Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
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.
Mol Ther ; 21(6): 1212-23, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23546299

RESUMO

Oncolytic adenoviruses and certain chemotherapeutics can induce autophagy and immunogenic cancer cell death. We hypothesized that the combination of oncolytic adenovirus with low-dose temozolomide (TMZ) is safe, effective, and capable of inducing antitumor immune responses. Metronomic low-dose cyclophosphamide (CP) was added to selectively reduce regulatory T-cells. Preclinically, combination therapy inhibited tumor growth, increased autophagy, and triggered immunogenic cell death as indicated by elevated calreticulin, adenosine triphosphate (ATP) release, and nuclear protein high-mobility group box-1 (HMGB1) secretion. A total of 41 combination treatments given to 17 chemotherapy-refractory cancer patients were well tolerated. We observed anti- and proinflammatory cytokine release, evidence of virus replication, and induction of neutralizing antibodies. Tumor cells showed increased autophagy post-treatment. Release of HMGB1 into serum--a possible indicator of immune response--increased in 60% of treatments, and seemed to correlate with tumor-specific T-cell responses, observed in 10/15 cases overall (P = 0.0833). Evidence of antitumor efficacy was seen in 67% of evaluable treatments with a trend for increased survival over matched controls treated with virus only. In summary, the combination of oncolytic adenovirus with low-dose TMZ and metronomic CP increased tumor cell autophagy, elicited antitumor immune responses, and showed promising safety and efficacy.


Assuntos
Adenoviridae/genética , Antineoplásicos/farmacologia , Autofagia/efeitos dos fármacos , Dacarbazina/análogos & derivados , Neoplasias/terapia , Terapia Viral Oncolítica/métodos , Trifosfato de Adenosina/metabolismo , Adenoviridae/fisiologia , Adolescente , Adulto , Idoso , Animais , Anticorpos Neutralizantes/sangue , Calreticulina/metabolismo , Morte Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Criança , Terapia Combinada/métodos , Ciclofosfamida/farmacologia , Citocinas/sangue , DNA Viral/sangue , Dacarbazina/farmacologia , Relação Dose-Resposta a Droga , Feminino , Proteína HMGB1/sangue , Proteína HMGB1/metabolismo , Humanos , Imuno-Histoquímica , Masculino , Camundongos , Camundongos Nus , Microscopia Eletrônica , Pessoa de Meia-Idade , Vírus Oncolíticos/genética , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/metabolismo , Temozolomida , Replicação Viral , Ensaios Antitumorais Modelo de Xenoenxerto , Adulto Jovem
3.
Clin Cancer Res ; 19(10): 2734-44, 2013 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-23493351

RESUMO

PURPOSE: Multiple injections of oncolytic adenovirus could enhance immunologic response. In the first part of this article, the focus was on immunologic aspects. Sixty patients previously naïve to oncolytic virus and who had white blood cells available were treated. Thirty-nine of 60 were assessed after a single virus administration, whereas 21 of 60 received a "serial treatment" consisting of three injections within 10 weeks. In the second part, we focused on 115 patients treated with a granulocyte macrophage colony-stimulating factor (GM-CSF)-coding capsid chimeric adenovirus, CGTG-102. RESULTS: Following serial treatment, both increase and decrease in antitumor T cells in blood were seen more frequently, findings which are compatible with induction of T-cell immunity and trafficking of T cells to tumors, respectively. Safety was good in both groups. In 115 patients treated with CGTG-102 (Ad5/3-D24-GMCSF), median overall survival was 111 days following single and 277 days after serial treatment in nonrandomized comparison. Switching the virus capsid for avoiding neutralizing antibodies in a serial treatment featuring three different viruses did not impact safety or efficacy. A correlation between antiviral and antitumor T cells was seen (P = 0.001), suggesting that viral oncolysis can result in epitope spreading and breaking of tumor-associated immunologic tolerance. Alternatively, some patients may be more susceptible to induction of T-cell immunity and/or trafficking. CONCLUSIONS: These results provide the first human data linking antiviral immunity with antitumor immunity, implying that oncolytic viruses could have an important role in cancer immunotherapy.


Assuntos
Neoplasias/imunologia , Neoplasias/terapia , Terapia Viral Oncolítica/métodos , Linfócitos T/imunologia , Adenoviridae/genética , Adenoviridae/imunologia , Adolescente , Adulto , Idoso , Proteínas do Capsídeo/genética , Proteínas do Capsídeo/imunologia , Criança , Anergia Clonal/imunologia , Ensaio de Imunoadsorção Enzimática , Feminino , Fator Estimulador de Colônias de Granulócitos e Macrófagos/genética , Fator Estimulador de Colônias de Granulócitos e Macrófagos/imunologia , Fator Estimulador de Colônias de Granulócitos e Macrófagos/metabolismo , Humanos , Interferon gama/imunologia , Interferon gama/metabolismo , Interleucina-10/imunologia , Interleucina-10/metabolismo , Masculino , Pessoa de Meia-Idade , Neoplasias/classificação , Vírus Oncolíticos/genética , Vírus Oncolíticos/imunologia , Análise de Sobrevida , Linfócitos T/metabolismo , Fatores de Tempo , Resultado do Tratamento , Fator de Necrose Tumoral alfa/imunologia , Fator de Necrose Tumoral alfa/metabolismo , Adulto Jovem
4.
Mol Ther ; 20(1): 221-9, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22044933

RESUMO

Calcium channel blockers including verapamil have been proposed to enhance release and antitumor efficacy of oncolytic adenoviruses in preclinical studies but this has not been studied in humans before. Here, we studied if verapamil leads to increased replication of oncolytic adenovirus in cancer patients, as measured by release of virions from tumor cells into the systemic circulation. The study was conducted as a matched case-control study of advanced cancer patients treated with oncolytic adenoviruses with or without verapamil. We observed that verapamil increased mean virus titers present in blood after treatment (P < 0.05). The frequency or severity of adverse events was not increased, nor were cytokine responses or neutralizing antibody levels different between groups. Signs of possible treatment-related clinical benefits were observed in both groups, but there was no significant difference in responses or survival. Thus, our data suggests that the combination of verapamil with oncolytic adenoviruses is safe and well tolerated. Moreover, verapamil treatment seems to result in higher virus titers in blood, indicating enhanced overall replication in tumors. A randomized trial is needed to confirm these findings and to study if enhanced replication results in benefits to patients.


Assuntos
Adenoviridae/genética , Bloqueadores dos Canais de Cálcio/uso terapêutico , Vetores Genéticos , Neoplasias/terapia , Vírus Oncolíticos/genética , Verapamil/uso terapêutico , Adenoviridae/imunologia , Adolescente , Adulto , Idoso , Anticorpos Neutralizantes/imunologia , Anticorpos Antivirais/imunologia , Bloqueadores dos Canais de Cálcio/efeitos adversos , Terapia Combinada , DNA Viral/sangue , Sinergismo Farmacológico , Feminino , Terapia Genética , Vetores Genéticos/administração & dosagem , Vetores Genéticos/efeitos adversos , Humanos , Mediadores da Inflamação/sangue , Masculino , Pessoa de Meia-Idade , Estadiamento de Neoplasias , Neoplasias/mortalidade , Neoplasias/patologia , Terapia Viral Oncolítica/efeitos adversos , Vírus Oncolíticos/imunologia , Análise de Sobrevida , Resultado do Tratamento , Verapamil/efeitos adversos , Carga Viral/efeitos dos fármacos , Adulto Jovem
5.
Int J Cancer ; 130(8): 1937-47, 2012 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-21630267

RESUMO

The safety of oncolytic viruses for treatment of cancer has been shown in clinical trials while antitumor efficacy has often remained modest. As expression of the coxsackie-adenovirus receptor may be variable in advanced tumors, we developed Ad5-D24-RGD, a p16/Rb pathway selective oncolytic adenovirus featuring RGD-4C modification of the fiber. This allows viral entry through alpha-v-beta integrins frequently highly expressed in advanced tumors. Advanced tumors are often immunosuppressive which results in lack of tumor eradication despite abnormal epitopes being present. Granulocyte-macrophage colony stimulating factor (GMCSF) is a potent activator of immune system with established antitumor properties. To stimulate antitumor immunity and break tumor associated immunotolerance, we constructed Ad5-RGD-D24-GMCSF, featuring GMCSF controlled by the adenoviral E3 promoter. Preliminary safety of Ad5-D24-RGD and Ad5-RGD-D24-GMCSF for treatment of human cancer was established. Treatments with Ad5-D24-RGD (N = 9) and Ad5-RGD-D24-GMCSF (N = 7) were well tolerated. Typical side effects were grade 1-2 fatigue, fever and injection site pain. 77% (10/13) of evaluable patients showed virus in circulation for at least 2 weeks. In 3 out of 6 evaluable patients, disease previously progressing stabilized after a single treatment with Ad5-RGD-D24-GMCSF. In addition, 2/3 patients had stabilization or reduction in tumor marker levels. All patients treated with Ad5-D24-RGD showed disease progression in radiological analysis, although 3/6 had temporary reduction or stabilization of marker levels. Induction of tumor and adenovirus specific immunity was demonstrated with ELISPOT in Ad5-RGD-D24-GMCSF treated patients. RGD modified oncolytic adenoviruses with or without GMCSF seem safe for further clinical development.


Assuntos
Fator Estimulador de Colônias de Granulócitos e Macrófagos/metabolismo , Neoplasias/terapia , Oligopeptídeos/metabolismo , Terapia Viral Oncolítica/métodos , Adenoviridae/genética , Adulto , Idoso , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Linhagem Celular Tumoral , DNA Viral/genética , Resistencia a Medicamentos Antineoplásicos , Fadiga/etiologia , Feminino , Febre/etiologia , Vetores Genéticos/administração & dosagem , Vetores Genéticos/genética , Fator Estimulador de Colônias de Granulócitos e Macrófagos/genética , Humanos , Integrinas/metabolismo , Masculino , Pessoa de Meia-Idade , Neoplasias/metabolismo , Neoplasias/virologia , Oligopeptídeos/genética , Terapia Viral Oncolítica/efeitos adversos , Vírus Oncolíticos/genética , Reação em Cadeia da Polimerase em Tempo Real , Resultado do Tratamento , Carga Viral , Replicação Viral/genética
6.
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
7.
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
8.
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
9.
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
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
12.
Eur J Cancer ; 46(3): 625-35, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-20006487

RESUMO

Head and neck squamous cell carcinoma (HNSCC) is a common and often devastating disease without curative treatment when advanced or recurrent. The aim of this study was to assess whether capsid-modified oncolytic adenoviruses have therapeutic efficacy in HNSCC low passage tumour cell cultures and if it could be further improved by combination with cetuximab, radiotherapy and chemotherapy. We investigated which adenoviral capsid modifications allow best gene transfer and cell killing of HNSCC substrates. Gene transfer was assessed using replication-deficient adenoviruses expressing luciferase. Cell killing was studied in vitro and in vivo using oncolytic adenoviruses, which kill tumour cell by viral replication. The most effective capsid-modified oncolytic adenoviruses were combined with HNSCC standard therapies and their efficacy was assessed in vitro as well as in vivo. Cell killing was assessed in vitro by MTS assay and in vivo by HNSCC subcutaneous tumour growth follow-up in nude mice. Cetuximab treatment was found to enrich CD133+ and CD44+ tumour-initiating type cells in tumours grown in mice. Capsid-modified viruses showed increased transduction and oncolysis of HNSCC substrates in comparison to Ad5-based agents. Polylysine (pK7)-modified oncolytic virus resulted in significant tumour growth reduction in vivo. Combination of chemotherapy (cisplatin and 5-fluorouracil), radiotherapy and cetuximab with oncolytic adenovirus therapy resulted in further increases in cell killing effect in vitro and complete eradication of tumours in vivo. Our pre-clinical data suggest that it is feasible and efficacious to combine oncolytic adenoviruses with HNSCC standard therapies into a multimodality treatment regimen for clinical testing in HNSCC patients.


Assuntos
Anticorpos Monoclonais/farmacologia , Antineoplásicos/farmacologia , Carcinoma de Células Escamosas/terapia , Neoplasias de Cabeça e Pescoço/terapia , Terapia Viral Oncolítica/métodos , Antígeno AC133 , Adenoviridae , Animais , Anticorpos Monoclonais Humanizados , Antígenos CD/análise , Carcinoma de Células Escamosas/patologia , Morte Celular , Cetuximab , Terapia Combinada , Estudos de Viabilidade , Feminino , Técnicas de Transferência de Genes , Glicoproteínas/análise , Neoplasias de Cabeça e Pescoço/patologia , Humanos , Receptores de Hialuronatos/análise , Camundongos , Camundongos Nus , Peptídeos/análise , Transplante Heterólogo , Células Tumorais Cultivadas
13.
Int J Radiat Oncol Biol Phys ; 78(1): 42-9, 2010 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-20004525

RESUMO

PURPOSE: To estimate the safety and tolerability of daily administration of 250 mg of gefitinib given concurrently with three-dimensional conformal radiotherapy for patients with nonmetastatic prostate cancer. METHODS AND MATERIALS: A total of 42 patients with T2-T3N0M0 tumors were treated in a nonrandomized single-center study. A prostate-specific antigen (PSA) level of <20 and a good performance status (WHO, 0-1) were required. Adjuvant or neoadjuvant hormone treatments were not allowed. A daily regimen of 250 mg of gefitinib was started 1 week before radiation therapy began and lasted for the duration of radiation therapy. A dose of 50.4 Gy (1.8 Gy/day) was administered to the tumor, prostate, and seminal vesicles, followed by a 22-Gy booster (2 Gy/day) for a total dose of 72.4 Gy. Correlative studies included analysis of epidermal growth factor receptor (EGFR), EGFRvIII, and phosphorylated EGFR in tumors and tumor necrosis factor, interleukin-1alpha (IL-1alpha), and IL-6 in serum. RESULTS: Maximum tolerated dose was not reached in phase I (12 patients), and 30 additional patients were treated in phase II. Thirty (71.4%) patients completed trial medication. Dose-limiting toxicities were recorded for 16 (38.1%) patients, the most common of which was a grade 3 to 4 increase in transaminase (6 patients). After a median follow-up of 38 months, there were no deaths due to prostate cancer. The estimated PSA relapse-free survival rate at 4 years (Kaplan-Meier) was 97%, the salvage therapy-free survival rate was 91%, and the overall survival rate was 87%. These figures compared favorably with those of matched patients treated with radiation only at higher doses. CONCLUSIONS: The combination of gefitinib and radiation is reasonably well tolerated and has promising activity against nonmetastatic prostate cancer.


Assuntos
Antineoplásicos/efeitos adversos , Neoplasias da Próstata/tratamento farmacológico , Neoplasias da Próstata/radioterapia , Quinazolinas/efeitos adversos , Idoso , Antineoplásicos/administração & dosagem , Terapia Combinada/efeitos adversos , Terapia Combinada/métodos , Intervalo Livre de Doença , Esquema de Medicação , Receptores ErbB/análise , Gefitinibe , Humanos , Interleucina-1alfa/sangue , Interleucina-6/sangue , Masculino , Dose Máxima Tolerável , Pessoa de Meia-Idade , Proteínas de Neoplasias/metabolismo , Estadiamento de Neoplasias , Antígeno Prostático Específico/sangue , Neoplasias da Próstata/metabolismo , Neoplasias da Próstata/patologia , Quinazolinas/administração & dosagem , Dosagem Radioterapêutica , Fator de Necrose Tumoral alfa/sangue
14.
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
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...