Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
Oncotarget ; 6(6): 3540-52, 2015 Feb 28.
Article in English | MEDLINE | ID: mdl-25686838

ABSTRACT

Oligometastasis is a clinically distinct subset of metastasis characterized by a limited number of metastases potentially curable with localized therapies. We analyzed pathways targeted by microRNAs over-expressed in clinical oligometastasis samples and identified suppression of cellular adhesion, invasion, and motility pathways in association with the oligometastatic phenotype. We identified miR-127-5p, miR-544a, and miR-655-3p encoded in the 14q32 microRNA cluster as co-regulators of multiple metastatic pathways through repression of shared target genes. These microRNAs suppressed cellular adhesion and invasion and inhibited metastasis development in an animal model of breast cancer lung colonization. Target genes, including TGFBR2 and ROCK2, were key mediators of these effects. Understanding the role of microRNAs expressed in oligometastases may lead to improved identification of and interventions for patients with curable metastatic disease, as well as an improved understanding of the molecular basis of this unique clinical entity.


Subject(s)
Chromosomes, Human, Pair 14 , MicroRNAs/genetics , Neoplasms/genetics , Neoplasms/pathology , Animals , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , Gene Expression , Heterografts , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Neoplasm Metastasis , Phenotype
2.
Sci Transl Med ; 6(229): 229ra42, 2014 Mar 26.
Article in English | MEDLINE | ID: mdl-24670686

ABSTRACT

Mutagenesis is a hallmark of malignancy, and many oncologic treatments function by generating additional DNA damage. Therefore, DNA damage repair is centrally important in both carcinogenesis and cancer treatment. Homologous recombination (HR) and nonhomologous end joining are alternative pathways of double-strand DNA break repair. We developed a method to quantify the efficiency of DNA repair pathways in the context of cancer therapy. The recombination proficiency score (RPS) is based on the expression levels for four genes involved in DNA repair pathway preference (Rif1, PARI, RAD51, and Ku80), such that high expression of these genes yields a low RPS. Carcinoma cells with low RPS exhibit HR suppression and frequent DNA copy number alterations, which are characteristic of error-prone repair processes that arise in HR-deficient backgrounds. The RPS system was clinically validated in patients with breast or non-small cell lung carcinomas (NSCLCs). Tumors with low RPS were associated with greater mutagenesis, adverse clinical features, and inferior patient survival rates, suggesting that HR suppression contributes to the genomic instability that fuels malignant progression. This adverse prognosis associated with low RPS was diminished if NSCLC patients received adjuvant chemotherapy, suggesting that HR suppression and associated sensitivity to platinum-based drugs counteract the adverse prognosis associated with low RPS. Therefore, RPS may help oncologists select which therapies will be effective for individual patients, thereby enabling more personalized care.


Subject(s)
Antineoplastic Agents/therapeutic use , DNA Repair/genetics , Drug Therapy , Gene Expression Regulation, Neoplastic , Neoplasms/drug therapy , Neoplasms/genetics , Antineoplastic Agents/pharmacology , Cell Line, Tumor , DNA Repair/drug effects , DNA Replication/drug effects , DNA Replication/genetics , Gene Expression Regulation, Neoplastic/drug effects , Genomic Instability/drug effects , Homologous Recombination/drug effects , Homologous Recombination/genetics , Humans , Prognosis , Stress, Physiological/drug effects , Stress, Physiological/genetics
3.
Proc Natl Acad Sci U S A ; 111(4): E484-91, 2014 Jan 28.
Article in English | MEDLINE | ID: mdl-24434553

ABSTRACT

An siRNA screen targeting 89 IFN stimulated genes in 14 different cancer cell lines pointed to the RIG-I (retinoic acid inducible gene I)-like receptor Laboratory of Genetics and Physiology 2 (LGP2) as playing a key role in conferring tumor cell survival following cytotoxic stress induced by ionizing radiation (IR). Studies on the role of LGP2 revealed the following: (i) Depletion of LGP2 in three cancer cell lines resulted in a significant increase in cell death following IR, (ii) ectopic expression of LGP2 in cells increased resistance to IR, and (iii) IR enhanced LGP2 expression in three cell lines tested. Studies designed to define the mechanism by which LGP2 acts point to its role in regulation of IFNß. Specifically (i) suppression of LGP2 leads to enhanced IFNß, (ii) cytotoxic effects following IR correlated with expression of IFNß inasmuch as inhibition of IFNß by neutralizing antibody conferred resistance to cell death, and (iii) mouse embryonic fibroblasts from IFN receptor 1 knockout mice are radioresistant compared with wild-type mouse embryonic fibroblasts. The role of LGP2 in cancer may be inferred from cumulative data showing elevated levels of LGP2 in cancer cells are associated with more adverse clinical outcomes. Our results indicate that cytotoxic stress exemplified by IR induces IFNß and enhances the expression of LGP2. Enhanced expression of LGP2 suppresses the IFN stimulated genes associated with cytotoxic stress by turning off the expression of IFNß.


Subject(s)
Cell Survival/physiology , DEAD-box RNA Helicases/physiology , Neoplasms, Experimental/pathology , RNA Helicases/physiology , Radiation, Ionizing , Animals , Apoptosis , Brain Neoplasms/pathology , DEAD Box Protein 58 , DEAD-box RNA Helicases/metabolism , Glioblastoma/pathology , Humans , Interferon Type I/biosynthesis , Mice , Mice, Knockout , Neoplasms, Experimental/metabolism , RNA Helicases/metabolism , Tumor Cells, Cultured
4.
PLoS One ; 7(12): e50141, 2012.
Article in English | MEDLINE | ID: mdl-23251360

ABSTRACT

RATIONALE: Strategies to stage and treat cancer rely on a presumption of either localized or widespread metastatic disease. An intermediate state of metastasis termed oligometastasis(es) characterized by limited progression has been proposed. Oligometastases are amenable to treatment by surgical resection or radiotherapy. METHODS: We analyzed microRNA expression patterns from lung metastasis samples of patients with ≤ 5 initial metastases resected with curative intent. RESULTS: Patients were stratified into subgroups based on their rate of metastatic progression. We prioritized microRNAs between patients with the highest and lowest rates of recurrence. We designated these as high rate of progression (HRP) and low rate of progression (LRP); the latter group included patients with no recurrences. The prioritized microRNAs distinguished HRP from LRP and were associated with rate of metastatic progression and survival in an independent validation dataset. CONCLUSION: Oligo- and poly- metastasis are distinct entities at the clinical and molecular level.


Subject(s)
Adenocarcinoma/genetics , Adenocarcinoma/secondary , Lung Neoplasms/genetics , Lung Neoplasms/secondary , Lung/pathology , MicroRNAs/genetics , Adenocarcinoma/mortality , Disease Progression , Humans , Lung/metabolism , Lung Neoplasms/mortality , MicroRNAs/metabolism , Survival Rate
5.
PLoS One ; 7(10): e46104, 2012.
Article in English | MEDLINE | ID: mdl-23056240

ABSTRACT

BACKGROUND: Vascular endothelial cells contribute to the pathogenesis of numerous human diseases by actively regulating the stromal inflammatory response; however, little is known regarding the role of endothelial inflammation in the growth of human tumors and its influence on the prognosis of human cancers. METHODS: Using an experimental model of tumor necrosis factor-alpha (TNF-α)-mediated inflammation, we characterized inflammatory gene expression in immunopurified tumor-associated endothelial cells. These genes formed the basis of a multivariate molecular predictor of overall survival that was trained and validated in four types of human cancer. RESULTS: We report that expression of experimentally derived tumor endothelial genes distinguished pathologic tissue specimens from normal controls in several human diseases associated with chronic inflammation. We trained these genes in human cancer datasets and defined a six-gene inflammatory signature that predicted significantly reduced overall survival in breast cancer, colon cancer, lung cancer, and glioma. This endothelial-derived signature predicted outcome independently of, but cooperatively with, standard clinical and pathological prognostic factors. Consistent with these findings, conditioned culture media from human endothelial cells stimulated by pro-inflammatory cytokines accelerated the growth of human colon and breast tumors in immunodeficient mice as compared with conditioned media from untreated endothelial cells. CONCLUSIONS: This study provides the first prognostic cancer gene signature derived from an experimental model of tumor-associated endothelial inflammation. These findings support the notion that activation of inflammatory pathways in non-malignant tumor-infiltrating endothelial cells contributes to tumor growth and progression in multiple human cancers. Importantly, these results identify endothelial-derived factors that could serve as potential targets for therapy in diverse human cancers.


Subject(s)
Endothelium, Vascular/metabolism , Inflammation/genetics , Neoplasms/genetics , Neovascularization, Pathologic/genetics , Adult , Aged , Animals , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , Cells, Cultured , Colonic Neoplasms/genetics , Colonic Neoplasms/pathology , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Endothelium, Vascular/pathology , Female , Gene Expression Profiling , Glioma/genetics , Glioma/pathology , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Inflammation/pathology , Kaplan-Meier Estimate , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Male , Mice , Middle Aged , Multivariate Analysis , Neoplasms/blood supply , Neoplasms/pathology , Neovascularization, Pathologic/pathology , Oligonucleotide Array Sequence Analysis , Tumor Necrosis Factor-alpha/pharmacology
6.
Oncol Rep ; 27(5): 1625-9, 2012 May.
Article in English | MEDLINE | ID: mdl-22294050

ABSTRACT

Signaling pathways that activate mTOR (mammalian target of rapamycin) are altered in many human cancers and these alterations are associated with prognosis and treatment response. mTOR inhibition can restore sensitivity to DNA damaging agents such as cisplatin. The rapamycin derivative everolimus exhibits antitumor activity and is approved for patients with renal cell cancer. Clinically, everolimus has also been evaluated in patients with advanced non-small cell lung cancer (NSCLC) that were refractory to chemotherapy and epidermal growth factor receptor tyrosine kinase inhibitors. We tested the effects of combined treatment with everolimus (RAD001) and fractionated radiation using a xenograft model of human NSCLC (A549 cells). In growth studies, mean tumor volume was reduced in the everolimus plus 30 Gy cohort with significant tumor growth suppression compared to 30 Gy alone (p=0015), or everolimus alone (p<0.001, ANOVA). everolimus (20 nM) significantly reduced protein levels of the mTOR downstream effector p70-S6K compared with radiation and vehicle (p=0.05, ANOVA) and significantly suppressed phospho-p70-S6K levels compared with all other treatments (p<0.001, ANOVA). We also evaluated everolimus and radiation effects on gene expression in A549 cells. Everolimus ± 5 Gy suppressed endothelin 1 and lactate dehydrogenase expression and increased VEGFA, p21, hypoxia-inducible factor-1α and SLC2A1 (facilitated glucose transporter 1). mTOR mRNA levels were unaffected while TNF-α levels were increased with everolimus + 5 Gy compared to either treatment alone. These findings suggest that everolimus increases the antitumor activity of radiation. Clinical trials combining everolimus with fractionated radiation in patients with NSCLC are warranted.


Subject(s)
Carcinoma, Non-Small-Cell Lung/drug therapy , Lung Neoplasms/drug therapy , Radiation-Sensitizing Agents/therapeutic use , Sirolimus/analogs & derivatives , Animals , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/radiotherapy , Cell Line, Tumor , Combined Modality Therapy , Everolimus , Female , Gene Expression/drug effects , Humans , Lung Neoplasms/genetics , Lung Neoplasms/radiotherapy , Mice , Mice, Nude , Phosphorylation/drug effects , Radiation, Ionizing , Radiation-Sensitizing Agents/pharmacology , Ribosomal Protein S6 Kinases, 70-kDa/metabolism , Sirolimus/pharmacology , Sirolimus/therapeutic use , Xenograft Model Antitumor Assays
7.
Mol Ther ; 20(5): 1046-55, 2012 May.
Article in English | MEDLINE | ID: mdl-22334019

ABSTRACT

Radiotherapy offers an effective treatment for advanced cancer but local and distant failures remain a significant challenge. Here, we treated melanoma and pancreatic carcinoma in syngeneic mice with ionizing radiation (IR) combined with the poly(ADP-ribose) polymerase inhibitor (PARPi) veliparib to inhibit DNA repair and promote accelerated senescence. Based on prior work implicating cytotoxic T lymphocytes (CTLs) as key mediators of radiation effects, we discovered that senescent tumor cells induced by radiation and veliparib express immunostimulatory cytokines to activate CTLs that mediate an effective antitumor response. When these senescent tumor cells were injected into tumor-bearing mice, an antitumor CTL response was induced which potentiated the effects of radiation, resulting in elimination of established tumors. Applied to human cancers, radiation-inducible immunotherapy may enhance radiotherapy responses to prevent local recurrence and distant metastasis.


Subject(s)
Benzimidazoles/pharmacology , Cancer Vaccines/therapeutic use , Immunotherapy/methods , Melanoma, Experimental/therapy , Pancreatic Neoplasms/therapy , Radiation-Sensitizing Agents/pharmacology , Animals , Cancer Vaccines/immunology , Cellular Senescence/drug effects , Cellular Senescence/radiation effects , Combined Modality Therapy , Cytokines/biosynthesis , Cytokines/immunology , Cytotoxicity, Immunologic , Female , Humans , Lymphocyte Activation , Melanoma, Experimental/immunology , Melanoma, Experimental/mortality , Mice , Neoplasm Transplantation , Pancreatic Neoplasms/immunology , Pancreatic Neoplasms/mortality , Poly(ADP-ribose) Polymerase Inhibitors , Survival Rate , T-Lymphocytes, Cytotoxic/immunology , Tumor Cells, Cultured
8.
Mol Cell ; 44(5): 785-96, 2011 Dec 09.
Article in English | MEDLINE | ID: mdl-22152481

ABSTRACT

The functional significance of the signaling pathway induced by O(6)-methylguanine (O(6)-MeG) lesions is poorly understood. Here, we identify the p50 subunit of NF-κB as a central target in the response to O(6)-MeG and demonstrate that p50 is required for S(N)1-methylator-induced cytotoxicity. In response to S(N)1-methylation, p50 facilitates the inhibition of NF-κB-regulated antiapoptotic gene expression. Inhibition of NF-κB activity is noted to be an S phase-specific phenomenon that requires the formation of O(6)-MeG:T mismatches. Chk1 associates with p50 following S(N)1-methylation, and phosphorylation of p50 by Chk1 results in the inhibition of NF-κB DNA binding. Expression of an unphosphorylatable p50 mutant blocks inhibition of NF-κB-regulated antiapoptotic gene expression and attenuates S(N)1-methylator-induced cytotoxicity. While O(6)-MeG:T-induced, p50-dependent signaling is not sufficient to induce cell death, this pathway sensitizes cells to the cytotoxic effects of DNA breaks.


Subject(s)
DNA Damage , DNA Methylation , NF-kappa B p50 Subunit/metabolism , Animals , Cell Death , Cell Line, Tumor , Humans , Mice , NF-kappa B p50 Subunit/antagonists & inhibitors , NF-kappa B p50 Subunit/deficiency
9.
PLoS One ; 6(12): e28650, 2011.
Article in English | MEDLINE | ID: mdl-22174856

ABSTRACT

BACKGROUND: Cancer staging and treatment presumes a division into localized or metastatic disease. We proposed an intermediate state defined by ≤ 5 cumulative metastasis(es), termed oligometastases. In contrast to widespread polymetastases, oligometastatic patients may benefit from metastasis-directed local treatments. However, many patients who initially present with oligometastases progress to polymetastases. Predictors of progression could improve patient selection for metastasis-directed therapy. METHODS: Here, we identified patterns of microRNA expression of tumor samples from oligometastatic patients treated with high-dose radiotherapy. RESULTS: Patients who failed to develop polymetastases are characterized by unique prioritized features of a microRNA classifier that includes the microRNA-200 family. We created an oligometastatic-polymetastatic xenograft model in which the patient-derived microRNAs discriminated between the two metastatic outcomes. MicroRNA-200c enhancement in an oligometastatic cell line resulted in polymetastatic progression. CONCLUSIONS: These results demonstrate a biological basis for oligometastases and a potential for using microRNA expression to identify patients most likely to remain oligometastatic after metastasis-directed treatment.


Subject(s)
MicroRNAs/genetics , Neoplasm Metastasis/genetics , Animals , Cell Line, Tumor , Cluster Analysis , Databases, Genetic , Disease Models, Animal , Disease Progression , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Lung/pathology , Mice , MicroRNAs/metabolism , Reproducibility of Results , Xenograft Model Antitumor Assays
10.
Mol Cancer Ther ; 10(7): 1185-93, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21571912

ABSTRACT

Radiation therapy remains a promising modality for curative treatment of localized prostate cancer, but dose-limiting toxicities significantly limit its effectiveness. Agents that enhance efficacy at lower radiation doses might have considerable value in increasing tumor control without compromising organ function. Here, we tested the hypothesis that the PARP inhibitor ABT-888 (veliparib) can enhance the response of prostate cancer cells and tumors to ionizing radiation (IR). Following exposure of DU-145 and PC-3 prostate cancer cell lines to the combination of 10 µmol/L ABT-888 and 6 Gy, we observed similar persistence between both cell lines of DNA damage foci and in vitro radiosensitization. We have previously observed that persistent DNA damage foci formed after ABT-888 plus IR efficiently promote accelerated cell senescence, but only PC-3 cells displayed the expected senescent response of G(2)-M arrest, induction of p21 and ß-galactosidase expression, and accumulation as large flat cells. In turn, combining ABT-888 with 6 Gy resulted in delayed tumor regrowth compared with either agent alone only in PC-3 xenograft tumors, whereas DU-145 tumors continued to grow. By 7 days after treatment with ABT-888 plus IR, PC-3 tumors contained abundant senescent cells displaying persistent DNA damage foci, but no evidence of senescence was noted in the DU-145 tumors. That equivalent radiosensitization by ABT-888 plus IR in vitro failed to predict comparable results with tumors in vivo suggests that the efficacy of PARP inhibitors may partially depend on a competent senescence response to accumulated DNA damage.


Subject(s)
Benzimidazoles/pharmacology , Enzyme Inhibitors/pharmacology , Poly(ADP-ribose) Polymerase Inhibitors , Prostatic Neoplasms/enzymology , Aging/drug effects , Aging/radiation effects , Animals , Cell Line, Tumor , DNA Breaks, Double-Stranded/drug effects , DNA Damage/drug effects , DNA Damage/radiation effects , Female , Humans , Male , Mice , Mice, Nude , Prostatic Neoplasms/genetics , Radiation, Ionizing , Tumor Burden/drug effects , Tumor Burden/radiation effects , Xenograft Model Antitumor Assays
11.
Cancer Res ; 70(15): 6277-82, 2010 Aug 01.
Article in English | MEDLINE | ID: mdl-20610628

ABSTRACT

Persistent DNA double-strand breaks (DSB) may determine the antitumor effects of ionizing radiation (IR) by inducing apoptosis, necrosis, mitotic catastrophe, or permanent growth arrest. IR induces rapid modification of megabase chromatin domains surrounding DSBs via poly-ADP-ribosylation, phosphorylation, acetylation, and protein assembly. The dynamics of these IR-induced foci (IRIF) have been implicated in DNA damage signaling and DNA repair. As an IRIF reporter, we tracked the relocalization of green fluorescent protein fused to a chromatin binding domain of the checkpoint adapter protein 53BP1 after IR of breast cancer cells and tumors. To block DSB repair in breast cancer cells and tumors, we targeted poly(ADP-ribose) polymerase (PARP) with ABT-888 (veliparib), one of several PARP inhibitors currently in clinical trials. PARP inhibition markedly enhanced IRIF persistence and increased breast cancer cell senescence both in vitro and in vivo, arguing for targeting IRIF resolution as a novel therapeutic strategy.


Subject(s)
Benzimidazoles/pharmacology , Breast Neoplasms/drug therapy , Breast Neoplasms/radiotherapy , Enzyme Inhibitors/pharmacology , Poly(ADP-ribose) Polymerase Inhibitors , Animals , Breast Neoplasms/enzymology , Breast Neoplasms/pathology , Cell Growth Processes/drug effects , Cell Growth Processes/radiation effects , Cell Line, Tumor , Cellular Senescence/drug effects , Cellular Senescence/radiation effects , Combined Modality Therapy , Dose-Response Relationship, Radiation , Female , Green Fluorescent Proteins/analysis , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Infrared Rays , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Mice , Mice, Nude , Recombinant Fusion Proteins/analysis , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Tumor Suppressor p53-Binding Protein 1 , Xenograft Model Antitumor Assays
12.
Mol Ther ; 18(5): 912-20, 2010 May.
Article in English | MEDLINE | ID: mdl-20197756

ABSTRACT

Ad.Egr-TNF is a radioinducible adenovector currently in phase 3 trials for inoperable pancreatic cancer. The combination of Ad.Egr-TNF and ionizing radiation (IR) contributes to local tumor control through the production of tumor necrosis factor-alpha (TNFalpha) in the tumor microenvironment. Moreover, clinical and preclinical studies with Ad.Egr-TNF/IR have suggested that this local approach suppresses the growth of distant metastatic disease; however, the mechanisms responsible for this effect remain unclear. These studies have been performed in wild-type (WT) and TNFR1,2(-/-) mice to assess the role of TNFalpha-induced signaling in the suppression of draining lymph node (DLN) metastases. The results demonstrate that production of TNFalpha in the tumor microenvironment induces expression of interferon (IFNbeta). In turn, IFNbeta stimulates the production of chemokines that recruit CD8(+) T cells to the tumor. The results further demonstrate that activation of tumor antigen-specific CD8(+) CTLs contributes to local antitumor activity and suppression of DLN metastases. These findings support a model in which treatment of tumors with Ad.Egr-TNF and IR is mediated by local and distant immune-mediated antitumor effects that suppress the development of metastases.


Subject(s)
CD8-Positive T-Lymphocytes/metabolism , Neoplasm Metastasis/prevention & control , Neoplasm Metastasis/therapy , Radiation, Ionizing , Tumor Necrosis Factor-alpha/metabolism , Adenoviridae/genetics , Animals , Cell Proliferation , Genetic Vectors/genetics , Humans , Interferon-beta/metabolism , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Mutant Strains , Neoplasm Metastasis/genetics , Reverse Transcriptase Polymerase Chain Reaction , Tumor Necrosis Factor-alpha/genetics
13.
PLoS One ; 4(6): e5821, 2009 Jun 08.
Article in English | MEDLINE | ID: mdl-19503789

ABSTRACT

BACKGROUND: Traditionally IFN/STAT1 signaling is connected with an anti-viral response and pro-apoptotic tumor-suppressor functions. Emerging functions of a constitutively activated IFN/STAT1 pathway suggest an association with an aggressive tumor phenotype. We hypothesized that tumor clones that constitutively overexpress this pathway are preferentially selected by the host microenvironment due to a resistance to STAT1-dependent cytotoxicity and demonstrate increased metastatic ability combined with increased resistance to genotoxic stress. METHODOLOGY/PRINCIPAL FINDINGS: Here we report that clones of B16F1 tumors grown in the lungs of syngeneic C57BL/6 mice demonstrate variable transcriptional levels of IFN/STAT1 pathway expression. Tumor cells that constitutively overexpress the IFN/STAT1 pathway (STAT1(H) genotype) are selected by the lung microenvironment. STAT1(H) tumor cells also demonstrate resistance to IFN-gamma (IFNgamma), ionizing radiation (IR), and doxorubicin relative to parental B16F1 and low expressors of the IFN/STAT1 pathway (STAT1(L) genotype). Stable knockdown of STAT1 reversed the aggressive phenotype and decreased both lung colonization and resistance to genotoxic stress. CONCLUSIONS: Our results identify a pathway activated by tumor-stromal interactions thereby selecting for pro-metastatic and therapy-resistant tumor clones. New therapies targeted against the IFN/STAT1 signaling pathway may provide an effective strategy to treat or sensitize aggressive tumor clones to conventional cancer therapies and potentially prevent distant organ colonization.


Subject(s)
STAT1 Transcription Factor/metabolism , Animals , Apoptosis , Doxorubicin/pharmacology , Interferon-gamma/metabolism , Lung/metabolism , Melanoma, Experimental/metabolism , Mice , Mice, Inbred C57BL , Neoplasm Metastasis , Neoplasm Transplantation , Phenotype , Radiation, Ionizing , Signal Transduction , Transcription, Genetic
14.
Int J Radiat Biol ; 85(5): 421-31, 2009 May.
Article in English | MEDLINE | ID: mdl-19437244

ABSTRACT

PURPOSE: To determine the mechanisms of Signal Transducer and Activator of Transcription 1 (Stat1)-associated radioresistance developed by nu61 tumour selected in vivo by fractionated irradiation of the parental radiosensitive tumour SCC61. MATERIALS AND METHODS: Radioresistence of nu61 and SCC61 in vitro was measured by clonogenic assay. Apoptotic response of nu61 and SCC61 cells to genotoxic stress was examined using caspase-based apoptotic assays. Co-cultivation of carboxyfluorescein diacetate, succinimidyl ester (CFDE-SE)-labeled nu61 with un-labeled SCC61 was performed at 1:1 ratio. Production of interleukin-6, interleukin-8 and soluble receptor of interleukin 6 (IL6, IL8 and sIL6R) was measured using Enzyme-Linked Immunosorbent Assay (ELISA). RESULTS: Radioresistant nu61 was also resistant to interferon-gamma (IFNgamma) and the death ligands of tumour necrosis factor alpha receptor (TNFR) family when compared to SCC61. This combined resistance is due to an impaired apoptotic response in nu61. Relative to SCC61, nu61 produced more IL6, IL8 and sIL6R. Using Stat1 knock-downs we demonstrated that IL6 and IL8 production is Stat1-dependent. Treatment with neutralising antibodies to IL6 and IL8, but not to either cytokine alone sensitised nu61 to genotoxic stress induced apoptosis. CONCLUSION: Nu61, which over-expresses Stat1 pathway, is deficient in apoptotic response to ionising radiation and cytotoxic ligands. This resistance to apoptosis is associated with Stat1-dependent production of IL6 and IL8 and suppression of caspases 8, 9 and 3.


Subject(s)
Apoptosis/drug effects , Apoptosis/radiation effects , Interleukins/metabolism , Neoplasms/pathology , Radiation Tolerance , STAT1 Transcription Factor/metabolism , Signal Transduction , Animals , Caspases/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/radiation effects , Coculture Techniques , Cytokines/toxicity , Cytotoxins/toxicity , DNA Damage , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/radiation effects , Humans , Interferon-gamma/pharmacology , Interleukin-6/metabolism , Interleukin-8/metabolism , Neoplasms/genetics , Radiation, Ionizing , Receptors, Tumor Necrosis Factor/metabolism , Signal Transduction/drug effects , Signal Transduction/radiation effects
15.
Int J Oncol ; 31(6): 1519-28, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17982679

ABSTRACT

TNFalpha was initially described as inducing necrotic death in tumors in vivo, and more recently as a cytokine that mediates cytoprotection and inflammation. The anti-tumor effects of TNFalpha are poorly characterized because TNFalpha-induced death of human tumor cells has largely been studied in the presence of agents that block transcription or protein synthesis. Also, most reports in model cell systems describe apoptosis within relatively early time points as the principal mode of cell death induced by TNFalpha. We investigated the cytotoxic effects of 10 ng/ml TNFalpha on human tumor cells of different histological types without concomitant exposure to these inhibitors. Eleven of 21 human tumor cell lines underwent TNFalpha-induced cell death which ranged from 41% to complete loss of viability. Only one cell line demonstrated caspase-dependent apoptosis within 24 h. Nine cell lines underwent death between 48 h and 21 days. Seven of these lines underwent caspase-3 independent death consistent with necrosis. One tumor line exhibited characteristics of senescence following TNFalpha exposure. Nine of 9 cell lines activated NF-kappaB following TNFalpha exposure by 24 h. In all cell lines studied, with the exception of the epidermoid carcinoma cell line that underwent early apoptosis, expression of one or more NF-kappaB target genes was demonstrated at 24-96 h. BMS-345541, a specific IKK inhibitor, increased TNFalpha killing in TNFalpha resistant tumor cell lines by increasing apoptosis, suggesting that inhibition of NF-kappaB may be an effective strategy to enhance the tumoricidal effects of TNFalpha.


Subject(s)
Apoptosis/drug effects , NF-kappa B/antagonists & inhibitors , Neoplasms/drug therapy , Tumor Necrosis Factor-alpha/pharmacology , Cell Line, Tumor , Cell Survival/drug effects , Cellular Senescence/drug effects , Humans , Imidazoles/pharmacology , NF-kappa B/physiology , Necrosis , Neoplasms/pathology , Quinoxalines/pharmacology
16.
Cancer Res ; 67(19): 9214-20, 2007 Oct 01.
Article in English | MEDLINE | ID: mdl-17909027

ABSTRACT

Elsewhere, we reported that multiple serial in vivo passage of a squamous cell carcinoma cells (SCC61) concurrent with ionizing radiation (IR) treatment resulted in the selection of radioresistant tumor (nu61) that overexpresses the signal transducer and activator of transcription 1 (Stat1)/IFN-dependent pathway. Here, we report that (a) the Stat1 pathway is induced by IR, (b) constitutive overexpression of Stat1 is linked with failure to transmit a cytotoxic signal by radiation or IFNs, (c) selection of parental cell line SCC61 against IFN-alpha and IFN-gamma leads to the same IR- and IFN-resistant phenotype as was obtained by IR selection, and (d) suppression of Stat1 by short hairpin RNA renders the IR-resistant nu61 cells radiosensitive to IR. We propose a model that transient induction of Stat1 by IFN, IR, or other stress signals activates cytotoxic genes and cytotoxic response. Constitutive overexpression of Stat1 on the other hand leads to the suppression of the cytotoxic response and induces prosurvival genes that, at high levels of Stat1, render the cells resistant to IR or other inducers of cell death.


Subject(s)
Carcinoma, Squamous Cell/metabolism , STAT1 Transcription Factor/metabolism , Animals , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/radiotherapy , Drug Resistance, Neoplasm , Female , Gene Expression/drug effects , Gene Expression/radiation effects , Humans , Interferon-alpha/pharmacology , Interferon-gamma/pharmacology , Mice , Mice, Nude , Neoplasm Transplantation , RNA, Small Interfering/genetics , Radiation Tolerance , STAT1 Transcription Factor/antagonists & inhibitors , STAT1 Transcription Factor/biosynthesis , STAT1 Transcription Factor/genetics , Transplantation, Heterologous
17.
Cancer Res ; 67(14): 6889-98, 2007 Jul 15.
Article in English | MEDLINE | ID: mdl-17638900

ABSTRACT

The alkylating agent temozolomide, commonly used in the treatment of malignant glioma, causes cellular cytotoxicity by forming O(6)-methylguanine adducts. In this report, we investigated whether temozolomide alters the activity of the transcription factor nuclear factor-kappaB (NF-kappaB). Temozolomide inhibits basal and tumor necrosis factor alpha (TNFalpha)-induced NF-kappaB transcriptional activity without altering phosphorylation or degradation of inhibitor of kappaB-alpha. Inhibition of NF-kappaB is secondary to attenuation of p65 DNA binding, not nuclear translocation. Inhibition of DNA binding is shown both in vitro, with gel shift studies and DNA binding assays, and in vivo at kappaB sites. Consistent with inhibition of NF-kappaB activity, temozolomide reduces basal and TNFalpha-induced kappaB-dependent gene expression. Temozolomide also inhibits NF-kappaB activated by inducers other than TNFalpha, including lipopolysaccharide, doxorubicin, and phorbol 12-myristate 13-acetate. The inhibitory action of temozolomide on NF-kappaB is observed to be maximal following pretreatment of cells with temozolomide for 16 h and is also seen with the S(N)1-type methylating agent methylnitrosourea. The ability of temozolomide to form O(6)-methylguanine adducts is important for inhibition of NF-kappaB as is the presence of a functioning mismatch repair system. Activation of NF-kappaB with TNFalpha before administration of temozolomide reduces the cytotoxicity of temozolomide, whereas 16-h pretreatment with temozolomide resensitizes cells to killing. This work shows a mechanism whereby O(6)-methylguanine adducts formed by temozolomide lead to inhibition of NF-kappaB activity and illustrates a link between mismatch repair processing of alkylator-induced DNA damage and cell death.


Subject(s)
Dacarbazine/analogs & derivatives , Guanine/analogs & derivatives , NF-kappa B/metabolism , Transcription Factor RelA/metabolism , Active Transport, Cell Nucleus , Antineoplastic Agents, Alkylating/pharmacology , Cell Line, Tumor , DNA Damage , Dacarbazine/pharmacology , Guanine/chemistry , Humans , Luciferases/metabolism , Microscopy, Fluorescence , Protein Binding , Subcellular Fractions/metabolism , Temozolomide , Tetradecanoylphorbol Acetate/pharmacology , Time Factors , Transcription Factor RelA/antagonists & inhibitors
18.
Cancer Res ; 65(20): 9479-84, 2005 Oct 15.
Article in English | MEDLINE | ID: mdl-16230412

ABSTRACT

Ionizing radiation potentiates the oncolytic activity of attenuated herpes simplex viruses in tumors exposed to irradiation at specific time intervals by inducing higher virus yields. Cell culture studies have shown that an attenuated virus lacking the viral gamma(1)34.5 genes underproduces late proteins whose synthesis depends on sustained synthesis of viral DNA. Here we report that ionizing radiation enhances gene expression from late viral promoters in transduced cells in the absence of other viral gene products. Consistent with this result, we show that in tumors infected with the attenuated virus, ionizing radiation increases 13.6-fold above baseline the gene expression from a late viral promoter as early as 2 hours after virus infection, an interval too short to account for viral DNA synthesis. The radiation-dependent up-regulation of late viral genes is mediated by the p38 pathway, inasmuch as the enhancement is abolished by p38 inhibitors or a p38 dominant-negative construct. The p38 pathway is not essential for wild-type virus gene expression. The results suggest that ionizing radiation up-regulates late promoters active in the course of viral DNA synthesis and provide a rationale for use of radiation to up-regulate cytotoxic genes introduced into tumor cells by viral vectors for cytoreductive therapy.


Subject(s)
Herpesvirus 1, Human/enzymology , Herpesvirus 1, Human/radiation effects , Neoplasms/therapy , Neoplasms/virology , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Chlorocebus aethiops , DNA, Viral/biosynthesis , DNA, Viral/genetics , Enzyme Activation , Gene Expression Regulation, Viral/radiation effects , HeLa Cells , Herpesvirus 1, Human/genetics , Humans , MAP Kinase Signaling System , Mice , Pancreatic Neoplasms/therapy , Pancreatic Neoplasms/virology , Phosphorylation , Promoter Regions, Genetic/radiation effects , Rabbits , Up-Regulation/radiation effects , Vero Cells , Xenograft Model Antitumor Assays , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors
19.
Cancer Res ; 65(8): 3146-54, 2005 Apr 15.
Article in English | MEDLINE | ID: mdl-15833844

ABSTRACT

We did expressional profiling on 24 paired samples of normal esophageal epithelium, Barrett's metaplasia, and esophageal adenocarcinomas. Matching tissue samples representing the three different histologic types were obtained from each patient undergoing esophagectomy for adenocarcinoma. Our analysis compared the molecular changes accompanying the transformation of normal squamous epithelium with Barrett's esophagus and adenocarcinoma in individual patients rather than in a random cohort. We tested the hypothesis that expressional profiling may reveal gene sets that can be used as molecular markers of progression from normal esophageal epithelium to Barrett's esophagus and adenocarcinoma. Expressional profiling was done using U133A GeneChip (Affymetrix), which represent approximately two thirds of the human genome. The final selection of 214 genes permitted the discrimination of differential gene expression of normal esophageal squamous epithelium, Barrett's esophagus, and adenocarcinoma using two-dimensional hierarchical clustering of selected genes. These data indicate that transformation of Barrett's esophagus to adenocarcinoma is associated with suppression of the genes involved in epidermal differentiation, including genes in 1q21 loci and corresponding to the epidermal differentiation complex. Correlation analysis of genes concordantly expressed in Barrett's esophagus and adenocarcinoma revealed 21 genes that represent potential genetic markers of disease progression and pharmacologic targets for treatment intervention. PCR analysis of genes selected based on DNA array experiments revealed that estimation of the ratios of GATA6 to SPRR3 allows discrimination among normal esophageal epithelium, Barrett's dysplasia, and adenocarcinoma.


Subject(s)
Adenocarcinoma/genetics , Barrett Esophagus/genetics , Cell Transformation, Neoplastic/genetics , Esophageal Neoplasms/genetics , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Barrett Esophagus/metabolism , Barrett Esophagus/pathology , Cell Differentiation/genetics , Cornified Envelope Proline-Rich Proteins , DNA-Binding Proteins/biosynthesis , DNA-Binding Proteins/genetics , Disease Progression , Epithelial Cells/metabolism , Epithelial Cells/physiology , Esophageal Neoplasms/metabolism , Esophageal Neoplasms/pathology , GATA6 Transcription Factor , Gene Expression , Gene Expression Profiling , Humans , Multigene Family , Proteins/genetics , Reverse Transcriptase Polymerase Chain Reaction , Transcription Factors/biosynthesis , Transcription Factors/genetics
20.
FEBS Lett ; 565(1-3): 167-70, 2004 May 07.
Article in English | MEDLINE | ID: mdl-15135073

ABSTRACT

We demonstrate that human umbilical vein endothelial cells (HUVEC) grown in co-culture (CC) with U87 glioblastoma cells transfected with green fluorescent protein (GFP-U87) exhibit resistance to radiation-mediated apoptosis. cDNA macroarray analysis reveals increases in the accumulation of RNAs for HUVEC genes encoding cell adhesion molecules, growth factor-related proteins, and cell cycle regulatory/DNA repair proteins. An increase in protein expression of integrin alphav, integrin beta1, MAPK(p42), Rad51, DNA-PK(CS), and ataxia telangiectasia gene (ATM) was detected in HUVEC grown in CC with GFP-U87 cells compared with HUVEC grown in mono-culture. Treatment with anti-VEGF antibody decreases the expression of integrin alphav, integrin beta1, DNA-PK(CS) and ATM with a corresponding increase in ionizing radiation (IR)-induced apoptosis. These data support the concept that endothelial cells growing in the tumor microenvironment may develop resistance to cytotoxic therapies due to the up-regulation by tumor cells of endothelial cells genes associated with survival.


Subject(s)
Apoptosis , Endothelial Cells/pathology , Glioblastoma/pathology , Ataxia Telangiectasia Mutated Proteins , Blotting, Western , Cell Adhesion , Cell Cycle , Cell Cycle Proteins , Cell Division , Cell Line, Tumor , Cells, Cultured , Coculture Techniques , DNA Repair , DNA, Complementary/metabolism , DNA-Activated Protein Kinase , DNA-Binding Proteins/metabolism , Endothelial Cells/radiation effects , Endothelium, Vascular/cytology , Glioblastoma/radiotherapy , Green Fluorescent Proteins , Humans , Infrared Rays , Integrin alphaV/biosynthesis , Integrin alphaVbeta3/metabolism , Integrin beta1/biosynthesis , Luminescent Proteins/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Nuclear Proteins , Oligonucleotide Array Sequence Analysis , Protein Serine-Threonine Kinases/metabolism , Rad51 Recombinase , Radiotherapy , Recombination, Genetic , Transcription, Genetic , Transfection , Tumor Suppressor Proteins , Umbilical Veins/cytology , Up-Regulation , Vascular Endothelial Growth Factor A/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...