 257.
[http://dx.doi.org/10.3390/genes13020257] [PMID: 35205302]
[131]
Ilyinskii PO, Michaud AM, Rizzo GL, et al. ImmTOR nanoparticles enhance AAV transgene expression after initial and repeat dosing in a mouse model of methylmalonic acidemia. Mol Ther Methods Clin Dev 2021; 22: 279-92.
[http://dx.doi.org/10.1016/j.omtm.2021.06.015] [PMID: 34485611]
[132]
Boutin S, Monteilhet V, Veron P, et al. Prevalence of serum IgG and neutralizing factors against adeno-associated virus (AAV) types 1, 2, 5, 6, 8, and 9 in the healthy population: Implications for gene therapy using AAV vectors. Hum Gene Ther 2010; 21(6): 704-12.
[http://dx.doi.org/10.1089/hum.2009.182] [PMID: 20095819]
[133]
Chicoine LG, Montgomery CL, Bremer WG, et al. Plasmapheresis eliminates the negative impact of AAV antibodies on microdystrophin gene expression following vascular delivery. Mol Ther 2014; 22(2): 338-47.
[http://dx.doi.org/10.1038/mt.2013.244] [PMID: 24196577]

Rights & Permissions Print Cite
© 2024 Bentham Science Publishers | Privacy Policy