Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
Mais filtros

Base de dados
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
PLoS Pathog ; 17(7): e1009544, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-34265018

RESUMO

SARS-CoV-2 variants have emerged with enhanced pathogenicity and transmissibility, and escape from pre-existing immunity, suggesting first-generation vaccines and monoclonal antibodies may now be less effective. Here we present an approach for preventing clinical sequelae and the spread of SARS-CoV-2 variants. First, we affinity matured an angiotensin-converting enzyme 2 (ACE2) decoy protein, achieving 1000-fold binding improvements that extend across a wide range of SARS-CoV-2 variants and distantly related, ACE2-dependent coronaviruses. Next, we demonstrated the expression of this decoy in proximal airway when delivered via intranasal administration of an AAV vector. This intervention significantly diminished clinical and pathologic consequences of SARS-CoV-2 challenge in a mouse model and achieved therapeutic levels of decoy expression at the surface of proximal airways when delivered intranasally to nonhuman primates. Importantly, this long-lasting, passive protection approach is applicable in vulnerable populations such as the elderly and immune-compromised that do not respond well to traditional vaccination. This approach could be useful in combating COVID-19 surges caused by SARS-CoV-2 variants and should be considered as a countermeasure to future pandemics caused by one of the many pre-emergent, ACE2-dependent CoVs that are poised for zoonosis.


Assuntos
Enzima de Conversão de Angiotensina 2 , Tratamento Farmacológico da COVID-19 , Dependovirus , Terapia Genética , Vetores Genéticos , SARS-CoV-2 , Administração Intranasal , Enzima de Conversão de Angiotensina 2/biossíntese , Enzima de Conversão de Angiotensina 2/genética , Animais , COVID-19/genética , COVID-19/metabolismo , Humanos , Camundongos , Camundongos Transgênicos , SARS-CoV-2/genética , SARS-CoV-2/metabolismo
2.
Hum Mol Genet ; 26(19): 3837-3849, 2017 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-28934395

RESUMO

The mucopolysaccharidoses (MPS) are rare genetic disorders marked by severe somatic and neurological symptoms. Development of treatments for the neurological manifestations of MPS has been hindered by the lack of objective measures of central nervous system disease burden. Identification of biomarkers for central nervous system disease in MPS patients would facilitate the evaluation of new agents in clinical trials. High throughput metabolite screening of cerebrospinal fluid (CSF) samples from a canine model of MPS I revealed a marked elevation of the polyamine, spermine, in affected animals, and gene therapy studies demonstrated that reduction of CSF spermine reflects correction of brain lesions in these animals. In humans, CSF spermine was elevated in neuropathic subtypes of MPS (MPS I, II, IIIA, IIIB), but not in subtypes in which cognitive function is preserved (MPS IVA, VI). In MPS I patients, elevated CSF spermine was restricted to patients with genotypes associated with CNS disease and was reduced following hematopoietic stem cell transplantation, which is the only therapy currently capable of improving cognitive outcomes. Additional studies in cultured neurons from MPS I mice showed that elevated spermine was essential for the abnormal neurite overgrowth exhibited by MPS neurons. These findings offer new insights into the pathogenesis of CNS disease in MPS patients, and support the use of spermine as a new biomarker to facilitate the development of next generation therapeutics for MPS.


Assuntos
Mucopolissacaridoses/metabolismo , Poliaminas/metabolismo , Adolescente , Animais , Biomarcadores/líquido cefalorraquidiano , Doenças do Sistema Nervoso Central/diagnóstico , Criança , Modelos Animais de Doenças , Cães , Terapia de Reposição de Enzimas/métodos , Feminino , Terapia Genética/métodos , Humanos , Masculino , Camundongos , Mucopolissacaridoses/líquido cefalorraquidiano , Mucopolissacaridose I/líquido cefalorraquidiano , Mucopolissacaridose I/diagnóstico , Mucopolissacaridose I/metabolismo , Espermina/análise , Espermina/líquido cefalorraquidiano , Espermina/química
3.
J Immunol ; 198(12): 4581-4587, 2017 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-28500077

RESUMO

CTL responses to the transgene product remain an active area of concern for the gene therapy field. A patient's underlying genetic mutation may influence the qualitative nature of these potentially destructive T cell responses. Individuals with a mutation that introduces a premature termination codon (PTC) that prevents synthesis of the full-length peptide are considered more likely to mount a transgene-specific T cell response because of a lack of immune tolerance to C-terminal epitopes as a consequence of absent endogenous Ag presentation. In this article, we demonstrate that a human ornithine transcarbamylase gene containing various PTC-inducing non-sense mutations is able to generate and present epitopes downstream of the termination codon. Generation of these epitopes occurs primarily from alternative translation start sites downstream of the stop codon. Furthermore, we show that expression of these genes from adeno-associated virus vectors in C57BL/6 mice is able to induce peripheral tolerance to epitopes downstream of the PTC. These results suggest that, despite the lack of full-length endogenous protein, patients with PTC-inducing non-sense mutations may still present T cell epitopes downstream of the premature termination site that may render the subject tolerant to wild-type transgene products.


Assuntos
Apresentação de Antígeno , Códon sem Sentido , Códon de Terminação , Epitopos de Linfócito T/imunologia , Tolerância Imunológica , Ornitina Carbamoiltransferase/genética , Animais , Linfócitos T CD8-Positivos , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Ornitina Carbamoiltransferase/metabolismo , Peptídeos/genética , Peptídeos/imunologia , Linfócitos T Citotóxicos
4.
Mol Ther ; 24(2): 206-216, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26447927

RESUMO

Mucopolysaccharidosis VII (MPS VII) is a lysosomal storage disease arising from mutations in ß-d-glucuronidase (GUSB), which results in glycosaminoglycan (GAG) accumulation and a variety of clinical manifestations including neurological disease. Herein, MPS VII dogs were injected intravenously (i.v.) and/or intrathecally (i.t.) via the cisterna magna with AAV9 or AAVrh10 vectors carrying the canine GUSB cDNA. Although i.v. injection alone at 3 days of age resulted in normal cerebrospinal fluid (CSF) GUSB activity, brain tissue homogenates had only ~1 to 6% normal GUSB activity and continued to have elevated GAG storage. In contrast, i.t. injection at 3 weeks of age resulted in CSF GUSB activity 44-fold normal while brain tissue homogenates had >100% normal GUSB activity and reduced GAGs compared with untreated dogs. Markers for secondary storage and inflammation were eliminated in i.t.-treated dogs and reduced in i.v.-treated dogs compared with untreated dogs. Given that i.t.-treated dogs expressed higher levels of GUSB in the CNS tissues compared to those treated i.v., we conclude that i.t. injection of AAV9 or AAVrh10 vectors is more effective than i.v. injection alone in the large animal model of MPS VII.


Assuntos
Doenças do Sistema Nervoso Central/terapia , Terapia Genética/métodos , Glucuronidase/genética , Mucopolissacaridose VII/terapia , Animais , Animais Recém-Nascidos , Doenças do Sistema Nervoso Central/genética , Doenças do Sistema Nervoso Central/metabolismo , Dependovirus/genética , Modelos Animais de Doenças , Cães , Vetores Genéticos/administração & dosagem , Glucuronidase/líquido cefalorraquidiano , Glicosaminoglicanos/metabolismo , Injeções Intravenosas , Injeções Espinhais , Masculino , Mucopolissacaridose VII/complicações , Mucopolissacaridose VII/genética , Mucopolissacaridose VII/metabolismo
5.
Proc Natl Acad Sci U S A ; 111(41): 14894-9, 2014 Oct 14.
Artigo em Inglês | MEDLINE | ID: mdl-25267637

RESUMO

Patients with mucopolysaccharidosis type I (MPS I), a genetic deficiency of the lysosomal enzyme α-l-iduronidase (IDUA), exhibit accumulation of glycosaminoglycans in tissues, with resulting diverse clinical manifestations including neurological, ocular, skeletal, and cardiac disease. MPS I is currently treated with hematopoietic stem cell transplantation or weekly enzyme infusions, but these therapies have significant drawbacks for patient safety and quality of life and do not effectively address some of the most critical clinical sequelae, such as life-threatening cardiac valve involvement. Using the naturally occurring feline model of MPS I, we tested liver-directed gene therapy as a means of achieving long-term systemic IDUA reconstitution. We treated four MPS I cats at 3-5 mo of age with an adeno-associated virus serotype 8 vector expressing feline IDUA from a liver-specific promoter. We observed sustained serum enzyme activity for 6 mo at ∼ 30% of normal levels in one animal, and in excess of normal levels in three animals. Remarkably, treated animals not only demonstrated reductions in glycosaminoglycan storage in most tissues, but most also exhibited complete resolution of aortic valve lesions, an effect that has not been previously observed in this animal model or in MPS I patients treated with current therapies. These data point to clinically meaningful benefits of the robust enzyme expression achieved with hepatic gene transfer that extend beyond the economic and quality of life advantages over lifelong enzyme infusions.


Assuntos
Doenças Cardiovasculares/terapia , Terapia Genética , Fígado/metabolismo , Mucopolissacaridose I/terapia , Animais , Valva Aórtica/metabolismo , Valva Aórtica/patologia , Doenças Cardiovasculares/patologia , Gatos , Dependovirus/genética , Feminino , Vetores Genéticos/metabolismo , Glicosaminoglicanos/metabolismo , Cofator II da Heparina/metabolismo , Iduronidase/sangue , Iduronidase/genética , Iduronidase/uso terapêutico , Fígado/patologia , Masculino , Dados de Sequência Molecular , Mucopolissacaridose I/sangue , Mucopolissacaridose I/patologia , Miocárdio/metabolismo , Miocárdio/patologia , Trombina/metabolismo , Distribuição Tecidual , Transdução Genética
6.
Mol Genet Metab ; 119(1-2): 124-30, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27386755

RESUMO

High fidelity animal models of human disease are essential for preclinical evaluation of novel gene and protein therapeutics. However, these studies can be complicated by exaggerated immune responses against the human transgene. Here we demonstrate that dogs with a genetic deficiency of the enzyme α-l-iduronidase (IDUA), a model of the lysosomal storage disease mucopolysaccharidosis type I (MPS I), can be rendered immunologically tolerant to human IDUA through neonatal exposure to the enzyme. Using MPS I dogs tolerized to human IDUA as neonates, we evaluated intrathecal delivery of an adeno-associated virus serotype 9 vector expressing human IDUA as a therapy for the central nervous system manifestations of MPS I. These studies established the efficacy of the human vector in the canine model, and allowed for estimation of the minimum effective dose, providing key information for the design of first-in-human trials. This approach can facilitate evaluation of human therapeutics in relevant animal models, and may also have clinical applications for the prevention of immune responses to gene and protein replacement therapies.


Assuntos
Terapia de Reposição de Enzimas , Iduronidase/genética , Doenças por Armazenamento dos Lisossomos/terapia , Mucopolissacaridose I/terapia , Animais , Modelos Animais de Doenças , Cães , Terapia Genética , Vetores Genéticos , Glicosaminoglicanos/metabolismo , Humanos , Iduronidase/deficiência , Iduronidase/uso terapêutico , Doenças por Armazenamento dos Lisossomos/genética , Doenças por Armazenamento dos Lisossomos/patologia , Mucopolissacaridose I/genética , Mucopolissacaridose I/patologia , Transgenes
7.
Mol Ther ; 23(8): 1298-1307, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26022732

RESUMO

The potential host immune response to a nonself protein poses a fundamental challenge for gene therapies targeting recessive diseases. We demonstrate in both dogs and nonhuman primates that liver-directed gene transfer using an adeno-associated virus (AAV) vector in neonates induces a persistent state of immunological tolerance to the transgene product, substantially improving the efficacy of subsequent vector administration targeting the central nervous system (CNS). We applied this approach to a canine model of mucopolysaccharidosis type I (MPS I), a progressive neuropathic lysosomal storage disease caused by deficient activity of the enzyme α-l-iduronidase (IDUA). MPS I dogs treated systemically in the first week of life with a vector expressing canine IDUA did not develop antibodies against the enzyme and exhibited robust expression in the CNS upon intrathecal AAV delivery at 1 month of age, resulting in complete correction of brain storage lesions. Newborn rhesus monkeys treated systemically with AAV vector expressing human IDUA developed tolerance to the transgene, resulting in high cerebrospinal fluid (CSF) IDUA expression and no antibody induction after subsequent CNS gene therapy. These findings suggest that inducing tolerance to the transgene product during a critical period in immunological development can improve the efficacy and safety of gene therapy.


Assuntos
Sistema Nervoso Central/metabolismo , Dependovirus/genética , Terapia Genética/métodos , Iduronidase/genética , Mucopolissacaridose I/genética , Mucopolissacaridose I/terapia , Animais , Animais Recém-Nascidos , Modelos Animais de Doenças , Cães , Feminino , Técnicas de Transferência de Genes , Vetores Genéticos , Células HEK293 , Humanos , Iduronidase/deficiência , Macaca mulatta , Transgenes
8.
Blood ; 121(12): 2224-33, 2013 Mar 21.
Artigo em Inglês | MEDLINE | ID: mdl-23325831

RESUMO

Recent clinical trials have shown that evasion of CD8(+) T-cell responses against viral capsid is critical for successful liver-directed gene therapy with adeno-associated viral (AAV) vectors for hemophilia. Preclinical models to test whether use of alternate serotypes or capsid variants could avoid this deleterious response have been lacking. Here, the ability of CD8(+) T cells ("cap-CD8," specific for a capsid epitope presented by human B*0702 or murine H2-L(d) molecules) to target AAV-infected hepatocytes was investigated. In a murine model based on adoptive transfer of ex vivo expanded cap-CD8, AAV2-transduced livers showed CD8(+) T-cell infiltrates, transaminitis, significant reduction in factor IX transgene expression, and loss of transduced hepatocytes. AAV8 gene transfer resulted in prolonged susceptibility to cap-CD8, consistent with recent clinical findings. In contrast, using an AAV2(Y-F) mutant capsid, which is known to be less degraded by proteasomes, preserved transgene expression and largely avoided hepatotoxicity. In vitro assays confirmed reduced major histocompatibility complex class I presentation of this capsid and killing of human or murine hepatocytes compared with AAV2. In conclusion, AAV capsids can be engineered to substantially reduce the risk of destruction by cytotoxic T lymphocytes, whereas use of alternative serotypes per se does not circumvent this obstacle.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Proteínas do Capsídeo/imunologia , Dependovirus/fisiologia , Terapia Genética/métodos , Vetores Genéticos/fisiologia , Hepatócitos/imunologia , Transferência Adotiva/métodos , Animais , Linfócitos T CD8-Positivos/metabolismo , Proteínas do Capsídeo/genética , Proteínas do Capsídeo/metabolismo , Células Cultivadas , Dependovirus/genética , Dependovirus/imunologia , Dependovirus/metabolismo , Engenharia Genética , Vetores Genéticos/genética , Hepatócitos/metabolismo , Humanos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Especificidade do Receptor de Antígeno de Linfócitos T/genética , Transdução Genética
9.
Mol Ther ; 22(12): 2018-2027, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25027660

RESUMO

Enzyme replacement therapy has revolutionized the treatment of the somatic manifestations of lysosomal storage diseases (LSD), although it has been ineffective in treating central nervous system (CNS) manifestations of these disorders. The development of neurotrophic vectors based on novel serotypes of adeno-associated viruses (AAV) such as AAV9 provides a potential platform for stable and efficient delivery of enzymes to the CNS. We evaluated the safety and efficacy of intrathecal delivery of AAV9 expressing α-l-iduronidase (IDUA) in a previously described feline model of mucopolysaccharidosis I (MPS I). A neurological phenotype has not been defined in these animals, so our analysis focused on the biochemical and histological CNS abnormalities characteristic of MPS I. Five MPS I cats were dosed with AAV9 vector at 4-7 months of age and followed for 6 months. Treated animals demonstrated virtually complete correction of biochemical and histological manifestations of the disease throughout the CNS. There was a range of antibody responses against IDUA in this cohort which reduced detectable enzyme without substantially reducing efficacy; there was no evidence of toxicity. This first demonstration of the efficacy of intrathecal gene therapy in a large animal model of a LSD should pave the way for translation into the clinic.


Assuntos
Gatos , Sistema Nervoso Central/patologia , Modelos Animais de Doenças , Terapia Genética/métodos , Iduronidase/sangue , Iduronidase/líquido cefalorraquidiano , Mucopolissacaridose I/terapia , Animais , Dependovirus/enzimologia , Dependovirus/genética , Vetores Genéticos/administração & dosagem , Injeções Espinhais , Mucopolissacaridose I/enzimologia , Mucopolissacaridose I/genética , Mucopolissacaridose I/patologia , Especificidade de Órgãos
10.
Res Sq ; 2023 Jun 29.
Artigo em Inglês | MEDLINE | ID: mdl-37461697

RESUMO

Deficiency in human mature frataxin (hFXN-M) protein is responsible for the devastating neurodegenerative and cardiodegenerative disease of Friedreich's ataxia (FRDA). It results primarily by epigenetic silencing the FXN gene due to up to 1400 GAA triplet repeats in intron 1 of both alleles of the gene; a subset of approximately 3% of FRDA patients have a mutation on one allele. FRDA patients die most commonly in their 30s from heart disease. Therefore, increasing expression of heart hFXN-M using gene therapy offers a way to prevent early mortality in FRDA. We used rhesus macaque monkeys to test the pharmacology of an adeno-associated virus (AAV)hu68.CB7.hFXN therapy. The advantage of using non-human primates for hFXN-M gene therapy studies is that hFXN-M and monkey FXN-M (mFXN-M) are 98.5% identical, which limits potential immunologic side-effects. However, this presented a formidable bioanalytical challenge in quantification of proteins with almost identical sequences. This was overcome by development of a species-specific quantitative mass spectrometry-based method, which revealed for the first time, robust transgene-specific human protein expression in monkey heart tissue. The dose response was non-linear resulting in a ten-fold increase in monkey heart hFXN-M protein expression with only a three-fold increase in dose of the vector.

11.
Hum Gene Ther ; 34(19-20): 1022-1032, 2023 10.
Artigo em Inglês | MEDLINE | ID: mdl-36719773

RESUMO

Advances in adeno-associated virus (AAV)-based gene therapy are transforming our ability to treat rare genetic disorders and address other unmet medical needs. However, the natural prevalence of anti-AAV neutralizing antibodies (NAbs) in humans currently limits the population who can benefit from AAV-based gene therapies. Neonatal Fc receptor (FcRn) plays an essential role in the long half-life of IgG, a key NAb. Researchers have developed several FcRn-inhibiting monoclonal antibodies to treat autoimmune diseases, as inhibiting the interaction between FcRn and IgG Fc can reduce circulating IgG levels to 20-30% of the baseline. We evaluated the utility of one such monoclonal antibody, M281, to reduce pre-existing NAb levels and to permit gene delivery to the liver and heart via systemic AAV gene therapy in mice and nonhuman primates. M281 successfully reduced NAb titers along with total IgG levels; it also enhanced gene delivery to the liver and other organs after intravenous administration of AAV in NAb-positive animals. These results indicate that mitigating pre-existing humoral immunity via disruption of the FcRn-IgG interaction may make AAV-based gene therapies effective in NAb-positive patients.


Assuntos
Terapia Genética , Imunidade Humoral , Imunoglobulina G , Animais , Camundongos , Anticorpos Neutralizantes/genética , Anticorpos Neutralizantes/imunologia , Anticorpos Antivirais , Dependovirus/genética , Dependovirus/imunologia , Terapia Genética/métodos , Vetores Genéticos/genética , Imunidade Humoral/genética , Imunidade Humoral/imunologia , Imunoglobulina G/genética , Imunoglobulina G/imunologia
12.
Commun Biol ; 6(1): 1093, 2023 10 27.
Artigo em Inglês | MEDLINE | ID: mdl-37891254

RESUMO

Deficiency in human mature frataxin (hFXN-M) protein is responsible for the devastating neurodegenerative and cardiodegenerative disease of Friedreich's ataxia (FRDA). It results primarily through epigenetic silencing of the FXN gene by GAA triplet repeats on intron 1 of both alleles. GAA repeat lengths are most commonly between 600 and 1200 but can reach 1700. A subset of approximately 3% of FRDA patients have GAA repeats on one allele and a mutation on the other. FRDA patients die most commonly in their 30s from heart disease. Therefore, increasing expression of heart hFXN-M using gene therapy offers a way to prevent early mortality in FRDA. We used rhesus macaque monkeys to test the pharmacology of an adeno-associated virus (AAV)hu68.CB7.hFXN therapy. The advantage of using non-human primates for hFXN-M gene therapy studies is that hFXN-M and monkey FXN-M (mFXN-M) are 98.5% identical, which limits potential immunologic side-effects. However, this presented a formidable bioanalytical challenge in quantification of proteins with almost identical sequences. This could be overcome by the development of a species-specific quantitative mass spectrometry-based method, which has revealed for the first time, robust transgene-specific human protein expression in monkey heart tissue. The dose response is non-linear resulting in a ten-fold increase in monkey heart hFXN-M protein expression with only a three-fold increase in dose of the vector.


Assuntos
Ataxia de Friedreich , Proteínas de Ligação ao Ferro , Animais , Humanos , Macaca mulatta , Proteínas de Ligação ao Ferro/genética , Coração , Ataxia de Friedreich/genética , Ataxia de Friedreich/terapia , Ataxia de Friedreich/metabolismo , Terapia Genética , Frataxina
13.
J Vet Intern Med ; 37(6): 2200-2210, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37847024

RESUMO

BACKGROUND: A treatment of chronic kidney disease (CKD)-associated anemia in cats is needed. SB-001 is an adeno-associated virus-vectored (AAV)-based gene therapeutic agent that is administered intramuscularly, causing the expression of feline erythropoietin. HYPOTHESIS/OBJECTIVE: We hypothesized that SB-001 injection would lead to a sustained increase in PCV in cats with CKD-associated anemia. ANIMALS: Twenty-three cats with International Renal Interest Society (IRIS) Stage 2 to 4 CKD-associated anemia were enrolled at 4 veterinary clinics. METHODS: In a prospective clinical trial, cats were treated with 1 of 3 regimens of SB-001 (Lo 1.2 × 109 genome copies [GCs] on Day 0; Lo ± Hi [supplemental 2nd dose of 3.65 × 109 GC on Day 42]; Hi 3.65 × 109 GC IM on Day 0) and followed for 70 days. RESULTS: A response to SB-001 at any time between Day 28 and Day 70 was seen in 86% (95% confidence interval 65, 97%) of all cats. There was a significant (P < .003) increase in PCV from Day 0 to Day 28 (mean increase 6 ± 6 percentage points [pp]; n = 21), Day 42 (8 ± 9 pp; n = 21), Day 56 (10 ± 11 pp; n = 17), and Day 70 (13 ± 14 pp, n = 14). Twelve cats were hypertensive at baseline, 4 of which developed encephalopathy during the study. An additional 6 cats became hypertensive during the study. CONCLUSIONS AND CLINICAL IMPORTANCE: Results of this study suggest that SB-001 therapy represents a suitable single injection treatment that can address nonregenerative anemia in cats with CKD. It was generally well tolerated; however, hypertension and encephalopathy developed in some cats as previously described in association with erythropoiesis-stimulating agent therapy.


Assuntos
Anemia , Encefalopatias , Doenças do Gato , Eritropoetina , Hipertensão , Insuficiência Renal Crônica , Gatos , Animais , Dependovirus/genética , Estudos Prospectivos , Insuficiência Renal Crônica/complicações , Insuficiência Renal Crônica/terapia , Insuficiência Renal Crônica/veterinária , Anemia/terapia , Anemia/veterinária , Eritropoetina/genética , Eritropoetina/uso terapêutico , Hipertensão/veterinária , Encefalopatias/veterinária , Terapia Genética/veterinária , Doenças do Gato/terapia
14.
Hum Gene Ther ; 34(21-22): 1095-1106, 2023 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-37624734

RESUMO

Based on studies in experimental animals demonstrating that administration of adeno-associated virus (AAV) vectors to the cerebrospinal fluid (CSF) is an effective route to transfer genes to the nervous system, there are increasing number of clinical trials using the CSF route to treat nervous system disorders. With the knowledge that the CSF turns over four to five times daily, and evidence in experimental animals that at least some of CSF administered AAV vectors are distributed to systemic organs, we asked: with AAV administration to the CSF, what fraction of the total dose remains in the nervous system and what fraction goes off target and is delivered systemically? To quantify the biodistribution of AAV capsids immediately after administration, we covalently labeled AAV capsids with iodine 124 (I-124), a cyclotron generated positron emitter, enabling quantitative positron emission tomography scanning of capsid distribution for up to 96 h after AAV vector administration. We assessed the biodistribution to nonhuman primates of I-124-labeled capsids from different AAV clades, including 9 (clade F), rh.10 (E), PHP.eB (F), hu68 (F), and rh91(A). The analysis demonstrated that 60-90% of AAV vectors administered to the CSF through either the intracisternal or intrathecal (lumbar) routes distributed systemically to major organs. These observations have potentially significant clinical implications regarding accuracy of AAV vector dosing to the nervous system, evoking systemic immunity at levels similar to that with systemic administration, and potential toxicity of genes designed to treat nervous system disorders being expressed in non-nervous system organs. Based on these data, individuals in clinical trials using AAV vectors administered to the CSF should be monitored for systemic as well as nervous system adverse events and CNS dosing considerations should account for a significant AAV systemic distribution.


Assuntos
Dependovirus , Doenças do Sistema Nervoso , Animais , Dependovirus/genética , Radioisótopos do Iodo , Capsídeo , Distribuição Tecidual , Transdução Genética , Terapia Genética/métodos , Tomografia por Emissão de Pósitrons , Vetores Genéticos/genética , Técnicas de Transferência de Genes
15.
Mol Ther Methods Clin Dev ; 27: 272-280, 2022 Dec 08.
Artigo em Inglês | MEDLINE | ID: mdl-36320416

RESUMO

Gene therapy using neurotropic adeno-associated virus vectors represents an emerging solution for genetic disorders affecting the central nervous system. The first approved central nervous system-targeting adeno-associated virus gene therapy, Zolgensma®, for treating spinal muscular atrophy is administered intravenously at high doses that cause liver-associated adverse events in 20%-30% of patients. Intrathecal routes of vector administration, such as the intra-cisterna magna route, provide efficient gene transduction to central nervous system cells while reducing off-target liver transduction. However, significant levels of liver transduction often occur upon intra-cisterna magna vector delivery in preclinical studies. Using vectors expressing monoclonal antibody transgenes, we examined whether passive transfer of adeno-associated virus-neutralizing antibodies as intravenous immunoglobulin before intrathecal adeno-associated virus delivery improved the safety of viral gene therapy targeting the central nervous system in mice and nonhuman primates. We used intracerebroventricular and intra-cisterna magna routes for vector administration to mice and nonhuman primates, respectively, and evaluated transgene expression and vector genome distribution. Our data indicate that pretreatment with intravenous immunoglobulin significantly reduced gene transduction to the liver and other peripheral organs but not to the central nervous system in both species. With further refinement, this method may improve the safety of adeno-associated virus-based, central nervous system-targeting gene therapies in clinical settings.

17.
Hum Gene Ther ; 31(21-22): 1169-1177, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-33045869

RESUMO

GM1 gangliosidosis is a rare neurodegenerative lysosomal storage disease caused by loss-of-function mutations in the gene encoding beta-galactosidase (ß-gal). There are no approved treatments for GM1 gangliosidosis. Previous studies in animal models have demonstrated that adeno-associated viral (AAV) vector-mediated gene transfer to the brain can restore ß-gal expression and prevent the onset of neurological signs. We developed an optimized AAV vector expressing human ß-gal and evaluated the efficacy of a single intracerebroventricular injection of this vector into the cerebrospinal fluid (CSF) of a murine disease model. The AAV vector administration into the CSF increased ß-gal activity in the brain, reduced neuronal lysosomal storage lesions, prevented the onset of neurological signs and gait abnormalities, and increased survival. These findings demonstrate the potential therapeutic activity of this vector and support its subsequent development for the treatment of GM1 gangliosidosis.


Assuntos
Encéfalo/metabolismo , Líquido Cefalorraquidiano/metabolismo , Dependovirus/genética , Gangliosidose GM1/terapia , Terapia Genética/métodos , Vetores Genéticos/administração & dosagem , beta-Galactosidase/fisiologia , Animais , Encéfalo/patologia , Líquido Cefalorraquidiano/citologia , Modelos Animais de Doenças , Gangliosidose GM1/genética , Gangliosidose GM1/metabolismo , Gangliosidose GM1/patologia , Vetores Genéticos/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , beta-Galactosidase/administração & dosagem , beta-Galactosidase/genética
18.
Ann Clin Transl Neurol ; 7(10): 1843-1853, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32937039

RESUMO

OBJECTIVE: Dominant loss-of-function mutations in the gene encoding the lysosomal protein, progranulin, cause 5-10% of frontotemporal dementia cases. As progranulin undergoes secretion and endocytosis, a small number of progranulin-expressing cells can potentially supply the protein to the entire central nervous system. Thus, gene therapy is a promising treatment approach. METHODS: We evaluated adeno-associated viral vector administration into the cerebrospinal fluid as a minimally invasive approach to deliver the granulin gene to the central nervous system in a murine disease model and nonhuman primates. RESULTS: In progranulin-deficient mice, vector delivery into the lateral cerebral ventricles increased progranulin levels in the cerebrospinal fluid and normalized histological and biochemical markers of progranulin deficiency. A single vector injection into the cisterna magna of nonhuman primates achieved CSF progranulin concentrations up to 40-fold higher than those of normal human subjects and exceeded CSF progranulin levels of successfully treated mice. Animals treated with an adeno-associated virus serotype 1 vector exhibited progranulin expression fivefold higher than those treated with an AAV5 vector or the AAV9 variant, AAVhu68, apparently due to remarkably efficient transduction of ependymal cells. Progranulin expression mediated by adeno-associated viral vectors was well tolerated in nonhuman primates with no evidence of dose-limiting toxicity, even at vector doses that induced supraphysiologic progranulin expression. INTERPRETATION: These findings support the development of AAV1-based gene therapy for frontotemporal dementia caused by progranulin deficiency.


Assuntos
Dependovirus/genética , Demência Frontotemporal/virologia , Mutação/genética , Progranulinas/genética , Animais , Dependovirus/patogenicidade , Demência Frontotemporal/genética , Terapia Genética/métodos , Camundongos Knockout , Progranulinas/metabolismo , Sorogrupo
19.
Mol Ther Methods Clin Dev ; 17: 969-974, 2020 Jun 12.
Artigo em Inglês | MEDLINE | ID: mdl-32420410

RESUMO

Preclinical studies have demonstrated that a single injection of an adeno-associated virus (AAV) vector into the cerebrospinal fluid (CSF) can achieve widespread gene transfer throughout the central nervous system. Successfully translating this approach to humans requires identifying factors that influence AAV distribution in the CSF so that optimal parameters can be replicated in the clinic. In the context of developing a motor neuron-targeted gene therapy for spinal muscular atrophy, we conducted studies in nonhuman primates to evaluate the impact of injection volume on spinal cord transduction after AAV delivery via lumbar puncture. Lumbar injection of an AAVhu68 vector targeted motor neurons throughout the spinal cord, but only in juvenile nonhuman primates administered large injection volumes, equivalent to about half of the total CSF volume. Upon repeating this study with clinically relevant injection volumes and larger animals, we found that lumbar puncture failed to achieve significant transduction of the spinal cord. In contrast, vector administered into the cisterna magna distributed reproducibly throughout the spinal cord in both juvenile and adult animals. These findings highlight the challenges of translating AAV delivery via lumbar puncture to humans and suggest that delivery into the cisterna magna may represent a more feasible alternative.

20.
Hum Gene Ther ; 31(15-16): 808-818, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32845779

RESUMO

The administration of adeno-associated virus (AAV) vectors to nonhuman primates (NHP) via the blood or cerebrospinal fluid (CSF) can lead to dorsal root ganglion (DRG) pathology. The pathology is minimal to moderate in most cases; clinically silent in affected animals; and characterized by mononuclear cell infiltrates, neuronal degeneration, and secondary axonopathy of central and peripheral axons on histopathological analysis. We aggregated data from 33 nonclinical studies in 256 NHP and performed a meta-analysis of the severity of DRG pathology to compare different routes of administration, dose, time course, study conduct, age of the animals, sex, capsid, promoter, capsid purification method, and transgene. DRG pathology was observed in 83% of NHP that were administered AAV through the CSF, and 32% of NHP that received an intravenous (IV) injection. We show that dose and age at injection significantly affected the severity whereas sex had no impact. DRG pathology was minimal at acute time points (i.e., <14 days), similar from one to 5 months post-injection, and was less severe after 6 months. Vector purification method had no impact, and all capsids and promoters that we tested resulted in some DRG pathology. The data presented here from five different capsids, five different promoters, and 20 different transgenes suggest that DRG pathology is almost universal after AAV gene therapy in nonclinical studies using NHP. None of the animals receiving a therapeutic transgene displayed any clinical signs. Incorporation of sensitive techniques such as nerve-conduction velocity testing can show alterations in a minority of animals that correlate with the severity of peripheral nerve axonopathy. Monitoring sensory neuropathies in human central nervous system and high-dose IV clinical studies seems prudent to determine the functional consequences of DRG pathology.


Assuntos
Dependovirus/genética , Gânglios Espinais/patologia , Técnicas de Transferência de Genes , Vetores Genéticos/administração & dosagem , Vetores Genéticos/genética , Condução Nervosa , Animais , Feminino , Gânglios Espinais/metabolismo , Macaca fascicularis , Macaca mulatta , Masculino , Transdução Genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA