Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 84
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
J Oral Pathol Med ; 45(6): 399-408, 2016 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-26493274

RESUMO

BACKGROUND: p16 overexpression is a highly sensitive yet moderately specific biomarker for predicting human papillomavirus (HPV)-associated oropharyngeal squamous cell carcinoma (OPSCC). Nuclear ß-catenin translocation has been linked to HPV-positive OPSCC. However, whether the strategy of combining ß-catenin with p16 can better predict HPV-associated OPSCC remains unknown. METHODS: We evaluated the expression of p16 and ß-catenin (nuclear and membrane) by immunohistochemistry staining in 101 OPSCC tissues and HPV status by HPV DNA in situ hybridization. Logistic regression models were used to evaluate single or multiple biomarkers for HPV prediction. The prediction power, sensitivity, and specificity were determined by receiver operating characteristic (ROC) analyses. RESULTS: Our data showed that upon univariate analysis, p16 and nuclear ß-catenin were positively correlated with HPV status, while membrane ß-catenin was inversely correlated with HPV status (P < 0.01). p16 showed the highest HPV predictive power, with area under the curve (AUC) of 0.9074 compared to 0.6762 for nuclear ß-catenin and 0.7635 for membrane ß-catenin, respectively, indicating differential accuracies for HPV prediction. Multivariable analysis showed that p16 was significantly correlated with HPV, while nuclear and membrane ß-catenin showed marginal significance. The three-biomarker model was similarly sensitive (98.9% vs. 100%) but more specific (88.9% vs. 81%) than p16 alone, which also showed a good predictive value for overall (P = 0.0002) survival and disease-free (P = 0.0158) survival. CONCLUSION: Our study suggests a novel model of combining p16 and subcellular ß-catenin for prediction of HPV-associatred OPSCC, and this finding deserves further validation.


Assuntos
Biomarcadores Tumorais/biossíntese , Carcinoma de Células Escamosas/diagnóstico , Inibidor p16 de Quinase Dependente de Ciclina/biossíntese , Neoplasias de Cabeça e Pescoço/diagnóstico , Neoplasias Orofaríngeas/diagnóstico , Papillomaviridae/isolamento & purificação , Infecções por Papillomavirus/diagnóstico , beta Catenina/biossíntese , Adulto , Idoso , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/patologia , Carcinoma de Células Escamosas/virologia , DNA Viral/genética , Feminino , Neoplasias de Cabeça e Pescoço/metabolismo , Neoplasias de Cabeça e Pescoço/patologia , Neoplasias de Cabeça e Pescoço/virologia , Humanos , Estimativa de Kaplan-Meier , Masculino , Pessoa de Meia-Idade , Estadiamento de Neoplasias , Neoplasias Orofaríngeas/metabolismo , Neoplasias Orofaríngeas/patologia , Neoplasias Orofaríngeas/virologia , Papillomaviridae/genética , Infecções por Papillomavirus/metabolismo , Infecções por Papillomavirus/patologia , Infecções por Papillomavirus/virologia , Valor Preditivo dos Testes , Carcinoma de Células Escamosas de Cabeça e Pescoço
2.
Cancer ; 121(2): 214-25, 2015 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-25209444

RESUMO

BACKGROUND: To understand the mechanism of frequent and early lymph node metastasis in high-risk human papillomavirus (HPV)-associated oropharyngeal squamous cell carcinoma (OPSCC), this study investigated whether ß-catenin is regulated by the HPV oncoprotein and contributes to OPSCC metastasis. METHODS: Expression levels of p16, ß-catenin, and epidermal growth factor receptor (EGFR) were examined in OPSCC samples (n = 208) by immunohistochemistry. The expression and subcellular localization of ß-catenin and EGFR activation were also studied in HPV-positive and HPV-negative head and neck squamous cell carcinoma cell lines with western blot analysis. HPV16 E6 small interfering RNA was used to elucidate the effect of the HPV oncoprotein on ß-catenin translocation. The involvement of EGFR in ß-catenin translocation was confirmed by treatment with erlotinib. Moreover, the invasive capacity was evaluated after HPV16 E6/E7 repression. RESULTS: The results showed that the membrane weighted index of ß-catenin was inversely correlated with p16 positivity (P < .001) and lymph node metastasis (P = .026), whereas nuclear staining of ß-catenin was associated with p16-positive OPSCC (P < .001). A low level of membrane ß-catenin expression was significantly associated with disease-free and overall survival (P < .0001 in both cases). Furthermore, the membrane weighted index of EGFR was inversely correlated with p16 positivity (P < .001) and positively correlated with membrane ß-catenin (P < .001). The in vitro study showed that HPV16 E6 repression led to reductions of phospho-EGFR and nuclear ß-catenin, which were also observed after erlotinib treatment, and inhibition of invasion. CONCLUSIONS: The findings suggest that HPV16 E6 mediates the translocation of ß-catenin to the nucleus, which may be regulated by activated EGFR.


Assuntos
Carcinoma de Células Escamosas/metabolismo , Receptores ErbB/metabolismo , Proteínas Oncogênicas Virais/metabolismo , Neoplasias Orofaríngeas/metabolismo , Proteínas E7 de Papillomavirus/metabolismo , Proteínas Repressoras/metabolismo , Translocação Genética , beta Catenina/genética , Adulto , Idoso , Idoso de 80 Anos ou mais , Antineoplásicos/uso terapêutico , Biomarcadores Tumorais/metabolismo , Western Blotting , Carcinoma de Células Escamosas/mortalidade , Carcinoma de Células Escamosas/patologia , Linhagem Celular Tumoral , Receptores ErbB/antagonistas & inibidores , Receptores ErbB/genética , Cloridrato de Erlotinib , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Imuno-Histoquímica , Estimativa de Kaplan-Meier , Metástase Linfática/genética , Metástase Linfática/patologia , Masculino , Pessoa de Meia-Idade , Neoplasias Orofaríngeas/mortalidade , Neoplasias Orofaríngeas/patologia , Valor Preditivo dos Testes , Prognóstico , Quinazolinas/uso terapêutico , RNA Interferente Pequeno/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , beta Catenina/análise , beta Catenina/metabolismo
3.
Apoptosis ; 20(7): 986-95, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25860284

RESUMO

Combinatorial approaches using two or more compounds are gaining increasing attention for cancer therapy. We have previously reported that the combination of the EGFR-TKI erlotinib and epigallocatechin-3-gallate (EGCG) exhibited synergistic chemopreventive effects in head and neck cancers by inducing the expression of Bim, p21, p27, and by inhibiting the phosphorylation of ERK and AKT and expression of Bcl-2. In the current study, we further investigated the mechanism of regulation of Bim, Bcl-2, p21 and p27, and their role in apoptosis. shRNA-mediated silencing of Bim significantly inhibited apoptosis induced by the combination of erlotinib and EGCG (p = 0.005). On the other hand, overexpression of Bcl-2 markedly protected cells from apoptosis (p = 0.003), whereas overexpression of constitutively active AKT only minimally protected cells from apoptosis induced by the combination of the two compounds. Analysis of mRNA expression by RT-PCR revealed that erlotinib, EGCG and their combination had no significant effects on the mRNA expression of Bim, p21, p27 or Bcl-2 suggesting the post-transcriptional regulation of these molecules. Furthermore, we found that erlotinib or the combination of EGCG and erlotinib inhibited the phosphorylation of Bim and stabilized Bim after inhibition of protein translation by cycloheximide. Taken together, our results strongly suggest that the combination of erlotinib and EGCG induces apoptosis of SCCHN cells by regulating Bim and Bcl-2 at the posttranscriptional level.


Assuntos
Anticarcinógenos/farmacologia , Apoptose/efeitos dos fármacos , Catequina/análogos & derivados , Cloridrato de Erlotinib/farmacologia , Neoplasias de Cabeça e Pescoço/metabolismo , Interferência de RNA/efeitos dos fármacos , Proteínas Reguladoras de Apoptose/biossíntese , Proteínas Reguladoras de Apoptose/metabolismo , Proteína 11 Semelhante a Bcl-2 , Catequina/farmacologia , Linhagem Celular Tumoral , Cicloeximida/farmacologia , Sinergismo Farmacológico , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Humanos , Proteínas de Membrana/biossíntese , Proteínas de Membrana/metabolismo , Fosforilação/efeitos dos fármacos , Biossíntese de Proteínas/efeitos dos fármacos , Inibidores da Síntese de Proteínas/farmacologia , Proteínas Proto-Oncogênicas/biossíntese , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo
4.
Int J Cancer ; 131(4): 956-69, 2012 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-21918971

RESUMO

An antibody simultaneously targeting epidermal growth factor receptor (EGFR) and vascular endothelial growth factor (VEGF), two major tumor growth-driving machineries, may provide a novel effective strategy for optimizing tumor targeting and maximizing potential clinical benefits. Human domain antibodies selected against VEGF and EGFR were formatted into a fully human dual-targeting IgG (DT-IgG) to directly target both antigens in a single molecule. We evaluated the efficacy of DT-IgG in comparison with bevacizumab and cetuximab alone and in combination in the lung cancer cell line A549 (low EGFR expression and KRAS mutant) and the head and neck squamous cell carcinoma (HNSCC) cell line Tu212 (high EGFR expression and KRAS wild type) in vitro and in vivo. DT-IgG suppressed Tu212 and A549 cell growth, inhibited EGFR activation and induced apoptosis as effectively as cetuximab, and neutralized VEGF as effectively as bevacizumab. DT-IgG induced EGFR-dependent VEGF internalization, constituting a novel antiangiogenesis mechanism. In xenograft models with lung and head and neck cancer cell lines, DT-IgG displayed efficacy equivalent to bevacizumab in diminishing tumor growth despite its short serum half-life (36 hr in rats) and both agents may constitute preferable alternatives to cetuximab in KRAS-mutant tumors. Immunofluorescence staining revealed that localization of DT-IgG was similar to that of cetuximab, largely associated with EGFR+tumor cells. Our proof of principle study suggests a DT-IgG against EGFR and VEGF as an alternative therapeutic strategy with potentially enhanced clinical benefit.


