Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
2.
Commun Biol ; 6(1): 509, 2023 05 11.
Article in English | MEDLINE | ID: mdl-37169941

ABSTRACT

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.


Subject(s)
Lung Neoplasms , Mice , Animals , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Lung Neoplasms/pathology , ErbB Receptors/genetics , Phosphatidylinositol 3-Kinases , Proto-Oncogene Proteins c-akt/genetics , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Drug Resistance, Neoplasm/genetics , Mutation , TOR Serine-Threonine Kinases/genetics , Phosphatidylinositols
3.
Commun Biol ; 5(1): 167, 2022 02 24.
Article in English | MEDLINE | ID: mdl-35210547

ABSTRACT

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.


Subject(s)
AMP-Activated Protein Kinases , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Proto-Oncogene Proteins p21(ras) , Tumor Suppressor Proteins , AMP-Activated Protein Kinases/genetics , AMP-Activated Protein Kinases/immunology , Animals , Antibodies, Monoclonal, Humanized/administration & dosage , Carboplatin/administration & dosage , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/therapy , Disease Models, Animal , Genes, Tumor Suppressor , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Lung Neoplasms/therapy , Mice , Proto-Oncogene Proteins p21(ras)/genetics , Proto-Oncogene Proteins p21(ras)/immunology , Tumor Microenvironment , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/immunology
4.
Oncogene ; 39(4): 801-813, 2020 01.
Article in English | MEDLINE | ID: mdl-31554935

ABSTRACT

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.


Subject(s)
Antineoplastic Agents/pharmacology , Lysosomes/metabolism , Neoplasms/drug therapy , Phosphotransferases (Alcohol Group Acceptor)/antagonists & inhibitors , Proteolysis , Unfolded Protein Response , eIF-2 Kinase/antagonists & inhibitors , Animals , Antineoplastic Agents/chemistry , Cell Line, Tumor , Cell Survival , Databases, Genetic/statistics & numerical data , Exosomes/metabolism , Female , Humans , Mice , Mice, SCID , Minor Histocompatibility Antigens/metabolism , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/pathology , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Prognosis , Protein Folding , Survival Rate , Xenograft Model Antitumor Assays , eIF-2 Kinase/metabolism
5.
Cancer Immunol Res ; 7(8): 1267-1279, 2019 08.
Article in English | MEDLINE | ID: mdl-31186248

ABSTRACT

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.


Subject(s)
Immunity , Lung Neoplasms/immunology , Lung Neoplasms/pathology , Animals , Antigens, Neoplasm/immunology , Biomarkers, Tumor , Disease Models, Animal , Female , Flow Cytometry , HLA Antigens/genetics , HLA Antigens/immunology , Hematopoietic Stem Cells , Humans , Lung Neoplasms/metabolism , Male , Mice , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Tumor Microenvironment , Xenograft Model Antitumor Assays
6.
Cancer Med ; 7(6): 2405-2414, 2018 06.
Article in English | MEDLINE | ID: mdl-29673125

ABSTRACT

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.


Subject(s)
Lung Neoplasms/drug therapy , Neoadjuvant Therapy/methods , Rad51 Recombinase/genetics , Adult , Aged , Aged, 80 and over , Female , Humans , Lung Neoplasms/pathology , Male , Middle Aged , Survival Rate
7.
Sci Rep ; 7(1): 9029, 2017 08 22.
Article in English | MEDLINE | ID: mdl-28831115

ABSTRACT

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.


Subject(s)
Biomarkers, Tumor/genetics , Down-Regulation , Frizzled Receptors/genetics , Lung Neoplasms/genetics , MicroRNAs/genetics , Polymorphism, Single Nucleotide , 3' Untranslated Regions , Cell Line, Tumor , Cell Movement , Cell Proliferation , Gene Expression Regulation, Neoplastic , Gene Regulatory Networks , Humans , Lung Neoplasms/pathology , Neoplasm Staging , Prognosis , Survival Analysis , Wnt Signaling Pathway
8.
Oncotarget ; 8(29): 48222-48239, 2017 Jul 18.
Article in English | MEDLINE | ID: mdl-28637023

ABSTRACT

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.


Subject(s)
Crystallins , Gene Transfer Techniques , Nanoparticles , RNA, Small Interfering/administration & dosage , RNAi Therapeutics , Animals , Cell Line, Tumor , Chloride Channels/chemistry , Drug Carriers , Drug Delivery Systems , Flow Cytometry , Gene Silencing , Humans , Mice , MicroRNAs/administration & dosage , MicroRNAs/chemistry , MicroRNAs/genetics , Microscopy, Fluorescence , Models, Animal , Nanoparticles/chemistry , Nanoparticles/ultrastructure , RNA Interference , RNA, Small Interfering/chemistry , RNA, Small Interfering/genetics , RNAi Therapeutics/adverse effects , RNAi Therapeutics/methods , Tissue Distribution , Xenograft Model Antitumor Assays
9.
Sci Rep ; 6: 35741, 2016 11 15.
Article in English | MEDLINE | ID: mdl-27845352

ABSTRACT

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.


Subject(s)
Auranofin/pharmacology , Carcinoma, Non-Small-Cell Lung/metabolism , ErbB Receptors/biosynthesis , Erlotinib Hydrochloride/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Lung Neoplasms/metabolism , Tumor Suppressor Proteins/biosynthesis , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line , ErbB Receptors/genetics , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Tumor Suppressor Proteins/genetics
10.
Oncotarget ; 7(25): 38235-38242, 2016 Jun 21.
Article in English | MEDLINE | ID: mdl-27203671

ABSTRACT

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.


Subject(s)
Lung Neoplasms/enzymology , Lung Neoplasms/radiotherapy , eIF-2 Kinase/metabolism , A549 Cells , Adenoviridae/genetics , Cell Line, Tumor , Cell Nucleus/enzymology , Genetic Vectors/genetics , Humans , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Radiation Tolerance , Transfection , eIF-2 Kinase/biosynthesis , eIF-2 Kinase/genetics
11.
Oncotarget ; 7(3): 3548-58, 2016 Jan 19.
Article in English | MEDLINE | ID: mdl-26657290

ABSTRACT

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.


Subject(s)
Auranofin/pharmacology , Carcinoma, Non-Small-Cell Lung/prevention & control , Lung Neoplasms/prevention & control , Phosphatidylinositol 3-Kinases/chemistry , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , TOR Serine-Threonine Kinases/antagonists & inhibitors , Animals , Antirheumatic Agents/pharmacology , Apoptosis/drug effects , Blotting, Western , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Cell Proliferation/drug effects , Flow Cytometry , Humans , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Mice , Phosphatidylinositol 3-Kinases/metabolism , Protein Array Analysis , Proto-Oncogene Proteins c-akt/metabolism , TOR Serine-Threonine Kinases/metabolism , Thioredoxin Reductase 1/metabolism , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
12.
Oncotarget ; 6(13): 11114-24, 2015 May 10.
Article in English | MEDLINE | ID: mdl-25798539

ABSTRACT

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.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Adenocarcinoma/metabolism , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Squamous Cell/metabolism , Glucose/deficiency , Lung Neoplasms/metabolism , eIF-2 Kinase/metabolism , AMP-Activated Protein Kinases/antagonists & inhibitors , Adenocarcinoma/mortality , Adenocarcinoma/pathology , Adenosine Monophosphate/metabolism , Adenosine Triphosphate/metabolism , Blotting, Western , Carcinoma, Non-Small-Cell Lung/mortality , Carcinoma, Non-Small-Cell Lung/pathology , Carcinoma, Squamous Cell/mortality , Carcinoma, Squamous Cell/pathology , Flow Cytometry , Humans , Immunoenzyme Techniques , Lactates/metabolism , Lung Neoplasms/mortality , Lung Neoplasms/pathology , Neoplasm Staging , Prognosis , Survival Rate , Tumor Cells, Cultured
13.
J Thorac Oncol ; 8(3): 301-8, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23370317

ABSTRACT

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.


Subject(s)
Adenocarcinoma/mortality , Biomarkers, Tumor/metabolism , Carcinoma, Non-Small-Cell Lung/mortality , Carcinoma, Squamous Cell/mortality , Lung Neoplasms/mortality , Receptor, EphA2/metabolism , eIF-2 Kinase/metabolism , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Female , Follow-Up Studies , Humans , Immunoenzyme Techniques , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Male , Neoplasm Staging , Prognosis , RNA, Small Interfering/genetics , Receptor, EphA2/antagonists & inhibitors , Receptor, EphA2/genetics , Survival Rate , Tissue Array Analysis , Tumor Cells, Cultured
14.
Small ; 6(9): 1022-31, 2010 May 07.
Article in English | MEDLINE | ID: mdl-20394071

ABSTRACT

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.


Subject(s)
Delayed-Action Preparations/administration & dosage , Delayed-Action Preparations/chemistry , Delayed-Action Preparations/radiation effects , Glioma/drug therapy , Nanostructures/chemistry , Paclitaxel/chemistry , Absorbable Implants , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/chemistry , Cell Line, Tumor , Diffusion , Glioma/pathology , Gold/chemistry , Hot Temperature , Humans , Infrared Rays , Macromolecular Substances/chemistry , Materials Testing , Microspheres , Molecular Conformation , Nanostructures/administration & dosage , Nanostructures/ultrastructure , Paclitaxel/administration & dosage , Surface Properties
15.
Mol Imaging Biol ; 11(6): 424-33, 2009.
Article in English | MEDLINE | ID: mdl-19424760

ABSTRACT

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.


Subject(s)
Gelatinases/chemistry , Peptides, Cyclic/chemistry , Amino Acid Sequence , Carbocyanines/metabolism , Cell Line, Tumor , Chemical Phenomena , Fluorescent Dyes/chemistry , Gelatinases/analysis , Gelatinases/metabolism , Glioma/pathology , Humans , Lung Neoplasms/pathology , Male , Molecular Structure , Molecular Weight , Peptides, Cyclic/isolation & purification , Prostatic Neoplasms/pathology , Protein Binding , Protein Stability , Spectroscopy, Near-Infrared , Structure-Activity Relationship
16.
Mol Imaging ; 6(6): 417-26, 2007.
Article in English | MEDLINE | ID: mdl-18053412

ABSTRACT

Phosphatidylserine (PS) is a well-characterized biomarker for apoptosis. Ligands that bind to PS can be used for noninvasive imaging of therapy-induced cell death, particularly apoptosis. In this study, we screened a random 12-mer peptide phage library on liposomes prepared from PS. One clone displaying the peptide SVSVGMKPSPRP (designated as PS3-10) bound to PS approximately 4-fold better than its binding to phosphatidylcholine and 18-fold better than to bovine serum albumin in a solid-phase binding assay. In addition, the binding of the corresponding PS3-10 peptide to PS was significantly higher than that of a scrambled peptide. PS3-10 phages, but not a control 4-2-2 phage, bound to aged red blood cells that had PS exposed on their surface. Binding of PS3-10 phages and PS3-10 peptide to TRAIL-induced apoptotic DLD1 cells was 3.2 and 5.4 times higher than their binding to untreated viable cells, respectively. Significantly, immunohistochemical staining confirmed selective binding of PS3-10 phages to apoptotic cells. Our data suggest that panning of phage display libraries may allow the selection of suitable peptide ligands for apoptotic cells and that PS3-10 peptide may serve as a template for further development of molecular probes for in vitro and in vivo imaging of apoptosis.


Subject(s)
Apoptosis/physiology , Bacteriophage M13/metabolism , Diagnostic Imaging/methods , Drug Delivery Systems , Peptide Library , Peptides/metabolism , Phosphatidylserines/metabolism , Amino Acid Sequence , Cellular Senescence/physiology , Erythrocytes/drug effects , Erythrocytes/physiology , Humans , Membrane Glycoproteins/metabolism , Peptides/analysis , Protein Binding , Sequence Homology, Amino Acid , TNF-Related Apoptosis-Inducing Ligand/pharmacology , Tumor Cells, Cultured
17.
Pharm Res ; 24(6): 1217-24, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17377742

ABSTRACT

PURPOSE: To develop an L-PG-based imaging probe suitable for assessing the degradation of L-PG in vivo. MATERIALS AND METHODS: Conjugates of L-PG and a near-infrared fluorescence (NIRF) dye, NIR813, were characterized with regard to quenching efficiency and degradability by cathepsin B (CB) and other proteases. The kinetics of L-PG-NIR813's degradation and its degradation in orthotopic human U87/TGL glioma in nude mice after intravenous injection was assessed using NIRF optical imaging (n = 3). RESULTS: The fluorescence signal from L-PG-NIR813 was efficiently quenched and activated at NIR813 loadings of 8-10%. Upon exposure to CB, the fluorescence intensity of L-PG-NIR813 increased 10-fold. L-PG-NIR813 was also degraded by another cysteine protease cathepsin L, but not by MMP-2, cathepsin E, cathepsin D, and plasmin. A selective CB inhibitor blocked the fluorescence activation. After intravenous injection, the degradation of L-PG-NIR813 was visualized primarily in the liver, which peaked at 4 h postinjection. Activation of L-PG-NIR813 but not D-PG-NIR813 was clearly seen in U87/TGL tumors. CONCLUSION: Our results indicate that L-PG-NIR813 may be used to monitor the in vivo degradation of L-PG-based polymeric drugs, and that this agent may prove useful in noninvasive imaging of protease activity, particularly that of cysteine proteases.


Subject(s)
Drug Carriers , Polyglutamic Acid/metabolism , Animals , Blotting, Western , Cell Line, Tumor , Fluorescent Dyes , Glioma/metabolism , Humans , Mice , Spectroscopy, Near-Infrared
18.
J Phys Chem C Nanomater Interfaces ; 111(17): 6245-6251, 2007 Apr 06.
Article in English | MEDLINE | ID: mdl-20165552

ABSTRACT

We describe the synthesis, characterization, and use of hybrid nanoparticles with a superparamagnetic iron oxide (SPIO) core and a gold nanoshell. These multifunctional nanoparticles, designated SPIO-Au nanoshells, displayed superparamagnetic characteristics and a significant absorbance in the near-infrared (NIR) region of the electromagnetic spectrum. In addition, they exhibited high transverse relaxivity, r2 , and a large r2/r1 ratio and therefore could be imaged by MRI to obtain T2-weighted images. Moreover, SPIO-Au nanoshells showed efficient photo-thermal effect when exposed to NIR light. The use of SPIO-Au nanoshells, with their combination of unique magnetic and optical properties, should enhance the efficacy of nanoshell-mediated photo-thermal therapy by making it possible to direct more nanoparticles to tumors through the application of external magnetic field and by permitting real-time in vivo MRI imaging of the distribution of the nanoparticles before, during, and after photo-thermal therapy.

19.
Mol Cancer Res ; 3(4): 219-26, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15831675

ABSTRACT

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.


Subject(s)
Adenovirus E1A Proteins/genetics , Apoptosis/physiology , Caspases/metabolism , Membrane Glycoproteins/pharmacology , Ovarian Neoplasms/therapy , Tumor Necrosis Factor-alpha/pharmacology , Adenovirus E1A Proteins/metabolism , Antibodies/pharmacology , Apoptosis Regulatory Proteins , Breast Neoplasms/therapy , Bystander Effect , Caspase 3 , Caspase Inhibitors , Cell Line, Tumor , Cysteine Proteinase Inhibitors/pharmacology , Female , Genetic Therapy/methods , Humans , Membrane Glycoproteins/immunology , Membrane Glycoproteins/metabolism , Oligopeptides/pharmacology , Recombinant Proteins/immunology , Recombinant Proteins/metabolism , Recombinant Proteins/pharmacology , TNF-Related Apoptosis-Inducing Ligand , Transfection , Tumor Necrosis Factor-alpha/immunology , Tumor Necrosis Factor-alpha/metabolism
20.
Oncogene ; 23(26): 4556-66, 2004 Jun 03.
Article in English | MEDLINE | ID: mdl-15122330

ABSTRACT

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.


Subject(s)
Breast Neoplasms/genetics , Genetic Therapy/methods , Nuclear Proteins/genetics , Alternative Splicing , Amino Acid Sequence , Animals , Antineoplastic Agents/pharmacology , Breast Neoplasms/therapy , CDC2 Protein Kinase/metabolism , CDC2-CDC28 Kinases/metabolism , Carcinogenicity Tests , Cyclin-Dependent Kinase 2 , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/metabolism , Female , Gene Expression Regulation, Neoplastic , Humans , Mice , Mice, Nude , Molecular Sequence Data , Nuclear Proteins/metabolism , Nuclear Proteins/pharmacology , Protein Isoforms , Retinoblastoma Protein/metabolism , Tumor Cells, Cultured , Tumor Suppressor Protein p53/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...