Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
PLoS One ; 7(6): e38557, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22745668

RESUMO

PURPOSE: To assess the cost effectiveness of adding cetuximab to platinum-based chemotherapy in first-line treatment of patients with recurrent or metastatic head and neck squamous cell carcinoma (HNSCC) from the perspective of the Canadian public healthcare system. METHODS: We developed a Markov state transition model to project the lifetime clinical and economic consequences of recurrent or metastatic HNSCC. Transition probabilities were derived from a phase III trial of cetuximab in patients with recurrent or metastatic HNSCC. Cost estimates were obtained from London Health Sciences Centre and the Ontario Case Costing Initiative, and expressed in 2011 CAD. A three year time horizon was used. Future costs and health benefits were discounted at 5%. RESULTS: In the base case, cetuximab plus platinum-based chemotherapy compared to platinum-based chemotherapy alone led to an increase of 0.093 QALY and an increase in cost of $36,000 per person, resulting in an incremental cost effectiveness ratio (ICER) of $386,000 per QALY gained. The cost effectiveness ratio was most sensitive to the cost per mg of cetuximab and the absolute risk of progression among patients receiving cetuximab. CONCLUSION: The addition of cetuximab to standard platinum-based chemotherapy in first-line treatment of patients with recurrent or metastatic HNSCC has an ICER that exceeds $100,000 per QALY gained. Cetuximab can only be economically attractive in this patient population if the cost of cetuximab is substantially reduced or if future research can identify predictive markers to select patients most likely to benefit from the addition of cetuximab to chemotherapy.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Protocolos de Quimioterapia Combinada Antineoplásica/economia , Neoplasias de Cabeça e Pescoço/tratamento farmacológico , Platina/uso terapêutico , Anticorpos Monoclonais Humanizados , Cetuximab , Análise Custo-Benefício , Humanos , Modelos Teóricos
2.
Cancer Cell Int ; 12(1): 27, 2012 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-22704542

RESUMO

BACKGROUND: Head and neck squamous cell carcinoma (HNSCC) is the 5th most common cancer worldwide. Locally advanced HNSCC are treated with either radiation or chemo-radiotherapy, but still associated with high mortality rate, underscoring the need to develop novel therapies. Oncolytic viruses have been garnering increasing interest as anti-cancer agents due to their preferential killing of transformed cells. In this study, we evaluated the therapeutic potential of mutant vesicular stomatitis virus (VSVΔ51) against the human hypopharyngeal FaDu tumour model in vitro and in vivo. RESULTS: Our data demonstrated high toxicity of the virus against FaDu cells in vitro, which was associated with induction of apoptosis. In vivo, systemic injection of 1 × 109 pfu had minimal effect on tumour growth; however, when combined with two doses of ionizing radiation (IR; 5 Gy each) or a single injection of the vascular disrupting agent (ZD6126), the virus exhibited profound suppression of tumour growth, which translated to a prolonged survival in the treated mice. Concordantly, VSVΔ51 combined with ZD6126 led to a significant increase in viral replication in these tumours. CONCLUSIONS: Our data suggest that the combinations of VSVΔ51 with either IR or ZD6126 are potentially novel therapeutic opportunities for HNSCC.

3.
Int J Radiat Oncol Biol Phys ; 81(4): 964-73, 2011 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-20932688

RESUMO

PURPOSE: To compare the quality-adjusted life expectancy and overall survival in patients with Stage I non-small-cell lung cancer (NSCLC) treated with either stereotactic body radiation therapy (SBRT) or surgery. METHODS AND MATERIALS: We constructed a Markov model to describe health states after either SBRT or lobectomy for Stage I NSCLC for a 5-year time frame. We report various treatment strategy survival outcomes stratified by age, sex, and pack-year history of smoking, and compared these with an external outcome prediction tool (Adjuvant! Online). RESULTS: Overall survival, cancer-specific survival, and other causes of death as predicted by our model correlated closely with those predicted by the external prediction tool. Overall survival at 5 years as predicted by baseline analysis of our model is in favor of surgery, with a benefit ranging from 2.2% to 3.0% for all cohorts. Mean quality-adjusted life expectancy ranged from 3.28 to 3.78 years after surgery and from 3.35 to 3.87 years for SBRT. The utility threshold for preferring SBRT over surgery was 0.90. Outcomes were sensitive to quality of life, the proportion of local and regional recurrences treated with standard vs. palliative treatments, and the surgery- and SBRT-related mortalities. CONCLUSIONS: The role of SBRT in the medically operable patient is yet to be defined. Our model indicates that SBRT may offer comparable overall survival and quality-adjusted life expectancy as compared with surgical resection. Well-powered prospective studies comparing surgery vs. SBRT in early-stage lung cancer are warranted to further investigate the relative survival, quality of life, and cost characteristics of both treatment paradigms.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/cirurgia , Técnicas de Apoio para a Decisão , Neoplasias Pulmonares/cirurgia , Cadeias de Markov , Pneumonectomia/métodos , Radiocirurgia/métodos , Idoso , Carcinoma Pulmonar de Células não Pequenas/mortalidade , Carcinoma Pulmonar de Células não Pequenas/patologia , Causas de Morte , Feminino , Humanos , Expectativa de Vida , Neoplasias Pulmonares/mortalidade , Neoplasias Pulmonares/patologia , Masculino , Estadiamento de Neoplasias , Pneumonectomia/mortalidade , Qualidade de Vida , Anos de Vida Ajustados por Qualidade de Vida , Radiocirurgia/mortalidade , Reprodutibilidade dos Testes , Terapia de Salvação/métodos , Sensibilidade e Especificidade , Fatores Sexuais , Fumar
4.
Int J Cancer ; 126(9): 2036-48, 2010 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-19739117

RESUMO

Polo-like kinase 1 (Plk1) is a critical regulator of many stages of mitosis; increasing evidence indicates that Plk1 overexpression correlates with poor clinical outcome, yet its mechanism of regulation remains unknown. Hence, a detailed evaluation was undertaken of Plk1 expression in human nasopharyngeal cancer (NPC), the cellular effects of targeting Plk1 using siRNA in combination with ionizing radiation (RT) and potential upstream microRNAs (miRs) that might regulate Plk1 expression. Using immunohistochemistry, Plk1 was observed to be overexpressed in 28 of 40 (70%) primary NPC biopsies, which in turn was associated with a higher likelihood of recurrence (p = 0.018). SiPlk1 significantly inhibited Plk1 mRNA and protein expression, and decreased Cdc25c levels in NPC cell lines. This depletion resulted in cytotoxicity of C666-1 cells, enhanced by the addition of RT, mediated by G2/M arrest, increased DNA double-strand breaks, apoptosis, and caspase activation. Immunofluorescence demonstrated that the G2/M arrest was associated with aberrant spindle formation, leading to mitotic arrest. In vivo, transfection of C666-1 cells and systemic delivery of siPlk1 decreased tumour growth. MicroRNA-100 (miR-100) was predicted to target Plk1 mRNA, which was indeed underexpressed in C666-1 cells, inversely correlating with Plk1 expression. Using luciferase constructs containing the 3'-UTR of Plk1 sequence, we document that miR-100 can directly target Plk1. Hence, our data demonstrate for the first time that underexpressed miR-100 leads to Plk1 overexpression, which in turn contributes to NPC progression. Targeting Plk1 will cause mitotic catastrophe, with significant cytotoxicity both in vitro and in vivo, underscoring the important therapeutic opportunity of Plk1 in NPC.


Assuntos
Proteínas de Ciclo Celular/genética , MicroRNAs/fisiologia , Neoplasias Nasofaríngeas/patologia , Proteínas Serina-Treonina Quinases/genética , Proteínas Proto-Oncogênicas/genética , Animais , Apoptose , Proteínas de Ciclo Celular/antagonistas & inibidores , Sobrevivência Celular/efeitos da radiação , Feminino , Regulação da Expressão Gênica , Histonas/análise , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Neoplasias Nasofaríngeas/enzimologia , Neoplasias Nasofaríngeas/terapia , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Proteínas Proto-Oncogênicas/antagonistas & inibidores , RNA Mensageiro/análise , RNA Interferente Pequeno/genética , Quinase 1 Polo-Like
5.
Clin Cancer Res ; 14(15): 4891-7, 2008 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-18676763

RESUMO

PURPOSE: Nasopharyngeal carcinoma (NPC) is a malignancy of the head and neck region that is associated with EBV latency. Curative treatments for NPC achieve modest survival rates, underscoring a need to develop novel therapies. We evaluated the therapeutic potential of a mutant vesicular stomatitis virus (VSVDelta51) as single treatment modality or in combination with ionizing radiation (RT) in NPC. EXPERIMENTAL DESIGN: MTS assay was used to assess cell viability in vitro; apoptosis was measured using propidium iodide staining and caspase activation. In vivo experiments were conducted using tumor-bearing nude mice with or without local RT (4 Gy). Apoptosis was assessed in excised tumor sections with terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling staining. RESULTS: Our data showed that NPC cells are exquisitely sensitive to VSVDelta51 oncolysis, which correlated with the presence of EBV. Efficacy of VSVDelta51 against NPC cells was further augmented when combined with RT. A single systemic injection of VSVDelta51 achieved 50% survival in treated mice, which increased to 83% when combined with local tumor RT. In addition to induction of apoptosis, an antiangiogenic effect of VSVDelta51 was observed in vivo, suggesting a novel tumoricidal mechanism for VSVDelta51. This virus also prevented growth of NPC sphere-forming cells in vitro, showing potential utility in targeting NPC-initiating cells. CONCLUSIONS: Our data represent the first report showing that EBV-positive NPC cells are exquisitely sensitive to VSVDelta51 oncolysis and documenting the successful utilization of this combinatorial regimen as a novel curative therapeutic strategy for NPC.


Assuntos
Carcinoma/terapia , Mutação , Neoplasias Nasofaríngeas/terapia , Vesiculovirus/metabolismo , Animais , Apoptose , Carcinoma/radioterapia , Linhagem Celular Tumoral , Sobrevivência Celular , Terapia Combinada/métodos , Humanos , Masculino , Camundongos , Camundongos Nus , Neoplasias Nasofaríngeas/radioterapia , Transplante de Neoplasias , Resultado do Tratamento , Vesiculovirus/genética
6.
Biotechniques ; 44(2): ix-xiv, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18422490

RESUMO

The colony formation assay (CFA) is the gold standard for measuring the effects of cytotoxic agents on cancer cells in vitro; however, in its traditional 6-well format, it is a time-consuming assay, particularly when evaluating combination therapies. In the interest of increased efficiency, the 6-well CFA was converted to a 96-well format using an automated colony counting algorithm. The 96-well CFA was validated using ionizing radiation therapy on the FaDu (human hypopharyngeal squamous cell) and A549 (human lung) cancer cell lines. Its ability to evaluate combination therapies was investigated by the generation of dose-response curves for the combination of cisplatin and radiation therapy on FaDu and A549 cells. The 96-well CFA was then transferred to a robotic platform for evaluating its potential as a high-throughput screening (HTS) readout. The LOPAC1280 library was screened against FaDu cells, and eight putative hits were identified. Using the 96-well CFA to validate the eight putative chemicals, six of the eight were confirmed, resulting in a positive hit rate of 75%. These data indicate that the 96-well CFA can be adopted as an efficient alternative assay to the 6-well CFA in evaluating single and combination therapies in vitro, providing a possible readout that could be used on a HTS platform.


Assuntos
Ensaio Tumoral de Célula-Tronco/instrumentação , Ensaio Tumoral de Célula-Tronco/métodos , Linhagem Celular Tumoral , Cisplatino/farmacologia , Terapia Combinada , Relação Dose-Resposta a Droga , Relação Dose-Resposta à Radiação , Humanos , Reprodutibilidade dos Testes , Bibliotecas de Moléculas Pequenas/análise , Ensaio Tumoral de Célula-Tronco/normas
7.
Photochem Photobiol Sci ; 7(6): 675-80, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18528551

RESUMO

Firefly luciferase catalyzes the emission of light from luciferin in the presence of oxygen and adenosine triphosphate. This bioluminescence is commonly employed in imaging mode to monitor tumor growth and treatment responses in vivo. A potential concern is that, since solid tumors are often hypoxic, either constitutively and/or as a result of treatment, the oxygen available for the bioluminescence reaction could be reduced to limiting levels, leading to underestimation of the actual number of luciferase-labeled cells during in vivo experiments. We present studies of the oxygen dependence of bioluminescence in vitro in rat 9 L gliosarcoma cells tagged with the firefly luciferase gene (9L(luc)). We demonstrate that the bioluminescence signal decreases at pO(2)

Assuntos
Gliossarcoma/metabolismo , Luciferases/metabolismo , Transgenes/genética , Trifosfato de Adenosina/metabolismo , Animais , Hipóxia Celular , Linhagem Celular Tumoral , Gliossarcoma/genética , Luciferases/genética , Medições Luminescentes , Oxigênio , Ratos , Transfecção
8.
Int J Nanomedicine ; 2(3): 461-78, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-18019844

RESUMO

Copolymers of 2-dimethyl(aminoethyl) methacrylate (PDMAEM) with N-isopropylacrylamide (NIPAM) were evaluated for their potential to enhance transgene expression of plasmid DNA (pDNA) and gene delivery by adenovirus vectors. The polymers of varying compositions and molecular weights (MW) were synthesized by free-radical polymerization. Polyelectrolyte complexes (PECs) were prepared with different charge (N:P) ratios of PNIPAM/ DMAEM to pDNA. Polymer-modified viral vectors based on non-replicating adenovirus serotype 5 (Ad5), (deltaE1/oriP/luc) or (deltaE1/CMV/luc) transcriptor/promoter/reporter were constructed by electrostatically coupling PNIPAM/DMAEM (Type 1) or PECs (oriP/luc, 6.6 kb) (Type II) to the viral capsid. The N:P value at complete condensation was lower for PECs with higher DMAEM content and MW. pDNA binding was enhanced by high MW PNIPAM/DMAEM. Circular dichroism spectroscopy revealed changes to the secondary structure of pDNA and adenovirus capsid proteins in the presence of PNIPAM/DMAEM. The toxicity of PNIPAM/DMAEM to CNE-1 nasopharyngeal cancer (NPC) cells diminished with decreasing DMAEM content and increasing MW The transfection efficiency ofC666-1 NPC cells by PECs increased with DMAEM content and MW. The transduction efficiency of CNE-1 NPC cells by Type I Ad5 vectors improved with DMAEM content, but was independent of MW. The transduction efficiency of Type II Ad5 in C666-1 cells approximated the sum of expression levels of the PECs and Ad5 vectors individually. PDMAEM and PNIPAM/DMAEM demonstrate both transfection and transduction enhancement activity of modified vectors in nasopharyngeal cancer cells in culture.


Assuntos
Resinas Acrílicas/química , Adenoviridae/genética , Portadores de Fármacos/química , Marcação de Genes/métodos , Terapia Genética/métodos , Metacrilatos/química , Neoplasias Nasofaríngeas/genética , Nylons/química , Plasmídeos/administração & dosagem , Transfecção/métodos , Linhagem Celular Tumoral , Portadores de Fármacos/administração & dosagem , Vetores Genéticos/genética , Humanos , Neoplasias Nasofaríngeas/terapia , Plasmídeos/química
9.
Clin Cancer Res ; 13(19): 5935-41, 2007 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-17908990

RESUMO

PURPOSE: The tumor microenvironment is complex and heterogeneous, populated by tortuous irregular vasculature, hypoxic cells, and necrotic regions. These factors can all contribute to the biodistribution difficulties encountered by most cancer therapeutic agents. Antisense oligodeoxynucleotides (ASO) are a class of therapeutics where limited information is available about their distribution within a solid tumor environment. EXPERIMENTAL DESIGN: To assess ASO distribution, a fluorescein-labeled phosphorothionated ASO based on the G3139 mismatch control was injected systemically (i.v.) into tumor-bearing severe combined immunodeficient mice. Hoechst 33342 was injected i.v. to visualize active vasculature. Unstained sections were imaged through tiled fluorescence stereomicroscopy and then quantitated using novel algorithms. Tumor sections from four human tumor models were examined (CaSki, DU-145, C666-1, and C15) for hypoxia, apoptosis/necrosis, and morphology. RESULTS: For all four tumors, ASO accumulated within regions of hypoxia, necrosis, and apoptosis. Scatter plots of ASO versus active vasculature generated for each individual tumor revealed a consistent pattern of distribution of the ASO within each model. In C666-1 xenografts, the slopes of these scatter plots were significantly reduced from 0.41 to 0.16 when pretreated with the antivascular agent ZD6126 48 h before ASO injection. This was accompanied by the formation of large disseminated necrotic regions in the tumor, along with a 13.1 mmHg reduction in interstitial fluid pressure. CONCLUSIONS: These data suggest the possibility that these algorithms might offer a generalizable and objective methodology to describe the distribution of molecular therapeutic agents within a tumor microenvironment and to quantitatively assess distribution changes in response to combination therapies.


Assuntos
Diagnóstico por Imagem/métodos , Oligonucleotídeos Antissenso/farmacologia , Algoritmos , Animais , Benzimidazóis/farmacologia , Linhagem Celular Tumoral , Proliferação de Células , Humanos , Hipóxia , Camundongos , Camundongos SCID , Necrose , Transplante de Neoplasias , Compostos Organofosforados/farmacologia , Ensaios Antitumorais Modelo de Xenoenxerto
10.
Mol Ther ; 15(5): 921-9, 2007 May.
Artigo em Inglês | MEDLINE | ID: mdl-17356543

RESUMO

To explore systemic utilization of Epstein-Barr virus (EBV)-specific transcriptionally targeted adenoviruses, three vectors were constructed to examine kinetics, specificity, and biodistribution: adv.oriP.luc, expressing luciferase under EBV-specific control; adv.SV40luc, expressing luciferase constitutively; and adv.oriP.E1A.oriP.luc, a conditionally replicating adenovirus, expressing both luciferase and E1A. Bioluminescence imaging (BLI) was conducted on tumor-bearing severe combined immunodeficient (SCID) mice (C666-1, EBV-positive human nasopharyngeal cancer) treated intravenously (i.v.) with 3 x 10(8) infectious units (ifu) of the adenoviral vectors. At 72 hours, adv.oriPluc demonstrated an 8.4-fold higher tumor signal than adv.SV40luc; adv.oriP.E1A.oriP.luc was 26.7-fold higher; however, a significant liver signal was also observed, necessitating further action to improve biodistribution. Several compounds were examined to this end, including norepinephrine, serotonin, clodronate liposomes, and STI571, to determine whether any of these measures could improve adenoviral biodistribution. Each of these interventions was assessed using BLI in mice i.v. injected with adv.oriP.luc. STI571 achieved the highest increase in tumor-to-liver ratio (TLR; 6.6-fold), which was associated with a 59% reduction in tumor interstitial fluid pressure (IFP) along with a decrease in platelet-derived growth factor-beta receptor (PDGF beta R) activation. This study reports the favorable modulation by STI571 of the biodistribution of adenoviral vectors, providing a potential approach to improving therapeutic outcome.


Assuntos
Terapia Genética/métodos , Neoplasias/terapia , Proteínas Recombinantes de Fusão/farmacocinética , Adenoviridae/genética , Animais , Antineoplásicos/farmacologia , Benzamidas , Linhagem Celular Tumoral , Ácido Clodrônico/farmacologia , Vetores Genéticos/genética , Humanos , Mesilato de Imatinib , Imuno-Histoquímica , Cinética , Fígado/metabolismo , Luciferases/genética , Luciferases/metabolismo , Medições Luminescentes/métodos , Masculino , Camundongos , Camundongos SCID , Microscopia de Fluorescência , Neoplasias/metabolismo , Fosforilação/efeitos dos fármacos , Piperazinas/farmacologia , Pirimidinas/farmacologia , Receptor beta de Fator de Crescimento Derivado de Plaquetas/metabolismo , Proteínas Recombinantes de Fusão/administração & dosagem , Proteínas Recombinantes de Fusão/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fatores de Tempo , Distribuição Tecidual/efeitos dos fármacos
11.
Mol Ther ; 15(5): 921-929, 2007 May.
Artigo em Inglês | MEDLINE | ID: mdl-28182894

RESUMO

See page 841 To explore systemic utilization of Epstein-Barr virus (EBV)-specific transcriptionally targeted adenoviruses, three vectors were constructed to examine kinetics, specificity, and biodistribution: adv.oriP.luc, expressing luciferase under EBV-specific control; adv.SV40luc, expressing luciferase constitutively; and adv.oriP.E1A.oriP.luc, a conditionally replicating adenovirus, expressing both luciferase and E1A. Bioluminescence imaging (BLI) was conducted on tumor-bearing severe combined immunodeficient (SCID) mice (C666-1, EBV-positive human nasopharyngeal cancer) treated intravenously (i.v.) with 3 × 108 infectious units (ifu) of the adenoviral vectors. At 72 hours, adv.oriPluc demonstrated an 8.4-fold higher tumor signal than adv.SV40luc; adv.oriP.E1A.oriP.luc was 26.7-fold higher; however, a significant liver signal was also observed, necessitating further action to improve biodistribution. Several compounds were examined to this end, including norepinephrine, serotonin, clodronate liposomes, and STI571, to determine whether any of these measures could improve adenoviral biodistribution. Each of these interventions was assessed using BLI in mice i.v. injected with adv.oriP.luc. STI571 achieved the highest increase in tumor-to-liver ratio (TLR; 6.6-fold), which was associated with a 59% reduction in tumor interstitial fluid pressure (IFP) along with a decrease in platelet-derived growth factor-ß receptor (PDGFßR) activation. This study reports the favorable modulation by STI571 of the biodistribution of adenoviral vectors, providing a potential approach to improving therapeutic outcome.

12.
Clin Cancer Res ; 12(19): 5726-32, 2006 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-17020977

RESUMO

PURPOSE: Nasopharyngeal cancer (NPC) is a malignant epithelial carcinoma which is intimately associated with EBV. The latent presence of EBV affects the function of p53, Bcl-2, and survivin. We thus investigated the relationship between EBV status, p53, Bcl-2, and survivin in biopsy specimens from patients with primary NPC. EXPERIMENTAL DESIGN: Archival formalin-fixed, paraffin-embedded NPC biopsies were evaluated in 80 patients treated with curative radiation from a single institution. The presence of EBV was determined using EBER in situ hybridization, whereas p53, Bcl-2, and survivin were assessed using immunohistochemistry. RESULTS: The majority of NPC specimens in this patient cohort were EBER-positive (64 of 78, or 82%), which in turn, was significantly associated with ethnicity (P = 0.0007), and WHO subtype 2A/2B (P = 0.04). EBER-positive tumors were also associated with p53 (P = 0.002), Bcl-2 (P = 0.04), and nuclear survivin (P = 0.03) expression. Patients with EBER-positive NPC fared better, with a 10-year overall survival of 68% versus 48% for EBER-negative patients (P = 0.03). For nuclear survivin, patients with either low or high nuclear survivin fared worse than patients with intermediate survivin expression (P = 0.05), suggesting that there is an optimal proportion of survivin-expressing cells for best function and clinical outcome. CONCLUSIONS: With an extended median follow-up time of 11.4 years, EBV status remains a strong predictor for overall survival in NPC. EBV-positive NPC has strong molecular associations with p53, Bcl-2, and survivin expression. Furthermore, we provide clinical data revealing the potentially dual nature of survivin in predicting clinical outcome.


Assuntos
Infecções por Vírus Epstein-Barr/metabolismo , Regulação Neoplásica da Expressão Gênica , Proteínas Associadas aos Microtúbulos/metabolismo , Neoplasias Nasofaríngeas/metabolismo , Proteínas de Neoplasias/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/virologia , DNA de Neoplasias/genética , Infecções por Vírus Epstein-Barr/virologia , Feminino , Herpesvirus Humano 4/genética , Herpesvirus Humano 4/isolamento & purificação , Humanos , Técnicas Imunoenzimáticas , Proteínas Inibidoras de Apoptose , Masculino , Pessoa de Meia-Idade , Neoplasias Nasofaríngeas/virologia , Prognóstico , Taxa de Sobrevida , Survivina
13.
Mol Cancer Ther ; 5(9): 2234-40, 2006 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16985057

RESUMO

Despite advances in surgery, radiation, and chemotherapy, novel therapeutics are needed for head and neck cancer treatment. The objective of this current study was to evaluate alexidine dihydrochloride as a novel compound lead for head and neck cancers. Using a tetrazolium-based assay, the dose required to reduce cell viability by 50% (ED50) was found to be approximately 1.8 micromol/L in FaDu (human hypopharyngeal squamous cancer) and approximately 2.6 micromol/L in C666-1 (human undifferentiated nasopharyngeal cancer) cells. In contrast, the ED50 values were much higher in untransformed cells, specifically at approximately 8.8 micromol/L in GM05757 (primary normal human fibroblast), approximately 8.9 micromol/L in HNEpC (primary normal human nasal epithelial), and approximately 19.6 micromol/L in NIH/3T3 (mouse embryonic fibroblast) cells. Alexidine dihydrochloride did not interfere with the activities of cisplatin, 5-fluorouracil, or radiation, and interacted in a less-than-additive manner. DNA content analyses and Hoechst 33342 staining revealed that this compound induced apoptosis. Alexidine dihydrochloride-induced mitochondrial damage was visualized using transmission electron microscopy. Mitochondrial membrane potential (DeltaPsiM) depolarization was detectable after only 3 hours of treatment, and was followed by cytosolic Ca2+ increase along with loss of membrane integrity/cell death. Caspase-2 and caspase-9 activities were detectable at 12 hours, caspase-8 at 24 hours, and caspase-3 at 48 hours. FaDu cell clonogenic survival was reduced to < 5% with 1 micromol/L alexidine dihydrochloride, and, correspondingly, this compound decreased the in vivo tumor-forming potential of FaDu cells. Thus, we have identified alexidine dihydrochloride as the first bisbiguanide compound with anticancer specificity.


Assuntos
Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Biguanidas/farmacologia , Carcinoma de Células Escamosas/tratamento farmacológico , Neoplasias Hipofaríngeas/tratamento farmacológico , Animais , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Biguanidas/administração & dosagem , Carcinoma de Células Escamosas/patologia , Caspase 2/metabolismo , Caspase 9/metabolismo , Cisplatino/administração & dosagem , Ativação Enzimática/efeitos dos fármacos , Feminino , Fluoruracila/administração & dosagem , Humanos , Neoplasias Hipofaríngeas/patologia , Potenciais da Membrana/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos SCID , Membranas Mitocondriais/efeitos dos fármacos , Membranas Mitocondriais/fisiologia , Células NIH 3T3 , Ensaios Antitumorais Modelo de Xenoenxerto
14.
Clin Cancer Res ; 12(18): 5557-69, 2006 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-17000693

RESUMO

PURPOSE: This study aims to identify a novel therapeutic agent for head and neck cancer and to evaluate its antitumor efficacy. EXPERIMENTAL DESIGN: A cell-based and phenotype-driven high-throughput screening of approximately 2,400 biologically active or clinically used compounds was done using a tetrazolium-based assay on FaDu (hypopharyngeal squamous cancer) and NIH 3T3 (untransformed mouse embryonic fibroblast) cells, with secondary screening done on C666-1 (nasopharyngeal cancer) and GM05757 (primary normal human fibroblast) lines. The "hit" compound was assayed for efficacy in combination with standard therapeutics on a panel of human cancer cell lines. Furthermore, its mode of action (using transmission electron microscopy and flow cytometry) and its in vivo efficacy (using xenograft models) were evaluated. RESULTS: Benzethonium chloride was identified as a novel cancer-specific compound. For benzethonium (48-hour incubation), the dose required to reduce cell viability by 50% was 3.8 micromol/L in FaDu, 42.2 micromol/L in NIH 3T3, 5.3 micromol/L in C666-1, and 17.0 micromol/L in GM05757. In vitro, this compound did not interfere with the effects of cisplatin, 5-fluorouracil, or gamma-irradiation. Benzethonium chloride induced apoptosis and activated caspases after 12 hours. Loss of mitochondrial membrane potential (DeltaPsiM) preceded cytosolic Ca2+ increase and cell death. In vivo, benzethonium chloride ablated the tumor-forming ability of FaDu cells, delayed the growth of xenograft tumors, and combined additively with local tumor radiation therapy. Evaluation of benzethonium chloride on the National Cancer Institute/NIH Developmental Therapeutics Program 60 human cancer cell lines revealed broad-range antitumor activity. CONCLUSIONS: This high-throughput screening identified a novel antimicrobial compound with significant broad-spectrum anticancer activity.


Assuntos
Antineoplásicos/isolamento & purificação , Benzetônio/isolamento & purificação , Ensaios de Seleção de Medicamentos Antitumorais/métodos , Análise Serial de Tecidos/métodos , Animais , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Apoptose/efeitos dos fármacos , Benzetônio/farmacologia , Benzetônio/uso terapêutico , Cálcio/metabolismo , Caspases/metabolismo , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/efeitos da radiação , Sobrevivência Celular/efeitos dos fármacos , Quimioterapia Combinada , Feminino , Neoplasias de Cabeça e Pescoço/tratamento farmacológico , Neoplasias de Cabeça e Pescoço/radioterapia , Humanos , Potenciais da Membrana/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos SCID , Membranas Mitocondriais/efeitos dos fármacos , Modelos Biológicos , Células NIH 3T3 , Neoplasias/tratamento farmacológico , Neoplasias/radioterapia , Radiossensibilizantes/farmacologia , Radiossensibilizantes/uso terapêutico , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto/métodos
15.
Clin Cancer Res ; 11(22): 8131-44, 2005 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-16299246

RESUMO

PURPOSE: A wide variety of tumors depend on the dysregulation of Bcl-2 family proteins for survival. The resulting apoptotic block can often provide a mechanism for resistance to anticancer treatments, such as chemotherapy and radiation. This current study evaluates the efficacy of combining systemically delivered Bcl-2 phosphorothioate antisense (Bcl-2 ASO) and radiation for nasopharyngeal cancer therapy. RESULTS: Antisense uptake was unaffected by 0, 3, or 6 Gy radiation. Radiation decreased the fraction of viable C666-1 cells to 60%, with a further decrease to 40% in combination with Bcl-2 ASO. Despite a modest in vitro effect, Bcl-2 ASO alone caused the regression of established xenograft tumors in mice, extending survival by 15 days in a C666-1 and by 6 days in a C15 model. The survival times for mice treated with both Bcl-2 ASO and radiation increased by 52 days in C666-1 and by 20 days in C15 tumors. This combination resulted in a more-than-additive effect in C666-1 tumors. Less impressive gains observed in C15 tumors might be attributable to higher expression of antiapoptotic Bcl-2 family proteins and limited drug distribution in the tumor. Retreatment of C666-1 tumors with the Bcl-2 ASO-radiation combination, however, was effective, resulting in mice surviving for >80 days relative to untreated controls. CONCLUSIONS: Our results show that the Bcl-2 ASO and radiation combination is a highly potent therapy for nasopharyngeal cancer. Further examination of combination therapy with radiation and other Bcl-2 family-targeted anticancer agents in both preclinical and clinical settings is definitely warranted.


Assuntos
Neoplasias Nasofaríngeas/tratamento farmacológico , Neoplasias Nasofaríngeas/radioterapia , Oligonucleotídeos Antissenso/farmacologia , Proteínas Proto-Oncogênicas c-bcl-2/genética , Animais , Apoptose/efeitos dos fármacos , Apoptose/efeitos da radiação , Vasos Sanguíneos/metabolismo , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos da radiação , Terapia Combinada , Feminino , Fluoresceína-5-Isotiocianato/química , Fluoresceína-5-Isotiocianato/farmacocinética , Humanos , Rim/metabolismo , Fígado/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos SCID , Microscopia de Fluorescência , Neoplasias Nasofaríngeas/patologia , Oligonucleotídeos Antissenso/genética , Oligonucleotídeos Antissenso/farmacocinética , Radiação Ionizante , Distribuição Tecidual , Resultado do Tratamento , Ensaios Antitumorais Modelo de Xenoenxerto/métodos
16.
Mol Ther ; 10(3): 533-44, 2004 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-15336653

RESUMO

We have previously demonstrated a 1000-fold induction of gene expression exclusive to Epstein-Barr virus (EBV)-positive nasopharyngeal carcinoma (NPC) cells using an adenoviral vector (ad5.oriP). This platform allows us to explore tumor-specific gene therapy with BimS (ad5.oriP.BimS), a potent proapoptotic Bcl-2 family member. Ad5.oriP.BimS (25 infectious units (ifu)/cell) reduced C666-1 viability in a time- and dose-dependent manner to 15% survival. The effect was enhanced with radiation (6 Gy). Three days after infection, the proportion of apoptotic cells increased from 3.5% (control) to 47.5% (25 ifu/cell). Confocal microscopy demonstrated Bim colocalization to the mitochondria within 18 h of ad5.oriP.BimS infection. Ad5.oriP.BimS induced a 2.8-, 2.1-, and 1.85-fold increase in caspase-3, -8, and -9 activities, respectively. When C666-1 cells were infected with ad5.oriP.BimS (20 ifu/cell), no tumors formed in 7/9 mice followed for 100 days. Six intratumoral injections of ad5.oriP.BimS (1.5 x 10(9) ifu/dose) in combination with radiation were sufficient to cause almost complete disappearance of established C666-1 or C15 xenograft tumors. Intravenous injections of ad5.oriP.BimS (10(9) ifu) induced mild perturbation in liver function tests, associated with hepatocyte apoptoses and mitoses. This vector appears to be safe and effectively cytotoxic to EBV-positive NPC cells both in vitro and in vivo, mediated primarily through the induction of apoptosis.


Assuntos
Apoptose , Terapia Genética , Neoplasias Nasofaríngeas/terapia , Proteínas Proto-Oncogênicas c-bcl-2/genética , Adenoviridae/genética , Adenoviridae/metabolismo , Animais , Caspases/metabolismo , Linhagem Celular , Terapia Combinada , Ativação Enzimática , Raios gama , Terapia Genética/efeitos adversos , Herpesvirus Humano 4 , Humanos , Camundongos , Microscopia Confocal , Mitocôndrias/metabolismo , Neoplasias Nasofaríngeas/genética , Neoplasias Nasofaríngeas/virologia , Regiões Promotoras Genéticas , Proteínas Proto-Oncogênicas c-bcl-2/biossíntese , Transplante Heterólogo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...