Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 53
Filtrar
1.
Mol Ther ; 27(7): 1313-1326, 2019 07 03.
Artigo em Inglês | MEDLINE | ID: mdl-31043342

RESUMO

Mutations within over 250 known genes are associated with inherited retinal degeneration. Clinical success following gene-replacement therapy for congenital blindness due to RPE65 mutations establishes a platform for the development of downstream treatments targeting other forms of inherited ocular disease. Unfortunately, several challenges relevant to complex disease pathology and limitations of current gene-transfer technologies impede the development of related strategies for each specific form of inherited retinal degeneration. Here, we describe a gene-augmentation strategy that delays retinal degeneration by stimulating features of anabolic metabolism necessary for survival and structural maintenance of photoreceptors. We targeted two critical points of regulation in the canonical insulin/AKT/mammalian target of rapamycin (mTOR) pathway with AAV-mediated gene augmentation in a mouse model of retinitis pigmentosa. AAV vectors expressing the serine/threonine kinase, AKT3, promote dramatic preservation of photoreceptor numbers, structure, and partial visual function. This protective effect was associated with successful reprogramming of photoreceptor metabolism toward pathways associated with cell growth and survival. Collectively, these findings underscore the importance of AKT activity and downstream pathways associated with anabolic metabolism in photoreceptor survival and maintenance.


Assuntos
Terapia Genética/métodos , Neuroproteção/genética , Células Fotorreceptoras de Vertebrados/metabolismo , Proteínas Proto-Oncogênicas c-akt/genética , Retinose Pigmentar/terapia , Transdução de Sinais/genética , Transdução Genética , Animais , Sobrevivência Celular/genética , Células Cultivadas , Nucleotídeo Cíclico Fosfodiesterase do Tipo 6/genética , Modelos Animais de Doenças , Vetores Genéticos , Gliose/genética , Gliose/terapia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Mutação Puntual , Degeneração Retiniana/terapia , Retinose Pigmentar/genética , Serina-Treonina Quinases TOR/metabolismo , Acuidade Visual/genética
2.
Mol Ther ; 26(6): 1581-1593, 2018 06 06.
Artigo em Inglês | MEDLINE | ID: mdl-29673930

RESUMO

Most genetically distinct inherited retinal degenerations are primary photoreceptor degenerations. We selected a severe early onset form of Leber congenital amaurosis (LCA), caused by mutations in the gene LCA5, in order to test the efficacy of gene augmentation therapy for a ciliopathy. The LCA5-encoded protein, Lebercilin, is essential for the trafficking of proteins and vesicles to the photoreceptor outer segment. Using the AAV serotype AAV7m8 to deliver a human LCA5 cDNA into an Lca5 null mouse model of LCA5, we show partial rescue of retinal structure and visual function. Specifically, we observed restoration of rod-and-cone-driven electroretinograms in about 25% of injected eyes, restoration of pupillary light responses in the majority of treated eyes, an ∼20-fold decrease in target luminance necessary for visually guided behavior, and improved retinal architecture following gene transfer. Using LCA5 patient-derived iPSC-RPEs, we show that delivery of the LCA5 cDNA restores lebercilin protein and rescues cilia quantity. The results presented in this study support a path forward aiming to develop safety and efficacy trials for gene augmentation therapy in human subjects with LCA5 mutations. They also provide the framework for measuring the effects of intervention in ciliopathies and other severe, early-onset blinding conditions.


Assuntos
Cegueira/metabolismo , Cegueira/terapia , Dependovirus/genética , Terapia Genética/métodos , Animais , Eletrorretinografia , Proteínas do Olho/genética , Proteínas do Olho/metabolismo , Feminino , Humanos , Amaurose Congênita de Leber/metabolismo , Amaurose Congênita de Leber/terapia , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Associadas aos Microtúbulos/genética , Proteínas Associadas aos Microtúbulos/metabolismo
3.
Blood ; 127(5): 565-71, 2016 Feb 04.
Artigo em Inglês | MEDLINE | ID: mdl-26702064

RESUMO

Factor VII (FVII) deficiency is a rare autosomal recessive bleeding disorder treated by infusion of fresh-frozen plasma, plasma-derived FVII concentrates and low-dose recombinant activated FVII. Clinical data suggest that a mild elevation of plasma FVII levels (>10% normal) results in improved hemostasis. Research dogs with a G96E missense FVII mutation (FVII-G96E) have <1% FVII activity. By western blot, we show that they have undetectable plasmatic antigen, thus representing the most prevalent type of human FVII deficiency (low antigen/activity). In these dogs, we determine the feasibility of a gene therapy approach using liver-directed, adeno-associated viral (AAV) serotype 8 vector delivery of a canine FVII (cFVII) zymogen transgene. FVII-G96E dogs received escalating AAV doses (2E11 to 4.95E13 vector genomes [vg] per kg). Clinically therapeutic expression (15% normal) was attained with as low as 6E11 vg/kg of AAV and has been stable for >1 year (ongoing) without antibody formation to the cFVII transgene. Sustained and supraphysiological expression of 770% normal was observed using 4.95E13 vg/kg of AAV (2.6 years, ongoing). No evidence of pathological activation of coagulation or detrimental animal physiology was observed as platelet counts, d-dimer, fibrinogen levels, and serum chemistries remained normal in all dogs (cumulative 6.4 years). We observed a transient and noninhibitory immunoglobulin G class 2 response against cFVII only in the dog receiving the highest AAV dose. In conclusion, in the only large-animal model representing the majority of FVII mutation types, our data are first to demonstrate the feasibility, safety, and long-term duration of AAV-mediated correction of FVII deficiency.


Assuntos
Deficiência do Fator VII/genética , Deficiência do Fator VII/terapia , Fator VII/genética , Terapia Genética , Vetores Genéticos/genética , Vetores Genéticos/uso terapêutico , Precursores de Proteínas/genética , Adenoviridae/genética , Animais , Cães , Deficiência do Fator VII/sangue , Expressão Gênica , Vetores Genéticos/administração & dosagem , Células HEK293 , Humanos , Mutação Puntual , Transgenes
4.
Blood ; 125(10): 1553-61, 2015 Mar 05.
Artigo em Inglês | MEDLINE | ID: mdl-25568350

RESUMO

Emerging successful clinical data on gene therapy using adeno-associated viral (AAV) vector for hemophilia B (HB) showed that the risk of cellular immune response to vector capsid is clearly dose dependent. To decrease the vector dose, we explored AAV-8 (1-3 × 10(12) vg/kg) encoding a hyperfunctional factor IX (FIX-Padua, arginine 338 to leucine) in FIX inhibitor-prone HB dogs. Two naïve HB dogs showed sustained expression of FIX-Padua with an 8- to 12-fold increased specific activity reaching 25% to 40% activity without antibody formation to FIX. A third dog with preexisting FIX inhibitors exhibited a transient anamnestic response (5 Bethesda units) at 2 weeks after vector delivery following by spontaneous eradication of the antibody to FIX by day 70. In this dog, sustained FIX expression reached ∼200% and 30% of activity and antigen levels, respectively. Immune tolerance was confirmed in all dogs after challenges with plasma-derived FIX concentrate. Shortening of the clotting times and lack of bleeding episodes support the phenotypic correction of the severe phenotype, with no clinical or laboratory evidence of risk of thrombosis. Provocative studies in mice showed that FIX-Padua exhibits similar immunogenicity and thrombogenicity compared with FIX wild type. Collectively, these data support the potential translation of gene-based strategies using FIX-Padua for HB.


Assuntos
Fator IX/antagonistas & inibidores , Terapia Genética/métodos , Hemofilia B/genética , Hemofilia B/terapia , Substituição de Aminoácidos , Animais , Capsídeo/imunologia , Citocinas/sangue , Dependovirus/genética , Dependovirus/imunologia , Modelos Animais de Doenças , Cães , Fator IX/genética , Fator IX/imunologia , Fator IX/uso terapêutico , Expressão Gênica , Vetores Genéticos/efeitos adversos , Vetores Genéticos/imunologia , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Proteínas Mutantes/genética , Proteínas Mutantes/imunologia , Proteínas Mutantes/uso terapêutico , Proteínas Recombinantes/genética , Proteínas Recombinantes/imunologia , Proteínas Recombinantes/uso terapêutico , Trombose/prevenção & controle , Pesquisa Translacional Biomédica
5.
Blood ; 126(15): 1777-84, 2015 Oct 08.
Artigo em Inglês | MEDLINE | ID: mdl-26297739

RESUMO

Site-specific genome editing provides a promising approach for achieving long-term, stable therapeutic gene expression. Genome editing has been successfully applied in a variety of preclinical models, generally focused on targeting the diseased locus itself; however, limited targeting efficiency or insufficient expression from the endogenous promoter may impede the translation of these approaches, particularly if the desired editing event does not confer a selective growth advantage. Here we report a general strategy for liver-directed protein replacement therapies that addresses these issues: zinc finger nuclease (ZFN) -mediated site-specific integration of therapeutic transgenes within the albumin gene. By using adeno-associated viral (AAV) vector delivery in vivo, we achieved long-term expression of human factors VIII and IX (hFVIII and hFIX) in mouse models of hemophilia A and B at therapeutic levels. By using the same targeting reagents in wild-type mice, lysosomal enzymes were expressed that are deficient in Fabry and Gaucher diseases and in Hurler and Hunter syndromes. The establishment of a universal nuclease-based platform for secreted protein production would represent a critical advance in the development of safe, permanent, and functional cures for diverse genetic and nongenetic diseases.


Assuntos
Albuminas/genética , Terapia de Reposição de Enzimas , Terapia Genética , Genoma , Fígado/metabolismo , Transgenes/fisiologia , Albuminas/metabolismo , Animais , Dependovirus/genética , Endonucleases , Doença de Fabry/genética , Doença de Fabry/terapia , Fator IX/genética , Fator VIII/genética , Doença de Gaucher/genética , Doença de Gaucher/terapia , Vetores Genéticos/administração & dosagem , Hemofilia A/genética , Hemofilia A/terapia , Hemofilia B/genética , Hemofilia B/terapia , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Lisossomos/enzimologia , Camundongos , Camundongos Endogâmicos C57BL , Mucopolissacaridose I/genética , Mucopolissacaridose I/terapia , Mucopolissacaridose II/genética , Mucopolissacaridose II/terapia , Regiões Promotoras Genéticas/genética , Edição de RNA , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Dedos de Zinco
6.
Nature ; 475(7355): 217-21, 2011 Jun 26.
Artigo em Inglês | MEDLINE | ID: mdl-21706032

RESUMO

Editing of the human genome to correct disease-causing mutations is a promising approach for the treatment of genetic disorders. Genome editing improves on simple gene-replacement strategies by effecting in situ correction of a mutant gene, thus restoring normal gene function under the control of endogenous regulatory elements and reducing risks associated with random insertion into the genome. Gene-specific targeting has historically been limited to mouse embryonic stem cells. The development of zinc finger nucleases (ZFNs) has permitted efficient genome editing in transformed and primary cells that were previously thought to be intractable to such genetic manipulation. In vitro, ZFNs have been shown to promote efficient genome editing via homology-directed repair by inducing a site-specific double-strand break (DSB) at a target locus, but it is unclear whether ZFNs can induce DSBs and stimulate genome editing at a clinically meaningful level in vivo. Here we show that ZFNs are able to induce DSBs efficiently when delivered directly to mouse liver and that, when co-delivered with an appropriately designed gene-targeting vector, they can stimulate gene replacement through both homology-directed and homology-independent targeted gene insertion at the ZFN-specified locus. The level of gene targeting achieved was sufficient to correct the prolonged clotting times in a mouse model of haemophilia B, and remained persistent after induced liver regeneration. Thus, ZFN-driven gene correction can be achieved in vivo, raising the possibility of genome editing as a viable strategy for the treatment of genetic disease.


Assuntos
Reparo do DNA/genética , Modelos Animais de Doenças , Marcação de Genes/métodos , Terapia Genética/métodos , Genoma/genética , Hemofilia B/genética , Hemostasia , Animais , Sequência de Bases , Linhagem Celular Tumoral , Quebras de DNA de Cadeia Dupla , Endonucleases/química , Endonucleases/genética , Endonucleases/metabolismo , Éxons/genética , Fator IX/análise , Fator IX/genética , Vetores Genéticos/genética , Células HEK293 , Hemofilia B/fisiopatologia , Humanos , Íntrons/genética , Fígado/metabolismo , Regeneração Hepática , Camundongos , Camundongos Endogâmicos C57BL , Mutação/genética , Fenótipo , Homologia de Sequência , Dedos de Zinco
7.
Blood ; 121(19): 3825-9, S1-3, 2013 May 09.
Artigo em Inglês | MEDLINE | ID: mdl-23515928

RESUMO

Severe deficiency of plasma ADAMTS13 activity causes thrombotic thrombocytopenic purpura (TTP), a life-threatening syndrome for which plasma is the only effective therapy currently available. As much as 5% of TTP cases are hereditary, resulting from mutations of the ADAMTS13 gene. Here, we report the efficacy and safety of recombinant adeno-associated virus serotype 8 (AAV8)-mediated expression of a murine ADAMTS13 variant (MDTCS), truncated after the spacer domain, in a murine model of TTP. Administration of AAV8-hAAT-mdtcs at doses greater than 2.6 × 10(11) vg/kg body weight resulted in sustained expression of plasma ADAMTS13 activity at therapeutic levels. Expression of the truncated ADAMTS13 variant eliminated circulating ultralarge von Willebrand factor multimers, prevented severe thrombocytopenia, and reduced mortality in Adamts13(-/-) disease-prone mice triggered by shigatoxin-2. These data support AAV vector-mediated expression of a comparable truncated ADAMTS13 variant as a novel therapeutic approach for hereditary TTP in humans.


Assuntos
Citoproteção/genética , Terapia Genética/métodos , Metaloendopeptidases/genética , Púrpura Trombocitopênica Trombótica/prevenção & controle , Toxina Shiga II/toxicidade , Proteína ADAMTS13 , Animais , Códon sem Sentido/fisiologia , Dependovirus , Vetores Genéticos/genética , Camundongos , Camundongos Knockout , Púrpura Trombocitopênica Trombótica/induzido quimicamente , Transformação Genética
8.
Blood ; 121(21): 4396-403, 2013 May 23.
Artigo em Inglês | MEDLINE | ID: mdl-23372167

RESUMO

Recombinant canine B-domain deleted (BDD) factor VIII (FVIII) is predominantly expressed as a single-chain protein and exhibits greater stability after activation compared with human FVIII-BDD. We generated a novel BDD-FVIII variant (FVIII-RH) with an amino acid change at the furin cleavage site within the B domain (position R1645H) that mimics the canine sequence (HHQR vs human RHQR). Compared with human FVIII-BDD, expression of FVIII-RH protein revealed a 2.5-fold increase in the single-chain form. Notably, FVIII-RH exhibited a twofold increase in biological activity compared with FVIII-BDD, likely due to its slower dissociation of the A2-domain upon thrombin activation. Injection of FVIII-RH protein in hemophilia A (HA) mice resulted in more efficacious hemostasis following vascular injury in both the macro- and microcirculation. These findings were successfully translated to adeno-associated viral (AAV)-based liver gene transfer in HA mice. Expression of circulating FVIII-RH was approximately twofold higher compared with AAV-FVIII-BDD-injected mice. Moreover, FVIII-RH exhibits superior procoagulant effects compared with FVIII-BDD following a series of hemostatic challenges. Notably, the immunogenicity of FVIII-RH did not differ from FVIII-BDD. Thus, FVIII-RH is an attractive bioengineered molecule for improving efficacy without increased immunogenicity and may be suitable for both protein- and gene-based strategies for HA.


Assuntos
Fator VIII/genética , Terapia Genética/métodos , Hemofilia A/genética , Hemofilia A/terapia , Animais , Cães , Fator VIII/química , Fator VIII/imunologia , Variação Genética , Hemostasia/genética , Hemostasia/fisiologia , Humanos , Tolerância Imunológica , Fígado/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fenótipo , Estrutura Terciária de Proteína/genética , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/imunologia , Especificidade da Espécie
9.
Blood ; 122(19): 3283-7, 2013 Nov 07.
Artigo em Inglês | MEDLINE | ID: mdl-24085764

RESUMO

Monogenic diseases, including hemophilia, represent ideal targets for genome-editing approaches aimed at correcting a defective gene. Here we report that systemic adeno-associated virus (AAV) vector delivery of zinc finger nucleases (ZFNs) and corrective donor template to the predominantly quiescent livers of adult mice enables production of high levels of human factor IX in a murine model of hemophilia B. Further, we show that off-target cleavage can be substantially reduced while maintaining robust editing by using obligate heterodimeric ZFNs engineered to minimize unwanted cleavage attributable to homodimerization of the ZFNs. These results broaden the therapeutic potential of AAV/ZFN-mediated genome editing in the liver and could expand this strategy to other nonreplicating cell types.


Assuntos
Endonucleases/genética , Fator IX/biossíntese , Terapia Genética/métodos , Vetores Genéticos , Genoma , Hemofilia B/terapia , Dedos de Zinco/genética , Animais , Dependovirus/genética , Modelos Animais de Doenças , Endonucleases/metabolismo , Fator IX/genética , Fator IX/metabolismo , Hemofilia B/genética , Hemofilia B/patologia , Fígado/metabolismo , Masculino , Camundongos , Camundongos Transgênicos , Multimerização Proteica
10.
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
11.
Blood ; 121(17): 3335-44, 2013 Apr 25.
Artigo em Inglês | MEDLINE | ID: mdl-23426947

RESUMO

Recombinant adeno-associated virus (rAAV) vectors encoding human factor VIII (hFVIII) were systematically evaluated for hemophilia A (HA) gene therapy. A 5.7-kb rAAV-expression cassette (rAAV-HLP-codop-hFVIII-N6) containing a codon-optimized hFVIII cDNA in which a 226 amino acid (aa) B-domain spacer replaced the entire B domain and a hybrid liver-specific promoter (HLP) mediated 10-fold higher hFVIII levels in mice compared with non-codon-optimized variants. A further twofold improvement in potency was achieved by replacing the 226-aa N6 spacer with a novel 17-aa peptide (V3) in which 6 glycosylation triplets from the B domain were juxtaposed. The resulting 5.2-kb rAAV-HLP-codop-hFVIII-V3 cassette was more efficiently packaged within AAV virions and mediated supraphysiologic hFVIII expression (732 ± 162% of normal) in HA knock-out mice following administration of 2 × 10(12) vector genomes/kg, a vector dose shown to be safe in subjects with hemophilia B. Stable hFVIII expression at 15 ± 4% of normal was observed at this dose in a nonhuman primate. hFVIII expression above 100% was observed in 3 macaques that received a higher dose of either this vector or the N6 variant. These animals developed neutralizing anti-FVIII antibodies that were abrogated with transient immunosuppression. Therefore, rAAV-HLP-codop-hFVIII-V3 substantially improves the prospects of effective HA gene therapy.


Assuntos
Dependovirus/genética , Fator VIII/farmacologia , Terapia Genética , Variação Genética/genética , Vetores Genéticos/administração & dosagem , Hemofilia A/terapia , Animais , Western Blotting , Fator VIII/genética , Fator VIII/imunologia , Glicosilação , Hemofilia A/genética , Humanos , Tolerância Imunológica , Fígado/metabolismo , Macaca mulatta , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fragmentos de Peptídeos/genética , Fragmentos de Peptídeos/metabolismo , Regiões Promotoras Genéticas/genética
12.
Mol Ther ; 22(1): 42-51, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24077034

RESUMO

Self-complementary adeno-associated viral (AAV) vectors expressing human factor IX (hF.IX) have achieved transient or sustained correction of hemophilia B in human volunteers. High doses of AAV2 or AAV8 vectors delivered to the liver caused in several patients an increase in transaminases accompanied by a rise in AAV capsid-specific T cells and a decrease in circulating hF.IX levels suggesting immune-mediated destruction of vector-transduced cells. Kinetics of these adverse events differed in patients receiving AAV2 or AAV8 vectors causing rise in transaminases at 3 versus 8 weeks after vector injection, respectively. To test if CD8+ T cells to AAV8 vectors, which are similar to AAV2 vectors are fully-gutted vectors and thereby fail to encode structural viral proteins, could cause damage at this late time point, we tested in a series of mouse studies how long major histocompatibility (MHC) class I epitopes within AAV8 capsid can be presented to CD8+ T cells. Our results clearly show that depending on the vectors' genome, CD8+ T cells can detect such epitopes on AAV8's capsid for up to 6 months indicating that the capsid of AAV8 degrades slowly in mice.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Capsídeo/imunologia , Dependovirus/imunologia , Vetores Genéticos/genética , Vetores Genéticos/imunologia , Genoma , Animais , Proteínas do Capsídeo/imunologia , Dependovirus/genética , Epitopos de Linfócito T , Vetores Genéticos/normas , Humanos , Memória Imunológica , Ativação Linfocitária/imunologia , Masculino , Camundongos , Controle de Qualidade , Especificidade do Receptor de Antígeno de Linfócitos T , Transdução Genética
13.
Mol Ther ; 21(1): 158-66, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22929660

RESUMO

There is considerable interest in the use of adeno-associated virus serotype 9 (AAV9) for neurological gene therapy partly because of its ability to cross the blood-brain barrier to transduce astrocytes and neurons. This raises the possibility that AAV9 might also transduce antigen-presenting cells (APC) in the brain and provoke an adaptive immune response. We tested this hypothesis by infusing AAV9 vectors encoding foreign antigens, namely human aromatic L-amino acid decarboxylase (hAADC) and green fluorescent protein (GFP), into rat brain parenchyma. Over ensuing weeks, both vectors elicited a prominent inflammation in transduced brain regions associated with upregulation of MHC II in glia and associated lymphocytic infiltration. Transduction of either thalamus or striatum with AAV9-hAADC evinced a significant loss of neurons and induction of anti-hAADC antibodies. We conclude that AAV9 transduces APC in the brain and, depending on the immunogenicity of the transgene, can provoke a full immune response that mediates significant brain pathology. We emphasize, however, that these observations do not preclude the use of AAV serotypes that can transduce APC. However, it does potentially complicate preclinical toxicology studies in which non-self proteins are expressed at a level sufficient to trigger cell-mediated and humoral immune responses.


Assuntos
Dependovirus/genética , Vetores Genéticos , Imunidade Celular , Proteínas/genética , Animais , Proteínas/imunologia , Ratos , Transdução Genética
14.
Mol Ther ; 21(9): 1727-37, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23857231

RESUMO

Immune responses directed against viral capsid proteins constitute a main safety concern in the use of adeno-associated virus (AAV) as gene transfer vectors in humans. Pharmacological immunosuppression has been proposed as a solution to the problem; however, the approach suffers from several potential limitations. Using MHC class II epitopes initially identified within human IgG, named Tregitopes, we showed that it is possible to modulate CD8+ T cell responses to several viral antigens in vitro. We showed that incubation of peripheral blood mononuclear cells with these epitopes triggers proliferation of CD4+CD25+FoxP3+ T cells that suppress killing of target cells loaded with MHC class I antigens in an antigen-specific fashion, through a mechanism that seems to require cell-to-cell contact. Expression of a construct encoding for the AAV capsid structural protein fused to Tregitopes resulted in reduction of CD8+ T cell reactivity against the AAV capsid following immunization with an adenoviral vector expressing capsid. This was accompanied by an increase in frequency of CD4+CD25+FoxP3+ T cells in spleens and lower levels of inflammatory infiltrates in injected tissues. This proof-of-concept study demonstrates modulation of CD8+ T cell reactivity to an antigen using regulatory T cell epitopes is possible.


Assuntos
Antígenos Virais/imunologia , Linfócitos T CD8-Positivos/imunologia , Proteínas do Capsídeo/imunologia , Dependovirus/imunologia , Epitopos de Linfócito T/imunologia , Vetores Genéticos , Imunoglobulina G/imunologia , Animais , Antígenos Virais/genética , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/metabolismo , Capsídeo/imunologia , Proteínas do Capsídeo/genética , Proteínas do Capsídeo/metabolismo , Células Cultivadas , Dependovirus/genética , Epitopos de Linfócito T/genética , Epitopos de Linfócito T/metabolismo , Terapia Genética , Antígenos de Histocompatibilidade Classe I/imunologia , Antígenos de Histocompatibilidade Classe II/imunologia , Humanos , Imunoglobulina G/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Baço/imunologia , Linfócitos T Reguladores/imunologia
15.
Blood ; 117(15): 3974-82, 2011 Apr 14.
Artigo em Inglês | MEDLINE | ID: mdl-21325603

RESUMO

Catalytic domain variants of activated factor VII (FVIIa) with enhanced hemostatic properties are highly attractive for the treatment of bleeding disorders via gene-based therapy. To explore this in a hemophilic mouse model, we characterized 2 variants of murine activated FVII (mFVIIa-VEAY and mFVIIa-DVQ) with modified catalytic domains, based on recombinant human FVIIa (rhFVIIa) variants. Using purified recombinant proteins, we showed that murine FVIIa (mFVIIa) and variants had comparable binding to human and murine tissue factor (TF) and exhibited similar extrinsic coagulant activity. In vitro in the absence of TF, the variants showed a 6- to 17-fold enhanced proteolytic and coagulant activity relative to mFVIIa, but increased inactivation by antithrombin. Gene delivery of mFVIIa-VEAY resulted in long-term, effective hemostasis at 5-fold lower expression levels relative to mFVIIa in hemophilia A mice or in hemophilia B mice with inhibitors to factor IX. However, expression of mFVIIa-VEAY at 14-fold higher than therapeutic levels resulted in a progressive mortality to 70% within 6 weeks after gene delivery. These results are the first demonstration of the hemostatic efficacy of continuous expression, in the presence or absence of inhibitors, of a high-activity gene-based FVIIa variant in an animal model of hemophilia.


Assuntos
Domínio Catalítico/genética , Fator VIIa/genética , Terapia Genética/métodos , Hemofilia A/terapia , Hemostasia/fisiologia , Animais , Linhagem Celular , Dependovirus/genética , Modelos Animais de Doenças , Fator VIIa/química , Regulação da Expressão Gênica , Técnicas de Transferência de Genes , Terapia Genética/mortalidade , Hemofilia A/sangue , Hemofilia A/genética , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Fenótipo , Plasmídeos/genética , Estrutura Terciária de Proteína
16.
Mol Ther ; 20(7): 1410-6, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22565846

RESUMO

Liver gene transfer for hemophilia B has shown very promising results in recent clinical studies. A potential complication of gene-based treatments for hemophilia and other inherited disorders, however, is the development of neutralizing antibodies (NAb) against the therapeutic transgene. The risk of developing NAb to the coagulation factor IX (F.IX) transgene product following adeno-associated virus (AAV)-mediated hepatic gene transfer for hemophilia is small but not absent, as formation of inhibitory antibodies to F.IX is observed in experimental animals following liver gene transfer. Thus, strategies to modulate antitransgene NAb responses are needed. Here, we used the anti-B cell monoclonal antibody rituximab (rtx) in combination with cyclosporine A (CsA) to eradicate anti-human F.IX NAb in rhesus macaques previously injected intravenously with AAV8 vectors expressing human F.IX. A short course of immunosuppression (IS) resulted in eradication of anti-F.IX NAb with restoration of plasma F.IX transgene product detection. In one animal, following IS anti-AAV6 antibodies also dropped below detection, allowing for successful AAV vector readministration and resulting in high levels (60% or normal) of F.IX transgene product in plasma. Though the number of animals is small, this study supports for the safety and efficacy of B cell-targeting therapies to eradicate NAb developed following AAV-mediated gene transfer.


Assuntos
Fator IX/imunologia , Técnicas de Transferência de Genes , Terapia Genética , Hemofilia B/terapia , Imunidade Humoral/efeitos dos fármacos , Animais , Anticorpos Monoclonais Murinos/farmacologia , Anticorpos Neutralizantes/imunologia , Linfócitos B/efeitos dos fármacos , Linfócitos B/imunologia , Linfócitos T CD4-Positivos/imunologia , Ciclosporinas/farmacologia , Dependovirus/genética , Fator IX/genética , Hemofilia B/genética , Fatores Imunológicos/farmacologia , Imunossupressores/farmacologia , Macaca mulatta , Rituximab , Transgenes
17.
Mol Ther ; 20(9): 1737-49, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22735378

RESUMO

The hepatitis C virus (HCV) chronically infects 2% of the world population and effective treatment is limited by long duration and significant side-effects. Here, we describe a novel drug, intended as a "single-shot " therapy, which expresses three short hairpin RNAs (shRNAs) that simultaneously target multiple conserved regions of the HCV genome as confirmed in vitro by knockdown of an HCV replicon system. Using a recombinant adeno-associated virus (AAV) serotype 8 vector for delivery, comprehensive transduction of hepatocytes was achieved in vivo in a nonhuman primate (NHP) model following a single intravenous injection. However, dose ranging studies performed in 13 NHP resulted in high-expression levels of shRNA from wild-type (wt) Pol III promoters and dose-dependent hepatocellular toxicity, the first demonstration of shRNA-related toxicity in primates, establishing that the hepatotoxicity arises from highly conserved features of the RNA interference (RNAi) pathway. In the second generation drug, each promoter was re-engineered to reduce shRNA transcription to levels that circumvent toxicity but still inhibit replicon activity. In vivo testing of this modified construct in 18 NHPs showed conservation of hepatocyte transduction but complete elimination of hepatotoxicity, even with sustained shRNA expression for 50 days. These data support progression to a clinical study for treatment of HCV infection.


Assuntos
Genoma Viral , Hepacivirus/genética , Hepatite C Crônica/terapia , Hepatócitos/virologia , Fígado/virologia , RNA Interferente Pequeno/genética , RNA Viral/antagonistas & inibidores , Animais , DNA Polimerase III/genética , Dependovirus/genética , Engenharia Genética , Terapia Genética , Vetores Genéticos , Hepatite C Crônica/virologia , Hepatócitos/patologia , Injeções Intravenosas , Fígado/patologia , Macaca fascicularis , Camundongos , Regiões Promotoras Genéticas , RNA Viral/genética , Replicon , Transdução Genética , Replicação Viral
18.
Blood ; 116(26): 5842-8, 2010 Dec 23.
Artigo em Inglês | MEDLINE | ID: mdl-20876851

RESUMO

Inhibitory antibodies to factor VIII (FVIII) are a major complication in the treatment of hemophilia A, affecting approximately 20% to 30% of patients. Current treatment for inhibitors is based on long-term, daily injections of large amounts of FVIII protein. Liver-directed gene therapy has been used to induce antigen-specific tolerance, but there are no data in hemophilic animals with pre-existing inhibitors. To determine whether sustained endogenous expression of FVIII could eradicate inhibitors, we injected adeno-associated viral vectors encoding canine FVIII (cFVIII) in 2 strains of inhibitor hemophilia A dogs. In 3 dogs, a transient increase in inhibitor titers (up to 7 Bethesda Units [BU]) at 2 weeks was followed by continuous decline to complete disappearance within 4-5 weeks. Subsequently, an increase in cFVIII levels (1.5%-8%), a shortening of clotting times, and a reduction (> 90%) of bleeding episodes were observed. Immune tolerance was confirmed by lack of antibody formation after repeated challenges with cFVIII protein and normal protein half-life. A fourth dog exhibited a strong early anamnestic response (216 BU), with slow decline to 0.8 BU and cFVIII antigen detection by 18 months after vector delivery. These data suggest that liver gene therapy has the potential to eradicate inhibitors and could improve the outcomes of hemophilia A patients.


Assuntos
Anticorpos Neutralizantes/imunologia , Fator VIII/imunologia , Fator VIII/farmacocinética , Terapia Genética , Hemofilia A/terapia , Fígado/fisiologia , Adenoviridae/genética , Animais , Cães , Fator VIII/genética , Citometria de Fluxo , Vetores Genéticos , Meia-Vida , Hemofilia A/genética , Hemofilia A/veterinária , Tolerância Imunológica , Masculino , Distribuição Tecidual
19.
Blood ; 115(23): 4678-88, 2010 Jun 10.
Artigo em Inglês | MEDLINE | ID: mdl-20335222

RESUMO

Muscle represents an important tissue target for adeno-associated viral (AAV) vector-mediated gene transfer of the factor IX (FIX) gene in hemophilia B (HB) subjects with advanced liver disease. Previous studies of direct intramuscular administration of an AAV-FIX vector in humans showed limited efficacy. Here we adapted an intravascular delivery system of AAV vectors encoding the FIX transgene to skeletal muscle of HB dogs. The procedure, performed under transient immunosuppression (IS), resulted in widespread transduction of muscle and sustained, dose-dependent therapeutic levels of canine FIX transgene up to 10-fold higher than those obtained by intramuscular delivery. Correction of bleeding time correlated clinically with a dramatic reduction of spontaneous bleeding episodes. None of the dogs (n = 14) receiving the AAV vector under transient IS developed inhibitory antibodies to canine FIX; transient inhibitor was detected after vector delivery without IS. The use of AAV serotypes with high tropism for muscle and low susceptibility to anti-AAV2 antibodies allowed for efficient vector administration in naive dogs and in the presence of low- but not high-titer anti-AAV2 antibodies. Collectively, these results demonstrate the feasibility of this approach for treatment of HB and highlight the importance of IS to prevent immune responses to the FIX transgene product.


Assuntos
Dependovirus , Fator IX/biossíntese , Terapia Genética , Vetores Genéticos , Hemofilia B/terapia , Terapia de Imunossupressão , Músculo Esquelético , Animais , Anticorpos Antivirais/sangue , Inibidores dos Fatores de Coagulação Sanguínea/sangue , Cães , Fator IX/genética , Hemofilia B/sangue , Hemofilia B/genética , Hemorragia/sangue , Hemorragia/genética , Hemorragia/terapia , Humanos , Injeções Intramusculares , Transdução Genética
20.
Mol Ther ; 19(3): 442-9, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21081906

RESUMO

Developing adeno-associated viral (AAV)-mediated gene therapy for hemophilia A (HA) has been challenging due to the large size of the factor VIII (FVIII) complementary DNA and the concern for the development of inhibitory antibodies to FVIII in HA patients. Here, we perform a systematic study in HA dogs by delivering a canine FVIII (cFVIII) transgene either as a single chain or two chains in an AAV vector. An optimized cFVIII single chain delivered using AAV serotype 8 (AAV8) by peripheral vein injection resulted in a dose-response with sustained expression of FVIII up to 7% (n = 4). Five HA dogs administered two-chain delivery using either AAV8 or AAV9 via the portal vein expressed long-term, vector dose-dependent levels of FVIII activity (up to 10%). In the two-chain approach, circulating cFVIII antigen levels were more than fivefold higher than activity. Notably, no long-term immune response to FVIII was observed in any of the dogs (1/9 dogs had a transient inhibitor). Long-term follow-up of the dogs showed a remarkable reduction (>90%) of bleeding episodes in a combined total of 24 years of observation. These data demonstrate that both approaches are safe and achieve dose-dependent therapeutic levels of FVIII expression, which supports translational studies of AAV-mediated delivery for HA.


Assuntos
Dependovirus , Fator VIII , Terapia Genética , Vetores Genéticos , Hemofilia A , Fígado , Animais , Dependovirus/genética , Modelos Animais de Doenças , Cães , Fator VIII/genética , Fator VIII/metabolismo , Regulação da Expressão Gênica , Ordem dos Genes , Técnicas de Transferência de Genes , Vetores Genéticos/genética , Hemofilia A/imunologia , Hemofilia A/prevenção & controle , Hemofilia A/terapia , Fígado/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fenótipo , Resultado do Tratamento
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA