Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Clin Cancer Res ; 23(16): 4716-4723, 2017 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-28420725

RESUMO

Purpose: Noninvasive drug biomarkers for the early assessment of tumor response can enable adaptive therapeutic decision-making and proof-of-concept studies for investigational drugs. Circulating tumor DNA (ctDNA) is released into the circulation by tumor cell turnover and has been shown to be detectable in urine.Experimental Design: We tested the hypothesis that dynamic changes in EGFR activating (exon 19del and L858R) and resistance (T790M) mutation levels detected in urine could inform tumor response within days of therapy for advanced non-small cell lung cancer (NSCLC) patients receiving osimertinib, a second-line third-generation anti-EGFR tyrosine kinase inhibitor.Results: Eight of nine evaluable NSCLC patients had detectable T790M-mutant DNA fragments in pretreatment baseline samples. Daily monitoring of mutations in urine indicated a pattern of intermittent spikes throughout week 1, suggesting apoptosis with an overall decrease in fragment numbers from baselines to day 7 preceding radiographic response assessed at 6 to 12 weeks.Conclusions: These findings suggest drug-induced tumor apoptosis within days of initial dosing. Daily sampling of ctDNA may enable early assessment of patient response and proof-of-concept studies for drug development. The modeling of tumor lysis through the day-to-day kinetics of ctDNA released into the blood and then into the urine is demonstrated in this proof-of-concept study in lung cancer patients receiving anti-EGFR tyrosine kinase inhibitors. This strategy may determine the specific clonal populations of cells which undergo apoptosis within the first week of therapy. This has important implications for developing combinational strategies to address inter- and intralesional heterogeneity and characterizing residual disease after initial drug exposure. Clin Cancer Res; 23(16); 4716-23. ©2017 AACR.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , DNA Tumoral Circulante/urina , DNA de Neoplasias/urina , Neoplasias Pulmonares/tratamento farmacológico , Piperazinas/uso terapêutico , Acrilamidas , Idoso , Compostos de Anilina , Apoptose/efeitos dos fármacos , Apoptose/genética , Carcinoma Pulmonar de Células não Pequenas/genética , Carcinoma Pulmonar de Células não Pequenas/urina , DNA Tumoral Circulante/genética , DNA de Neoplasias/genética , Monitoramento de Medicamentos , Resistencia a Medicamentos Antineoplásicos/genética , Receptores ErbB/antagonistas & inibidores , Receptores ErbB/genética , Éxons/genética , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/urina , Pessoa de Meia-Idade , Terapia de Alvo Molecular , Mutação , Inibidores de Proteínas Quinases/uso terapêutico , Fatores de Tempo , Resultado do Tratamento
2.
Clin Cancer Res ; 23(14): 3657-3666, 2017 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-28096270

RESUMO

Purpose: Tumor-derived cell-free DNA (cfDNA) from urine of patients with cancer offers noninvasive biological material for detection of cancer-related molecular abnormalities such as mutations in Exon 2 of KRASExperimental Design: A quantitative, mutation-enrichment next-generation sequencing test for detecting KRASG12/G13 mutations in urine cfDNA was developed, and results were compared with clinical testing of archival tumor tissue and plasma cfDNA from patients with advanced cancer.Results: With 90 to 110 mL of urine, the KRASG12/G13 cfDNA test had an analytical sensitivity of 0.002% to 0.006% mutant copies in wild-type background. In 71 patients, the concordance between urine cfDNA and tumor was 73% (sensitivity, 63%; specificity, 96%) for all patients and 89% (sensitivity, 80%; specificity, 100%) for patients with urine samples of 90 to 110 mL. Patients had significantly fewer KRASG12/G13 copies in urine cfDNA during systemic therapy than at baseline or disease progression (P = 0.002). Compared with no changes or increases in urine cfDNA KRASG12/G13 copies during therapy, decreases in these measures were associated with longer median time to treatment failure (P = 0.03).Conclusions: A quantitative, mutation-enrichment next-generation sequencing test for detecting KRASG12/G13 mutations in urine cfDNA had good concordance with testing of archival tumor tissue. Changes in mutated urine cfDNA were associated with time to treatment failure. Clin Cancer Res; 23(14); 3657-66. ©2017 AACR.


Assuntos
Biomarcadores Tumorais/urina , Ácidos Nucleicos Livres/urina , Neoplasias/genética , Proteínas Proto-Oncogênicas p21(ras)/urina , Adulto , Idoso , Idoso de 80 Anos ou mais , Biomarcadores Tumorais/genética , Ácidos Nucleicos Livres/genética , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Mutação , Neoplasias/patologia , Neoplasias/urina , Proteínas Proto-Oncogênicas p21(ras)/genética
3.
J Thorac Oncol ; 11(10): 1690-700, 2016 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-27468937

RESUMO

INTRODUCTION: In approximately 60% of patients with NSCLC who are receiving EGFR tyrosine kinase inhibitors, resistance develops through the acquisition of EGFR T790M mutation. We aimed to demonstrate that a highly sensitive and quantitative next-generation sequencing analysis of EGFR mutations from urine and plasma specimens is feasible. METHODS: Short footprint mutation enrichment next-generation sequencing assays were used to interrogate EGFR activating mutations and the T790M resistance mutation in urine or plasma specimens from patients enrolled in TIGER-X (NCT01526928), a phase 1/2 clinical study of rociletinib in previously treated patients with EGFR mutant-positive advanced NSCLC. RESULTS: Of 63 patients, 60 had evaluable tissue specimens. When the tissue result was used as a reference, the sensitivity of EGFR mutation detection in urine was 72% (34 of 47 specimens) for T790M, 75% (12 of 16) for L858R, and 67% (28 of 42) for exon 19 deletions. With specimens that met a recommended volume of 90 to 100 mL, the sensitivity was 93% (13 of 14 specimens) for T790M, 80% (four of five) for L858R, and 83% (10 of 12) for exon 19 deletions. A comparable sensitivity of EGFR mutation detection was observed in plasma: 93% (38 of 41 specimens) for T790M, 100% (17 of 17) for L858R, and 87% (34 of 39) for exon 19 deletions. Together, urine and plasma testing identified 12 additional T790M-positive cases that were either undetectable or inadequate by tissue test. In nine patients monitored while receiving treatment with rociletinib, a rapid decrease in urine T790M levels was observed by day 21. CONCLUSIONS: DNA derived from NSCLC tumors can be detected with high sensitivity in urine and plasma, enabling diagnostic detection and monitoring of therapeutic response from these noninvasive "liquid biopsy" samples.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/genética , Receptores ErbB/sangue , Receptores ErbB/urina , Neoplasias Pulmonares/genética , Adulto , Idoso , Idoso de 80 Anos ou mais , Carcinoma Pulmonar de Células não Pequenas/patologia , Método Duplo-Cego , Feminino , Humanos , Neoplasias Pulmonares/patologia , Masculino , Pessoa de Meia-Idade , Mutação , Estudos Retrospectivos
4.
Biomicrofluidics ; 6(2): 24133, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23805171

RESUMO

Isolation and enumeration of circulating tumor cells (CTCs) are used to monitor metastatic disease progression and guide cancer therapy. However, currently available technologies are limited to cells expressing specific cell surface markers, such as epithelial cell adhesion molecule (EpCAM) or have limited specificity because they are based on cell size alone. We developed a device, ApoStream(™) that overcomes these limitations by exploiting differences in the biophysical characteristics between cancer cells and normal, healthy blood cells to capture CTCs using dielectrophoretic technology in a microfluidic flow chamber. Further, the system overcomes throughput limitations by operating in continuous mode for efficient isolation and enrichment of CTCs from blood. The performance of the device was optimized using a design of experiment approach for key operating parameters such as frequency, voltage and flow rates, and buffer formulations. Cell spiking studies were conducted using SKOV3 or MDA-MB-231 cell lines that have a high and low expression level of EpCAM, respectively, to demonstrate linearity and precision of recovery independent of EpCAM receptor levels. The average recovery of SKOV3 and MDA-MB-231 cancer cells spiked into approximately 12 × 10(6) peripheral blood mononuclear cells obtained from 7.5 ml normal human donor blood was 75.4% ± 3.1% (n = 12) and 71.2% ± 1.6% (n = 6), respectively. The intra-day and inter-day precision coefficients of variation of the device were both less than 3%. Linear regression analysis yielded a correlation coefficient (R(2)) of more than 0.99 for a spiking range of 4-2600 cells. The viability of MDA-MB-231 cancer cells captured with ApoStream was greater than 97.1% and there was no difference in cell growth up to 7 days in culture compared to controls. The ApoStream device demonstrated high precision and linearity of recovery of viable cancer cells independent of their EpCAM expression level. Isolation and enrichment of viable cancer cells from ApoStream enables molecular characterization of CTCs from a wide range of cancer types.

5.
Cancer Res ; 71(10): 3494-504, 2011 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-21467165

RESUMO

The acquisition of the metastatic melanoma phenotype is associated with increased expression of the melanoma cell adhesion molecule MCAM/MUC18 (CD146). However, the mechanism by which MUC18 contributes to melanoma metastasis remains unclear. Herein, we stably silenced MUC18 expression in two metastatic melanoma cell lines, A375SM and C8161, and conducted cDNA microarray analysis. We identified and validated that the transcriptional regulator, inhibitor of DNA binding-1 (Id-1), previously shown to function as an oncogene in several malignancies, including melanoma, was downregulated by 5.6-fold following MUC18 silencing. Additionally, we found that MUC18 regulated Id-1 expression at the transcriptional level via ATF-3, which itself was upregulated by 6.9-fold in our cDNA microarray analysis. ChIP analysis showed increased binding of ATF-3 to the Id-1 promoter after MUC18 silencing. To complement these studies, we rescued the expression of MUC18, which reversed the expression patterns of Id-1 and ATF-3. Moreover, we showed that MUC18 promotes melanoma invasion through Id-1, as overexpression of Id-1 in MUC18-silenced cells resulted in increased MMP-2 expression and activity. To our knowledge, this is the first demonstration that MUC18 is involved in cell signaling regulating the expression of Id-1 and ATF-3, thus contributing to melanoma metastasis.


Assuntos
Fator 3 Ativador da Transcrição/metabolismo , Antígeno CD146/metabolismo , Regulação Neoplásica da Expressão Gênica , Proteína 1 Inibidora de Diferenciação/fisiologia , Melanoma/metabolismo , Animais , Linhagem Celular Tumoral , Imunoprecipitação da Cromatina , Progressão da Doença , Feminino , Humanos , Proteína 1 Inibidora de Diferenciação/genética , Melanoma/patologia , Camundongos , Camundongos Endogâmicos BALB C , Metástase Neoplásica , Transplante de Neoplasias
6.
Proc Natl Acad Sci U S A ; 108(2): 626-31, 2011 Jan 11.
Artigo em Inglês | MEDLINE | ID: mdl-21187389

RESUMO

The thrombin receptor protease activated receptor-1 (PAR-1) is overexpressed in metastatic melanoma cell lines and tumor specimens. Previously, we demonstrated a significant reduction in tumor growth and experimental lung metastasis after PAR-1 silencing via systemic delivery of siRNA encapsulated into nanoliposomes. Gene expression profiling identified a 40-fold increase in expression of Maspin in PAR-1-silenced metastatic melanoma cell lines. Maspin promoter activity was significantly increased after PAR-1 silencing, suggesting that PAR1 negatively regulates Maspin at the transcriptional level. ChIP analyses revealed that PAR-1 decreases binding of Ets-1 and c-Jun transcription factors to the Maspin promoter, both known to activate Maspin transcription. PAR-1 silencing did not affect Ets-1 or c-Jun expression; rather it resulted in increased expression of the chromatin remodeling complex CBP/p300, as well as decreased activity of the CBP/p300 inhibitor p38, resulting in increased binding of Ets-1 and c-Jun to the Maspin promoter and higher Maspin expression. Functionally, Maspin expression reduced the invasive capability of melanoma cells after PAR-1 silencing, which was abrogated after rescuing with PAR-1. Furthermore, tumor growth and experimental lung metastasis was significantly decreased after expressing Maspin in a metastatic melanoma cell line. Moreover, silencing Maspin in PAR-1-silenced cells reverted the inhibition of tumor growth and experimental lung metastasis. Herein, we demonstrate a mechanism by which PAR-1 negatively regulates the expression of the Maspin tumor-suppressor gene in the acquisition of the metastatic melanoma phenotype, thus attributing an alternative function to PAR-1 other than coagulation.


Assuntos
Regulação Neoplásica da Expressão Gênica , Genes Supressores de Tumor , Melanoma/patologia , Receptor PAR-1/metabolismo , Serpinas/metabolismo , Animais , Cromatina/química , Progressão da Doença , Humanos , Neoplasias Pulmonares/patologia , Camundongos , Camundongos Nus , Metástase Neoplásica , Fenótipo , Proteína Proto-Oncogênica c-ets-1/metabolismo , Proteínas Proto-Oncogênicas c-jun/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Transdução de Sinais
7.
PLoS One ; 5(8): e12452, 2010 Aug 27.
Artigo em Inglês | MEDLINE | ID: mdl-20805990

RESUMO

BACKGROUND: The loss of AP-2alpha and increased activity of cAMP-responsive element binding (CREB) protein are two hallmarks of malignant progression of cutaneous melanoma. However, the molecular mechanism responsible for the loss of AP-2alpha during melanoma progression remains unknown. METHODOLOGY/PRINCIPAL FINDINGS: Herein, we demonstrate that both inhibition of PKA-dependent CREB phosphorylation, as well as silencing of CREB expression by shRNA, restored AP-2alpha protein expression in two metastatic melanoma cell lines. Moreover, rescue of CREB expression in CREB-silenced cell lines downregulates expression of AP-2alpha. Loss of AP-2alpha expression in metastatic melanoma occurs via a dual mechanism involving binding of CREB to the AP-2alpha promoter and CREB-induced overexpression of another oncogenic transcription factor, E2F-1. Upregulation of AP-2alpha expression following CREB silencing increases endogenous p21(Waf1) and decreases MCAM/MUC18, both known to be downstream target genes of AP-2alpha involved in melanoma progression. CONCLUSIONS/SIGNIFICANCE: Since AP-2alpha regulates several genes associated with the metastatic potential of melanoma including c-KIT, VEGF, PAR-1, MCAM/MUC18, and p21(Waf1), our data identified CREB as a major regulator of the malignant melanoma phenotype.


Assuntos
Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Regulação Neoplásica da Expressão Gênica , Melanoma/genética , Fator de Transcrição AP-2/genética , Fator de Transcrição AP-2/metabolismo , Animais , Antígeno CD146/metabolismo , Linhagem Celular Tumoral , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Progressão da Doença , Fator de Transcrição E2F1/metabolismo , Humanos , Melanoma/metabolismo , Melanoma/patologia , Metástase Neoplásica , Fosforilação , Regiões Promotoras Genéticas/genética , Transcrição Gênica , Regulação para Cima
8.
Cancer Res ; 69(16): 6730-7, 2009 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-19679555

RESUMO

Protease-activated receptor-1 (PAR-1) is a key player in melanoma metastasis with higher expression seen in metastatic melanoma cell lines and tissue specimens. cDNA microarray and Western blot analyses reveal that the gap junctional intracellular communication molecule connexin 43 (Cx-43), known to be involved in tumor cell diapedesis and attachment to endothelial cells, is significantly decreased after PAR-1 silencing in metastatic melanoma cell lines. Furthermore, Cx-43 promoter activity was significantly inhibited in PAR-1-silenced cells, suggesting that PAR-1 regulates Cx-43 at the transcriptional level. Chromatin immunoprecipitation studies showed a reduction in the binding of SP-1 and AP-1 transcription factors to the promoter of Cx-43. Both transcription factors have been shown previously to be required for maximal Cx-43 promoter activity. These results were corroborated by mutating the AP-1 and SP-1 binding sites resulting in decreased Cx-43 promoter activity in PAR-1-positive cells. Moreover, as Cx-43 has been shown to facilitate arrest of circulating tumor cells at the vascular endothelium, melanoma cell attachment to endothelial cells was significantly decreased in PAR-1-silenced cells, with this effect being abrogated after PAR-1 rescue. Herein, we report that up-regulation of PAR-1 expression, seen in melanoma progression, mediates high levels of Cx-43 expression. As both SP-1 and AP-1 transcription factors act as positive regulators of Cx-43, our data provide a novel mechanism for the regulation of Cx-43 expression by PAR-1. Indeed, Cx-43 expression was restored following PAR-1 rescue in PAR-1-silenced cells. Taken together, our data support the tumor promoting function of Cx-43 in melanoma.


Assuntos
Conexina 43/genética , Regulação Neoplásica da Expressão Gênica , Melanoma/genética , Melanoma/patologia , Receptor PAR-1/fisiologia , Adesão Celular/efeitos dos fármacos , Células Cultivadas , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Células Endoteliais/fisiologia , Regulação Neoplásica da Expressão Gênica/fisiologia , Humanos , Melanoma/metabolismo , Metástase Neoplásica , Regiões Promotoras Genéticas , Ligação Proteica , Interferência de RNA/fisiologia , RNA Interferente Pequeno/farmacologia , Receptor PAR-1/antagonistas & inibidores , Receptor PAR-1/genética , Receptor PAR-1/metabolismo , Ativação Transcricional , Transfecção
9.
J Biol Chem ; 284(42): 28845-55, 2009 Oct 16.
Artigo em Inglês | MEDLINE | ID: mdl-19703903

RESUMO

The cellular and molecular pathways that regulate platelet activation, blood coagulation, and inflammation are emerging as critical players in cancer progression and metastasis. Here, we demonstrate a novel signaling mechanism whereby protease-activated receptor 1 (PAR1) mediates expression of melanoma cell adhesion molecule MCAM/MUC18 (MUC18), a critical marker of melanoma metastasis, via activation of platelet-activating factor receptor (PAFR) and cAMP-responsive element-binding protein (CREB). We found that PAR1 silencing with small hairpin RNA inhibits MUC18 expression in metastatic melanoma cells by inhibiting CREB phosphorylation, activity, and binding to the MUC18 promoter. We further demonstrate that the PAF/PAFR pathway mediates MUC18 expression downstream of PAR1. Indeed, PAR1 silencing down-regulates PAFR expression and PAF production, PAFR silencing blocks MUC18 expression, and re-expression of PAFR in PAR1-silenced cells rescues MUC18 expression. We further demonstrate that the PAR1-PAFR-MUC18 pathway mediates melanoma cell adhesion to microvascular endothelial cells, transendothelial migration, and metastatic retention in the lungs. Rescuing PAFR expression in PAR1-silenced cells fully restores metastatic phenotype of melanoma, indicating that PAFR plays critical role in the molecular mechanism of PAR1 action. Our results link the two pro-inflammatory G-protein-coupled receptors, PAR1 and PAFR, with the metastatic dissemination of melanoma and suggest that PAR1, PAFR, and MUC18 are attractive therapeutic targets for preventing melanoma metastasis.


Assuntos
Melanoma/metabolismo , Glicoproteínas da Membrana de Plaquetas/metabolismo , Receptor PAR-1/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Neoplasias Cutâneas/metabolismo , Antígeno CD146/metabolismo , Proteína de Ligação a CREB/metabolismo , Linhagem Celular Tumoral , Inativação Gênica , Humanos , Melanoma/patologia , Metástase Neoplásica , Regiões Promotoras Genéticas , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Neoplasias Cutâneas/patologia , Fatores de Transcrição/metabolismo
10.
J Biol Chem ; 284(38): 26194-206, 2009 Sep 18.
Artigo em Inglês | MEDLINE | ID: mdl-19632997

RESUMO

Metastatic progression of melanoma is associated with overexpression and activity of cAMP-response element-binding protein (CREB). However, the mechanism by which CREB contributes to tumor progression and metastasis remains unclear. Here, we demonstrate that stably silencing CREB expression in two human metastatic melanoma cell lines, A375SM and C8161-c9, suppresses tumor growth and experimental metastasis. Analysis of cDNA microarrays revealed that CREB silencing leads to increased expression of cysteine-rich protein 61 (CCN1/CYR61) known to mediate adhesion, chemostasis, survival, and angiogenesis. Promoter analysis and chromatin immunoprecipitation assays demonstrated that CREB acts as a negative regulator of CCN1/CYR61 transcription by directly binding to its promoter. Re-expression of CREB in CREB-silenced cells rescued the low CCN1/CYR61 expression phenotype. CCN1/CYR61 overexpression resulted in reduced tumor growth and metastasis and inhibited the activity of matrix metalloproteinase-2. Furthermore, its overexpression decreased melanoma cell motility and invasion through Matrigel, which was abrogated by silencing CCN1/CYR61 in low metastatic melanoma cells. Moreover, a significant decrease in angiogenesis as well as an increase in apoptosis was seen in tumors overexpressing CCN1/CYR61. Our results demonstrate that CREB promotes melanoma growth and metastasis by down-regulating CCN1/CYR61 expression, which acts as a suppressor of melanoma cell motility, invasion and angiogenesis.


Assuntos
Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Proteína Rica em Cisteína 61/metabolismo , Regulação Neoplásica da Expressão Gênica , Inativação Gênica , Genes Supressores de Tumor , Melanoma/metabolismo , Animais , Linhagem Celular Tumoral , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/genética , Proteína Rica em Cisteína 61/genética , Regulação Enzimológica da Expressão Gênica/genética , Humanos , Metaloproteinase 2 da Matriz/biossíntese , Metaloproteinase 2 da Matriz/genética , Melanoma/genética , Melanoma/patologia , Camundongos , Camundongos Nus , Metástase Neoplásica , Transplante de Neoplasias , Neovascularização Patológica/genética , Neovascularização Patológica/metabolismo , Transplante Heterólogo
11.
Pigment Cell Melanoma Res ; 22(3): 257-67, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19368690

RESUMO

Metastatic melanoma is extremely refractory to existing chemotherapeutic drugs and bioimmune adjuvant therapies, and the life span of patients with metastatic melanoma is often measured in months. Understanding the mechanisms responsible for the development of tumor metastasis is critical for finding successful curative measures. An expending amount of data reveal the importance of inflammatory microenvironment and stroma in cancer initiation and progression, which brings new directions and approaches to cancer treatment. This review will summarize current data on the role of the tumor microenvironment in shaping the metastatic phenotype of melanoma.


Assuntos
Inflamação/patologia , Melanoma/patologia , Invasividade Neoplásica/patologia , Metástase Neoplásica/patologia , Quimiocinas/imunologia , Progressão da Doença , Humanos , Sistema Imunitário/fisiologia , Interleucinas/imunologia , Queratinócitos/metabolismo , Melanoma/tratamento farmacológico , Fator de Ativação de Plaquetas/metabolismo , Ativação Plaquetária , Receptor PAR-1/metabolismo , Células Estromais/metabolismo
12.
Cancer Res ; 68(21): 9078-86, 2008 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-18974154

RESUMO

The thrombin receptor [protease-activated receptor-1 (PAR-1)] is overexpressed in highly metastatic melanoma cell lines and in patients with metastatic lesions. Activation of PAR-1 leads to cell signaling and up-regulation of genes involved in adhesion, invasion, and angiogenesis. Herein, we stably silence PAR-1 through the use of lentiviral short hairpin RNA and found significant decreases in both tumor growth (P < 0.01) and metastasis (P < 0.001) of highly metastatic melanoma cell lines in vivo. The use of viruses for therapy is not ideal as it can induce toxic immune responses and possible gene alterations following viral integration. Therefore, we also used systemic delivery of PAR-1 small interfering RNA (siRNA) incorporated into neutral liposomes [1,2-dioleoyl-sn-glycero-3-phosphatidylcholine (DOPC)] to decrease melanoma growth and metastasis in vivo. Significant decreases in tumor growth, weight, and metastatic lung colonies (P < 0.001 for all) were found in mice treated with PAR-1 siRNA-DOPC. The in vivo effects of PAR-1 on invasion and angiogenesis were analyzed via immunohistochemistry. Concomitant decreases in vascular endothelial growth factor, interleukin-8, and matrix metalloproteinase-2 expression levels, as well as decreased blood vessel density (CD31), were found in tumor samples from PAR-1 siRNA-treated mice, suggesting that PAR-1 is a regulator of melanoma cell growth and metastasis by affecting angiogenic and invasive factors. We propose that siRNA incorporated into DOPC nanoparticles could be delivered systemically and used as a new modality for melanoma treatment.


Assuntos
Divisão Celular , Melanoma/patologia , Metástase Neoplásica , RNA Interferente Pequeno/administração & dosagem , Receptor PAR-1/genética , Animais , Sequência de Bases , Linhagem Celular Tumoral , Feminino , Humanos , Imuno-Histoquímica , Lipossomos , Camundongos , Camundongos Nus , Reação em Cadeia da Polimerase Via Transcriptase Reversa
13.
Am J Pathol ; 173(6): 1839-52, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18988806

RESUMO

Galectin-3 (Gal-3) is a beta-galactoside-binding protein that is involved in cancer progression and metastasis. Using a progressive human melanoma tissue microarray, we previously demonstrated that melanocytes accumulate Gal-3 during the progression from benign to dysplastic nevi to melanoma and further to metastatic melanoma. Herein, we show that silencing of Gal-3 expression with small hairpin RNA results in a loss of tumorigenic and metastatic potential of melanoma cells. In vitro, Gal-3 silencing resulted in loss of tumor cell invasiveness and capacity to form tube-like structures on collagen ("vasculogenic mimicry"). cDNA microarray analysis after Gal-3 silencing revealed that Gal-3 regulates the expression of multiple genes, including endothelial cell markers that appear to be aberrantly expressed in highly aggressive melanoma cells, causing melanoma cell plasticity. These genes included vascular endothelial-cadherin, which plays a pivotal role in vasculogenic mimicry, as well as interleukin-8, fibronectin-1, endothelial differentiation sphingolipid G-protein receptor-1, and matrix metalloproteinase-2. Chromatin immunoprecipitation assays and promoter analyses revealed that Gal-3 silencing resulted in a decrease of vascular endothelial-cadherin and interleukin-8 promoter activities due to enhanced recruitment of transcription factor early growth response-1. Moreover, transient overexpression of early growth response-1 in C8161-c9 cells resulted in a loss of vascular endothelial-cadherin and interleukin-8 promoter activities and protein expression. Thus, Gal-3 plays an essential role during the acquisition of vasculogenic mimicry and angiogenic properties associated with melanoma progression.


Assuntos
Galectina 3/metabolismo , Regulação Neoplásica da Expressão Gênica , Melanoma/metabolismo , Neovascularização Patológica , Animais , Antígenos CD/metabolismo , Apoptose , Caderinas/metabolismo , Linhagem Celular , Proliferação de Células , Feminino , Galectina 3/genética , Perfilação da Expressão Gênica , Inativação Gênica , Humanos , Interleucina-8/metabolismo , Melanoma/genética , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Microvasos/anatomia & histologia , Metástase Neoplásica , Transplante de Neoplasias , Análise de Sequência com Séries de Oligonucleotídeos
14.
Am J Clin Dermatol ; 9(5): 307-11, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18717605

RESUMO

The incidence of melanoma has been steadily increasing over the last 3 decades. Currently, there are several approved treatments for metastatic melanoma, including chemotherapy and biologic therapy as both single treatments and in combination, but none is associated with a significant increase in survival. The chemotherapeutic agent dacarbazine is the standard treatment for metastatic melanoma, with a response rate of 15-20%, although most responses are not sustained. One of the main problems with melanoma treatment is chemotherapeutic resistance. The mechanisms of resistance of melanoma cells to chemotherapy have yet to be elucidated. Following treatment with dacarbazine, melanoma cells activate the extracellular signal-regulated kinase pathway, which results in over-expression and secretion of interleukin (IL)-8 and vascular endothelial growth factor. Melanoma cells utilize this mechanism to escape from the cytotoxic effect of the drug. We have previously reported on the development of fully human neutralizing antibodies against IL-8 (anti-IL-8-monoclonal-antibody [ABX-IL8]). In preclinical studies, ABX-IL8 inhibited tumor growth, angiogenesis, and metastasis of human melanoma in vivo. We propose that combination treatment with dacarbazine and IL-8 will potentiate the cytotoxic effect of the drug. Furthermore, formation of metastasis is a multistep process that includes melanoma cell adhesion to endothelial cells. Melanoma cell adhesion molecule (MUC18) mediates these processes in melanoma and is therefore a good target for eliminating metastasis. We have developed a fully human antibody against MUC18 that has shown promising results in preclinical studies. Since resistance is one of the major obstacles in the treatment of melanoma, we propose that utilization of antibodies against IL-8 or MUC18 alone, or as part of a 'cocktail' in combination with dacarbazine, may be a new treatment modality for metastatic melanoma that overcomes resistance of the disease to chemotherapy and significantly improves survival of patients.


Assuntos
Imunoterapia/métodos , Interleucina-8/imunologia , Melanoma/terapia , Neoplasias Cutâneas/terapia , Anticorpos Monoclonais/uso terapêutico , Antineoplásicos/uso terapêutico , Antígeno CD146/imunologia , Dacarbazina/uso terapêutico , Progressão da Doença , Resistencia a Medicamentos Antineoplásicos , Humanos , Melanoma/imunologia , Melanoma/patologia , Metástase Neoplásica/prevenção & controle , Neoplasias Cutâneas/imunologia , Neoplasias Cutâneas/patologia
16.
Cancer Biol Ther ; 7(7): 997-1003, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18698165

RESUMO

Malignant progression and tumor metastasis is a complex process enabled by the intrinsic genomic instability of the tumor cells and supported by a plethora of recruited cell types within the tumor microenvironment. Propelled by a selective pressure to overcome tissue homeostatic mechanisms, a metastatic "super-cell" emerges whose phenotype is associated with the cellular capacity for uncontrolled growth, resistance to apoptosis, high invasive potential, and effective neoangiogenesis. While genetic alterations arise early in the course of melanoma development, the progression toward metastatic disease is accompanied by deregulated expression of a number of transcription factors. In melanoma, acquisition of the metastatic phenotype involves the loss of Activator Protein-2alpha (AP-2alpha) and gain in expression of cAMP-responsive element (CRE)-binding protein/Activating Transcription Factor-1 (CREB/ATF-1) family transcription factors. Together with the upregulation of ATF-2, SNAIL/SLUG, Nuclear Factor kappaB, STAT3 and 5, and other transcription factors, these changes result in the aberrant expression of cellular adhesion molecules, matrix-degrading enzymes as well as other factors that enable a complex interaction of metastatic cells with the extracellular milieu. Similar to the case of oncogene addiction, the tumorigenicity and metastatic potential of melanoma can be greatly inhibited by altering the activity of the above-named transcription factors, therefore indicating new treatment prospects.


Assuntos
Regulação Neoplásica da Expressão Gênica , Melanoma/metabolismo , Transcrição Gênica , Fator 2 Ativador da Transcrição/metabolismo , Animais , Apoptose , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Humanos , Camundongos , Modelos Biológicos , NF-kappa B/metabolismo , Metástase Neoplásica , Neovascularização Patológica , Fenótipo , Fator de Transcrição AP-2/metabolismo
17.
Adv Exp Med Biol ; 624: 265-82, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18348463

RESUMO

The p53 tumor suppressor gene and gene product are among the most diverse and complex been shown to have a direct correlation with cancer development and have been shown to occur in nearly 50% of all cancers. p53 mutations are particularly common in skin cancers and UV irradiation has been shown to be a primary cause of specific 'signature' mutations that can result in oncogenic transformation. There are certain 'hot-spots' in the p53 gene where mutations are commonly found that result in a mutated dipyrimidine site. This review discusses the role of p53 from normal function and its dysfunction in precancerous lesions, nonmelanoma and melanoma skin cancers. Additionally, molecules that associate with p53 and alter its function to produce neoplastic conditions are also explored in this chapter.


Assuntos
Carcinoma Basocelular/patologia , Carcinoma de Células Escamosas/patologia , Melanoma/patologia , Fragmentos de Peptídeos/genética , Neoplasias Cutâneas/patologia , Proteína Supressora de Tumor p53/genética , Animais , Carcinoma Basocelular/genética , Carcinoma de Células Escamosas/genética , Transformação Celular Neoplásica/genética , Humanos , Melanoma/genética , Camundongos , Neoplasias Cutâneas/genética , Raios Ultravioleta/efeitos adversos
18.
Cancer Microenviron ; 1(1): 103-11, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19308689

RESUMO

Melanoma growth, angiogenesis and metastatic progression are strongly promoted by the inflammatory tumor microenvironment due to high levels of cytokine and chemokine secretion by the recruited inflammatory and stromal cells. In addition, platelets and molecular components of procoagulant pathways have been recently emerging as critical players of tumor growth and metastasis. In particular, thrombin, through the activity of its receptor protease-activated receptor-1 (PAR-1), regulates tumor cell adhesion to platelets and endothelial cells, stimulates tumor angiogenesis, and promotes tumor growth and metastasis. Notably, in many tumor types including melanoma, PAR-1 expression directly correlates with their metastatic phenotype and is directly responsible for the expression of interleukin-8, matrix metalloproteinase-2 (MMP-2), vascular endothelial growth factor, platelet-derived growth factor, and integrins. Another proinflammatory receptor-ligand pair, platelet-activating factor (PAF) and its receptor (PAFR), have been shown to act as important modulators of tumor cell adhesion to endothelial cells, angiogenesis, tumor growth and metastasis. PAF is a bioactive lipid produced by a variety of cells from membrane glycerophospholipids in the same reaction that releases arachidonic acid, and can be secreted by platelets, inflammatory cells, keratinocytes and endothelial cells. We have demonstrated that in metastatic melanoma cells, PAF stimulates the phosphorylation of cyclic adenosine monophosphate response element-binding protein (CREB) and activating transcription factor 1 (ATF-1), which results in overexpression of MMP-2 and membrane type 1-MMP (membrane type 1-MMP). Since only metastatic melanoma cells overexpress CREB/ATF-1, we propose that metastatic melanoma cells are better equipped than their non-metastatic counterparts to respond to PAF within the tumor microenvironment. The evidence supporting the hypothesis that the two G-protein coupled receptors, PAR-1 and PAFR, contribute to the acquisition of the metastatic phenotype of melanoma is presented and discussed.

19.
Mol Carcinog ; 46(8): 671-8, 2007 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17570501

RESUMO

Human melanoma represents the fastest growing malignancy in the US. The etiology of melanoma is highly debated as is the role of ultraviolet (UV) radiation in the initiation and progression of melanoma. This article discusses data from UV exposure and its relationship to the development of melanoma from various models of melanoma as well as various genetic alterations seen in oncogenic transformation of melanocytes. Genetic alterations such as the p16(INK4a) deletion, melanocortin 1 receptor (MC1R), RAS, and v-raf murine sarcoma viral oncogene homolog B1 (BRAF) may be indicative of a predisposition to melanoma development. Historical research as well as current data on the significance of the hot spot mutation in BRAF is discussed in its relative potential to the activating mutation in RAS.


Assuntos
Modelos Animais de Doenças , Melanoma/genética , Neoplasias Cutâneas/genética , Animais , Humanos , Melanoma/patologia , Proteínas Proto-Oncogênicas B-raf/fisiologia , Neoplasias Cutâneas/patologia
20.
Cancer Res ; 67(11): 5258-66, 2007 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-17545605

RESUMO

Melanoma is a very aggressive and highly angiogenic tumor in which standard treatments have had only limited success. Patients with advanced disease have a 5-year survival rate of 5%. In search for alternatives, we identified a natural product extracted from the fungus Aspergillus niger, termed ACTIBIND, that inhibits tumor growth and metastasis of melanoma in vivo. ACTIBIND, a T2 RNase, exerts antitumorigenic and antiangiogenic activities by competing with the angiogenic factor angiogenin (itself an RNase homologue). Thus, there was decreased expression and activity of the matrix metalloproteinase 2 in melanoma and vascular endothelial cells, decreased vascularization, and increased tumor cell apoptosis in vivo. ACTIBIND significantly inhibited angiogenesis in an in vivo angiogenesis assay with sponges containing angiogenin. In vitro, ACTIBIND was internalized by both melanoma and human umbilical vein endothelial cells, reached the cell nuclei, and inhibited the activity of angiogenin response elements in a dose-dependent manner. Collectively, our data indicate that ACTIBIND should be tested for its potential as a new antiangiogenic modality for the treatment of melanoma.


Assuntos
Endorribonucleases/farmacologia , Melanoma/tratamento farmacológico , Ribonuclease Pancreático/farmacologia , Animais , Apoptose/efeitos dos fármacos , Aspergillus niger/enzimologia , Processos de Crescimento Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Interações Medicamentosas , Endorribonucleases/metabolismo , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Células Endoteliais/patologia , Humanos , Luciferases/metabolismo , Masculino , Metaloproteinase 2 da Matriz/biossíntese , Metaloproteinase 2 da Matriz/metabolismo , Inibidores de Metaloproteinases de Matriz , Melanoma/irrigação sanguínea , Melanoma/patologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Metástase Neoplásica , Neovascularização Patológica/tratamento farmacológico , Neovascularização Patológica/patologia , Ribonuclease Pancreático/antagonistas & inibidores , Ribonuclease Pancreático/metabolismo , Transcrição Gênica
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...