Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Clin Cancer Res ; 14(1): 188-98, 2008 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-18172270

RESUMO

PURPOSE: Our preclinical experiments indicated that Romidepsin (Depsipeptide FK228; DP) mediates growth arrest and apoptosis in cultured lung cancer cells. A phase II trial was done to examine clinical and molecular responses mediated by this histone deacetylase inhibitor in lung cancer patients. EXPERIMENTAL DESIGN: Nineteen patients with neoplasms refractory to standard therapy received 4-h DP infusions (17.8 mg/m(2)) on days 1 and 7 of a 21-day cycle. Each full course of therapy consisted of two identical 21-day cycles. Plasma DP levels were evaluated by liquid chromatography-mass spectrometry techniques. A variety of molecular end points were assessed in tumor biopsies via immunohistochemistry techniques. Long oligo arrays were used to examine gene expression profiles in laser-captured tumor cells before and after DP exposure, relative to lung cancer cells and adjacent normal bronchial epithelia from patients undergoing pulmonary resections. RESULTS: Nineteen patients were evaluable for toxicity assessment; 18 were evaluable for treatment response. Myelosuppression was dose limiting in one individual. No significant cardiac toxicities were observed. Maximum steady-state plasma DP concentrations ranged from 384 to 1,114 ng/mL. No objective responses were observed. Transient stabilization of disease was noted in nine patients. DP enhanced acetylation of histone H4, increased p21 expression in lung cancer cells, and seemed to shift global gene expression profiles in these cells toward those detected in normal bronchial epithelia. CONCLUSION: Although exhibiting minimal clinical efficacy at this dose and schedule, DP mediates biological effects that may warrant further evaluation of this histone deacetylase inhibitor in combination with novel-targeted agents in lung cancer patients.


Assuntos
Antibióticos Antineoplásicos/uso terapêutico , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Carcinoma de Células Pequenas/tratamento farmacológico , Depsipeptídeos/uso terapêutico , Neoplasias Pulmonares/tratamento farmacológico , Acetilação/efeitos dos fármacos , Adulto , Idoso , Antibióticos Antineoplásicos/farmacocinética , Depsipeptídeos/farmacocinética , Feminino , Expressão Gênica/efeitos dos fármacos , Perfilação da Expressão Gênica , Histonas/efeitos dos fármacos , Humanos , Imuno-Histoquímica , Lasers , Masculino , Microdissecção , Pessoa de Meia-Idade , Análise de Sequência com Séries de Oligonucleotídeos , Reação em Cadeia da Polimerase Via Transcriptase Reversa
2.
Cancer J ; 13(2): 105-13, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17476138

RESUMO

BACKGROUND: Whereas histone deacetylase inhibitors are known to modulate chromatin structure, the precise mechanisms by which these novel agents induce apoptosis in cancer cells remain unknown. Previously we reported that depsipeptide FK228 depletes epidermal growth factor receptor (EGFR), erbB2, and Raf-1 kinases in non-small cell lung cancer cells. In the present study we sought to further define the mechanisms by which FK228 modulates oncoprotein signaling and to ascertain whether altered signal transduction contributes to FK228-mediated apoptosis in lung cancer cells. METHODS: Cultured non-small cell lung cancer cells were treated with FK228 alone or FK228 with a variety of kinase inhibitors. Proliferation and apoptosis mediated by drug exposure were assessed by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium, and Apo-BrdU techniques. Western blot and kinase assays were used to evaluate EGFR-related signal transduction pathways. Lung cancer cells were transduced with adenoviral vectors expressing activated AKT or mitogen-activated protein kinase kinase (MEK) 1 or beta-galactosidase to determine whether constitutive activation of mitogen-activated protein kinase signaling could abrogate FK228-mediated apoptosis. RESULTS: FK228 treatment induced time-dependent apoptosis in lung cancer cells expressing wild-type or mutant EGFR. FK228 inhibited a variety of EGFR-related pathways including Src, RAF-MEK-extracellular signal-regulated kinase (ERK) 1/2 and phosphatidyl inositol-3 kinase (PI3K)/AKT, resulting in down-regulation of Bcl-2 and Bcl-xL and up-regulation of Bax. The kinase inhibitors AG1478, AG825, PD98059, and LY294002 markedly enhanced FK228-induced apoptosis in lung cancer cells. Coincident with inhibition of ERK1/2 and PI3K/AKT survival pathways, FK228 enhanced p38 and stress-activated protein kinase/c-Jun NH2-terminal kinase stress signaling. Constitutive expression of MEK1 but not AKT markedly reduced FK228-mediated apoptosis in lung cancer cells. CONCLUSIONS: FK228 inhibits EGFR expression and modulates a variety of downstream mediators regulating proliferation and stress responses in lung cancer cells. These data highlight the significance of MEK signaling with respect to FK228-mediated apoptosis and support evaluation of histone deacetylase inhibitors in conjunction with agents specifically targeting mitogen-activated protein kinases in patients with lung cancer.


Assuntos
Apoptose/efeitos dos fármacos , Carcinoma Pulmonar de Células não Pequenas/patologia , Depsipeptídeos/farmacologia , Inibidores Enzimáticos/farmacologia , Neoplasias Pulmonares/patologia , Transdução de Sinais/efeitos dos fármacos , Cromonas/farmacologia , Regulação para Baixo , Receptores ErbB/antagonistas & inibidores , Receptores ErbB/genética , MAP Quinases Reguladas por Sinal Extracelular/efeitos dos fármacos , Flavonoides/farmacologia , Inibidores de Histona Desacetilases , Humanos , Morfolinas/farmacologia , Mutação , Proteínas Proto-Oncogênicas c-akt/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-bcl-2/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Quinazolinas , Receptor ErbB-2/antagonistas & inibidores , Células Tumorais Cultivadas , Tirfostinas/farmacologia , Regulação para Cima , Proteína X Associada a bcl-2/efeitos dos fármacos , Proteína X Associada a bcl-2/metabolismo , Proteína bcl-X/efeitos dos fármacos , Proteína bcl-X/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/efeitos dos fármacos
3.
Clin Cancer Res ; 12(19): 5777-85, 2006 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-17020984

RESUMO

PURPOSE: The DNA methylation paradox, manifested as derepression of cancer-testis antigens, and silencing of tumor suppressors during malignant transformation, provides the rationale for the utilization of chromatin remodeling agents for cancer therapy. A phase I trial was done to examine pharmacokinetics, toxicities, and gene expression mediated by 5-aza-2'-deoxycytidine (DAC) in patients with thoracic malignancies. EXPERIMENTAL DESIGN: Thirty-five patients with cancers refractory to standard therapy received continuous 72-hour DAC infusions using a phase I dose-escalation schema. Each full course of therapy consisted of two identical 35-day cycles. Plasma DAC levels were evaluated by liquid chromatography-mass spectrometry techniques. Quantitative reverse transcription-PCR, methylation-specific PCR, and immunohistochemical techniques were used to evaluate NY-ESO-1, MAGE-3, and p16 expression in tumor biopsies. Long oligonucleotide arrays were used to evaluate gene expression profiles in laser-captured tumor cells before and after DAC exposure. RESULTS: Thirty-five patients were evaluable for toxicities; 25 were evaluable for treatment response. Myelosuppression constituted dose-limiting toxicity. The maximum tolerated dose of DAC was 60 to 75 mg/m(2) depending on the number of prior cytotoxic chemotherapy regimens. No objective responses were observed. Plasma DAC concentrations approximated thresholds for gene induction in cultured cancer cells. Target gene induction was observed in 36% of patients. Posttreatment antibodies to NY-ESO-1 were detected in three patients exhibiting NY-ESO-1 induction in their tumor tissues. Complex, heterogeneous gene expression profiles were observed in pretreatment and posttreatment tissues. CONCLUSION: Prolonged DAC infusions can modulate gene expression in primary thoracic malignancies. These findings support further evaluation of DNA-demethylating agents alone or in combination with other regimens targeting induced gene products for the treatment of these neoplasms.


Assuntos
Antimetabólitos Antineoplásicos/farmacologia , Azacitidina/análogos & derivados , Neoplasias Esofágicas/genética , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Neoplasias Pulmonares/genética , Neoplasias Pleurais/genética , Adulto , Idoso , Antígenos de Neoplasias/genética , Antígenos de Neoplasias/metabolismo , Azacitidina/farmacologia , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Carcinoma Pulmonar de Células não Pequenas/genética , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Carcinoma de Células Escamosas/tratamento farmacológico , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/metabolismo , Metilases de Modificação do DNA/antagonistas & inibidores , Decitabina , Neoplasias Esofágicas/tratamento farmacológico , Neoplasias Esofágicas/metabolismo , Feminino , Genes p16/fisiologia , Humanos , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/metabolismo , Masculino , Dose Máxima Tolerável , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Mesotelioma/tratamento farmacológico , Mesotelioma/genética , Mesotelioma/metabolismo , Pessoa de Meia-Idade , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Neoplasias Pleurais/tratamento farmacológico , Neoplasias Pleurais/metabolismo , Ativação Transcricional
4.
J Thorac Cardiovasc Surg ; 131(2): 298-306, 2006 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-16434257

RESUMO

OBJECTIVE: DNA methyltransferase (DNMT)1, DNMT3b, or both, facilitate malignant transformation through chromatin remodeling mechanisms. The present study was undertaken to examine the effects of antisense-mediated inhibition of DNMT expression in cultured thoracic malignancies. METHODS: CALU-6 and A549 lung cancer, SKGT5 and BIC esophageal adenocarcinoma, and H2373 and H2052 malignant pleural mesothelioma (MPM) cells, as well as normal human bronchial epithelial (NHBE) cells, were transfected with phosphorothioate-modified antisense oligos targeting DNMT1, DNMT3b, or both, or mismatch oligos. Quantitative reverse transcription-polymerase chain reaction, Western blotting, trypan blue exclusion, and ApoBrdU techniques were used to evaluate DNMT expression, proliferation, and apoptosis after antisense oligo transfections. Gene expression profiles were assessed by using long-oligo array techniques. RESULTS: Antisense oligos mediated specific and dose-dependent depletion of DNMT1 and DNMT3b, resulting in pronounced inhibition of proliferation of all thoracic cancer lines, but not NHBE cells. Depletion of DNMT1 or DNMT3b coincided with dramatic, caspase-dependent, p53-independent apoptosis in 4 of the 6 thoracic cancer lines. The antiproliferative effects of the antisense oligos were not attributable to induction of RASSF1A, p16, or p21 tumor suppressor genes, and did not coincide with demethylation of genes encoding cancer-testis antigens. DNA methyltransferase knockdown mediated induction of numerous genes regulating response to genotoxic stress. Gene expression profiles after DNMT1, DNMT3b, or combined DNMT1/3b depletion were remarkably similar, yet distinctly different from expression profiles mediated by 5 aza 2' deoxycytidine. CONCLUSIONS: Antisense oligos targeting DNMT1 and DNMT3b induce genomic stress, and mediate potent growth inhibition in lung and esophageal cancer and MPM cells. These findings support further evaluation of DNMT knockdown strategies for cancer therapy.


Assuntos
Adenocarcinoma/fisiopatologia , Apoptose/fisiologia , DNA (Citosina-5-)-Metiltransferases/fisiologia , Neoplasias Esofágicas/fisiopatologia , Neoplasias Pulmonares/fisiopatologia , Mesotelioma/fisiopatologia , Neoplasias Pleurais/fisiopatologia , Adenocarcinoma/patologia , Linhagem Celular Tumoral , Proliferação de Células , DNA (Citosina-5-)-Metiltransferase 1 , Neoplasias Esofágicas/patologia , Expressão Gênica , Humanos , Neoplasias Pulmonares/patologia , Mesotelioma/patologia , Oligodesoxirribonucleotídeos Antissenso/farmacologia , Neoplasias Pleurais/patologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transfecção , Células Tumorais Cultivadas , DNA Metiltransferase 3B
5.
Cancer Res ; 66(2): 1105-13, 2006 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-16424047

RESUMO

Recent studies suggest that immunotherapy targeting specific tumor-associated antigens (TAAs) may be beneficial in cancer patients. However, most of these TAAs are tumor type specific and heterogeneous among patients, thus limiting their applications. Here, we describe the de novo induction of a cancer/testis antigen (CTA) for immunotherapy of tumors of various histologies. The murine CTA P1A, normally expressed only in a few tumor lines, could be induced de novo in all P1A-negative cancer lines of eight histologic origins in vitro and in various murine xenografts by systemic administration of 5-aza-2'-deoxycytidine. The induction of P1A expression correlated strongly with demethylation of the CpG island in the promoter region of this gene. The induced antigen was processed and presented properly for recognition by H-2L(d)-restricted P1A-specific CTLs. The combination of a demethylating agent and adoptive transfer of P1A-specific CTL effectively treated lung metastases in syngeneic mice challenged with P1A-negative 4T1 mammary carcinoma cells. These data show a novel strategy of combined chemoimmunotherapy of cancer targeting a CTA induced de novo in a broad range of tumor histologies, and support further evaluation of chromatin-remodeling agents for human cancer therapy.


Assuntos
Antígenos de Neoplasias/biossíntese , Antimetabólitos Antineoplásicos/farmacologia , Azacitidina/análogos & derivados , Imunoterapia Adotiva , Animais , Antígenos de Neoplasias/fisiologia , Azacitidina/farmacologia , Cromatina/metabolismo , Ilhas de CpG , Metilação de DNA , Decitabina , Perfilação da Expressão Gênica , Humanos , Neoplasias Pulmonares/imunologia , Neoplasias Pulmonares/secundário , Neoplasias Pulmonares/terapia , Neoplasias Mamárias Animais/imunologia , Neoplasias Mamárias Animais/patologia , Neoplasias Mamárias Animais/terapia , Camundongos , Neoplasias/imunologia , Neoplasias/terapia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transplante Heterólogo , Células Tumorais Cultivadas
6.
Cancer Res ; 65(17): 7763-74, 2005 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-16140944

RESUMO

Regulatory sequences recognized by the unique pair of paralogous factors, CTCF and BORIS, have been implicated in epigenetic regulation of imprinting and X chromosome inactivation. Lung cancers exhibit genome-wide demethylation associated with derepression of a specific class of genes encoding cancer-testis (CT) antigens such as NY-ESO-1. CT genes are normally expressed in BORIS-positive male germ cells deficient in CTCF and meCpG contents, but are strictly silenced in somatic cells. The present study was undertaken to ascertain if aberrant activation of BORIS contributes to derepression of NY-ESO-1 during pulmonary carcinogenesis. Preliminary experiments indicated that NY-ESO-1 expression coincided with derepression of BORIS in cultured lung cancer cells. Quantitative reverse transcription-PCR analysis revealed robust, coincident induction of BORIS and NY-ESO-1 expression in lung cancer cells, but not normal human bronchial epithelial cells following 5-aza-2'-deoxycytidine (5-azadC), Depsipeptide FK228 (DP), or sequential 5-azadC/DP exposure under clinically relevant conditions. Bisulfite sequencing, methylation-specific PCR, and chromatin immunoprecipitation (ChIP) experiments showed that induction of BORIS coincided with direct modulation of chromatin structure within a CpG island in the 5'-flanking noncoding region of this gene. Cotransfection experiments using promoter-reporter constructs confirmed that BORIS modulates NY-ESO-1 expression in lung cancer cells. Gel shift and ChIP experiments revealed a novel CTCF/BORIS-binding site in the NY-ESO-1 promoter, which unlike such sites in the H19-imprinting control region and X chromosome, is insensitive to CpG methylation in vitro. In vivo occupancy of this site by CTCF was associated with silencing of the NY-ESO-1 promoter, whereas switching from CTCF to BORIS occupancy coincided with derepression of NY-ESO-1. Collectively, these data indicate that reciprocal binding of CTCF and BORIS to the NY-ESO-1 promoter mediates epigenetic regulation of this CT gene in lung cancer cells, and suggest that induction of BORIS may be a novel strategy to augment immunogenicity of pulmonary carcinomas.


Assuntos
Antígenos de Neoplasias/genética , Proteínas de Ligação a DNA/metabolismo , Neoplasias Pulmonares/genética , Proteínas de Membrana/genética , Proteínas Repressoras/metabolismo , Antígenos de Neoplasias/biossíntese , Antígenos de Neoplasias/metabolismo , Sequência de Bases , Fator de Ligação a CCCTC , Linhagem Celular Tumoral , Imunoprecipitação da Cromatina , Metilação de DNA , Proteínas de Ligação a DNA/biossíntese , Proteínas de Ligação a DNA/genética , Regulação Neoplásica da Expressão Gênica , Inativação Gênica , Histonas/metabolismo , Humanos , Imuno-Histoquímica , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patologia , Proteínas de Membrana/antagonistas & inibidores , Proteínas de Membrana/biossíntese , Proteínas de Membrana/metabolismo , Dados de Sequência Molecular , Reação em Cadeia da Polimerase , Regiões Promotoras Genéticas , Ligação Proteica , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Sulfitos/farmacologia
7.
Oncogene ; 24(14): 2386-97, 2005 Mar 31.
Artigo em Inglês | MEDLINE | ID: mdl-15735751

RESUMO

cDNA arrays were used to examine gene induction in CALU-6 and H460 lung cancer cells mediated by sequential 5-aza 2'-deoxycytidine (DAC)/depsipeptide FK228 (DP) exposure in order to identify translational end points for clinical trials evaluating these agents. In both cell lines, sequential DAC/DP treatment induced expression of tissue factor pathway inhibitor-2 (TFPI-2), an inhibitor of Factor VII: tissue factor signal transduction known to diminish the malignant phenotype of cancer cells. TFPI-2 expression was diminished or absent in 16 of 32 cell lines established from thoracic malignancies. Sequential DAC/DP treatment induced TFPI-2 in cancer cells deficient for TFPI-2 expression in the basal state. Promoter methylation coincided with loss of TFPI-2 expression in a number of cancer lines. TFPI-2 promoter methylation was observed in one of five pulmonary adenocarcinomas, and seven of seven esophageal adenocarcinomas, but not corresponding normal tissues. DP enhanced acetylation of TFPI-2-associated histones in CALU-6 cells. DP or PDBU, alone, induced TFPI-2 expression in cancer cells deficient for TFPI-2 expression in the absence of promoter methylation. In these cells, DP-mediated TFPI-2 induction was abrogated by calphostin. Induction of TFPI-2 by distinct, yet cooperative mechanisms involving chromatin remodeling and PKC signaling strengthens the preclinical rationale for sequential administration of DNA demethylating agents and HDAC inhibitors in cancer patients. Furthermore, induction of TFPI-2 may be a useful surrogate marker of treatment response in individuals receiving sequential DAC/DP infusions.


Assuntos
Azacitidina/análogos & derivados , Azacitidina/farmacologia , Depsipeptídeos/farmacologia , Neoplasias Esofágicas/metabolismo , Glicoproteínas/genética , Neoplasias Pulmonares/metabolismo , Sequência de Bases , Linhagem Celular Tumoral , Primers do DNA , Decitabina , Neoplasias Esofágicas/patologia , Humanos , Neoplasias Pulmonares/patologia
8.
Clin Cancer Res ; 10(5): 1813-25, 2004 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-15014036

RESUMO

PURPOSE: Recent insights regarding the pathogenesis of malignant pleural mesothelioma (MPM) provide new opportunities for targeted molecular therapies for this highly lethal disease. The present study was undertaken to examine the effects of the histone deacetylase inhibitor, Depsipeptide (DP) FK228, in conjunction with the cyclin-dependent kinase inhibitor, Flavopiridol (FLA), in cultured MPM cells. EXPERIMENTAL DESIGN: Proliferation and apoptosis in drug-treated, virally transduced, or control cells were assessed by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide and Apo-bromodeoxyuridine techniques. Western blot and ELISA techniques were used to examine signal transduction and cell cycle-related protein levels in MPM cells exposed to DP and/or FLA in the presence or absence of calphostin, phorbol-12,13-dibutyrate, 5,6-dichloro-1-beta-D-ribofuranosyl-benzimidazole, or adenoviral p21 transduction. RESULTS: DP (1-50 ng/ml x 6 h) or FLA (100-200 nM x 72 h) alone, mediated low-level, dose-dependent growth inhibition in MPM cells. In contrast, sequential DP/FLA treatment mediated marked growth inhibition and apoptosis in these cell lines. The cytotoxic effects of DP/FLA were considerably less pronounced in cultured normal cells. The proapoptotic effects of DP/FLA treatment coincided with inhibition of DP-mediated induction of p21 by FLA. Overexpression of p21 by adenoviral gene transfer techniques rendered MPM cells refractory to the cytotoxic effects of this treatment regimen. In p21 reporter assays, promoter activation by DP was antagonized by FLA. The magnitude of inhibition of DP-mediated p21 induction by FLA exceeded that observed with the pTEFb antagonist 5,6-dichloro-1-beta-D-ribofuranosyl-benzimidazole. Calphostin C abrogated p21 induction mediated by DP and enhanced DP-mediated apoptosis in a manner comparable with FLA in MPM cells; in contrast, phorbol-12,13-dibutyrate blocked FLA-mediated inhibition of p21 induction by DP and markedly protected these cells from the apoptotic effects of sequential DP/FLA. CONCLUSIONS: FLA abrogates DP-mediated induction of p21 expression, in part, via inhibition of protein kinase C signaling and markedly potentiates the cytotoxic effects of DP in MPM cells.


Assuntos
Apoptose/efeitos dos fármacos , Divisão Celular/efeitos dos fármacos , Ciclinas/genética , Flavonoides/farmacologia , Piperidinas/farmacologia , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Inibidor de Quinase Dependente de Ciclina p21 , Inibidores Enzimáticos , Humanos , Mesotelioma , Neoplasias Pleurais
9.
J Thorac Cardiovasc Surg ; 127(2): 365-75, 2004 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-14762343

RESUMO

BACKGROUND: Constitutive activation of the phosphoinositide 3-kinase/protein kinase B survival signal transduction pathway influences the intrinsic chemoresistance of cancer cells. This study evaluates the effect of LY294002, a pharmacologic inhibitor of phosphoinositide 3-kinase, on the sensitivity of lung and esophageal cancer cells to paclitaxel (Taxol) in vitro. Materials and methods Cell viability and apoptosis of cancer cells treated with paclitaxel + LY294002 combinations were quantitated by methyl-thiazol-diphenyl-tetrazolium and terminal deoxynucleotidyltransferase-mediated dUTP nick-end labeling-based ApoBrdU assays, respectively. The effect of LY294002-mediated phosphoinositide 3-kinase inhibition on protein kinase B (Akt) activation and nuclear factor-kappaB signaling was determined by Western blot analysis. Nuclear factor-kappaB transcription activity in cultured cancer cells either at baseline or after treatments with LY294002 or BAY11-0782 (a pharmacologic inhibitor of nuclear factor-kappaB) was determined by the nuclear factor-kappaB-Luciferase reporter system. RESULTS: A 4- to more than 20-fold reduction of paclitaxel IC(50) values was observed in cancer cells treated with paclitaxel + LY294002 combinations. This was paralleled with synergistic induction of apoptosis. LY294002 treatment caused a significant dose-dependent inhibition of protein kinase B (Akt) activation and suppression of nuclear factor-kappaB transcriptional activity that was accompanied by elevation of IkappaB, the intrinsic inhibitor of nuclear factor-kappaB, and concomitant reduction of nuclear factor-kappaB-regulated antiapoptotic proteins cIAP1, cIAP2, and BclXL. Direct inhibition of nuclear factor-kappaB activity by BAY11-0782 also resulted in profound enhancement of paclitaxel sensitivity and paclitaxel-mediated induction of apoptosis in lung and esophageal cancer cells. CONCLUSION: LY294002-mediated inhibition of the phosphoinositide 3-kinase/protein kinase B-dependent survival pathway with secondary suppression of nuclear factor-kappaB transcriptional activity was associated with enhancement of paclitaxel cytotoxicity in lung and esophageal cancer cells. Direct inhibition of nuclear factor-kappaB by BAY11-0782 also sensitized these cancer cells to paclitaxel, indicating that nuclear factor-kappaB may be the crucial intermediary step connecting phosphoinositide 3-kinase/protein kinase B (Akt) to the intrinsic susceptibility of cancer cells to chemotherapeutic agents.


Assuntos
Antineoplásicos Fitogênicos/toxicidade , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Neoplasias Esofágicas/tratamento farmacológico , Neoplasias Pulmonares/tratamento farmacológico , Paclitaxel/toxicidade , Fosfatidilinositol 3-Quinases/efeitos dos fármacos , Inibidores de Fosfoinositídeo-3 Quinase , Proteínas Serina-Treonina Quinases , Proteínas Proto-Oncogênicas/antagonistas & inibidores , Proteínas Proto-Oncogênicas/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Antineoplásicos Fitogênicos/administração & dosagem , Apoptose/efeitos dos fármacos , Carcinoma Pulmonar de Células não Pequenas/enzimologia , Sobrevivência Celular/efeitos dos fármacos , Cromonas/administração & dosagem , Relação Dose-Resposta a Droga , Regulação para Baixo/efeitos dos fármacos , Quimioterapia Combinada , Ativação Enzimática/efeitos dos fármacos , Inibidores Enzimáticos/administração & dosagem , Neoplasias Esofágicas/enzimologia , Humanos , Neoplasias Pulmonares/enzimologia , Morfolinas/administração & dosagem , NF-kappa B/efeitos dos fármacos , NF-kappa B/metabolismo , Paclitaxel/administração & dosagem , Fosfatidilinositol 3-Quinases/metabolismo , Fosforilação/efeitos dos fármacos , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-akt , Estatística como Assunto , Fatores de Tempo , Falha de Tratamento , Células Tumorais Cultivadas
10.
J Natl Cancer Inst ; 94(7): 504-13, 2002 Apr 03.
Artigo em Inglês | MEDLINE | ID: mdl-11929951

RESUMO

BACKGROUND: Histone deacetylases (HDACs) modulate chromatin structure by regulating acetylation of core histone proteins. HDAC inhibitors, such as depsipeptide FR901228 (FK228), induce growth arrest and apoptosis in a variety of human cancer cells by mechanisms that cannot be attributed solely to histone acetylation. This study evaluated the mechanisms by which FK228 mediates apoptosis in non-small-cell lung cancer (NSCLC) cells. METHODS: Proliferation and apoptosis were assessed in a panel of NSCLC cell lines that vary in the expression of the growth-regulating proteins p53, pRb, and K-Ras treated with a clinically relevant dose of FK228 (25 ng/mL). Western blot and immunoprecipitation techniques were used to analyze expression of cell-cycle proteins (cyclin A, cyclin E, p53, and p21), signaling-related proteins (ErbB1, ErbB2, and Raf-1), activity of extracellular signal-regulated kinase 1 and 2 (ERK1/2), binding of mutant p53 and Raf-1 to heat shock protein (Hsp)90, and acetylation of Hsp90. RESULTS: FK228 treatment inhibited growth and induced apoptosis in NSCLC cells expressing wild-type or mutant p53. FK228 treatment led to altered expression of cyclin A, cyclin E, and p21, and to reduced expression of mutant, but not wild-type, p53. FK228-treated cells also were depleted of ErbB1, ErbB2, and Raf-1 proteins, and exhibited lower ERK1/2 activity. FK228 treatment also inhibited the binding of mutant p53 and Raf-1 to Hsp90; this inhibition was associated with acetylation of Hsp90. CONCLUSIONS: FK228 depletes the levels of several oncoproteins that are normally stabilized by binding to Hsp90 in cancer cells. The resulting ability of FK228 to diminish signal transduction via pathways involving Raf-1 and ERK may contribute to the potency and specificity of this novel antitumor agent.


Assuntos
Antibacterianos/farmacologia , Antibióticos Antineoplásicos/farmacologia , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Depsipeptídeos , Receptores ErbB/metabolismo , Neoplasias Pulmonares/metabolismo , Peptídeos Cíclicos , Proteínas Proto-Oncogênicas c-raf/metabolismo , Receptor ErbB-2/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Northern Blotting , Western Blotting , Carcinoma Pulmonar de Células não Pequenas/genética , Proteínas de Ciclo Celular/metabolismo , Receptores ErbB/genética , Proteínas de Choque Térmico HSP90/metabolismo , Humanos , Neoplasias Pulmonares/genética , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Testes de Precipitina , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Proto-Oncogênicas c-raf/genética , RNA Mensageiro/metabolismo , Receptor ErbB-2/genética , Células Tumorais Cultivadas/efeitos dos fármacos , Proteína Supressora de Tumor p53/genética
11.
J Thorac Cardiovasc Surg ; 123(2): 295-302, 2002 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-11828289

RESUMO

OBJECTIVE: Although well characterized in several solid tumors, the effects of Fas/Fas ligand interactions in malignant pleural mesothelioma cells have not been defined. The present study was undertaken to examine the functional status of the Fas/Fas ligand pathway in malignant pleural mesothelioma cells and to determine the feasibility of targeting this death-signal pathway for molecular intervention in patients with mesotheliomas. METHODS: Fas expression in primary normal human bronchial epithelial cells and 6 malignant pleural mesothelioma cell lines was quantified by means of flow cytometry. The caspase components of the Fas-mediated apoptotic pathway were evaluated by means of Western blot techniques. Soluble Fas ligand-mediated cytotoxicity and apoptosis were evaluated by means of MTS and TUNEL assays, respectively. Cisplatin (3 microg/mL) and lymphokine-activated killer cells were used to enhance mesothelioma sensitivity to soluble Fas ligand. An H2373 nude mouse xenograft model of malignant pleural mesothelioma was established to assess the in vivo effects of soluble Fas ligand. RESULTS: Four of 6 malignant pleural mesothelioma lines exhibited high levels of Fas expression, and 2 of 4 were inherently susceptible to soluble Fas ligand-mediated cytotoxicity (soluble Fas ligand 50% inhibitory concentration, < 15 ng/mL). Two soluble Fas ligand refractory cell lines (H2052 and H513) exhibited high levels of Fas receptor. Pretreatment with cisplatin resulted in a reduction of 50% inhibitory concentration from infinity to 4.17 +/- 0.14 ng/mL and 10.23 +/- 1.58 ng/mL, respectively. Two additional soluble Fas ligand refractory cell lines (H2595 and REN) expressed low levels of Fas. Exposure of these cells to lymphokine-activated killer cells or lymphokine-activated killer cell-conditioned medium followed by a 24-hour treatment with cisplatin resulted in a significant reduction in 50% inhibitory concentration of soluble Fas ligand and pronounced induction of apoptosis. Intraperitoneally administered soluble Fas ligand mediated regression of H2373 xenografts. CONCLUSION: The Fas/Fas ligand pathway in mesothelioma cells is either intrinsically intact or can be rendered functional with chemotherapeutic agents or immune effector cells. These preclinical data support further evaluation of strategies to enhance Fas-mediated apoptosis in mesotheliomas.


Assuntos
Apoptose , Mesotelioma/patologia , Neoplasias Pleurais/patologia , Receptor fas/metabolismo , Animais , Western Blotting , Caspases/metabolismo , Linhagem Celular , Cisplatino/farmacologia , Proteína Ligante Fas , Citometria de Fluxo , Humanos , Células Matadoras Ativadas por Linfocina/metabolismo , Glicoproteínas de Membrana/metabolismo , Camundongos , Camundongos Nus , Transplante Heterólogo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...