Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Mol Ther Methods Clin Dev ; 32(1): 101195, 2024 Mar 14.
Artículo en Inglés | MEDLINE | ID: mdl-38327805

RESUMEN

Patients with pre-existing immunity to adeno-associated virus (AAV) are currently unable to receive systemic gene transfer therapies. In this nonhuman primate study, we investigated the impact of immunosuppression strategies on gene transfer therapy safety and efficacy and analyzed plasmapheresis as a potential pretreatment for circumvention of pre-existing immunity or redosing. In part 1, animals received delandistrogene moxeparvovec (SRP-9001), an AAVrh74-based gene transfer therapy for Duchenne muscular dystrophy. Cohort 1 (control, n = 2) received no immunosuppression; cohorts 2-4 (n = 3 per cohort) received prednisone at different time points; and cohort 5 (n = 3) received rituximab, sirolimus, and prednisone before and after dosing. In part 2, cohorts 2-4 underwent plasmapheresis before redosing; cohort 5 was redosed without plasmapheresis. We analyzed safety, immune response (humoral and cell-mediated responses and complement activation), and vector genome distribution. After 2 or 3 plasmapheresis exchanges, circulating anti-AAVrh74 antibodies were reduced, and animals were redosed. Plasmapheresis was well tolerated, with no abnormal clinical or immunological observations. Cohort 5 (redosed with high anti-AAVrh74 antibody titers) had hypersensitivity reactions, which were controlled with treatment. These findings suggest that plasmapheresis is a safe and effective method to reduce anti-AAV antibody levels in nonhuman primates prior to gene transfer therapy. The results may inform human studies involving redosing or circumvention of pre-existing immunity.

2.
Nat Med ; 30(1): 199-206, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38177855

RESUMEN

Limb-girdle muscular dystrophy 2E/R4 is caused by mutations in the ß-sarcoglycan (SGCB) gene, leading to SGCB deficiency and consequent muscle loss. We developed a gene therapy approach based on functional replacement of the deficient SCB protein. Here we report interim results from a first-in-human, open-label, nonrandomized, phase 1/2 trial evaluating the safety and efficacy of bidridistrogene xeboparvovec, an adeno-associated virus-based gene therapy containing a codon-optimized, full-length human SGCB transgene. Patients aged 4-15 years with confirmed SGCB mutations at both alleles received one intravenous infusion of either 1.85 × 1013 vector genome copies kg-1 (Cohort 1, n = 3) or 7.41 × 1013 vector gene copies kg-1 (Cohort 2, n = 3). Primary endpoint was safety, and secondary endpoint was change in SGCB expression in skeletal muscle from baseline to Day 60. We report interim Year 2 results (trial ongoing). The most frequent treatment-related adverse events were vomiting (four of six patients) and gamma-glutamyl transferase increase (three of six patients). Serious adverse events resolved with standard therapies. Robust SGCB expression was observed: Day 60 mean (s.d.) percentage of normal expression 36.2% (2.7%) in Cohort 1 and 62.1% (8.7%) in Cohort 2. Post hoc exploratory analysis showed preliminary motor improvements using the North Star Assessment for Limb-girdle Type Muscular Dystrophies maintained through Year 2. The 2-year safety and efficacy of bidridistrogene xeboparvovec support clinical development advancement. Further studies are necessary to confirm the long-term safety and efficacy of this gene therapy. ClinicalTrials.gov registration: NCT03652259 .


Asunto(s)
Distrofia Muscular de Cinturas , Sarcoglicanopatías , Humanos , Distrofia Muscular de Cinturas/genética , Distrofia Muscular de Cinturas/metabolismo , Distrofia Muscular de Cinturas/terapia , Sarcoglicanopatías/genética , Sarcoglicanopatías/metabolismo , Sarcoglicanopatías/terapia , Músculo Esquelético/metabolismo , Terapia Genética/efectos adversos , Terapia Genética/métodos
3.
Muscle Nerve ; 69(1): 93-98, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-37577753

RESUMEN

INTRODUCTION/AIMS: Delandistrogene moxeparvovec is indicated in the United States for the treatment of ambulatory pediatric patients aged 4 through 5 years with Duchenne muscular dystrophy (DMD) with a confirmed mutation in the DMD gene. Long-term delandistrogene moxeparvovec microdystrophin protein (a shortened dystrophin that retains key functional domains of the wild-type protein) expression may positively alter disease progression in patients with DMD. We evaluated long-term safety and functional outcomes of delandistrogene moxeparvovec in patients with DMD. METHODS: An open-label, phase 1/2a, nonrandomized controlled trial (Study 101; NCT03375164) enrolled ambulatory males, ≥4 to <8 years old, with DMD. Patients received a single intravenous infusion (2.0 × 1014 vg/kg by supercoiled quantitative polymerase chain reaction) of delandistrogene moxeparvovec and prednisone (1 mg/kg/day) 1 day before to 30 days after treatment. The primary endpoint was safety. Functional outcomes were change from baseline in North Star Ambulatory Assessment (NSAA) and timed function tests. RESULTS: Four patients (mean age, 5.1 years) were enrolled. There were 18 treatment-related adverse events; all occurred within 70 days posttreatment and resolved. Mean NSAA total score increased from 20.5 to 27.5, baseline to year 4, with a mean (standard deviation) change of +7.0 (2.9). Post hoc analysis demonstrated a statistically significant and clinically meaningful 9-point difference in NSAA score, relative to a propensity-score-weighted external control cohort (least-squares mean [standard error] = 9.4 [3.4]; P = .0125). DISCUSSION: Gene transfer therapy with delandistrogene moxeparvovec treatment is well tolerated, with a favorable safety profile. Functional improvements are sustained through 4 years, suggesting delandistrogene moxeparvovec may positively alter disease progression.


Asunto(s)
Distrofia Muscular de Duchenne , Niño , Preescolar , Humanos , Masculino , Progresión de la Enfermedad , Terapia Genética/efectos adversos , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/terapia , Distrofia Muscular de Duchenne/metabolismo , Prednisona/uso terapéutico
4.
Biol Open ; 12(9)2023 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-37670674

RESUMEN

Robust expression of shortened, functional dystrophin provided impetus to develop adeno-associated virus (AAV)-based constructs for clinical application. Because several cassettes are being tested in clinical trials, this study compared the efficacies of four shortened dystrophin-promoter combinations with implications for outcomes in clinical trials: MHCK7 or MCK promoter with a shortened dystrophin transgene containing the N-terminus and spectrin repeats R1, R2, R3 and R24 (rAAVrh74.MHCK7.micro-dystrophin and rAAVrh74.MCK.micro-dystrophin, respectively); shortened dystrophin construct containing the neuronal nitric oxide (nNOS) binding site (rAAVrh74.MHCK7.DV.mini-dystrophin); and shortened dystrophin containing the C-terminus (rAAVrh74.MHCK7.micro-dystrophin.Cterm). Functional and histological benefit were examined at 4 weeks following intramuscular delivery in mdx mice. rAAVrh74.MHCK7.micro-dystrophin provided the most robust transgene expression and significantly increased specific force output in the tibialis anterior muscle. Muscle environment was normalized (i.e. reductions in central nucleation), indicating functional and histological advantages of rAAVrh74.MHCK7.micro-dystrophin. Thus, promoter choice and transgene design are critical for optimal dystrophin expression/distribution for maximal functional improvement.


Asunto(s)
Distrofina , Distrofia Muscular de Duchenne , Ratones , Animales , Distrofina/genética , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/terapia , Ratones Endogámicos mdx , Dependovirus/genética , Citoesqueleto de Actina , Modelos Animales de Enfermedad
5.
Ann Neurol ; 94(5): 955-968, 2023 11.
Artículo en Inglés | MEDLINE | ID: mdl-37539981

RESUMEN

OBJECTIVE: Delandistrogene moxeparvovec is approved in the USA for the treatment of ambulatory patients (4-5 years) with Duchenne muscular dystrophy. ENDEAVOR (SRP-9001-103; NCT04626674) is a single-arm, open-label study to evaluate delandistrogene moxeparvovec micro-dystrophin expression, safety, and functional outcomes following administration of commercial process delandistrogene moxeparvovec. METHODS: In cohort 1 of ENDEAVOR (N = 20), eligible ambulatory males, aged ≥4 to <8 years, received a single intravenous infusion of delandistrogene moxeparvovec (1.33 × 1014 vg/kg). The primary endpoint was change from baseline (CFBL) to week 12 in delandistrogene moxeparvovec micro-dystrophin by western blot. Additional endpoints evaluated included: safety; vector genome copies; CFBL to week 12 in muscle fiber-localized micro-dystrophin by immunofluorescence; and functional assessments, including North Star Ambulatory Assessment, with comparison with a propensity score-weighted external natural history control. RESULTS: The 1-year safety profile of commercial process delandistrogene moxeparvovec in ENDEAVOR was consistent with safety data reported in other delandistrogene moxeparvovec trials (NCT03375164 and NCT03769116). Delandistrogene moxeparvovec micro-dystrophin expression was robust, with sarcolemmal localization at week 12; mean (SD) CFBL in western blot, 54.2% (42.6); p < 0.0001. At 1 year, patients demonstrated stabilized or improved North Star Ambulatory Assessment total scores; mean (SD) CFBL, +4.0 (3.5). Treatment versus a propensity score-weighted external natural history control demonstrated a statistically significant difference in least squares mean (standard error) CFBL in North Star Ambulatory Assessment, +3.2 (0.6) points; p < 0.0001. INTERPRETATION: Results confirm efficient transduction of muscle by delandistrogene moxeparvovec. One-year post-treatment, delandistrogene moxeparvovec was well tolerated, and demonstrated stabilized or improved motor function, suggesting a clinical benefit for patients with Duchenne muscular dystrophy. ANN NEUROL 2023;94:955-968.


Asunto(s)
Distrofia Muscular de Duchenne , Masculino , Humanos , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/terapia , Distrofina/genética , Terapia Genética/métodos , Infusiones Intravenosas , Fibras Musculares Esqueléticas
6.
Front Cell Dev Biol ; 11: 1167762, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37497476

RESUMEN

Introduction: Delandistrogene moxeparvovec (SRP-9001) is an investigational gene transfer therapy designed for targeted expression of SRP-9001 dystrophin protein, a shortened dystrophin retaining key functional domains of the wild-type protein. Methods: This Phase 2, double-blind, two-part (48 weeks per part) crossover study (SRP-9001-102 [Study 102]; NCT03769116) evaluated delandistrogene moxeparvovec in patients, aged ≥4 to <8 years with Duchenne muscular dystrophy. Primary endpoints (Part 1) were change from baseline (CFBL) in SRP-9001 dystrophin expression (Week 12), by Western blot, and in North Star Ambulatory Assessment (NSAA) score (Week 48). Safety assessments included treatment-related adverse events (TRAEs). Patients were randomized and stratified by age to placebo (n = 21) or delandistrogene moxeparvovec (n = 20) and crossed over for Part 2. Results: SRP-9001 dystrophin expression was achieved in all patients: mean CFBL to Week 12 was 23.82% and 39.64% normal in Parts 1 and 2, respectively. In Part 1, CFBL to Week 48 in NSAA score (least-squares mean, LSM [standard error]) was +1.7 (0.6) with treatment versus +0.9 (0.6) for placebo; p = 0.37. Disparity in baseline motor function between groups likely confounded these results. In 4- to 5-year-olds with matched baseline motor function, CFBL to Week 48 in NSAA scores was significantly different (+2.5 points; p = 0.0172), but not significantly different in 6-to-7-year-olds with imbalanced baseline motor function (-0.7 points; p = 0.5384). For patients treated with delandistrogene moxeparvovec in Part 2, CFBL to Week 48 in NSAA score was +1.3 (2.7), whereas for those treated in Part 1, NSAA scores were maintained. As all patients in Part 2 were exposed to treatment, results were compared with a propensity-score-weighted external control (EC) cohort. The LSM difference in NSAA score between the Part 2 treated group and EC cohort was statistically significant (+2.0 points; p = 0.0009). The most common TRAEs were vomiting, decreased appetite, and nausea. Most occurred within the first 90 days and all resolved. Discussion: Results indicate robust expression of SRP-9001 dystrophin and overall stabilization in NSAA up to 2 years post-treatment. Differences in NSAA between groups in Part 1 were not significant for the overall population, likely because cohorts were stratified only by age, and other critical prognostic factors were not well matched at baseline.

7.
Mol Ther Methods Clin Dev ; 28: 284-299, 2023 Mar 09.
Artículo en Inglés | MEDLINE | ID: mdl-36816759

RESUMEN

Limb-girdle muscular dystrophy (LGMD) type 2C/R5 results from mutations in the γ-sarcoglycan (SGCG) gene and is characterized by muscle weakness and progressive wasting. Loss of functional γ-sarcoglycan protein in the dystrophin-associated protein complex destabilizes the sarcolemma, leading to eventual myofiber death. The SGCG knockout mouse (SGCG -/-) has clinical-pathological features that replicate the human disease, making it an ideal model for translational studies. We designed a self-complementary rAAVrh74 vector containing a codon-optimized human SGCG transgene driven by the muscle-specific MHCK7 promoter (SRP-9005) to investigate adeno-associated virus (AAV)-mediated SGCG gene transfer in SGCG -/- mice as proof of principle for LGMD 2C/R5. Gene transfer therapy resulted in widespread transgene expression in skeletal muscle and heart, improvements in muscle histopathology characterized by decreased central nuclei and fibrosis, and normalized fiber size. Histopathologic improvements were accompanied by functional improvements, including increased ambulation and force production and resistance to injury of the tibialis anterior and diaphragm muscles. This study demonstrates successful systemic delivery of the hSGCG transgene in SGCG -/- mice, with functional protein expression, reconstitution of the sarcoglycan complex, and corresponding physiological and functional improvements, which will help establish a minimal effective dose for translation of SRP-9005 gene transfer therapy in patients with LGMD 2C/R5.

8.
Ind Health ; 61(5): 307-319, 2023 Sep 22.
Artículo en Inglés | MEDLINE | ID: mdl-35934791

RESUMEN

Due to the COVID-19 pandemic, the number of employees in flexible work from home has increased markedly along with a reliance on information communication technologies. This study investigated the role of an organisational factor, psychosocial safety climate (PSC; the climate for worker psychological health and safety), as an antecedent of these new kinds of demands (specifically work from home digital job demands) and their effect on work-life conflict. Data were gathered via an online survey of 2,177 employees from 37 Australian universities. Multilevel modelling showed that university level PSC to demands, y=-0.09, SE=0.03, p<0.01, and demands to work-life conflict, y=0.51, SE=0.19, p<0.05, relationships were significant. Supporting the antecedent theory, university level PSC was significantly indirectly related to work-life conflict via demands (LL -0.10 UL -0.01). Against expectations PSC did not moderate the demand to work-life conflict relationship. The results imply that targeting PSC could help prevent work from home digital job demands, and therefore, work-life conflict. Further research is needed on the role of digital job resources as flexible and hybrid work takes hold post COVID.


Asunto(s)
COVID-19 , Cultura Organizacional , Humanos , Estrés Psicológico/psicología , Equilibrio entre Vida Personal y Laboral , Pandemias , Teletrabajo , Australia/epidemiología , COVID-19/epidemiología , Encuestas y Cuestionarios , Satisfacción en el Trabajo
9.
Artículo en Inglés | MEDLINE | ID: mdl-36294252

RESUMEN

Healthcare sector organizations have long been facing the issue of productivity loss due to presenteeism which is affected by psychosocial safety climate (PSC) and work intensification. Presenteeism has visibly increased among nurses during COVID-19 pandemic period. Grounded in COR theory and sensemaking theory, the current study aimed to examine the role PSC plays as driver or moderator to reduce presenteeism by lessening work intensification over time and the impact of work intensification over time on presenteeism during the COVID-19 pandemic. Adopting a time-lagged research design, this study gathered data from randomly selected registered nurses, practicing in Québec, Canada in two phases, i.e., 800 at Time 1 and 344 at Time 2 through email surveys. The study results showed that (1) PSC reduces presenteeism over time by reducing work intensification at time 1; (2) PSC moderates the relationship between work intensification at time 1 and work intensification at time 2; and (3) PSC as moderator also lessens the detrimental effect of work intensification at time 2 on presenteeism at time 2. Presenteeism among nurses affects their health and psychological well-being. We find that PSC is likely an effective organizational tool particularly in crises situations, by providing an organizational mechanism to assist nurses cope (through a resource caravan, management support) with managing intensified work.


Asunto(s)
COVID-19 , Presentismo , Humanos , Cultura Organizacional , Pandemias/prevención & control , Encuestas y Cuestionarios , Lugar de Trabajo/psicología
10.
Australas J Ageing ; 41(3): 440-447, 2022 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-35289048

RESUMEN

OBJECTIVE: The aim of the present study was to explore the impact of the COVID-19 pandemic on the financial well-being of older Australian retirees. METHODS: Thirty Australian retirees (16 females and 14 males), older than 65 years of age, were asked 'Have your finances been affected by the events surrounding COVID-19?'. Data were analysed using Braun and Clarke's six-step approach, and Bronfenbrenner's socio-ecological model was utilised to analyse thematic responses at individual, household, community and societal levels. RESULTS: Two COVID-19-related themes emerged from interviews: COVID-19 and increased financial stress and COVID-19 and frustration with digital banking. CONCLUSIONS: This study revealed that the financial well-being of older Australians and especially self-funded retirees has been negatively impacted by the COVID-19 pandemic. Participants felt financially worse off primarily due to the volatility of the financial markets, the need to support adult children and the increased cost of living. Also, participants expressed their hesitation and frustration with digital banking services, and their desire for greater personal contact with financial institutions, particularly during times of uncertainty.


Asunto(s)
COVID-19 , Administración Financiera , Australia/epidemiología , Femenino , Humanos , Masculino , Pandemias
11.
Neurodegener Dis Manag ; 11(5): 411-429, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34472379

RESUMEN

Limb-girdle muscular dystrophies (LGMDs) represent a major group of muscle disorders. Treatment is sorely needed and currently expanding based on safety and efficacy adopting principles of single-dosing gene therapy for monogenic autosomal recessive disorders. Gene therapy has made in-roads for LGMD and this review describes progress that has been achieved for these conditions. This review first provides a background on the definition and classification of LGMDs. The major effort focuses on progress in LGMD gene therapy, from experimental studies to clinical trials. The disorders discussed include the LGMDs where the most work has been done including calpainopathies (LGMD2A/R1), dysferlinopathies (LGMD2B/R2) and sarcoglycanopathies (LGMD2C/R5, LGMD2D/R3, LGMD2E/R4). Early success in clinical trials provides a template to move the field forward and potentially apply emerging technology like CRISPR/Cas9 that may enhance the scope and efficacy of gene therapy applied to patient care.


Lay abstract Limb-girdle muscular dystrophy is a term that is applied to a group of relatively rare forms of muscular dystrophy. The term 'LGMD' was introduced in the 1950's, but there were no strict rules for defining the condition. This changed as a result of the 229th European Neuromuscular Center International Workshop in 2017 providing a clear definition and classification discussed in this article. Limb-girdle muscular dystrophy is now recognized as a genetic muscle disease with an elevated serum creatine kinase and dystrophic changes on muscle histology. Most treatments up to now rely on supportive measures for heart and lungs and assisting the physical limitations. Medications have not proven to be beneficial to stop progression of disease. This article focuses on new innovations of treatment that target the effected gene and the use special methods to replace the abnormal gene.


Asunto(s)
Distrofia Muscular de Cinturas , Sarcoglicanopatías , Humanos , Distrofia Muscular de Cinturas/genética , Distrofia Muscular de Cinturas/terapia
12.
J Neuromuscul Dis ; 8(4): 489-494, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33780374

RESUMEN

Recombinant micro-dystrophin genes are designed to treat Duchenne muscular dystrophy (DMD) by retaining dystrophin domains believed to play key functional roles while fitting the packaging capacity of adeno-associated virus vectors. Domains R1-3 are important for muscle force generation and for association with the sarcolemma, but the nature of this interaction is not fully understood. We measured lipid-binding affinity of 3 peptides containing different spectrin-like repeat modules (R1-3; R1-2; and R1, 2, 22). Lipid-binding affinity was highest with R1-3, suggesting that the complete R1-R3 region could be beneficial and should be considered for inclusion in micro-dystrophin constructs.


Asunto(s)
Distrofina/genética , Distrofia Muscular de Duchenne/genética , Espectrina/genética , Humanos , Lípidos/genética , Sarcolema/genética
13.
Hum Gene Ther ; 32(7-8): 375-389, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33397205

RESUMEN

Duchenne muscular dystrophy (DMD) is a rare, X-linked, fatal, degenerative neuromuscular disease caused by mutations in the DMD gene. More than 2,000 mutations of the DMD gene are responsible for progressive loss of muscle strength, loss of ambulation, and generally respiratory and cardiac failure by age 30. Recently, gene transfer therapy has received widespread interest as a disease-modifying treatment for all patients with DMD. We designed an adeno-associated virus vector (rAAVrh74) containing a codon-optimized human micro-dystrophin transgene driven by a skeletal and cardiac muscle-specific promoter, MHCK7. To test the efficacy of rAAVrh74.MHCK7.micro-dystrophin, we evaluated systemic injections in mdx (dystrophin-null) mice at low (2 × 1012 vector genome [vg] total dose, 8 × 1013 vg/kg), intermediate (6 × 1012 vg total dose, 2 × 1014 vg/kg), and high doses (1.2 × 1013 vg total dose, 6 × 1014 vg/kg). Three months posttreatment, specific force increased in the diaphragm (DIA) and tibialis anterior muscle, with intermediate and high doses eliciting force outputs at wild-type (WT) levels. Histological improvement included reductions in fibrosis and normalization of myofiber size, specifically in the DIA, where results for low and intermediate doses were not significantly different from the WT. Significant reduction in central nucleation was also observed, although complete normalization to WT was not seen. No vector-associated toxicity was reported either by clinical or organ-specific laboratory assessments or following formal histopathology. The findings in this preclinical study provided proof of principle for safety and efficacy of systemic delivery of rAAVrh74.MHCK7.micro-dystrophin at high vector titers, supporting initiation of a Phase I/II safety study in boys with DMD.


Asunto(s)
Distrofina , Distrofia Muscular de Duchenne , Animales , Modelos Animales de Enfermedad , Distrofina/genética , Terapia Genética , Humanos , Ratones , Ratones Endogámicos mdx , Músculo Esquelético , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/terapia
14.
Hum Gene Ther ; 32(7-8): 390-404, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33349138

RESUMEN

Limb-girdle muscular dystrophy type 2D/R3 (LGMD2D/R3) is a progressive muscular dystrophy that manifests with muscle weakness, respiratory abnormalities, and in rare cases cardiomyopathy. LGMD2D/R3 is caused by mutations in the SGCA gene resulting in loss of protein and concomitant loss of some or all components of the dystrophin-associated glycoprotein complex. The sgca-null (sgca-/-) mouse recapitulates the clinical phenotype of patients with LGMD2D/R3, including dystrophic features such as muscle necrosis and fibrosis, elevated serum creatine kinase (CK), and reduction in the generation of absolute muscle force and locomotor activity. Thus, sgca-/- mice provide a relevant model to test the safety and efficacy of gene transfer. We designed a self-complementary AAVrh74 vector containing a codon-optimized full-length human SGCA (hSGCA) transgene driven by a muscle-specific promoter, shortened muscle creatine kinase (tMCK). In this report, we test the efficacy and safety of scAAVrh74.tMCK.hSGCA in sgca-/- mice using a dose-escalation design to evaluate a single systemic injection of 1.0 × 1012, 3.0 × 1012, and 6.0 × 1012 vg total dose compared with vehicle-treatment and wild-type mice. In sgca-/- mice, treatment with scAAVrh74.tMCK.hSGCA resulted in robust expression of α-sarcoglycan protein at the sarcolemma membrane in skeletal muscle at all doses tested. In addition, scAAVrh74.tMCK.hSGCA was effective in improving the histopathology of limb and diaphragm muscle of sgca-/- mice, as indicated by reductions in fibrosis, central nucleation, and normalization of myofiber size. These molecular changes were concomitant with significant increases in specific force generation in the diaphragm and tibialis anterior muscle, protection against eccentric force loss, and reduction in serum CK. Locomotor activity was improved at all doses of vector-treated compared with vehicle-treated sgca-/- mice. Lastly, vector toxicity was not detected in a serum chemistry panel and by gross necropsy. Collectively, these findings provide support for a systemic delivery of scAAVrh74.tMCK.hSGCA in a clinical setting for the treatment of LGMD2D/R3.


Asunto(s)
Distrofia Muscular de Cinturas , Sarcoglicanopatías , Animales , Terapia Genética , Humanos , Ratones , Músculo Esquelético , Distrofia Muscular de Cinturas/genética , Distrofia Muscular de Cinturas/terapia , Sarcoglicanopatías/genética , Sarcoglicanopatías/terapia , Sarcoglicanos/genética
15.
JAMA Neurol ; 77(9): 1122-1131, 2020 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-32539076

RESUMEN

Importance: Micro-dystrophin gene transfer shows promise for treating patients with Duchenne muscular dystrophy (DMD) using recombinant adeno-associated virus serotype rh74 (rAAVrh74) and codon-optimized human micro-dystrophin driven by a skeletal and cardiac muscle-specific promoter with enhanced cardiac expression (MHCK7). Objective: To identify the 1-year safety and tolerability of intravenous rAAVrh74.MHCK7.micro-dystrophin in patients with DMD. Design, Setting, and Participants: This open-label, phase 1/2a nonrandomized controlled trial was conducted at the Nationwide Children's Hospital in Columbus, Ohio. It began on November 2, 2017, with a planned duration of follow-up of 3 years, ending in March 2021. The first 4 patients who met eligibility criteria were enrolled, consisting of ambulatory male children with DMD without preexisting AAVrh74 antibodies and a stable corticosteroid dose (≥12 weeks). Interventions: A single dose of 2.0 × 1014 vg/kg rAAVrh74.MHCK7.micro-dystrophin was infused through a peripheral limb vein. Daily prednisolone, 1 mg/kg, started 1 day before gene delivery (30-day taper after infusion). Main Outcomes and Measures: Safety was the primary outcome. Secondary outcomes included micro-dystrophin expression by Western blot and immunohistochemistry. Functional outcomes measured by North Star Ambulatory Assessment (NSAA) and serum creatine kinase were exploratory outcomes. Results: Four patients were included (mean [SD] age at enrollment, 4.8 [1.0] years). All adverse events (n = 53) were considered mild (33 [62%]) or moderate (20 [38%]), and no serious adverse events occurred. Eighteen adverse events were considered treatment related, the most common of which was vomiting (9 of 18 events [50%]). Three patients had transiently elevated γ-glutamyltransferase, which resolved with corticosteroids. At 12 weeks, immunohistochemistry of gastrocnemius muscle biopsy specimens revealed robust transgene expression in all patients, with a mean of 81.2% of muscle fibers expressing micro-dystrophin with a mean intensity of 96% at the sarcolemma. Western blot showed a mean expression of 74.3% without fat or fibrosis adjustment and 95.8% with adjustment. All patients had confirmed vector transduction and showed functional improvement of NSAA scores and reduced creatine kinase levels (posttreatment vs baseline) that were maintained for 1 year. Conclusions and Relevance: This trial showed rAAVrh74.MHCK7.micro-dystrophin to be well tolerated and have minimal adverse events; the safe delivery of micro-dystrophin transgene; the robust expression and correct localization of micro-dystrophin protein; and improvements in creatine kinase levels and NSAA scores. These findings suggest that rAAVrh74.MHCK7.micro-dystrophin can provide functional improvement that is greater than that observed under standard of care. Trial Registration: ClinicalTrials.gov Identifier: NCT03375164.


Asunto(s)
Distrofina , Terapia Genética/métodos , Distrofia Muscular de Duchenne/terapia , Evaluación de Resultado en la Atención de Salud , Niño , Preescolar , Dependovirus , Distrofina/genética , Estudios de Seguimiento , Técnicas de Transferencia de Gen , Terapia Genética/efectos adversos , Vectores Genéticos , Humanos , Masculino , Músculo Esquelético/metabolismo , Distrofia Muscular de Duchenne/genética , Proyectos Piloto
16.
Expert Opin Biol Ther ; 20(3): 263-274, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-32031420

RESUMEN

Introduction: The development of adeno-associated virus (AAV) vectors as safe vehicles for in vivo delivery of therapeutic genes has been a major milestone in the advancement of gene therapy, enabling a promising strategy for ameliorating a wide range of diseases, including Duchenne muscular dystrophy (DMD).Areas covered: Based on experience with the development of a gene transfer therapy agent for DMD, we discuss ways in which gene therapy for rare disease challenges traditional clinical development paradigms, and recommend a step-wise approach for design and evaluation to support broader applicability of gene therapy.Expert opinion: The gene therapy development approach should intentionally design the therapeutic construct and the clinical study to systematically evaluate agent delivery, safety, and efficacy. Rigorous preclinical work is essential for establishing an effective gene delivery platform and determining the efficacious dose. Clinical studies should thoroughly evaluate transduction, on-target transgene expression at the tissue and cellular level, and functional efficacy.


Asunto(s)
Distrofina/genética , Distrofia Muscular de Duchenne/terapia , Ensayos Clínicos como Asunto , Dependovirus/genética , Distrofina/uso terapéutico , Terapia Genética/efectos adversos , Vectores Genéticos/genética , Vectores Genéticos/metabolismo , Humanos , Distrofia Muscular de Duchenne/genética , Evaluación de Resultado en la Atención de Salud , Bibliotecas de Moléculas Pequeñas/uso terapéutico
17.
Hum Gene Ther ; 29(7): 749-762, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-28707952

RESUMEN

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.


Asunto(s)
ADN Complementario/administración & dosificación , Disferlina/genética , Terapia Genética , Distrofia Muscular de Cinturas/terapia , Animales , ADN Complementario/genética , Dependovirus/genética , Modelos Animales de Enfermedad , Disferlina/administración & dosificación , Regulación de la Expresión Génica , Vectores Genéticos/uso terapéutico , Humanos , Masculino , Ratones , Músculo Esquelético , Distrofia Muscular de Cinturas/genética , Distrofia Muscular de Cinturas/patología , Mutación
18.
BMC Cell Biol ; 14: 39, 2013 Sep 21.
Artículo en Inglés | MEDLINE | ID: mdl-24053798

RESUMEN

BACKGROUND: Differentiation and fusion of skeletal muscle myoblasts into multi-nucleated myotubes is required for neonatal development and regeneration in adult skeletal muscle. Herein, we report novel findings that protein kinase C theta (PKCθ) regulates myoblast differentiation via phosphorylation of insulin receptor substrate-1 and ERK1/2. RESULTS: In this study, PKCθ knockdown (PKCθshRNA) myotubes had reduced inhibitory insulin receptor substrate-1 ser1095 phosphorylation, enhanced myoblast differentiation and cell fusion, and increased rates of protein synthesis as determined by [3H] phenylalanine incorporation. Phosphorylation of insulin receptor substrate-1 ser632/635 and extracellular signal-regulated kinase1/2 (ERK1/2) was increased in PKCθshRNA cells, with no change in ERK5 phosphorylation, highlighting a PKCθ-regulated myogenic pathway. Inhibition of PI3-kinase prevented cell differentiation and fusion in control cells, which was attenuated in PKCθshRNA cells. Thus, with reduced PKCθ, differentiation and fusion occur in the absence of PI3-kinase activity. Inhibition of the ERK kinase, MEK1/2, impaired differentiation and cell fusion in control cells. Differentiation was preserved in PKCθshRNA cells treated with a MEK1/2 inhibitor, although cell fusion was blunted, indicating PKCθ regulates differentiation via IRS1 and ERK1/2, and this occurs independently of MEK1/2 activation. CONCLUSION: Cellular signaling regulating the myogenic program and protein synthesis are complex and intertwined. These studies suggest that PKCθ regulates myogenic and protein synthetic signaling via the modulation of IRS1and ERK1/2 phosphorylation. Myotubes lacking PKCθ had increased rates of protein synthesis and enhanced myotube development despite reduced activation of the canonical anabolic-signaling pathway. Further investigation of PKCθ regulated signaling may reveal important interactions regulating skeletal muscle health in an insulin resistant state.


Asunto(s)
Proteínas Sustrato del Receptor de Insulina/genética , Isoenzimas/genética , Proteína Quinasa 1 Activada por Mitógenos/genética , Proteína Quinasa 3 Activada por Mitógenos/genética , Desarrollo de Músculos/genética , Fibras Musculares Esqueléticas/metabolismo , Mioblastos/metabolismo , Proteína Quinasa C/genética , Animales , Diferenciación Celular , Fusión Celular , Células Cultivadas , Regulación del Desarrollo de la Expresión Génica , Proteínas Sustrato del Receptor de Insulina/metabolismo , Isoenzimas/antagonistas & inhibidores , Isoenzimas/metabolismo , MAP Quinasa Quinasa 1/genética , MAP Quinasa Quinasa 1/metabolismo , MAP Quinasa Quinasa 2/genética , MAP Quinasa Quinasa 2/metabolismo , Ratones , Proteína Quinasa 1 Activada por Mitógenos/antagonistas & inhibidores , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/antagonistas & inhibidores , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Fibras Musculares Esqueléticas/citología , Mioblastos/citología , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación , Biosíntesis de Proteínas , Proteína Quinasa C/antagonistas & inhibidores , Proteína Quinasa C/metabolismo , Proteína Quinasa C-theta , Inhibidores de Proteínas Quinasas/farmacología , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Transducción de Señal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...