Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Int J Mol Sci ; 24(12)2023 Jun 14.
Artigo em Inglês | MEDLINE | ID: mdl-37373272

RESUMO

Antibodies are considered highly specific therapeutic agents in cancer medicines, and numerous formats have been developed. Among them, bispecific antibodies (BsAbs) have gained a lot of attention as a next-generation strategy for cancer therapy. However, poor tumor penetration is a major challenge because of their large size and thus contributes to suboptimal responses within cancer cells. On the other hand, affibody molecules are a new class of engineered affinity proteins and have achieved several promising results with their applications in molecular imaging diagnostics and targeted tumor therapy. In this study, an alternative format for bispecific molecules was constructed and investigated, named ZLMP110-277 and ZLMP277-110, that targets Epstein-Barr virus latent membrane protein 1 (LMP1) and latent membrane protein 2 (LMP2). Surface plasmon resonance (SPR), indirect immunofluorescence assay, co-immunoprecipitation, and near-infrared (NIR) imaging clearly demonstrated that ZLMP110-277 and ZLMP277-110 have good binding affinity and specificity for both LMP1 and LMP2 in vitro and in vivo. Moreover, ZLMP110-277 and ZLMP277-110, especially ZLMP277-110, significantly reduced the cell viability of C666-1 and CNE-2Z as compared to their monospecific counterparts. ZLMP110-277 and ZLMP277-110 could inhibit phosphorylation of proteins modulated by the MEK/ERK/p90RSK signaling pathway, ultimately leading to suppression of oncogene nuclear translocations. Furthermore, ZLMP110-277 and ZLMP277-110 showed significant antitumor efficacy in nasopharyngeal carcinoma-bearing nude mice. Overall, our results demonstrated that ZLMP110-277 and ZLMP277-110, especially ZLMP277-110, are promising novel prognostic indicators for molecular imaging and targeted tumor therapy of EBV-associated nasopharyngeal carcinoma.


Assuntos
Carcinoma , Infecções por Vírus Epstein-Barr , Neoplasias Nasofaríngeas , Animais , Camundongos , Carcinoma Nasofaríngeo , Herpesvirus Humano 4/fisiologia , Carcinoma/patologia , Neoplasias Nasofaríngeas/patologia , Camundongos Nus , Proteínas da Matriz Viral/metabolismo
2.
Microbiol Spectr ; 11(1): e0356222, 2023 02 14.
Artigo em Inglês | MEDLINE | ID: mdl-36511681

RESUMO

The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) pandemic has been an unprecedented public health disaster in human history, and its spike (S) protein is the major target for vaccines and antiviral drug development. Although widespread vaccination has been well established, the viral gene is prone to rapid mutation, resulting in multiple global spread waves. Therefore, specific antivirals are needed urgently, especially those against variants. In this study, the domain of the receptor binding motif (RBM) and fusion peptide (FP) (amino acids [aa] 436 to 829; denoted RBMFP) of the SARS-CoV-2 S protein was expressed as a recombinant RBMFP protein in Escherichia coli and identified as being immunogenic and antigenically active. Then, the RBMFP proteins were used for phage display to screen the novel affibody. After prokaryotic expression and selection, four novel affibody molecules (Z14, Z149, Z171, and Z327) were obtained. Through surface plasmon resonance (SPR) and pseudovirus neutralization assay, we showed that affibody molecules specifically bind to the RBMFP protein with high affinity and neutralize against SARS-CoV-2 pseudovirus infection. Especially, Z14 and Z171 displayed strong neutralizing activities against Delta and Omicron variants. Molecular docking predicted that affibody molecule interaction sites with RBM overlapped with ACE2. Thus, the novel affibody molecules could be further developed as specific neutralization agents against SARS-CoV-2 variants. IMPORTANCE SARS-CoV-2 and its variants are threatening the whole world. Although a full dose of vaccine injection showed great preventive effects and monoclonal antibody reagents have also been used for a specific treatment, the global pandemic persists. So, developing new vaccines and specific agents are needed urgently. In this work, we expressed the recombinant RBMFP protein as an antigen, identified its antigenicity, and used it as an antigen for affibody phage-display selection. After the prokaryotic expression, the specific affibody molecules were obtained and tested for pseudovirus neutralization. Results showed that the serum antibody induced by RBMFP neutralized Omicron variants. The screened affibody molecules specifically bound the RBMFP of SARS-CoV-2 with high affinity and neutralized the Delta and Omicron pseudovirus in vitro. So, the RBMFP induced serum provides neutralizing effects against pseudovirus in vitro, and the affibodies have the potential to be developed into specific prophylactic agents for SARS-CoV-2 and its variants.


Assuntos
COVID-19 , SARS-CoV-2 , Glicoproteína da Espícula de Coronavírus , Humanos , Anticorpos Neutralizantes , Anticorpos Antivirais , COVID-19/prevenção & controle , Simulação de Acoplamento Molecular , Testes de Neutralização/métodos , Proteínas Recombinantes , SARS-CoV-2/genética , SARS-CoV-2/imunologia , Glicoproteína da Espícula de Coronavírus/genética , Glicoproteína da Espícula de Coronavírus/imunologia
3.
Front Cell Infect Microbiol ; 12: 1078504, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36683690

RESUMO

Nasopharyngeal carcinoma (NPC), is an Epstein-Barr virus (EBV) associated malignancy most common in Southern China and Southeast Asia. In southern China, it is one of the major causes of cancer-related death. Despite improvement in radiotherapy and chemotherapy techniques, locoregional recurrence and distant metastasis remains the major causes for failure of treatment in NPC patients. Therefore, finding new specific drug targets for treatment interventions are urgently needed. Here, we report three potential ZLMP1-C affibody molecules (ZLMP1-C15, ZLMP1-C114 and ZLMP1-C277) that showed specific binding interactions for recombinant and native EBV LMP1 as determined by epitope mapping, co-localization and co-immunoprecipitation assays. The ZLMP1-C affibody molecules exhibited high antitumor effects on EBV-positive NPC cell lines and displayed minimal cytotoxicity towards EBV-negative NPC cell line. Moreover, ZLMP1-C277 showed higher antitumor efficacy than ZLMP1-C15 and ZLMP1-C114 affibody molecules. The ability of ZLMP1-C277 decrease the phosphorylation levels of up-stream activator phospho-Raf-1(Ser338), phospho-MEK1/2(Ser217/Ser221), phospho-ERK1/2(Thr202/Thr204), thereby leading to downstream suppression of phospho-p90RSK(Ser380) and transcription factor c-Fos. Importantly, tumor growth was reduced in tumor-bearing mice treated with ZLMP1-C277 and caused no apparent toxicity. Taken together, our findings provide evidence that ZLMP1-C277 as a promising therapeutic agent in EBV-associated NPC.


Assuntos
Infecções por Vírus Epstein-Barr , Carcinoma Nasofaríngeo , Neoplasias Nasofaríngeas , Animais , Humanos , Camundongos , Linhagem Celular Tumoral , Proliferação de Células , Infecções por Vírus Epstein-Barr/tratamento farmacológico , Herpesvirus Humano 4/metabolismo , Sistema de Sinalização das MAP Quinases , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Carcinoma Nasofaríngeo/tratamento farmacológico , Carcinoma Nasofaríngeo/virologia , Neoplasias Nasofaríngeas/tratamento farmacológico , Neoplasias Nasofaríngeas/virologia , Recidiva Local de Neoplasia , Proteínas Quinases S6 Ribossômicas 90-kDa/metabolismo , Proteínas Quinases S6 Ribossômicas 90-kDa/farmacologia , Proteínas Quinases S6 Ribossômicas 90-kDa/uso terapêutico , Ensaios Antitumorais Modelo de Xenoenxerto
4.
Appl Microbiol Biotechnol ; 105(19): 7283-7293, 2021 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-34505914

RESUMO

Nasopharyngeal carcinoma (NPC) is consistently associated with Epstein-Barr virus (EBV) latent infection and is common in Southern China and Southeast Asia. The viral latent membrane proteins LMP1 and LMP2 are persistently expressed in NPC tissues; the cytoplasmic domain of LMP1 (LMP1 C-terminal) and LMP2A (LMP2A N-terminal) proteins is essential for maintenance of latency and can alter host cell signaling to facilitate tumor growth and progression. Thus, targeting LMP1 or LMP2 oncoprotein has been an increasing interest for diagnosis and targeted therapy of NPC. Affibody molecules, a new class of small-affinity engineered scaffold proteins, have demonstrated high potential for therapeutics, diagnostics, and biotechnological applications. More recently, radiolabelled HER2-specific affibody molecules have demonstrated to be useful in imaging of HER2 expressing tumor. In this study, we report three novel EBV LMP1 C-terminal (EBV LMP1-C) domain affibody molecules (ZLMP1-C15, ZLMP1-C114, and ZLMP1-C277) were selected by biopanning from a random-peptide displayed phage library and used for molecular imaging in tumor-bearing nude mice. Surface plasmon resonance (SPR), indirect immunofluorescence, and immunohistochemistry (IHC) clearly showed that all three selected affibody molecules have high affinity and specificity in binding to EBV LMP1 protein. Moreover, in vivo tumor imaging revealed that Dylight-755-labeled affibody molecules accumulated rapidly in tumor site after injection (1 h) and then were continuously maintained for 24 h in EBV-positive NPC xenograft mice model. In conclusion, our findings highlight the potential use of ZLMP1-C affibody molecules as tumor-specific molecular imaging agents of EBV-associated NPC.Key points• We screened three novel affibody molecules (ZLMP1-C15, ZLMP1-C114, and ZLMP1-C277) targeting EBV LMP1-C terminal domain• ZLMP1-C recognize the recombinant and native LMP1-C with high affinity and specificity• ZLMP1-C can be used for molecular imaging.


Assuntos
Infecções por Vírus Epstein-Barr , Neoplasias Nasofaríngeas , Animais , Infecções por Vírus Epstein-Barr/diagnóstico por imagem , Herpesvirus Humano 4 , Camundongos , Camundongos Nus , Imagem Molecular , Carcinoma Nasofaríngeo , Neoplasias Nasofaríngeas/diagnóstico por imagem
5.
J Org Chem ; 84(7): 4040-4049, 2019 04 05.
Artigo em Inglês | MEDLINE | ID: mdl-30854850

RESUMO

A novel oxidation of benzylic C-H bonds for the synthesis of diverse six-membered N-heteroaromatic aldehydes and ketones has been developed. The obvious advantages of this approach are the simple operation, mild reaction conditions, and without use of toxic reagent and transition metal. The present method should provide a useful access for the synthesis and modification of N-heterocycles.

6.
Mol Ther ; 18(5): 965-76, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20354504

RESUMO

The development of leukemia as a consequence of vector-mediated genotoxicity in gene therapy trials for X-linked severe combined immunodeficiency (SCID-X1) has prompted substantial research effort into the design and safety testing of integrating vectors. An important element of vector design is the selection and evaluation of promoter-enhancer elements with sufficient strength to drive reliable immune reconstitution, but minimal propensity for enhancer-mediated insertional mutagenesis. In this study, we set out to explore the effect of promoter-enhancer selection on the efficacy and safety of human immunodeficiency virus-1-derived lentiviral vectors in gammac-deficient mice. We observed incomplete or absent T- and B-cell development in mice transplanted with progenitors expressing gammac from the phosphoglycerate kinase (PGK) and Wiscott-Aldrich syndrome (WAS) promoters, respectively. In contrast, functional T- and B-cell compartments were restored in mice receiving an equivalent vector containing the elongation factor-1-alpha (EF1alpha) promoter; however, 4 of 14 mice reconstituted with this vector subsequently developed lymphoma. Extensive analyses failed to implicate insertional mutagenesis or gammac overexpression as the underlying mechanism. These findings highlight the need for detailed mechanistic analysis of tumor readouts in preclinical animal models assessing vector safety, and suggest the existence of other ill-defined risk factors for oncogenesis, including replicative stress, in gene therapy protocols targeting the hematopoietic compartment.


Assuntos
Subunidade gama Comum de Receptores de Interleucina/fisiologia , Lentivirus/genética , Linfoma/etiologia , Linfoma/genética , Mutagênese Insercional , Doenças por Imunodeficiência Combinada Ligada ao Cromossomo X/terapia , Animais , Células Cultivadas , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Terapia Genética/efeitos adversos , Vetores Genéticos/efeitos adversos , Vetores Genéticos/genética , Subunidade gama Comum de Receptores de Interleucina/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos SCID , Análise de Sequência com Séries de Oligonucleotídeos , Reação em Cadeia da Polimerase , Regiões Promotoras Genéticas/genética , Doenças por Imunodeficiência Combinada Ligada ao Cromossomo X/genética
7.
Immunol Cell Biol ; 88(2): 228-32, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19935770

RESUMO

Genetic fusion of tandem repeats of the complement molecule C3d has been shown to considerably enhance immune responses to genetic vaccines. We have investigated the applicability of this approach to augment humoral immune responses toward vaccines delivered by recombinant adeno-associated virus (AAV) vectors. C3d(3)-fusion was found to markedly decrease antibody responses to merozoite surface protein 4/5 from Plasmodium yoelii and contrasted with greater than 50-fold enhancement in responses when this strategy was similarly applied to another AAV-encoded model antigen, hen egg lysozyme. These data indicate that the efficacy of the C3d(3) strategy operates in an antigen-dependent manner. Additional studies also showed that homologous recombination events between the C3d tandem repeats occurred during vector packaging and transduction resulting in expression of C3d(1)-, C3d(2)-, C3d(3)- and C3d(4)-fused antigen. This is the first report to apply the C3d approach to augment responses against a recombinant viral vector system and the consequences of these findings are discussed.


Assuntos
Antígenos/imunologia , Complemento C3d/imunologia , Dependovirus/genética , Imunidade Humoral/imunologia , Proteínas Recombinantes de Fusão/imunologia , Transgenes/imunologia , Vacinas de DNA/imunologia , Animais , Formação de Anticorpos/imunologia , Linhagem Celular , Dependovirus/imunologia , Vetores Genéticos/administração & dosagem , Vetores Genéticos/imunologia , Humanos , Camundongos , Titulometria , Vacinas Virais/imunologia
8.
Liver Transpl ; 15(2): 233-41, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19177450

RESUMO

The aim of this study was to evaluate the ability of local overexpression of indoleamine dioxygenase (IDO) to abrogate rat liver transplant rejection by the use of an adeno-associated virus vector [recombinant adeno-associated virus 2/8 (rAAV2/8)] to deliver the transgene to the allograft prior to transplantation. A green fluorescent protein (GFP)-expressing vector [recombinant adeno-associated virus 2/8-liver-specific promoter 1-enhanced green fluorescent protein (rAAV2/8-LSP1-eGFP)] was used to examine the kinetics of expression and optimal dosing for transduction of Piebald Virol Glaxo (PVG) rat livers. A vector encoding the rat IDO gene (rAAV2/8-LSP1-rIDO) was constructed and tested by its ability to induce tryptophan catabolism and kynurenine production in vitro and in vivo. PVG donor rats were injected, via the portal vein, with rAAV2/8-LSP1-rIDO 2 weeks before transplantation into PVG strain isograft or Lewis (LEW) strain allograft recipients. With the enhanced GFP vector, 29.5% and 47.4% of hepatocytes were found to express GFP at 3 and 6 weeks after injection, respectively. In untransplanted PVG animals, the rAAV2/8-LSP1-rIDO vector induced, 3 weeks after administration, a 1.8-fold increase (P = 0.0161) in liver IDO activity, which was associated with a fall in serum tryptophan to 0.5 times the baseline level (P < 0.001). PVG recipients of PVG liver isografts pretreated with the IDO-expressing vector had a 45% lower level of serum tryptophan than recipients of isografts pretreated with the GFP-expressing vector (P = 0.03). LEW recipients of PVG liver allografts pretreated with the rat IDO vector had a median survival time of 12 days, whereas recipients of allografts pretreated with rAAV2/8-LSP1-eGFP had a median survival time of 13 days (P = 0.38). Both groups displayed similar histological features of acute cellular rejection. In conclusion, rAAV2/8 vectors produce highly efficient, though delayed, hepatocyte transduction in vivo and provide a useful gene delivery tool for transplantation models. However, gene delivery using IDO was unsuccessful in prolonging rat liver allograft survival.


Assuntos
Regulação Enzimológica da Expressão Gênica , Rejeição de Enxerto/prevenção & controle , Indolamina-Pirrol 2,3,-Dioxigenase/biossíntese , Indolamina-Pirrol 2,3,-Dioxigenase/genética , Transplante de Fígado/efeitos adversos , Animais , Dependovirus/genética , Terapia Genética/métodos , Vetores Genéticos/genética , Rejeição de Enxerto/enzimologia , Rejeição de Enxerto/genética , Sobrevivência de Enxerto/genética , Ratos , Transplante Homólogo , Regulação para Cima
9.
J Gene Med ; 9(6): 429-39, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17410609

RESUMO

BACKGROUND: Development of effective and durable gene therapy for treatment of the respiratory manifestations of cystic fibrosis remains a formidable challenge. Obstacles include difficulty in achieving efficient gene transfer to mature airway epithelium and the need to stably transduce self-renewing epithelial progenitor cells in order to avoid loss of transgene expression through epithelial turnover. Targeting the developing airway epithelium during fetal life offers the prospect of circumventing these challenges. METHODS: In the current study we investigated vesicular stomatitis virus glycoprotein (VSVg)-pseudotyped HIV-1-derived lentivirus vector-mediated gene transfer to the airway epithelium of mid-gestation fetal lambs, both in vitro and in vivo. In the in vitro studies epithelial sheet explants and lung organ culture were used to examine transduction of the proximal and more distal airway epithelium, respectively. For the in vivo studies, vector was delivered directly into the proximal airway. RESULTS: We found that even during the early pseudoglandular and canalicular phases of lung development, occurring through mid-gestation, the proximal bronchial airway epithelium was relatively mature and highly resistant to lentivirus-mediated transduction. In contrast, the more distal bronchiolar airway epithelium was relatively permissive for transduction although the absolute levels achieved remained low. CONCLUSION: This result is promising as the bronchiolar airway epithelium is a major site of pathology in the cystic fibrosis airway, and much higher levels of transduction are likely to be achieved by developing strategies that increase the amount of vector reaching the more distal airway after intratracheal delivery.


Assuntos
Feto/metabolismo , Vetores Genéticos/genética , Lentivirus/genética , Mucosa Respiratória/embriologia , Mucosa Respiratória/metabolismo , Ovinos/metabolismo , Transdução Genética , Animais , Líquidos Corporais/metabolismo , Brônquios/citologia , Brônquios/metabolismo , Células Cultivadas , Células Epiteliais/citologia , Células Epiteliais/metabolismo , Feminino , Sangue Fetal/metabolismo , Feto/citologia , Glicoproteínas/metabolismo , Humanos , Gravidez , Mucosa Respiratória/citologia , Vírus da Estomatite Vesicular Indiana/química
10.
Vaccine ; 25(6): 1014-22, 2007 Jan 22.
Artigo em Inglês | MEDLINE | ID: mdl-17081661

RESUMO

This study explores the utility of recombinant adeno-associated virus (rAAV) as a genetic vaccine delivery system using muscle as a target tissue. A single injection of rAAV encoding the malarial antigens MSP4 (Plasmodium falciparum) or MSP4/5 (Plasmodium yoelii) stimulated long-term antigen-specific antibody responses. Anti-MSP4/5 immunity stimulated by AAV was not protective against P. yoelii infection and efforts taken to augment antibody responses against MSP4/5, either by priming with plasmid DNA or AAV and boosting with rAAV were unsuccessful. Alternative strategies such as inclusion of genetic adjuvants into the AAV vector will be necessary to stimulate an adequate level of anti-malarial protective immunity in this model.


Assuntos
Antígenos de Protozoários/imunologia , Dependovirus/genética , Vacinas Antimaláricas/administração & dosagem , Malária/imunologia , Proteínas de Membrana/imunologia , Plasmodium falciparum/imunologia , Plasmodium yoelii/imunologia , Proteínas de Protozoários/imunologia , Adjuvantes Imunológicos/farmacologia , Animais , Formação de Anticorpos/imunologia , Antígenos de Protozoários/genética , Dependovirus/imunologia , Epitopos , Feminino , Vetores Genéticos/genética , Vetores Genéticos/imunologia , Células HeLa , Humanos , Injeções Intramusculares , Malária/parasitologia , Malária/prevenção & controle , Vacinas Antimaláricas/genética , Vacinas Antimaláricas/imunologia , Proteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos BALB C , Plasmodium falciparum/parasitologia , Plasmodium yoelii/parasitologia , Proteínas de Protozoários/genética
11.
Hum Gene Ther ; 16(8): 947-56, 2005 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16076253

RESUMO

Peripheral nervous system (PNS) sensory neurons are directly involved in the pathophysiology of a number of debilitating inherited and acquired neurological conditions. The lack of effective treatments for many such conditions provides a strong rationale for exploring novel therapeutic approaches, including gene therapy. Friedreich ataxia (FRDA), a sensory neuropathy, is a progressive neurodegenerative disease associated with a loss of large sensory neurons from the dorsal root ganglia. Because a mouse model for this well-characterized disease has been generated, we elected to use FRDA as a model disease. In previous studies we achieved efficient and sustained delivery of a reporter gene to PNS sensory neurons, using recombinant adeno-associated viral (AAV) and lentiviral (LV) vectors. In the current study, AAV and LV vectors encoding the human frataxin cDNA were constructed and assessed for frataxin expression and function in primary FRDA patient fibroblast cell lines. FRDA fibroblasts have been shown to exhibit subtle biochemical changes, including increased mitochondrial iron and sensitivity to oxidant stress. Despite the inherent difficulty in working with primary cells, transduction of patient fibroblasts with either vector resulted in the expression of appropriately localized frataxin and partial reversal of phenotype.


Assuntos
Ataxia de Friedreich/genética , Ataxia de Friedreich/terapia , Técnicas de Transferência de Genes , Terapia Genética/métodos , Proteínas de Ligação ao Ferro/genética , Estresse Oxidativo , Adenoviridae/genética , DNA Complementar/genética , Células Epiteliais/fisiologia , Fibroblastos , Ataxia de Friedreich/fisiopatologia , Vetores Genéticos , Células HeLa , Humanos , Técnicas In Vitro , Ferro/análise , Lentivirus/genética , Mitocôndrias/química , Fenótipo , Resultado do Tratamento , Frataxina
12.
Med J Aust ; 182(9): 458-63, 2005 May 02.
Artigo em Inglês | MEDLINE | ID: mdl-15865589

RESUMO

OBJECTIVE: To report the outcome of gene therapy in an infant with X-linked severe combined immunodeficiency (SCID-X1), which typically causes a lack of T and natural killer (NK) cells. DESIGN AND SETTING: Ex-vivo culture and gene transfer procedures were performed at The Children's Hospital at Westmead, Sydney, NSW, in March 2002. Follow-up to March 2005 (36 months) is available. PATIENT: A 9-month-old male infant with confirmed SCID-X1 (including complete absence of T cells) with an NK+ phenotype (a less common variant of SCID-X1), and no HLA-identical sibling donor available for conventional bone marrow transplantation. PROCEDURE: CD34+ haemopoietic progenitor cells were isolated from harvested bone marrow and cultured with cytokines to stimulate cellular replication. Cells were then genetically modified by exposure to a retrovirus vector encoding human gamma c (the common gamma chain of several interleukin receptors; mutations affecting the gamma c gene cause SCID-X1). Gene-modified cells (equivalent to 1.3 x 10(6) CD34+/gamma c+ cells/kg) were returned to the infant via a central line. RESULTS: T cells were observed in peripheral blood 75 days after treatment, and levels increased rapidly to 0.46 x 10(9) CD3+ cells/L at 5 months. Within 2 weeks of the appearance of T cells, there was a distinct clinical improvement, with early weight gain and clearance of rotavirus from the gut. However, T-cell levels did not reach the reference range, and immune reconstitution remained incomplete. The infant failed to thrive and developed weakness, hypertonia and hyperreflexia in the legs, possibly the result of immune dysregulation. He went on to receive a bone marrow transplant from a matched unrelated donor 26 months after gene therapy. CONCLUSIONS: This is the first occasion that gene therapy has been used to treat a genetic disease in Australia. Only partial immunological reconstitution was achieved, most likely because of the relatively low dose of gene-corrected CD34+ cells re-infused, although viral infection during the early phase of T-cell reconstitution and the infant's NK+ phenotype may also have exerted an effect.


Assuntos
Técnicas de Transferência de Genes , Terapia Genética/métodos , Imunodeficiência Combinada Severa/terapia , Antígenos CD34 , Vetores Genéticos , Células-Tronco Hematopoéticas/imunologia , Humanos , Lactente , Células Matadoras Naturais/imunologia , Contagem de Linfócitos , Masculino , Fenótipo , Imunodeficiência Combinada Severa/genética , Subpopulações de Linfócitos T , Resultado do Tratamento
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...