Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
2.
Clin Cancer Res ; 13(10): 2890-6, 2007 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-17504988

RESUMO

PURPOSE: EGFR gene mutations and increased EGFR copy number have been associated with favorable response to epidermal growth factor receptor (EGFR) tyrosine kinase inhibitors (EGFR-TKI) in patients with non-small-cell lung cancer (NSCLC). In contrast, KRAS mutation has been shown to predict poor response to such therapy. We tested the utility of combinations of these three markers in predicting response and survival in patients with NSCLC treated with EGFR-TKIs. EXPERIMENTAL DESIGN: Patients with advanced NSCLC treated with EGFR-TKI with available archival tissue specimens were included. EGFR and KRAS mutations were analyzed using PCR-based sequencing. EGFR copy number was analyzed using fluorescence in situ hybridization. RESULTS: The study included 73 patients, 59 of whom had all three potential markers successfully analyzed. EGFR mutation was detected in 7 of 71 patients (9.8%), increased EGFR copy number in 32 of 59 (54.2%), and KRAS mutation in 16 of 70 (22.8%). EGFR mutation (P<0.0001) but not increased EGFR copy number (P=0.48) correlated with favorable response. No survival benefit was detected in patients with either of these features. KRAS mutation correlated with progressive disease (P=0.04) and shorter median time to progression (P=0.0025) but not with survival. Patients with both EGFR mutation and increased EGFR copy number had a >99.7% chance of objective response, whereas patients with KRAS mutation with or without increased EGFR copy number had a >96.5% chance of disease progression. CONCLUSION: KRAS mutation should be included as indicator of resistance in the panel of markers used to predict response to EGFR-TKIs in NSCLC.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Resistencia a Medicamentos Antineoplásicos/genética , Receptores ErbB/antagonistas & inibidores , Neoplasias Pulmonares/tratamento farmacológico , Inibidores de Proteínas Quinases/uso terapêutico , Proteínas Proto-Oncogênicas/genética , Proteínas ras/genética , Progressão da Doença , Receptores ErbB/genética , Cloridrato de Erlotinib , Feminino , Gefitinibe , Dosagem de Genes , Humanos , Masculino , Pessoa de Meia-Idade , Mutação , Prognóstico , Proteínas Proto-Oncogênicas p21(ras) , Quinazolinas/uso terapêutico , Resultado do Tratamento
3.
Cancer ; 109(11): 2357-64, 2007 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-17457827

RESUMO

BACKGROUND: Despite recommendations to immunize all patients at an increased risk of influenza complications, the vaccine utilization among high-risk nonelderly adults remains low and its cost-effectiveness is unclear. In the current study, the authors analyzed the cost-effectiveness of influenza vaccination in working-age (ages 20-64 years) cancer patients. METHODS: The authors developed a decision-analytic model, from the societal perspective, using epidemiologic, vaccine effectiveness, resource utilization, cost, survival, and utility data from published sources, supplemented with data collected from the authors' own institutional accounting system. Two strategies were compared: influenza vaccination of working-age cancer patients and no vaccination. The base-case patient was assumed to be a 51-year-old cancer patient (the mean age for the National Cancer Institute's Surveillance, Epidemiology, and End Results [SEER] population of working-age patients within 5 years of cancer diagnosis). RESULTS: The effectiveness of the influenza vaccine was 6.02 quality-adjusted life-years (QALYs) at a cost of $30.10. The effectiveness of the no vaccination strategy was 6.01 QALYs at a cost of $27.86. Compared with the no vaccination strategy, the incremental cost-effectiveness ratio of vaccinating working-age cancer patients would be $224.00 per QALY gained. Using the benchmark of $50,000 per QALY, the model was only sensitive to changes in cancer survival (threshold of 2.8 months). CONCLUSIONS: The influenza vaccine is cost-effective for working-age cancer patients with a life expectancy of >or=3 months. All working-age cancer patients who are within 5 years of cancer diagnosis and have a life expectancy of at least 3 months should be vaccinated against influenza.


Assuntos
Vacinas contra Influenza/economia , Influenza Humana/economia , Neoplasias/patologia , Vacinação/economia , Adulto , Distribuição por Idade , Análise Custo-Benefício , Técnicas de Apoio para a Decisão , Feminino , Política de Saúde/economia , Humanos , Vacinas contra Influenza/administração & dosagem , Influenza Humana/prevenção & controle , Expectativa de Vida , Masculino , Pessoa de Meia-Idade , Modelos Econométricos , Anos de Vida Ajustados por Qualidade de Vida , Estados Unidos/epidemiologia
4.
Mol Ther ; 15(2): 287-94, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17235306

RESUMO

Bevacizumab, a humanized monoclonal antibody against vascular endothelial growth factor (VEGF), has shown antitumor activity by inhibiting tumor angiogenesis in preclinical and clinical studies. However, bevacizumab monotherapy does not induce complete tumor regression. Therefore, additional treatments must be combined with bevacizumab to promote tumor regression. We previously showed that melanoma differentiation associated gene-7 (mda-7) protein exerts potent antitumor and antiangiogenic activity. Thus, in this study, we investigated the therapeutic effects of mda-7 in combination with bevacizumab using lung cancer as a model. In vitro, treatment of human umbilical vein endothelial cells with conditioned medium from Ad-mda7 plus bevacizumab-treated lung tumor cells showed reduced VEGF ligand-receptor binding, and decreased cell survival, resulting in growth arrest and apoptosis. In vivo, treatment of subcutaneous lung tumor xenografts with bevacizumab plus Ad-mda7 resulted in significant tumor growth inhibition and improved survival compared to tumor growth in control mice. Furthermore, tumors in all the Ad-mda7 plus bevacizumab-treated mice completely regressed, and these were tumor free through the study's end. Molecular analysis showed enhanced tumor cell apoptosis and reduced VEGF and CD31 expression in Ad-mda7 plus bevacizumab-treated tumors. Thus, Ad-mda7 and bevacizumab treatment produces a synergistic and complete therapeutic effect against human lung cancer.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Interleucinas/genética , Neoplasias Pulmonares/terapia , Ensaios Antitumorais Modelo de Xenoenxerto , Adenoviridae/genética , Inibidores da Angiogênese/farmacologia , Inibidores da Angiogênese/uso terapêutico , Animais , Anticorpos Monoclonais/farmacologia , Anticorpos Monoclonais Humanizados , Apoptose/efeitos dos fármacos , Apoptose/genética , Bevacizumab , Western Blotting , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/genética , Terapia Combinada , Ensaio de Imunoadsorção Enzimática , Feminino , Terapia Genética/métodos , Humanos , Imuno-Histoquímica , Interleucinas/fisiologia , Neoplasias Pulmonares/patologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Molécula-1 de Adesão Celular Endotelial a Plaquetas/análise , Fator A de Crescimento do Endotélio Vascular/análise , Fator A de Crescimento do Endotélio Vascular/imunologia
5.
Clin Genitourin Cancer ; 5(2): 144-9, 2006 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17026803

RESUMO

BACKGROUND: This is a phase I/II trial of thalidomide with estramustine and paclitaxel in men with androgen-independent prostate cancer (AIPC) who underwent previous chemotherapy. PATIENTS AND METHODS: Men with progressive AIPC were treated with oral thalidomide (200 mg, 400 mg, or 600 mg daily), intravenous paclitaxel (100 mg/m2 over 3 hours on days 3 and 10), and oral estramustine (140 mg 3 times daily on days 1-5 and days 8-12) every 21 days. RESULTS: Phase I: first cycle dose-limiting toxicity occurred in 0 of 3 patients at 200 mg thalidomide daily, 0 of 3 at 400 mg daily, and 1 of 3 at 600 mg daily (the designated maximum tolerated dose). Phase II: twenty-nine of 38 evaluable patients (76%; 95% confidence interval, 67%-87%) experienced a 50% decrease in prostate-specific antigen level. Five of 18 patients (28%) with measurable disease exhibited an objective response. Nine of 14 patients (64%) with disease refractory to previous taxane therapy had 50% decreases in prostate-specific antigen level. Grade 3/4 adverse events included neutropenia (9 of 39 [23%]), fatigue (9 of 39 [23%]), dyspnea (8 of 39 [21%]), and thromboembolic events (7 of 39 [18%]). Cumulative dose-limiting toxicity rates were minimal (13%) with thalidomide at 200 mg daily. CONCLUSION: The profile of activity of thalidomide/paclitaxel/estramustine in taxane-refractory AIPC warrants further investigation.


Assuntos
Adenocarcinoma/tratamento farmacológico , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Neoplasias da Próstata/tratamento farmacológico , Adenocarcinoma/secundário , Idoso , Idoso de 80 Anos ou mais , Hidrocarbonetos Aromáticos com Pontes/efeitos adversos , Progressão da Doença , Estramustina/administração & dosagem , Humanos , Masculino , Pessoa de Meia-Idade , Neoplasias Hormônio-Dependentes/tratamento farmacológico , Paclitaxel/administração & dosagem , Antígeno Prostático Específico/sangue , Neoplasias da Próstata/patologia , Taxa de Sobrevida , Taxoides/efeitos adversos , Talidomida/administração & dosagem , Fatores de Tempo , Resultado do Tratamento
6.
Mol Cancer Ther ; 4(8): 1146-56, 2005 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16093430

RESUMO

We investigated integrin-linked kinase (ILK), a focal adhesion serine-threonine protein kinase, as a new molecular target for treating anaplastic thyroid cancer. ILK mediates cell growth and survival signals and is overexpressed in a number of cancers. Therefore, we hypothesized that inhibition of ILK leads to growth arrest and apoptosis of thyroid cancer cells. According to Western blotting, the level of ILK protein was highly expressed in one papillary (NPA187) and four of five (Hth74, DRO, ARO, KAT4, and K4) anaplastic thyroid cancer cell lines. Immunohistochemical analysis of a human tissue microarray revealed that ILK was highly expressed in anaplastic thyroid cancer but not in normal human thyroid tissue. Treating thyroid cancer cell lines with a new ILK inhibitor, QLT0267, inhibited epidermal growth factor-induced phosphorylation of AKT, inhibited cell growth, and induced apoptosis in the NPA187, DRO, and K4 cell lines. QLT0267 also inhibited the kinase activity of immunoprecipitated ILK in four of five cell lines. Tumor volumes in mice treated with QLT0267 were significantly reduced compared with those in untreated mice. In immunohistochemical studies, QLT0267 suppressed phosphorylated p-AKT and angiogenesis (i.e., reduced mean vascular density) and induced apoptosis in both tumor cells and tumor-associated endothelial cells of the thyroid DRO xenografts. In summary, we found that ILK expression and activity were elevated in human anaplastic thyroid cancer and ILK inhibition led to growth arrest and apoptosis in vitro and in vivo. Our results provide preliminary evidence that ILK is a potential therapeutic target for treating anaplastic thyroid cancer.


Assuntos
Antineoplásicos/uso terapêutico , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Neoplasias da Glândula Tireoide/tratamento farmacológico , Neoplasias da Glândula Tireoide/enzimologia , Animais , Antineoplásicos/farmacologia , Apoptose , Células Endoteliais/efeitos dos fármacos , Fator de Crescimento Epidérmico/metabolismo , Humanos , Masculino , Camundongos , Neovascularização Patológica/tratamento farmacológico , Fosforilação/efeitos dos fármacos , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-akt , Neoplasias da Glândula Tireoide/irrigação sanguínea , Análise Serial de Tecidos , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
7.
Cancer ; 104(3): 618-28, 2005 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-15973737

RESUMO

BACKGROUND: Although patients with cancer generally respond favorably to vaccination, they may not receive annual influenza vaccinations. The current population-based study described the epidemiology and outcomes of potentially preventable, serious influenza-related infections in patients with cancer. METHODS: From the Nationwide Inpatient Sample, the authors created a subsample that included discharges with any International Classification of Diseases, ninth revision, diagnosis code for cancer and principal diagnosis code for influenza, bronchopneumonia, or pneumonia caused by an unspecified organism. From the latter two diagnosis codes, the authors estimated excess cases during the influenza season for each year and stratum, then selected a random sample from fall and winter discharges. Subset analyses included weighted sample means, frequencies, and analysis of variance values. The authors converted charges to costs using cost-to-charge ratios and inflated these to 2003 U.S. dollars. Hospitalization and mortality rates were calculated using 5-year cancer prevalence estimates. RESULTS: The estimated mean annual hospital discharges of patients with cancer with potentially preventable, serious influenza-related infections numbered 16,000. The average length and cost per stay were 6 days and > USD 6300, respectively. Approximately 9% of patients died in the hospital and 31% needed further skilled care. The estimated age-specific rates for hospitalization and death per 100,000 in the prevalent cancer population were 219 and 17.4, respectively, for patients age < 65 years and 623 and 59.4, respectively, for those age > or = 65 years. Hospitalization costs averaged USD 1300 more for patients age < 65 years. CONCLUSIONS: Death from influenza-related infections occurred in an estimated 9% of patients with cancer hospitalized for such. Using recommended vaccination schedules for patients with cancer and their contacts reduced hospitalizations, treatment delays, and deaths in this highly susceptible population.


Assuntos
Hospitalização/estatística & dados numéricos , Influenza Humana/epidemiologia , Neoplasias/epidemiologia , Neoplasias/virologia , Orthomyxoviridae/patogenicidade , Adolescente , Adulto , Distribuição por Idade , Idoso , Idoso de 80 Anos ou mais , Criança , Pré-Escolar , Humanos , Lactente , Recém-Nascido , Influenza Humana/prevenção & controle , Tempo de Internação , Pessoa de Meia-Idade , Neoplasias/complicações , Fatores de Risco , Estados Unidos , Vacinação
8.
Clin Cancer Res ; 11(5): 1713-21, 2005 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-15755992

RESUMO

PURPOSE: To develop an orthotopic model of anaplastic thyroid carcinoma (ATC) in athymic nude mice. EXPERIMENTAL DESIGN: Various thyroid carcinoma cell lines were injected into the thyroid gland of athymic nude mice to determine whether such injection was technically feasible. ATC cells were then injected into the thyroid gland or the subcutis of nude mice at various concentrations, and the mice were then followed for tumor development. The tumors were examined histopathologically for local invasion or regional or distant metastasis. RESULTS: Injection of tumor cells into the thyroid glands of nude mice was technically feasible and resulted in the formation of thyroid tumors. The ATC cell line DRO showed significantly higher tumorigenicity in the thyroid gland than in the subcutis. In contrast, oral squamous cell carcinoma cell line TU167 shows no significantly higher tumorigenicity in the thyroid gland than in the subcutis. ATC tumors established in the thyroid gland also produced symptomatic compression of the esophagus and the trachea. Local invasion of the larynx and trachea was as well as high rates of pulmonary metastasis were also observed. Immunohistochemical staining showed higher microvessel density as well as higher expression of vascular endothelial growth factor and interleukin-8 in the orthotopic thyroid tumors than in ectopic tumors. CONCLUSION: An orthotopic model of ATC in athymic nude mice was developed that closely recapitulates the clinical findings of human ATC. This model should facilitate the understanding of the pathogenesis of ATC and aid in the development of novel therapies against ATC.


Assuntos
Carcinoma/patologia , Carcinoma/veterinária , Modelos Animais de Doenças , Neoplasias da Glândula Tireoide/patologia , Neoplasias da Glândula Tireoide/veterinária , Animais , Regulação Neoplásica da Expressão Gênica , Humanos , Imuno-Histoquímica , Masculino , Camundongos , Camundongos Nus , Invasividade Neoplásica , Metástase Neoplásica , Transplante Heterólogo , Células Tumorais Cultivadas
9.
Clin Cancer Res ; 11(5): 1963-73, 2005 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-15756022

RESUMO

PURPOSE: We investigated whether concomitant blockade of the epidermal growth factor receptor (EGFR) and vascular endothelial growth factor receptor (VEGFR) signaling pathways by AEE788, a dual inhibitor of EGFR and VEGFR tyrosine kinases, would inhibit the growth of cutaneous squamous cell carcinoma (SCC) cells and human cutaneous cancer xenografts in nude mice. EXPERIMENTAL DESIGN: We examined the effects of AEE788 on the phosphorylation of EGFR and VEGFR-2 in cutaneous SCC cells expressing EGFR and VEGFR-2 and cutaneous SCC cell growth and apoptosis. We assessed the in vivo antitumor effects of AEE788 in a xenograft model in nude mice. AEE788 (50 mg/kg) was given orally thrice weekly to mice that had been s.c. injected with Colo16 tumor cells. Mechanisms of in vivo AEE788 activity were determined by immunohistochemical analysis. RESULTS: Treatment of cutaneous SCC cells with AEE788 led to dose-dependent inhibition of EGFR and VEGFR-2 phosphorylation, growth inhibition, and induction of apoptosis. In mice treated with AEE788, tumor growth was inhibited by 54% at 21 days after the start of treatment compared with control mice (P < 0.01). Immunohistochemical analysis revealed that AEE788 inhibited phosphorylation of EGFR and VEGFR and induced apoptosis of tumor cells and tumor-associated endothelial cells. CONCLUSIONS: In addition to inhibiting cutaneous cancer cell growth by blocking EGFR and VEGFR signaling pathways in vitro, AEE788 inhibited in vivo tumor growth by inducing tumor and endothelial cell apoptosis.


Assuntos
Apoptose , Carcinoma de Células Escamosas/patologia , Purinas/farmacologia , Neoplasias Cutâneas/patologia , Administração Oral , Animais , Proliferação de Células , Células Endoteliais , Receptores ErbB/antagonistas & inibidores , Humanos , Camundongos , Camundongos Nus , Purinas/administração & dosagem , Receptores de Fatores de Crescimento do Endotélio Vascular/antagonistas & inibidores , Transdução de Sinais , Transplante Heterólogo
10.
Clin Cancer Res ; 10(24): 8594-602, 2004 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-15623643

RESUMO

PURPOSE: No effective treatment options currently are available to patients with anaplastic thyroid cancer (ATC), resulting in high mortality rates. Epidermal growth factor (EGF) has been shown to play a role in the pathogenesis of many types of cancer, and its receptor (EGFR) provides an attractive target for molecular therapy. EXPERIMENTAL DESIGN: The expression of EGFR was determined in ATC in vitro and in vivo and in human tissue arrays of ATC. We assessed the potential of the EGFR inhibitor gefitinib ("Iressa," ZD1839) to inhibit EGFR activation in vitro and in vivo, inhibit ATC cellular proliferation, induce apoptosis, and reduce the growth of ATC cells in vivo when administered alone and in combination with paclitaxel. RESULTS: EGFR was overexpressed in ATC cell lines in vitro and in vivo and in human ATC specimens. Activation of EGFR by EGF was blocked by the addition of gefitinib. In vitro studies showed that gefitinib greatly inhibited cellular proliferation and induced apoptosis in ATC cell lines and slowed tumor growth in a nude mouse model of thyroid carcinoma cells injected subcutaneously. CONCLUSIONS: ATC cells consistently overexpress EGFR, rendering this receptor a potential target for molecular therapy. Gefitinib effectively blocks activation of EGFR by EGF, inhibits ATC cellular proliferation, and induces apoptosis in vitro. Our in vivo results show that gefitinib has significant antitumor activity against ATC in a subcutaneous nude mouse tumor model and therefore is a potential candidate for human clinical trials.


Assuntos
Antineoplásicos/uso terapêutico , Apoptose/efeitos dos fármacos , Carcinoma/tratamento farmacológico , Receptores ErbB/antagonistas & inibidores , Quinazolinas/uso terapêutico , Neoplasias da Glândula Tireoide/tratamento farmacológico , Animais , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Carcinoma/metabolismo , Carcinoma/prevenção & controle , Carcinoma Papilar/tratamento farmacológico , Carcinoma Papilar/metabolismo , Carcinoma Papilar/prevenção & controle , Proliferação de Células/efeitos dos fármacos , Fator de Crescimento Epidérmico/metabolismo , Fator de Crescimento Epidérmico/farmacologia , Receptores ErbB/metabolismo , Gefitinibe , Humanos , Camundongos , Camundongos Nus , Paclitaxel/administração & dosagem , Fosforilação/efeitos dos fármacos , Neoplasias da Glândula Tireoide/metabolismo , Neoplasias da Glândula Tireoide/prevenção & controle , Fator de Crescimento Transformador alfa/metabolismo , Fator de Crescimento Transformador alfa/farmacologia , Células Tumorais Cultivadas
11.
Cancer Res ; 64(21): 7977-84, 2004 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-15520205

RESUMO

Expression of the epidermal growth factor (EGF) and activation of its receptor (EGFR), a tyrosine kinase, are associated with progressive growth of head and neck cancer. Expression of the vascular endothelial growth factor (VEGF) is associated with angiogenesis and progressive growth of tumor. The tyrosine kinase inhibitor NVP-AEE788 (AEE788) blocks the EGF and VEGF signaling pathways. We examined the effects of AEE788 administered alone, or with paclitaxel (Taxol), on the progression of human head and neck cancer implanted orthotopically into nude mice. Cells of two different human oral cancer lines, JMAR and MDA1986, were injected into the tongues of nude mice. Mice with established tumors were randomized to receive three times per week oral AEE788, once weekly injected paclitaxel, AEE788 plus paclitaxel, or placebo. Oral tumors were resected at necropsy. Kinase activity, cell proliferation, apoptosis, and mean vessel density were determined by immunohistochemical immunofluorescent staining. AEE788 inhibited cell growth, induced apoptosis, and reduced the phosphorylation of EGFR, VEGFR-2, AKT, and mitogen-activated protein kinase in both cell lines. Mice treated with AEE788 and AEE788 plus paclitaxel had decreased microvessel density, decreased proliferative index, and increased apoptosis. Hence, AEE788 inhibited tumor vascularization and growth and prolonged survival. Inhibition of EGFR and VEGFR phosphorylation by AEE788 effectively inhibits cellular proliferation of squamous cell carcinoma of the head and neck, induces apoptosis of tumor endothelial cells and tumor cells, and is well tolerated in mice. These data recommend the consideration of patients with head and neck cancer for inclusion in clinical trials of AEE788.


Assuntos
Carcinoma de Células Escamosas/tratamento farmacológico , Receptores ErbB/antagonistas & inibidores , Neoplasias Bucais/tratamento farmacológico , Purinas/uso terapêutico , Receptores de Fatores de Crescimento do Endotélio Vascular/antagonistas & inibidores , Inibidores da Angiogênese/uso terapêutico , Animais , Linhagem Celular Tumoral , Neoplasias de Cabeça e Pescoço/tratamento farmacológico , Humanos , Imuno-Histoquímica , Camundongos , Camundongos Nus , Paclitaxel/farmacologia , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia
12.
Cancer Gene Ther ; 11(11): 733-9, 2004 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-15486560

RESUMO

Lung cancer is one of the leading causes of death in the world. The underlying cause for lung cancer has been attributed to various factors that include alteration and mutation in the tumor suppressor genes. Restoration of normal function of the tumor suppressor gene is a potential therapeutic strategy. Recent studies have identified a group of candidate tumor suppressor genes on human chromosome 3p21.3 that are frequently deleted in human lung and breast cancers. Among the various genes identified in the 3p21.3 region, we tested the antitumor activity of the FUS1 gene in two human non-small-cell lung cancer (NSCLC) xenografts in vivo. Intratumoral administration of FUS1 gene complexed to DOTAP:cholesterol (DOTAP:Chol) liposome into subcutaneous H1299 and A549 lung tumor xenograft resulted in significant (P = .02) inhibition of tumor growth. Furthermore, intravenous injections of DOTAP:Chol-FUS1 complex into mice bearing experimental A549 lung metastasis demonstrated significant (P = .001) decrease in the number of metastatic tumor nodules. Finally, lung tumor-bearing animals when treated with DOTAP:Chol-FUS1 complex demonstrate prolonged survival (median survival time: 80 days, P = .01) compared to control animals. This result demonstrates the potent tumor suppressive activity of the FUS1 gene and is a promising therapeutic agent for treatment of primary and disseminated human lung cancer.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/genética , Cromossomos Humanos Par 3 , Genes Supressores de Tumor , Terapia Genética , Vetores Genéticos , Neoplasias Pulmonares/terapia , Proteínas Supressoras de Tumor/genética , Carcinoma Pulmonar de Células não Pequenas/patologia , Humanos , Lipossomos , Neoplasias Pulmonares/patologia , Células Tumorais Cultivadas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...