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 Ther Oncolytics ; 5: 41-61, 2017 Jun 16.
Artigo em Inglês | MEDLINE | ID: mdl-28480327

RESUMO

Oncolytic vaccinia virus (VACV) therapy is an alternative cancer treatment modality that mediates targeted tumor destruction through a tumor-selective replication and an induction of anti-tumor immunity. We developed a humanized tumor mouse model with subcutaneous human tumors to analyze the interactions of VACV with the developing tumors and human immune system. A successful systemic reconstitution with human immune cells including functional T cells as well as development of tumors infiltrated with human T and natural killer (NK) cells was observed. We also demonstrated successful in vivo colonization of such tumors with systemically administered VACVs. Further, a new recombinant GLV-1h376 VACV encoding for a secreted human CTLA4-blocking single-chain antibody (CTLA4 scAb) was tested. Surprisingly, although proving CTLA4 scAb's in vitro binding ability and functionality in cell culture, beside the significant increase of CD56bright NK cell subset, GLV-1h376 was not able to increase cytotoxic T or overall NK cell levels at the tumor site. Importantly, the virus-encoded ß-glucuronidase as a measure of viral titer and CTLA4 scAb amount was demonstrated. Therefore, studies in our "patient-like" humanized tumor mouse model allow the exploration of newly designed therapy strategies considering the complex relationships between the developing tumor, the oncolytic virus, and the human immune system.

3.
PLoS One ; 10(9): e0137573, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26348361

RESUMO

Blood tests are necessary, easy-to-perform and low-cost alternatives for monitoring of oncolytic virotherapy and other biological therapies in translational research. Here we assessed three candidate proteins with the potential to be used as biomarkers in biological fluids: two glucuronidases from E. coli (GusA) and Staphylococcus sp. RLH1 (GusPlus), and the luciferase from Gaussia princeps (GLuc). The three genes encoding these proteins were inserted individually into vaccinia virus GLV-1h68 genome under the control of an identical promoter. The three resulting recombinant viruses were used to infect tumor cells in cultures and human tumor xenografts in nude mice. In contrast to the actively secreted GLuc, the cytoplasmic glucuronidases GusA and GusPlus were released into the supernatants only as a result of virus-mediated oncolysis. GusPlus resulted in the most sensitive detection of enzyme activity under controlled assay conditions in samples containing as little as 1 pg/ml of GusPlus, followed by GusA (25 pg/ml) and GLuc (≥375 pg/ml). Unexpectedly, even though GusA had a lower specific activity compared to GusPlus, the substrate conversion in the serum of tumor-bearing mice injected with the GusA-encoding virus strains was substantially higher than that of GusPlus. This was attributed to a 3.2 fold and 16.2 fold longer half-life of GusA in the blood stream compared to GusPlus and GLuc respectively, thus a more sensitive monitor of virus replication than the other two enzymes. Due to the good correlation between enzymatic activity of expressed marker gene and virus titer, we conclude that the amount of the biomarker protein in the body fluid semiquantitatively represents the amount of virus in the infected tumors which was confirmed by low light imaging. We found GusA to be the most reliable biomarker for monitoring oncolytic virotherapy among the three tested markers.


Assuntos
Biomarcadores Tumorais/genética , Glucuronidase/genética , Luciferases/genética , Neoplasias/terapia , Terapia Viral Oncolítica , Animais , Linhagem Celular Tumoral , Escherichia coli/enzimologia , Glucuronidase/biossíntese , Humanos , Luciferases/biossíntese , Camundongos , Neoplasias/genética , Neoplasias/virologia , Vírus Oncolíticos/genética , Staphylococcus/enzimologia , Vaccinia virus/genética , Ensaios Antitumorais Modelo de Xenoenxerto
4.
Viruses ; 7(7): 4075-92, 2015 Jul 20.
Artigo em Inglês | MEDLINE | ID: mdl-26205404

RESUMO

Virotherapy on the basis of oncolytic vaccinia virus (VACV) strains is a novel approach for canine cancer therapy. Here we describe, for the first time, the characterization and the use of VACV strain GLV-5b451 expressing the anti-vascular endothelial growth factor (VEGF) single-chain antibody (scAb) GLAF-2 as therapeutic agent against different canine cancers. Cell culture data demonstrated that GLV-5b451 efficiently infected and destroyed all four tested canine cancer cell lines including: mammary carcinoma (MTH52c), mammary adenoma (ZMTH3), prostate carcinoma (CT1258), and soft tissue sarcoma (STSA-1). The GLV-5b451 virus-mediated production of GLAF-2 antibody was observed in all four cancer cell lines. In addition, this antibody specifically recognized canine VEGF. Finally, in canine soft tissue sarcoma (CSTS) xenografted mice, a single systemic administration of GLV-5b451 was found to be safe and led to anti-tumor effects resulting in the significant reduction and substantial long-term inhibition of tumor growth. A CD31-based immuno-staining showed significantly decreased neo-angiogenesis in GLV-5b451-treated tumors compared to the controls. In summary, these findings indicate that GLV-5b451 has potential for use as a therapeutic agent in the treatment of CSTS.


Assuntos
Doenças do Cão/terapia , Neoplasias/veterinária , Terapia Viral Oncolítica , Vírus Oncolíticos/genética , Anticorpos de Cadeia Única/imunologia , Vaccinia virus/genética , Fator A de Crescimento do Endotélio Vascular/imunologia , Animais , Doenças do Cão/genética , Doenças do Cão/metabolismo , Cães , Feminino , Camundongos , Camundongos Nus , Neoplasias/genética , Neoplasias/imunologia , Neoplasias/terapia , Vírus Oncolíticos/metabolismo , Anticorpos de Cadeia Única/genética , Anticorpos de Cadeia Única/uso terapêutico , Vaccinia virus/metabolismo
5.
Methods Mol Biol ; 1317: 225-37, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26072410

RESUMO

Herein we describe the use of the vaccinia virus strain GLV-1h68 as a theragnostic agent in cancer models. To date, GLV-1h68 has been used successfully in more than 50 xenograft tumor models. The recombinant vaccinia virus strain has been equipped with heterologous expression cassettes for a luciferase-fluorescent protein fusion gene, bacterial beta-galactosidase, as well as a bacterial glucuronidase. The methods to investigate and monitor GLV-1h68 mediated virotherapy, including optical imaging and biomarker analysis, will be presented in detail.


Assuntos
Terapia Viral Oncolítica/métodos , Vaccinia virus/fisiologia , Animais , Linhagem Celular , Ensaios Enzimáticos , Glucuronidase/metabolismo , Humanos , Luciferases/metabolismo , Camundongos Nus , Imagem Óptica , Vaccinia virus/genética , Ensaios Antitumorais Modelo de Xenoenxerto
6.
Mol Ther Oncolytics ; 2: 15003, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-27119102

RESUMO

The tumor microenvironment plays an important role in tumor growth and progression. Here we demonstrate that vaccinia virus-mediated, constitutively expressed intratumoral antibodies against vascular endothelial growth factor (VEGF), epidermal growth factor receptor (EGFR), and fibroblast activation protein (FAP) significantly improved tumor regression and oncolytic virotherapy through suppression of angiogenesis, cell proliferation, and stromagenesis in virus-colonized tumors. In contrast to the tumor growth inhibition by the three tumor growth-inhibiting antibodies individually, when two of the three antibodies were expressed simultaneously by single vaccinia virus strains tumor regression was further enhanced. These findings strongly indicate that interference with the two tumor growth-stimulating mechanisms did in fact result in enhanced therapeutic efficacy in tumor xenograft models and may lead to an effective therapy in patients with cancer.

7.
Breast Cancer Res Treat ; 148(3): 489-99, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25391896

RESUMO

Vascular endothelial growth factor (VEGF) expression is higher in triple-negative breast cancers (TNBC) compared to other subtypes and is reported to predict incidence of distant metastases and shorter overall survival. We investigated the therapeutic impact of a vaccinia virus (VACV) GLV-1h164 (derived from its parent virus GLV-1h100), encoding a single-chain antibody (scAb) against VEGF (GLAF-2) in an orthotopic TNBC murine model. GLV-1h164 was tested against multiple TNBC cell lines. Viral infectivity, cytotoxicity, and replication were determined. Mammary fat pad tumors were generated in athymic nude mice using MDA-MB-231 cells. Xenografts were treated with GLV-1h164, GLV-1h100, or PBS and followed for tumor growth. Viral infectivity was time- and concentration-dependent. GLV-1h164 killed TNBC cell lines in a dose-dependent fashion with greater than 90% cytotoxicity within 4 days at a multiplicity of infection of 5.0. In vitro, cytotoxicity of GLV-1h164 was identical to GLV-1h100. GLV-1h164 replicated efficiently in all cell lines with an over 400-fold increase in copy numbers from the initial viral dose within 4 days. In vivo, mean tumor volumes after 2 weeks of treatment were 73, 191, and 422 mm(3) (GLV-1h164, GLV-1h100, and PBS, respectively) (p < 0.05). Both in vivo Doppler ultrasonography and immuno-staining showed decreased neo-angiogenesis in GLV-1h164-treated tumors compared to both GLV-1h100 and PBS controls (p < 0.05). This is the first study to demonstrate efficient combination of oncolytic and anti-angiogenic activity of a novel VACV on TNBC xenografts. Our results suggest that GLV-1h164 is a promising therapeutic agent that warrants testing for patients with TNBC.


Assuntos
Neovascularização Patológica/terapia , Vírus Oncolíticos/genética , Neoplasias de Mama Triplo Negativas/terapia , Vaccinia virus/genética , Inibidores da Angiogênese/administração & dosagem , Inibidores da Angiogênese/genética , Animais , Linhagem Celular Tumoral , Feminino , Humanos , Camundongos , Neovascularização Patológica/genética , Neovascularização Patológica/virologia , Terapia Viral Oncolítica/métodos , Neoplasias de Mama Triplo Negativas/patologia , Neoplasias de Mama Triplo Negativas/virologia , Fator A de Crescimento do Endotélio Vascular/biossíntese , Ensaios Antitumorais Modelo de Xenoenxerto
8.
PLoS One ; 9(8): e104337, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25093734

RESUMO

Virotherapy on the basis of oncolytic vaccinia virus (VACV) infection is a promising approach for cancer therapy. In this study we describe the establishment of a new preclinical model of feline mammary carcinoma (FMC) using a recently established cancer cell line, DT09/06. In addition, we evaluated a recombinant vaccinia virus strain, GLV-5b451, expressing the anti-vascular endothelial growth factor (VEGF) single-chain antibody (scAb) GLAF-2 as an oncolytic agent against FMC. Cell culture data demonstrate that GLV-5b451 virus efficiently infected, replicated in and destroyed DT09/06 cancer cells. In the selected xenografts of FMC, a single systemic administration of GLV-5b451 led to significant inhibition of tumor growth in comparison to untreated tumor-bearing mice. Furthermore, tumor-specific virus infection led to overproduction of functional scAb GLAF-2, which caused drastic reduction of intratumoral VEGF levels and inhibition of angiogenesis. In summary, here we have shown, for the first time, that the vaccinia virus strains and especially GLV-5b451 have great potential for effective treatment of FMC in animal model.


Assuntos
Vetores Genéticos/genética , Neoplasias Mamárias Animais/genética , Neoplasias Mamárias Animais/patologia , Vírus Oncolíticos/genética , Vaccinia virus/genética , Animais , Gatos , Linhagem Celular , Linhagem Celular Tumoral , Sobrevivência Celular/genética , Chlorocebus aethiops , Feminino , Expressão Gênica , Vetores Genéticos/administração & dosagem , Vetores Genéticos/farmacocinética , Neoplasias Mamárias Animais/imunologia , Neoplasias Mamárias Animais/terapia , Camundongos , Camundongos Nus , Neovascularização Patológica/genética , Neovascularização Patológica/terapia , Terapia Viral Oncolítica , Anticorpos de Cadeia Única/genética , Anticorpos de Cadeia Única/metabolismo , Transdução Genética , Fator A de Crescimento do Endotélio Vascular/antagonistas & inibidores , Fator A de Crescimento do Endotélio Vascular/genética , Fator A de Crescimento do Endotélio Vascular/metabolismo , Replicação Viral , Ensaios Antitumorais Modelo de Xenoenxerto
9.
Int J Cancer ; 133(12): 2989-99, 2013 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-23729266

RESUMO

Oncolytic viruses are currently in clinical trials for a variety of tumors, including high grade gliomas. A characteristic feature of high grade gliomas is their high vascularity and treatment approaches targeting tumor endothelium are under investigation, including bevacizumab. The aim of this study was to improve oncolytic viral therapy by combining it with ionizing radiation and to radiosensitize tumor vasculature through a viral encoded anti-angiogenic payload. Here, we show how vaccinia virus-mediated expression of a single-chain antibody targeting VEGF resulted in radiosensitization of the tumor-associated vasculature. Cell culture experiments demonstrated that purified vaccinia virus encoded antibody targeting VEGF reversed VEGF-induced radioresistance specifically in endothelial cells but not tumor cells. In a subcutaneous model of U-87 glioma, systemically administered oncolytic vaccinia virus expressing anti-VEGF antibody (GLV-1h164) in combination with fractionated irradiation resulted in enhanced tumor growth inhibition when compared to nonanti-VEGF expressing oncolytic virus (GLV-1h68) and irradiation. Irradiation of tumor xenografts resulted in an increase in VACV replication of both GLV-1h68 and GLV-1h164. However, GLV-1h164 in combination with irradiation resulted in a drastic decrease in intratumoral VEGF levels and tumor vessel numbers in comparison to GLV-1h68 and irradiation. These findings demonstrate the incorporation of an oncolytic virus expressing an anti-VEGF antibody (GLV-1h164) into a fractionated radiation scheme to target tumor cells by enhanced VACV replication in irradiated tumors as well as to radiosensitize tumor endothelium which results in enhanced efficacy of combination therapy of human glioma xenografts.


Assuntos
Endotélio Vascular/efeitos da radiação , Glioma/terapia , Terapia Viral Oncolítica/métodos , Tolerância a Radiação , Vaccinia virus/genética , Fator A de Crescimento do Endotélio Vascular/antagonistas & inibidores , Animais , Linhagem Celular Tumoral , Glioma/irrigação sanguínea , Humanos , Masculino , Camundongos , Fator A de Crescimento do Endotélio Vascular/fisiologia
10.
J Transl Med ; 11: 106, 2013 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-23635329

RESUMO

BACKGROUND: Malignant pleural effusion (MPE) is associated with advanced stages of lung cancer and is mainly dependent on invasion of the pleura and expression of vascular endothelial growth factor (VEGF) by cancer cells. As MPE indicates an incurable disease with limited palliative treatment options and poor outcome, there is an urgent need for new and efficient treatment options. METHODS: In this study, we used subcutaneously generated PC14PE6 lung adenocarcinoma xenografts in athymic mice that developed subcutaneous malignant effusions (ME) which mimic pleural effusions of the orthotopic model. Using this approach monitoring of therapeutic intervention was facilitated by direct observation of subcutaneous ME formation without the need of sacrificing mice or special imaging equipment as in case of MPE. Further, we tested oncolytic virotherapy using Vaccinia virus as a novel treatment modality against ME in this subcutaneous PC14PE6 xenograft model of advanced lung adenocarcinoma. RESULTS: We demonstrated significant therapeutic efficacy of Vaccinia virus treatment of both advanced lung adenocarcinoma and tumor-associated ME. We attribute the efficacy to the virus-mediated reduction of tumor cell-derived VEGF levels in tumors, decreased invasion of tumor cells into the peritumoral tissue, and to viral infection of the blood vessel-invading tumor cells. Moreover, we showed that the use of oncolytic Vaccinia virus encoding for a single-chain antibody (scAb) against VEGF (GLAF-1) significantly enhanced mono-therapy of oncolytic treatment. CONCLUSIONS: Here, we demonstrate for the first time that oncolytic virotherapy using tumor-specific Vaccinia virus represents a novel and promising treatment modality for therapy of ME associated with advanced lung cancer.


Assuntos
Neoplasias Pulmonares/terapia , Terapia Viral Oncolítica/métodos , Derrame Pleural Maligno/terapia , Animais , Linhagem Celular Tumoral , Feminino , Citometria de Fluxo , Humanos , Injeções Subcutâneas , Imageamento por Ressonância Magnética , Camundongos , Camundongos Nus , Vírus Oncolíticos/metabolismo , Anticorpos de Cadeia Única/química , Resultado do Tratamento , Vaccinia virus/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
11.
Bioengineered ; 4(2): 84-9, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23093804

RESUMO

Virotherapy on the basis of oncolytic vaccinia virus (VACV) strains is one novel approach for canine cancer therapy. In this study we described for the first time the characterization and the use of new VACV strain LIVP6.1.1 as an oncolytic agent against canine cancer in a panel of four canine cancer cell lines including: soft tissue sarcoma (STSA-1), melanoma (CHAS), osteosarcoma (D-17) and prostate carcinoma (DT08/40). Cell culture data demonstrated that LIVP6.1.1 efficiently infected and destroyed all four tested canine cancer cell lines. In two different xenograft models on the basis of the canine soft tissue sarcoma STSA-1 and the prostate carcinoma DT08/40 cell lines, a systemic administration of the LIVP6.1.1 virus was found to be safe and led to anti-tumor and immunological effects resulting in the significant reduction of tumor growth in comparison to untreated control mice. In summary, the pre-clinical evaluation has demonstrated the efficacy of LIVP6.1.1 for canine cancer therapy. Furthermore, a clinical trial with canine cancer patients has already been started.


Assuntos
Neoplasias/terapia , Vírus Oncolíticos/fisiologia , Vaccinia virus/fisiologia , Animais , Cães , Camundongos , Neoplasias/genética , Terapia Viral Oncolítica/métodos , Vírus Oncolíticos/genética , Vaccinia virus/genética , Ensaios Antitumorais Modelo de Xenoenxerto
12.
PLoS One ; 7(10): e47472, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23091626

RESUMO

Virotherapy using oncolytic vaccinia virus (VACV) strains is one promising new strategy for cancer therapy. We have previously reported that oncolytic vaccinia virus strains expressing an anti-VEGF (Vascular Endothelial Growth Factor) single-chain antibody (scAb) GLAF-1 exhibited significant therapeutic efficacy for treatment of human tumor xenografts. Here, we describe the use of oncolytic vaccinia virus GLV-1h109 encoding GLAF-1 for canine cancer therapy. In this study we analyzed the virus-mediated delivery and production of scAb GLAF-1 and the oncolytic and immunological effects of the GLV-1h109 vaccinia virus strain against canine soft tissue sarcoma and canine prostate carcinoma in xenograft models. Cell culture data demonstrated that the GLV-1h109 virus efficiently infect, replicate in and destroy both tested canine cancer cell lines. In addition, successful expression of GLAF-1 was demonstrated in virus-infected canine cancer cells and the antibody specifically recognized canine VEGF. In two different xenograft models, the systemic administration of the GLV-1h109 virus was found to be safe and led to anti-tumor and immunological effects resulting in the significant reduction of tumor growth in comparison to untreated control mice. Furthermore, tumor-specific virus infection led to a continued production of functional scAb GLAF-1, resulting in inhibition of angiogenesis. Overall, the GLV-1h109-mediated cancer therapy and production of immunotherapeutic anti-VEGF scAb may open the way for combination therapy concept i.e. vaccinia virus mediated oncolysis and intratumoral production of therapeutic drugs in canine cancer patients.


Assuntos
Doenças do Cão/terapia , Vetores Genéticos/genética , Neoplasias/veterinária , Terapia Viral Oncolítica , Anticorpos de Cadeia Única/genética , Vaccinia virus/genética , Fator A de Crescimento do Endotélio Vascular/antagonistas & inibidores , Animais , Linhagem Celular , Sobrevivência Celular , Doenças do Cão/imunologia , Doenças do Cão/patologia , Cães , Feminino , Expressão Gênica , Ordem dos Genes , Vetores Genéticos/administração & dosagem , Vetores Genéticos/farmacocinética , Glucuronidase/genética , Glucuronidase/metabolismo , Humanos , Camundongos , Camundongos Nus , Neoplasias/imunologia , Neoplasias/terapia , Anticorpos de Cadeia Única/metabolismo , Carga Tumoral , Fator A de Crescimento do Endotélio Vascular/imunologia , Replicação Viral , Ensaios Antitumorais Modelo de Xenoenxerto
13.
BMC Microbiol ; 11: 163, 2011 Jul 11.
Artigo em Inglês | MEDLINE | ID: mdl-21745384

RESUMO

BACKGROUND: Specific cell targeting is an important, yet unsolved problem in bacteria-based therapeutic applications, like tumor or gene therapy. Here, we describe the construction of a novel, internalin A and B (InlAB)-deficient Listeria monocytogenes strain (Lm-spa+), which expresses protein A of Staphylococcus aureus (SPA) and anchors SPA in the correct orientation on the bacterial cell surface. RESULTS: This listerial strain efficiently binds antibodies allowing specific interaction of the bacterium with the target recognized by the antibody. Binding of Trastuzumab (Herceptin®) or Cetuximab (Erbitux®) to Lm-spa+, two clinically approved monoclonal antibodies directed against HER2/neu and EGFR/HER1, respectively, triggers InlAB-independent internalization into non-phagocytic cancer cell lines overexpressing the respective receptors. Internalization, subsequent escape into the host cell cytosol and intracellular replication of these bacteria are as efficient as of the corresponding InlAB-positive, SPA-negative parental strain. This specific antibody/receptor-mediated internalization of Lm-spa+ is shown in the murine 4T1 tumor cell line, the isogenic 4T1-HER2 cell line as well as the human cancer cell lines SK-BR-3 and SK-OV-3. Importantly, this targeting approach is applicable in a xenograft mouse tumor model after crosslinking the antibody to SPA on the listerial cell surface. CONCLUSIONS: Binding of receptor-specific antibodies to SPA-expressing L. monocytogenes may represent a promising approach to target L. monocytogenes to host cells expressing specific receptors triggering internalization.


Assuntos
Anticorpos Antibacterianos/metabolismo , Proteínas de Bactérias/genética , Endocitose , Listeria monocytogenes/patogenicidade , Proteínas de Membrana/deficiência , Proteína Estafilocócica A/metabolismo , Animais , Linhagem Celular Tumoral , Receptores ErbB/imunologia , Feminino , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos SCID , Ligação Proteica , Receptor ErbB-2/imunologia , Proteína Estafilocócica A/genética
14.
Proc Natl Acad Sci U S A ; 106(31): 12915-20, 2009 Aug 04.
Artigo em Inglês | MEDLINE | ID: mdl-19617539

RESUMO

We previously reported that the replication-competent vaccinia virus (VACV) GLV-1h68 shows remarkable oncolytic activity and efficacy in different animal models as a single treatment modality and also in combination with chemotherapy [Yu YA, et al. (2009) Mol Cancer Ther 8:141-151]. Here, we report the construction of 3 VACV strains encoding GLAF-1, a previously undescribed engineered single-chain antibody (scAb). This unique scAb is transcribed from 3 vaccinia promoters (synthetic early, early/late, and late) and directed against both human and murine VEGFs. The expression of GLAF-1 was demonstrated in cell cultures. Also, the replication efficiency of all GLAF-1-expressing VACV strains in cell culture was similar to that of the parental GLV-1h68 virus. Successful tumor-specific delivery and continued production of functional scAb derived from individual VACV strains were obtained in tumor xenografts following a single intravenous injection of the virus. The VACV strains expressing the scAb exhibited significantly enhanced therapeutic efficacy in comparison to treatment of human tumor xenografts with the parental virus GLV-1h68. This enhanced efficacy was comparable to the concomitant treatment of tumors with a one-time i.v. injection of GLV-1h68 and multiple i.p. injections of Avastin. Taken together, the VACV-mediated delivery and production of immunotherapeutic anti-VEGF scAb in colonized tumors may open the way for a unique therapy concept: tumor-specific, locally amplified drug therapy in humans.


Assuntos
Anticorpos/genética , Neoplasias Experimentais/terapia , Terapia Viral Oncolítica , Vírus Oncolíticos/genética , Vaccinia virus/genética , Fator A de Crescimento do Endotélio Vascular/imunologia , Animais , Anticorpos Monoclonais/uso terapêutico , Anticorpos Monoclonais Humanizados , Bevacizumab , Linhagem Celular , Chlorocebus aethiops , Feminino , Humanos , Masculino , Camundongos , Transplante de Neoplasias , Neoplasias Experimentais/irrigação sanguínea , Transplante Heterólogo , Vaccinia virus/fisiologia , Fator A de Crescimento do Endotélio Vascular/antagonistas & inibidores , Replicação Viral
15.
Int J Med Microbiol ; 298(1-2): 45-58, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17936682

RESUMO

Listeria monocytogenes is a facultative intracellular bacterium that enters a variety of non-professional mammalian cells by triggered phagocytosis ("zipper mechanism") and replicates in the cytosol of the infected host cells. Therefore, it is a promising vaccine vector for the presentation of passenger antigens to the MHC class II and especially class I pathways. Here, we review recent progress made in our laboratory on the development of novel attenuated L. monocytogenes carrier strains for the delivery of heterologous antigens or antigen-encoding DNA and RNA to eukaryotic host cells. Based on the deletion of the chromosomal copy of the tryptophanyl-tRNA synthetase gene (trpS) and plasmid-based in trans complementation of the same, we were able to establish a balanced-lethal plasmid system in L. monocytogenes. Safety concerns in the antigen delivery in vivo were addressed by chromosomal deletion of genes in the basic branch of the aromatic amino acid pathway, resulting in safe, attenuated L. monocytogenes carrier strains. Furthermore, plasmid-based expression of a cytosolically expressed phage lysin resulted in a self-destructing carrier strain that has been successfully used for the delivery of antigens as well as antigen-encoding plasmid DNA and particularly mRNA, therefore overcoming bottlenecks that have been shown to exist for bacteria-mediated DNA delivery.


Assuntos
Antígenos/imunologia , Imunização/métodos , Listeria monocytogenes/imunologia , Plasmídeos/imunologia , Vacinas de DNA/imunologia , Vacinas/imunologia , Animais , RNA Mensageiro/imunologia , Linfócitos T/imunologia , Vacinas Atenuadas/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...