Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
Hum Gene Ther ; 26(3): 134-44, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25557131

RESUMO

For long it has been recognized that tumor necrosis factor alpha (TNFa) has anticancer characteristics, and its use as a cancer therapeutic was proposed already in the 1980s. However, its systemic toxicity has limited its usability. Oncolytic viruses, selectively cancer-killing viruses, have shown great potency, and one of their most useful aspects is their ability to produce high amounts of transgene products locally, resulting in high local versus systemic concentrations. Therefore, the overall magnitude of tumor cell killing results from the combination of oncolysis, transgene-mediated direct effect such as TNFa-mediated apoptosis, and, perhaps most significantly, from activation of the host immune system against the tumor. We generated a novel chimeric oncolytic adenovirus expressing human TNFa, Ad5/3-D24-hTNFa, whose efficacy and immunogenicity were tested in vitro and in vivo. The hTNFa-expressing adenovirus showed increased cancer-eradicating potency, which was shown to be because of elevated apoptosis and necrosis rates and induction of various immune responses. Interestingly, we saw increase in immunogenic cell death markers in Ad5/3-d24-hTNFa-treated cells. Moreover, tumors treated with Ad5/3-D24-hTNFa displayed enhanced presence of OVA-specific cytotoxic T cells. We thus can conclude that tumor eradication and antitumor immune responses mediated by Ad5/3-d24-hTNFa offer a new potential drug candidate for cancer therapy.


Assuntos
Adenoviridae/genética , Terapia Genética/métodos , Vetores Genéticos/uso terapêutico , Neoplasias/genética , Neoplasias/terapia , Terapia Viral Oncolítica/métodos , Fator de Necrose Tumoral alfa/metabolismo , Apoptose/imunologia , Linhagem Celular Tumoral , Vetores Genéticos/imunologia , Células HEK293 , Humanos , Neoplasias/imunologia , Estatísticas não Paramétricas , Linfócitos T Citotóxicos/imunologia , Fator de Necrose Tumoral alfa/genética
2.
Duodecim ; 130(8): 823-31, 2014.
Artigo em Finlandês | MEDLINE | ID: mdl-24822333

RESUMO

INTRODUCTION: The aim was to elucidate the costs and clinical results of sterilization. MATERIAL AND METHODS: A retrospective analysis was carried out on sterilizations conducted at the Hyvinkää hospital in 2006 to 2007 by tubal ligation with clips and by microimplants. RESULTS: Total costs obtained for microimplant sterilization per patient were 1,146 Euros and for clip sterilization 1,712 Euros. Postoperative pain was significantly less in the microimplant group, and adverse effects associated with the procedure were more common in the clip sterilization group. CONCLUSIONS: Microimplant sterilization performed on an outpatient basis is more cost-effective than laparoscopic clip sterilization.


Assuntos
Histeroscopia/economia , Laparoscopia/economia , Esterilização Tubária/economia , Esterilização Tubária/métodos , Análise Custo-Benefício , Feminino , Finlândia , Humanos , Dor Pós-Operatória/economia , Estudos Retrospectivos , Esterilização Tubária/efeitos adversos
3.
PLoS One ; 7(3): e32871, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22412937

RESUMO

Oncolytic adenoviruses can be engineered for better tumor selectivity, gene delivery and be armed for imaging and concentrating radionuclides into tumors for synergistic oncolysis. We constructed Ad5/3-hTERT-hNIS where replication is controlled by hTERT-promoter. Ad5/3-hTERT-hNIS expresses hNIS for imaging of transgene expression and for treatment of infected tumors by radioiodine. Ad5/3-hTERT-hNIS efficiently killed prostate cancer cells and induced iodine uptake in vitro and in vivo after intratumoral virus administration. Survival of mice treated with intravenous Ad5/3-hTERT-hNIS significantly prolonged survival over mock or radioiodine only but the combination of virus with radioiodine was not more effective than virus alone. Temporal and spatial changes in hNIS-expression during therapy were detected with SPECT, demonstrating feasibility of evaluation of the combination therapy with hNIS-expressing adenoviruses and radioiodide.


Assuntos
Adenoviridae/genética , Vetores Genéticos/genética , Radioisótopos do Iodo/metabolismo , Imagem Multimodal , Vírus Oncolíticos/genética , Tomografia por Emissão de Pósitrons , Simportadores/genética , Tomografia Computadorizada por Raios X , Animais , Linhagem Celular , Expressão Gênica , Terapia Genética , Vetores Genéticos/administração & dosagem , Humanos , Injeções Intravenosas , Radioisótopos do Iodo/uso terapêutico , Masculino , Camundongos , Camundongos Nus , Imagem Molecular , Terapia Viral Oncolítica , Neoplasias da Próstata/genética , Neoplasias da Próstata/mortalidade , Neoplasias da Próstata/terapia , Simportadores/metabolismo , Resultado do Tratamento
4.
Int J Radiat Oncol Biol Phys ; 83(1): 376-84, 2012 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-22019240

RESUMO

PURPOSE: In the present study, we evaluated the combination of replication-deficient adenoviruses and radiotherapy in vitro. The purpose of the present study was to analyze the mechanism of radiation-mediated upregulation of adenoviral transgene expression. METHODS AND MATERIALS: Adenoviral transgene expression (luciferase or green fluorescent protein) was studied with and without radiation in three cell lines: breast cancer M4A4-LM3, prostate cancer PC-3MM2, and lung cancer LNM35/enhanced green fluorescent protein. The effect of the radiation dose, modification of the viral capsid, and five different transgene promoters were studied. The cellular responses were studied using mass spectrometry and immunofluorescence analysis. Double strand break repair was modulated by inhibitors of heat shock protein 90, topoisomerase-I, and DNA protein kinase, and transgene expression was measured. RESULTS: We found that a wide range of radiation doses increased adenoviral transgene expression regardless of the cell line, transgene, promoter, or viral capsid modification. Treatment with adenovirus, radiation, and double strand break repair inhibitors resulted in persistence of double strand breaks and subsequent increases in adenovirus transgene expression. CONCLUSIONS: Radiation-induced enhancement of adenoviral transgene expression is linked to DNA damage recognition and repair. Radiation induces a global cellular response that results in increased production of RNA and proteins, including adenoviral transgene products. This study provides a mechanistic rationale for combining radiation with adenoviral gene delivery.


Assuntos
Adenovírus Humanos/efeitos da radiação , Reparo do DNA , Vírus Defeituosos/efeitos da radiação , Expressão Gênica/efeitos da radiação , Terapia Genética/métodos , Vetores Genéticos/efeitos da radiação , Transgenes/efeitos da radiação , Adenovírus Humanos/genética , Linhagem Celular Tumoral , Terapia Combinada/métodos , Quebras de DNA de Cadeia Dupla , Vírus Defeituosos/genética , Expressão Gênica/genética , Regulação Viral da Expressão Gênica/genética , Regulação Viral da Expressão Gênica/efeitos da radiação , Vetores Genéticos/genética , Proteínas de Fluorescência Verde/genética , Proteínas de Choque Térmico HSP90/antagonistas & inibidores , Proteínas de Choque Térmico HSP90/metabolismo , Humanos , Luciferases/genética , Inibidores de Proteínas Quinases/farmacologia , Doses de Radiação , Inibidores da Topoisomerase I/farmacologia , Transgenes/genética , Regulação para Cima/genética , Regulação para Cima/efeitos da radiação , Replicação Viral/genética , Replicação Viral/efeitos da radiação
5.
Mol Ther ; 19(9): 1737-46, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21673660

RESUMO

Patients with advanced solid tumors refractory to and progressing after conventional therapies were treated with three different regimens of low-dose cyclophosphamide (CP) in combination with oncolytic adenovirus. CP was given with oral metronomic dosing (50 mg/day, N = 21), intravenously (single 1,000 mg dose, N = 7) or both (N = 7). Virus was injected intratumorally. Controls (N = 8) received virus without CP. Treatments were well tolerated and safe regardless of schedule. Antibody formation and virus replication were not affected by CP. Metronomic CP (oral and oral + intravenous schedules) decreased regulatory T cells (T(regs)) without compromising induction of antitumor or antiviral T-cell responses. Oncolytic adenovirus given together with metronomic CP increased cytotoxic T cells and induced Th1 type immunity on a systemic level in most patients. All CP regimens resulted in higher rates of disease control than virus only (all P < 0.0001) and the best progression-free (PFS) and overall survival (OS) was seen in the oral + intravenous group. One year PFS and OS were 53 and 42% (P = 0.0016 and P < 0.02 versus virus only), respectively, both which are unusually high for chemotherapy refractory patients. We conclude that low-dose CP results in immunological effects appealing for oncolytic virotherapy. While these first-in-human data suggest good safety, intriguing efficacy and extended survival, the results should be confirmed in a randomized trial.


Assuntos
Antineoplásicos/administração & dosagem , Ciclofosfamida/administração & dosagem , Neoplasias/tratamento farmacológico , Terapia Viral Oncolítica/métodos , Linfócitos T Reguladores/imunologia , Adenoviridae/genética , Adolescente , Adulto , Idoso , Animais , Antineoplásicos/imunologia , Antineoplásicos/uso terapêutico , Linhagem Celular Tumoral , Criança , Terapia Combinada , Cricetinae , Ciclofosfamida/imunologia , Ciclofosfamida/uso terapêutico , Relação Dose-Resposta a Droga , Esquema de Medicação , Feminino , Vetores Genéticos , Humanos , Masculino , Mesocricetus , Pessoa de Meia-Idade , Neoplasias/imunologia , Resultado do Tratamento , Adulto Jovem
7.
Mol Pharm ; 8(1): 93-103, 2011 Feb 07.
Artigo em Inglês | MEDLINE | ID: mdl-20964369

RESUMO

Oncolytic adenoviruses are an emerging treatment option for advanced and refractory cancer. Such patients are often treated with corticosteroids to ameliorate tumor associated symptoms. Thus, it is important to evaluate whether safety is affected by immunosuppression possibly induced by corticosteroids. Concurrent low-dose cyclophosphamide, appealing for its immunomodulatory effects, could also impact safety. In a retrospective case-control study, we evaluated the effect of systemic corticosteroid use in cancer patients receiving oncolytic virotherapy. Four treatment groups were identified: (1) oncolytic adenovirus with oral glucocorticoids, (2) virus alone, (3) virus with glucocorticoids and cyclophosphamide and (4) virus with cyclophosphamide. Adverse events, neutralizing antibody titers, viral DNA in circulation and tumor responses were evaluated. The most common adverse effects were grade 1-2 fatigue, nausea, fever and abdominal pain. Common asymptomatic findings included self-limiting grade 1-3 hyponatremia and aspartate aminotransferase increase. Safety was good and no significant differences were observed between the groups. All patients had an increase in neutralizing antibody titers post-treatment, and no trends for differences between groups were observed. There were fewer post-treatment virus genomes circulating in patients receiving glucocorticoids when compared to their control groups. Overall, glucocorticoid use in cancer patients receiving oncolytic adenovirus, with or without low-dose cyclophosphamide, seems safe.


Assuntos
Adenoviridae/genética , Glucocorticoides/uso terapêutico , Neoplasias/tratamento farmacológico , Neoplasias/terapia , Terapia Viral Oncolítica/métodos , Adulto , Idoso , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Terapia Viral Oncolítica/efeitos adversos , Estudos Retrospectivos , Resultado do Tratamento
8.
Int J Radiat Oncol Biol Phys ; 78(4): 1201-9, 2010 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-20832189

RESUMO

PURPOSE: Radiotherapy is widely used for treatment of many tumor types, but it can damage normal tissues. It has been proposed that cancer cells can be selectively sensitized to radiation by adenovirus replication or by using radiosensitizing transgenes. Adenoviral proteins E1B55K, E4orf3, and E4orf6 play a role in radiosensitization, by targeting the Mre11, Rad50, and NBS1 complex (MRN) and inhibiting DNA double-strand break (DSB) repair. We hypothesize that combined with irradiation, these adenoviral proteins increase cell killing through the impairment of DSB repair. METHODS AND MATERIALS: We assessed the radiosensitizing/additive potential of replication-deficient adenoviruses expressing E1B55K, E4orf3, and E4orf6 proteins. Combination treatments with low-dose external photon beam radiotherapy were studied in prostate cancer (PC-3MM2 and DU-145), breast cancer (M4A4-LM3), and head and neck cancer (UT-SCC8) cell lines. We further demonstrated radiosensitizing or additive effects in mice with PC-3MM2 tumors. RESULTS: We show enhanced cell killing with adenovirus and radiation combination treatment. Co-infection with several of the viruses did not further increase cell killing, suggesting that both E4orf6 and E4orf3 are potent in MRN inhibition. Our results show that adenoviral proteins E4orf3 and E4orf6, but not E1B55K, are effective also in vivo. Enhanced cell killing was due to inhibition of DSB repair resulting in persistent double-strand DNA damage, indicated by elevated phospho-H2AX levels at 24 h after irradiation. CONCLUSIONS: This knowledge can be applied for improving the treatment of malignant tumors, such as prostate cancer, for development of more effective combination therapies and minimizing radiation doses and reducing side effects.


Assuntos
Proteínas E4 de Adenovirus/fisiologia , Adenovírus Humanos/metabolismo , Neoplasias/radioterapia , Tolerância a Radiação/fisiologia , Proteínas Virais/fisiologia , Proteínas E4 de Adenovirus/metabolismo , Adenovírus Humanos/genética , Animais , Neoplasias da Mama/radioterapia , Linhagem Celular Tumoral , Quebras de DNA de Cadeia Dupla , Reparo do DNA , Feminino , Neoplasias de Cabeça e Pescoço/radioterapia , Histonas/metabolismo , Humanos , Masculino , Camundongos , Camundongos Nus , Neoplasias/genética , Neoplasias/metabolismo , Neoplasias/virologia , Neoplasias da Próstata/radioterapia , Dosagem Radioterapêutica , Distribuição Aleatória , Proteínas Virais/metabolismo
9.
Mol Ther ; 18(10): 1874-84, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20664527

RESUMO

Augmenting antitumor immunity is a promising way to enhance the potency of oncolytic adenoviral therapy. Granulocyte-macrophage colony-stimulating factor (GMCSF) can mediate antitumor effects by recruiting natural killer cells and by induction of tumor-specific CD8(+) cytotoxic T-lymphocytes. Serotype 5 adenoviruses (Ad5) are commonly used in cancer gene therapy. However, expression of the coxsackie-adenovirus receptor is variable in many advanced tumors and preclinical data have demonstrated an advantage for replacing the Ad5 knob with the Ad3 knob. Here, a 5/3 capsid chimeric and p16-Rb pathway selective oncolytic adenovirus coding for GMCSF was engineered and tested preclinically. A total of 21 patients with advanced solid tumors refractory to standard therapies were then treated intratumorally and intravenously with Ad5/3-D24-GMCSF, which was combined with low-dose metronomic cyclophosphamide to reduce regulatory T cells. No severe adverse events occurred. Analysis of pretreatment samples of malignant pleural effusion and ascites confirmed the efficacy of Ad5/3-D24-GMCSF in transduction and cell killing. Evidence of biological activity of the virus was seen in 13/21 patients and 8/12 showed objective clinical benefit as evaluated by radiology with Response Evaluation Criteria In Solid Tumors (RECIST) criteria. Antiadenoviral and antitumoral immune responses were elicited after treatment. Thus, Ad5/3-D24-GMCSF seems safe in treating cancer patients and promising signs of efficacy were seen.


Assuntos
Adenoviridae/genética , Terapia Genética/métodos , Fator Estimulador de Colônias de Granulócitos e Macrófagos/metabolismo , Neoplasias/terapia , Terapia Viral Oncolítica/métodos , Adolescente , Adulto , Idoso , Animais , Linhagem Celular , Linhagem Celular Tumoral , Cricetinae , Ciclofosfamida/uso terapêutico , Feminino , Fator Estimulador de Colônias de Granulócitos e Macrófagos/genética , Humanos , Imunossupressores/uso terapêutico , Masculino , Mesocricetus , Pessoa de Meia-Idade , Neoplasias/tratamento farmacológico , Ensaios Antitumorais Modelo de Xenoenxerto , Adulto Jovem
10.
Cancer Res ; 70(11): 4297-309, 2010 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-20484030

RESUMO

Granulocyte macrophage colony-stimulating factor (GMCSF) can mediate antitumor effects by recruiting natural killer cells and by induction of tumor-specific cytotoxic T-cells through antigen-presenting cells. Oncolytic tumor cell-killing can produce a potent costimulatory danger signal and release of tumor epitopes for antigen-presenting cell sampling. Therefore, an oncolytic adenovirus coding for GMCSF was engineered and shown to induce tumor-specific immunity in an immunocompetent syngeneic hamster model. Subsequently, 20 patients with advanced solid tumors refractory to standard therapies were treated with Ad5-D24-GMCSF. Of the 16 radiologically evaluable patients, 2 had complete responses, 1 had a minor response, and 5 had disease stabilization. Responses were frequently seen in injected and noninjected tumors. Treatment was well tolerated and resulted in the induction of both tumor-specific and virus-specific immunity as measured by ELISPOT and pentamer analysis. This is the first time that oncolytic virus-mediated antitumor immunity has been shown in humans. Ad5-D24-GMCSF is promising for further clinical testing.


Assuntos
Adenoviridae/genética , Fator Estimulador de Colônias de Granulócitos/genética , Imunoterapia/métodos , Neoplasias/terapia , Terapia Viral Oncolítica/métodos , Adenoviridae/imunologia , Adenoviridae/metabolismo , Animais , Cricetinae , Epitopos de Linfócito T/imunologia , Fator Estimulador de Colônias de Granulócitos/biossíntese , Fator Estimulador de Colônias de Granulócitos/imunologia , Humanos , Proteínas Inibidoras de Apoptose , Proteínas Associadas aos Microtúbulos/imunologia , Neoplasias/genética , Neoplasias/imunologia , Neoplasias/virologia , Survivina , Linfócitos T/imunologia , Transfecção
11.
Clin Cancer Res ; 16(11): 3035-43, 2010 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-20501623

RESUMO

PURPOSE: Twenty-one patients with cancer were treated with a single round of oncolytic adenovirus ICOVIR-7. EXPERIMENTAL DESIGN: ICOVIR-7 features an RGD-4C modification of the fiber HI-loop of serotype 5 adenovirus for enhanced entry into tumor cells. Tumor selectivity is mediated by an insulator, a modified E2F promoter, and a Rb-binding site deletion of E1A, whereas replication is optimized with E2F binding hairpins and a Kozak sequence. ICOVIR-7 doses ranged from 2 x 10(10) to 1 x 10(12) viral particles. All patients had advanced and metastatic solid tumors refractory to standard therapies. RESULTS: ICOVIR-7 treatment was well tolerated with mild to moderate fever, fatigue, elevated liver transaminases, chills, and hyponatremia. One patient had grade 3 anemia but no other serious side effects were seen. At baseline, 9 of 21 of patients had neutralizing antibody titers against the ICOVIR-7 capsid. Treatment resulted in neutralizing antibody titer induction within 4 weeks in 16 of 18 patients. No elevations of serum proinflammatory cytokine levels were detected. Viral genomes were detected in the circulation in 18 of 21 of patients after injection and 7 of 15 of the samples were positive 2 to 4 weeks later suggesting viral replication. CONCLUSIONS: Overall, objective evidence of antitumor activity was seen in 9 of 17 evaluable patients. In radiological analyses, 5 of 12 evaluable patients had stabilization or reduction in tumor size. These consisted of one partial response, two minor responses and two cases of stable disease, all occurring in patients who had progressive disease before treatment. In summary, ICOVIR-7 treatment is apparently safe, resulting in anticancer activity, and is therefore promising for further clinical testing.


Assuntos
Adenoviridae , Neoplasias/terapia , Terapia Viral Oncolítica/métodos , Adolescente , Adulto , Idoso , Anticorpos Antivirais/análise , Criança , Feminino , Humanos , Interleucinas/sangue , Masculino , Pessoa de Meia-Idade , Terapia Viral Oncolítica/efeitos adversos , Retratamento , Resultado do Tratamento , Replicação Viral
13.
Int J Cancer ; 125(10): 2441-9, 2009 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-19672857

RESUMO

New treatment approaches are needed for hormone refractory prostate cancer. Oncolytic adenoviruses are promising anti-cancer agents, and their efficacy can be improved by combining with conventional therapies such as ionizing radiation. The aim of this study was to determine the timing of oncolytic adenovirus treatment with regard to radiation and study the mechanisms of synergy in combination treatment. Prostate cancer cells were infected with oncolytic adenoviruses, irradiated and synergy mechanisms were assessed. In vivo models of combination treatment were tested. Radiation and oncolytic viruses were synergistic when viral infection was scheduled 24 hr after irradiation. Combination of oncolytic adenovirus with radiotherapy significantly increased antitumor efficacy in vivo compared to either agent alone. Microarray analysis showed dysregulated pathways including cell cycle, mTOR and antigen processing pathways. Functional analysis showed that adenoviral infection was accompanied with degradation of proteins involved in DNA break repair. Mre11 was degraded for subsequent inactivation of Chk2-Thr68 in combination treated cells, while gammaH2AX-Ser139 was elevated implicating the persistence of DNA double strand breaks. Increased autophagocytosis was seen in combination treated cells. Combination treatment did not increase apoptosis or virus replication. The results provide evidence of the antitumor efficacy of combining oncolytic adenoviruses with irradiation as a therapeutic strategy for the treatment of prostate cancer. Further, these findings propose a molecular mechanism that may be important in radiation induced cell death, autophagy and viral cytopathic effect.


Assuntos
Autofagia , Proteínas de Ligação a DNA/antagonistas & inibidores , Terapia Viral Oncolítica , Neoplasias da Próstata/terapia , Radiação Ionizante , Adenoviridae/genética , Animais , Apoptose , Terapia Combinada , Proteínas de Ligação a DNA/metabolismo , Sinergismo Farmacológico , Perfilação da Expressão Gênica , Humanos , Proteína Homóloga a MRE11 , Masculino , Camundongos , Camundongos Nus , Análise de Sequência com Séries de Oligonucleotídeos , Vírus Oncolíticos , Neoplasias da Próstata/metabolismo , Neoplasias da Próstata/patologia , Células Tumorais Cultivadas , Replicação Viral , Irradiação Corporal Total , Ensaios Antitumorais Modelo de Xenoenxerto
14.
J Gene Med ; 11(11): 966-77, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19670332

RESUMO

BACKGROUND: Rapid clearance of adenoviruses from blood by macrophage lineage cells of the liver and spleen, and binding to platelets, hinder their successful systemic use for cancer gene therapy. Vitamin K dependent coagulation factors are important mediators for the adenovirus liver tropism. Here we aim to determine the effects of coagulation factor, thrombocyte and liver macrophage (Kupffer cell) ablation on biodistribution of serotype 5 adenoviruses in mice with orthotopic breast tumors. METHODS: Prior to intravenous injection of adenoviruses, mice bearing orthotopic breast tumors were pretreated with warfarin to inhibit vitamin K dependent coagulation factor synthesis, an anti-platelet antibody causing thrombocytopenia or an inhibitor of the Kupffer cell scavenger receptor or their combination. Virus availability in blood after injection was determined from blood samples and transgene expression in tissues analyzed 72 hours afterwards with In Vivo Imaging and luciferase assays. RESULTS: Warfarin pretreatment reduced gene delivery to liver, spleen and lung. Kupffer cell ablation increased persistence of adenoviruses in blood but didn't affect biodistribution significantly. Depletion of Kupffer cells combined with thrombocytopenia doubled the systemic gene delivery of 5/3 chimeric adenovirus to tumors (p < 0.05). Triple ablation of platelets, Kupffer cells and coagulation factors increased the tumor to liver ratio of systemic adenovirus gene delivery by 81% (p < 0.05). CONCLUSIONS: Depletion of coagulation factors can reduce transduction of liver, spleen and lung. Kupffer cell depletion is the most feasible method of increasing amount adenovirus in systemic blood flow and in combination with ablation of thrombocytes can increase the transduction of adenovirus to tumors.


Assuntos
Adenoviridae/genética , Neoplasias Mamárias Experimentais/genética , Vírus Oncolíticos/genética , Transdução Genética , Adenoviridae/metabolismo , Animais , Fatores de Coagulação Sanguínea/antagonistas & inibidores , Plaquetas/efeitos dos fármacos , Feminino , Células de Kupffer/efeitos dos fármacos , Neoplasias Mamárias Experimentais/metabolismo , Neoplasias Mamárias Experimentais/terapia , Camundongos , Terapia Viral Oncolítica/métodos , Varfarina/farmacologia
15.
Clin Cancer Res ; 15(17): 5396-403, 2009 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-19706820

RESUMO

PURPOSE: Oncolytic adenoviruses are promising tools for cancer therapy. Although several clinical reports have indicated both safety and promising antitumor capabilities for these viruses, there are only a few examples of complete tumor eradication. Thus, the antitumor efficacy of oncolytic adenoviruses needs to be improved. One potentially useful approach is combination with radiotherapy. EXPERIMENTAL DESIGN: To target systemically administered radioiodide to tumors, we created Ad5/3-Delta24-human sodium iodide symporter (hNIS), a Rb-p16 pathway selective infectivity enhanced oncolytic adenovirus encoding hNIS. RESULTS: Ad5/3-Delta24-hNIS replication effectively killed prostate cancer cells in vitro and in vivo. Also, the virus-mediated radioiodide uptake into prostate cancer cells in vitro and into tumors in vivo. Furthermore, Ad5/3-Delta24-hNIS with radioiodide was significantly more effective than virus alone in mice with prostate cancer xenografts. CONCLUSIONS: These results suggest that oncolytic adenovirus-mediated targeted radiotherapy might be a potentially useful option for enhancing the efficacy or adenoviral virotherapy.


Assuntos
Adenocarcinoma/terapia , Neoplasias Pulmonares/terapia , Terapia Viral Oncolítica , Neoplasias da Próstata/terapia , Simportadores/genética , Adenocarcinoma/radioterapia , Adenoviridae/genética , Animais , Linhagem Celular Tumoral , Terapia Combinada , Terapia Genética , Humanos , Isótopos de Iodo/farmacologia , Isótopos de Iodo/uso terapêutico , Neoplasias Pulmonares/radioterapia , Masculino , Camundongos , Vírus Oncolíticos/genética , Neoplasias da Próstata/radioterapia , Ensaios Antitumorais Modelo de Xenoenxerto
16.
Hum Gene Ther ; 20(6): 611-20, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19239383

RESUMO

Despite some advances, patients with advanced renal cell carcinoma (RCC) cannot usually be cured. Alteration of the natural tropism of adenoviruses may permit more specific gene transfer to target tissues. The aim of this study was to use novel targeting moieties for adenoviral gene therapy of RCC. Previous work in rats suggested that use of Ad5/19p (Ad5 capsid with Ad19p fiber) with kidney vascular targeting moieties HTTHREP (HTT), HITSLLS (HIT), and APASLYN (APA) placed into the fiber knob might be useful for targeting kidney vasculature. Therefore, we sought to investigate the utility of Ad5/19p variants for gene delivery to human RCC cell lines, clinical samples, and orthotopic murine models of metastatic RCC. Six different human RCC cell lines were infected but only Ad5/19p-HIT showed increased transduction, and only in one cell line. Thus, we analyzed human normal and cancerous kidney specimens fresh from patients, which might better mimic the three-dimensional architecture of clinical tumors and found that Ad5/19p-HIT showed transduction levels similar to Ad5. In mice, we found that intraperitoneal and intravenous Ad5/19p-HIT transduced tumors at levels comparable to Ad5, and that intratumoral Ad5/19p-HIT was superior to Ad5. Liver tropism was significantly reduced in comparison with Ad5. Improvements in tumor-to-liver transduction ratios suggested that Ad5/19p-HIT may be promising for systemic gene delivery to kidney tumors.


Assuntos
Adenoviridae/genética , Neoplasias Renais/terapia , Fígado/metabolismo , Mutação/genética , Peptídeos/farmacologia , Sorotipagem , Animais , Linhagem Celular Tumoral , Modelos Animais de Doenças , Feminino , Técnicas de Transferência de Genes , Humanos , Injeções Intraperitoneais , Injeções Intravenosas , Fígado/efeitos dos fármacos , Camundongos , Camundongos SCID , Pessoa de Meia-Idade , Especificidade de Órgãos/efeitos dos fármacos , Peptídeos/administração & dosagem , Ratos , Transdução Genética
17.
Mol Cancer Ther ; 6(10): 2728-36, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17938266

RESUMO

Renal cancer is a common and deadly disease that lacks curative treatments when metastatic. Here, we have used oncolytic adenoviruses, a promising developmental approach whose safety has recently been validated in clinical trials. Although preliminary clinical efficacy data exist for selected tumor types, potency has generally been less than impressive. One important reason may be that expression of the primary receptor, coxsackie-adenovirus receptor, is often low on many or most advanced tumors, although not evaluated in detail with renal cancer. Here, we tested if fluorescence-assisted cell sorting could be used to predict efficacy of a panel of infectivity-enhanced capsid-modified marker gene expressing adenoviruses in renal cancer cell lines, clinical specimens, and subcutaneous and orthotopic murine models of peritoneally metastatic renal cell cancer. The respective selectively oncolytic adenoviruses were tested for killing of tumor cells in these models, and biodistribution after locoregional delivery was evaluated. In vivo replication was analyzed with noninvasive imaging. Ad5/3-Delta24, Ad5-Delta24RGD, and Ad5.pK7-Delta24 significantly increased survival of mice compared with mock or wild-type virus and 50% of Ad5/3-Delta24 treated mice were alive at 320 days. Because renal tumors are often highly vascularized, we investigated if results could be further improved by adding bevacizumab, a humanized antivascular endothelial growth factor antibody. The combination was well tolerated but did not improve survival, suggesting that the agents may be best used in sequence instead of together. These results set the stage for clinical testing of oncolytic adenoviruses for treatment of metastatic renal cancer currently lacking other treatment options.


Assuntos
Adenoviridae/genética , Capsídeo/metabolismo , Carcinoma de Células Renais/terapia , Neoplasias Renais/terapia , Terapia Viral Oncolítica , Neoplasias Peritoneais/terapia , Transgenes/fisiologia , Inibidores da Angiogênese/uso terapêutico , Animais , Anticorpos Monoclonais/uso terapêutico , Anticorpos Monoclonais Humanizados , Apoptose/efeitos dos fármacos , Bevacizumab , Capsídeo/química , Carcinoma de Células Renais/secundário , Terapia Combinada , Proteína de Membrana Semelhante a Receptor de Coxsackie e Adenovirus , Modelos Animais de Doenças , Feminino , Citometria de Fluxo , Técnicas de Transferência de Genes , Proteoglicanas de Heparan Sulfato/metabolismo , Humanos , Neoplasias Renais/patologia , Luciferases/metabolismo , Camundongos , Camundongos Nus , Camundongos SCID , Neoplasias Peritoneais/secundário , Receptores Virais/metabolismo , Células Tumorais Cultivadas , Fator A de Crescimento do Endotélio Vascular/antagonistas & inibidores , Fator A de Crescimento do Endotélio Vascular/imunologia , Replicação Viral , Ensaios Antitumorais Modelo de Xenoenxerto
18.
Mol Cancer Ther ; 6(2): 742-51, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17308070

RESUMO

Hormone refractory metastatic prostate cancer is a deadly disease that currently lacks curative treatments. Conditionally replicating adenoviruses (CRAds) are promising new agents against cancer due to their innate capability to cause oncolysis of tumor cells. Their antitumor effect is determined in part by their capacity for infecting cancer cells. However, the respective primary receptor, the coxsackie-adenovirus receptor (CAR), is variably expressed in many cancer types. We created Ad5/3Delta24hCG, a novel CRAd retargeted to the adenovirus serotype 3 receptor, which has been reported to be highly expressed in tumors. Furthermore, we added a transgene for the beta-chain of human chorionic gonadotropin (hCGbeta), whose expression was tightly coupled to virus replication. Ad5/3Delta24hCG was found effective in killing prostate cancer cells, and oncolysis was seen in concordance with hCGbeta production. In a s.c. in vivo model of hormone refractory prostate cancer, Ad5/3Delta24hCG treatment resulted in statistically significant tumor growth inhibition. Moreover, i.v. injection of Ad5/3Delta24hCG prolonged the survival of mice with hormone refractory prostate cancer metastatic to the lung. Detection of hCGbeta in serum samples confirmed viral replication in vivo. Infection of human clinical samples of cancerous and normal prostatic tissue resulted in effective hCGbeta production in cancer tissue, whereas it remained low in nonmalignant tissue, suggesting cancer-specific replication. These results suggest that Ad5/3Delta24hCG is a potent virus for the treatment of hormone refractory prostate cancer in vitro and in vivo. These preclinical data set the stage for translation into clinical studies.


Assuntos
Adenoviridae/fisiologia , Gonadotropina Coriônica/farmacologia , Neoplasias Pulmonares/terapia , Terapia Viral Oncolítica , Neoplasias da Próstata/terapia , Replicação Viral , Animais , Sobrevivência Celular , Células Cultivadas , Gonadotropina Coriônica/genética , Proteína de Membrana Semelhante a Receptor de Coxsackie e Adenovirus , DNA Viral/genética , Terapia Genética , Vetores Genéticos , Humanos , Rim/metabolismo , Rim/virologia , Neoplasias Pulmonares/secundário , Neoplasias Pulmonares/virologia , Masculino , Camundongos , Camundongos Nus , Oligopeptídeos/metabolismo , Neoplasias da Próstata/metabolismo , Neoplasias da Próstata/virologia , Receptores Virais/metabolismo , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
19.
Blood Coagul Fibrinolysis ; 16(2): 119-24, 2005 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15741799

RESUMO

We carried out a prospective cohort study to determine whether the plasma levels of fibrinogen, plasminogen, factor VII and lipoprotein (a) are predictors of ischemic stroke and all cardiovascular disease (CVD) events. The FINRISK '92 Hemostasis Study included a random sample of 2372 participants, who were followed-up from winter 1992 to 31 December 2001. During the follow-up, 75 ischemic stroke and 145 coronary events occurred. Of these, 169 were observed among participants free of known CVD at baseline. In this group, fibrinogen and plasminogen were positively associated with the risk of a CVD event with hazard ratios of 1.22 [95% confidence interval (CI), 1.05-1.41] and 1.22 (95% CI, 1.03-1.44), respectively, after adjusting for age, sex and conventional risk factors. Factor VII:C was associated with risk of a future CVD event only among persons with positive history of CVD at baseline (hazard ratio, 1.32; 95% CI, 1.00-1.73). Factor VII antigen was not associated with CVD risk. None of the measured hemostatic factors was a predictor of ischemic stroke events, with the possible exception of lipoprotein (a), which had a borderline significant association (hazard ratio, 1.25; 95% CI, 0.99-1.58). In conclusion, the present study supports the observations that fibrinogen and plasminogen are significant predictors of CVD events, independently of conventional risk factors.


Assuntos
Fibrinogênio/análise , Plasminogênio/análise , Acidente Vascular Cerebral/sangue , Biomarcadores/sangue , Estudos de Coortes , Feminino , Humanos , Masculino , Fatores de Risco , Acidente Vascular Cerebral/diagnóstico
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...