Assuntos
Receptores ErbB/imunologia , Neoplasias de Cabeça e Pescoço/terapia , Imunoglobulina G/uso terapêutico , Neoplasias Pulmonares/terapia , Fator A de Crescimento do Endotélio Vascular/imunologia , Animais , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/uso terapêutico , Anticorpos Monoclonais Humanizados/imunologia , Anticorpos Monoclonais Humanizados/uso terapêutico , Apoptose , Bevacizumab , Linhagem Celular Tumoral , Proliferação de Células , Cetuximab , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Neoplasias de Cabeça e Pescoço/patologia , Humanos , Imunoglobulina G/imunologia , Neoplasias Pulmonares/patologia , Masculino , Ratos , Ratos Sprague-Dawley , Ensaios Antitumorais Modelo de Xenoenxerto
5.
J Exp Clin Cancer Res ; 41(1): 248, 2022 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-35965328

RESUMO

FAT atypical cadherin 1 (FAT1) is among the most frequently mutated genes in many types of cancer. Its highest mutation rate is found in head and neck squamous cell carcinoma (HNSCC), in which FAT1 is the second most frequently mutated gene. Thus, FAT1 has great potential to serve as a target or prognostic biomarker in cancer treatment. FAT1 encodes a member of the cadherin-like protein family. Under normal physiological conditions, FAT1 serves as a molecular "brake" on mitochondrial respiration and acts as a receptor for a signaling pathway regulating cell-cell contact interaction and planar cell polarity. In many cancers, loss of FAT1 function promotes epithelial-mesenchymal transition (EMT) and the formation of cancer initiation/stem-like cells. However, in some types of cancer, overexpression of FAT1 leads to EMT. The roles of FAT1 in cancer progression, which seems to be cancer-type specific, have not been clarified. To further study the function of FAT1 in cancers, this review summarizes recent relevant literature regarding this protein. In addition to phenotypic alterations due to FAT1 mutations, several signaling pathways and tumor immune systems known or proposed to be regulated by this protein are presented. The potential impact of detecting or targeting FAT1 mutations on cancer treatment is also prospectively discussed.


Assuntos
Caderinas , Neoplasias de Cabeça e Pescoço , Caderinas/genética , Caderinas/metabolismo , Transição Epitelial-Mesenquimal/genética , Humanos , Transdução de Sinais , Carcinoma de Células Escamosas de Cabeça e Pescoço
6.
J Biol Chem ; 285(45): 34557-65, 2010 Nov 05.
Artigo em Inglês | MEDLINE | ID: mdl-20826787

RESUMO

Natural dietary agents have drawn a great deal of attention toward cancer prevention because of their wide safety margin. However, single agent intervention has failed to bring the expected outcome in clinical trials; therefore, combinations of chemopreventive agents are gaining increasingly popularity. In the present study, we investigated a combinatorial approach using two natural dietary polyphenols, luteolin and EGCG, and found that their combination at low doses (at which single agents induce minimal apoptosis) synergistically increased apoptosis (3-5-fold more than the additive level of apoptosis) in both head and neck and lung cancer cell lines. This combination also significantly inhibited growth of xenografted tumors in nude mice. The in vivo findings also were supported by significant inhibition of Ki-67 expression and increase in TUNEL-positive cells in xenografted tissues. Mechanistic studies revealed that the combination induced mitochondria-dependent apoptosis in some cell lines and mitochondria-independent apoptosis in others. Moreover, we found more efficient stabilization and ATM-dependent Ser(15) phosphorylation of p53 due to DNA damage by the combination, and ablation of p53 using shRNA strongly inhibited apoptosis as evidenced by decreased poly(ADP-ribose) polymerase and caspase-3 cleavage. In addition, we observed mitochondrial translocation of p53 after treatment with luteolin or the combination of EGCG and luteolin. Taken together, our results for the first time suggest that the combination of luteolin and EGCG has synergistic/additive growth inhibitory effects and provides an important rationale for future chemoprevention trials of head and neck and lung cancers.


Assuntos
Anticarcinógenos/farmacologia , Catequina/análogos & derivados , Neoplasias de Cabeça e Pescoço/metabolismo , Neoplasias de Cabeça e Pescoço/prevenção & controle , Luteolina/farmacologia , Proteína Supressora de Tumor p53/metabolismo , Animais , Anticarcinógenos/agonistas , Apoptose/efeitos dos fármacos , Proteínas Mutadas de Ataxia Telangiectasia , Caspase 3/metabolismo , Catequina/agonistas , Catequina/farmacologia , Proteínas de Ciclo Celular/farmacologia , Linhagem Celular Tumoral , Dano ao DNA/efeitos dos fármacos , Proteínas de Ligação a DNA/farmacologia , Sinergismo Farmacológico , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Antígeno Ki-67/biossíntese , Luteolina/agonistas , Camundongos , Camundongos Nus , Mitocôndrias/metabolismo , Transplante de Neoplasias , Fosforilação/efeitos dos fármacos , Proteínas Serina-Treonina Quinases/farmacologia , Estabilidade Proteica/efeitos dos fármacos , Proteínas Supressoras de Tumor/farmacologia , Ensaios Antitumorais Modelo de Xenoenxerto
7.
Nat Rev Drug Discov ; 7(9): 771-82, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18758474

RESUMO

Nanoparticles--particles in the size range 1-100 nm--are emerging as a class of therapeutics for cancer. Early clinical results suggest that nanoparticle therapeutics can show enhanced efficacy, while simultaneously reducing side effects, owing to properties such as more targeted localization in tumours and active cellular uptake. Here, we highlight the features of nanoparticle therapeutics that distinguish them from previous anticancer therapies, and describe how these features provide the potential for therapeutic effects that are not achievable with other modalities. While large numbers of preclinical studies have been published, the emphasis here is placed on preclinical and clinical studies that are likely to affect clinical investigations and their implications for advancing the treatment of patients with cancer.


Assuntos
Antineoplásicos/administração & dosagem , Nanopartículas , Neoplasias/tratamento farmacológico , Animais , Ensaios Clínicos como Assunto , Sistemas de Liberação de Medicamentos , Avaliação Pré-Clínica de Medicamentos , Humanos
8.
Oncol Rep ; 45(5)2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33864659

RESUMO

Despite widespread interest in chemoprevention and therapy due to the high margin of safety of dietary natural compounds, clinical intervention with single agents has failed to yield the expected outcomes, mostly due to poor bioavailability and low potency. Combinations of natural agents with synergistic effects are gaining increasing attention. In the present study, in vitro and in vivo antitumor effects of a combination of two natural dietary agents, green tea epigallocatechin gallate (EGCG) and resveratrol were investigated. It was revealed that their combination at low doses (at which single agents induce minimal apoptosis) synergistically increased apoptosis (combination index < 1) in head and neck cancer cell lines. Synergistic apoptosis was also supported by caspase­3 and PARP cleavage. The combination also significantly inhibited growth of xenografted head and neck tumors in nude mice as supported by significant inhibition of tumor volume, tumor weight and Ki67 expression, and increase in TUNEL­positive cells. Mechanistic studies revealed that the combination inhibited AKT­mTOR signaling both in vitro and in vivo. In addition, overexpression of constitutively active AKT protected cells from apoptosis induced by the combination of EGCG and resveratrol. Collectively, the present results for the first time suggest that the combination of EGCG and resveratrol has synergistic growth inhibitory effects and provide an important rationale for future clinical development for chemoprevention and treatment of head and neck cancer.


Assuntos
Anticarcinógenos/farmacologia , Catequina/análogos & derivados , Neoplasias de Cabeça e Pescoço/prevenção & controle , Resveratrol/farmacologia , Animais , Anticarcinógenos/uso terapêutico , Apoptose/efeitos dos fármacos , Carcinogênese/efeitos dos fármacos , Catequina/farmacologia , Catequina/uso terapêutico , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sinergismo Farmacológico , Neoplasias de Cabeça e Pescoço/patologia , Humanos , Camundongos , Resveratrol/uso terapêutico , Transdução de Sinais/efeitos dos fármacos , Chá/química , Ensaios Antitumorais Modelo de Xenoenxerto
9.
PLoS One ; 15(9): e0238497, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32986729

RESUMO

Human papilloma virus (HPV) causes a subset of head and neck squamous cell carcinomas (HNSCC) of the oropharynx. We combined targeted DNA- and genome-wide RNA-sequencing to identify genetic variants and gene expression signatures respectively from patients with HNSCC including oropharyngeal squamous cell carcinomas (OPSCC). DNA and RNA were purified from 35- formalin fixed and paraffin embedded (FFPE) HNSCC tumor samples. Immuno-histochemical evaluation of tumors was performed to determine the expression levels of p16INK4A and classified tumor samples either p16+ or p16-. Using ClearSeq Comprehensive Cancer panel, we examined the distribution of somatic mutations. Somatic single-nucleotide variants (SNV) were called using GATK-Mutect2 ("tumor-only" mode) approach. Using RNA-seq, we identified a catalog of 1,044 and 8 genes as significantly expressed between p16+ and p16-, respectively at FDR 0.05 (5%) and 0.1 (10%). The clinicopathological characteristics of the patients including anatomical site, smoking and survival were analyzed when comparing p16+ and p16- tumors. The majority of tumors (65%) were p16+. Population sequence variant databases, including gnomAD, ExAC, COSMIC and dbSNP, were used to identify the mutational landscape of somatic sequence variants within sequenced genes. Hierarchical clustering of The Cancer Genome Atlas (TCGA) samples based on HPV-status was observed using differentially expressed genes. Using RNA-seq in parallel with targeted DNA-seq, we identified mutational and gene expression signatures characteristic of p16+ and p16- HNSCC. Our gene signatures are consistent with previously published data including TCGA and support the need to further explore the biologic relevance of these alterations in HNSCC.


Assuntos
Inibidor p16 de Quinase Dependente de Ciclina/genética , Neoplasias de Cabeça e Pescoço/genética , Carcinoma de Células Escamosas de Cabeça e Pescoço/genética , Adulto , Idoso , Inibidor p16 de Quinase Dependente de Ciclina/metabolismo , DNA Viral/genética , Gerenciamento de Dados , Bases de Dados de Ácidos Nucleicos , Testes Diagnósticos de Rotina , Feminino , Expressão Gênica , Perfilação da Expressão Gênica , Neoplasias de Cabeça e Pescoço/metabolismo , Neoplasias de Cabeça e Pescoço/virologia , Humanos , Masculino , Pessoa de Meia-Idade , Mutação , Neoplasias Orofaríngeas/genética , Neoplasias Orofaríngeas/metabolismo , Neoplasias Orofaríngeas/virologia , Papillomaviridae/genética , Infecções por Papillomavirus/genética , Infecções por Papillomavirus/metabolismo , Infecções por Papillomavirus/virologia , Carcinoma de Células Escamosas de Cabeça e Pescoço/metabolismo , Carcinoma de Células Escamosas de Cabeça e Pescoço/virologia , Transcriptoma
10.
J Recept Signal Transduct Res ; 29(3-4): 182-94, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19604037

RESUMO

Targeting dysregulated signaling pathways in tumors has led to the development of a novel class of signal transduction inhibitors, including inhibitors of the epidermal growth factor (EGF) receptor (EGFR). To dissect oncogenic pathways, identify key pathway determinants, and evaluate the efficacy of targeted agents, it is vital to develop technologies that allow the detection of temporal signaling events under physiological conditions. Here we report the application of a label-free optical biosensor to reveal the rapid response of cancer cells to EGF, expressed as a dynamic mass redistribution (DMR) signal. In response to EGF, squamous cell carcinoma of the head and neck cells exhibited a rapid rise in DMR signal, whereas lung adenocarcinoma cells showed a biphasic DMR profile, suggesting a cell type-dependent DMR response. Pharmacological studies suggested the importance of EGFR and the phosphatidylinositol-3 kinase pathway in mediating the EGF-induced DMR response. The defined DMR signatures offer a simple yet sensitive tool for evaluating EGFR-targeted agents, as shown with gefitinib and erlotinib. The assay can also be used for cell-based high-throughput screening of EGF pathway inhibitors, as demonstrated by its robust performance in a 384-well plate format (Z' > 0.5). This technology is applicable to other oncogenic pathways for the discovery of novel therapeutic agents for the treatment of various cancers.


Assuntos
Técnicas Biossensoriais , Receptores ErbB/metabolismo , Neoplasias/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Transdução de Sinais/efeitos dos fármacos , Antineoplásicos/isolamento & purificação , Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Fator de Crescimento Epidérmico/farmacologia , Cloridrato de Erlotinib , Gefitinibe , Humanos , Inibidores de Proteínas Quinases/farmacologia , Quinazolinas/farmacologia , Transdução de Sinais/fisiologia
11.
Nanotechnology ; 20(22): 225102, 2009 Jun 03.
Artigo em Inglês | MEDLINE | ID: mdl-19433879

RESUMO

Nanoparticle quantum dots (QDs) provide sharper and more photostable fluorescent signals than organic dyes, allowing quantification of multiple biomarkers simultaneously. In this study, we quantified the expression of epidermal growth factor receptor (EGFR) and E-cadherin (E-cad) in the same cells simultaneously by using secondary antibody-conjugated QDs with two different emission wavelengths (QD605 and QD565) and compared the cellular distribution of EGFR and E-cad between EGFR-tyrosine kinase inhibitor (TKI)-insensitive and -sensitive lung and head and neck cancer cell lines. Relocalization of EGFR and E-cad upon treatment with the EGFR-TKI erlotinib in the presence of EGF was visualized and analyzed quantitatively. Our results showed that QD-immunocytochemistry (ICC)-based technology can not only quantify basal levels of multiple biomarkers but also track the localization of the biomarkers upon biostimulation. With this new technology we found that in EGFR-TKI-insensitive cells, EGFR and E-cad were located mainly in the cytoplasm; while in sensitive cells, they were found mainly on the cell membrane. After induction with EGF, both EGFR and E-cad internalized to the cytoplasm, but the internalization capability in sensitive cells was greater than that in insensitive cells. Quantification also showed that inhibition of EGF-induced EGFR and E-cad internalization by erlotinib in the sensitive cells was stronger than that in the insensitive cells. These studies demonstrate substantial differences between EGFR-TKI-insensitive and -sensitive cancer cells in EGFR and E-cad expression and localization both at the basal level and in response to EGF and erlotinib. QD-based analysis facilitates the understanding of the features of EGFR-TKI-insensitive versus -sensitive cancer cells and may be used in the prediction of patient response to EGFR-targeted therapy.


Assuntos
Caderinas/metabolismo , Receptores ErbB/metabolismo , Imuno-Histoquímica/métodos , Neoplasias/metabolismo , Pontos Quânticos , Quinazolinas/farmacologia , Antineoplásicos/farmacologia , Carcinoma de Células Escamosas/metabolismo , Linhagem Celular Tumoral , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Endossomos/metabolismo , Cloridrato de Erlotinib , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Neoplasias de Cabeça e Pescoço/metabolismo , Humanos , Neoplasias Pulmonares/metabolismo , Lisossomos/metabolismo , Inibidores de Proteínas Quinases/farmacologia , Proteínas Tirosina Quinases/antagonistas & inibidores , Transdução de Sinais/efeitos dos fármacos
12.
Clin Cancer Res ; 14(5): 1310-6, 2008 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-18316549

RESUMO

Cancer nanotherapeutics are rapidly progressing and are being implemented to solve several limitations of conventional drug delivery systems such as nonspecific biodistribution and targeting, lack of water solubility, poor oral bioavailability, and low therapeutic indices. To improve the biodistribution of cancer drugs, nanoparticles have been designed for optimal size and surface characteristics to increase their circulation time in the bloodstream. They are also able to carry their loaded active drugs to cancer cells by selectively using the unique pathophysiology of tumors, such as their enhanced permeability and retention effect and the tumor microenvironment. In addition to this passive targeting mechanism, active targeting strategies using ligands or antibodies directed against selected tumor targets amplify the specificity of these therapeutic nanoparticles. Drug resistance, another obstacle that impedes the efficacy of both molecularly targeted and conventional chemotherapeutic agents, might also be overcome, or at least reduced, using nanoparticles. Nanoparticles have the ability to accumulate in cells without being recognized by P-glycoprotein, one of the main mediators of multidrug resistance, resulting in the increased intracellular concentration of drugs. Multifunctional and multiplex nanoparticles are now being actively investigated and are on the horizon as the next generation of nanoparticles, facilitating personalized and tailored cancer treatment.


Assuntos
Antineoplásicos/administração & dosagem , Sistemas de Liberação de Medicamentos , Nanopartículas/administração & dosagem , Neoplasias/tratamento farmacológico , Humanos , Nanomedicina
13.
Cancer Res ; 67(15): 7518-24, 2007 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-17671223

RESUMO

Squamous cell carcinoma of the head and neck (SCCHN) metastasizes to the lymph nodes and lungs. We have generated previously an orthotopic mouse model for head and neck metastasis and did in vivo selection of SCCHN cells through four rounds of serial metastases. A subpopulation of 686LN cells with high metastatic potential (686LN-Ms) was isolated. When the highly metastatic cells were compared with their low metastatic parental cells (686LN-Ps), we found that CXC chemokine receptor-4 (CXCR4) mRNA levels were significantly higher in the 686LN-Ms cells than the 686LN-Ps cells. Interestingly, the metastatic subclones had lost epithelial morphology and acquired mesenchymal features, which were maintained during cell expansion in vitro. This was featured by decreased E-cadherin and involucrin and increased vimentin and integrin beta(1). These results imply that CXCR4 and epithelial-mesenchymal transition markers can be potential biomarkers to identify the subpopulation of cells with high metastatic potential. Using the orthotopic SCCHN animal model, we showed that anti-CXCR4 treatment suppressed primary tumor growth by inhibiting tumor angiogenesis and prevented lung metastasis. Because the reduction of metastasis seen in the treated group could have resulted from 2-fold reduction in primary tumor size compared with that in the control group, we examined the effects of the CXCR4 antagonist in an experimental metastatic animal model in which 686LN-Ms cells were i.v. injected. 686LN-Ms cells failed to metastasize in the CXCR4 antagonist-treated group, whereas they metastasized to the lungs in the control group. Our data indicate that CXCR4 is an important target to inhibit tumor progression in SCCHN.


Assuntos
Carcinoma de Células Escamosas/metabolismo , Neoplasias de Cabeça e Pescoço/prevenção & controle , Neoplasias Pulmonares/prevenção & controle , Oligopeptídeos/uso terapêutico , Receptores CXCR4/metabolismo , Animais , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/metabolismo , Northern Blotting , Western Blotting , Carcinoma de Células Escamosas/prevenção & controle , Carcinoma de Células Escamosas/secundário , Divisão Celular/fisiologia , Feminino , Neoplasias de Cabeça e Pescoço/metabolismo , Neoplasias de Cabeça e Pescoço/patologia , Humanos , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/secundário , Camundongos , Camundongos Nus , Dados de Sequência Molecular , Neovascularização Patológica/prevenção & controle , Análise de Sequência com Séries de Oligonucleotídeos , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptores CXCR4/antagonistas & inibidores , Receptores CXCR4/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução Genética
14.
Vis Comput Ind Biomed Art ; 2(1): 18, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-32190408

RESUMO

It can be challenging to detect tumor margins during surgery for complete resection. The purpose of this work is to develop a novel learning method that learns the difference between the tumor and benign tissue adaptively for cancer detection on hyperspectral images in an animal model. Specifically, an auto-encoder network is trained based on the wavelength bands on hyperspectral images to extract the deep information to create a pixel-wise prediction of cancerous and benign pixel. According to the output hypothesis of each pixel, the misclassified pixels would be reclassified in the right prediction direction based on their adaptive weights. The auto-encoder network is again trained based on these updated pixels. The learner can adaptively improve the ability to identify the cancer and benign tissue by focusing on the misclassified pixels, and thus can improve the detection performance. The adaptive deep learning method highlighting the tumor region proved to be accurate in detecting the tumor boundary on hyperspectral images and achieved a sensitivity of 92.32% and a specificity of 91.31% in our animal experiments. This adaptive learning method on hyperspectral imaging has the potential to provide a noninvasive tool for tumor detection, especially, for the tumor whose margin is indistinct and irregular.

15.
J Biophotonics ; 11(3)2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-28921845

RESUMO

Hyperspectral imaging (HSI) holds the potential for the noninvasive detection of cancers. Oral cancers are often diagnosed at a late stage when treatment is less effective and the mortality and morbidity rates are high. Early detection of oral cancer is, therefore, crucial in order to improve the clinical outcomes. To investigate the potential of HSI as a noninvasive diagnostic tool, an animal study was designed to acquire hyperspectral images of in vivo and ex vivo mouse tongues from a chemically induced tongue carcinogenesis model. A variety of machine-learning algorithms, including discriminant analysis, ensemble learning, and support vector machines, were evaluated for tongue neoplasia detection using HSI and were validated by the reconstructed pathological gold-standard maps. The diagnostic performance of HSI, autofluorescence imaging, and fluorescence imaging were compared in this study. Color-coded prediction maps were generated to display the predicted location and distribution of premalignant and malignant lesions. This study suggests that hyperspectral imaging combined with machine-learning techniques can provide a noninvasive tool for the quantitative detection and delineation of squamous neoplasia.


Assuntos
Carcinogênese , Carcinoma de Células Escamosas/diagnóstico por imagem , Carcinoma de Células Escamosas/patologia , Imagem Óptica , Neoplasias da Língua/diagnóstico por imagem , Neoplasias da Língua/patologia , Animais , Modelos Animais de Doenças , Processamento de Imagem Assistida por Computador , Camundongos
16.
Artigo em Inglês | MEDLINE | ID: mdl-30220773

RESUMO

Hyperspectral imaging (HSI), a non-contact optical imaging technique, has been recently used along with machine learning technique to provide diagnostic information about ex-vivo surgical specimens for optical biopsy. The computer-aided diagnostic approach requires accurate ground truths for both training and validation. This study details a processing pipeline for registering the cancer-normal margin from a digitized histological image to the gross-level HSI of a tissue specimen. Our work incorporates an initial affine and control-point registration followed by a deformable Demons-based registration of the moving mask obtained from the histological image to the fixed mask made from the HS image. To assess registration quality, Dice similarity coefficient (DSC) measures the image overlap, visual inspection is used to evaluate the margin, and average target registration error (TRE) of needle-bored holes measures the registration error between the histologic and HSI images. Excised tissue samples from seventeen patients, 11 head and neck squamous cell carcinoma (HNSCCa) and 6 thyroid carcinoma, were registered according to the proposed method. Three registered specimens are illustrated in this paper, which demonstrate the efficacy of the registration workflow. Further work is required to apply the technique to more patient data and investigate the ability of this procedure to produce suitable gold standards for machine learning validation.

17.
Clin Cancer Res ; 24(4): 858-869, 2018 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-29180609

RESUMO

Purpose: Previous studies revealed diverging results regarding the role of survivin in squamous cell carcinoma of the head and neck (SCCHN). This study aimed to evaluate the clinical significance of survivin expression in SCCHN; the function of survivin in DNA-damage repair following ionizing radiation therapy (RT) in SCCHN cells; and the potential of honokiol to enhance RT through downregulation of survivin.Experimental Design: Expression of survivin in SCCHN patient primary tumor tissues (n = 100) was analyzed and correlated with clinical parameters. SCCHN cell lines were used to evaluate the function of survivin and the effects of honokiol on survivin expression in vitro and in vivoResults: Overexpression of survivin was significantly associated with lymph nodes' metastatic status (P = 0.025), worse overall survival (OS), and disease-free survival (DFS) in patients receiving RT (n = 65, OS: P = 0.024, DFS: P = 0.006) and in all patients with SCCHN (n = 100, OS: P = 0.002, DFS: P = 0.003). In SCCHN cells, depletion of survivin led to increased DNA damage and cell death following RT, whereas overexpression of survivin increased clonogenic survival. RT induced nuclear accumulation of survivin and its molecular interaction with γ-H2AX and DNA-PKCs. Survivin specifically bound to DNA DSB sites induced by I-SceI endonuclease. Honokiol (which downregulates survivin expression) in combination with RT significantly augmented cytotoxicity in SCCHN cells with acquired radioresistance and inhibited growth in SCCHN xenograft tumors.Conclusions: Survivin is a negative prognostic factor and is involved in DNA-damage repair induced by RT. Targeting survivin using honokiol in combination with RT may provide novel therapeutic opportunities. Clin Cancer Res; 24(4); 858-69. ©2017 AACR.


Assuntos
Compostos de Bifenilo/farmacologia , Carcinoma de Células Escamosas/terapia , Neoplasias de Cabeça e Pescoço/terapia , Lignanas/farmacologia , Survivina/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto , Animais , Antineoplásicos Fitogênicos/farmacologia , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/metabolismo , Linhagem Celular Tumoral , Quimiorradioterapia , Regulação para Baixo , Feminino , Neoplasias de Cabeça e Pescoço/genética , Neoplasias de Cabeça e Pescoço/metabolismo , Humanos , Estimativa de Kaplan-Meier , Camundongos Nus , Radiossensibilizantes/farmacologia , Survivina/genética , Carga Tumoral/efeitos dos fármacos , Carga Tumoral/genética , Carga Tumoral/efeitos da radiação
18.
Artigo em Inglês | MEDLINE | ID: mdl-30220770

RESUMO

Hyperspectral imaging (HSI) is an emerging imaging modality that can provide a noninvasive tool for cancer detection and image-guided surgery. HSI acquires high-resolution images at hundreds of spectral bands, providing big data to differentiating different types of tissue. We proposed a deep learning based method for the detection of head and neck cancer with hyperspectral images. Since the deep learning algorithm can learn the feature hierarchically, the learned features are more discriminative and concise than the handcrafted features. In this study, we adopt convolutional neural networks (CNN) to learn the deep feature of pixels for classifying each pixel into tumor or normal tissue. We evaluated our proposed classification method on the dataset containing hyperspectral images from 12 tumor-bearing mice. Experimental results show that our method achieved an average accuracy of 91.36%. The preliminary study demonstrated that our deep learning method can be applied to hyperspectral images for detecting head and neck tumors in animal models.

19.
Clin Lung Cancer ; 18(3): e203-e210, 2017 05.
Artigo em Inglês | MEDLINE | ID: mdl-28073681

RESUMO

OBJECTIVES: Compelling evidence demonstrates that CXC-chemokine receptor 4 (CXCR4) is involved in tumor invasion, angiogenesis, metastasis, and resistance to chemotherapy in addition to being one of the coreceptors for T-tropic human immunodeficiency virus entry into T cells. However, it remains controversial as to how to identify functionally activated CXCR4 in tumor biopsies, which would assist in determining which patients may benefit from potential CXCR4-targeted therapy. MATERIALS AND METHODS: Immunohistochemistry (IHC) staining on archival tissues of patients with non-small-cell lung cancer (NSCLC) was used to detect a panel of biomarkers, including phospho-ERK1/2, phospho-AKT, and E-cadherin, which are relevant to downstream signaling of CXCR4 and epithelial to mesenchymal transition (EMT). We also examined whether subcellular localization of CXCR4 could help define possible activation of CXCR4. RESULTS: A total of 94 primary tumor tissue samples from patients with NSCLC were included. Sixty-six patients had both cytomembranous and nuclear staining of CXCR4, 22 had solely nuclear staining, 5 had solely cytomembranous staining, and 1 had negative staining. Cytoplasmic location of CXCR4 with or without nuclear location was associated with loss of the epithelial marker E-cadherin (P = .0015) and activation of ERK1/2 (P = .0121) and AKT (P = .0024), suggesting EMT in these tumors; whereas tumors with only nuclear location of CXCR4 were more indolent and preserved an epithelial phenotype. CONCLUSIONS: Our study suggests that different subcellular localization of CXCR4 may be associated with different activation states; cytoplasmic CXCR4 seems to correlate with biomarker changes associated with EMT in NSCLC.


Assuntos
Biomarcadores Tumorais/metabolismo , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Citoplasma/metabolismo , Epitélio/metabolismo , Neoplasias Pulmonares/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Receptores CXCR4/metabolismo , Idoso , Carcinoma Pulmonar de Células não Pequenas/diagnóstico , Transição Epitelial-Mesenquimal , Epitélio/patologia , Feminino , Humanos , Imuno-Histoquímica , Neoplasias Pulmonares/diagnóstico , Sistema de Sinalização das MAP Quinases , Masculino , Transporte Proteico
20.
Mol Cancer Ther ; 16(4): 729-738, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-28119490

RESUMO

We previously reported that the EGFR-targeted inhibitor erlotinib induces G1 arrest of squamous cell carcinoma of the head and neck (SCCHN) cell lines without inducing significant apoptosis. Large-scale genomic studies suggest that >50% of SCCHN cases have activation of PI3K pathways. This study investigated whether cotargeting of EGFR and PI3K has synergistic antitumor effects and apoptosis induction. We examined growth suppression, apoptosis, and signaling pathway modulation resulting from single and combined targeting of EGFR and PI3K with erlotinib and BKM120, respectively, in a panel of SCCHN cell lines and a xenograft model of SCCHN. In a panel of 12 cell lines, single targeting of EGFR with erlotinib or PI3K with BKM120 suppressed cellular growth without inducing significant apoptosis. Cotargeting of EGFR and PI3K synergistically inhibited SCCHN cell line and xenograft tumor growth, but induced variable apoptosis; some lines were highly sensitive, others were resistant. Mechanistic studies revealed that the combination inhibited both axes of the mTORC1 (S6 and 4EBP1) pathway in apoptosis-sensitive cell lines along with translational inhibition of Bcl-2, Bcl-xL, and Mcl-1, but failed to inhibit p-4EBP1, Bcl-2, Bcl-xL, and Mcl-1 in an apoptosis-resistant cell line. siRNA-mediated knockdown of eIF4E inhibited Bcl-2 and Mcl-1 and sensitized this cell line to apoptosis. Our results strongly suggest that cotargeting of EGFR and PI3K is synergistic and induces apoptosis of SCCHN cell lines by inhibiting both axes of the AKT-mTOR pathway and translational regulation of antiapoptotic Bcl-2 proteins. These findings may guide the development of clinical trials using this combination of agents. Mol Cancer Ther; 16(4); 729-38. ©2017 AACR.


Assuntos
Aminopiridinas/administração & dosagem , Protocolos de Quimioterapia Combinada Antineoplásica/administração & dosagem , Carcinoma de Células Escamosas/tratamento farmacológico , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Cloridrato de Erlotinib/administração & dosagem , Neoplasias de Cabeça e Pescoço/tratamento farmacológico , Morfolinas/administração & dosagem , Proteínas Proto-Oncogênicas c-akt/genética , Serina-Treonina Quinases TOR/genética , Aminopiridinas/farmacologia , Animais , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Apoptose , Carcinoma de Células Escamosas/genética , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Citoproteção/efeitos dos fármacos , Sinergismo Farmacológico , Cloridrato de Erlotinib/farmacologia , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Neoplasias de Cabeça e Pescoço/genética , Humanos , Camundongos , Morfolinas/farmacologia , Transdução de Sinais/efeitos dos fármacos , Carcinoma de Células Escamosas de Cabeça e Pescoço , Ensaios Antitumorais Modelo de Xenoenxerto
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA