Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
Gene Ther ; 19(1): 78-85, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21716299

RESUMO

The ability of transient immunosuppression with a combination of a non-depleting anti-CD4 (NDCD4) antibody and cyclosporine (CyA) to abrogate immune reactivity to both adeno-associated viral vector (AAV) and its transgene product was evaluated. This combination of immunosuppressants resulted in a 20-fold reduction in the resulting anti-AAV8 antibody titres, to levels in naïve mice, following intravenous administration of 2 × 10(12) AAV8 vector particles per kg to immunocompetent mice. This allowed efficient transduction upon secondary challenge with vector pseudotyped with the same capsid. Persistent tolerance did not result, however, as an anti-AAV8 antibody response was elicited upon rechallenge with AAV8 without immunosuppression. The route of vector administration, vector dose, AAV serotype or the concomitant administration of adenoviral vector appeared to have little impact on the ability of the NDCD4 antibody and CyA combination to moderate the primary humoral response to AAV capsid proteins. The combination of NDCD4 and CyA also abrogated the humoral response to the transgene product, that otherwise invariably would occur, following intramuscular injection of AAV5, leading to stable transgene expression. These observations could significantly improve the prospects of using rAAV vectors for chronic disorders by allowing for repeated vector administration and avoiding the development of antibodies to the transgene product.


Assuntos
Anticorpos Antivirais/imunologia , Proteínas do Capsídeo/imunologia , Ciclosporina/farmacologia , Dependovirus/metabolismo , Terapia Genética/métodos , Imunidade Humoral , Adenoviridae/genética , Adenoviridae/metabolismo , Animais , Anticorpos Antivirais/administração & dosagem , Linfócitos T CD4-Positivos/imunologia , Proteínas do Capsídeo/genética , Proteínas do Capsídeo/metabolismo , Ciclosporina/administração & dosagem , Dependovirus/genética , Dependovirus/imunologia , Técnicas de Transferência de Genes , Vetores Genéticos/administração & dosagem , Vetores Genéticos/genética , Vetores Genéticos/imunologia , Vetores Genéticos/metabolismo , Humanos , Terapia de Imunossupressão , Injeções Intramusculares , Injeções Intravenosas , Interferon beta/genética , Interferon beta/imunologia , Interferon beta/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Proteínas Recombinantes/genética , Proteínas Recombinantes/imunologia , Proteínas Recombinantes/metabolismo , Transgenes
2.
Gene Ther ; 17(4): 486-93, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20010626

RESUMO

Gene therapy for inherited retinal degeneration in which expression of a mutant allele has a gain-of-function effect on photoreceptor cells is likely to depend on efficient silencing of the mutated allele. Peripherin-2 (Prph2, also known as peripherin/RDS) is an abundantly expressed photoreceptor-specific gene. In humans, gain-of-function mutations in PRPH2 result in both autosomal dominant retinitis pigmentosa and dominant maculopathies. Gene-silencing strategies for these conditions include RNA interference by short hairpin RNAs (shRNAs). Recent evidence suggests that microRNA (miRNA)-based hairpins may offer a safer and more effective alternative. In this study, we used for the first time a virally transferred miRNA-based hairpin to silence Prph2 in the murine retina. The results show that an miRNA-based shRNA can efficiently and specifically silence Prph2 in vivo as early as 3 weeks after AAV2/8-mediated subretinal delivery, leading to a nearly 50% reduction of photoreceptor cells after 5 weeks. We conclude that miRNA-based hairpins can achieve rapid and robust gene silencing after efficient vector-mediated delivery to the retina. The rationale of using an miRNA-based template to improve the silencing efficiency of a hairpin may prove valuable for allele-specific silencing in which the choice for an RNAi target is limited and offers an alternative therapeutic strategy for the treatment of dominant retinopathies.


Assuntos
Terapia Genética/métodos , Proteínas de Filamentos Intermediários/genética , Glicoproteínas de Membrana/genética , MicroRNAs/genética , Proteínas do Tecido Nervoso/genética , Interferência de RNA , Degeneração Retiniana/terapia , Animais , Pareamento de Bases , Sequência de Bases , Western Blotting , Primers do DNA/genética , Dependovirus , Imuno-Histoquímica , Camundongos , Dados de Sequência Molecular , Periferinas , Degeneração Retiniana/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa
3.
Gene Ther ; 16(1): 60-9, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18701909

RESUMO

A number of distinct factors acting at different stages of the adeno-associated virus vector (AAV)-mediated gene transfer process were found to influence murine hepatocyte transduction. Foremost among these was the viral capsid protein. Self-complementary (sc) AAV pseudotyped with capsid from serotype 8 or rh.10 mediated fourfold greater hepatocyte transduction for a given vector dose when compared with vector packaged with AAV7 capsid. An almost linear relationship between vector dose and transgene expression was noted for all serotypes with vector doses as low as 1 x 10(7) vg per mouse (4 x 10(8) vg kg(-1)) mediating therapeutic levels of human FIX (hFIX) expression. Gender significantly influenced scAAV-mediated transgene expression, with twofold higher levels of expression observed in male compared with female mice. Pretreatment of mice with the proteasome inhibitor bortezomib increased scAAV-mediated hFIX expression from 4+/-0.6 to 9+/-2 microg ml(-1) in female mice, although the effect of this agent was less profound in males. Exposure of mice to adenovirus 10-20 weeks after gene transfer with AAV vectors augmented AAV transgene expression twofold by increasing the level of proviral mRNA. Hence, optimization of individual steps in the AAV gene transfer process can further enhance the potency of AAV-mediated transgene expression, thus increasing the probability of successful gene therapy.


Assuntos
Dependovirus/genética , Terapia Genética/métodos , Vetores Genéticos/administração & dosagem , Fígado/metabolismo , Transdução Genética/métodos , Animais , Anticorpos Antivirais/análise , Ácidos Borônicos/farmacologia , Bortezomib , Dependovirus/imunologia , Dependovirus/metabolismo , Fator IX/genética , Fator IX/metabolismo , Feminino , Expressão Gênica , Vetores Genéticos/genética , Vetores Genéticos/imunologia , Humanos , Injeções Intravenosas , Fígado/imunologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Inibidores de Proteases/farmacologia , Pirazinas/farmacologia , Transgenes
4.
Gene Ther ; 15(6): 463-7, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18004402

RESUMO

To date adeno-associated viral (AAV) vectors are the only gene therapy vectors that have been shown to efficiently transduce photoreceptor cells and have thus become the most commonly used vector for ocular transduction. Various AAV serotypes have been evaluated in the eye, the first of which was AAV2, which is able to transduce photoreceptors, retinal pigment epithelium (RPE) and retinal ganglion cells. AAV serotypes 1 and 4, as well as AAV2 pseudotyped with these capsids, only transduce the RPE. AAV serotype 5 and AAV2/5 transduce the photoreceptors as well as RPE, but not retinal ganglion cells. Here, we assessed the capacity of the novel serotype AAV2/8 to transduce various ocular tissues of the adult murine retina by administering AAV2/8 green fluorescent protein intravitreally, subretinally and intracamerally. We also determined the kinetics and efficiency of self-complementary AAV (scAAV) vectors of serotypes 2/2, 2/5 and 2/8 and compared them with single-stranded AAV (ssAAV). We found that ssAAV2/8 transduces photoreceptors and RPE more efficiently than ssAAV2/2 and ssAAV2/5, and that scAAV2/8 had faster onset and higher transgene expression than ssAAV2/8. This improved transduction efficiency might facilitate the development of improved gene therapy protocols for inherited retinal degenerations, particularly those caused by defects in photoreceptor-specific genes.


Assuntos
Dependovirus/genética , Terapia Genética/métodos , Vetores Genéticos/administração & dosagem , Degeneração Retiniana/terapia , Transdução Genética/métodos , Animais , DNA Complementar , DNA de Cadeia Simples , Fundo de Olho , Expressão Gênica , Vetores Genéticos/genética , Proteínas de Fluorescência Verde/genética , Camundongos , Microscopia de Fluorescência , Epitélio Pigmentado Ocular/metabolismo , Células Ganglionares da Retina/metabolismo , Transgenes
5.
Leukemia ; 21(10): 2181-91, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17657224

RESUMO

We describe a new model of myeloma bone disease in which beta2m NOD/SCID mice injected with KMS-12-BM cells develop medullary disease after tail vein administration. Micro-computed tomography analysis demonstrated significant bone loss in the tibiae and vertebrae of diseased animals compared to controls, with loss of cortical bone (P<0.01), as well as trabecular bone volume, thickness and number (P<0.05 for all). Bone marrow of diseased animals demonstrated an increase in osteoclasts (P<0.01) and reduction in osteoblasts (P<0.01) compared to control animals. Both bone loss and osteoclast increase correlated with the degree of disease involvement. Mesenchymal stem cells (MSCs) were lentivirally transduced to express human osteoprotegerin (hOPG). Systemic administration of OPG expressing MSC reduced osteoclast activation (P<0.01) and trabecular bone loss in the vertebrae (P<0.05) and tibiae of diseased animals, to levels comparable to non-diseased controls. Because of its predominantly medullary involvement and quantifiable parameters of bone disease, the KMS-12-BM xenogeneic model provides unique opportunities to test therapies targeted at the bone marrow microenvironment.


Assuntos
Modelos Animais de Doenças , Lentivirus/genética , Células-Tronco Mesenquimais/citologia , Mieloma Múltiplo/patologia , Mieloma Múltiplo/terapia , Osteoprotegerina/biossíntese , Animais , Osso e Ossos/metabolismo , Linhagem Celular , Terapia Genética/métodos , Humanos , Cinética , Lentivirus/metabolismo , Células-Tronco Mesenquimais/metabolismo , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Transplante de Neoplasias , Osteoblastos/metabolismo , Osteoclastos/metabolismo , Tíbia/patologia
6.
Cancer Gene Ther ; 13(1): 99-106, 2006 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-16052229

RESUMO

Type I interferons (alpha/beta) have significant antitumor activity although their short half-life and systemic side effects have limited their clinical utility. An alternative dosing schedule of continuous, low-level delivery, as is achieved by gene therapy, rather than intermittent, high concentration pulsed-dosing, might avoid the toxicity of interferon while maintaining its antitumor efficacy. We have tested a gene therapy approach in murine tumor models to treat malignancies that have shown responsiveness to interferon in clinical trials. The tumor cell lines used were moderately sensitive to the direct effects of human interferon-beta (hIFN-beta) in vitro. For in vivo testing, systemic delivery of hIFN-beta was generated following liver-targeted delivery of adeno-associated virus (AAV) vector carrying the hIFN-beta transgene. This prevented engraftment of subcutaneous human gliomas, and orthotopic, localized (intrarenal) and disseminated (primarily pulmonary) human renal cell carcinomas; and caused regression of established tumors at these sites. In a syngeneic, immunocompetent model of melanoma, AAV IFN-beta treatment limited subcutaneous tumor growth and prevented disseminated disease. A significant decrease in mean intratumoral vessel density was demonstrated in hIFN-beta-treated tumors, suggesting that in addition to a direct tumoricidal effect, the antitumor efficacy of AAV IFN-beta in this study was due to its ability to inhibit angiogenesis.


Assuntos
Dependovirus/metabolismo , Terapia Genética/métodos , Vetores Genéticos/metabolismo , Interferon beta/metabolismo , Animais , Carcinoma de Células Renais/metabolismo , Carcinoma de Células Renais/terapia , Glioblastoma/metabolismo , Glioblastoma/terapia , Humanos , Masculino , Camundongos , Modelos Genéticos , Metástase Neoplásica/prevenção & controle , Sensibilidade e Especificidade
7.
Leukemia ; 24(4): 806-12, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20130599

RESUMO

Acute lymphoblastic leukemia (ALL) harboring the t(4;11) translocation is associated with a very poor prognosis; innovative treatment strategies are required to improve the current 5-year survival rate of 30-40%. Interferon beta (IFN beta) has shown promise in the treatment of both solid and hematologic malignancies, although the short half-life and toxicity associated with high doses have limited its clinical utility. To overcome these limitations, we investigated the effect of continuous, gene transfer-mediated delivery of IFN beta using adeno-associated virus (AAV)-mediated expression, on ALL cells with the t(4;11) translocation. We found that this method of IFN beta delivery resulted in complete remission of leukemia in a murine model. However, leukemic cells eventually became resistant to IFN beta and relapse was observed. Activation of NF-kappaB was identified as a mechanism for IFN beta resistance, and inhibition of NF-kappaB activity in resistant cells sensitized cells to IFN beta. IFN beta combined with agents that inhibit NF-kappaB could have therapeutic potential in the treatment of children with mixed lineage leukemia subtype ALL.


Assuntos
Antineoplásicos/farmacologia , Regulação Leucêmica da Expressão Gênica , Rearranjo Gênico , Interferon beta/farmacologia , Proteína de Leucina Linfoide-Mieloide/genética , NF-kappa B/metabolismo , Leucemia-Linfoma Linfoblástico de Células Precursoras/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Apoptose/efeitos dos fármacos , Western Blotting , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Cromossomos Humanos Par 11/genética , Cromossomos Humanos Par 4/genética , Dependovirus/genética , Resistencia a Medicamentos Antineoplásicos , Ensaio de Desvio de Mobilidade Eletroforética , Citometria de Fluxo , Humanos , Proteínas Quinases JNK Ativadas por Mitógeno/genética , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Masculino , Camundongos , Camundongos SCID , Proteína de Leucina Linfoide-Mieloide/metabolismo , NF-kappa B/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamento farmacológico , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Translocação Genética , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
8.
Anticancer Res ; 30(9): 3301-8, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20944101

RESUMO

BACKGROUND: The vascular niche necessary for cancer stem cell maintenance is a potential target for cancer therapy. MATERIALS AND METHODS: Human glioma xenografts were treated with IFN-ß delivered systemically via a liver-targeted, adeno-associated viral vector. The vascular niche was examined with immunofluorescence for glioma stem cells, endothelial cells, and perivascular cells. RESULTS: Although IFN-ß was not directly toxic to glioma stem cells in vitro, IFN-ß decreased tumor size and the number of stem cells recovered in both heterotopic and orthotopic models. Treatment with IFN-ß increased perivascular cells investing the tumor vasculature (6-fold) distancing stem cells from endothelial cells. Additionally, vascular smooth muscle cells co-cultured between stem cells and endothelial cells decreased stem cell recovery. CONCLUSION: Continuous delivery of IFN-ß decreased the number of stem cells in glioma xenografts by disrupting the vascular niche through an increase in perivascular cells, which created a barrier between the glioma stem cells and the endothelial cells.


Assuntos
Antineoplásicos/farmacologia , Neoplasias Encefálicas/irrigação sanguínea , Glioma/irrigação sanguínea , Interferon beta/farmacologia , Células-Tronco Neoplásicas/efeitos dos fármacos , Animais , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/metabolismo , Comunicação Celular/efeitos dos fármacos , Técnicas de Cocultura , Células Endoteliais/efeitos dos fármacos , Imunofluorescência , Glioma/tratamento farmacológico , Glioma/metabolismo , Humanos , Masculino , Camundongos , Camundongos SCID , Pericitos/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto
9.
Haematologica ; 78(3): 162-6, 1993.
Artigo em Inglês | MEDLINE | ID: mdl-8375745

RESUMO

BACKGROUND AND METHODS: We designed a chemotherapy schedule similar to that used in childhood acute lymphoblastic leukaemia for the management of patients with Philadelphia chromosome positive chronic myeloid leukaemia in lymphoid blast transformation. It includes the use of intensive systemic chemotherapy and intrathecal methotrexate followed by 'maintenance' chemotherapy for 2 years or allogeneic bone marrow transplantation. RESULTS AND CONCLUSIONS: Of the four patients treated with this schedule, two survive at 30 and 36 months from transformation respectively. The overall median survival is 19 months. A coordinated approach to this relatively rare clinical problem may be useful.


Assuntos
Leucemia Mielogênica Crônica BCR-ABL Positiva/tratamento farmacológico , Ativação Linfocitária , Adulto , Transplante de Medula Óssea , Feminino , Humanos , Leucemia Mielogênica Crônica BCR-ABL Positiva/cirurgia , Masculino , Metotrexato/administração & dosagem , Metotrexato/uso terapêutico , Pessoa de Meia-Idade , Indução de Remissão
10.
Vox Sang ; 87(2): 73-81, 2004 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-15355497

RESUMO

Gene therapy is a new and exciting therapeutic concept that offers the promise of cure for an array of inherited, malignant and infectious disorders. After years of failure, substantial progress in the efficiency of gene-transfer technology has recently resulted in impressive clinical success in infants with immunodeficiency. Two of these children have, however, subsequently developed leukaemia as a result of insertional mutagenesis, raising concerns about the safety of genetic therapeutics. The purpose of this article is to review the current status of gene therapy in light of recent successes and tragedies, and to consider the challenges faced by this relatively new field.


Assuntos
Terapia Genética/tendências , Criança , Ensaios Clínicos como Assunto , Previsões , Técnicas de Transferência de Genes , Terapia Genética/efeitos adversos , Vetores Genéticos , Hemofilia A/terapia , Humanos , Neoplasias/terapia
11.
Haemophilia ; 10(4): 309-18, 2004 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15230943

RESUMO

That gene therapy offers the promise of a cure for haemophilia was apparent more than a decade ago. After years of failure, substantial progress in the efficiency of gene transfer technology has recently resulted in impressive success in animal models with haemophilia. However, fears of the risks intrinsic to such therapy have been raised by the fate of two children cured of immune deficiency by gene transfer who have, however, subsequently developed leukaemia as a result of insertional mutagenesis. The purpose of this review is to outline the current status of gene therapy in light of recent successes and tragedies and to consider the prospects for curing haemophilia in the short-to-medium term.


Assuntos
Terapia Genética/métodos , Hemofilia A/terapia , Adenoviridae/genética , Terapia Genética/efeitos adversos , Vetores Genéticos/genética , Vetores Genéticos/imunologia , Humanos , Retroviridae/genética
12.
Br J Haematol ; 88(1): 122-8, 1994 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-7803234

RESUMO

Tissue factor (TF) is a cellular receptor and cofactor for factor VII/VIIa which initiates the blood coagulation cascade. We have investigated the role of 5'-flanking DNA sequences in regulating the expression of the human TF gene in human umbilical vein endothelial cells (HUVEC). Using a chloramphenicol acetyltransferase (CAT) reporter gene, we attempted to transfect primary cultured HUVEC (passage 3-4) with calcium phosphate coprecipitation, DEAE Dextran, lipopolyamine-coated DNA or electroporation. Electroporation in HEPES-buffered saline of 1 x 10(7) cells at 200V and 250 microF was found to be optimal. Using these conditions, varying lengths of TF 5'-flanking sequences coupled to the CAT reporter gene were tested in transient expression studies. CAT expression corrected for variation in transfection efficiency and cell viability revealed that the sequences between -111 and +14 base pairs are essential for minimal transcriptional activity. This region contains consensus sequences for a TATA box and three Sp1 binding sites. A domain from -382 to -111bp, which contains two AP-1 consensus elements, promoted high levels of gene expression. This transcriptional activity was repressed by 50% with constructs containing sequences between -550 and -382 bp. A further 2-fold drop in transcription activity was attributed to the region between -948 and -550 bp. These results suggest that the basal transcription of the human TF gene in HUVEC is mediated through at least two negative regulatory elements upstream of the proximal promoter domain. The proximal promoter region which contains two AP-1 sites is essential for efficient transcription.


Assuntos
Endotélio Vascular/citologia , Técnicas de Transferência de Genes , Regiões Promotoras Genéticas , Tromboplastina/genética , Veias Umbilicais/citologia , Sequência de Bases , Células Cultivadas , Eletroporação , Regulação da Expressão Gênica , Humanos , Sequências Reguladoras de Ácido Nucleico , Transcrição Gênica
13.
Gene Ther ; 7(3): 183-95, 2000 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-10694794

RESUMO

Recombinant adeno-associated viral (rAAV) vectors have been evaluated for their ability to transduce primitive hematopoietic cells. Early studies documented rAAV-mediated gene expression during progenitor derived colony formation in vitro, but studies examining genome integration and long-term gene expression in hematopoietic cells have yielded conflicting results. Such studies were performed with crude vector preparations. Using improved methodology, we have generated high titer, biologically active preparations of rAAV free of wild-type AAV (less than 1/107particles) and adenovirus. Transduction of CD34+ cells from umbilical cord blood was evaluated with a bicistronic rAAV vector encoding the green fluorescent protein (GFP) and a trimetrexate resistant variant of dihydrofolate reductase (DHFR). Freshly isolated, quiescent CD34+ cells were resistant to transduction (less than 4%), but transduction increased to 23 +/- 2% after 2 days of cytokine stimulation and was further augmented by addition of tumor necrosis factor alpha (51 +/- 4%) at a multiplicity of infection of 106. rAAV-mediated gene expression was transient in that progenitor derived colony formation was inhibited by trimetrexate. Primitive CD34+ and CD34+, CD38- subsets were sequentially transduced with a rAAV vector encoding the murine ecotropic receptor followed by transduction with an ecotropic retroviral vector encoding GFP and DHFR. Under optimal conditions 41 +/- 7% of CD34+ progenitors and 21 +/- 6% of CD34+, CD38- progenitors became trimetrexate resistant. These results document that highly purified rAAV transduce primitive human hematopoietic cells efficiently but gene expression appears to be transient. Gene Therapy (2000) 7, 183-195.


Assuntos
Adenoviridae/genética , Antígenos CD34/genética , Antígenos CD , Antígenos de Diferenciação/genética , Vetores Genéticos/genética , Células-Tronco Hematopoéticas/fisiologia , NAD+ Nucleosidase/genética , ADP-Ribosil Ciclase , ADP-Ribosil Ciclase 1 , Animais , Células Cultivadas , Sangue Fetal , Técnicas de Transferência de Genes , Vírus Auxiliares/genética , Humanos , Glicoproteínas de Membrana , Camundongos , Transdução Genética/genética , Fator de Necrose Tumoral alfa/farmacologia
14.
Blood ; 97(5): 1258-65, 2001 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-11222368

RESUMO

Long-term expression of coagulation factor IX (FIX) has been observed in murine and canine models following administration of recombinant adeno-associated viral (rAAV) vectors into either the portal vein or muscle. These studies were designed to evaluate factors that influence rAAV-mediated FIX expression. Stable and persistent human FIX (hFIX) expression (> 22 weeks) was observed from 4 vectors after injection into the portal circulation of immunodeficient mice. The level of expression was dependent on promoter with the highest expression, 10% of physiologic levels, observed with a vector containing the cytomegalovirus (CMV) enhancer/beta-actin promoter complex (CAGG). The kinetics of expression after injection of vector particles into muscle, tail vein, or portal vein were similar with hFIX detectable at 2 weeks and reaching a plateau by 8 weeks. For a given dose, intraportal administration of rAAV CAGG-FIX resulted in a 1.5-fold or 4-fold higher level of hFIX compared to tail vein or intramuscular injections, respectively. Polymerase chain reaction analysis demonstrated predominant localization of the rAAV FIX genome in liver and spleen after tail vein injection with a higher proportion in liver after portal vein injection. Therapeutic levels of hFIX were detected in the majority of immunocompetent mice (21 of 22) following intravenous administration of rAAV vector without the development of anti-hFIX antibodies, but hFIX was not detected in 14 immunocompetent mice following intramuscular administration, irrespective of strain. Instead, neutralizing anti-hFIX antibodies were detected in all the mice. These observations may have important implications for hemophilia B gene therapy with rAAV vectors.


Assuntos
Fator IX/genética , Vetores Genéticos/administração & dosagem , Vetores Genéticos/farmacocinética , Transdução Genética/métodos , Animais , Anticorpos/análise , Anticorpos/sangue , DNA Complementar/farmacocinética , DNA Recombinante , Dependovirus/genética , Fator IX/imunologia , Humanos , Injeções Intramusculares , Injeções Intravenosas , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos SCID , Veia Porta , Fatores de Tempo , Distribuição Tecidual
15.
Gene Ther ; 6(8): 1456-68, 1999 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-10467370

RESUMO

One factor limiting the ability to modify human repopulating hematopoietic cells genetically with retroviral vectors is the relatively low expression of the cognate viral receptor. We have tested sequential transduction of human hematopoietic cells with an adenoviral vector encoding the ecotropic retroviral receptor followed by transduction with an ecotropic retroviral vector. Adenoviral transduction of K562 erythroleukemia cells was highly efficiently with >95% of cells expressing the ecotropic receptor at a multiplicity of infection (MOI) of 103with a correspondingly high transduction with a retroviral vector. Ecotropic receptor expression in CD34+ cells following transduction with adenoviral vectors was increased by at least two-fold (from 20 to 48%) by replacing the RSV promoter with the CMV E1a promoter, resulting in a parallel increase in retroviral transduction efficiency. Replacing the head portion of the fiber protein in conventional adenoviral vectors (serotype 5) with the corresponding portion from an adenoviral 3 serotype resulted in ecotropic receptor expression in 60% of CD34+ cells at an MOI of 104 and a retroviral transduction of 60% of hematopoietic clonogenic progenitors. The sequential transduction strategy also resulted in efficient transduction of the primitive CD34+CD38- subset suggesting that it may hold promise for genetic modification of human hematopoietic stem cells.


Assuntos
Adenoviridae/genética , Vetores Genéticos/genética , Células-Tronco Hematopoéticas/fisiologia , Glicoproteínas de Membrana/genética , Receptores Virais/genética , Transdução Genética/genética , Animais , Antígenos CD34/metabolismo , Células Cultivadas , Humanos , Camundongos , Proteínas Recombinantes de Fusão/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA