Systemic Delivery of Dysferlin Overlap Vectors Provides Long-Term Gene Expression and Functional Improvement for Dysferlinopathy

Rachael A Potter, Danielle A Griffin, Patricia C Sondergaard, Ryan W Johnson, Eric R Pozsgai, Kristin N Heller, Ellyn L Peterson, Kimmo K Lehtimäki, Hillarie P Windish, Plavi J Mittal, Douglas E Albrecht, Jerry R Mendell, Louise R Rodino-Klapac, Rachael A Potter, Danielle A Griffin, Patricia C Sondergaard, Ryan W Johnson, Eric R Pozsgai, Kristin N Heller, Ellyn L Peterson, Kimmo K Lehtimäki, Hillarie P Windish, Plavi J Mittal, Douglas E Albrecht, Jerry R Mendell, Louise R Rodino-Klapac

Abstract

Dysferlinopathies comprise a family of disorders caused by mutations in the dysferlin (DYSF) gene, leading to a progressive dystrophy characterized by chronic muscle fiber loss, fat replacement, and fibrosis. To correct the underlying histopathology and function, expression of full-length DYSF is required. Dual adeno-associated virus vectors have been developed, defined by a region of homology, to serve as a substrate for reconstitution of the full 6.5 kb dysferlin cDNA. Previous work studied the efficacy of this treatment through intramuscular and regional delivery routes. To maximize clinical efficacy, dysferlin-deficient mice were treated systemically to target all muscles through the vasculature for efficacy and safety studies. Mice were evaluated at multiple time points between 4 and 13 months post treatment for dysferlin expression and functional improvement using magnetic resonance imaging and magnetic resonance spectroscopy and membrane repair. A systemic dose of 6 × 1012 vector genomes resulted in widespread gene expression in the muscles. Treated muscles showed a significant decrease in central nucleation, collagen deposition, and improvement of membrane repair to wild-type levels. Treated gluteus muscles were significantly improved compared to placebo-treated muscles and were equivalent to wild type in volume, intra- and extramyocellular lipid accumulation, and fat percentage using magnetic resonance imaging and magnetic resonance spectroscopy. Dual-vector treatment allows for production of full-length functional dysferlin with no toxicity. This confirms previous safety data and validates translation of systemic gene delivery for dysferlinopathy patients.

Keywords: AAV; LGMD2B; dysferlin; gene therapy; systemic delivery.

Conflict of interest statement

Dr. Rodino-Klapac is the inventor of the AAV.dysferlin dual vector technology. This technology has been licensed exclusively to Myonexus Therapeutics, Inc. Dr. Rodino-Klapac is a co-founder and Chief Scientific Officer for Myonexus Therapeutics, Inc. No competing financial interests exist for the remaining authors.

Figures

Figure 1.
Figure 1.
Sustained long-term expression following intramuscular delivery in dysferlin null mice. Four-week-old Dysf–/– mice were injected with 2 × 1011 vg total dose of AAVrh74.MHCK7.DYSF.DV into the left tibialis anterior (TA) muscle and followed for 3–24 months to monitor expression and histology (n = 3 per time point). (A) Hematoxylin and eosin (H&E) staining demonstrates a significant reduction in centrally located nuclei in the treated TA compared to (B) contralateral control TA muscles injected with saline. (C) Anti-dysferlin immunofluorescence staining demonstrates dysferlin transgene expression was sustained throughout all time points (3–24 months). (D) At 6, 9, 12, 18, and 24 months post delivery, there was a significant decrease in central nucleation to <5%. Excluding the 3 month time point, all values were significant (p ≤ 0.01) for central nucleation quantification. (E) There was no significant decrease in the percentage of dysferlin-positive fivers over time, and contralateral (right side) muscles demonstrated no dysferlin expression. Two-way analyses of variance (ANOVAs) were utilized to identify differences in the means between treated and untreated cohorts. Error bars are represented as mean ± standard error of the mean (SEM; p < 0.05).
Figure 2.
Figure 2.
Intravenous delivery of AAVrh74.MHCK7.DYSF.DV improves histological parameters in BlaJ mice. (A) H&E staining of the psoas muscle 28 weeks post treatment demonstrated improvement in fiber size distribution and reduced central nuclei following intravenous delivery of 6 × 1012 vg total dose of AAVrh74.MHCK7.DYSF.DV in treated BlaJ mice versus saline-treated mice. (B) Picosirius red staining for collagen demonstrated reduction in fibrosis in AAVrh74.MHCK7.DYSF.DV-treated psoas muscles. (C) Following AAVrh74.MHCK7.DYSF.DV delivery, there was a trend toward normal fiber diameters, with a significant increase in mean fiber diameter in treated muscle (27.8 ± 0.17 μm) versus control (18.4 ± 0.099 μm), and reduced central nucleation in the TA, quadriceps, triceps, and psoas muscles. (E) Additionally, there was a significant decrease from 37.0% to 9.7% collagen deposition in treated mice compared to saline-treated mice. Two-way ANOVAs were utilized with post hoc analysis to locate differences in central nucleation and fiber diameters. Error bars are represented as mean ± SEM (p < 0.05).
Figure 3.
Figure 3.
Long-term improvement in collagen deposition and immune staining with AAVrh74.MHCK7.DYSF.DV treatment. (A) At 15 months of age, H&E staining demonstrated normalization of histology in treated psoas muscles (BlaJ-AAV). (B) Picosirius staining in the gluteus muscle demonstrated a reduction in collagen deposition. (C) Following long-term treatment in systemically treated mice, there was a trend toward normalization of fiber diameters and (D) reduced centrally located nuclei in the quadriceps muscle, as shown with H&E staining. (E) Finally, there was a 9.0% reduction in collagen deposition using picosirius staining in the gluteus muscle. One-way ANOVAs were utilized with post hoc analysis to locate differences in the means. Error bars are represented as mean ± SEM (p < 0.05).
Figure 4.
Figure 4.
Long-term expression and normalization of centrally located nuclei in AAVrh74.MHCK7.DYSF.DV-treated mice. (A) Robust dysferlin expression was seen following systemic delivery (6 × 1012 dose) using anti-dysferlin immune staining with an N-terminal antibody. Gastrocnemius (GAS) and TA muscles are shown for representation. (B) Sections were co-stained with DAPI to quantify dysferlin positive fibers co-localized with centrally located nuclei. (C) There was widespread dysferlin transgene expression at 15 months of age >5% in all tissues, with the diaphragm expressing the highest at 53%. (D) It is demonstrated that the majority of fibers with centrally located nuclei are absent of dysferlin expression (<0.5% are positive for dysferlin and centrally located nuclei). (E) Full-length dysferlin in treated muscles at 15 months of age was confirmed by Western blot analysis (TA and GAS) compared to wild-type and BlaJ-untreated muscle.
Figure 5.
Figure 5.
Long-term improvements using magnetic resonance imaging (MRI) in systemically treated mice with AAVrh74.MHCK7.DYSF.DV. (A) Animals underwent MRI after 7, 9, 12, and 15 months of age and demonstrated improvements in gluteus volume (p < 0.001), (B) decreased gluteus fat (p < 0.0001), (C) increased water content to wild-type levels (p = 0.223; p < 0.01), and (D) decreased fat percentage significantly at the 15-month time points (p < 0.0001). (E) MR-spectroscopy was used to evaluate metabolite accumulation and decreased extramyocellular lipid accumulation was demonstrated in the gluteus muscle (p < 0.0001), as well as decreased creatine levels in the muscle to wild-type levels (p = 0.021) (F). Fifteen-month-old BLAJ animals were unable to undergo magnetic resonance spectroscopy evaluation due to excessive fat infiltration in the saline-treated cohort. Two-way ANOVAs with Tukey post hoc analysis were utilized to locate differences in the means. Error bars are represented as mean ± SEM (p < 0.05).
Figure 6.
Figure 6.
Systemic expression and histological improvements in an aged cohort treated with AAVrh74.MHCK7.DYSF.DV. (A) Animals treated at 6 months of age and sacrificed at 9 months of age demonstrated reductions in centrally located nuclei in psoas muscle using H&E staining. (B) There was slightly reduced collagen deposition between 6 and 9 months of age with treatment. (C) Dysferlin transgene expression was noted across all muscles following AAV treatment in this aged cohort, which led to significant improvement in membrane repair capacity in flexor digitorum brevis muscles (p < 0.05 at 190 s).
Figure 7.
Figure 7.
Systemic delivery in rhesus macaque with AAVrh74.MHCK7.DYSF.DV. (A) Anti-dysferlin immunofluorescence staining in untreated naïve rhesus macaque diaphragm and quadriceps muscles (left panel) and systemically treated diaphragm, quadriceps, and hamstring demonstrates robust dysferlin expression. (B) Enzyme-linked immunosorbent spot assay analysis of T-cell response to dysferlin and AAV peptide pools demonstrates no adverse T-cell response in the presence of (C) widespread vector genome (vg/μg of genomic DNA) biodistribution quantified using quantitative polymerase chain reaction (qPCR). Vastus lateralis control and heart control in qPCR biodistribution refers to an uninjected male rhesus macaque.

References

    1. Bansal D, Miyake K, Vogel SS, et al. . Defective membrane repair in dysferlin-deficient muscular dystrophy. Nature 2003;423:168–172
    1. Moore SA, Shilling CJ, Westra S, et al. . Limb-girdle muscular dystrophy in the United States. J Neuropathol Exp Neurol 2006;65:995–1003
    1. Rosales XQ, Gastier-Foster JM, Lewis S, et al. . Novel diagnostic features of dysferlinopathies. Muscle Nerve 2010;42:14–21
    1. Rosales XQ, Moser SJ, Tran T, et al. . Cardiovascular magnetic resonance of cardiomyopathy in limb girdle muscular dystrophy 2B and 2I. J Cardiovasc Magn Reson 2011;13:39.
    1. Emmerling VV, Fischer S, Stiefel F, et al. . Temperature-sensitive miR-483 is a conserved regulator of recombinant protein and viral vector production in mammalian cells. Biotechnol Bioeng 2016;113:830–841
    1. Aoki M, Liu J, Richard I, et al. . Genomic organization of the dysferlin gene and novel mutations in Miyoshi myopathy. Neurology 2001;57:271–278
    1. Liu J, Aoki M, Illa I, et al. . Dysferlin, a novel skeletal muscle gene, is mutated in Miyoshi myopathy and limb girdle muscular dystrophy. Nat Genet 1998;20:31–36
    1. Liu J, Wu C, Bossie K, et al. . Generation of a 3-Mb PAC contig spanning the Miyoshi myopathy/limb-girdle muscular dystrophy (MM/LGMD2B) locus on chromosome 2p13. Genomics 1998;49:23–29
    1. Grose WE, Clark KR, Griffin D, et al. . Homologous recombination mediates functional recovery of dysferlin deficiency following AAV5 gene transfer. PLoS One 2012;7:e39233.
    1. Lennon NJ, Kho A, Bacskai BJ, et al. . Dysferlin interacts with annexins A1 and A2 and mediates sarcolemmal wound-healing. J Biol Chem 2003;278:50466–50473
    1. Sondergaard PC, Griffin D, Pozsgai E, et al. . AAV.dysferlin overlap vectors restore function in dysferlinopathy animal models. Ann Clin Transl Neurol 2015;2:256–270
    1. Kerr J, Ward C, Bloch R. Dysferlin at transverse tubules regulates Ca2+ homeostasis in skeletal muscle. Front Physiol 2014;5.
    1. Lek A, Evesson F, Sutton B, et al. . Ferlins: regulators of vesicle fusion for auditory neurotransmission, receptor trafficking and membrane repair. Traffic 2012;13:185–194
    1. Oulhen N, Onorato T, Ramos I, et al. . Dysferlin is essential for endocytosis in the sea star oocyte. Dev Biol 2014;388:94–102
    1. Chiu YH, Hornsey MA, Klinge L, et al. . Attenuated muscle regeneration is a key factor in dysferlin-deficient muscular dystrophy. Hum Mol Genet 2009;18:1976–1989
    1. de Morree A, Flix B, Bagaric I, et al. . Dysferlin regulates cell adhesion in human monocytes. J Biol Chem 2013;288:14147–14157
    1. Mariano A, Henning A, Han R. Dysferlin-deficient muscular dystrophy and innate immune activation. FEBS J 2013;280:4165–4176
    1. Millay DP, Maillet M, Roche JA, et al. . Genetic manipulation of dysferlin expression in skeletal muscle: novel insights into muscular dystrophy. Am J Pathol 2009;175:1817–1823
    1. Lostal W, Bartoli M, Roudaut C, et al. . Lack of correlation between outcomes of membrane repair assay and correction of dystrophic changes in experimental therapeutic strategy in dysferlinopathy. PLoS One 2012;7:e38036.
    1. Walter MC, Reilich P, Thiele S, et al. . Treatment of dysferlinopathy with deflazacort: a double-blind, placebo-controlled clinical trial. Orphanet J Rare Dis 2013;8:26.
    1. Gallardo E, Rojas-Garcia R, de Luna N, et al. . Inflammation in dysferlin myopathy: immunohistochemical characterization of 13 patients. Neurology 2001;57:2136–2138
    1. Fanin M, Angelini C. Muscle pathology in dysferlin deficiency. Neuropathol Appl Neurobiol 2002;28:461–470
    1. Barthelemy F, Wein N, Krahn M, et al. . Translational research and therapeutic perspectives in dysferlinopathies. Mol Med 2011;17:875–882
    1. He B, Tang RH, Weisleder N, et al. . Enhancing muscle membrane repair by gene delivery of MG53 ameliorates muscular dystrophy and heart failure in delta-Sarcoglycan-deficient hamsters. Mol Ther 2012;20:727–735
    1. Krahn M, Wein N, Bartoli M, et al. . A naturally occurring human minidysferlin protein repairs sarcolemmal lesions in a mouse model of dysferlinopathy. Sci Transl Med 2010;2:50ra69
    1. Lostal W, Bartoli M, Bourg N, et al. . Efficient recovery of dysferlin deficiency by dual adeno-associated vector-mediated gene transfer. Hum Mol Genet 2010;19:1897–1907
    1. Hofhuis J, Bersch K, Bussenschutt R, et al. . Dysferlin mediates membrane tubulation and links T-tubule biogenesis to muscular dystrophy. J Cell Sci 2017;130:841–852
    1. Chicoine LG, Rodino-Klapac LR, Shao G, et al. . Vascular delivery of rAAVrh74.MCK.GALGT2 to the gastrocnemius muscle of the rhesus macaque stimulates the expression of dystrophin and laminin alpha2 surrogates. Mol Ther 2014;22:713–724
    1. Gao G, Vandenberghe LH, Wilson JM. New recombinant serotypes of AAV vectors. Curr Gene Ther 2005;5:285–297
    1. Heller KN, Montgomery CL, Janssen PM, et al. . AAV-mediated overexpression of human alpha7 integrin leads to histological and functional improvement in dystrophic mice. Mol Ther 2013;21:520–525
    1. Sahenk Z, Galloway G, Clark KR, et al. . AAV1.NT-3 gene therapy for Charcot–Marie–Tooth neuropathy. Mol Ther 2014;22:511–521
    1. Grieger JC, Soltys SM, Samulski RJ. Production of recombinant adeno-associated virus vectors using suspension HEK293 cells and continuous harvest of vector from the culture media for GMP FIX and FLT1 clinical vector. Mol Ther 2016;24:287–297
    1. Emmerling VV, Pegel A, Milian EG, et al. . Rational plasmid design and bioprocess optimization to enhance recombinant adeno-associated virus (AAV) productivity in mammalian cells. Biotechnol J 2016;11:290–297
    1. Qu W, Wang M, Wu Y, et al. . Scalable downstream strategies for purification of recombinant adeno- associated virus vectors in light of the properties. Curr Pharm Biotechnol 2015;16:684–695
    1. Foust KD, Wang X, McGovern VL, et al. . Rescue of the spinal muscular atrophy phenotype in a mouse model by early postnatal delivery of SMN. Nat Biotechnol 2010;28:271–274
    1. Wang D, Zhong L, Nahid MA, et al. . The potential of adeno-associated viral vectors for gene delivery to muscle tissue. Expert Opin Drug Deliv 2014;11:345–364
    1. Rodino-Klapac LR, Janssen PM, Montgomery CL, et al. . A translational approach for limb vascular delivery of the micro-dystrophin gene without high volume or high pressure for treatment of Duchenne muscular dystrophy. J Transl Med 2007;5:45.
    1. Pozsgai ER, Griffin DA, Heller KN, et al. . Systemic AAV-mediated beta-sarcoglycan delivery targeting cardiac and skeletal muscle ameliorates histological and functional deficits in LGMD2E mice. Mol Ther 2017;25:855–869
    1. Clark KR, Liu X, McGrath JP, et al. . Highly purified recombinant adeno-associated virus vectors are biologically active and free of detectable helper and wild-type viruses. Hum Gene Ther 1999;10:1031–1039
    1. Ghosh A, Yue Y, Long C, et al. . Efficient whole-body transduction with trans-splicing adeno-associated viral vectors. Mol Ther 2007;15:750–755
    1. Emmerling VV, Fischer S, Kleemann M, et al. . miR-483 is a self-regulating microRNA and can activate its own expression via USF1 in HeLa cells. Int J Biochem Cell Biol 2016;80:81–86
    1. Toromanoff A, Cherel Y, Guilbaud M, et al. . Safety and efficacy of regional intravenous (RI) versus intramuscular (IM) delivery of rAAV1 and rAAV8 to nonhuman primate skeletal muscle. Mol Ther 2008;16:1291–1299
    1. Rodino-Klapac LR, Montgomery CL, Bremer WG, et al. . Persistent expression of FLAG-tagged micro dystrophin in nonhuman primates following intramuscular and vascular delivery. Mol Ther 2010;18:109–117
    1. Schnepp BC, Jensen RL, Chen CL, et al. . Characterization of adeno-associated virus genomes isolated from human tissues. J Virol 2005;79:14793–14803
    1. Labikova J, Vcelakova J, Ulmannova T, et al. . The cytokine production of peripheral blood mononuclear cells reflects the autoantibody profile of patients suffering from type 1 diabetes. Cytokine 2014;69:189–195
    1. Roche JA, Ru LW, O'Neill AM, et al. . Unmasking potential intracellular roles for dysferlin through improved immunolabeling methods. J Histochem Cytochem 2011;59:964–975
    1. Vontzalidis A, Terzis G, Manta P. Increased dysferlin expression in Duchenne muscular dystrophy. Anal Quant Cytopathol Histpathol 2014;36:15–22
    1. Nagy N, Nonneman RJ, Llanga T, et al. . Hip region muscular dystrophy and emergence of motor deficits in dysferlin-deficient Bla/J mice. Physiol Rep 2017;5

Source: PubMed

3
Abonneren