Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
1.
Drug Chem Toxicol ; 40(4): 406-409, 2017 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-28100091

RESUMO

CONTEXT: Polyethylenimine (PEI) is a cationic polymer commonly used in gene transfer. Although numerous investigations have indicated that PEI can induce apoptosis/necrosis but the mechanism of its cytotoxicity is still poorly understood. OBJECTIVE: The purpose of this study was to investigate the effects of PEI/DNA complexes on the expression of apoptotic genes in human colon adenocarcinoma cells (HT29). METHODS: HT29 cells were exposed to PEI/DNA complex (C/P = 0.8) for 24 h. Then, qRT PCR was used to assess the expression of 26 apoptotic-related genes. RESULT: Analysis of the transcript level of genes revealed that while the expression of anti-apoptotic genes such as Bclx, Bcl2, NFkB, and AIF was not significantly reduced but the expression of pro-apoptotic genes such as Fasl, Bax, TNFR1, DR4, Casp8, and cytochrome C was considerably increased in transfected HT29 cell lines. CONCLUSIONS: Our results showed that PEI could increase the level of pro-apoptotic genes and decrease antiapoptotic genes as a possible mechanism involved in PEI cytotoxicity.


Assuntos
Proteínas Reguladoras de Apoptose/metabolismo , DNA Recombinante/efeitos adversos , Regulação da Expressão Gênica/efeitos dos fármacos , Nanopartículas/efeitos adversos , Polietilenoimina/efeitos adversos , Apoptose/efeitos dos fármacos , Proteínas Reguladoras de Apoptose/genética , Linhagem Celular Tumoral , DNA Recombinante/química , Perfilação da Expressão Gênica , Técnicas de Transferência de Genes/efeitos adversos , Genes Reporter/efeitos dos fármacos , Humanos , Nanopartículas/química , Polietilenoimina/química , RNA Mensageiro/metabolismo
2.
Hum Gene Ther Clin Dev ; 27(1): 27-36, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-27003752

RESUMO

Applied Genetic Technologies Corporation (AGTC) is developing rAAV2tYF-PR1.7-hCNGB3, a recombinant adeno-associated virus (rAAV) vector expressing the human CNGB3 gene, for treatment of achromatopsia, an inherited retinal disorder characterized by markedly reduced visual acuity, extreme light sensitivity, and absence of color discrimination. We report here results of a study evaluating safety and biodistribution of rAAV2tYF-PR1.7-hCNGB3 in CNGB3-deficient mice. Three groups of animals (n = 35 males and 35 females per group) received a subretinal injection in one eye of 1 µl containing either vehicle or rAAV2tYF-PR1.7-hCNGB3 at one of two dose concentrations (1 × 10(12) or 4.2 × 10(12) vg/ml) and were euthanized 4 or 13 weeks later. There were no test-article-related changes in clinical observations, body weights, food consumption, ocular examinations, clinical pathology parameters, organ weights, or macroscopic observations at necropsy. Cone-mediated electroretinography (ERG) responses were detected after vector administration in the treated eyes in 90% of animals in the higher dose group and 31% of animals in the lower dose group. Rod-mediated ERG responses were reduced in the treated eye for all groups, with the greatest reduction in males given the higher dose of vector, but returned to normal by the end of the study. Microscopic pathology results demonstrated minimal mononuclear cell infiltrates in the retina and vitreous of some animals at the interim euthanasia and in the vitreous of some animals at the terminal euthanasia. Serum anti-AAV antibodies developed in most vector-injected animals. No animals developed antibodies to hCNGB3. Biodistribution studies demonstrated high levels of vector DNA in vector-injected eyes but little or no vector DNA in nonocular tissue. These results support the use of rAAV2tYF-PR1.7-hCNGB3 in clinical studies in patients with achromatopsia caused by CNGB3 mutations.


Assuntos
Defeitos da Visão Cromática/terapia , Canais de Cátion Regulados por Nucleotídeos Cíclicos/genética , DNA Recombinante/efeitos adversos , Dependovirus/genética , Terapia Genética , Vetores Genéticos/efeitos adversos , Animais , Defeitos da Visão Cromática/genética , Canais de Cátion Regulados por Nucleotídeos Cíclicos/deficiência , Canais de Cátion Regulados por Nucleotídeos Cíclicos/metabolismo , DNA Recombinante/administração & dosagem , Feminino , Vetores Genéticos/administração & dosagem , Humanos , Injeções Intraoculares , Masculino , Camundongos , Retina/metabolismo
3.
Hum Gene Ther Clin Dev ; 27(1): 37-48, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-27003753

RESUMO

Applied Genetic Technologies Corporation (AGTC) is developing rAAV2tYF-PR1.7-hCNGB3, a recombinant adeno-associated viral (rAAV) vector expressing the human CNGB3 gene, for treatment of achromatopsia, an inherited retinal disorder characterized by markedly reduced visual acuity, extreme light sensitivity, and absence of color discrimination. We report here results of a study evaluating the safety and biodistribution of rAAV2tYF-PR1.7-hCNGB3 in cynomolgus macaques. Three groups of animals (n = 2 males and 2 females per group) received a subretinal injection in one eye of 300 µl containing either vehicle or rAAV2tYF-PR1.7-hCNGB3 at one of two concentrations (4 × 10(11) or 4 × 10(12) vector genomes/ml) and were evaluated over a 3-month period before being euthanized. Administration of rAAV2tYF-PR1.7-hCNGB3 was associated with a dose-related anterior and posterior segment inflammatory response that was greater than that observed in eyes injected with the vehicle control. Most manifestations of inflammation improved over time except that vitreous cells persisted in vector-treated eyes until the end of the study. One animal in the lower vector dose group was euthanized on study day 5, based on a clinical diagnosis of endophthalmitis. There were no test article-related effects on intraocular pressure, visual evoked potential responses, hematology or clinical chemistry parameters, or gross necropsy observations. Histopathological examination demonstrated minimal mononuclear infiltrates in all vector-injected eyes. Serum anti-AAV antibodies developed in all vector-injected animals. No animals developed antibodies to CNGB3. Biodistribution studies demonstrated high levels of vector DNA in the injected eye but minimal or no vector DNA in any other tissue. These results support the use of rAAV2tYF-PR1.7-hCNGB3 in clinical studies in patients with achromatopsia caused by CNGB3 mutations.


Assuntos
Defeitos da Visão Cromática/terapia , Canais de Cátion Regulados por Nucleotídeos Cíclicos/genética , DNA Recombinante/efeitos adversos , Dependovirus/genética , Terapia Genética , Vetores Genéticos/efeitos adversos , Animais , Canais de Cátion Regulados por Nucleotídeos Cíclicos/metabolismo , DNA Recombinante/administração & dosagem , Feminino , Vetores Genéticos/administração & dosagem , Humanos , Injeções Intraoculares , Macaca fascicularis , Masculino
4.
Carbohydr Polym ; 136: 791-802, 2016 Jan 20.
Artigo em Inglês | MEDLINE | ID: mdl-26572414

RESUMO

Polyethylenimine (PEI) has been used as a vehicle to deliver genes to cancer cells and somatic cells. In this study, cationic polymers of PEI were shielded with anionic polymers of hyaluronic acid (HA) to safely and effectively deliver genes into human mesenchymal stem cells (hMSCs). HA interacted with CD44 in the plasma membranes of hMSCs to facilitate the internalization of HA-shielded PEI/pDNA complexes. The HA-shielded PEI/pDNA nanogels were confirmed by size changes, ζ-potential, and gel retardation assays. HA-shielded nanogels were easily internalized by hMSCs, and this was reduced by pretreatment with a specific monoclonal antibody that blocked CD44. By shielding PEI/pDNA complexes with HA, nanogels were easily internalized to hMSCs when it did not blocked by anti-CD44. These shielded nanogels were also easily internalized by HeLa cells, and this was reduced by pretreatment with an anti-CD44 monoclonal antibody. Following internalization of the SOX9 gene, chondrogenesis of hMSCs was increased, as determined by RT-PCR, real-time quantitative PCR, and histological analyses.


Assuntos
Géis/química , Terapia Genética/métodos , Receptores de Hialuronatos/metabolismo , Ácido Hialurônico/farmacologia , Células-Tronco Mesenquimais/metabolismo , Polietilenoimina/química , Diferenciação Celular , Células Cultivadas , DNA Recombinante/administração & dosagem , DNA Recombinante/efeitos adversos , Géis/farmacologia , Células HeLa , Humanos , Ácido Hialurônico/química , Masculino , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/efeitos dos fármacos , Plasmídeos/administração & dosagem , Plasmídeos/efeitos adversos , Ligação Proteica , Fatores de Transcrição SOX9/genética , Fatores de Transcrição SOX9/metabolismo , Adulto Jovem
5.
Hum Gene Ther Clin Dev ; 25(4): 202-11, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25211193

RESUMO

X-linked retinoschisis (XLRS) is a retinal disease caused by mutations in the gene encoding the protein retinoschisin (RS1) and one of the most common causes of macular degeneration in young men. Currently, no FDA-approved treatments are available for XLRS and a replacement gene therapy could provide a promising strategy. We have developed a novel gene therapy approach for XLRS, based on the administration of AAV8-scRS/IRBPhRS, an adeno-associated viral vector coding the human RS1 protein, via the intravitreal route. On the basis of our prior study in an Rs1-KO mouse, this construct transduces efficiently all the retinal layers, resulting in an RS1 expression similar to that observed in the wild-type and improving retinal structure and function. In support of a clinical trial, we carried out a study to evaluate the ocular safety of intravitreal administration of AAV8-scRS/IRBPhRS into 39 New Zealand White rabbits. Two dose levels of vector, 2e(10) and 2e(11) vector genomes per eye (vg/eye), were tested and ocular inflammation was monitored over a 12-week period by serial ophthalmological and histopathological analysis. A mild ocular inflammatory reaction, consisting mainly of vitreous infiltrates, was observed within 4 weeks from injection, in both 2e(10) and 2e(11) vg/eye groups and was likely driven by the AAV8 capsid. At 12-week follow-up, ophthalmological examination revealed no clinical signs of vitreitis in either of the dose groups. However, while vitreous inflammatory infiltrate was significantly reduced in the 2e(10) vg/eye group at 12 weeks, some rabbits in the higher dose group still showed persistence of inflammatory cells, histologically. In conclusion, intravitreal administration of AAV8-scRS/IRBPhRS into the rabbit eye produces a mild and transient intraocular inflammation that resolves, at a 2e(10) vg/eye dose, within 3 months, and does not cause irreversible tissue damages. These data support the initiation of a clinical trial of intravitreal administration of AAV8-scRS/IRBPhRS in XLRS patients.


Assuntos
DNA Recombinante/efeitos adversos , Dependovirus/genética , Proteínas do Olho/genética , Terapia Genética , Vetores Genéticos/efeitos adversos , Retinosquise/terapia , Animais , DNA Recombinante/administração & dosagem , DNA Recombinante/genética , Dependovirus/metabolismo , Proteínas do Olho/metabolismo , Vetores Genéticos/administração & dosagem , Vetores Genéticos/genética , Injeções Intravítreas , Coelhos
7.
Curr Gene Ther ; 13(6): 434-52, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24195602

RESUMO

It is hoped that the use of gene transfer technology to treat both monogenetic and acquired diseases may soon become a common therapy option in medicine. For gene therapy to achieve this objective, any gene delivery method will have to meet several criteria, including ease of manufacturing, efficient gene transfer to target tissue, long-term gene expression to alleviate the disease, and most importantly safety in patients. Viral vectors are an attractive choice for use in gene therapy protocols due to their relative efficiency in gene delivery. Since there is inherent risk in using viruses, investigators in the gene therapy community have devoted extensive efforts toward reengineering viral vectors for enhance safety. Here we review the approaches and technologies that are being evaluated for the use of recombinant vectors based upon adeno-associated virus (AAV) in the treatment of a variety of human diseases. AAV is currently the only known human DNA virus that is non-pathogenic and AAV-based vectors are classified as Risk Group 1 agents for all laboratory and animal studies carried out in the US. Although its apparent safety in natural infection and animals appears well documented, we examine the accumulated knowledge on the biology and vectorology of AAV, lessons learned from gene therapy clinical trials, and how this information is impacting current vector design and manufacturing with an overall emphasis on biosafety.


Assuntos
Dependovirus/genética , Terapia Genética/efeitos adversos , Vetores Genéticos/efeitos adversos , Animais , Ensaios Clínicos Fase I como Assunto , Ensaios Clínicos Fase II como Assunto , DNA Recombinante/administração & dosagem , DNA Recombinante/efeitos adversos , DNA Recombinante/genética , Técnicas de Transferência de Genes/efeitos adversos , Terapia Genética/métodos , Vetores Genéticos/genética , Substâncias Perigosas/efeitos adversos , Humanos
8.
Hum Reprod ; 28(9): 2398-406, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23820419

RESUMO

STUDY QUESTION: Is targeted adenovirus vector, Ad-SSTR-RGD-TK (Adenovirus -human somatostatin receptor subtype 2- arginine, glycine and aspartate-thymidine kinase), given in combination with ganciclovir (GCV) against immortalized human leiomyoma cells (HuLM) a potential therapy for uterine fibroids? SUMMARY ANSWER: Ad-SSTR-RGD-TK/GCV, a targeted adenovirus, effectively reduces cell growth in HuLM cells and to a significantly greater extent than in human uterine smooth muscle cells (UtSM). WHAT IS KNOWN ALREADY: Uterine fibroids (leiomyomas), a major cause of morbidity and the most common indication for hysterectomy in premenopausal women, are well-defined tumors, making gene therapy a suitable and potentially effective non-surgical approach for treatment. Transduction of uterine fibroid cells with adenoviral vectors such as Ad-TK/GCV (herpes simplex virus thymidine kinase gene) decreases cell proliferation. STUDY DESIGN, SIZE, DURATION: An in vitro cell culture method was set up to compare and test the efficacy of a modified adenovirus vector with different multiplicities of infection in two human immortalized cell lines for 5 days. PARTICIPANTS/MATERIALS, SETTING, METHODS: Immortalized human leiomyoma cells and human uterine smooth muscle cells were infected with different multiplicities of infection (MOI) (5-100 plaque-forming units (pfu)/cell) of a modified Ad-SSTR-RGD-TK vector and subsequently treated with GCV. For comparison, HuLM and UtSM cells were transfected with Ad-TK/GCV and Ad-LacZ/GCV. Cell proliferation was measured using the CyQuant assay in both cell types. Additionally, western blotting was used to assess the expression of proteins responsible for regulating proliferation and apoptosis in the cells. MAIN RESULTS AND THE ROLE OF CHANCE: Transduction of HuLM cells with Ad-SSTR-RGD-TK/GCV at 5, 10, 50 and 100 pfu/cell decreased cell proliferation by 28, 33, 45, and 84%, respectively (P < 0.05) compared with untransfected cells, whereas cell proliferation in UtSM cells transfected with the same four MOIs of Ad-SSTR-RGD-TK/GCV compared with that of untransfected cells was decreased only by 8, 23, 25, and 28%, respectively (P < 0.01). Western blot analysis showed that, in comparison with the untargeted vector Ad-TK, Ad-SSTR-RGD-TK/GCV more effectively reduced expression of proteins that regulate the cell cycle (Cyclin D1) and proliferation (PCNA, Proliferating Cell Nuclear Antigen), and it induced expression of the apoptotic protein BAX, in HuLM cells. LIMITATIONS, REASONS FOR CAUTION: Results from this study need to be replicated in an appropriate animal model before testing this adenoviral vector in a human trial. WIDER IMPLICATIONS OF THE FINDINGS: Effective targeting of gene therapy to leiomyoma cells enhances its potential as a non-invasive treatment of uterine fibroids.


Assuntos
DNA Recombinante/metabolismo , Regulação Neoplásica da Expressão Gênica , Vetores Genéticos/metabolismo , Leiomioma/metabolismo , Miométrio/metabolismo , Transdução Genética , Neoplasias Uterinas/metabolismo , Adenoviridae/genética , Adenoviridae/metabolismo , Adenoviridae/patogenicidade , Apoptose , Proteínas Reguladoras de Apoptose/genética , Proteínas Reguladoras de Apoptose/metabolismo , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Linhagem Celular Transformada , Linhagem Celular Tumoral , Proliferação de Células , DNA Recombinante/efeitos adversos , DNA Recombinante/uso terapêutico , Feminino , Terapia Genética/efeitos adversos , Terapia Genética/métodos , Vetores Genéticos/efeitos adversos , Vetores Genéticos/uso terapêutico , Humanos , Leiomioma/terapia , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Neoplasias Uterinas/terapia
9.
Immunotherapy ; 3(1): 17-21, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21174553

RESUMO

In the USA, the incidence of cutaneous melanoma is increasing rapidly. It has been shown to be responsive to immune-stimulating drugs. Allovectin-7 allows the immune system to recognize metastatic melanoma lesions as foreign by incorporating a MHC class I complex into the tumor through direct injection. Once transfected, tumor biopsies have shown an increased presence of cytotoxic T lymphocytes in the tumor beds themselves. Phase I and II trials have shown local and systemic response to these tumors, with an excellent safety profile. Currently, a Phase III trial has completed enrollment and is set to determine the safety and efficacy of treatment in comparison with standard chemotherapy.


Assuntos
DNA Recombinante/uso terapêutico , Antígeno HLA-B7/imunologia , Lipídeos/uso terapêutico , Melanoma/terapia , Linfócitos T Citotóxicos/imunologia , Ensaios Clínicos como Assunto , DNA Recombinante/administração & dosagem , DNA Recombinante/efeitos adversos , Terapia Genética/métodos , Antígeno HLA-B7/genética , Humanos , Imunoterapia/métodos , Lipídeos/administração & dosagem , Lipídeos/efeitos adversos , Lipossomos , Melanoma/imunologia , Metástase Neoplásica/imunologia , Metástase Neoplásica/terapia , Plasmídeos , Neoplasias Cutâneas/terapia , Resultado do Tratamento
10.
Expert Opin Biol Ther ; 10(5): 841-51, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20367461

RESUMO

IMPORTANCE OF THE FIELD: Immunotherapy for cancer has been investigated for several decades, achieving limited success. The development of effective new immunotherapeutic agents has reignited interest in the filed. Intralesional injection of plasmids in order to transfect genes capable of stimulating or augmenting immune recognition and destruction of tumors is a relatively new approach. AREAS COVERED IN THIS REVIEW: Our objective is to discuss the role velimogene aliplasmid (Allovectin-7, Vical Incorporated), a plasmid-lipid complex containing the DNA sequences encoding HLA-B7 and beta2 microglobulin, as an immunotherapeutic agent. WHAT THE READER WILL GAIN: Intralesional velimogene aliplasmid induces anti-tumor responses in a proportion of melanoma patients with locoregional and limited distant metastases. Preclinical data and the results of Phase I, II and III clinical trials with this drug are reviewed. The limited data in other malignancies is also reviewed. Velimogene aliplasmid in humans appears safe, with minimal drug-related adverse events. TAKE HOME MESSAGE: Velimogene aliplasmid has activity in melanoma with local and limited distant disease associated with an excellent safety profile. The activity of this approach is also being investigated in other malignancies.


Assuntos
Vacinas Anticâncer/uso terapêutico , DNA Recombinante/uso terapêutico , Terapia Genética/métodos , Imunoterapia/métodos , Lipídeos/uso terapêutico , Melanoma/terapia , Animais , Vacinas Anticâncer/efeitos adversos , DNA Recombinante/efeitos adversos , Medicina Baseada em Evidências , Terapia Genética/efeitos adversos , Antígeno HLA-B7/genética , Antígeno HLA-B7/imunologia , Humanos , Imunoterapia/efeitos adversos , Lipídeos/efeitos adversos , Melanoma/imunologia , Melanoma/secundário , Resultado do Tratamento , Microglobulina beta-2/genética , Microglobulina beta-2/imunologia
11.
Expert Opin Biol Ther ; 8(6): 839-44, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18476795

RESUMO

BACKGROUND: Patients with metastatic melanoma are immunosuppressed by the growing tumor. Allovectin-7 therapy is a form of active immunotherapy that aims at immunization of the host with substances designed to elicit an immune reaction that will eliminate or slow down the growth and spread of the cancer. OBJECTIVE: to describe the rationale for immunotherapy with Allovectin-7 and assess its safety profile and efficacy based on the results of completed melanoma clinical trials. METHODS: we reviewed both the published medical literature and the results of trials pending publication. RESULTS/CONCLUSION: Allovectin-7 is a safe and active immunotherapeutic agent. It induces local and systemic durable responses in patients with metastatic melanoma.


Assuntos
Vacinas Anticâncer/uso terapêutico , DNA Recombinante/uso terapêutico , Vetores Genéticos/uso terapêutico , Antígeno HLA-B7/genética , Imunoterapia Ativa , Lipídeos/uso terapêutico , Melanoma/secundário , Neoplasias Cutâneas/secundário , Vacinas de DNA/uso terapêutico , Microglobulina beta-2/genética , Animais , Vacinas Anticâncer/administração & dosagem , Vacinas Anticâncer/efeitos adversos , Ensaios Clínicos como Assunto/estatística & dados numéricos , DNA Recombinante/administração & dosagem , DNA Recombinante/efeitos adversos , Ensaios de Seleção de Medicamentos Antitumorais , Vetores Genéticos/imunologia , Antígeno HLA-B7/imunologia , Humanos , Imunoterapia Ativa/efeitos adversos , Injeções Intralesionais , Lipídeos/administração & dosagem , Lipídeos/efeitos adversos , Neoplasias Pulmonares/imunologia , Neoplasias Pulmonares/secundário , Neoplasias Pulmonares/terapia , Macaca fascicularis , Melanoma/imunologia , Melanoma/terapia , Camundongos , Fosfatidiletanolaminas/administração & dosagem , Compostos de Amônio Quaternário/administração & dosagem , Neoplasias Cutâneas/imunologia , Neoplasias Cutâneas/terapia , Evasão Tumoral , Vacinas de DNA/administração & dosagem , Vacinas de DNA/efeitos adversos , Microglobulina beta-2/imunologia
12.
Med Hypotheses ; 64(6): 1105-8, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-15823695

RESUMO

Over the past decade, there has been growing interest in the use of antibodies against intracellular targets. This is currently achieved through recombinant expression of the single chain variable fragment (scFv) antibody format within the cell, which is commonly referred to as an intrabody. This possesses a number of inherent advantages over RNA interference (iRNA). Firstly, the high specificity and affinity of intrabodies to target antigens is well-established, whereas iRNA has been frequently shown to exert multiple non-specific effects. Secondly, intrabodies being proteins possess a much longer active half-life compared to iRNA. Thirdly, when the active half-life of the intracellular target molecule is long, gene silencing through iRNA would be slow to yield any effect, whereas the effects of intrabody expression would be almost instantaneous. Lastly, it is possible to design intrabodies to block certain binding interactions of a particular target molecule, while sparing others. There is, however, various technical challenges faced with intrabody expression through the application of recombinant DNA technology. In particular, protein conformational folding and structural stability of the newly-synthesized intrabody within the cell is affected by reducing conditions of the intracellular environment. Also, there are overwhelming safety concerns surrounding the application of transfected recombinant DNA in human clinical therapy, which is required to achieve intrabody expression within the cell. Of particular concern are the various viral-based vectors that are commonly-used in genetic manipulation. A novel approach around these problems would be to look at the possibility of fusing protein transduction domains (PTD) to scFv antibodies, to create a 'cell-permeable' antibody or 'Transbody'. PTD are short peptide sequences that enable proteins to translocate across the cell membrane and be internalized within the cytosol, through atypical secretory and internalization pathways. There are a number of distinct advantages that a 'Transbody' would possess over conventional intrabodies expressed within the cell. For a start, 'correct' conformational folding and disulfide bond formation can take place prior to introduction into the target cell. More importantly, the use of cell-permeable antibodies or 'Transbodies' would avoid the overwhelming safety and ethical concerns surrounding the direct application of recombinant DNA technology in human clinical therapy, which is required for intrabody expression within the cell. 'Transbodies' introduced into the cell would possess only a limited active half-life, without resulting in any permanent genetic alteration. This would allay any safety concerns with regards to their application in human clinical therapy.


Assuntos
Anticorpos Monoclonais/genética , Genes de Imunoglobulinas , Genes Sintéticos , Fragmentos de Imunoglobulinas/genética , Região Variável de Imunoglobulina/genética , Estrutura Terciária de Proteína , Proteínas Recombinantes de Fusão/genética , Animais , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/farmacocinética , Permeabilidade da Membrana Celular , DNA Recombinante/administração & dosagem , DNA Recombinante/efeitos adversos , DNA Recombinante/genética , DNA Recombinante/uso terapêutico , Sistemas de Liberação de Medicamentos , Desenho de Fármacos , Humanos , Fragmentos de Imunoglobulinas/imunologia , Região Variável de Imunoglobulina/imunologia , Conformação Proteica , Dobramento de Proteína , Proteínas Recombinantes de Fusão/imunologia , Transfecção
13.
Diabet Med ; 22(1): 102-6, 2005 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-15606700

RESUMO

AIMS: Allergy to recombinant human (rDNA) insulin preparations is a rare complication of insulin therapy. However, insulin preparations contain several allergens, and several disorders can resemble insulin allergy. Studies evaluating the diagnostic procedures on suspected insulin allergy are extremely few. METHODS: Since January 1998, we have used a standardized investigative procedure during admittance to the medical ward allowing observation and repeated recording of reactions to intradermal skin test (performed with a commercially available kit containing isolated insulin allergens). Data on all investigated cases until April 2003 were collected retrospectively, and self-reported efficacy of intervention was compared to clinical data. RESULTS: Twenty-two patients were included. In nine (41%) cases, non-insulin allergic causes were discovered and successfully treated: poor injection technique (n = 5), skin disease (n = 3) and other systemic allergy (n = 1). Nine other patients were found to be allergic to protamine (n = 3) or rDNA insulin (n = 6), and specific treatment was associated with relief in 8 patients (89%). Four patients had local reactions of unknown causes but symptom relief was obtained in three cases by unspecific therapy. Overall, 20 (91%) reported relief of symptoms. CONCLUSION: Our standardized investigative procedure of suspected insulin preparation (IP) allergy was associated with relief of symptoms in > 90% of patients. IP allergy was diagnosed in 41%, and intradermal testing with isolated insulin allergens was a prerequisite in identification of culprit allergen and targeting of treatment.


Assuntos
DNA Recombinante/efeitos adversos , Hipersensibilidade a Drogas/diagnóstico , Hipoglicemiantes/efeitos adversos , Insulina/efeitos adversos , Adulto , Idoso , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Prognóstico , Estudos Retrospectivos , Testes Cutâneos
14.
Trends Pharmacol Sci ; 23(6): 254-6, 2002 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-12084625

RESUMO

Many biopharmaceuticals have been reported to induce the production of antibodies. Several factors influence immunogenicity, including the methods of production, formulation and storage as well as patient characteristics (e.g. type of disease and genetic background). Because immunogenicity is unpredictable, testing for antibodies should always be part of the evaluation of new biopharmaceuticals, although the biological consequences of these antibodies are rarely severe.


Assuntos
Formação de Anticorpos/imunologia , DNA Recombinante/imunologia , Animais , Biofarmácia , DNA Recombinante/efeitos adversos , DNA Recombinante/uso terapêutico , Humanos , Modelos Animais
19.
Hum Gene Ther ; 9(2): 249-69, 1998 Jan 20.
Artigo em Inglês | MEDLINE | ID: mdl-9472784

RESUMO

UNLABELLED: GTAB1001: A Double-Blind, Placebo Controlled, Dose Ranging Study to Evaluate the Safety and Biological Efficacy of the Lipid-DNA Complex GR213487B in the Nasal Epithelium of Adult Patients with Cystic Fibrosis. OBJECTIVES: To evaluate the effectiveness of various dosages of the lipid-DNA complex GR213487B (0.4375mg and either 4.0mg or 0.0625mg) for producing CFTR gene transfer and correcting the chloride ion transport defect in the nasal epithelium of patients with cystic fibrosis. To assess the safety and tolerability of the lipid-DNA complex GR213487B when applied to the nasal epithelium of patients with cystic fibrosis. DESIGN: Single-center, double-blind, placebo controlled, dose ranging study. DURATION: Pre-treatment evaluations will be performed during two outpatient study visits (ie. between Day -7 to -3 and at Day -2). Patients will be admitted to the Clinical Research Unit (CRU) at the University of North Carolina at Chapel Hill on Day -1 for additional pre-treatment evaluations performed the day prior to administration of double-blind treatment (ie. gene transfer) on Treatment Day 0. Patients will remain in the CRU for 7 days (Day -1 to Day 6) and will be discharged on Day 6. Patients will subsequently be followed on an outpatient basis but will return for another assessment between Days 9-11, and may also return to the CRU for two optional study visits on Days 14 and 21. All patients will return to the CRU on an out-patient basis for follow-up evaluations on Day 28 +/- 3. SETTING: Patients will receive in-patient treatment in the CRU at the University of North Carolina at Chapel Hill and will remain in the CRU for 7 days. PATIENTS: A target enrollment of 12 evaluable patients is planned. STUDY TREATMENTS: Patients who meet all entry criteria will complete pre-treatment assessments, which will take place between Day -7 to Day -1, and will serve as a baseline for specific evaluations and to ensure clinical stability. Patients will return on Day -1 for admission to the CRU the day prior to gene transfer. Each nostril of the patients will be randomly assigned in a double blind manner to receive either GR213487B liquid nasal spray or the lipid alone (ie. control administered as liposome), by topical application directed at the inferior turbinate. The first four patients will receive an initial dosage of GR213487B containing 0.4375 mg of DNA. The decision to proceed to administer a higher dose (ie. 4.0mg DNA) or a lower dose (ie. 0.0625mg DNA) in the subsequent eight patients will be determined by the Principal Investigator in association with an FDA officer serving as an independent Clinical Ombudsman, according to the study plan (see Section 5.5 and Appendix 3-Dosing Flow Chart). MEASUREMENTS: Efficacy Evaluations The primary variables to determine the efficacy of transgene expression will be: * Evidence of vector derived CFTR (cystic fibrosis transmembrane conductance regulator) mRNA, as measured by reverse transcriptase polymerase chain reaction (RT-PCR) in nasal epithelial cells obtained from nasal scrapes on Day 3 and, nasal biopsies on Day 5, if sufficient tissue is available. * Correction of chloride ion transport across the nasal epithelium as measured by the transepithelial electrical potential difference (TEPD). The baseline TEPD will initially be measured, and again subsequently following perfusion of: --zero chloride perfusion containing amiloride (to induce chloride secretion) --zero chloride perfusion containing amiloride and isoproterenol (to increase cAMP-mediated chloride secretion) Secondary measures to determine the efficacy of gene transfer will be: * Evidence of delivery of plasmid DNA in the nasal lavage (Day 1-5, Day 9-11 and Day 28) * Evidence of vector derived CFTR mRNA from nasal scrapes performed after the nasal biopsy (ie. Day 9-11 and/or Day 28) * Percentage of cells from nasal biopsies expressing vector derived CFTR mRNA as measured by in situ hybridization * Evidence of vector derived CFTR


Assuntos
Regulador de Condutância Transmembrana em Fibrose Cística/administração & dosagem , Regulador de Condutância Transmembrana em Fibrose Cística/genética , Fibrose Cística/tratamento farmacológico , DNA Recombinante/administração & dosagem , Técnicas de Transferência de Genes , Lipossomos/administração & dosagem , Mucosa Nasal/metabolismo , Adulto , Animais , Protocolos Clínicos , Fibrose Cística/genética , Regulador de Condutância Transmembrana em Fibrose Cística/efeitos adversos , DNA Recombinante/efeitos adversos , Método Duplo-Cego , Esquema de Medicação , Epitélio/metabolismo , Feminino , Terapia Genética/métodos , Humanos , Lipossomos/efeitos adversos , Masculino , Seleção de Pacientes , Projetos Piloto , Ratos
20.
Hypertens Res ; 20(4): 269-77, 1997 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-9453262

RESUMO

Adrenomedullin (ADM) is a potent vasoactive peptide. In this study, we explored the effects of a continuous supply of ADM by somatic gene delivery on spontaneously hypertensive rats (SHR). DNA constructs containing the human ADM cDNA fused to either the cytomegalovirus promoter (CMV-cADM) or Rous Sarcoma virus 3'-long terminal repeat (RSV-cADM) were intravenously injected into SHR through the tail vein. Expression of human ADM in SHR was identified in the kidney, adrenal gland, heart, and lung by radioimmunoassay and reverse transcription-polymerase chain reaction followed by Southern blot analysis. A single injection of ADM plasmid DNA in young adult SHR (7 wk old) caused a significant reduction in systolic blood pressure for up to 5 wk (p < 0.05). A second injection of CMV-cADM 5 wk after the first delivery resulted in a further reduction in blood pressure for another 3 wk (p < 0.001). A maximal blood pressure reduction of 22 mmHg in SHR was observed 7 wk after injection of CMV-cADM plasmid DNA (185 +/- 1.7 mmHg, n = 6, p < 0.001), and a reduction of 15 mmHg was observed after injection of RSV-cADM (192 +/- 2.7 mmHg, n = 6, p < 0.001), as compared with control rats given vector DNA (207 +/- 2.4 mmHg, n = 6). Similarly, injection of CMV-cADM plasmid DNA in adult SHR (10 wk old) resulted in a significant reduction in blood pressure for up to 6 wk. Antibodies to either human ADM or its plasmid DNA were not detected in rat sera after the second injection. These studies indicate that intravenous injection of the human ADM gene in hypertensive rats results in expression of the foreign gene and induces a long-lasting reduction in blood pressure.


Assuntos
Pressão Sanguínea/efeitos dos fármacos , Terapia Genética , Hipertensão/terapia , Peptídeos/genética , Adrenomedulina , Animais , Formação de Anticorpos , Vírus do Sarcoma Aviário/genética , Pressão Sanguínea/genética , Citomegalovirus/genética , DNA Complementar/genética , DNA Recombinante/efeitos adversos , DNA Recombinante/genética , DNA Recombinante/uso terapêutico , Expressão Gênica , Humanos , Hipotensão/etiologia , Injeções Intravenosas , Rim/citologia , Rim/embriologia , Rim/enzimologia , Pulmão/enzimologia , RNA Mensageiro/análise , RNA Mensageiro/genética , Ratos , Ratos Endogâmicos SHR , Baço/enzimologia , Distribuição Tecidual , Transfecção , beta-Galactosidase/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...