Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
1.
Commun Biol ; 6(1): 509, 2023 05 11.
Artigo em Inglês | MEDLINE | ID: mdl-37169941

RESUMO

Osimertinib sensitive and resistant NSCLC NCI-H1975 clones are used to model osimertinib acquired resistance in humanized and non-humanized mice and delineate potential resistance mechanisms. No new EGFR mutations or loss of the EGFR T790M mutation are found in resistant clones. Resistant tumors grown under continuous osimertinib pressure both in humanized and non-humanized mice show aggressive tumor regrowth which is significantly less sensitive to osimertinib as compared with parental tumors. 3-phosphoinositide-dependent kinase 1 (PDK1) is identified as a potential driver of osimertinib acquired resistance, and its selective inhibition by BX795 and CRISPR gene knock out, sensitizes resistant clones. In-vivo inhibition of PDK1 enhances the osimertinib sensitivity against osimertinib resistant xenograft and a patient derived xenograft (PDX) tumors. PDK1 knock-out dysregulates PI3K/Akt/mTOR signaling, promotes cell cycle arrest at the G1 phase. Yes-associated protein (YAP) and active-YAP are upregulated in resistant tumors, and PDK1 knock-out inhibits nuclear translocation of YAP. Higher expression of PDK1 and an association between PDK1 and YAP are found in patients with progressive disease following osimertinib treatment. PDK1 is a central upstream regulator of two critical drug resistance pathways: PI3K/AKT/mTOR and YAP.


Assuntos
Neoplasias Pulmonares , Camundongos , Animais , Humanos , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patologia , Receptores ErbB/genética , Fosfatidilinositol 3-Quinases , Proteínas Proto-Oncogênicas c-akt/genética , Inibidores de Proteínas Quinases/farmacologia , Inibidores de Proteínas Quinases/uso terapêutico , Resistencia a Medicamentos Antineoplásicos/genética , Mutação , Serina-Treonina Quinases TOR/genética , Fosfatidilinositóis
2.
Commun Biol ; 5(1): 167, 2022 02 24.
Artigo em Inglês | MEDLINE | ID: mdl-35210547

RESUMO

KRAS/LKB1 (STK11) NSCLC metastatic tumors are intrinsically resistant to anti-PD-1 or PD-L1 immunotherapy. In this study, we use a humanized mouse model to show that while carboplatin plus pembrolizumab reduce tumor growth moderately and transiently, the addition of the tumor suppressor gene TUSC2, delivered systemically in nanovesicles, to this combination, eradicates tumors in the majority of animals. Immunoprofiling of the tumor microenvironment shows the addition of TUSC2 mediates: (a) significant infiltration of reconstituted human functional cytotoxic T cells, natural killer cells, and dendritic cells; (b) induction of antigen-specific T cell responses; (c) enrichment of functional central and memory effector T cells; and (d) decreased levels of PD-1+ T cells, myeloid-derived suppressor cells, Tregs, and M2 tumor associated macrophages. Depletion studies show the presence of functional central and memory effector T cells are required for the efficacy. TUSC2 sensitizes KRAS/LKB1 tumors to carboplatin plus pembrolizumab through modulation of the immune contexture towards a pro-immune tumor microenvironment.


Assuntos
Proteínas Quinases Ativadas por AMP , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Carcinoma Pulmonar de Células não Pequenas , Neoplasias Pulmonares , Proteínas Proto-Oncogênicas p21(ras) , Proteínas Supressoras de Tumor , Proteínas Quinases Ativadas por AMP/genética , Proteínas Quinases Ativadas por AMP/imunologia , Animais , Anticorpos Monoclonais Humanizados/administração & dosagem , Carboplatina/administração & dosagem , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Carcinoma Pulmonar de Células não Pequenas/genética , Carcinoma Pulmonar de Células não Pequenas/terapia , Modelos Animais de Doenças , Genes Supressores de Tumor , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/terapia , Camundongos , Proteínas Proto-Oncogênicas p21(ras)/genética , Proteínas Proto-Oncogênicas p21(ras)/imunologia , Microambiente Tumoral , Proteínas Supressoras de Tumor/genética , Proteínas Supressoras de Tumor/imunologia
3.
Small ; 6(9): 1022-31, 2010 May 07.
Artigo em Inglês | MEDLINE | ID: mdl-20394071

RESUMO

Despite advances in controlled drug delivery, reliable methods for activatable, high-resolution control of drug release are needed. The hypothesis that the photothermal effect mediated by a near-infrared (NIR) laser and hollow gold nanospheres (HAuNSs) could modulate the release of anticancer agents is tested with biodegradable and biocompatible microspheres (1-15 microm) containing the antitumor drug paclitaxel (PTX) and HAuNSs (approximately 35 nm in diameter), which display surface plasmon absorbance in the NIR region. HAuNS-containing microspheres exhibit a NIR-induced thermal effect similar to that of plain HAuNSs. Rapid, repetitive PTX release from the PTX/HAuNS-containing microspheres is observed upon irradiation with NIR light (808 nm), whereas PTX release is insignificant when the NIR light is switched off. The release of PTX from the microspheres is readily controlled by the output power of the NIR laser, duration of irradiation, treatment frequency, and concentration of HAuNSs embedded inside the microspheres. In vitro, cancer cells incubated with PTX/HAuNS-loaded microspheres and irradiated with NIR light display significantly greater cytotoxic effects than cells incubated with the microspheres alone or cells irradiated with NIR light alone, owing to NIR-light-triggered drug release. Treatment of human U87 gliomas and MDA-MB-231 mammary tumor xenografts in nude mice with intratumoral injections of PTX/HAuNS-loaded microspheres followed by NIR irradiation results in significant tumor-growth delay compared to tumors treated with HAuNS-loaded microspheres (no PTX) and NIR irradiation or with PTX/HAuNS-loaded microspheres alone. The data support the feasibility of a therapeutic approach in which NIR light is used for simultaneous modulation of drug release and induction of photothermal cell killing.


Assuntos
Preparações de Ação Retardada/administração & dosagem , Preparações de Ação Retardada/química , Preparações de Ação Retardada/efeitos da radiação , Glioma/tratamento farmacológico , Nanoestruturas/química , Paclitaxel/química , Implantes Absorvíveis , Antineoplásicos/administração & dosagem , Antineoplásicos/química , Linhagem Celular Tumoral , Difusão , Glioma/patologia , Ouro/química , Temperatura Alta , Humanos , Raios Infravermelhos , Substâncias Macromoleculares/química , Teste de Materiais , Microesferas , Conformação Molecular , Nanoestruturas/administração & dosagem , Nanoestruturas/ultraestrutura , Paclitaxel/administração & dosagem , Propriedades de Superfície
4.
Oncogene ; 39(4): 801-813, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31554935

RESUMO

The role of RNA-dependent protein kinase R (PKR) and its association with misfolded protein expression in cancer cells are unclear. Herein we report that PKR regulates misfolded protein clearance by preventing it release through exosomes and promoting lysosomal degradation of misfolded prion proteins in cancer cells. We demonstrated that PKR contributes to the lysosome function and regulates misfolded prion protein clearance. We hypothesized that PKR-associated lysosome function is critical for cancer but not normal cell survival, representing an effective approach for highly targeted cancer therapy. In screening a compound library, we identified two PKR-associated compounds 1 and 2 (Pac 1 and 2) did not affect normal cells but selectively induced cell death in cancer cells depending on their PKR expression status. Pac 1 significantly inhibited the growth of human lung and breast xenograft tumors in mice with no toxicity. Pac 1 binds to PI4K2A and disrupts the PKR/PI4K2A-associated lysosome complex, contributing to destabilization of cancer cell lysosomes and triggering cell death. We observed that PKR and PI4K2A play significant prognostic roles in breast cancer patients. These results demonstrate that targeting of a PI4K2A/PKR lysosome complex may be an effective approach for cancer therapy.


Assuntos
Antineoplásicos/farmacologia , Lisossomos/metabolismo , Neoplasias/tratamento farmacológico , Fosfotransferases (Aceptor do Grupo Álcool)/antagonistas & inibidores , Proteólise , Resposta a Proteínas não Dobradas , eIF-2 Quinase/antagonistas & inibidores , Animais , Antineoplásicos/química , Linhagem Celular Tumoral , Sobrevivência Celular , Bases de Dados Genéticas/estatística & dados numéricos , Exossomos/metabolismo , Feminino , Humanos , Camundongos , Camundongos SCID , Antígenos de Histocompatibilidade Menor/metabolismo , Neoplasias/genética , Neoplasias/metabolismo , Neoplasias/patologia , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , Prognóstico , Dobramento de Proteína , Taxa de Sobrevida , Ensaios Antitumorais Modelo de Xenoenxerto , eIF-2 Quinase/metabolismo
5.
Cancer Immunol Res ; 7(8): 1267-1279, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31186248

RESUMO

Human tumor xenograft models do not replicate the human immune system and tumor microenvironment. We developed an improved humanized mouse model, derived from fresh cord blood CD34+ stem cells (CD34+ HSC), and combined it with lung cancer cell line-derived human xenografts or patient-derived xenografts (Hu-PDX). Fresh CD34+ HSCs could reconstitute detectable mature human leukocytes (hCD45+) in mice at four weeks without the onset of graft-versus-host disease (GVHD). Repopulated human T cells, B cells, natural killer (NK) cells, dendritic cells (DC), and myeloid-derived suppressor cells (MDSC) increased in peripheral blood, spleen, and bone marrow over time. Although cultured CD34+ HSCs labeled with luciferase could be detected in mice, the cultured HSCs did not develop into mature human immune cells by four weeks, unlike fresh CD34+ HSCs. Ex vivo, reconstituted T cells, obtained from the tumor-bearing humanized mice, secreted IFNγ upon treatment with phorbol myristate acetate (PMA) or exposure to human A549 lung tumor cells and mediated antigen-specific CTL responses, indicating functional activity. Growth of engrafted PDXs and tumor xenografts was not dependent on the human leukocyte antigen status of the donor. Treatment with the anti-PD-1 checkpoint inhibitors pembrolizumab or nivolumab inhibited tumor growth in humanized mice significantly, and correlated with an increased number of CTLs and decreased MDSCs, regardless of the donor HLA type. In conclusion, fresh CD34+HSCs are more effective than their expanded counterparts in humanizing mice, and do so in a shorter time. The Hu-PDX model provides an improved platform for evaluation of immunotherapy.


Assuntos
Imunidade , Neoplasias Pulmonares/imunologia , Neoplasias Pulmonares/patologia , Animais , Antígenos de Neoplasias/imunologia , Biomarcadores Tumorais , Modelos Animais de Doenças , Feminino , Citometria de Fluxo , Antígenos HLA/genética , Antígenos HLA/imunologia , Células-Tronco Hematopoéticas , Humanos , Neoplasias Pulmonares/metabolismo , Masculino , Camundongos , Linfócitos T/imunologia , Linfócitos T/metabolismo , Microambiente Tumoral , Ensaios Antitumorais Modelo de Xenoenxerto
6.
Cancer Med ; 7(6): 2405-2414, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29673125

RESUMO

In a previous study, we determined that major pathologic response (MPR) as indicated by the percentage of residual viable tumor cells predicted overall survival (OS) in patients with non-small-cell lung cancer (NSCLC) who received neoadjuvant chemotherapy. In this study, we assessed whether two genes and five protein biomarkers could predict MPR and OS in 98 patients with NSCLC receiving neoadjuvant chemotherapy. We collected formalin-fixed, paraffin-embedded specimens of resected NSCLC tumors from 98 patients treated with neoadjuvant chemotherapy. We identified mutations in KRAS and EGFR genes using pyrosequencing and examined the expression of protein markers VEGFR2, EZH2, ERCC1, RAD51, and PKR using immunohistochemistry. We assessed whether gene mutation status or protein expression was associated with MPR or OS. We observed that KRAS mutation tended to be associated with OS (P = .06), but EGFR mutation was not associated with OS. We found that patients with high RAD51 expression levels had a poorer prognosis than did those with low RAD51 expression. We also observed that RAD51 expression was associated with MPR. MPR and RAD51 expression were associated with OS in univariate and multivariate analyses (P = .04 and P = .02, respectively). Combination of MPR with RAD51 is a significant predictor of prognosis in patients with NSCLC who received neoadjuvant chemotherapy. We demonstrated that MPR or RAD51 expression was associated with OS in patients with NSCLC receiving neoadjuvant chemotherapy. Prediction of a patient's prognosis could be improved by combined assessment of MPR and RAD51 expression.


Assuntos
Neoplasias Pulmonares/tratamento farmacológico , Terapia Neoadjuvante/métodos , Rad51 Recombinase/genética , Adulto , Idoso , Idoso de 80 Anos ou mais , Feminino , Humanos , Neoplasias Pulmonares/patologia , Masculino , Pessoa de Meia-Idade , Taxa de Sobrevida
8.
Sci Rep ; 7(1): 9029, 2017 08 22.
Artigo em Inglês | MEDLINE | ID: mdl-28831115

RESUMO

Through a new hypothesis-driven and microRNA-pathway-based SNP (miR-SNP) association study we identified a novel miR-SNP (rs713065) in the 3'UTR region of FZD4 gene linked with decreased risk of death in early stage NSCLC patients. We determined biological function and mechanism of action of this FZD4-miR-SNP biomarker in a cellular platform. Our data suggest that FZD4-miR-SNP loci may significantly influence overall survival in NSCLC patients by specifically interacting with miR-204 and modulating FZD4 expression and cellular function in the Wnt-signaling-driven tumor progression. Our findings are bridging the gap between the discovery of epidemiological SNP biomarkers and their biological function and will enable us to develop novel therapeutic strategies that specifically target epigenetic markers in the oncogenic Wnt/FZD signaling pathways in NSCLC.


Assuntos
Biomarcadores Tumorais/genética , Regulação para Baixo , Receptores Frizzled/genética , Neoplasias Pulmonares/genética , MicroRNAs/genética , Polimorfismo de Nucleotídeo Único , Regiões 3' não Traduzidas , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Regulação Neoplásica da Expressão Gênica , Redes Reguladoras de Genes , Humanos , Neoplasias Pulmonares/patologia , Estadiamento de Neoplasias , Prognóstico , Análise de Sobrevida , Via de Sinalização Wnt
9.
Oncotarget ; 8(29): 48222-48239, 2017 Jul 18.
Artigo em Inglês | MEDLINE | ID: mdl-28637023

RESUMO

RNA interference (RNAi)-based therapeutics have been used to silence the expression of targeted pathological genes. Small interfering RNA (siRNAs) and microRNA (miRNAs) inhibitor have performed this function. However, short half-life, poor cellular uptake, and nonspecific distribution of small RNAs call for the development of novel delivery systems to facilitate the use of RNAi. We developed a novel cationic liquid crystalline nanoparticle (CLCN) to efficiently deliver synthetic siRNAs and miRNAs. CLCNs were prepared by using high-speed homogenization and assembled with synthetic siRNA or miRNA molecules in nuclease-free water to create CLCN/siRNA or miRNA complexes. The homogeneous and stable CLCNs and CLCN-siRNA complexes were about 100 nm in diameter, with positively charged surfaces. CLCNs are nontoxic and are taken up by human cells though endocytosis. Significant inhibition of gene expression was detected in transiently transfected lung cancer H1299 cells treated with CLCNs/anti-GFP complexes 24 hours after transfection. Biodistribution analysis showed that the CLCNs and CLCNs-RNAi complexes were successfully delivered to various organs and into the subcutaneous human lung cancer H1299 tumor xenografts in mice 24 hours after systemic administration. These results suggest that CLCNs are a unique and advanced delivery system capable of protecting RNAi from degradation and of efficiently delivering RNAi in vitro and in vivo.


Assuntos
Cristalinas , Técnicas de Transferência de Genes , Nanopartículas , RNA Interferente Pequeno/administração & dosagem , Terapêutica com RNAi , Animais , Linhagem Celular Tumoral , Canais de Cloreto/química , Portadores de Fármacos , Sistemas de Liberação de Medicamentos , Citometria de Fluxo , Inativação Gênica , Humanos , Camundongos , MicroRNAs/administração & dosagem , MicroRNAs/química , MicroRNAs/genética , Microscopia de Fluorescência , Modelos Animais , Nanopartículas/química , Nanopartículas/ultraestrutura , Interferência de RNA , RNA Interferente Pequeno/química , RNA Interferente Pequeno/genética , Terapêutica com RNAi/efeitos adversos , Terapêutica com RNAi/métodos , Distribuição Tecidual , Ensaios Antitumorais Modelo de Xenoenxerto
10.
Mol Cancer Res ; 3(4): 219-26, 2005 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15831675

RESUMO

Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) has been shown to induce apoptosis of cancer cells. Sensitization of cancer cells to TRAIL, particularly TRAIL-resistant cancer cells, could improve the effectiveness of TRAIL as an anticancer agent. The adenovirus type 5 E1A that associates with anticancer activities including sensitization to apoptosis induced by tumor necrosis factor is currently being tested in clinical trials. In this study, we investigated the sensitivity to TRAIL in the E1A transfectants ip1-E1A2 and 231-E1A cells and the parental TRAIL-resistant human ovarian cancer SKOV3.ip1 and TRAIL-sensitive human breast cancer MDA-MB-231 cells. The results indicated that the percentage of TRAIL-induced apoptotic cells was significantly higher in the E1A transfectants of both cell lines than it was in the parental cell lines. To further investigate the cellular mechanism of this effect, we found that E1A enhances TRAIL-induced activation of caspase-8, caspase-9, and caspase-3. Inhibition of caspase-3 activity by a specific inhibitor, Z-DEVD-fmk, abolished TRAIL-induced apoptosis. In addition, E1A enhanced TRAIL expression in ip1-E1A2 cells, but not in 231-E1A cells, and the anti-TRAIL neutralizing antibody N2B2 blocked the E1A-mediated bystander effect in vitro. Taken together, these results suggest that E1A sensitizes both TRAIL-sensitive and TRAIL-resistant cancer cells to TRAIL-induced apoptosis, which occurs through the enhancement of caspase activation; activation of caspase-3 is required for TRAIL-induced apoptosis; and E1A-induced TRAIL expression is involved in the E1A-mediated bystander effect. Combination of E1A and TRAIL could be an effective treatment for cancer.


Assuntos
Proteínas E1A de Adenovirus/genética , Apoptose/fisiologia , Caspases/metabolismo , Glicoproteínas de Membrana/farmacologia , Neoplasias Ovarianas/terapia , Fator de Necrose Tumoral alfa/farmacologia , Proteínas E1A de Adenovirus/metabolismo , Anticorpos/farmacologia , Proteínas Reguladoras de Apoptose , Neoplasias da Mama/terapia , Efeito Espectador , Caspase 3 , Inibidores de Caspase , Linhagem Celular Tumoral , Inibidores de Cisteína Proteinase/farmacologia , Feminino , Terapia Genética/métodos , Humanos , Glicoproteínas de Membrana/imunologia , Glicoproteínas de Membrana/metabolismo , Oligopeptídeos/farmacologia , Proteínas Recombinantes/imunologia , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/farmacologia , Ligante Indutor de Apoptose Relacionado a TNF , Transfecção , Fator de Necrose Tumoral alfa/imunologia , Fator de Necrose Tumoral alfa/metabolismo
11.
Cancer Res ; 62(5): 1311-4, 2002 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-11888897

RESUMO

The breast cancer susceptibility gene BRCA2 has been suggested to function as a "caretaker" of the genome. Cells without wild-type BRCA2 are deficient in repairing DNA damage. However, whether BRCA2 can also suppress oncogenesis by regulating cell proliferation remains to be determined. To address this question, the expression of wild-type BRCA2 protein was reconstituted, in an either constitutive or regulated manner, in the pancreatic cancer cell line Capan-1, which expresses only a mutant BRCA2. Expression of wild-type BRCA2 inhibited cell proliferation in culture and suppressed tumor growth in animals. Our results showed that, in addition to the DNA repair function, BRCA2 also suppresses tumor development by inhibiting cancer cell growth.


Assuntos
Genes BRCA2/fisiologia , Neoplasias Pancreáticas/prevenção & controle , Animais , Divisão Celular , Feminino , Camundongos , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patologia , Transfecção , Células Tumorais Cultivadas
12.
Cancer Res ; 64(5): 1788-93, 2004 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-14996741

RESUMO

Gene therapy provides a novel treatment approach to cancer patients. Ideally, expression of therapeutic genes driven by cancer-specific promoters would only target tumors resulting in minimal toxicity to normal tissues. While there is a need of more effective and tolerable treatments for ovarian cancer patients, we aimed to identify gene promoters with high activity in ovarian tumors that can be potentially used in gene therapy to drive the expression of a therapeutic gene in tumors. To identify such promoters, a literature search was performed to reveal genes that are preferentially expressed in ovarian cancer compared with normal ovarian tissue. We found that the ceruloplasmin promoter drove up to 30-fold higher luciferase expression in ovarian cancer cells compared with immortalized normal cells. Furthermore, deletion studies revealed an activator protein-1 (AP-1) site in the ceruloplasmin promoter to be critical for optimal ceruloplasmin promoter activity. Ceruloplasmin promoter activity was significantly activated by 1-O-tetradecanoyl phorbol-13-acetate, a c-jun activator, and conversely suppressed by SP600125, a c-jun inhibitor. Consistently, the ceruloplasmin AP-1 site was specifically recognized by c-jun both in vitro and in vivo. Immunohistochemical analyses of human ovarian cancer specimens showed a direct correlation (r = 0.7, P = 0.007) between expression levels of c-jun and ceruloplasmin. In nude mice carrying SKOV3.ip1 xenografts, the ceruloplasmin promoter demonstrated significantly higher activities in tumors compared with normal organs. Together, these results suggest that the ceruloplasmin promoter activity is significantly enhanced in ovarian cancer and therefore may be exploited as a promising cancer-specific promoter in developing new gene therapy strategies for ovarian cancer.


Assuntos
Ceruloplasmina/genética , Terapia Genética , Neoplasias Ovarianas/terapia , Regiões Promotoras Genéticas , Linhagem Celular Tumoral , Feminino , Genes jun , Humanos , Neoplasias Ovarianas/genética , Transcrição Gênica
13.
Oncotarget ; 7(25): 38235-38242, 2016 Jun 21.
Artigo em Inglês | MEDLINE | ID: mdl-27203671

RESUMO

We have previously demonstrated that radiation induced cell death in PKR (-/-) deficient mouse embryo fibroblasts (MEFs) but not in PKR (+/+) wild type MEFs. Our study indicated that PKR can also be involved in survival pathways following radiation therapy through activation of the AKT survival pathways in these MEFs is mediated in part through PKR. The role of PKR on radiation sensitivity in cancer cells has not been evaluated. In this study, we demonstrated that radiation treatment causes nuclear translocation of PKR in human lung cancer cells. The transduction of lung cancer cells with a dominant negative adenoviral PKR vector blocks nuclear translocation of PKR and leads to the reversal of radiation resistance. Plasmid transduction of lung cancer cells with nuclear targeted wild type PKR vectors also increased radiation resistance. This effect is selectively abrogated by plasmid transduction of dominant negative PKR vectors which restore radiation sensitivity. These findings suggest a novel role for PKR in lung cancer cells as a mediator of radiation resistance possibly through translocation of the protein product to the nucleus.


Assuntos
Neoplasias Pulmonares/enzimologia , Neoplasias Pulmonares/radioterapia , eIF-2 Quinase/metabolismo , Células A549 , Adenoviridae/genética , Linhagem Celular Tumoral , Núcleo Celular/enzimologia , Vetores Genéticos/genética , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patologia , Tolerância a Radiação , Transfecção , eIF-2 Quinase/biossíntese , eIF-2 Quinase/genética
14.
Sci Rep ; 6: 35741, 2016 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-27845352

RESUMO

Expression of the TUSC2/FUS1 tumor suppressor gene in TUSC2 deficient EGFR wildtype lung cancer cells increased sensitivity to erlotinib. Microarray mRNA expression analysis of TUSC2 inducible lung cancer cells treated with erlotinib uncovered defects in the response to oxidative stress suggesting that increasing reactive oxygen species (ROS) would enhance therapeutic efficacy. Addition of the thioredoxin reductase 1 inhibitor (TXNRD1) auranofin (AF) to NSCLC cells treated with combination of TUSC2 forced expression with erlotinib increased tumor cell apoptosis and inhibited colony formation. TXNRD1 overexpression rescued tumors from AF-TUSC2-erlotinib induced apoptosis. Neutralizing ROS with nordihydroguaiaretic acid (NDGA) abrogated cell death induced by AF-TUSC2-erlotinib, indicating a regulatory role for ROS in the efficacy of the three drug combination. Isobologram-based statistical analysis of this combination demonstrated superior synergism, compared with each individual treatment at lower concentrations. In NSCLC tumor xenografts, tumor growth was markedly inhibited and animal survival was prolonged over controls by AF-TUSC2-erlotinib. Microarray mRNA expression analysis uncovered oxidative stress and DNA damage gene signatures significantly upregulated by AF-TUSC2-erlotinib compared to TUSC2-erlotinib. Pathway analysis showed the highest positive z-score for the NRF2-mediated oxidative stress response. Taken together these findings show that the combination of TUSC2-erlotinib induces additional novel vulnerabilities that can be targeted with AF.


Assuntos
Auranofina/farmacologia , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Receptores ErbB/biossíntese , Cloridrato de Erlotinib/farmacologia , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Neoplasias Pulmonares/metabolismo , Proteínas Supressoras de Tumor/biossíntese , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Carcinoma Pulmonar de Células não Pequenas/genética , Carcinoma Pulmonar de Células não Pequenas/patologia , Linhagem Celular , Receptores ErbB/genética , Humanos , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patologia , Proteínas Supressoras de Tumor/genética
15.
Oncotarget ; 7(3): 3548-58, 2016 Jan 19.
Artigo em Inglês | MEDLINE | ID: mdl-26657290

RESUMO

Auranofin, a gold complex that has been used to treat rheumatoid arthritis in clinics and has documented pharmacokinetic and safety profiles in humans, has recently been investigated for its anticancer activity in leukemia and some solid cancers. However, auranofin's single agent activity in lung cancer is not well characterized. To determine whether auranofin has single agent activity in lung cancer, we evaluated auranofin's activity in a panel of 10 non-small cell lung cancer (NSCLC) cell lines. Cell viability analysis revealed that auranofin induced growth inhibition in a subset of NSCLC cell lines with a half maximal inhibitory concentration (IC50) below 1.0 µM. Treatment with auranofin elicited apoptosis and necroptosis in auranofin-sensitive cell lines. Moreover, the susceptibility of NSCLC cells to auranofin was inversely correlated with TXNRD1 expression in the cells. Transient transfection of the TXNRD1-expressing plasmid in auranofin-sensitive Calu3 cells resulted in partial resistance, indicating that high TXNRD level is one of causal factors for resistance to auranofin. Further mechanistic characterization with proteomic analysis revealed that auranofin inhibits expression and/or phosphorylation of multiple key nodes in the PI3K/AKT/mTOR pathway, including S6, 4EBP1, Rictor, p70S6K, mTOR, TSC2, AKT and GSK3. Ectopic expression of TXNRD1 partially reversed auranofin-mediated PI3K/AKT/mTOR inhibition, suggesting that TXNRD1 may participate in the regulation of PI3K/AKT/mTOR pathway. Administration of auranofin to mice with xenograft tumors derived from NSCLC cells significantly suppressed tumor growth without inducing obvious toxic effects. Our results demonstrated feasibility of repurposing auranofin for treatment of lung cancer.


Assuntos
Auranofina/farmacologia , Carcinoma Pulmonar de Células não Pequenas/prevenção & controle , Neoplasias Pulmonares/prevenção & controle , Fosfatidilinositol 3-Quinases/química , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , Serina-Treonina Quinases TOR/antagonistas & inibidores , Animais , Antirreumáticos/farmacologia , Apoptose/efeitos dos fármacos , Western Blotting , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Carcinoma Pulmonar de Células não Pequenas/patologia , Proliferação de Células/efeitos dos fármacos , Citometria de Fluxo , Humanos , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patologia , Camundongos , Fosfatidilinositol 3-Quinases/metabolismo , Análise Serial de Proteínas , Proteínas Proto-Oncogênicas c-akt/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Tiorredoxina Redutase 1/metabolismo , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
16.
Oncogene ; 23(26): 4556-66, 2004 Jun 03.
Artigo em Inglês | MEDLINE | ID: mdl-15122330

RESUMO

We identified IFIX as a new member of the hematopoietic interferon (IFN)-inducible nuclear protein with the 200-amino-acid repeat (HIN-200) family. Six different alternatively spliced forms of mRNA are transcribed from the IFIX gene, which are predicted to encode six different isoforms of IFIX proteins (IFIXalpha1, alpha2, beta1, beta2, gamma1, and gamma2). The IFIX proteins are primarily localized in the nucleus. They share a common N-terminal region that contains a predicted pyrin domain and a putative nuclear localization signal. Unlike IFIXalpha and IFIXbeta, IFIXgamma isoforms do not have the 200-amino-acid signature motif. Interestingly, the expression of IFIX was reduced in most human breast tumors and breast cancer cell lines. Expression of IFIXalpha1, the longest isoform of IFIX, in human breast cancer cell lines reduced their anchorage-dependent and -independent growth in vitro and tumorigenicity in nude mice. Moreover, a liposome-mediated IFIXalpha1 gene transfer suppressed the growth of already-formed tumors in a breast cancer xenograft model. IFIXalpha1 appears to suppress the growth of breast cancer cells in a pRB- and p53-independent manner by increasing the expression of the cyclin-dependent kinase inhibitor p21(CIP1), which leads to the reduction of the kinase activity of both Cdk2 and p34(Cdc2). Together, our results show that IFIXalpha1 possesses a tumor-suppressor activity and suggest IFIXalpha1 may be used as a therapeutic agent in cancer treatment.


Assuntos
Neoplasias da Mama/genética , Terapia Genética/métodos , Proteínas Nucleares/genética , Processamento Alternativo , Sequência de Aminoácidos , Animais , Antineoplásicos/farmacologia , Neoplasias da Mama/terapia , Proteína Quinase CDC2/metabolismo , Quinases relacionadas a CDC2 e CDC28/metabolismo , Testes de Carcinogenicidade , Quinase 2 Dependente de Ciclina , Inibidor de Quinase Dependente de Ciclina p21 , Ciclinas/metabolismo , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Camundongos , Camundongos Nus , Dados de Sequência Molecular , Proteínas Nucleares/metabolismo , Proteínas Nucleares/farmacologia , Isoformas de Proteínas , Proteína do Retinoblastoma/metabolismo , Células Tumorais Cultivadas , Proteína Supressora de Tumor p53/metabolismo
17.
Clin Cancer Res ; 8(10): 3290-7, 2002 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-12374701

RESUMO

PURPOSE AND EXPERIMENTAL DESIGN: p202, a mouse IFN-inducible protein, is a member of the 200-amino acid repeat family. Enforced p202 expression in stable cancer cell lines resulted in growth inhibition in vitro and tumor suppression in vivo. However, to study the immediate effect of p202 and test the potential efficacy of p202 treatment, an efficient gene delivery system for p202 is required. For these purposes, an adenoviral vector expressing the p202 gene (Ad-p202) was generated. We examined the effects of Ad-p202 infection on human breast cancer cells. Furthermore, we tested the efficacy of Ad-p202 treatment on breast and pancreatic cancer xenograft models. RESULTS: We found that Ad-p202 infection induces growth inhibition and sensitizes the otherwise resistant cells to tumor necrosis factor alpha-induced apoptosis. In addition, we demonstrated for the first time that Ad-p202 infection induces apoptosis and that activation of caspases is required for the full apoptotic effect. More importantly, we showed the efficacy of Ad-p202 treatment on breast cancer xenograft models, and this antitumor effect correlated well with enhanced apoptosis in Ad-p202-treated tumors. CONCLUSIONS: We conclude that Ad-p202 is a potent growth-inhibitory, proapoptotic, and tumor-suppressing agent. Ad-p202 may be further developed into an efficient therapeutic agent for human cancer gene therapy.


Assuntos
Apoptose , Neoplasias da Mama/terapia , Proteínas de Transporte/genética , Terapia Genética/métodos , Peptídeos e Proteínas de Sinalização Intracelular , Proteínas Nucleares/genética , Neoplasias Pancreáticas/terapia , Fosfoproteínas/genética , Adenoviridae/genética , Animais , Antineoplásicos/farmacologia , Neoplasias da Mama/patologia , Neoplasias da Mama/prevenção & controle , Caspases/metabolismo , Ensaio de Desvio de Mobilidade Eletroforética , Feminino , Técnicas de Transferência de Genes , Vetores Genéticos , Humanos , Técnicas In Vitro , Camundongos , Camundongos Nus , NF-kappa B/metabolismo , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patologia , Transfecção , Células Tumorais Cultivadas , Fator de Necrose Tumoral alfa/farmacologia , Proteína 1 de Ligação à Proteína Supressora de Tumor p53 , Ensaios Antitumorais Modelo de Xenoenxerto
18.
Oncotarget ; 6(13): 11114-24, 2015 May 10.
Artigo em Inglês | MEDLINE | ID: mdl-25798539

RESUMO

We have demonstrated that RNA-dependent protein kinase (PKR) and its downstream protein p-eIF2α are independent prognostic markers for overall survival in lung cancer. In the current study, we further investigate the interaction between PKR and AMPK in lung tumor tissue and cancer cell lines. We examined PKR protein expression in 55 frozen primary lung tumor tissues by Western blotting and analyzed the association between PKR expression and expression of 139 proteins on tissue samples examined previously by Reverse Phase Protein Array (RPPA) from the same 55 patients. We observed that biomarkers were either positively (phosphorylated AMP-activated kinase(T172) [p-AMPK]) or negatively (insulin receptor substrate 1, meiotic recombination 11, ATR interacting protein, telomerase, checkpoint kinase 1, and cyclin E1) correlated with PKR. We further confirmed that induction of PKR with expression vectors in lung cancer cells causes activation of the AMPK protein independent of the LKB1, TAK1, and CaMKKß pathway. We found that PKR causes nutrient depletion, which increases AMP levels and decreases ATP levels, causing AMPK phosphorylation. We further demonstrated that inhibiting AMPK expression with compound C or siRNA enhanced PKR-mediated cell death. We next explored the combination of PKR and p-AMPK expression in NSCLC patients and observed that expression of p-AMPK predicted a poor outcome for adenocarcinoma patients with high PKR expression and a better prognosis for those with low PKR expression. These findings were consistent with our in vitro results. AMPK might rescue cells facing metabolic stresses, such as ATP depletion caused by PKR. Our data indicate that PKR causes nutrient depletion, which induces the phosphorylation of AMPK. AMPK might act as a protective response to metabolic stresses, such as nutrient deprivation.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Adenocarcinoma/metabolismo , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Carcinoma de Células Escamosas/metabolismo , Glucose/deficiência , Neoplasias Pulmonares/metabolismo , eIF-2 Quinase/metabolismo , Proteínas Quinases Ativadas por AMP/antagonistas & inibidores , Adenocarcinoma/mortalidade , Adenocarcinoma/patologia , Monofosfato de Adenosina/metabolismo , Trifosfato de Adenosina/metabolismo , Western Blotting , Carcinoma Pulmonar de Células não Pequenas/mortalidade , Carcinoma Pulmonar de Células não Pequenas/patologia , Carcinoma de Células Escamosas/mortalidade , Carcinoma de Células Escamosas/patologia , Citometria de Fluxo , Humanos , Técnicas Imunoenzimáticas , Lactatos/metabolismo , Neoplasias Pulmonares/mortalidade , Neoplasias Pulmonares/patologia , Estadiamento de Neoplasias , Prognóstico , Taxa de Sobrevida , Células Tumorais Cultivadas
19.
J Thorac Oncol ; 8(3): 301-8, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23370317

RESUMO

INTRODUCTION: RNA-dependent protein kinase (PKR) is an independent prognostic variable in patients with non-small-cell lung cancer (NSCLC). In the current study, we investigated the correlation between PKR and 25 other biomarkers for NSCLC, identified the markers that could further improve the prognostic significance of PKR and elucidated the mechanisms of interaction between these markers and PKR. METHODS: Tissue microarray samples obtained from 218 patients with lung cancer were stained with an anti-PKR antibody and antibodies against 25 biomarkers. Immunohistochemical expression was scored and used for Kaplan-Meier survival analysis. The interaction between PKR and EphA2 in NSCLC cell lines was examined. RESULTS: We found that PKR was associated with EphA2 and that the prognostic information regarding NSCLC provided by the combination of PKR and EphA2 (P/E) was significantly more accurate than that provided by either marker alone. The 5-year overall survival rate in patients with PKR/EphA2 (20%) was significantly lower than that of patients with PKR/EphA2 (74%), patients with PKR/EphA2 (55%), and patients with PKR/EphA2 (55%) (p < 0.0001). We also found that the PKR:EphA2 (P/E) ratio was significantly associated with prognosis (p < 0.0001). Univariate and multivariate Cox analyses revealed that this P/E combination or ratio was an independent predictor of overall survival. In addition, induction of PKR expression reduced EphA2 protein expression levels in NSCLC cell lines. CONCLUSIONS: PKR/EphA2 is a significant predictor of prognosis for NSCLC. PKR/EphA2 may be a promising approach to improving screening efficiency and predicting prognosis in patients with NSCLC.


Assuntos
Adenocarcinoma/mortalidade , Biomarcadores Tumorais/metabolismo , Carcinoma Pulmonar de Células não Pequenas/mortalidade , Carcinoma de Células Escamosas/mortalidade , Neoplasias Pulmonares/mortalidade , Receptor EphA2/metabolismo , eIF-2 Quinase/metabolismo , Adenocarcinoma/metabolismo , Adenocarcinoma/patologia , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Carcinoma Pulmonar de Células não Pequenas/patologia , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/patologia , Feminino , Seguimentos , Humanos , Técnicas Imunoenzimáticas , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patologia , Masculino , Estadiamento de Neoplasias , Prognóstico , RNA Interferente Pequeno/genética , Receptor EphA2/antagonistas & inibidores , Receptor EphA2/genética , Taxa de Sobrevida , Análise Serial de Tecidos , Células Tumorais Cultivadas
20.
Mol Imaging Biol ; 11(6): 424-33, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19424760

RESUMO

PURPOSE: The purpose of the study is to synthesize and characterize near-infrared (NIR) fluorescence imaging probes targeted to gelatinases. PROCEDURES: A phage display-selected cyclic peptide containing the His-Try-Gly-Phe (HWGF) motif was used as the lead compound. Structure-activity relationship analysis was used to identify stable and potent gelatinase inhibitors suitable for NIR imaging applications. RESULTS: Replacing the S-S bond in cyclic peptide c(CTTHWGFTLC)NH(2) (C1) with an amide bond between the epsilon-amino group of Lys and the side chain of Asp resulted in a significant increase in stability and a fourfold increase in gelatinase inhibition of the resulting peptide, c(KAHWGFTLD)NH(2) (C6). Conjugation of Cy5.5 to C6 led to Cy5.5-C6, which was selectively taken up by MMP-2 expressing human glioma U87 cells. In vivo, selective accumulation of Cy5.5-C6, but not Cy5.5-C1 or a Cy5.5-scrambled peptide conjugate, was visualized in intratibial prostate PC-3 tumors 48 h after their intravenous injection. Moreover, Cy5.5-C6 was readily visualized in orthotopically inoculated U87 brain tumors. CONCLUSIONS: Cy5.5-C6 may be a useful agent for molecular imaging of gelatinases. The approach of producing stable cyclic peptides through side chain amide linkage should be applicable to other peptide-based imaging agents.


Assuntos
Gelatinases/química , Peptídeos Cíclicos/química , Sequência de Aminoácidos , Carbocianinas/metabolismo , Linhagem Celular Tumoral , Fenômenos Químicos , Corantes Fluorescentes/química , Gelatinases/análise , Gelatinases/metabolismo , Glioma/patologia , Humanos , Neoplasias Pulmonares/patologia , Masculino , Estrutura Molecular , Peso Molecular , Peptídeos Cíclicos/isolamento & purificação , Neoplasias da Próstata/patologia , Ligação Proteica , Estabilidade Proteica , Espectroscopia de Luz Próxima ao Infravermelho , Relação Estrutura-Atividade
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA