Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Proc Natl Acad Sci U S A ; 114(16): 4213-4218, 2017 04 18.
Artigo em Inglês | MEDLINE | ID: mdl-28373570

RESUMO

Duchenne muscular dystrophy (DMD) is a lethal genetic disorder caused by an absence of the dystrophin protein in bodywide muscles, including the heart. Cardiomyopathy is a leading cause of death in DMD. Exon skipping via synthetic phosphorodiamidate morpholino oligomers (PMOs) represents one of the most promising therapeutic options, yet PMOs have shown very little efficacy in cardiac muscle. To increase therapeutic potency in cardiac muscle, we tested a next-generation morpholino: arginine-rich, cell-penetrating peptide-conjugated PMOs (PPMOs) in the canine X-linked muscular dystrophy in Japan (CXMDJ) dog model of DMD. A PPMO cocktail designed to skip dystrophin exons 6 and 8 was injected intramuscularly, intracoronarily, or intravenously into CXMDJ dogs. Intravenous injections with PPMOs restored dystrophin expression in the myocardium and cardiac Purkinje fibers, as well as skeletal muscles. Vacuole degeneration of cardiac Purkinje fibers, as seen in DMD patients, was ameliorated in PPMO-treated dogs. Although symptoms and functions in skeletal muscle were not ameliorated by i.v. treatment, electrocardiogram abnormalities (increased Q-amplitude and Q/R ratio) were improved in CXMDJ dogs after intracoronary or i.v. administration. No obvious evidence of toxicity was found in blood tests throughout the monitoring period of one or four systemic treatments with the PPMO cocktail (12 mg/kg/injection). The present study reports the rescue of dystrophin expression and recovery of the conduction system in the heart of dystrophic dogs by PPMO-mediated multiexon skipping. We demonstrate that rescued dystrophin expression in the Purkinje fibers leads to the improvement/prevention of cardiac conduction abnormalities in the dystrophic heart.


Assuntos
Cardiomiopatias/terapia , Peptídeos Penetradores de Células/farmacologia , Distrofina/metabolismo , Éxons , Morfolinos/farmacologia , Distrofia Muscular Animal/terapia , Distrofia Muscular de Duchenne/terapia , Animais , Cardiomiopatias/etiologia , Modelos Animais de Doenças , Cães , Feminino , Terapia Genética , Masculino , Músculo Esquelético/metabolismo , Músculo Esquelético/patologia , Distrofia Muscular Animal/complicações , Distrofia Muscular Animal/genética , Distrofia Muscular de Duchenne/complicações , Distrofia Muscular de Duchenne/genética
2.
N Engl J Med ; 373(4): 339-48, 2015 Jul 23.
Artigo em Inglês | MEDLINE | ID: mdl-26200980

RESUMO

BACKGROUND: AVI-7288 is a phosphorodiamidate morpholino oligomer with positive charges that targets the viral messenger RNA that encodes Marburg virus (MARV) nucleoprotein. Its safety in humans is undetermined. METHODS: We assessed the efficacy of AVI-7288 in a series of studies involving a lethal challenge with MARV in nonhuman primates. The safety of AVI-7288 was evaluated in a randomized, multiple-ascending-dose study in which 40 healthy humans (8 humans per dose group) received 14 once-daily infusions of AVI-7288 (1 mg, 4 mg, 8 mg, 12 mg, or 16 mg per kilogram of body weight) or placebo, in a 3:1 ratio. We estimated the protective dose in humans by comparing pharmacokinetic variables in infected nonhuman primates, uninfected nonhuman primates, and uninfected humans. RESULTS: Survival in infected nonhuman primates was dose-dependent, with survival rates of 0%, 30%, 59%, 87%, 100%, and 100% among monkeys treated with 0 mg, 3.75 mg, 7.5 mg, 15 mg, 20 mg, and 30 mg of AVI-7288 per kilogram, respectively (P<0.001 with the use of the log-rank test for the comparison of survival across groups). No safety concern was identified at doses up to 16 mg per kilogram per day in humans. No serious adverse events were reported. Drug exposure (the area under the curve) was dose-dependent in both nonhuman primates and humans; drug clearance was independent of dose but was higher in nonhuman primates than in humans. The protective dose in humans was initially estimated, on the basis of exposure, to be 9.6 mg per kilogram per day (95% confidence interval, 6.6 to 12.5) for 14 days. Monte Carlo simulations supported a dose of 11 mg per kilogram per day to match the geometric mean protective exposure in nonhuman primates. CONCLUSIONS: This study shows that, on the basis of efficacy in nonhuman primates and pharmacokinetic data in humans, AVI-7288 has potential as postexposure prophylaxis for MARV infection in humans. (Funded by the Department of Defense; ClinicalTrials.gov number, NCT01566877.).


Assuntos
Antivirais/administração & dosagem , Doença do Vírus de Marburg/tratamento farmacológico , Marburgvirus , Morfolinos/administração & dosagem , Animais , Antivirais/efeitos adversos , Antivirais/farmacocinética , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Humanos , Estimativa de Kaplan-Meier , Macaca fascicularis , Doença do Vírus de Marburg/mortalidade , Marburgvirus/genética , Morfolinos/efeitos adversos , Morfolinos/farmacocinética , RNA Mensageiro , RNA Viral
3.
Antimicrob Agents Chemother ; 58(11): 6639-47, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25155593

RESUMO

Two identical single-ascending-dose studies evaluated the safety and pharmacokinetics (PK) of AVI-6002 and AVI-6003, two experimental combinations of phosphorodiamidate morpholino oligomers with positive charges (PMOplus) that target viral mRNA encoding Ebola virus and Marburg virus proteins, respectively. Both AVI-6002 and AVI-6003 were found to suppress disease in virus-infected nonhuman primates in previous studies. AVI-6002 (a combination of AVI-7537 and AVI-7539) or AVI-6003 (a combination of AVI-7287 and AVI-7288) were administered as sequential intravenous (i.v.) infusions of a 1:1 fixed dose ratio of the two subcomponents. In each study, 30 healthy male and female subjects between 18 and 50 years of age were enrolled in six-dose escalation cohorts of five subjects each and received a single i.v. infusion of active study drug (0.005, 0.05, 0.5, 1.5, 3, and 4.5 mg/kg per component) or placebo in a 4:1 ratio. Both AVI-6002 and AVI-6003 were safe and well tolerated at the doses studied. A maximum tolerated dose was not observed in either study. The four chemically similar PMOplus components exhibited generally similar PK profiles. The mean peak plasma concentration and area under the concentration-time curve values of the four components exhibited dose-proportional PK. The estimated plasma half-life of all four components was 2 to 5 h. The safety of the two combinations and the PK of the four components were similar, regardless of the target RNA sequence.


Assuntos
Doença pelo Vírus Ebola/tratamento farmacológico , Doença do Vírus de Marburg/tratamento farmacológico , Morfolinos/farmacocinética , Adulto , Animais , Área Sob a Curva , Método Duplo-Cego , Ebolavirus/efeitos dos fármacos , Ebolavirus/genética , Feminino , Doença pelo Vírus Ebola/virologia , Humanos , Infusões Intravenosas , Masculino , Doença do Vírus de Marburg/virologia , Marburgvirus/efeitos dos fármacos , Marburgvirus/genética , Pessoa de Meia-Idade , Morfolinos/efeitos adversos , Morfolinos/sangue , Placebos , Adulto Jovem
4.
Ann Neurol ; 74(5): 637-47, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23907995

RESUMO

OBJECTIVE: In prior open-label studies, eteplirsen, a phosphorodiamidate morpholino oligomer, enabled dystrophin production in Duchenne muscular dystrophy (DMD) with genetic mutations amenable to skipping exon 51. The present study used a double-blind placebo-controlled protocol to test eteplirsen's ability to induce dystrophin production and improve distance walked on the 6-minute walk test (6MWT). METHODS: DMD boys aged 7 to 13 years, with confirmed deletions correctable by skipping exon 51 and ability to walk 200 to 400 m on 6 MWT, were randomized to weekly intravenous infusions of 30 or 50 mg/kg/wk eteplirsen or placebo for 24 weeks (n = 4/group). Placebo patients switched to 30 or 50 mg/kg eteplirsen (n=2/group) at week 25; treatment was open label thereafter. All patients had muscle biopsies at baseline and week 48. Efficacy included dystrophin-positive fibers and distance walked on the 6MWT. RESULTS: At week 24, the 30 mg/kg eteplirsen patients were biopsied, and percentage of dystrophin-positive fibers was increased to 23% of normal; no increases were detected in placebo-treated patients (p≤0.002). Even greater increases occurred at week 48 (52% and 43% in the 30 and 50 mg/kg cohorts, respectively), suggesting that dystrophin increases with longer treatment. Restoration of functional dystrophin was confirmed by detection of sarcoglycans and neuronal nitric oxide synthase at the sarcolemma. Ambulation-evaluable eteplirsen-treated patients experienced a 67.3 m benefit compared to placebo/delayed patients (p≤0.001). INTERPRETATION: Eteplirsen restored dystrophin in the 30 and 50 mg/kg/wk cohorts, and in subsequently treated, placebo-controlled subjects. Duration, more than dose, accounted for dystrophin production, also resulting in ambulation stability. No severe adverse events were encountered.


Assuntos
Músculo Esquelético/patologia , Distrofia Muscular de Duchenne/tratamento farmacológico , Oligonucleotídeos/uso terapêutico , Adolescente , Criança , Método Duplo-Cego , Distrofina/genética , Humanos , Masculino , Morfolinos , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/patologia , Mutação , Resultado do Tratamento
5.
Cancer Treat Res ; 158: 213-33, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24222360

RESUMO

Alterations in pre-mRNA splicing can have profound effects on gene expression and lead to cellular transformation. Oligonucleotide therapeutics are drugs that manipulate gene expression and improve the disease state. Antisense oligonucleotides hybridize with a target mRNA to downregulate gene expression via an RNase H-dependent mechanism. Additionally, RNase H-independent splice switching oligonucleotides (SSO) modulate alternative or aberrant splicing, to favor the therapeutically relevant splicing product. This chapter summarizes the progress made in the application of these oligonucleotide drugs in the treatment of cancer.


Assuntos
Oligonucleotídeos Antissenso , Oligonucleotídeos , Humanos , Neoplasias/tratamento farmacológico , Oligonucleotídeos Antissenso/uso terapêutico , RNA Mensageiro
6.
Int J Toxicol ; 30(3): 322-33, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21540335

RESUMO

AVI-4658 is a phosphorodiamidate morpholino oligomer (PMO) designed to induce skipping of dystrophin exon 51 and restore its expression in patients with Duchenne muscular dystrophy (DMD). Preclinically, restoration of dystrophin in the dystrophic mdx mouse model requires skipping of exon 23, achieved with the mouse-specific PMO, AVI-4225. Herein, we report the potential toxicological consequences of exon skipping and dystrophin restoration in mdx mice using AVI-4225. We also evaluated the toxicological effects of AVI-4658 in both mdx and wild-type mice. In both studies, animals were dosed once weekly for 12 weeks up to the maximum feasible dose of 960 mg/kg per injection. Both AVI-4658 and AVI-4225 were well-tolerated at all doses. Findings in AVI-4225-treated animals were generally limited to mild renal tubular basophilia/vacuolation, without any significant changes in renal function and with evidence of reversing. No toxicity associated with the mechanism of action of AVI-4225 in a dystrophic animal was observed.


Assuntos
Éxons , Morfolinos/toxicidade , Oligonucleotídeos/toxicidade , Animais , Camundongos , Camundongos Endogâmicos mdx , Tamanho do Órgão/efeitos dos fármacos
7.
Int J Toxicol ; 30(3): 313-21, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21540336

RESUMO

AVI-4658 is a phosphorodiamidate morpholino oligomer (PMO) drug designed to restore dystrophin expression in a subset of patients with Duchenne muscular dystrophy (DMD). Previous reports demonstrated this clinical proof-of-principle in patients with DMD following intramuscular injection of AVI-4658. This preclinical study evaluated the toxicity and toxicokinetic profile of AVI-4658 when administered either intravenously (IV) or subcutaneously (SC) to cynomolgus monkeys once weekly over 12 weeks, at doses up to the maximum feasible dose of 320 mg/kg per injection. No drug-related effects were noted on survival, clinical observations, body weight, food consumption, opthalmoscopic or electrocardiographic evaluations, hematology, clinical chemistry, urinalysis, organ weights, and macroscopic evaluations. Drug-related microscopic renal effects were dose-dependent, apparently reversible, and included basophilic granules (minimal), basophilic tubules (minimal to moderate), and tubular vacuolation (minimal to mild). These data establish the tolerability of AVI-4658 at doses up to and including the maximum feasible dose of 320 mg/kg by IV bolus or SC injection.


Assuntos
Distrofia Muscular de Duchenne/tratamento farmacológico , Oligonucleotídeos/toxicidade , Animais , Sequência de Bases , Feminino , Macaca fascicularis , Masculino , Morfolinos , Oligonucleotídeos/administração & dosagem , Oligonucleotídeos/farmacocinética , Oligonucleotídeos/uso terapêutico
8.
Int J Toxicol ; 29(2): 143-56, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20110565

RESUMO

Duchenne muscular dystrophy (DMD) is caused by dystrophin gene mutations. Restoration of dystrophin by exon skipping was demonstrated with the phosphorodiamidate morpholino oligomers (PMO) class of splice-switching oligomers, in both mouse and dog disease models. The authors report the results of Good Laboratory Practice-compliant safety pharmacology and genotoxicity evaluations of AVI-4658, a PMO under clinical evaluation for DMD. In cynomolgus monkeys, no test article-related effects were seen on cardiovascular, respiratory, global neurological, renal, or liver parameters at the maximum feasible dose (320 mg/kg). Genotoxicity battery showed that AVI-4658 has no genotoxic potential at up to 5000 microg/mL in an in vitro mammalian chromosome aberration test and a bacterial reverse mutation assay. In the mouse bone marrow erythrocyte micronucleus test, a single intravenous injection up to 2000 mg/kg was generally well tolerated and resulted in no mutagenic potential. These results allowed initiation of systemic clinical trials in DMD patients in the United Kingdom.


Assuntos
Mutagênicos/toxicidade , Oligonucleotídeos/farmacologia , Animais , Cães , Macaca fascicularis , Masculino , Camundongos , Morfolinos , Testes de Mutagenicidade , Oligonucleotídeos/toxicidade
9.
Int J Cancer ; 124(4): 772-7, 2009 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-19035464

RESUMO

The oncogene HER2 is overexpressed in a variety of human tumors, providing a target for anti-cancer molecular therapies. Here, we employed a 2'-O-methoxyethyl (MOE) splice switching oligonucleotide, SSO111, to induce skipping of exon 15 in HER2 pre-mRNA, leading to significant downregulation of full-length HER2 mRNA, and simultaneous upregulation of Delta15HER2 mRNA. SSO111 treatment of SK-BR-3 cells, which highly overexpress HER2, led to inhibition of cell proliferation and induction of apoptosis. The novel Delta15HER2 mRNA encodes a soluble, secreted form of the receptor. Treating SK-BR-3 cells with exogenous Delta15HER2 protein reduced membrane-bound HER2 and decreased HER3 transphosphorylation. Delta15HER2 protein thus has similar activity to an autoinhibitory, natural splice variant of HER2, Herstatin, and to the breast cancer drug Herceptin. Both SSO111 and Delta15HER2 may be potential candidates for the development of novel HER2-targeted cancer therapeutics.


Assuntos
Processamento Alternativo , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Precursores de RNA/metabolismo , Receptor ErbB-2/fisiologia , Anticorpos Monoclonais/farmacologia , Anticorpos Monoclonais Humanizados , Antineoplásicos/farmacologia , Apoptose , Linhagem Celular Tumoral , Sobrevivência Celular , Éxons , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/química , Oligonucleotídeos/química , Oligorribonucleotídeos/farmacologia , Fosfatidilinositol 3-Quinases/metabolismo , Fosforilação , Receptor ErbB-2/genética , Trastuzumab
10.
Mol Ther ; 16(7): 1316-1322, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-28178484

RESUMO

Tumor necrosis factor-α (TNF-α) is a key mediator of inflammatory diseases, including rheumatoid arthritis (RA), and anti-TNF-α drugs such as etanercept are effective treatments. Splice-switching oligonucleotides (SSOs) are a new class of drugs designed to induce therapeutically favorable splice variants of targeted genes. In this work, we used locked nucleic acid (LNA)-based SSOs to modulate splicing of TNF receptor 2 (TNFR2) pre-mRNA. The SSO induced skipping of TNFR2 exon 7, which codes the transmembrane domain (TM), switching endogenous expression from the membrane-bound, functional form to a soluble, secreted form (Δ7TNFR2). This decoy receptor protein accumulated in the circulation of treated mice, antagonized TNF-α, and altered disease in two mouse models: TNF-α-induced hepatitis and collagen-induced arthritis (CIA). This is the first report of upregulation of the endogenous, circulating TNF-α antagonist by oligonucleotide-induced splicing modulation.

11.
Mol Ther ; 16(9): 1624-9, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18545222

RESUMO

Cell-penetrating peptides (CPPs), containing arginine (R), 6-aminohexanoic acid (X), and/or beta-alanine (B) conjugated to phosphorodiamidate morpholino oligomers (PMOs), enhance their delivery in cell culture. In this study, the potency, functional biodistribution, and toxicity of these conjugates were evaluated in vivo, in EGFP-654 transgenic mice that ubiquitously express the aberrantly spliced EGFP-654 pre-mRNA reporter. Correct splicing and enhanced green fluorescence protein (EGFP) upregulation serve as a positive readout for peptide-PMO (PPMO) entry into cells and access to EGFP-654 pre-mRNA in the nucleus. Intraperitoneal injections of a series of PPMOs, A-N (12 mg/kg), administered once a day for four successive days resulted in splicing correction in numerous tissues. PPMO-B was highly potent in the heart, diaphragm, and quadriceps, which are key muscles in the treatment of Duchenne muscular dystrophy. We therefore investigated PPMO M23D-B, designed to force skipping of stop-codon containing dystrophin exon 23, in an mdx mouse model of the disease. Systemic delivery of M23D-B yielded persistent exon 23 skipping, yielding high and sustained dystrophin protein expression in body-wide muscles, including cardiac muscle, without detectable toxicity. The rescued dystrophin reduced serum creatinine kinase to near-wild-type levels, indicating improvement in muscle integrity. This is the first report of oligonucleotide-mediated exon skipping and dystrophin protein induction in the heart of treated animals.


Assuntos
Distrofina/fisiologia , Morfolinas/uso terapêutico , Músculo Esquelético/efeitos dos fármacos , Músculo Esquelético/metabolismo , Distrofia Muscular Animal/terapia , Oligonucleotídeos/uso terapêutico , Fragmentos de Peptídeos/uso terapêutico , Alanina Transaminase/sangue , Animais , Aspartato Aminotransferases/sangue , Imunofluorescência , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos mdx , Camundongos Knockout , Morfolinas/administração & dosagem , Distrofia Muscular Animal/genética , Distrofia Muscular Animal/patologia , Oligonucleotídeos/administração & dosagem , Oligonucleotídeos/genética
12.
Mol Ther ; 16(7): 1316-22, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18461057

RESUMO

Tumor necrosis factor-alpha (TNF-alpha) is a key mediator of inflammatory diseases, including rheumatoid arthritis (RA), and anti-TNF-alpha drugs such as etanercept are effective treatments. Splice-switching oligonucleotides (SSOs) are a new class of drugs designed to induce therapeutically favorable splice variants of targeted genes. In this work, we used locked nucleic acid (LNA)-based SSOs to modulate splicing of TNF receptor 2 (TNFR2) pre-mRNA. The SSO induced skipping of TNFR2 exon 7, which codes the transmembrane domain (TM), switching endogenous expression from the membrane-bound, functional form to a soluble, secreted form (Delta7TNFR2). This decoy receptor protein accumulated in the circulation of treated mice, antagonized TNF-alpha, and altered disease in two mouse models: TNF-alpha-induced hepatitis and collagen-induced arthritis (CIA). This is the first report of upregulation of the endogenous, circulating TNF-alpha antagonist by oligonucleotide-induced splicing modulation.


Assuntos
Artrite Experimental/tratamento farmacológico , Hepatite/tratamento farmacológico , Oligonucleotídeos/uso terapêutico , Sítios de Splice de RNA , Receptores Tipo II do Fator de Necrose Tumoral/genética , Fator de Necrose Tumoral alfa/antagonistas & inibidores , Animais , Linhagem Celular , Modelos Animais de Doenças , Hepatócitos/metabolismo , Humanos , Camundongos , Camundongos Endogâmicos , Receptores Tipo II do Fator de Necrose Tumoral/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Regulação para Cima
13.
J Clin Invest ; 112(4): 481-6, 2003 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-12925686

RESUMO

An estimated 60% of all human genes undergo alternative splicing, a highly regulated process that produces splice variants with different functions. Such variants have been linked to a variety of cancers, and genetic diseases such as thalassemia and cystic fibrosis. This Perspective describes a promising approach to RNA repair based on the use of antisense oligonucleotides to modulate alternative splicing and engender the production of therapeutic gene products.


Assuntos
Processamento Alternativo , Antígenos de Superfície , Oligonucleotídeos Antissenso/uso terapêutico , Carboxipeptidases/genética , Regulador de Condutância Transmembrana em Fibrose Cística/genética , Distrofina/genética , Globinas/genética , Glutamato Carboxipeptidase II , Humanos , Ligantes , Modelos Biológicos , Modelos Genéticos , Doenças Renais Policísticas/genética , Proteínas Proto-Oncogênicas c-bcl-2/genética , Proteínas Proto-Oncogênicas c-myc/genética , RNA Mensageiro/metabolismo , Receptor de Insulina/genética , Receptores de GABA-A/genética , Receptores de Interleucina/genética , Receptores de Interleucina-5 , Proteína bcl-X , Proteínas tau/genética
14.
Nat Biotechnol ; 20(12): 1228-33, 2002 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-12426578

RESUMO

Systemically injected 2'-O-methoxyethyl (2'-O-MOE)-phosphorothioate and PNA-4K oligomers (peptide nucleic acid with four lysines linked at the C terminus) exhibited sequence-specific antisense activity in a number of mouse organs. Morpholino oligomers were less effective, whereas PNA oligomers with only one lysine (PNA-1K) were completely inactive. The latter result indicates that the four-lysine tail is essential for the antisense activity of PNA oligomers in vivo. These results were obtained in a transgenic mouse model designed as a positive readout test for activity, delivery, and distribution of antisense oligomers. In this model, the expressed gene (EGFP-654) encoding enhanced green fluorescence protein (EGFP) is interrupted by an aberrantly spliced mutated intron of the human beta-globin gene. Aberrant splicing of this intron prevented expression of EGFP-654 in all tissues, whereas in tissues and organs that took up a splice site-targeted antisense oligomer, correct splicing was restored and EGFP-654 expression upregulated. The sequence-specific ability of PNA-4K and the 2'-O-MOE oligomers to upregulate EGFP-654 provides strong evidence that systemically delivered, chemically modified oligonucleotides affect gene expression by sequence-specific true antisense activity, validating their application as potential therapeutics.


Assuntos
Fibroblastos/fisiologia , Regulação da Expressão Gênica/efeitos dos fármacos , Fígado/fisiologia , Oligonucleotídeos Antissenso/administração & dosagem , Animais , Células Cultivadas , Fibroblastos/efeitos dos fármacos , Terapia Genética/métodos , Injeções Intravenosas , Fígado/efeitos dos fármacos , Camundongos , Camundongos Transgênicos , Oligonucleotídeos Antissenso/farmacocinética , Especificidade de Órgãos , Ácidos Nucleicos Peptídicos/administração & dosagem , Ácidos Nucleicos Peptídicos/farmacocinética , Distribuição Tecidual , Regulação para Cima/efeitos dos fármacos
15.
World J Pediatr ; 13(3): 197-201, 2017 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-28466241

RESUMO

BACKGROUND: Newborn screening for Duchenne muscular dystrophy (DMD) is currently being initiated in Zhejiang Province, China and is under consideration in other countries, including the United States. As China begins to implement DMD newborn screening (DMD-NBS), there is ongoing discussion regarding the steps forward for follow up care of positively identified patients as well as false positive and false negative results. DATA SOURCES: Relevant papers related to DMD-NBS, and NBS in China were reviewed in PubMed. RESULTS: The current state of DMD-NBS is discussed, along with the steps needed to effectively screen infants for this disease in China, recommendations for establishment of follow up care in patients with positive and negative screens, and measurement of patient outcomes. CONCLUSIONS: Zhejiang Province, China is ready to implement DMD-NBS. Future challenges that exist for this program, and other countries, include the ability to track patients, assist with access to care, and ensure adequate follow-up care according to evidence-based guidelines. In addition, China's large rural population, lack of specialty providers, and difficulty in educating patients regarding the benefits of treatment create challenges that will need to be addressed.


Assuntos
Distrofia Muscular de Duchenne/diagnóstico , Triagem Neonatal/métodos , China , Humanos , Recém-Nascido
16.
Nucleic Acids Res ; 30(21): 4592-8, 2002 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-12409448

RESUMO

A T-->G mutation at nucleotide 705 of human beta-globin intron 2 creates an aberrant 5' splice site and activates a cryptic 3' splice site upstream. In consequence, the pre-mRNA is spliced via aberrant splice sites, despite the presence of the still functional correct sites. Surprisingly, when IVS2-705 HeLa or K562 cells were cultured at temperatures below 30 degrees C, aberrant splicing was inhibited and correct splicing was restored. Similar temperature effects were seen for another beta-globin pre-mRNA, IVS2-745, and in a construct in which a beta-globin intron was inserted into a coding sequence of EGFP. Temperature-induced alternative splicing was affected by the nature of the internal aberrant splice sites flanking the correct sites and by exonic sequences. The results indicate that in the context of thalassemic splicing mutations and possibly in other alternatively spliced pre-mRNAs, temperature is one of the parameters that affect splice site selection.


Assuntos
Globinas/genética , Precursores de RNA/metabolismo , Sítios de Splice de RNA/genética , Splicing de RNA , RNA Mensageiro/metabolismo , Temperatura , Células HeLa , Humanos , Íntrons/genética , Células K562 , Mutação/genética , Precursores de RNA/genética , RNA Mensageiro/genética
17.
J Neuromuscul Dis ; 3(3): 381-393, 2016 08 30.
Artigo em Inglês | MEDLINE | ID: mdl-27854228

RESUMO

BACKGROUND: Phosphorodiamidate morpholino oligomers (PMOs) are a class of exon skipping drugs including eteplirsen, which has shown considerable promise for treatment of the degenerative neuromuscular disease, Duchenne musculardystrophy (DMD). OBJECTIVE: Toxicity studies in non-human primates (NHPs) of 12 weeks duration with two new PMOs for DMD, SRP-4045 and SRP-4053, along with results from a chronic study in NHPs of 39 weeks duration for eteplirsen, are described here. METHODS: PMOs were administered once-weekly by bolus intravenous (IV) injections to male NHPs. Endpoints evaluated included plasma exposures, clinical observations, body weight/food consumption, eye exams, electrocardiograms, male reproductive hormones/endpoints, complement alternative pathway, clinical pathology, urinalysis, and macroscopic/light microscopic pathology. RESULTS: Findings in these studies were limited to the kidneys, with a common presentation of tubular basophilia, vacuolation, and/or minimal degeneration that was considered non-adverse. No necrosis, glomerular lesions, or effects on renal function tests such as serum creatinine or urea nitrogen were observed, suggesting that PMO-related kidney findings are not likely to develop into frank nephrotoxicity. There were no adverse effects on other potential target organs after repeated IV injections at the highest dose levels tested, 320 mg/kg. CONCLUSIONS: Nonclinical results in NHPs for these three PMOs, together with the excellent clinical safety established for eteplirsen to date, suggest that once-weekly IV administration of PMOs for lifetime durations at therapeutic doses will be well tolerated by patients with DMD.


Assuntos
Rim/efeitos dos fármacos , Morfolinos/toxicidade , Distrofia Muscular de Duchenne/tratamento farmacológico , Animais , Basófilos/efeitos dos fármacos , Basófilos/patologia , Peso Corporal/efeitos dos fármacos , Eletrocardiografia , Éxons , Coração/efeitos dos fármacos , Túbulos Renais/efeitos dos fármacos , Túbulos Renais/patologia , Macaca fascicularis , Masculino , Vacúolos/efeitos dos fármacos , Vacúolos/patologia
18.
Curr Opin Biotechnol ; 13(5): 468-72, 2002 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-12459339

RESUMO

To achieve effective modulation of gene expression by antisense oligonucleotides, novel oligonucleotide chemistries that do not promote RNase H degradation of target RNA are needed. In addition to short-term oligonucleotide effects, long-term gene regulation can be accomplished by intracellularly expressed antisense RNAs delivered by viral vectors.


Assuntos
Regulação da Expressão Gênica/efeitos dos fármacos , Oligonucleotídeos Antissenso/química , Oligonucleotídeos Antissenso/genética , Biossíntese de Proteínas , RNA Antissenso/química , RNA Antissenso/genética , Terapia Genética/métodos , Oligonucleotídeos Antissenso/farmacologia , Processamento Pós-Transcricional do RNA/genética , RNA Antissenso/farmacologia , Ribonuclease H/química , Ribonuclease H/genética , Ribonuclease H/metabolismo
19.
Cardiovasc Res ; 85(3): 444-53, 2010 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-19815563

RESUMO

AIMS: The cardiomyopathy found in Duchenne muscular dystrophy (DMD) is responsible for death due to heart failure in approximately 30% of patients and additionally contributes to many DMD morbidities. Strategies to bypass DMD-causing mutations to allow an increase in body-wide dystrophin have proved promising, but increasing cardiac dystrophin continues to be challenging. The purpose of this study was to determine if therapeutic restoration of cardiac dystrophin improved the significant cardiac hypertrophy and diastolic dysfunction identified in X-linked muscular dystrophy (mdx) dystrophin-null mouse due to a truncation mutation over time after treatment. METHODS AND RESULTS: Mice lacking dystrophin due to a truncation mutation (mdx) were given an arginine-rich, cell-penetrating, peptide-conjugated phosphorodiamidate morpholino oligomer (PPMO) that delivered a splice-switching oligonucleotide-mediated exon skipping therapy to restore dystrophin in mdx mice before the development of detectable cardiomyopathy. PPMO successfully restored cardiac dystrophin expression, preserved cardiac sarcolemma integrity, and prevented the development of cardiac pathology that develops in mdx-null mice over time. By echocardiography and Doppler analysis of the mitral valve, we identified that PPMO treatment of mdx mice prevented the cardiac hypertrophy and diastolic dysfunction identified in sham-treated, age-matched mdx mice, characteristic of DMD patients early in the disease process, in as little as 5-6 weeks after the initiation of treatment. Surprisingly, despite the short-term replacement of cardiac dystrophin (<1% present after 12 weeks by immunodetection), PPMO therapy also provided a durable cardiac improvement in cardiac hypertrophy and diastolic dysfunction for up to 7 months after the initiation of treatment. CONCLUSION: These results demonstrate for the first time that PPMO-mediated exon skipping therapy early in the course of DMD may effectively prevent or slow down associated cardiac hypertrophy and diastolic dysfunction with significant long-term impact.


Assuntos
Cardiomiopatias/terapia , Distrofina/genética , Terapia Genética/métodos , Morfolinas/administração & dosagem , Distrofia Muscular de Duchenne/terapia , Animais , Cardiomegalia/prevenção & controle , Diástole , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos mdx , Morfolinos , Distrofia Muscular de Duchenne/complicações , Distrofia Muscular de Duchenne/genética
20.
Ann N Y Acad Sci ; 1175: 55-60, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19796077

RESUMO

Steric-blocking oligos can correct reading frame errors or skip premature termination codons. For Duchenne muscular dystrophy (DMD), systemic administration of oligos produces limited delivery into muscle cells. Conjugation to a cell-penetrating peptide greatly enhances muscle uptake of morpholino oligos. A peptide-morpholino conjugate (PPMO) restored dystrophin in mdx mice to > 80% and 50% of normal levels in skeletal and cardiac muscles, respectively, after a single intravenous 30-mg/kg injection. Six injections over 3 months restored dystrophin to nearly normal levels in all muscles. One PPMO injection daily at 12 mg/kg each for 4 days caused exon skipping clearly detectable in the muscles of the mdx mice 9 weeks later, showing prolonged activity. PPMO significantly improved muscle pathology, strength and function, and the survival rate of mice whose hearts were challenged by chemical-induced heart failure. No toxicity or immunogenicity was detected. Our studies demonstrated that muscle functions can be restored with a low dose of PPMO, making it a promising therapeutic for DMD.


Assuntos
Distrofina/genética , Morfolinas/uso terapêutico , Distrofia Muscular de Duchenne/terapia , Animais , Códon sem Sentido/genética , Modelos Animais de Doenças , Camundongos , Camundongos Endogâmicos mdx , Morfolinas/química , Morfolinas/metabolismo , Distrofia Muscular de Duchenne/genética , Oligonucleotídeos/química , Oligonucleotídeos/metabolismo , Oligonucleotídeos/uso terapêutico , Peptídeos/química , Peptídeos/metabolismo , Peptídeos/uso terapêutico
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA