Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
1.
Arch Virol ; 160(10): 2525-34, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26215439

RESUMO

The existence of multiple antigenically distinct types and subtypes of influenza viruses allows the construction of a multivalent vector system for the mucosal delivery of foreign sequences. Influenza A viruses have been exploited successfully for the expression of extraneous antigens as well as immunostimulatory molecules. In this study, we describe the development of an influenza B virus vector whose functional part of the interferon antagonist NS1 was replaced by human interleukin 2 (IL2) as a genetic adjuvant. We demonstrate that IL2 expressed by this viral vector displays immune adjuvant activity in immunized mice. Animals vaccinated with the IL2 viral vector showed an increased hemagglutination inhibition antibody response and higher protective efficacy after challenge with a wild-type influenza B virus when compared to mice vaccinated with a control virus. Our results demonstrate that it is feasible to construct influenza B vaccine strains expressing immune-potentiating foreign sequences from the NS genomic segment. Based on these data, it is now hypothetically possible to create a trivalent (or quadrivalent) live attenuated influenza vaccine in which each component expresses a selected genetic adjuvant with tailored expression levels.


Assuntos
Vetores Genéticos/imunologia , Vírus da Influenza B/imunologia , Influenza Humana/prevenção & controle , Adjuvantes Imunológicos/administração & dosagem , Animais , Anticorpos Antivirais/imunologia , Feminino , Vetores Genéticos/administração & dosagem , Vetores Genéticos/genética , Humanos , Imunização , Vírus da Influenza B/genética , Vírus da Influenza B/fisiologia , Influenza Humana/imunologia , Influenza Humana/virologia , Interleucina-2/genética , Interleucina-2/imunologia , Camundongos
2.
J Gen Virol ; 95(Pt 2): 337-349, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24222196

RESUMO

The development of influenza virus vectors with long insertions of foreign sequences remains difficult due to the small size and instable nature of the virus. Here, we used the influenza virus inherent property of self-optimization to generate a vector stably expressing long transgenes from the NS1 protein ORF. This was achieved by continuous selection of bright fluorescent plaques of a GFP-expressing vector during multiple passages in mouse B16f1 cells. The newly generated vector acquired stability in IFN-competent cell lines and in vivo in murine lungs. Although improved vector fitness was associated with the appearance of four coding mutations in the polymerase (PB2), haemagglutinin and non-structural (NS) segments, the stability of the transgene expression was dependent primarily on the single mutation Q20R in the nuclear export protein (NEP). Importantly, a longer insert, such as a cassette of 1299 nt encoding two Mycobacterium tuberculosis Esat6 and Ag85A proteins, could substitute for the GFP transgene. Thus, the inherent property of the influenza virus to adapt can also be used to adjust a vector backbone to give stable expression of long transgenes.


Assuntos
Transporte Ativo do Núcleo Celular , Expressão Gênica , Vírus da Influenza A/genética , Vírus da Influenza A/fisiologia , Proteínas não Estruturais Virais/genética , Proteínas não Estruturais Virais/metabolismo , Replicação Viral , Animais , Linhagem Celular , Genes Reporter , Vetores Genéticos , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Camundongos , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Carga Viral
3.
J Virol ; 85(5): 2469-73, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21177819

RESUMO

Segment 8 of the influenza A virus codes for two proteins (NS1 and NS2/NEP) via splicing. Here, we developed a viral vector expressing a cytokine or chemokine instead of the interferon antagonist NS1. To achieve both the desired genetic stability and high transgene expression levels, NS2/NEP mRNA splicing efficacy had to be fine-tuned by modification of splicing elements. Expression levels of secreted foreign proteins could be further enhanced by fusing the N-terminal 13 amino acids of NS1 with an IgK-derived secretion signal peptide. Thus, the first start codon was used for translation initiation of both NS2/NEP and the foreign protein.


Assuntos
Vírus Defeituosos/genética , Expressão Gênica , Vetores Genéticos/genética , Vírus da Influenza A/genética , Splicing de RNA , Vírus Defeituosos/fisiologia , Vetores Genéticos/fisiologia , Vírus da Influenza A/fisiologia , Replicação Viral
4.
J Virol ; 85(21): 11139-45, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21849438

RESUMO

In general, antibiotics are not rated as substances that inhibit or support influenza virus replication. We describe here the enhancing effect of the polyene antibiotic amphotericin B (AmB) on influenza virus growth in Vero cells. We show that isolation rates of influenza A and B viruses from clinical samples can be dramatically enhanced by adding AmB to the culture medium. We demonstrate that AmB promotes the viral uptake and endocytic processing of the virus particles. This effect is specific for Vero and human nasal epithelial cells and was not observed in Madin-Darby canine kidney cells. The effect of AmB was subtype specific and more prominent for human seasonal influenza strains but absent for H5N1 human viruses. The AmB-enhancing effect seemed to be solely due to the viral hemagglutinin function. Our results indicate that the use of AmB may facilitate influenza virus isolation and production in Vero cells.


Assuntos
Anfotericina B/metabolismo , Antifúngicos/metabolismo , Vírus da Influenza A/efeitos dos fármacos , Vírus da Influenza A/crescimento & desenvolvimento , Vírus da Influenza B/efeitos dos fármacos , Vírus da Influenza B/crescimento & desenvolvimento , Replicação Viral/efeitos dos fármacos , Animais , Linhagem Celular , Chlorocebus aethiops , Cães , Endocitose/efeitos dos fármacos , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/virologia , Humanos , Vírus da Influenza A/isolamento & purificação , Vírus da Influenza B/isolamento & purificação , Influenza Humana/virologia
5.
J Infect Dis ; 201(3): 354-62, 2010 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-20039806

RESUMO

BACKGROUND. The nonstructural protein NS1 of influenza virus counteracts the interferon-mediated immune response of the host. By deleting the open reading frame of NS1, we have generated a novel type of influenza vaccine. We studied the safety and immunogenicity of an influenza strain lacking the NS1 gene (DeltaNS1-H1N1) in healthy volunteers. METHODS. Healthy seronegative adult volunteers were randomized to receive either a single intranasal dose of the DeltaNS1-H1N1 A/New Caledonia vaccine at 1 of 5 dose levels (6.4, 6.7, 7.0, 7.4, and 7.7 log(10) median tissue culture infective dose) (n = 36 recipients) or placebo (n = 12 recipients). RESULTS. Intranasal vaccination with the replication-deficient DeltaNS1-H1N1 vaccine was well tolerated. Rhinitis-like symptoms and headache were the most common adverse events identified during the 28-day observation period. Adverse events were similarly distributed between the treatment and placebo groups. Vaccine-specific local and serum antibodies were induced in a dose-dependent manner. In the highest dose group, vaccine-specific antibodies were detected in 10 of 12 volunteers. Importantly, the vaccine also induced neutralizing antibodies against heterologous drift variants. CONCLUSIONS. We show that vaccination with an influenza virus strain lacking the viral interferon antagonist NS1 induces statistically significant levels of strain-specific and cross-neutralizing antibodies despite the highly attenuated replication-deficient phenotype. Further studies are warranted to determine whether these results translate into protection from influenza virus infection. TRIAL REGISTRATION. ClinicalTrials.gov identifier: NCT00724997 .


Assuntos
Vírus da Influenza A Subtipo H1N1/imunologia , Vacinas contra Influenza/imunologia , Influenza Humana/prevenção & controle , Vacinas Atenuadas/imunologia , Proteínas não Estruturais Virais/genética , Adulto , Anticorpos Antivirais/sangue , Anticorpos Antivirais/isolamento & purificação , Relação Dose-Resposta Imunológica , Método Duplo-Cego , Deleção de Genes , Humanos , Vírus da Influenza A Subtipo H1N1/genética , Vacinas contra Influenza/administração & dosagem , Vacinas contra Influenza/efeitos adversos , Líquido da Lavagem Nasal/imunologia , Líquido da Lavagem Nasal/virologia , Vacinas Atenuadas/administração & dosagem , Vacinas Atenuadas/efeitos adversos , Eliminação de Partículas Virais
6.
PLoS One ; 16(11): e0260155, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34797850

RESUMO

Bovine papillomaviruses types 1 and 2 (BPV1, BPV2) commonly induce skin tumours termed sarcoids in horses and other equids. Sarcoids seriously compromise the health and welfare of affected individuals due to their propensity to resist treatment and reoccur in a more severe form. We have developed influenza (Flu) A and B virus vectors that harbour a truncated NS1 gene (iNS) assuring interferon induction and co-express shuffled BPV1 E6 and E7 antigens for sarcoid immunotherapy. In a safety trial involving 12 healthy horses, intradermal administration of iNSA/E6E7equ and iNSB/E6E7equ was well tolerated, with the only transient side effect being mild fever in four horses. Repeated screening of secretions and faeces by RT-PCR and plaque assay revealed no virus shedding, thus also confirming biological safety. In a patient trial involving 29 horses bearing BPV1-induced single or multiple sarcoids, at least one lesion per horse was intratumourally injected and then boosted with iNSA/E6E7equ and/or iNSB/E6E7equ. The treatment induced a systemic antitumour response as reflected by the synchronous regression of injected and non-injected lesions. Irrespective of vaccination schemes, complete tumour regression was achieved in 10/29 horses. In 10/29 horses, regression is still ongoing (May 2021). Intriguingly, scrapings collected from former tumour sites in two patients tested negative by BPV1 PCR. Nine severely affected individuals with a history of unsuccessful therapeutic attempts did not (6/29) or only transiently (3/29) respond to the treatment. INSA/E6E7equ and iNSB/E6E7equ proved safe and effective in significantly reducing the tumour burden even in severe cases.


Assuntos
Papillomavirus Bovino 1/imunologia , Neoplasias/imunologia , Neoplasias/virologia , Sarcoidose/patologia , Sarcoidose/virologia , Animais , Chlorocebus aethiops , Vetores de Doenças , Feminino , Cavalos , Humanos , Influenza Humana/imunologia , Masculino , Orthomyxoviridae/imunologia , Vacinação/métodos , Células Vero , Eliminação de Partículas Virais/imunologia
7.
Vaccine ; 37(28): 3722-3729, 2019 06 19.
Artigo em Inglês | MEDLINE | ID: mdl-31155415

RESUMO

BACKGROUND: Traditional inactivated influenza vaccines are the type of vaccines that were most frequently developed for immunization against the highly pathogenic avian H5N1 influenza virus. However, clinical trials with inactivated influenza vaccines for H5N1 indicated that high doses and at least two immunizations are required for an effective immune response (Nicholson et al., 2001; Treanor, Campbell et al., 2006; Treanor, Schiff et al., 2006; Ehrlich et al., 2008). We investigated the safety and immunogenicity of a live attenuated H5N1 vaccine (delNS1-H5N1) lacking the interferon antagonist nonstructural protein 1 (NS1). METHODS: We conducted a double-blind, placebo-controlled, phase 1 study in healthy adult participants who were randomly assigned at a 2:1 ratio to receive two immunizations of delNS1-H5N1 vaccine at 6.8 log10 50% tissue culture infectious doses (TCID50)/subject or 7.5 log10 TCID50/subject, or placebo. RESULTS: Intranasal vaccination with the live attenuated delNS1-H5N1 vaccine was safe and well tolerated. The most common adverse events identified were symptoms associated with mild influenza infections, such as increased body temperature (>37.0 °C), pharyngeal erythema, rhinitis and throat irritation, and were reported within 7 days after the first immunization. delNS1-H5N1 was able to induce significant vaccine-specific serum antibody titers even at the lower dose level of 6.8 log10 TCID50/subject. Seroconversion occurred in 75% of study participants after only one immunization with 7.5 log10 TCID50/subject. Vaccine-specific local IgA responses were observed in 41.7% of individuals that showed serum antibody responses after 2nd immunization. CONCLUSIONS: We show that vaccination with a live attenuated H5N1 influenza vaccine lacking NS1 is safe and induces significant levels of vaccine-specific antibodies even after one immunization. The safety and immunogenicity data indicate that delNS1-H5N1 has the potential to fulfil the unmet need for an effective influenza vaccine in pandemic situations. (ClinicalTrials.gov identifier NCT03745274).


Assuntos
Formação de Anticorpos/imunologia , Virus da Influenza A Subtipo H5N1/imunologia , Vacinas contra Influenza/imunologia , Influenza Humana/imunologia , Influenza Humana/prevenção & controle , Proteínas não Estruturais Virais/genética , Adolescente , Adulto , Anticorpos Antivirais/imunologia , Método Duplo-Cego , Feminino , Humanos , Imunoglobulina A/imunologia , Virus da Influenza A Subtipo H5N1/genética , Vacinas contra Influenza/genética , Masculino , Pessoa de Meia-Idade , Vacinação/métodos , Vacinas Atenuadas/genética , Vacinas Atenuadas/imunologia , Adulto Jovem
8.
Vet Comp Oncol ; 17(3): 211-220, 2019 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-30719836

RESUMO

Canine oral malignant melanoma (COMM) is a potentially lethal cancer disease. We established primary cell lines from mostly amelanotic primary COMM and metastases and assessed lesions and derived cells for Melan A, PNL2 and CD146 expression. Then, migration and invasion of CD146-enriched vs -depleted COMM cells were analysed. Epithelial-to-mesenchymal transition (EMT) was addressed by Vimentin-staining and MMP2/MMP9 zymography. Phagocytic behaviour was analysed by histopathological examination and phagocytosis assay. While Melan A- and PNL2-staining yielded inconsistent data, 100% of COMM sections and primary cells showed CD146 expression, suggesting that this protein may serve as a prognostic marker. An overall correlation between CD146-expression and migration/invasion was not observed. All primary cell lines consistently expressed Vimentin and secreted biologically active MMP2, indicating that they had undergone EMT. Importantly, COMM sections exhibited cell-in-cell structures, and all primary cell lines exhibited phagocytic activity, supporting the concept that cell cannibalism may have a role in COMM progression.


Assuntos
Doenças do Cão , Melanoma/veterinária , Células-Tronco Mesenquimais/fisiologia , Neoplasias Bucais/veterinária , Fagocitose/fisiologia , Animais , Antígeno CD146/genética , Antígeno CD146/metabolismo , Adesão Celular , Movimento Celular/fisiologia , Células Cultivadas , Cães , Regulação Neoplásica da Expressão Gênica/fisiologia , Metaloproteinase 2 da Matriz/genética , Metaloproteinase 2 da Matriz/metabolismo , Metaloproteinase 9 da Matriz/genética , Metaloproteinase 9 da Matriz/metabolismo , Invasividade Neoplásica , Vimentina/genética , Vimentina/metabolismo
9.
Oncogene ; 22(43): 6725-38, 2003 Oct 02.
Artigo em Inglês | MEDLINE | ID: mdl-14555986

RESUMO

Hepatocytes adopt an invasive and metastatic phenotype caused by the cooperation of transforming growth factor (TGF)-beta and oncogenic Ha-Ras. In the initial phase of this process, c-Fos is rapidly induced by TGF-beta, but then decreases to undetectable levels. Here, we investigated the functional implications of c-Fos activation and its contribution to hepatocellular tumorigenesis. By employing conditional c-Fos expression, we observed that continuous activation of c-Fos and consequently AP-1 activity leads to depolarization of differentiated murine epithelial hepatocytes. Most remarkably, this change in morphology was associated with inhibition of proliferation and induction of cell death. Coexpression of antiapoptotic Bcl-XL or scavenging of reactive oxygen species was sufficient to prevent the c-Fos-mediated phenotype. In contrast, the cooperation of c-Fos with oncogenic Ha-Ras or a Ras mutant selectively activating the MAPK pathway even enhanced c-Fos-induced effects. Showing the negative role in hepatocellular tumorigenesis, c-Fos repressed oncogenic Ras-driven anchorage-independent growth in vitro and strongly suppressed tumour formation in vivo. Taken together, we demonstrate that c-Fos modulates plasticity of epithelial hepatocytes and acts tumour suppressive in neoplastic hepatocytes by stimulating cell cycle inhibition and cell death.


Assuntos
Carcinoma Hepatocelular/metabolismo , Neoplasias Hepáticas/metabolismo , Proteínas Proto-Oncogênicas c-fos/metabolismo , Animais , Apoptose , Proteínas Estimuladoras de Ligação a CCAAT/metabolismo , Morte Celular , Divisão Celular , Linhagem Celular , Sobrevivência Celular , Transformação Celular Neoplásica , Células Epiteliais , Citometria de Fluxo , Genes ras , Sistema de Sinalização das MAP Quinases , Camundongos , Camundongos Endogâmicos BALB C , Fenótipo , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Espécies Reativas de Oxigênio , Transdução de Sinais , Fatores de Tempo , Fator de Transcrição AP-1/metabolismo , Fator de Transcrição CHOP , Fatores de Transcrição/metabolismo , Transfecção , Fator de Crescimento Transformador beta/metabolismo , Regulação para Cima , Proteína bcl-X
10.
Stem Cells Dev ; 14(5): 556-63, 2005 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16305340

RESUMO

Stem cell factor (SCF) stimulation of the receptor tyrosine kinase c-kit has effects on the proliferation, differentiation, and apoptotic regulation of hematopoietic progenitor cell populations. Rat bone marrow myelomonocytic stem cells (MSC) isolated in vitro by wheat germ agglutinin culture exclusively undergo self-renewal divisions when stimulated by SCF but bipotentially differentiate in the presence of dexamethasone or 1alpha,25-dihydroxyvitamin D(3) to granulocytes and macrophages, respectively. We show here that withdrawal of SCF from MSC induces rapid apoptosis in all stages of the cell cycle accompanied by development of an ultrastructural apoptotic morphology. To investigate immediate-early gene induction during MSC apoptosis, a differential display polymerase chain reaction (DD-PCR) screen coupled with rapid amplification of cDNA ends (RACE) PCR was performed. An immediate-early apoptosis response gene was isolated from growth factor-deprived MSC that was not expressed during self-renewal or differentiation induction cultures containing SCF. The protein contains a PEST region enriched in proline, glutamic acid, serine, and threonine residues common to proteins with a high turnover and has a cytoplasmic, vesicular localization in apoptotic MSC shown by immunohistochemistry. The human orthologous gene, isolated by RACE PCR, shows 86% homology to the rat protein and high similarity with a human uncharacterized hypothalamus predicted protein (HSMNP1) localized to the long arm of chromosome 20. Because deletions in this region are a common occurrence in a wide range of myeloproliferative disorders characterized by treatment resistance to apoptosis, HSMNP1 expression may play a role in normal and pathological myeloid development.


Assuntos
Apoptose/fisiologia , Cromossomos Humanos Par 20 , Células Precursoras Eritroides/fisiologia , Genes Precoces , Sequência de Aminoácidos , Animais , Diferenciação Celular/fisiologia , Células Precursoras Eritroides/ultraestrutura , Perfilação da Expressão Gênica , Humanos , Marcação In Situ das Extremidades Cortadas , Dados de Sequência Molecular , Ratos , Alinhamento de Sequência , Análise de Sequência de DNA , Fator de Células-Tronco/metabolismo
11.
Clin Cancer Res ; 10(12 Pt 1): 4185-91, 2004 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-15217956

RESUMO

PURPOSE: Little is known about the role that Mcl-1, an antiapoptotic Bcl-2 family member, plays in solid tumor biology and susceptibility to anticancer therapy. We observed that the Mcl-1 protein is widely expressed in human sarcoma cell lines of different histological origin (n = 7). Because the expression of antiapoptotic Bcl-2 family proteins can significantly contribute to the chemoresistance of human malignancies, we used an antisense strategy to address this issue in sarcoma. EXPERIMENTAL DESIGN: SCID mice (n = 6/group) received s.c. injections of SW872 liposarcoma cells. After development of palpable tumors, mice were treated by s.c.-implanted miniosmotic pumps prefilled with saline or antisense or universal control oligonucleotides (20 mg/kg/day for 2 weeks). On days 2, 6, and 10, mice were treated with low-dose cyclophosphamide (35 mg/kg i.p) or saline control. During the experiments, tumor weight was assessed twice weekly by caliper measurements. On day 14, animals were sacrificed. Tumors were weighed and fixed in formalin for immunohistochemistry and terminal deoxynucleotidyl transferase-mediated dUTP nick end-labeling analysis. RESULTS: Mcl-1 antisense oligonucleotides specifically reduced Mcl-1 protein expression but produced no reduction in tumor weight compared with saline-treated control animals. Cyclophosphamide monotreatment caused only modest tumor weight reduction compared with saline control. However, use of Mcl-1 antisense oligonucleotides combined with cyclophosphamide clearly enhanced tumor cell apoptosis and significantly reduced tumor weight by more than two-thirds compared with respective control treatments. CONCLUSION: A combination of Mcl-1 antisense oligonucleotides with low-dose cyclophosphamide provides a synergistic antitumor effect and might qualify as a promising strategy to overcome chemoresistance in human sarcoma.


Assuntos
Ciclofosfamida/uso terapêutico , Resistencia a Medicamentos Antineoplásicos , Proteínas de Neoplasias/metabolismo , Oligonucleotídeos Antissenso/uso terapêutico , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Sarcoma/tratamento farmacológico , Sarcoma/metabolismo , Animais , Apoptose , Western Blotting , Linhagem Celular Tumoral , Relação Dose-Resposta a Droga , Regulação para Baixo , Feminino , Humanos , Imuno-Histoquímica , Marcação In Situ das Extremidades Cortadas , Camundongos , Camundongos SCID , Proteína de Sequência 1 de Leucemia de Células Mieloides , Proteínas de Neoplasias/genética , Transplante de Neoplasias , Oligonucleotídeos/química , Proteínas Proto-Oncogênicas c-bcl-2/genética , Estaurosporina/farmacologia , Fatores de Tempo
12.
PLoS One ; 10(9): e0138722, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26381401

RESUMO

Persistent infection with high-risk human papillomavirus (HPV) types, most often HPV16 and HPV18, causes all cervical and most anal cancers, and a subset of vulvar, vaginal, penile and oropharyngeal carcinomas. Two prophylactic virus-like particle (VLPs)-based vaccines, are available that protect against vaccine type-associated persistent infection and associated disease, yet have no therapeutic effect on existing lesions or infections. We have generated recombinant live-attenuated influenza A viruses expressing the HPV16 oncogenes E6 and E7 as experimental immunotherapeutic vaccine candidates. The influenza A virus life cycle lacks DNA intermediates as important safety feature. Different serotypes were generated to ensure efficient prime and boost immunizations. The immune response to vaccination in C57BL/6 mice was characterized by peptide ELISA and IFN-γ ELISpot, demonstrating induction of cell-mediated immunity to HPV16 E6 and E7 oncoproteins. Prophylactic and therapeutic vaccine efficacy was analyzed in the murine HPV16-positive TC-1 tumor challenge model. Subcutaneous (s.c.) prime and boost vaccinations of mice with recombinant influenza A serotypes H1N1 and H3N2, followed by challenge with TC-1 cells resulted in complete protection or significantly reduced tumor growth as compared to control animals. In a therapeutic setting, s.c. vaccination of mice with established TC-1 tumors decelerated tumor growth and significantly prolonged survival. Importantly, intralesional vaccine administration induced complete tumor regression in 25% of animals, and significantly reduced tumor growth in 50% of mice. These results suggest recombinant E6E7 influenza viruses as a promising new approach for the development of a therapeutic vaccine against HPV-induced disease.


Assuntos
Vacinas Anticâncer/imunologia , Papillomavirus Humano 16/imunologia , Vírus da Influenza A/imunologia , Infecções por Papillomavirus/prevenção & controle , Vacinas contra Papillomavirus/imunologia , Vacinação , Animais , Feminino , Imunidade Celular/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Infecções por Papillomavirus/imunologia , Neoplasias do Colo do Útero/prevenção & controle , Neoplasias do Colo do Útero/virologia , Vacinas Atenuadas/imunologia
13.
J Invest Dermatol ; 120(6): 1081-6, 2003 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-12787138

RESUMO

It is well established that high expression of the antiapoptotic Bcl-2 family proteins Bcl-2 and Bcl-xL can significantly contribute to chemoresistance in a number of human malignancies. Much less is known about the role the more recently described Bcl-2 family member Mcl-1 might play in tumor biology and resistance to chemotherapy. Using an antisense strategy, we here address this issue in melanoma, a paradigm of a treatment-resistant malignancy. After in vitro proof of principle supporting an antisense mechanism of action with specific reduction of Mcl-1 protein as a consequence of nuclear uptake of the Mcl-1 antisense oligonucleotides employed, antisense and universal control oligonucleotides were administered systemically in combination with dacarbazine in a human melanoma SCID mouse xenotransplantation model. Dacarbazine, available now for more than three decades, still remains the most active single agent for treatment of advanced melanoma. Mcl-1 antisense oligonucleotides specifically reduced target protein expression as well as the apoptotic threshold of melanoma xenotransplants. Combined Mcl-1 antisense oligonucleotide plus dacarbazine treatment resulted in enhanced tumor cell apoptosis and led to a significantly reduced mean tumor weight (mean 0.16 g, 95% confidence interval 0.08-0.26) compared to the tumor weight in universal control oligonucleotide plus dacarbazine treated animals (mean 0.35 g, 95% confidence interval 0.2-0.44) or saline plus dacarbazine treated animals (mean 0.39 g, 95% confidence interval 0.25-0.53). We thus show that Mcl-1 is an important factor contributing to the chemoresistance of human melanoma in vivo. Antisense therapy against the Mcl-1 gene product, possibly in combination with antisense strategies targeting other antiapoptotic Bcl-2 family members, appears to be a rational and promising approach to help overcome treatment resistance of malignant melanoma.


Assuntos
Melanoma/tratamento farmacológico , Oligonucleotídeos Antissenso/uso terapêutico , Proteínas Proto-Oncogênicas c-bcl-2 , Neoplasias Cutâneas/tratamento farmacológico , Animais , Antineoplásicos Alquilantes/uso terapêutico , Apoptose , Dacarbazina/uso terapêutico , Sinergismo Farmacológico , Quimioterapia Combinada , Humanos , Imuno-Histoquímica , Marcação In Situ das Extremidades Cortadas , Melanoma/metabolismo , Melanoma/fisiopatologia , Camundongos , Camundongos SCID , Proteína de Sequência 1 de Leucemia de Células Mieloides , Proteínas de Neoplasias/antagonistas & inibidores , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Transplante de Neoplasias , Oligonucleotídeos Antissenso/farmacocinética , Neoplasias Cutâneas/metabolismo , Neoplasias Cutâneas/fisiopatologia , Transplante Heterólogo , Células Tumorais Cultivadas
14.
Cancer Gene Ther ; 9(8): 673-80, 2002 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-12136428

RESUMO

We have recently developed surface-shielded transferrin-polyethylenimine (Tf-PEI)/DNA delivery systems that target reporter gene expression to distant tumors after systemic application. In the present study, we used surface-shielded Tf-PEI/DNA complexes for delivering the gene for a highly potent cytokine, tumor necrosis factor-alpha (TNFalpha). TNFalpha is known for its ability to induce hemorrhagic tumor necrosis and tumor regression. However, the therapeutic application of TNFalpha is hampered by its high systemic toxicity dictating the need to target TNFalpha activity to the tumor. Systemic application of surface-shielded Tf-PEI complexes with the TNFalpha gene resulted in preferential expression of TNFalpha in the tumor without detectable TNFalpha serum levels, in contrast to the application of nontargeted complexes. Tumor-targeted TNFalpha gene delivery induced pronounced hemorrhagic tumor necrosis and inhibition of tumor growth in three murine tumor models of different tissue origins, Neuro2a neuroblastoma, MethA fibrosarcoma, and M-3 melanoma, with complete tumor regressions observed in the MethA model. No systemic TNF-related toxicity was observed due to the localization of the TNFalpha activity to the tumor. Targeted gene therapy may be an attractive strategy applicable to highly active, yet toxic, molecules such as TNFalpha.


Assuntos
Neuroblastoma/patologia , Fator de Necrose Tumoral alfa/genética , Animais , Fibrossarcoma/genética , Fibrossarcoma/patologia , Terapia Genética , Melanoma Experimental/genética , Melanoma Experimental/patologia , Camundongos , Neuroblastoma/genética , Neuroblastoma/metabolismo , Neuroblastoma/terapia , Células Tumorais Cultivadas , Fator de Necrose Tumoral alfa/metabolismo
15.
J Control Release ; 91(1-2): 173-81, 2003 Aug 28.
Artigo em Inglês | MEDLINE | ID: mdl-12932649

RESUMO

Surface-shielded DNA delivery systems have been synthesized with virus-like characteristics that target gene expression into distant tumor tissues. Polyethylenimine (PEI)/DNA complexes ('polyplexes') conjugated with the cell-binding ligand transferrin (Tf) or epidermal growth factor (EGF) were used to achieve receptor-mediated endocytosis. The surface charge of the complexes was masked by covalently linking PEI to polyethylene glycol (PEG). Three alternatives for generating these surface-shielded formulations were utilized, attaching ligand and PEG molecules to PEI either before or after DNA complex formation. The stabilized formulations could be ultra-concentrated, stored frozen, and applied systemically after thawing. Intravenous injection of Tf-PEG-coated polyplexes resulted in gene transfer to subcutaneous Neuro2a neuroblastoma tumors of syngeneic A/J mice; EGF-PEG-coated polyplexes were intravenously applied for targeting human hepatocellular carcinoma xenografts in SCID mice. In these models, luciferase marker gene expression levels in tumor tissues were 10- to 100-fold higher than in other organ tissues. Repeated systemic application of Tf-PEG-PEI/DNA complexes encoding tumor necrosis factor alpha (TNF-alpha) into tumor-bearing mice induced tumor necrosis and inhibition of tumor growth in three murine tumor models of different tissue origin (Neuro2a, M-3 or B16 melanoma).


Assuntos
DNA/administração & dosagem , Terapia Genética/métodos , Polietilenoglicóis/química , Polietilenoimina/química , Animais , Fenômenos Químicos , Físico-Química , DNA/química , Fator de Crescimento Epidérmico/química , Excipientes , Feminino , Congelamento , Marcação de Genes , Humanos , Indicadores e Reagentes , Ligantes , Camundongos , Camundongos Endogâmicos A , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Transplante de Neoplasias , Tamanho da Partícula , Transfecção , Células Tumorais Cultivadas , Fator de Necrose Tumoral alfa/genética , Ultrafiltração
16.
J Soc Gynecol Investig ; 10(7): 412-7, 2003 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-14519482

RESUMO

OBJECTIVE: Changes in serum levels of angiogenic factors such as vascular endothelial growth factor or placental growth factor were associated with preeclampsia. The aim of our study was to investigate the concentrations of endostatin, a potent endogenous inhibitor of angiogenesis and endothelial or tumor cell growth, in preeclamptic patients. METHODS: Levels of soluble endostatin were determined by enzyme-linked immunosorbent assay and Western blot analysis in sera of nonpregnant women, healthy pregnant women, and patients with different forms of preeclampsia. RESULTS: Statistical analyses of age-matched study groups revealed elevated medians of endostatin concentration for severely (30.5 ng/mL) and mildly (26.05 ng/mL) preeclamptic patients compared with healthy pregnant (17.2 ng/mL) and nonpregnant women (17.2 ng/mL). Western blot analysis confirmed the up-regulation of soluble endostatin molecules (24 kD) in sera of severely preeclamptic patients. CONCLUSION: Elevated concentrations of endostatin could play a role in the pathophysiology of preeclampsia by counteracting the effects of vascular endothelial growth factor. The inhibitor could also be responsible for the observed growth-inhibitory effects of preeclamptic plasma on endothelial cells.


Assuntos
Endostatinas/sangue , Pré-Eclâmpsia/sangue , Adulto , Western Blotting , Ensaio de Imunoadsorção Enzimática , Feminino , Idade Gestacional , Humanos , Modelos Logísticos , Gravidez , Valores de Referência
17.
Eur J Gastroenterol Hepatol ; 15(2): 119-26, 2003 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-12560754

RESUMO

OBJECTIVE: CD40, a member of the tumour necrosis factor receptor family, plays a major role in adaptive immune responses and contributes to cancer surveillance. Conflicting results have been reported recently on the expression and function of CD40 in carcinomas. The aim of the present study was to investigate the role of CD40 in human hepatoma. DESIGN/METHODS: CD40 expression was examined in hepatomas and derived cell lines by immunohistochemistry, flow cytometry and reverse transcriptase polymerase chain reaction. We investigated in hepatoma cell lines the regulation of CD40 by pro-inflammatory cytokines and the effects of its ligation with soluble CD40L on the expression of co-stimulatory and pro-apoptotic cell-surface molecules and survival. RESULTS: CD40 was detected with a similar frequency of about 40% in hepatoma specimens and derived cell lines but not in normal hepatocytes. Tumour necrosis factor alpha and its combination with interferon gamma upregulated CD40 only in intrinsically positive cell lines. CD40 ligation had no effect on cell viability or surface expression of CD54, CD80, CD86 or CD95. CONCLUSIONS: CD40 is expressed variably in human hepatoma and enhanced by distinct pro-inflammatory cytokines. The lack of detectable effects of CD40 ligation does not support a major role of this molecule in hepatocellular carcinoma biology.


Assuntos
Antígenos CD40/fisiologia , Carcinoma Hepatocelular/metabolismo , Neoplasias Hepáticas/metabolismo , Adulto , Idoso , Antígenos CD40/análise , Carcinoma Hepatocelular/imunologia , Carcinoma Hepatocelular/patologia , Feminino , Citometria de Fluxo/métodos , Humanos , Interferon gama/metabolismo , Neoplasias Hepáticas/imunologia , Neoplasias Hepáticas/patologia , Masculino , Pessoa de Meia-Idade , Reação em Cadeia da Polimerase Via Transcriptase Reversa/métodos , Células Tumorais Cultivadas , Fator de Necrose Tumoral alfa/metabolismo , Regulação para Cima
18.
Vaccine ; 31(52): 6194-200, 2013 Dec 16.
Artigo em Inglês | MEDLINE | ID: mdl-24183981

RESUMO

BACKGROUND: The non-structural protein NS1 of the influenza virus counteracts the interferon-mediated immune response of the host. We investigated the safety and immunogenicity of a trivalent formulation containing influenza H1N1, H3N2 and B strains lacking NS1 (delNS1-trivalent). METHODS: Healthy adult study participants who were seronegative for at least one strain present in the vaccine formulation were randomized to receive a single intranasal dose of delNS1-trivalent vaccine at 7.0log10 TCID50/subject (n=39) or placebo (n=41). RESULTS: Intranasal vaccination with the live replication-deficient delNS1-trivalent vaccine was well tolerated with no treatment-related serious adverse events. The most common adverse events identified, i.e. headache, oropharyngeal pain and rhinitis-like symptoms, were mainly mild and transient and distributed similarly in the treatment and placebo groups. Significant vaccine-specific immune responses were induced. Pre-existing low antibody titers or seronegativity for the corresponding vaccine strain yielded better response rates. CONCLUSIONS: We show that vaccination with a replication-deficient trivalent influenza vaccine containing H1N1, H3N2 and B strains lacking NS1 is safe and induces significant levels of antibodies (ClinicalTrials.gov identifier NCT01369862).


Assuntos
Vacinas contra Influenza/efeitos adversos , Vacinas contra Influenza/imunologia , Proteínas não Estruturais Virais/deficiência , Administração Intranasal , Adolescente , Adulto , Anticorpos Antivirais/sangue , Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos/epidemiologia , Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos/patologia , Feminino , Voluntários Saudáveis , Humanos , Vírus da Influenza A Subtipo H1N1/genética , Vírus da Influenza A Subtipo H1N1/imunologia , Vírus da Influenza A Subtipo H3N2/genética , Vírus da Influenza A Subtipo H3N2/imunologia , Vírus da Influenza B/genética , Vírus da Influenza B/imunologia , Vacinas contra Influenza/administração & dosagem , Masculino , Placebos/administração & dosagem , Vacinas Atenuadas/administração & dosagem , Vacinas Atenuadas/imunologia
19.
PLoS One ; 7(5): e36506, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22563505

RESUMO

Oncolytic influenza A viruses with deleted NS1 gene (delNS1) replicate selectively in tumour cells with defective interferon response and/or activated Ras/Raf/MEK/ERK signalling pathway. To develop a delNS1 virus with specific immunostimulatory properties, we used an optimised technology to insert the interleukin-15 (IL-15) coding sequence into the viral NS gene segment (delNS1-IL-15). DelNS1 and delNS1-IL-15 exerted similar oncolytic effects. Both viruses replicated and caused caspase-dependent apoptosis in interferon-defective melanoma cells. Virus replication was required for their oncolytic activity. Cisplatin enhanced the oncolytic activity of delNS1 viruses. The cytotoxic drug increased delNS1 replication and delNS1-induced caspase-dependent apoptosis. Interference with MEK/ERK signalling by RNAi-mediated depletion or the MEK inhibitor U0126 did not affect the oncolytic effects of the delNS1 viruses. In oncolysis sensitive melanoma cells, delNS1-IL-15 (but not delNS1) infection resulted in the production of IL-15 levels ranging from 70 to 1140 pg/mL in the cell culture supernatants. The supernatants of delNS1-IL-15-infected (but not of delNS1-infected) melanoma cells induced primary human natural killer cell-mediated lysis of non-infected tumour cells. In conclusion, we constructed a novel oncolytic influenza virus that combines the oncolytic activity of delNS1 viruses with immunostimulatory properties through production of functional IL-15. Moreover, we showed that the oncolytic activity of delNS1 viruses can be enhanced in combination with cytotoxic anti-cancer drugs.


Assuntos
Vírus da Influenza A Subtipo H1N1/genética , Interleucina-15/genética , Vírus Oncolíticos/genética , Proteínas não Estruturais Virais/genética , Clorometilcetonas de Aminoácidos/farmacologia , Animais , Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Western Blotting , Inibidores de Caspase , Caspases/metabolismo , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Chlorocebus aethiops , Cisplatino/farmacologia , Inibidores de Cisteína Proteinase/farmacologia , Deleção de Genes , Humanos , Vírus da Influenza A Subtipo H1N1/crescimento & desenvolvimento , Interleucina-15/metabolismo , Melanoma/genética , Melanoma/patologia , Melanoma/virologia , Proteína Quinase 1 Ativada por Mitógeno/genética , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/genética , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Vírus Oncolíticos/crescimento & desenvolvimento , Interferência de RNA , Células Vero , Replicação Viral/efeitos dos fármacos
20.
Vaccine ; 29(19): 3517-24, 2011 Apr 27.
Artigo em Inglês | MEDLINE | ID: mdl-21406268

RESUMO

The isolation and cultivation of human influenza viruses in embryonated hen eggs or cell lines often leads to amino acid substitutions in the haemagglutinin (HA) molecule. We found that the propagation of influenza A H3N2 viruses on Vero cells may trigger the appearance of HA destabilising mutations, affecting viral resistance to low pH or high temperature treatment. Two ΔNS1 reassortants, containing the HA sequences identical to the original human H3N2 influenza virus isolates were constructed. Passages of these viruses on Vero cells led to the appearance of single mutations in the HA(1) L194P or HA(2) G75R subunits that impaired virus stability. The original HA sequences and the stable phenotypes of the primary isolates were preserved if reassortants were passaged by infection at pH 5.6 and cultivation in medium at pH 6.5. Corresponding ΔNS1 reassortants were compared for their immunogenicity in ferrets upon intranasal immunisation. Vaccine candidates containing HA mutations demonstrated significantly lower immunogenicity compared to those without mutations. Thus, the retaining of the original HA sequences of human viruses during vaccine production might be crucial for the efficacy of live attenuated influenza vaccines.


Assuntos
Glicoproteínas de Hemaglutininação de Vírus da Influenza/genética , Vírus da Influenza A Subtipo H3N2/genética , Administração Intranasal , Animais , Anticorpos Neutralizantes/sangue , Anticorpos Antivirais/sangue , Linhagem Celular , Chlorocebus aethiops , Feminino , Furões , Glicoproteínas de Hemaglutininação de Vírus da Influenza/imunologia , Concentração de Íons de Hidrogênio , Imunidade Humoral , Imunização , Vírus da Influenza A Subtipo H3N2/imunologia , Vacinas contra Influenza/genética , Vacinas contra Influenza/imunologia , Masculino , Mutação , Vírus Reordenados/genética , Vírus Reordenados/imunologia , Células Vero
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa