1. |
Ryder S, Leadley RM, Armstrong N, et al. The burden, epidemiology, costs and treatment for Duchenne muscular dystrophy: an evidence review. Orphanet J Rare Dis, 2017, 12(1): 79.
|
2. |
Okubo M, Minami N, Goto K, et al. Genetic diagnosis of Duchenne/Becker muscular dystrophyusing next-generation sequencing: validation analysis of DMD mutations. J Hum Genet, 2016, 61(6): 483-489.
|
3. |
ansović I, Barišić I, Dumić K. Improved detection of deletions and duplications in the DMD gene using the multiplex ligation-dependent probe amplification (MLPA) method. Biochem Genet, 2013, 51(3/4): 189-201.
|
4. |
Ankala A, da Silva C, Gualandi F, et al. A comprehensive genomic approach for neuromuscular diseases gives a high diagnostic yield. Ann Neurol, 2015, 77(2): 206-214.
|
5. |
Chamberlain JS, Metzger J, Reyes M, et al. Dystrophin-deficient mdx mice display a reduced life span and are susceptible to spontaneous rhabdomyosarcoma. FASEB J, 2007, 21(9): 2195-2204.
|
6. |
McGreevy JW, Hakim CH, McIntosh MA, et al. Animal models of Duchenne muscular dystrophy: from basic mechanisms to gene therapy. Dis Model Mech, 2015, 8(3): 195-213.
|
7. |
Duan D. Systemic AAV micro-dystrophin gene therapy for Duchenne muscular dystrophy. Mol Ther, 2018, 26(10): 2337-2356.
|
8. |
Leborgne C, Latournerie V, Boutin S, et al. Prevalence and long-term monitoring of humoral immunity against adeno-associated virus in Duchenne muscular dystrophy patients. Cell Immunol, 2019, 342: 103780.
|
9. |
Niks EH, Aartsma-Rus A. Exon skipping: a first in class strategy for Duchenne muscular dystrophy. Expert Opin Biol Ther, 2017, 17(2): 225-236.
|
10. |
Echigoya Y, Lim KRQ, Nakamura A, et al. Multiple exon skipping in the Duchenne muscular dystrophy hot spots: prospects and challenges. J Pers Med, 2018, 8(4): 41.
|
11. |
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.
|
12. |
Aartsma-Rus A, Krieg AM. FDA approves eteplirsen for Duchenne muscular dystrophy: the next chapter in the eteplirsen saga. Nucleic Acid Ther, 2017, 27(1): 1-3.
|
13. |
Charleston JS, Schnell FJ, Dworzak J, et al. Eteplirsen treatment for Duchenne muscular dystrophy: exon skipping and dystrophin production. Neurology, 2018, 90(24): e2146-e2154.
|
14. |
Goemans N, Mercuri E, Belousova E, et al. A randomized placebo-controlled phase 3 trial of an antisense oligonucleotide, drisapersen, in Duchenne muscular dystrophy. Neuromuscul Disord, 2018, 28(1): 4-15.
|
15. |
Lim KRQ, Yoon C, Yokota T. Applications of CRISPR/Cas9 for the treatment of Duchenne muscular dystrophy. J Pers Med, 2018, 8(4): 38.
|
16. |
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: 14454.
|
17. |
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): 400-403.
|
18. |
Hakim CH, Wasala NB, Nelson CE, et al. AAV CRISPR editing rescues cardiac and muscle function for 18 months in dystrophic mice. JCI insight, 2018, 3(23): e124297.
|
19. |
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.
|
20. |
Kemaladewi DU, Cohn RD. Exon snipping in Duchenne muscular dystrophy. Trends Mol Med, 2016, 22(3): 187-189.
|
21. |
El Refaey M, Xu L, Gao Y, et al. In vivo genome editing restores dystrophin expression and cardiac function in dystrophic mice. Circ Res, 2017, 121(8): 923-929.
|
22. |
Maggio I, Liu J, Janssen JM, et al. Adenoviral vectors encoding CRISPR/Cas9 multiplexes rescue dystrophin synthesis in unselected populations of DMD muscle cells. Sci Rep, 2016, 6: 37051.
|
23. |
Arnett AL, Konieczny P, Ramos JN, et al. Adeno-associated viral (AAV) vectors do not efficiently target muscle satellite cells. Mol Ther Methods Clin Dev, 2014, 1: 14038.
|
24. |
Aartsma-Rus A, Ginjaar IB, Bushby K. The importance of genetic diagnosis for Duchenne muscular dystrophy. J Med Genet, 2016, 53(3): 145-151.
|
25. |
Karijolich J, Yu YT. Therapeutic suppression of premature termination codons: mechanisms and clinical considerations (review). Int J Mol Med, 2014, 34(2): 355-362.
|
26. |
Welch EM, Barton ER, Zhuo J, et al. PTC124 targets genetic disorders caused by nonsense mutations. Nature, 2007, 447(7140): 87-91.
|
27. |
McDonald CM, Campbell C, Torricelli RE, et al. Ataluren in patients with nonsense mutation Duchenne muscular dystrophy (ACT DMD): a multicentre, randomised, double-blind, placebo-controlled, phase 3 trial. Lancet, 2017, 390(10101): 1489-1498.
|
28. |
Skuk D, Tremblay JP. Cell therapy in muscular dystrophies: many promises in mice and dogs, few facts in patients. Expert Opin Biol Ther, 2015, 15(9): 1307-1319.
|
29. |
Young CS, Hicks MR, Ermolova NV, et al. A single CRISPR-Cas9 deletion strategy that targets the majority of DMD patients restores dystrophin function in hiPSC-derived muscle cells. Cell Stem Cell, 2016, 18(4): 533-540.
|
30. |
Cossu G, Previtali SC, Napolitano S, et al. Intra-arterial transplantation of HLA-matched donor mesoangioblasts in Duchenne muscular dystrophy. EMBO Mol Med, 2016, 8(12): 1470-1471.
|
31. |
Kennedy TL, Guiraud S, Edwards B, et al. Micro-utrophin improves cardiac and skeletal muscle function of severely affected D2/mdx mice. Mol Ther Methods Clin Dev, 2018, 11: 92-105.
|
32. |
Shin J, Tajrishi MM, Ogura Y, et al. Wasting mechanisms in muscular dystrophy. Int J Biochem Cell Biol, 2013, 45(10): 2266-2279.
|
33. |
McDonald CM, Henricson EK, Abresch RT, et al. Long-term effects of glucocorticoids on function, quality of life, and survival in patients with Duchenne muscular dystrophy: a prospective cohort study. Lancet, 2018, 391(10119): 451-461.
|
34. |
Newton R, Holden NS. Separating transrepression and transactivation: a distressing divorce for the glucocorticoid receptor?. Mol Pharmacol, 2007, 72(4): 799-809.
|
35. |
Yoon SH, Grynpas MD, Mitchell J. Growth hormone increases bone toughness and decreases muscle inflammation in glucocorticoid-treated mdx mice, model of Duchenne muscular dystrophy. J Bone Miner Res, 2019, 34(8): 1473-1486.
|
36. |
Baudy AR, Reeves EK, Damsker JM, et al. Δ-9, 11 modification of glucocorticoids dissociates nuclear factor-κB inhibitory efficacy from glucocorticoid response element-associated side effects. J Pharmacol Exp Ther, 2012, 343(1): 225-232.
|
37. |
Hoffman EP, Riddle V, Siegler MA, et al. Phase 1 trial of vamorolone, a first-in-class steroid, shows improvements in side effects via biomarkers bridged to clinical outcomes. Steroids, 2018, 134: 43-52.
|
38. |
Hoffman EP, Schwartz BD, Mengle-Gaw LJ, et al. Vamorolone trial in Duchenne muscular dystrophy shows dose-related improvement of muscle function. Neurology, 2019, 93(13): e1312-e1323.
|
39. |
Signorelli M, Ayoglu B, Johansson C, et al. Longitudinal serum biomarker screening identifies malate dehydrogenase 2 as candidate prognostic biomarker for Duchenne muscular dystrophy. J Cachexia Sarcopenia Muscle, 2020, 11(2): 505-517.
|
40. |
Chrzanowski SM, Darras BT, Rutkove SB. The value of imaging and composition-based biomarkers in Duchenne muscular dystrophy clinical trials. Neurotherapeutics, 2020, 17(1): 142-152.
|
41. |
Leitner ML, Kapur K, Darras BT, et al. Electrical impedance myography for reducing sample size in Duchenne muscular dystrophy trials. Ann Clin Transl Neurol, 2020, 7(1): 4-14.
|
42. |
Regensburger AP, Fonteyne LM, Jüngert J, et al. Detection of collagens by multispectral optoacoustic tomography as an imaging biomarker for Duchenne muscular dystrophy. Nat Med, 2019, 25(12): 1905-1915.
|
43. |
Barnard AM, Willcocks RJ, Triplett WT, et al. MR biomarkers predict clinical function in Duchenne muscular dystrophy. Neurology, 2020, 94(9): e897-e909.
|