Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 117
Filtrer
2.
Muscle Nerve ; 70(1): 60-70, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38482981

RÉSUMÉ

INTRODUCTION/AIMS: Eteplirsen, approved in the US for patients with Duchenne muscular dystrophy (DMD) with exon 51 skip-amenable variants, is associated with attenuated ambulatory/pulmonary decline versus DMD natural history (NH). We report overall survival in a US cohort receiving eteplirsen and contextualize these outcomes versus DMD NH. METHODS: US patients with DMD receiving eteplirsen were followed through a patient support program, with data collected on ages at eteplirsen initiation and death/end of follow-up. Individual DMD NH data were extracted by digitizing Kaplan-Meier (KM) curves from published systematic and targeted literature reviews. Overall survival age was analyzed using KM curves and contextualized with DMD NH survival curves; subanalyses considered age groups and duration of eteplirsen exposure. Overall survival time from treatment initiation was also evaluated. RESULTS: A total of 579 eteplirsen-treated patients were included. During a total follow-up of 2119 person-years, median survival age was 32.8 years. DMD NH survival curves extracted from four publications (follow-up for 1224 DMD NH controls) showed overall pooled median survival age of 27.4 years. Eteplirsen-treated patients had significantly longer survival from treatment initiation versus age-matched controls (age-adjusted hazard ratio [HR], 0.65; 95% confidence interval [CI], 0.44-0.98; p < .05). Longer treatment exposure was associated with improved survival (HR, 0.15; 95% CI, 0.05-0.41; p < .001). Comparisons using different DMD NH cohorts to address common risks of bias yielded consistent findings. DISCUSSION: Data suggest eteplirsen may prolong survival in patients with DMD across a wide age range. As more data become available, the impact of eteplirsen on survival will be further elucidated.


Sujet(s)
Myopathie de Duchenne , Myopathie de Duchenne/traitement médicamenteux , Myopathie de Duchenne/mortalité , Humains , Mâle , Enfant , Adolescent , Adulte , Enfant d'âge préscolaire , Jeune adulte , Morpholinos/usage thérapeutique , Femelle , Études de cohortes , Études de suivi , Estimation de Kaplan-Meier
3.
EMBO Mol Med ; 16(4): 1027-1045, 2024 Apr.
Article de Anglais | MEDLINE | ID: mdl-38448545

RÉSUMÉ

Clinical deployment of oligonucleotides requires delivery technologies that improve stability, target tissue accumulation and cellular internalization. Exosomes show potential as ideal delivery vehicles. However, an affordable generalizable system for efficient loading of oligonucleotides on exosomes remain lacking. Here, we identified an Exosomal Anchor DNA Aptamer (EAA) via SELEX against exosomes immobilized with our proprietary CP05 peptides. EAA shows high binding affinity to different exosomes and enables efficient loading of nucleic acid drugs on exosomes. Serum stability of thrombin inhibitor NU172 was prolonged by exosome-loading, resulting in increased blood flow after injury in vivo. Importantly, Duchenne Muscular Dystrophy PMO can be readily loaded on exosomes via EAA (EXOEAA-PMO). EXOEAA-PMO elicited significantly greater muscle cell uptake, tissue accumulation and dystrophin expression than PMO in vitro and in vivo. Systemic administration of EXOEAA-PMO elicited therapeutic levels of dystrophin restoration and functional improvements in mdx mice. Altogether, our study demonstrates that EAA enables efficient loading of different nucleic acid drugs on exosomes, thus providing an easy and generalizable strategy for loading nucleic acid therapeutics on exosomes.


Sujet(s)
Exosomes , Myopathie de Duchenne , Animaux , Souris , Dystrophine/génétique , Souris de lignée mdx , Exosomes/métabolisme , Morpholinos/métabolisme , Morpholinos/pharmacologie , Morpholinos/usage thérapeutique , Myopathie de Duchenne/traitement médicamenteux , Myopathie de Duchenne/génétique , Oligonucléotides/métabolisme , Oligonucléotides/usage thérapeutique
4.
Skelet Muscle ; 13(1): 19, 2023 11 18.
Article de Anglais | MEDLINE | ID: mdl-37980539

RÉSUMÉ

BACKGROUND: The lack of functional dystrophin protein in Duchenne muscular dystrophy (DMD) causes chronic skeletal muscle inflammation and degeneration. Therefore, the restoration of functional dystrophin levels is a fundamental approach for DMD therapy. Electrical impedance myography (EIM) is an emerging tool that provides noninvasive monitoring of muscle conditions and has been suggested as a treatment response biomarker in diverse indications. Although magnetic resonance imaging (MRI) of skeletal muscles has become a standard measurement in clinical trials for DMD, EIM offers distinct advantages, such as portability, user-friendliness, and reduced cost, allowing for remote monitoring of disease progression or response to therapy. To investigate the potential of EIM as a biomarker for DMD, we compared longitudinal EIM data with MRI/histopathological data from an X-linked muscular dystrophy (mdx) mouse model of DMD. In addition, we investigated whether EIM could detect dystrophin-related changes in muscles using antisense-mediated exon skipping in mdx mice. METHODS: The MRI data for muscle T2, the magnetic resonance spectroscopy (MRS) data for fat fraction, and three EIM parameters with histopathology were longitudinally obtained from the hindlimb muscles of wild-type (WT) and mdx mice. In the EIM study, a cell-penetrating peptide (Pip9b2) conjugated antisense phosphorodiamidate morpholino oligomer (PPMO), designed to induce exon-skipping and restore functional dystrophin production, was administered intravenously to mdx mice. RESULTS: MRI imaging in mdx mice showed higher T2 intensity at 6 weeks of age in hindlimb muscles compared to WT mice, which decreased at ≥ 9 weeks of age. In contrast, EIM reactance began to decline at 12 weeks of age, with peak reduction at 18 weeks of age in mdx mice. This decline was associated with myofiber atrophy and connective tissue infiltration in the skeletal muscles. Repeated dosing of PPMO (10 mg/kg, 4 times every 2 weeks) in mdx mice led to an increase in muscular dystrophin protein and reversed the decrease in EIM reactance. CONCLUSIONS: These findings suggest that muscle T2 MRI is sensitive to the early inflammatory response associated with dystrophin deficiency, whereas EIM provides a valuable biomarker for the noninvasive monitoring of subsequent changes in skeletal muscle composition. Furthermore, EIM reactance has the potential to monitor dystrophin-deficient muscle abnormalities and their recovery in response to antisense-mediated exon skipping.


Sujet(s)
Dystrophine , Myopathie de Duchenne , Souris , Animaux , Dystrophine/génétique , Dystrophine/métabolisme , Souris de lignée mdx , Impédance électrique , Souris de lignée C57BL , Myopathie de Duchenne/imagerie diagnostique , Myopathie de Duchenne/génétique , Myopathie de Duchenne/anatomopathologie , Muscles squelettiques/métabolisme , Morpholinos/pharmacologie , Morpholinos/usage thérapeutique , Myographie , Marqueurs biologiques
5.
Cells ; 12(19)2023 10 02.
Article de Anglais | MEDLINE | ID: mdl-37830609

RÉSUMÉ

Antisense oligonucleotide-based (ASO) therapeutics have emerged as a promising strategy for the treatment of human disorders. Charge-neutral PMOs have promising biological and pharmacological properties for antisense applications. Despite their great potential, the efficient delivery of these therapeutic agents to target cells remains a major obstacle to their widespread use. Cellular uptake of naked PMO is poor. Cell-penetrating peptides (CPPs) appear as a possibility to increase the cellular uptake and intracellular delivery of oligonucleotide-based drugs. Among these, the DG9 peptide has been identified as a versatile CPP with remarkable potential for enhancing the delivery of ASO-based therapeutics due to its unique structural features. Notably, in the context of phosphorodiamidate morpholino oligomers (PMOs), DG9 has shown promise in enhancing delivery while maintaining a favorable toxicity profile. A few studies have highlighted the potential of DG9-conjugated PMOs in DMD (Duchenne Muscular Dystrophy) and SMA (Spinal Muscular Atrophy), displaying significant exon skipping/inclusion and functional improvements in animal models. The article provides an overview of a detailed understanding of the challenges that ASOs face prior to reaching their targets and continued advances in methods to improve their delivery to target sites and cellular uptake, focusing on DG9, which aims to harness ASOs' full potential in precision medicine.


Sujet(s)
Peptides de pénétration cellulaire , Amyotrophie spinale , Myopathie de Duchenne , Animaux , Humains , Oligonucléotides antisens/usage thérapeutique , Peptides de pénétration cellulaire/composition chimique , Oligonucléotides , Morpholinos/usage thérapeutique , Myopathie de Duchenne/traitement médicamenteux , Myopathie de Duchenne/génétique , Amyotrophie spinale/thérapie
6.
J Biosci ; 482023.
Article de Anglais | MEDLINE | ID: mdl-37846020

RÉSUMÉ

Duchenne muscular dystrophy (DMD) is an X-linked genetic disease primarily affecting boys causing loss of the dystrophin protein, ultimately leading to muscle wastage and death by cardiac or respiratory failure. The genetic mutation involved can be overcome with antisense oligonucleotides which bind to a pre-mRNA and results in reading frame restoration by exon skipping. Phosphorodiamidate morpholino oligonucleotides (PMOs) are a class of antisense agents with a neutral backbone derived from RNA which can induce effective exon skipping. In this review, the evolution of PMOs in exon skipping therapy for the last two decades has been detailed with the gradual structural and functional advancements. Even though the success rate of PMObased therapy has been high with four FDA approved drugs, several key challenges are yet to overcome, one being the dystrophin restoration in cardiac muscle. The current scenario in further improvement of PMOs has been discussed along with the future perspectives that have the potential to revolutionize the therapeutic benefits in DMD.


Sujet(s)
Dystrophine , Myopathie de Duchenne , Mâle , Humains , Morpholinos/génétique , Morpholinos/usage thérapeutique , Dystrophine/génétique , Dystrophine/métabolisme , Myopathie de Duchenne/génétique , Myopathie de Duchenne/thérapie , Oligonucléotides antisens/génétique , Oligonucléotides antisens/usage thérapeutique , Exons/génétique
7.
Eur J Cell Biol ; 102(2): 151326, 2023 Jun.
Article de Anglais | MEDLINE | ID: mdl-37295266

RÉSUMÉ

Spinal muscular atrophy (SMA), the most common genetic cause of infantile death, is caused by a mutation in the survival of motor neuron 1 gene (SMN1), leading to the death of motor neurons and progressive muscle weakness. SMN1 normally produces an essential protein called SMN. Although humans possess a paralogous gene called SMN2, ∼90% of the SMN it produces is non-functional. This is due to a mutation in SMN2 that causes the skipping of a required exon during splicing of the pre-mRNA. The first treatment for SMA, nusinersen (brand name Spinraza), was approved by the FDA in 2016 and by the EMU in 2017. Nusinersen is an antisense oligonucleotide-based therapy that alters the splicing of SMN2 to make functional full-length SMN protein. Despite the recent advancements in antisense oligonucleotide therapy and SMA treatment development, nusinersen is faced with a multitude of challenges, such as intracellular and systemic delivery. In recent years, the use of peptide-conjugated phosphorodiamidate morpholino oligomers (PPMOs) in antisense therapy has gained interest. These are antisense oligonucleotides conjugated to cell-penetrating peptides such as Pips and DG9, and they have the potential to address the challenges associated with delivery. This review focuses on the historic milestones, development, current challenges, and future perspectives of antisense therapy for SMA.


Sujet(s)
Amyotrophie spinale , Oligonucléotides antisens , Humains , Oligonucléotides antisens/génétique , Oligonucléotides antisens/usage thérapeutique , Amyotrophie spinale/traitement médicamenteux , Amyotrophie spinale/génétique , Morpholinos/génétique , Morpholinos/usage thérapeutique , Motoneurones/métabolisme , Épissage des ARN
8.
Neuromuscul Disord ; 33(6): 476-483, 2023 06.
Article de Anglais | MEDLINE | ID: mdl-37207382

RÉSUMÉ

Eteplirsen is FDA-approved for the treatment of Duchenne muscular dystrophy (DMD) in exon 51 skip-amenable patients. Previous studies in boys > 4 years of age indicate eteplirsen is well tolerated and attenuates pulmonary and ambulatory decline compared with matched natural history cohorts. Here the safety, tolerability and pharmacokinetics of eteplirsen in boys aged 6-48 months is evaluated. In this open-label, multicenter, dose-escalation study (NCT03218995), boys with a confirmed mutation of the DMD gene amenable to exon 51 skipping (Cohort 1: aged 24-48 months, n = 9; Cohort 2: aged 6 to < 24 months, n = 6) received ascending doses (2, 4, 10, 20, 30 mg/kg) of once-weekly eteplirsen intravenously over 10 weeks, continuing at 30 mg/kg up to 96 weeks. Endpoints included safety (primary) and pharmacokinetics (secondary). All 15 participants completed the study. Eteplirsen was well tolerated with no treatment-related discontinuations, deaths or evidence of kidney toxicity. Most treatment-emergent adverse events were mild; most common were pyrexia, cough, nasopharyngitis, vomiting, and diarrhea. Eteplirsen pharmacokinetics were consistent between both cohorts and with previous clinical experience in boys with DMD > 4 years of age. These data support the safety and tolerability of eteplirsen at the approved 30-mg/kg dose in boys as young as 6 months old.


Sujet(s)
Myopathie de Duchenne , Mâle , Humains , Enfant d'âge préscolaire , Nourrisson , Myopathie de Duchenne/traitement médicamenteux , Myopathie de Duchenne/génétique , Morpholinos/usage thérapeutique , Exons , Mutation , Dystrophine/génétique
9.
Methods Mol Biol ; 2587: 209-237, 2023.
Article de Anglais | MEDLINE | ID: mdl-36401033

RÉSUMÉ

Antisense oligonucleotides (ASOs) have shown great therapeutic potential in the treatment of many neuromuscular diseases including myotonic dystrophy 1 (DM1). However, systemically delivered ASOs display poor biodistribution and display limited penetration into skeletal muscle. The conjugation of cell-penetrating peptides (CPPs) to phosphorodiamidate morpholino oligonucleotides (PMOs), a class of ASOs with a modified backbone, can be used to enhance ASO skeletal muscle penetration. Peptide-PMOs (P-PMOs) have been shown to be highly effective in correcting the DM1 skeletal muscle phenotype in both murine and cellular models of DM1 and at a molecular and functional level. Here we describe the synthesis and conjugation of P-PMOs and methods for analyzing their biodistribution and toxicity in the HSA-LR DM1 mouse model and their efficacy both in vitro and in vivo using FISH and RT-PCR splicing analysis.


Sujet(s)
Peptides de pénétration cellulaire , Dystrophie myotonique , Souris , Animaux , Morpholinos/génétique , Morpholinos/usage thérapeutique , Morpholinos/composition chimique , Dystrophie myotonique/génétique , Dystrophie myotonique/thérapie , Distribution tissulaire , Oligonucléotides antisens/génétique , Oligonucléotides antisens/usage thérapeutique , Peptides de pénétration cellulaire/composition chimique
10.
Am J Physiol Gastrointest Liver Physiol ; 324(1): G60-G77, 2023 01 01.
Article de Anglais | MEDLINE | ID: mdl-36410025

RÉSUMÉ

Primary sclerosing cholangitis (PSC) is characterized by increased ductular reaction (DR), liver fibrosis, hepatic total bile acid (TBA) levels, and mast cell (MC) infiltration. Apical sodium BA transporter (ASBT) expression increases in cholestasis, and ileal inhibition reduces PSC phenotypes. FVB/NJ and multidrug-resistant 2 knockout (Mdr2-/-) mice were treated with control or ASBT Vivo-Morpholino (VM). We measured 1) ASBT expression and MC presence in liver/ileum; 2) liver damage/DR; 3) hepatic fibrosis/inflammation; 4) biliary inflammation/histamine serum content; and 5) gut barrier integrity/hepatic bacterial translocation. TBA/BA composition was measured in cholangiocyte/hepatocyte supernatants, intestine, liver, serum, and feces. Shotgun analysis was performed to ascertain microbiome changes. In vitro, cholangiocytes were treated with BAs ± ASBT VM, and histamine content and farnesoid X receptor (FXR) signaling were determined. Treated cholangiocytes were cocultured with MCs, and FXR signaling, inflammation, and MC activation were measured. Human patients were evaluated for ASBT/MC expression and histamine/TBA content in bile. Control patient- and PSC patient-derived three-dimensional (3-D) organoids were generated; ASBT, chymase, histamine, and fibroblast growth factor-19 (FGF19) were evaluated. ASBT VM in Mdr2-/- mice decreased 1) biliary ASBT expression, 2) PSC phenotypes, 3) hepatic TBA, and 4) gut barrier integrity compared with control. We found alterations between wild-type (WT) and Mdr2-/- mouse microbiome, and ASBT/MC and bile histamine content increased in cholestatic patients. BA-stimulated cholangiocytes increased MC activation/FXR signaling via ASBT, and human PSC-derived 3-D organoids secrete histamine/FGF19. Inhibition of hepatic ASBT ameliorates cholestatic phenotypes by reducing cholehepatic BA signaling, biliary inflammation, and histamine levels. ASBT regulation of hepatic BA signaling offers a therapeutic avenue for PSC.NEW & NOTEWORTHY We evaluated knockdown of the apical sodium bile acid transporter (ASBT) using Vivo-Morpholino in Mdr2KO mice. ASBT inhibition decreases primary sclerosing cholangitis (PSC) pathogenesis by reducing hepatic mast cell infiltration, altering bile acid species/cholehepatic shunt, and regulating gut inflammation/dysbiosis. Since a large cohort of PSC patients present with IBD, this study is clinically important. We validated findings in human PSC and PSC-IBD along with studies in novel human 3-D organoids formed from human PSC livers.


Sujet(s)
Angiocholite sclérosante , Cholestase , Maladies inflammatoires intestinales , Humains , Animaux , Souris , Angiocholite sclérosante/traitement médicamenteux , Angiocholite sclérosante/génétique , Angiocholite sclérosante/anatomopathologie , Acides et sels biliaires , Histamine , Morpholinos/usage thérapeutique , Foie/métabolisme , Cholestase/anatomopathologie , Cirrhose du foie/anatomopathologie , Inflammation/anatomopathologie , Protéines de transport membranaire , Maladies inflammatoires intestinales/anatomopathologie
11.
Methods Mol Biol ; 2587: 31-41, 2023.
Article de Anglais | MEDLINE | ID: mdl-36401022

RÉSUMÉ

Viltolarsen is a phosphorodiamidate morpholino antisense oligonucleotide (PMO) designed to skip exon 53 of the DMD gene for the treatment of Duchenne muscular dystrophy (DMD), one of the most common lethal genetic disorders characterized by progressive degeneration of skeletal muscles and cardiomyopathy. It was developed by Nippon Shinyaku in collaboration with the National Center of Neurology and Psychiatry (NCNP) in Japan based on the preclinical studies conducted in the DMD dog model at the NCNP. After showing hopeful results in pre-clinical trials and several clinical trials across North America and Japan, it received US Food and Drug Administration (FDA) approval for DMD in 2020. Viltolarsen restores the reading frame of the DMD gene by skipping  exon 53 and produces a truncated but functional form of dystrophin. It can treat approximately 8-10% of the DMD patient population. This paper aims to summarize the development of viltolarsen from preclinical trials to clinical trials to, finally, FDA approval, and discusses the challenges that come with fighting DMD using antisense therapy.


Sujet(s)
Myopathie de Duchenne , Oligonucléotides , États-Unis , Humains , Chiens , Animaux , Food and Drug Administration (USA) , Oligonucléotides/usage thérapeutique , Morpholinos/génétique , Morpholinos/usage thérapeutique , Myopathie de Duchenne/traitement médicamenteux , Myopathie de Duchenne/génétique
12.
Methods Mol Biol ; 2587: 125-139, 2023.
Article de Anglais | MEDLINE | ID: mdl-36401027

RÉSUMÉ

Phosphorodiamidate morpholino oligomer (PMO)-mediated exon skipping is a therapeutic approach that applies to many Duchenne muscular dystrophy (DMD) patients harboring out-of-frame deletion mutations in the DMD gene. In particular, PMOs for skipping exon 44 have been developing in clinical trials, such as the drug NS-089/NCNP-02. Two exon 53 skipping PMOs, golodirsen and viltolarsen, have received conditional approval for treating patients due to their ability to restore dystrophin protein expression. Although promising, further development of exon-skipping technology is needed for patients to have more therapeutic benefit. This chapter describes evaluation methods of exon 44 and 53 skipping PMOs in immortalized DMD patient-derived skeletal muscle cells. We introduce how to quantify exon-skipping efficiencies and dystrophin rescue levels represented by RT-PCR and western blotting, respectively. The screening methods using immortalized patient myotubes can serve to find exon-skipping PMO drug candidates.


Sujet(s)
Dystrophine , Myopathie de Duchenne , Humains , Dystrophine/génétique , Dystrophine/métabolisme , Oligonucléotides antisens/usage thérapeutique , Exons/génétique , Myopathie de Duchenne/génétique , Myopathie de Duchenne/thérapie , Myopathie de Duchenne/métabolisme , Morpholinos/usage thérapeutique , Fibres musculaires squelettiques/métabolisme
13.
Nucleic Acid Ther ; 32(5): 391-400, 2022 10.
Article de Anglais | MEDLINE | ID: mdl-35861718

RÉSUMÉ

We report a novel method to inhibit epidermal growth factor receptor (EGFR) signaling using custom morpholino antisense oligonucleotides (ASOs) to drive expression of dominant negative mRNA isoforms of EGFR by ASO-induced exon skipping within the transmembrane (16) or tyrosine kinase domains (18 and 21). In vivo ASO formulations induced >95% exon skipping in several models of nonsmall cell lung cancer (NSCLC) and were comparable in efficacy to erlotinib in reducing colony formation, cell viability, and migration in EGFR mutant NSCLC (PC9). However, unlike erlotinib, ASOs maintained their efficacy in both erlotinib-resistant subclones (PC9-GR) and wild-type overexpressing EGFR models (H292), in which erlotinib had no significant effect. The most dramatic ASO-induced phenotype resulted from targeting the EGFR kinase domain directly, which resulted in maximal inhibition of phosphorylation of EGFR, Akt, and Erk in both PC9 and PC9GR cells. Phosphoproteomic mass spectrometry confirmed highly congruent impacts of exon 16-, 18-, and 21-directed ASOs compared with erlotinib on PC9 genome-wide cell signaling. Furthermore, EGFR-directed ASOs had no impact in EGFR-independent NSCLC models, confirming an EGFR-specific therapeutic mechanism. Further exploration of synergy of ASOs with existing tyrosine kinase inhibitors may offer novel clinical models to improve EGFR-targeted therapies for both mutant and wild-type NSCLC patients.


Sujet(s)
Carcinome pulmonaire non à petites cellules , Tumeurs du poumon , Humains , Carcinome pulmonaire non à petites cellules/traitement médicamenteux , Carcinome pulmonaire non à petites cellules/génétique , Lignée cellulaire tumorale , Résistance aux médicaments antinéoplasiques/génétique , Récepteurs ErbB/génétique , Récepteurs ErbB/métabolisme , Chlorhydrate d'erlotinib/pharmacologie , Chlorhydrate d'erlotinib/usage thérapeutique , Tumeurs du poumon/traitement médicamenteux , Tumeurs du poumon/génétique , Tumeurs du poumon/métabolisme , Morpholinos/usage thérapeutique , Mutation , Oligonucléotides antisens/génétique , Oligonucléotides antisens/pharmacologie , Oligonucléotides antisens/usage thérapeutique , Inhibiteurs de protéines kinases/pharmacologie , Inhibiteurs de protéines kinases/usage thérapeutique , Protéines proto-oncogènes c-akt/génétique , Protéines proto-oncogènes c-akt/métabolisme , Isoformes d'ARN , Transduction du signal
14.
EMBO Rep ; 23(6): e53955, 2022 06 07.
Article de Anglais | MEDLINE | ID: mdl-35393769

RÉSUMÉ

Duchenne muscular dystrophy (DMD) is a systemic progressive muscular disease caused by frame-disrupting mutations in the DMD gene. Although exon-skipping antisense oligonucleotides (AOs) are clinically approved and can correct DMD, insufficient muscle delivery limits efficacy. If AO activity can be enhanced by safe dietary supplements, clinical trials for efficacy can be undertaken rapidly to benefit patients. We showed previously that intravenous glycine enhanced phosphorodiamidate morpholino oligomer (PMO) delivery to peripheral muscles in mdx mice. Here, we demonstrate that the combination of oral glycine and metformin with intravenous PMO enhances PMO activity, dystrophin restoration, extends lifespan, and improves body-wide function and phenotypic rescue of dystrophin /utrophin double knock-out (DKO) mice without any overt adverse effects. The DKO mice treated with the combination without altering the approved administration protocol of PMO show improved cardio-respiratory and behavioral functions. Metformin and glycine individually are ineffective in DMD patients, but the combination of PMO with clinically-approved oral glycine and metformin might improve the efficacy of the treatment also in DMD patients. Our data suggest that this combination therapy might be an attractive therapy for DMD and potentially other muscle diseases requiring systemic treatment with AOs.


Sujet(s)
Dystrophine , Metformine , Animaux , Dystrophine/génétique , Thérapie génétique/méthodes , Glycine/usage thérapeutique , Humains , Metformine/usage thérapeutique , Souris , Souris de lignée mdx , Morpholinos/génétique , Morpholinos/usage thérapeutique , Muscles squelettiques , Utrophine/génétique
15.
Methods Mol Biol ; 2434: 191-205, 2022.
Article de Anglais | MEDLINE | ID: mdl-35213018

RÉSUMÉ

Antisense oligonucleotides (AONs) are small synthetic molecules of therapeutic interest for a variety of human disease. Their ability to bind mRNA and affect its splicing gives AONs potential use for exon skipping therapies aimed at restoring the dystrophin transcript reading frame for Duchenne muscular dystrophy (DMD) patients. The neutrally charged phosphorodiamidate morpholino oligomers (PMOs) are a stable and relatively nontoxic AON modification. To assess exon skipping efficiency in vitro, it is important to deliver them to target cells. Here, we describe a method for the delivery of PMOs to myoblasts by electroporation. The described protocol for the Amaxa 4D X unit nucleofector system allows efficient processing of 16 samples in one nucleocuvette strip, aiding in high-throughput PMO efficacy screens.


Sujet(s)
Thérapie génétique , Myopathie de Duchenne , Dystrophine/génétique , Dystrophine/métabolisme , Électroporation , Thérapie génétique/méthodes , Humains , Morpholinos/génétique , Morpholinos/usage thérapeutique , Myopathie de Duchenne/traitement médicamenteux , Myopathie de Duchenne/thérapie , Myoblastes/métabolisme
16.
Curr Pharm Biotechnol ; 23(15): 1813-1823, 2022.
Article de Anglais | MEDLINE | ID: mdl-35034590

RÉSUMÉ

The dramatically increasing levels of antibiotic resistance are being seen worldwide and are a significant threat to public health. Antibiotic and drug resistance is seen in various bacterial species. Antibiotic resistance is associated with increased morbidity and mortality and increased treatment costs. Antisense-related technologies include oligonucleotides that interfere with gene transcription and expression; these oligonucleotides can help treat antibiotic-resistant bacteria. The important oligonucleotides include Peptide Nucleic Acids (PNAs), Phosphorodiamidate Morpholino Oligomers (PPMOs), and Locked Nucleic Acids (LNAs). Typically, the size of these structures (oligonucleotides) is 10 to 20 bases. PNAs, PPMOs, and LNAs are highlighted in this review as targets for genes that cause the gene to be destroyed and impede bacterial growth. These results open a new perspective for therapeutic intervention. Future studies need to examine different aspects of antisense agents, such as the safety, toxicity, and pharmacokinetic properties of antisense agents in clinical treatment.


Sujet(s)
Antibactériens , Acides nucléiques peptidiques , Antibactériens/pharmacologie , Antibactériens/usage thérapeutique , Morpholinos/génétique , Morpholinos/usage thérapeutique , Morpholinos/composition chimique , Oligonucléotides antisens/génétique , Oligonucléotides antisens/pharmacologie , Oligonucléotides antisens/usage thérapeutique , Bactéries/génétique , Oligonucléotides
17.
Mol Ther ; 30(3): 1288-1299, 2022 03 02.
Article de Anglais | MEDLINE | ID: mdl-34808387

RÉSUMÉ

Spinal muscular atrophy (SMA) is a motor neuron disease and the leading genetic cause of infant mortality. Recently approved SMA therapies have transformed a deadly disease into a survivable one, but these compounds show a wide spectrum of clinical response and effective rescue only in the early stages of the disease. Therefore, safe, symptomatic-suitable, non-invasive treatments with high clinical impact across different phenotypes are urgently needed. We conjugated antisense oligonucleotides with Morpholino (MO) chemistry, which increase SMN protein levels, to cell-penetrating peptides (CPPs) for better cellular distribution. Systemically administered MOs linked to r6 and (RXRRBR)2XB peptides crossed the blood-brain barrier and increased SMN protein levels remarkably, causing striking improvement of survival, neuromuscular function, and neuropathology, even in symptomatic SMA animals. Our study demonstrates that MO-CPP conjugates can significantly expand the therapeutic window through minimally invasive systemic administration, opening the path for clinical applications of this strategy.


Sujet(s)
Peptides de pénétration cellulaire , Amyotrophie spinale , Animaux , Peptides de pénétration cellulaire/génétique , Modèles animaux de maladie humaine , Humains , Morpholinos/génétique , Morpholinos/usage thérapeutique , Amyotrophie spinale/génétique , Amyotrophie spinale/métabolisme , Amyotrophie spinale/thérapie , Oligonucléotides antisens/génétique , Oligonucléotides antisens/usage thérapeutique , Phénotype
18.
Brasília; CONITEC; 2022.
Non conventionel de Portugais | BRISA/RedTESA | ID: biblio-1443379

RÉSUMÉ

CONDIÇÃO CLÍNICA: A distrofia muscular de Duchenne (DMD) faz parte do espectro das distrofinopatias. As distrofinopatias são um grupo de doenças neuromusculares com herança ligada ao cromossomo X, causadas por mutações no gene que codifica a distrofina (DMD), uma proteína estrutural das células musculares. Em quadros leves das distrofinopatias pode-se notar o aumento assintomático da concentração sérica de creatina fosfoquinase (CK), cãibras musculares e mioglobinúria. Enquanto nos casos graves estão as doenças musculares progressivas, especialmente DMD. A distrofia muscular de Becker, apesar de também ser progressiva e grave, é uma forma de distrofinopatia mais branda comparada à DMD. Ela é caracterizada por fraqueza muscular esquelética de início tardio com gravidade e/ou curso variáveis. TRATAMENTO: Não há cura para a DMD, e as intervenções são baseadas na prevenção e tratamento das complicações. Atualmente, a terapia padrão geralmente é baseada em corticosteróides. Os corticosteróides são capazes de retardar o declínio da força e da função muscular, proporcionando uma redução do risco de escoliose e estabilização da função pulmonar. Pode haver benefícios para a função cardíaca, embora as evidências sejam limitadas. De acordo com uma revisão sistemática Cochrane não há consenso sobre a idade ou estágio funcional para o início do tratamento com corticosteroides sendo necessários mais estudos para determinar as condições ideias para iniciá-lo, ainda que haja autores que defendam que deveria ocorrer antes do declínio físico substancial, normalmente aos 5 anos de idade. .ESTRATÉGIA DE BUSCA: Uma busca foi realizada no banco de dados eletrônico ClinicalTrials.gov, em 14 de julho de 2021. O termo empregado foi: "Duchenne Muscular Dystrophy". Foram considerados os estudos dos medicamentos a partir da fase 1/2 de pesquisa clínica, com a DMD como alvo e sem registro para essa indicação terapêutica no Brasil. Dos 314 cadastros de estudos, 157 atendiam ao critério de elegibilidade referente à fase da pesquisa. Destes, apenas os estudos em andamento ou concluídos nos últimos 5 anos foram selecionados, restando 79 protocolos. A fim de mapear os resultados publicados para esses ensaios clínicos, utilizaram-se os códigos de registro do ClinicalTrials.gov nas bases de dados MEDLINE (via PubMed), EMBASE e Google Acadêmico. Na ausência de resultados, buscas complementares foram feitas com o nome do medicamento associado à indicação. Pipelines de desenvolvimento de medicamentos para DMD também foram revisados. Por fim, os sítios eletrônicos da Agência Nacional de Vigilância Sanitária (Anvisa, Food and Drug Administration (FDA, EUA) e European Medicines Agency (EMA) foram consultadas com o intuito de verificar os registros. O levantamento de resultados publicados e do registro dos medicamentos aconteceu entre 14 de junho e 17 de julho de 2021. MEDICAMENTOS: Ainda assim, os avanços na tecnologia devem ser reconhecidos. De 1998 a 2021, treze oligonucleotídeos antisense foram aprovados pela FDA, sendo quatro deles para DMD. Dos treze, dez receberam aprovação a partir de 2016. Esses resultados, somados aos ensaios clínicos em andamento e aos esforços para amadurecimento das tecnologias, consolidam a perspectiva desta abordagem terapêutica no tratamento da DMD. Por fim, é importante destacar que o pequeno número de pacientes incluídos nos estudos de doenças raras é agravado no caso das estratégias terapêuticas baseadas no genótipo. No caso da DMD, cada uma das tecnologias incluídas neste informe aplica-se a subgrupos com 8-15% do total de pacientes com a doença. INFORMAÇÕES ADICIONAIS: Uma série de tratamentos emergentes para DMD foram identificados durante o mapeamento de tecnologias para este informe, incluindo terapias com oligonucleotídeos antisense, substituição de genes mediada por vírus adeno-associado recombinante (AAVr) e diversos medicamentos para o tratamento sintomático da doença. No entanto, a maior parte encontra-se nas primeiras fases de desenvolvimento, não apresenta resultados publicados ou apenas resultados preliminares foram divulgados. Diante deste cenário, este informe abordou exclusivamente os medicamentos voltados à causa-base da doença e que foram recém-registrados nas agências FDA e/ou EMA. CONSIDERAÇÕES FINAIS: Os quatro oligômeros fosforodiamidato morfolino que receberam aprovação acelerada da FDA para o tratamento de DMD apresentaram aumento da produção de distrofina e parecem ser bem tolerados. No entanto, conforme previsto no Programa de Aprovação Acelerada da FDA, a continuação das aprovações depende da verificação de um benefício clínico em ensaios de confirmação. O eteplirsen e o golodirsen são os únicos oligômeros fosforodiamidato morfolino que apresentam avaliações publicadas acerca do seu efeito funcional. Até a última atualização deste informe, os benefícios clínicos desses medicamentos não estão bem estabelecidos, uma vez que houve declínio na deambulação e função pulmonar dos pacientes tratados, ainda que a maior parte dos valores tenham sido inferiores àqueles encontrados nos controles e na história natural publicada, especialmente para TC6. Para que ocorra a oferta desses medicamentos no SUS, é necessária a análise pela Conitec, conforme disposto na Lei nº 12.401/2011, que alterou a Lei nº 8.080/1990. Os relatórios de recomendação da Conitec levam em consideração as evidências científicas sobre eficácia, a acurácia, a efetividade e a segurança do medicamento, e, também, a avaliação econômica comparativa dos benefícios e dos custos em relação às tecnologias já incorporadas e o impacto da incorporação da tecnologia no SUS.


Sujet(s)
Humains , Myopathie de Duchenne/traitement médicamenteux , Morpholinos/usage thérapeutique , Brésil , Efficacité en Santé Publique , Analyse coût-bénéfice/économie , Projets d'Innovation et de Développement Technologiques
19.
Nat Commun ; 12(1): 4396, 2021 07 20.
Article de Anglais | MEDLINE | ID: mdl-34285203

RÉSUMÉ

Rapid development of antisense therapies can enable on-demand responses to new viral pathogens and make personalized medicine for genetic diseases practical. Antisense phosphorodiamidate morpholino oligomers (PMOs) are promising candidates to fill such a role, but their challenging synthesis limits their widespread application. To rapidly prototype potential PMO drug candidates, we report a fully automated flow-based oligonucleotide synthesizer. Our optimized synthesis platform reduces coupling times by up to 22-fold compared to previously reported methods. We demonstrate the power of our automated technology with the synthesis of milligram quantities of three candidate therapeutic PMO sequences for an unserved class of Duchenne muscular dystrophy (DMD). To further test our platform, we synthesize a PMO that targets the genomic mRNA of SARS-CoV-2 and demonstrate its antiviral effects. This platform could find broad application not only in designing new SARS-CoV-2 and DMD antisense therapeutics, but also for rapid development of PMO candidates to treat new and emerging diseases.


Sujet(s)
Techniques de chimie synthétique/instrumentation , Chimie pharmaceutique/instrumentation , Tests de criblage à haut débit/instrumentation , Morpholinos/synthèse chimique , Oligonucléotides antisens/synthèse chimique , Animaux , COVID-19/virologie , Chlorocebus aethiops , Maladies transmissibles émergentes/traitement médicamenteux , Maladies transmissibles émergentes/microbiologie , Modèles animaux de maladie humaine , Tests de criblage à haut débit/méthodes , Humains , Morpholinos/pharmacologie , Morpholinos/usage thérapeutique , Myopathie de Duchenne/traitement médicamenteux , Myopathie de Duchenne/génétique , Oligonucléotides antisens/pharmacologie , Oligonucléotides antisens/usage thérapeutique , Médecine de précision/méthodes , ARN messager/antagonistes et inhibiteurs , ARN viral/antagonistes et inhibiteurs , SARS-CoV-2/génétique , Facteurs temps , Cellules Vero , Traitements médicamenteux de la COVID-19
20.
J Neuromuscul Dis ; 8(6): 989-1001, 2021.
Article de Anglais | MEDLINE | ID: mdl-34120909

RÉSUMÉ

BackgroundEteplirsen received accelerated FDA approval for treatment of Duchenne muscular dystrophy (DMD) with mutations amenable to exon 51 skipping, based on demonstrated dystrophin production.ObjectiveTo report results from PROMOVI, a phase 3, multicenter, open-label study evaluating efficacy and safety of eteplirsen in a larger cohort.MethodsAmbulatory patients aged 7-16 years, with confirmed mutations amenable to exon 51 skipping, received eteplirsen 30 mg/kg/week intravenously for 96 weeks. An untreated cohort with DMD not amenable to exon 51 skipping was also enrolled.Results78/79 eteplirsen-treated patients completed 96 weeks of treatment. 15/30 untreated patients completed the study; this cohort was considered an inappropriate control group because of genotype-driven differences in clinical trajectory. At Week 96, eteplirsen-treated patients showed increased exon skipping (18.7-fold) and dystrophin protein (7-fold) versus baseline. Post-hoc comparisons with patients from eteplirsen phase 2 studies (4658-201/202) and mutation-matched external natural history controls confirmed previous results, suggesting clinically notable attenuation of decline on the 6-minute walk test over 96 weeks (PROMOVI: -68.9 m; phase 2 studies: -67.3 m; external controls: -133.8 m) and significant attenuation of percent predicted forced vital capacity annual decline (PROMOVI: -3.3%, phase 2 studies: -2.2%, external controls: -6.0%; p < 0.001). Adverse events were generally mild to moderate and unrelated to eteplirsen. Most frequent treatment-related adverse events were headache and vomiting; none led to treatment discontinuation.ConclusionsThis large, multicenter study contributes to the growing body of evidence for eteplirsen, confirming a positive treatment effect, favorable safety profile, and slowing of disease progression versus natural history.


Sujet(s)
Morpholinos/usage thérapeutique , Myopathie de Duchenne/traitement médicamenteux , Adolescent , Enfant , Évolution de la maladie , Dystrophine , Exons , Humains , Mâle , Mutation , Capacité vitale
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...