Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Mol Pain ; 13: 1744806917731696, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28929859

RESUMO

In the absence of infection, the pathophysiology of endotracheal tube-induced sore throat pain is unclear. Activated neutrophils release elastase, reactive oxygen species, and inflammatory cytokines known to contribute to neuropathic pain. Sterile tissue injury can cause the release of damage-associated molecular patterns such as mitochondrial DNA that promote neutrophil activation. We hypothesized that endotracheal tube-induced sore throat pain is linked to mitochondrial DNA-mediated neutrophil inflammation. A nonrandomized prospective survey for sore throat pain was conducted in 31 patients who required short-term intubation and had no evidence of upper airway infection. Patterns of neutrophil abundance, activation, and mitochondrial DNA levels were analyzed in tracheal lavage fluid following intubation and prior to extubation. Thirteen of 31 patients reported sore throat pain. Sore throat patients had high neutrophilia with elevated adhesion molecule and TLR9 expression and constitutive reactive oxygen species generation. Tracheal lavage fluid from sore throat patients accumulated mitochondrial DNA and stimulated neutrophils to release mediators associated with pain in a TLR9- and DNAse-dependent fashion. Endotracheal tube-induced sore throat is linked to the release of mitochondrial DNA and can drive TLR9-mediated inflammatory responses by neutrophils reported to cause pain. Mitigating the effects of cell-free mitochondrial DNA may prove beneficial for the prevention of endotracheal tube-mediated sore throat pain.


Assuntos
DNA Mitocondrial/metabolismo , Inflamação/etiologia , Intubação Intratraqueal/efeitos adversos , Dor/etiologia , Faringite/etiologia , Adulto , Citocinas/metabolismo , Demografia , Feminino , Células HEK293 , Humanos , Masculino , Pessoa de Meia-Idade , Neutrófilos/metabolismo , Transdução de Sinais , Receptor Toll-Like 9/metabolismo , Adulto Jovem
2.
Hum Gene Ther ; 25(2): 121-31, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24195644

RESUMO

Several cancer vaccine efforts have been directed to simultaneously cotarget multiple tumor antigens, with the intent to achieve broader immune responses and more effective control of cancer growth. Genetic cancer vaccines based on in vivo muscle electro-gene-transfer of plasmid DNA (DNA-EGT) and adenoviral vectors represent promising modalities to elicit powerful immune responses against tumor-associated antigens (TAAs) such as carcinoembryonic antigen (CEA) and human epidermal growth factor receptor-2 (HER2)/neu. Combinations of these modalities of immunization (heterologous prime-boost) can induce superior immune reactions as compared with single-modality vaccines. We have generated a dual component-dual target genetic cancer vaccine consisting of a DNA moiety containing equal amounts of two plasmids, one encoding the extracellular and transmembrane domains of HER2 (ECD.TM) and the other encoding CEA fused to the B subunit of Escherichia coli heat-labile toxin (LTB), and of an adenoviral subtype 6 dicistronic vector carrying the same two tumor antigens gene constructs. The CEA/HER2 vaccine was tested in two different CEA/HER2 double-transgenic mouse models and in NOD/scid-DR1 mice engrafted with the human immune system. The immune response was measured by enzyme-linked immunospot assay, flow cytometry, and ELISA. The CEA/HER2 vaccine was able to break immune tolerance against both antigens. Induction of a T cell and antibody immune response was detected in immune-tolerant mice. Most importantly, the vaccine was able to slow the growth of HER2/neu⁺ and CEA⁺ tumors. A significant T cell response was measured in NOD/scid-DR1 mice engrafted with human cord blood cells. In conclusion, the CEA/HER2 genetic vaccine was immunogenic and able to confer significant therapeutic effects. These data warrant the evaluation of this vaccination strategy in human clinical trials.


Assuntos
Vacinas Anticâncer/imunologia , Antígeno Carcinoembrionário/imunologia , Receptor ErbB-2/imunologia , Adenoviridae/genética , Animais , Vacinas Anticâncer/genética , Antígeno Carcinoembrionário/genética , Modelos Animais de Doenças , Feminino , Ordem dos Genes , Técnicas de Transferência de Genes , Vetores Genéticos/administração & dosagem , Vetores Genéticos/genética , Humanos , Masculino , Camundongos , Camundongos Transgênicos , Neoplasias/genética , Neoplasias/imunologia , Neoplasias/mortalidade , Neoplasias/terapia , Ratos , Receptor ErbB-2/genética , Vacinas de DNA/genética , Vacinas de DNA/imunologia
3.
Hum Gene Ther ; 24(8): 728-38, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23902422

RESUMO

Client-owned pet dogs represent exceptional translational models for advancement of cancer research because they reflect the complex heterogeneity observed in human cancer. We have recently shown that a genetic vaccine targeting dog telomerase reverse transcriptase (dTERT) and based on adenovirus DNA electro-gene-transfer (Ad/DNA-EGT) technology can induce strong cell-mediated immune responses against this tumor antigen and increase overall survival of dogs affected by B-cell lymphosarcoma (LSA) in comparison with historical controls when combined with a cyclophosphamide, vincristine, and prednisone (COP) chemotherapy regimen. Here, we have conducted a double-arm clinical trial with an extended number of LSA patients, measured the antigen-specific immune response, and evaluated potential toxic effects of the immunotherapy along with a follow-up of patients survival for 3.5 years. The immune response was measured by enzyme-linked immunospot assay. The expression of dTERT was quantified by quantitative polymerase chain reaction. Changes in hematological parameters, local/systemic toxicity or organic dysfunction and fever were monitored over time during the treatment. dTERT-specific cell-mediated immune responses were induced in almost all treated animals. No adverse effects were observed in any dog patient that underwent treatment. The overall survival time of vaccine/COP-treated dogs was significantly increased over the COP-only cohort (>76.1 vs. 29.3 weeks, respectively, p<0.0001). There was a significant association between dTERT expression levels in LSA cells and overall survival among vaccinated patients. In conclusion, Ad/DNA-EGT-based cancer vaccine against dTERT in combination with COP chemotherapy is safe and significantly prolongs the survival of LSA canine patients. These data confirm the therapeutic efficacy of dTERT vaccine and support the evaluation of this approach for other cancer types as well as the translation of this approach to human clinical trials.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/normas , Vacinas Anticâncer/genética , Vacinas Anticâncer/uso terapêutico , Marcação de Genes , Linfoma de Células B/terapia , Telomerase/genética , Telomerase/metabolismo , Animais , Cães , Feminino , Imunoterapia , Linfoma de Células B/mortalidade , Masculino , Reação em Cadeia da Polimerase em Tempo Real , Análise de Sobrevida
4.
Oncoimmunology ; 1(8): 1258-1270, 2012 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-23243589

RESUMO

Functional T-cell epitope discovery is a key process for the development of novel immunotherapies, particularly for cancer immunology. In silico epitope prediction is a common strategy to try to achieve this objective. However, this approach suffers from a significant rate of false-negative results and epitope ranking lists that often are not validated by practical experience. A high-throughput platform for the identification and prioritization of potential T-cell epitopes is the iTopia(TM) Epitope Discovery System(TM), which allows measuring binding and stability of selected peptides to MHC Class I molecules. So far, the value of iTopia combined with in silico epitope prediction has not been investigated systematically. In this study, we have developed a novel in silico selection strategy based on three criteria: (1) predicted binding to one out of five common MHC Class I alleles; (2) uniqueness to the antigen of interest; and (3) increased likelihood of natural processing. We predicted in silico and characterized by iTopia 225 candidate T-cell epitopes and fixed-anchor analogs from three human tumor-associated antigens: CEA, HER2 and TERT. HLA-A2-restricted fragments were further screened for their ability to induce cell-mediated responses in HLA-A2 transgenic mice. The iTopia binding assay was only marginally informative while the stability assay proved to be a valuable experimental screening method complementary to in silico prediction. Thirteen novel T-cell epitopes and analogs were characterized and additional potential epitopes identified, providing the basis for novel anticancer immunotherapies. In conclusion, we show that combination of in silico prediction and an iTopia-based assay may be an accurate and efficient method for MHC Class I epitope discovery among tumor-associated antigens.

5.
Mol Ther ; 18(8): 1559-67, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20531395

RESUMO

Canine cancers occur with an incidence similar to that of humans and share many features with human malignancies including histological appearance, tumor genetics, biological behavior, and response to conventional therapies. As observed in humans, the telomerase reverse transcriptase (TERT) activity is largely confined to tumor tissues and absent in the majority of normal dog tissues. Therefore, dog TERT (dTERT) can constitute a valid target for translational cancer immunotherapy. We have evaluated the ability of adenovirus serotype 6 (Ad6) and DNA electroporation (DNA-EP) to induce immune responses against dTERT in dogs affected by malignant lymphoma (ML). The vaccine was combined with standard chemotherapy regimen [cyclophosphamide, vincristine, prednisone (COP)]. dTERT-specific immune response was induced in 13 out of 14 treated animals (93%) and remained detectable and long-lasting with the absence of autoimmunity or other side effects. Most interestingly, the survival time of vaccine/Chemo-treated dogs was significantly increased over historic controls of Chemo-treated animals (>97.8 versus 37 weeks, respectively, P = 0.001). Our results show that Ad6/DNA-EP-based cancer vaccine against dTERT overcomes host immune tolerance, should be combined with chemotherapy, induces long-lasting immune responses, and significantly prolongs the survival of ML canine patients. These data support further evaluation of this approach in human clinical trials.


Assuntos
Vacinas Anticâncer/imunologia , Imunoterapia/métodos , Linfoma de Células B/imunologia , Telomerase/imunologia , Adenoviridae/genética , Animais , Cães , Eletroporação , Humanos , Linfoma de Células B/metabolismo
6.
Vaccine ; 28(20): 3522-30, 2010 Apr 30.
Artigo em Inglês | MEDLINE | ID: mdl-20332048

RESUMO

The telomerase reverse transcriptase (TERT) is an attractive target for cancer vaccination because its expression is reactivated in most tumors. In this study, we have evaluated the ability of a genetic vaccine targeting murine TERT (mTERT) based on DNA electroporation (DNA-EP) and adenovirus serotype 6 (Ad6) to exert therapeutic effects in combination with a novel TLR9 agonist, referred to as immune modulatory oligonucleotide (IMO), as an adjuvant. IMO was administered to mice at the same time as vaccine. IMO induced dose-dependent cytokine secretion and activation of NK cells. Most importantly, vaccination of mice with IMO in combination with mTERT vaccine conferred therapeutic benefit in tumor bearing animals and this effect was associated with increased NK, DC and T cell tumor infiltration. These data show that appropriate combination of a DNA-EP/Ad6-based cancer vaccine against TERT with IMO induces multiple effects on innate and adaptive immune responses resulting in a significant antitumor efficacy.


Assuntos
Vacinas Anticâncer/imunologia , Telomerase/imunologia , Receptor Toll-Like 9/agonistas , Vacinas de DNA/imunologia , Imunidade Adaptativa , Adenoviridae , Adjuvantes Imunológicos/genética , Animais , Vacinas Anticâncer/genética , Citocinas/imunologia , Citocinas/metabolismo , Células Dendríticas/imunologia , Eletroporação , Feminino , Imunidade Inata , Células Matadoras Naturais/imunologia , Melanoma Experimental/imunologia , Melanoma Experimental/prevenção & controle , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Oligonucleotídeos/imunologia , Linfócitos T/imunologia , Vacinas de DNA/genética , Ensaios Antitumorais Modelo de Xenoenxerto
7.
Vaccine ; 28(5): 1201-8, 2010 Feb 03.
Artigo em Inglês | MEDLINE | ID: mdl-19944791

RESUMO

Pet dogs represent a valuable pre-clinical model to assess the efficacy of oncology drugs. Additionally, canine cancers occur with an incidence similar to that of humans and share many features with human malignancies including histological appearance, tumor genetics, biological behavior and response to conventional therapies. The telomerase reverse transcriptase (TERT) is reactivated in most of human and dog tumors. Similarly, HER-2/neu oncoprotein is overexpressed in a proportion of canine breast cancers. Therefore, TERT and HER-2/neu can constitute valid tumor associated antigens (TAA), suitable targets for translational cancer immunotherapy in dogs. In this study, we have evaluated the ability of DNA electroporation (DNA-EP) and Adenovirus serotype 6 (Ad6) to induce immune responses against dog TERT (dTERT) and HER-2/neu in healthy dogs. Vaccination was effective in all treated animals and the adaptive immune response remained detectable and long-lasting in the absence of autoimmunity or other side-effects. Our results show that DNA-EP/Ad6-based cancer vaccine induces adaptive immune responses against TAA in canine subjects and support further evaluation of this approach in cancer dog patients.


Assuntos
Vacinas Anticâncer/imunologia , Imunização , Neoplasias Mamárias Animais/prevenção & controle , Receptor ErbB-2/imunologia , Imunidade Adaptativa , Adenoviridae , Animais , Vacinas Anticâncer/genética , Doenças do Cão , Cães , Eletroporação , Feminino , Humanos , Neoplasias Mamárias Animais/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Receptor ErbB-2/genética , Telomerase
8.
Expert Opin Biol Ther ; 9(10): 1279-87, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19645630

RESUMO

Adenovirus (Ad)-based gene transfer has been successfully utilised in gene therapy and vaccine applications. To date, an increasing number of human clinical trials utilise recombinant Ad-based vectors as a gene transfer platform. In particular, progress has been made recently in utilising Ad-based vectors as a vaccine platform in HIV, cancer immunotherapy approaches and in vaccination for other infections. Despite these successes, the scientific and bio-industrial communities have recently recognised that innate and pre-existing immunity against Ad vectors can constitute a serious obstacle to the development and application of this technology. It is essential to overcome vector-mediated immune responses, such as production of inflammatory cytokines and pre-existing immunity to Ad, because the induction of these responses not only shortens the period of gene expression but also leads to serious side effects. This review focuses on the biology of Ad infection and the approaches that are being adopted to overcome immunity against the Ad-based vectors.


Assuntos
Adenoviridae/genética , Engenharia Genética , Vetores Genéticos/imunologia , Terapia Genética , Humanos , Neoplasias/terapia , Vacinas/genética
9.
Mol Ther ; 17(10): 1804-13, 2009 10.
Artigo em Inglês | MEDLINE | ID: mdl-19623161

RESUMO

The human telomerase reverse transcriptase (hTERT) is an attractive target for human cancer vaccination because its expression is reactivated in most human tumors. We have evaluated the ability of DNA electroporation (DNA-EP) and adenovirus serotype 6 (Ad6) to induce immune responses against hTERT in nonhuman primates (NHPs) (Macaca mulatta). Vaccination was effective in all treated animals, and the adaptive immune response remained detectable and long lasting without side effects. To further enhance the efficacy of the hTERT vaccine, we evaluated the combination of hTERT vaccine and a novel TLR9 agonist, referred to as immunomodulatory oligonucleotide (IMO). Monkeys were dosed weekly with IMO concurrently with the vaccine regimen and showed increases in cytokine secretion and activation of natural killer (NK) cells compared with the group that received vaccine alone. Using a peptide array, a specific profile of B-cell reactive epitopes was identified when hTERT vaccine was combined with IMO. The combination of IMO with hTERT genetic vaccine did not impact vaccine-induced TERT-specific cell-mediated immunity. Our results show that appropriate combination of a DNA-EP/Ad6-based cancer vaccine against hTERT with IMO induces multiple effects on innate and adaptive immune responses in NHPs.


Assuntos
Imunidade Inata/efeitos dos fármacos , Fatores Imunológicos/farmacologia , Telomerase/imunologia , Receptor Toll-Like 9/agonistas , Adenoviridae/genética , Animais , Eletroporação , Epitopos de Linfócito B/imunologia , Imunidade Celular/imunologia , Imunidade Inata/genética , Interferon-alfa/metabolismo , Interleucina-10/metabolismo , Interleucina-12/metabolismo , Interleucina-6/metabolismo , Macaca mulatta , Telomerase/genética
10.
J Immunother ; 32(7): 744-54, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19561534

RESUMO

Human leukocyte antigen (HLA)-A2.1 transgenic mice (HHD) represent a valuable model to study and predict the immunogenicity of vaccines against pathogens. However, HHD mice are unsuitable for in vivo studies of cancer vaccines against human tumor-associated antigens because they lack T-cell tolerance that is key to define the potency of the treatment. In this study, we developed HHD/carcinoembryonic antigen P(CEA) hybrid mice by breeding transgenic mice homozygous for CEA with HHD. These mice express human CEA, present epitopes solely through HLA-A2.1 molecules and constitute a unique in vivo animal model to study HLA-A2.1-restricted immune response of a human CEA-based vaccine. Owing to the immune tolerance, HHD/CEA mice show a limited immune response and expansion of a different and restricted T-cell receptor repertoire after antigen-specific stimulation. Our data show that genetic vectors expressing CEA and peptide-based vaccines are able to efficiently break immune tolerance against CEA and to elicit strong immune response against HLA-A2.1-restricted CEA epitopes. Most importantly, efficient lysis of human CEA+/HLA-A2.1+ tumor cells was observed and significant protection against HHD/CEA tumor cells was achieved in HHD/CEA-vaccinated mice. Hence, HHD/CEA provides a relevant model for the evaluation of the potential efficacy of human CEA-based vaccines.


Assuntos
Vacinas Anticâncer/imunologia , Antígeno Carcinoembrionário/imunologia , Antígeno HLA-A2/imunologia , Melanoma Experimental/imunologia , Adenoviridae/genética , Animais , Vacinas Anticâncer/administração & dosagem , Antígeno Carcinoembrionário/genética , Antígeno Carcinoembrionário/metabolismo , Linhagem Celular Tumoral , Citotoxicidade Imunológica/imunologia , Ensaio de Imunoadsorção Enzimática , Epitopos/imunologia , Fezes/química , Feminino , Citometria de Fluxo , Trato Gastrointestinal/metabolismo , Antígenos H-2/imunologia , Antígeno HLA-A2/genética , Antígeno HLA-A2/metabolismo , Humanos , Masculino , Melanoma Experimental/patologia , Melanoma Experimental/prevenção & controle , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Receptores de Antígenos de Linfócitos T/imunologia , Receptores de Antígenos de Linfócitos T/metabolismo , Transfecção , Vacinação/métodos
11.
Clin Cancer Res ; 15(12): 4104-13, 2009 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-19509157

RESUMO

PURPOSE: Matrix metalloproteinases (MMP) are zinc-dependent endopeptidases that mediate numerous physiologic and pathologic processes, including matrix degradation, tissue remodeling, inflammation, and tumor metastasis. To develop a vaccine targeting stromal antigens expressed by cancer-associated fibroblasts, we focused on MMP11 (or stromelysin 3). MMP11 expression correlates with aggressive profile and invasiveness of different types of carcinoma. EXPERIMENTAL DESIGN: To show the efficacy of a vaccine targeting MMP11, we constructed a series of plasmid DNA vectors expressing murine MMP11. Mice were vaccinated by i.m. injection followed by in vivo DNA electroporation. A chemically induced, MMP11-overexpressing colon cancer model was established and characterized. Antibody and T-cell responses were determined, and immunoreactive epitopes were characterized. To analyze the possible use of MMP11 as tumor-associated antigen in cancer patients, HLA-A2.1 transgenic mice (HHD) were used to identify reactive epitopes as tools to assess immunogenicity in humans. RESULTS: Using microarray, we confirmed the overexpression of MMP11 mRNA in a large panel of human tumor samples. MMP11 vaccine induced cell mediated and antibody immune response and exerted significant antitumoral protection in mice with colon cancer in prophylactic and therapeutic settings. HHD transgenic mice were vaccinated with a plasmid encoding human MMP11, and a HLA-A2.1--restricted epitope (hMMP(237)) was identified. hMMP(237) was shown to be immunogenic in human peripheral blood mononuclear cells (PBMC) by in vitro priming. CONCLUSION: Our study describes the identification of MMP11 as a novel broadly expressed tumor associated antigen as target candidate for cancer immunotherapy.


Assuntos
Antígenos de Neoplasias/imunologia , Vacinas Anticâncer/uso terapêutico , Metaloproteinase 11 da Matriz/imunologia , Inibidores de Metaloproteinases de Matriz , Neoplasias/terapia , Animais , Epitopos/imunologia , Antígeno HLA-A2/imunologia , Humanos , Imunoterapia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Transgênicos , Neoplasias/imunologia , Transfecção
12.
Clin Cancer Res ; 15(5): 1575-84, 2009 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-19240169

RESUMO

PURPOSE: Oligodeoxynucleotides containing unmethylated CpG dinucleotides induce innate and adaptive immunity through Toll-like receptor 9 (TLR9). In the present study, we have examined the ability of a novel agonist of TLR9, called immunomodulatory oligonucleotide (IMO), to enhance effects of a HER-2/neu plasmid DNA electroporation/adenovirus (DNA-EP/Ad) vaccine. EXPERIMENTAL DESIGN: BALB/NeuT mice were treated with DNA-EP vaccine alone, IMO alone, or the combination of two agents starting at week 13, when all mice showed mammary neoplasia. Tumor growth and survival were documented. Antibody and CD8+ T-cell responses were determined. Peptide microarray analysis of sera was carried out to identify immunoreactive epitopes. Additionally, microCT and microPET imaging was carried out in an advanced-stage tumor model starting treatment at week 17 in BALB/NeuT mice. RESULTS: The combination of DNA-EP and IMO resulted in significant tumor regression or delay to tumor progression. 2-Deoxy-2-[18F]fluoro-D-glucose microPET and microCT imaging of mice showed reduced tumor size in the DNA-EP/IMO combination treatment group. Mice treated with the combination produced greater antibody titers with IgG2a isotype switch and antibody-dependent cellular cytotoxicity activity than did mice treated with DNA-EP vaccine. An immunogenic B-cell linear epitope, r70, within the HER-2 dimerization domain was identified through microarray analysis. Heterologous DNA-EP/Ad vaccination combined with IMO increased mice survival. CONCLUSION: The combination of HER-2/neu genetic vaccine and novel agonist of TLR9 had potent antitumor activity associated with antibody isotype switch and antibody-dependent cellular cytotoxicity activities. These results support possible clinical trials of the combination of DNA-EP/Ad-based cancer vaccines and IMO.


Assuntos
Neoplasias Mamárias Experimentais/imunologia , Neoplasias Mamárias Experimentais/terapia , Plasmídeos/administração & dosagem , Receptor ErbB-2/imunologia , Receptor Toll-Like 9/fisiologia , Vacinas de DNA/uso terapêutico , Adenoviridae/genética , Fosfatase Alcalina/metabolismo , Animais , Citotoxicidade Celular Dependente de Anticorpos , Células Apresentadoras de Antígenos/imunologia , Terapia Combinada , DNA/administração & dosagem , Dimerização , Eletroporação , Ensaio de Imunoadsorção Enzimática , Feminino , Interleucina-12/metabolismo , Células Matadoras Naturais/imunologia , Células Matadoras Naturais/metabolismo , Neoplasias Mamárias Experimentais/genética , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Transgênicos , Fragmentos de Peptídeos/genética , Fragmentos de Peptídeos/imunologia , Fragmentos de Peptídeos/metabolismo , Tomografia por Emissão de Pósitrons , Ratos
13.
Vaccine ; 27(9): 1293-300, 2009 Feb 25.
Artigo em Inglês | MEDLINE | ID: mdl-19162112

RESUMO

The use of adenovirus (Ad) as vaccine vectors is hindered by pre-existing immunity to human Ads in most of the human population. In order to overcome this limitation, uncommon alternative Ad serotypes need to be utilized. In this study, an E1-E3 deleted recombinant Ad based on the chimpanzee serotype 3 (ChAd3) was engineered to express human carcinoembryonic antigen (CEA) protein or rat neu extracellular/transmembrane domains (ECD.TM). ChAd3 vectors were tested in CEA transgenic (CEA.Tg) and BALB/NeuT mice, which show immunologic tolerance to these antigens. ChAd3 is capable of inducing an immune response comparable to that of hAd5 serotype-based vectors, thus breaking tolerance to tumor associated antigens (TAAs) and achieving anti-tumor effects. Of importance is that ChAd3 can overcome hAd5 pre-existing immunity and work in conjunction with DNA electroporation (DNA-EP) and other Ad vaccines based on common human serotypes.


Assuntos
Adenoviridae/imunologia , Vacinas Anticâncer/imunologia , Antígeno Carcinoembrionário/genética , Pan troglodytes/imunologia , Linfócitos T/imunologia , Adenoviridae/genética , Animais , Vacinas Anticâncer/genética , Antígeno Carcinoembrionário/imunologia , Vetores Genéticos , Humanos , Imunidade , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Pan troglodytes/genética , Plasmídeos
14.
Hum Gene Ther ; 20(2): 125-36, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18937552

RESUMO

A genetic vaccine platform based on DNA electroporation (DNA-EP) and adenovirus (Ad) was used to generate immune response against human carcinoembryonic antigen (CEA) and antitumor effects in murine models with spontaneous tumors arising in an orthotopic location. CEA transgenic (CEA.Tg) mice treated with the carcinogen 1,2-dimethylhydrazine developed CEA-overexpressing tumors that resembled human sporadic colorectal cancer. APC1638N/CEA hybrid mice, generated by crossing mice carrying the adenomatous polyposis coli (Apc1638N) gene mutation with CEA.Tg mice, are representative of human familial polyposis and develop polyps that overexpress the antigen. In both models, the DNA-EP/Ad vaccine succeeded in breaking immune tolerance and achieved significant antitumor effects in therapeutic settings. Our data suggest that genetic vaccines targeting CEA may be feasible strategies against gut tumors that overexpress the antigen. In addition, these models are powerful systems for evaluating antigen-specific tumor immunity and assessing therapeutic vaccine strategies for human colorectal cancer.


Assuntos
Pólipos Adenomatosos/terapia , Vacinas Anticâncer/uso terapêutico , Antígeno Carcinoembrionário/metabolismo , Neoplasias Colorretais/terapia , Vetores Genéticos , Pólipos Adenomatosos/imunologia , Pólipos Adenomatosos/patologia , Adenoviridae/genética , Animais , Vacinas Anticâncer/genética , Antígeno Carcinoembrionário/genética , Neoplasias Colorretais/imunologia , Neoplasias Colorretais/patologia , Modelos Animais de Doenças , Vetores Genéticos/genética , Humanos , Imuno-Histoquímica , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Transgênicos , Padrões de Referência , Regulação para Cima
15.
Biochim Biophys Acta ; 1584(1): 46-54, 2002 Sep 05.
Artigo em Inglês | MEDLINE | ID: mdl-12213492

RESUMO

Inositide-specific phospholipase C (PLC) signaling constitutes a central intermediate in a number of cellular functions among which the control of cell growth raises a particular interest. Indeed, we have previously shown that nuclear phospholipase C beta1 (PLC beta1) is central for the regulation of mitogen-induced cell growth. We have also assigned by fluorescence in situ hybridization (FISH) analysis the PLC beta1 to human chromosome 20p12. In this study, we have carried out a detailed analysis of the human gene, showing the existence of alternative splicing, which gives rise, besides the two forms (1a and 1b) already shown in rodents, to a new 600 bp smaller form coding for a 110 kDa protein. We have also identified a new exon at the 5', showing no homology with the rodent sequence. Here we provide the complete determination of the exon/intron structure of the gene spanning 250 kb of DNA. We found that the exons are quite small, ranging from 49 to 222 bp, while the introns vary between 108 bp and 34,400 bp. The availability of the understanding of the genome organization of this inositide-specific PLC, which represents a key step of the cell cycle related signaling, could actually pave the way for further genetic analysis of p12 region of human chromosome 20 in diseases involving alterations of the control of cell growth such as malignancies.


Assuntos
Isoenzimas/genética , Fosfolipases Tipo C/genética , Regiões 3' não Traduzidas , Regiões 5' não Traduzidas , Adulto , Processamento Alternativo , Sequência de Bases , Northern Blotting , Western Blotting , Clonagem Molecular , DNA Complementar/biossíntese , DNA Complementar/química , Feto , Amplificação de Genes , Humanos , Isoenzimas/química , Dados de Sequência Molecular , Fosfolipase C beta , Regiões Promotoras Genéticas , Transfecção , Fosfolipases Tipo C/química
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...