Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 40
1.
Mol Carcinog ; 63(5): 897-911, 2024 May.
Article En | MEDLINE | ID: mdl-38353358

Increasing evidence has demonstrated that glutaminase (GLS) as a key mitochondrial enzyme plays a pivotal role in glutaminolysis, which widely participates in glutamine metabolism serving as main energy sources and building blocks for tumor growth. However, the roles and molecular mechanisms of GLS in esophageal squamous cell carcinoma (ESCC) remains unknown. Here, we found that GLS was highly expressed in ESCC tissues and cells. GLS inhibitor CB-839 significantly suppressed cell proliferation, colony formation, migration and invasion of ESCC cells, whereas GLS overexpression displayed the opposite effects. In addition, CB-839 markedly suppressed glucose consumption and lactate production, coupled with the downregulation of glycolysis-related proteins HK2, PFKM, PKM2 and LDHA, whereas GLS overexpression exhibited the adverse results. In vivo animal experiment revealed that CB-839 dramatically suppressed tumor growth, whereas GLS overexpression promoted tumor growth in ESCC cells xenografted nude mice. Mechanistically, GLS was localized in mitochondria of ESCC cells, which interacted with PDK1 protein. CB-839 attenuated the interaction of GLS and PDK1 in ESCC cells by suppressing PDK1 expression, which further evoked the downregulation of p-PDHA1 (s293), however, GLS overexpression markedly enhanced the level of p-PDHA1 (s293). These findings suggest that interaction of GLS with PDK1 accelerates the glycolysis of ESCC cells by inactivating PDH enzyme, and thus targeting GLS may be a novel therapeutic approach for ESCC patients.


Benzeneacetamides , Esophageal Neoplasms , Esophageal Squamous Cell Carcinoma , Glutaminase , Glycolysis , Pyruvate Dehydrogenase Acetyl-Transferring Kinase , Thiadiazoles , Animals , Humans , Mice , Cell Line, Tumor , Cell Movement , Cell Proliferation , Esophageal Neoplasms/genetics , Esophageal Neoplasms/metabolism , Esophageal Neoplasms/pathology , Esophageal Squamous Cell Carcinoma/genetics , Esophageal Squamous Cell Carcinoma/metabolism , Esophageal Squamous Cell Carcinoma/pathology , Gene Expression Regulation, Neoplastic , Glutaminase/genetics , Glutaminase/metabolism , Glycolysis/genetics , Mice, Nude , Pyruvate Dehydrogenase Acetyl-Transferring Kinase/genetics , Pyruvate Dehydrogenase Acetyl-Transferring Kinase/metabolism
2.
J Biol Chem ; 297(5): 101163, 2021 11.
Article En | MEDLINE | ID: mdl-34481841

Inactivation of p53 is present in almost every tumor, and hence, p53-reactivation strategies are an important aspect of cancer therapy. Common mechanisms for p53 loss in cancer include expression of p53-negative regulators such as MDM2, which mediate the degradation of wildtype p53 (p53α), and inactivating mutations in the TP53 gene. Currently, approaches to overcome p53 deficiency in these cancers are limited. Here, using non-small cell lung cancer and glioblastoma multiforme cell line models, we show that two alternatively spliced, functional truncated isoforms of p53 (p53ß and p53γ, comprising exons 1 to 9ß or 9γ, respectively) and that lack the C-terminal MDM2-binding domain have markedly reduced susceptibility to MDM2-mediated degradation but are highly susceptible to nonsense-mediated decay (NMD), a regulator of aberrant mRNA stability. In cancer cells harboring MDM2 overexpression or TP53 mutations downstream of exon 9, NMD inhibition markedly upregulates p53ß and p53γ and restores activation of the p53 pathway. Consistent with p53 pathway activation, NMD inhibition induces tumor suppressive activities such as apoptosis, reduced cell viability, and enhanced tumor radiosensitivity, in a relatively p53-dependent manner. In addition, NMD inhibition also inhibits tumor growth in a MDM2-overexpressing xenograft tumor model. These results identify NMD inhibition as a novel therapeutic strategy for restoration of p53 function in p53-deficient tumors bearing MDM2 overexpression or p53 mutations downstream of exon 9, subgroups that comprise approximately 6% of all cancers.


Gene Expression Regulation, Neoplastic , Mutation , Nonsense Mediated mRNA Decay , Proto-Oncogene Proteins c-mdm2 , Tumor Suppressor Protein p53 , A549 Cells , Animals , Humans , Mice , Protein Isoforms/biosynthesis , Protein Isoforms/genetics , Proto-Oncogene Proteins c-mdm2/genetics , Proto-Oncogene Proteins c-mdm2/metabolism , Tumor Suppressor Protein p53/biosynthesis , Tumor Suppressor Protein p53/genetics
3.
J Clin Endocrinol Metab ; 106(11): e4652-e4665, 2021 10 21.
Article En | MEDLINE | ID: mdl-34147031

CONTEXT: Anaplastic thyroid cancer (ATC) is a rare, aggressive, and deadly disease. Robust preclinical thyroid cancer models are needed to adequately develop and study novel therapeutic agents. Patient-derived xenograft (PDX) models may resemble patient tumors by recapitulating key genetic alterations and gene expression patterns, making them excellent preclinical models for drug response evaluation. OBJECTIVE: We developed distinct ATC PDX models concurrently with cell lines and characterized them in vitro and in vivo. METHODS: Fresh thyroid tumor from patients with a preoperative diagnosis of ATC was surgically collected and divided for concurrent cell line and PDX model development. Cell lines were created by generating single cells through enzymatic digestion. PDX models were developed following direct subcutaneous implantation of fresh tumor on the flank of immune compromised/athymic mice. RESULTS: Six ATC PDX models and 4 cell lines were developed with distinct genetic profiles. Mutational characterization showed one BRAF/TP53/CDKN2A, one BRAF/CDKN2A, one BRAF/TP53, one TP53 only, one TERT-promoter/HRAS, and one TERT-promoter/KRAS/TP53/NF2/NFE2L2 mutated phenotype. Hematoxylin-eosin staining comparing the PDX models to the original patient surgical specimens show remarkable resemblance, while immunohistochemistry stains for important biomarkers were in full concordance (cytokeratin, TTF-1, PAX8, BRAF). Short tandem repeats DNA fingerprinting analysis of all PDX models and cell lines showed strong concordance with the original tumor. PDX successful establishment rate was 32%. CONCLUSION: We have developed and characterized 6 novel ATC PDX models with 4 matching cell lines. Each PDX model harbors a distinct genetic profile, making them excellent tools for preclinical therapeutic trials.


Biomarkers, Tumor/metabolism , Disease Models, Animal , Gene Expression Regulation, Neoplastic , Phenotype , Thyroid Carcinoma, Anaplastic/pathology , Thyroid Neoplasms/pathology , Aged , Animals , Apoptosis , Biomarkers, Tumor/genetics , Cell Proliferation , Female , Humans , Male , Mice , Middle Aged , Prognosis , Survival Rate , Thyroid Carcinoma, Anaplastic/genetics , Thyroid Carcinoma, Anaplastic/metabolism , Thyroid Neoplasms/genetics , Thyroid Neoplasms/metabolism , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
4.
J Clin Endocrinol Metab ; 106(10): 2962-2978, 2021 09 27.
Article En | MEDLINE | ID: mdl-34120183

BACKGROUND: Despite the use of aggressive multimodality treatment, most anaplastic thyroid carcinoma (ATC) patients die within a year of diagnosis. Although the combination of BRAF and MEK inhibitors has recently been approved for use in BRAF-mutated ATC, they remain effective in a minority of patients who are likely to develop drug resistance. There remains a critical clinical need for effective systemic agents for ATC with a reasonable toxicity profile to allow for rapid translational development. MATERIAL AND METHODS: Twelve human thyroid cancer cell lines with comprehensive genomic characterization were used in a high-throughput screening (HTS) of 257 compounds to select agents with maximal growth inhibition. Cell proliferation, colony formation, orthotopic thyroid models, and patient-derived xenograft (PDX) models were used to validate the selected agents. RESULTS: Seventeen compounds were effective, and docetaxel, LBH-589, and pralatrexate were selected for additional in vitro and in vivo analysis as they have been previously approved by the US Food and Drug Administration for other cancers. Significant tumor growth inhibition (TGI) was detected in all tested models treated with LBH-589; pralatrexate demonstrated significant TGI in the orthotopic papillary thyroid carcinoma model and 2 PDX models; and docetaxel demonstrated significant TGI only in the context of mutant TP53. CONCLUSIONS: HTS identified classes of systemic agents that demonstrate preferential effectiveness against aggressive thyroid cancers, particularly those with mutant TP53. Preclinical validation in both orthotopic and PDX models, which are accurate in vivo models mimicking tumor microenvironment, may support initiation of early-phase clinical trials in non-BRAF mutated or refractory to BRAF/MEK inhibition ATC.


Antineoplastic Agents/pharmacology , High-Throughput Screening Assays , Protein Kinase Inhibitors/pharmacology , Thyroid Carcinoma, Anaplastic/drug therapy , Thyroid Neoplasms/drug therapy , Animals , Carcinogenicity Tests , Cell Line, Tumor , Disease Models, Animal , Humans , Proto-Oncogene Proteins B-raf/drug effects , Tumor Microenvironment/drug effects
5.
J Cancer Res Clin Oncol ; 147(10): 2913-2922, 2021 Oct.
Article En | MEDLINE | ID: mdl-34160678

PURPOSE: Assessing the downstaging effects of neoadjuvant chemotherapy (NACT) in patients with locally advanced nasopharyngeal carcinoma (LANPC) and predicting response to treatment remain challenging. The present study aimed to evaluate the long-term prognosis of downstaging after NACT in patients with LANPC and to investigate the prognostic value of post-NACT tumor downstaging on treatment outcomes in the era of concurrent chemoradiotherapy (CCRT). METHODS: This retrospective study included 226 patients with stage III (n = 188) and IVA (n = 38) NPC admitted to Haikou People's Hospital between 1 October 2009 and 1 October 2012. The patients were grouped as downstaging or no after NACT. Overall survival (OS), locoregional failure-free survival (LFFS), and distant failure-free survival (DFFS) were analyzed. RESULTS: Among 226 patients, 196 (86.7%) were in the downstaging group and 30 (13.3%) were in the non-downstaging group. The longest follow-up was 76 months, and the median was 45 months. The 3-year OS rates of the downstaging group and non-downstaging group were 91.0% (95% CI 0.89-0.93) and 69.5% (95% CI 0.66-0.72) (P = 0.005). The 5-year OS rates were 81.6% (95% CI 0.78-0.86) and 53.3% (95% CI 0.49-0.61) (P = 0.001). N downstaging (3-year OS, HR 0.491, 95% CI 0.221-0.881, P = 0.022; 5-year OS, HR = 0.597, 95% CI 0.378-0.878, P = 0.021) was independently associated with OS. CONCLUSION: In the treatment of LANPC, the patients with downstaging after NACT have a better prognosis than those without downstaging. This study suggests that NACT can improve the prognosis for patients with LANPC if there is downstaging.


Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Chemotherapy, Adjuvant/mortality , Nasopharyngeal Carcinoma/mortality , Nasopharyngeal Neoplasms/mortality , Neoadjuvant Therapy/mortality , Adolescent , Adult , Aged , Cisplatin/administration & dosage , Female , Fluorouracil/administration & dosage , Follow-Up Studies , Humans , Male , Middle Aged , Nasopharyngeal Carcinoma/drug therapy , Nasopharyngeal Carcinoma/pathology , Nasopharyngeal Neoplasms/drug therapy , Nasopharyngeal Neoplasms/pathology , Prognosis , Retrospective Studies , Survival Rate , Young Adult
6.
Am J Cancer Res ; 10(3): 908-924, 2020.
Article En | MEDLINE | ID: mdl-32266099

Epithelial ovarian cancer is characterized by universal TP53 mutations, which result in G1/S checkpoint deficiencies. Therefore, it is hypothesized that the abrogation of the G2/M checkpoint with Wee1 inhibitor might preferentially sensitize TP53-defective ovarian cancer cells. Given the extremely high molecular diversity in ovarian cancer, one approach to improving the clinical efficacy is to identify drug combinations that either broaden the applicable spectrum or circumvent resistance. Here, through a high-throughput unbiased proteomic profiling (RPPA), we found the complementary activated mTOR pathway contributes greatly to Wee1 inhibitor resistance. A combination of Wee1 and mTOR inhibits synergistically inhibiting tumor growth in ovarian cancer cell lines and patient-derived xenograft that closely mimic the heterogeneity of patient tumors. Mechanistically, dual Wee1/mTOR inhibition induced massive DNA replication stress, leading to fork stalling and DNA damage. Moreover, we found that the addition of nucleotide metabolic substrate dNTPs alleviated replication stress, restored the cell cycle and reduced apoptosis to some extent, supporting dNTPs depletion is necessary for the synergy between Wee1 and mTOR inhibits. These results suggest that our study opening up a wider therapeutic window of Wee1 inhibitor for the treatment in epithelial ovarian cancers.

7.
EMBO Mol Med ; 11(5)2019 05.
Article En | MEDLINE | ID: mdl-31040125

To address the need for improved systemic therapy for non-small-cell lung cancer (NSCLC), we previously demonstrated that mesenchymal NSCLC was sensitive to polo-like kinase (Plk1) inhibitors, but the mechanisms of resistance in epithelial NSCLC remain unknown. Here, we show that cMet was differentially regulated in isogenic pairs of epithelial and mesenchymal cell lines. Plk1 inhibition inhibits cMet phosphorylation only in mesenchymal cells. Constitutively active cMet abrogates Plk1 inhibitor-induced apoptosis. Likewise, cMet silencing or inhibition enhances Plk1 inhibitor-induced apoptosis. Cells with acquired resistance to Plk1 inhibitors are more epithelial than their parental cells and maintain cMet activation after Plk1 inhibition. In four animal NSCLC models, mesenchymal tumors were more sensitive to Plk1 inhibition alone than were epithelial tumors. The combination of cMet and Plk1 inhibition led to regression of tumors that did not regrow when drug treatment was stopped. Plk1 inhibition did not affect HGF levels but did decrease vimentin phosphorylation, which regulates cMet phosphorylation via ß1-integrin. This research defines a heretofore unknown mechanism of ligand-independent activation of cMet downstream of Plk1 and an effective combination therapy.


Apoptosis , Cell Cycle Proteins/antagonists & inhibitors , Protein Serine-Threonine Kinases/antagonists & inhibitors , Proto-Oncogene Proteins c-met/metabolism , Proto-Oncogene Proteins/antagonists & inhibitors , Vimentin/metabolism , Animals , Apoptosis/drug effects , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Drug Resistance, Neoplasm/drug effects , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Epithelial-Mesenchymal Transition/drug effects , Female , Humans , Integrin beta1/metabolism , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Mice, Nude , Phenotype , Phosphorylation/drug effects , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Pteridines/pharmacology , Transforming Growth Factor beta/pharmacology , Xenograft Model Antitumor Assays , Polo-Like Kinase 1
8.
Clin Cancer Res ; 25(11): 3329-3340, 2019 06 01.
Article En | MEDLINE | ID: mdl-30770351

PURPOSE: Head and neck squamous cell carcinoma (HNSCC) is driven largely by the loss of tumor suppressor genes, including NOTCH1, but lacks a biomarker-driven targeted therapy. Although the PI3K/mTOR pathway is frequently altered in HNSCC, the disease has modest clinical response rates to PI3K/mTOR inhibitors and lacks validated biomarkers of response. We tested the hypothesis that an unbiased pharmacogenomics approach to PI3K/mTOR pathway inhibitors would identify novel, clinically relevant molecular vulnerabilities in HNSCC with loss of tumor suppressor function.Experimental Design: We assessed the degree to which responses to PI3K/mTOR inhibitors are associated with gene mutations in 59 HNSCC cell lines. Apoptosis in drug-sensitive cell lines was confirmed in vitro and in vivo. NOTCH1 pathway components and PDK1 were manipulated with drugs, gene editing, knockdown, and overexpression. RESULTS: PI3K/mTOR inhibition caused apoptosis and decreased colony numbers in HNSCC cell lines harboring NOTCH1 loss-of-function mutations (NOTCH1 MUT) and reduced tumor size in subcutaneous and orthotopic xenograft models. In all cell lines, NOTCH1 MUT was strongly associated with sensitivity to six PI3K/mTOR inhibitors. NOTCH1 inhibition or knockout increased NOTCH1 WT HNSCC sensitivity to PI3K/mTOR inhibition. PDK1 levels dropped following PI3K/mTOR inhibition in NOTCH1 MUT but not NOTCH1 WT HNSCC, and PDK1 overexpression rescued apoptosis in NOTCH1 MUT cells. PDK1 and AKT inhibitors together caused apoptosis in NOTCH1 WT HNSCC but had little effect as single agents. CONCLUSIONS: Our findings suggest that NOTCH1 MUT predicts response to PI3K/mTOR inhibitors, which may lead to the first biomarker-driven targeted therapy for HNSCC, and that targeting PDK1 sensitizes NOTCH1 WT HNSCC to PI3K/mTOR pathway inhibitors.


Phosphatidylinositol 3-Kinases/metabolism , Pyruvate Dehydrogenase Acetyl-Transferring Kinase/metabolism , Receptor, Notch1/genetics , Squamous Cell Carcinoma of Head and Neck/etiology , Squamous Cell Carcinoma of Head and Neck/metabolism , TOR Serine-Threonine Kinases/metabolism , Animals , Apoptosis/genetics , CRISPR-Cas Systems , Cell Line, Tumor , Cell Proliferation/drug effects , Disease Models, Animal , Dose-Response Relationship, Drug , Gene Editing , Gene Expression , Gene Knockdown Techniques , Humans , Loss of Function Mutation , Mice , Protein Kinase Inhibitors/pharmacology , Receptor, Notch1/metabolism , Signal Transduction/drug effects , Squamous Cell Carcinoma of Head and Neck/drug therapy , Squamous Cell Carcinoma of Head and Neck/pathology
9.
Biotechnol Appl Biochem ; 66(3): 309-315, 2019 May.
Article En | MEDLINE | ID: mdl-30624798

Oval cells, a kind of hepatic progenitor cell quiescent at normal condition, activates to proliferate and differentiate into hepatocytes under severe and long-term liver injury, which usually raises severe inflammation. However, how oval cell survives in the inflammatory milieu interne is still unclear. Tumor necrosis factor α (TNFα), mimicking inflammatory hepatic milieu interne, was used to treat oval cell line, WB-F344, to test the protective function of matrilin-2. In this study, our data suggested that matrilin-2 prevented TNFα-induced apoptosis in WB-F344 cells via inhibiting ASK1/MKK7/JNK pathway. In conclusion, we determined that matrilin-2 plays the key role in maintaining the survival of oval cell and guarantees its proliferation under various injury factors.


Apoptosis/drug effects , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , Tumor Necrosis Factor-alpha/pharmacology , Animals , Cell Survival/drug effects , Cells, Cultured , JNK Mitogen-Activated Protein Kinases/metabolism , Matrilin Proteins/metabolism , Rats , Rats, Inbred F344
10.
Neoplasia ; 20(10): 975-984, 2018 10.
Article En | MEDLINE | ID: mdl-30157470

Mouse models are powerful tools to study lung cancer initiation and progression in vivo and have contributed significantly to recent advances in therapy. Using micro-computed tomography to monitor and study parenchymal and extra-parenchymal metastases in existing murine models of lung cancer is challenging owing to a lack of radiographic contrast and difficulty in achieving respiratory gating. To facilitate the analysis of these in vivo imaging studies and study of tumor progression in murine models we developed a novel, rapid, semi-automated method of calculating thoracic tumor burden from computed tomography images. This method, in which commercially available software is used to calculate the mass of the thoracic cavity (MTC), takes into account the aggregate tumor burden in the thoracic cavity. The present study showed that in tumor-free mice, the MTC does not change over time and is not affected by breathing, whereas in tumor-bearing mice, the increase in the MTC is a measure of tumor mass that correlates well with tumor burden measured by lung weight. Tumor burden calculated with our MTC method correlated with that measured by lung weight as well as or better than that calculated using four established methods. To test this method, we assessed metastatic tumor development and response to a pharmacologic PLK1 inhibitor in an orthotopic xenograft mouse model. PLK1 inhibition significantly inhibited tumor growth. Our results demonstrate that the MTC method can be used to study dynamic changes in tumor growth and response to therapeutics in genetically engineered mouse models and orthotopic xenograft mouse models of lung cancer.


Lung Neoplasms/diagnostic imaging , Neoplasms, Experimental/diagnostic imaging , Thoracic Cavity/diagnostic imaging , Tumor Burden , X-Ray Microtomography/methods , Animals , Antineoplastic Agents/pharmacology , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Male , Mice, Inbred Strains , Mice, Transgenic , Neoplasms, Experimental/pathology , Pteridines/pharmacology , Thoracic Cavity/pathology , Thoracic Neoplasms/diagnostic imaging , Tumor Burden/drug effects
11.
Cancer Lett ; 431: 64-72, 2018 09 01.
Article En | MEDLINE | ID: mdl-29807113

To address the unmet need for effective biomarker-driven targeted therapy for human papillomavirus (HPV)-associated head and neck squamous cell carcinoma (HNSCC) and cervical cancer, we conducted a high-throughput drug screen using 1122 compounds in 13 HPV-positive and 11 matched HPV-negative cell lines. The most effective drug classes were inhibitors of polo-like kinase, proteasomes, histone deacetylase, and Aurora kinases. Treatment with a pan-Aurora inhibitor, danusertib, led to G2M arrest and apoptosis in vitro. Furthermore, danusertib decreased tumor size compared with controls in patient derived xenograft models of HNSCC. To identify biomarkers predicting response, we determined associations between mutations and drug sensitivity. Our data and the Genomics of Drug Sensitivity in Cancer database showed that cancer cells with KMT2D mutations were more sensitive to Aurora kinase inhibitors than were cells without mutations. Knockdown of KMT2D in wild-type cells led to increased Aurora kinase inhibitor-induced apoptosis. We identified Aurora kinase inhibitors as effective and understudied drugs in HNSCC and CESC. This is the first published study to demonstrate that mutations in KMT2D, which are common in many cancers, correlate with drug sensitivity in two independent datasets.


Aurora Kinase A/antagonists & inhibitors , Carcinoma, Squamous Cell/genetics , DNA-Binding Proteins/genetics , Mutation , Neoplasm Proteins/genetics , Papillomavirus Infections/genetics , Animals , Apoptosis , Area Under Curve , Benzamides/pharmacology , Biomarkers/metabolism , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/virology , Cell Cycle , Cell Line , Cell Proliferation , Drug Evaluation, Preclinical , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Mice , Neoplasm Transplantation , Papillomaviridae , Papillomavirus Infections/drug therapy , Pharmacogenetics , Pyrazoles/pharmacology , Uterine Cervical Neoplasms
12.
Oncotarget ; 8(49): 86369-86383, 2017 Oct 17.
Article En | MEDLINE | ID: mdl-29156801

Human cancer cell lines are the most frequently used preclinical models in the study of cancer biology and the development of therapeutics. Although anatomically diverse, human papillomavirus (HPV)-driven cancers have a common etiology and similar mutations that overlap with but are distinct from those found in HPV-negative cancers. Building on prior studies that have characterized subsets of head and neck squamous cell carcinoma (HNSCC) and cervical squamous cell carcinoma (CESC) cell lines separately, we performed genomic, viral gene expression, and viral integration analyses on 74 cell lines that include all readily-available HPV-positive (9 HNSCC, 8 CESC) and CESC (8 HPV-positive, 2 HPV-negative) cell lines and 55 HPV-negative HNSCC cell lines. We used over 700 human tumors for comparison. Mutation patterns in the cell lines were similar to those of human tumors. We confirmed HPV viral protein and mRNA expression in the HPV-positive cell lines. We found HPV types in three CESC cell lines that are distinct from those previously reported. We found that cell lines and tumors had similar patterns of viral gene expression; there were few sites of recurrent HPV integration. As seen in tumors, HPV integration did appear to alter host gene expression in cell lines. The HPV-positive cell lines had higher levels of p16 and lower levels of Rb protein expression than did the HPV-negative lines. Although the number of HPV-positive cell lines is limited, our results suggest that these cell lines represent suitable models for studying HNSCC and CESC, both of which are common and lethal.

13.
Sci Rep ; 7(1): 12890, 2017 10 10.
Article En | MEDLINE | ID: mdl-29018259

Synchronous gastric tumors that consist of both gastrointestinal stromal tumor (GIST) and adenocarcinoma are rare. We studied the clinicopathological and molecular characteristics of six cases containing both gastric adenocarcinoma and GIST. By means of immunohistochemical analysis, all GIST cells expressed CD117, CD34 and Dog1 in all six synchronous gastric adenocarcinomas with GIST, and in GIST alone. Sequencing analysis demonstrated that exon 11 c-kit mutations were present in two of six synchronous tumors and four of five GISTs. One of the two exon 11 c-kit mutations in synchronous adenocarcinomas with GISTs was an uncommon mutation of CTT > CCA at amino acid 576, and the other was a GTT deletion at amino acid 560. The mutation was a homozygous A > G mutation in exon 12 (amino acid 567) of PDGFR-α. We concluded that the exon 11 mutations were the most important in both cases of synchronous gastric adenocarcinoma with GIST and GIST alone. The mutation rate was higher in GIST alone than in synchronous adenocarcinoma with GIST.


Adenocarcinoma/genetics , Adenocarcinoma/pathology , Gastrointestinal Stromal Tumors/genetics , Gastrointestinal Stromal Tumors/pathology , Neoplasms, Multiple Primary/genetics , Neoplasms, Multiple Primary/pathology , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology , Aged , Base Sequence , DNA, Neoplasm/genetics , Female , Humans , Male , Middle Aged , Mutation/genetics , Oncogenes
15.
Cancer Lett ; 392: 71-82, 2017 04 28.
Article En | MEDLINE | ID: mdl-28126323

The genomic alterations identified in head and neck squamous cell carcinoma (HNSCC) tumors have not resulted in any changes in clinical care, making the development of biomarker-driven targeted therapy for HNSCC a major translational gap in knowledge. To fill this gap, we used 59 molecularly characterized HNSCC cell lines and found that mutations of AJUBA, SMAD4 and RAS predicted sensitivity and resistance to treatment with inhibitors of polo-like kinase 1 (PLK1), checkpoint kinases 1 and 2, and WEE1. Inhibition or knockdown of PLK1 led to cell-cycle arrest at the G2/M transition and apoptosis in sensitive cell lines and decreased tumor growth in an orthotopic AJUBA-mutant HNSCC mouse model. AJUBA protein expression was undetectable in most AJUBA-mutant HNSCC cell lines, and total PLK1 and Bora protein expression were decreased. Exogenous expression of wild-type AJUBA in an AJUBA-mutant cell line partially rescued the phenotype of PLK1 inhibitor-induced apoptosis and decreased PLK1 substrate inhibition, suggesting a threshold effect in which higher drug doses are required to affect PLK1 substrate inhibition. PLK1 inhibition was an effective therapy for HNSCC in vitro and in vivo. However, biomarkers to guide such therapy are lacking. We identified AJUBA, SMAD4 and RAS mutations as potential candidate biomarkers of response of HNSCC to treatment with these mitotic inhibitors.


Antineoplastic Agents/pharmacology , Carcinoma, Squamous Cell/drug therapy , Cell Cycle Proteins/antagonists & inhibitors , Cell Proliferation/drug effects , Head and Neck Neoplasms/drug therapy , LIM Domain Proteins/genetics , Mutation , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Proto-Oncogene Proteins/antagonists & inhibitors , Pteridines/pharmacology , Animals , Apoptosis/drug effects , Carcinoma, Squamous Cell/enzymology , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/pathology , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Checkpoint Kinase 1/antagonists & inhibitors , Checkpoint Kinase 1/metabolism , Checkpoint Kinase 2/antagonists & inhibitors , Checkpoint Kinase 2/metabolism , Dose-Response Relationship, Drug , G2 Phase Cell Cycle Checkpoints/drug effects , Genotype , Head and Neck Neoplasms/enzymology , Head and Neck Neoplasms/genetics , Head and Neck Neoplasms/pathology , Humans , Mice, Nude , Molecular Targeted Therapy , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/metabolism , Phenotype , Protein Serine-Threonine Kinases/metabolism , Protein-Tyrosine Kinases/antagonists & inhibitors , Protein-Tyrosine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Pyrimidinones , RNA Interference , Signal Transduction/drug effects , Smad4 Protein/genetics , Squamous Cell Carcinoma of Head and Neck , Thiophenes/pharmacology , Time Factors , Transfection , Tumor Burden/drug effects , Urea/analogs & derivatives , Urea/pharmacology , Xenograft Model Antitumor Assays , ras Proteins/genetics , Polo-Like Kinase 1
17.
Clin Cancer Res ; 22(7): 1674-1686, 2016 Apr 01.
Article En | MEDLINE | ID: mdl-26597303

PURPOSE: To identify new therapeutic targets for non-small cell lung cancer (NSCLC), we systematically searched two cancer cell line databases for sensitivity data on a broad range of drugs. We identified polo-like kinase 1 (PLK1) as the most promising target for further investigation based on a subset of sensitive NSCLC cell lines and inhibitors that were in advanced clinical development. EXPERIMENTAL DESIGN: To identify potential biomarkers of response of NSCLC to PLK1 inhibition and mechanisms of PLK1 inhibitor-induced apoptosis, integrated analysis of gene and protein expression, gene mutations, and drug sensitivity was performed using three PLK1 inhibitors (volasertib, BI2536, and GSK461364) with a large panel of NSCLC cell lines. RESULTS: The NSCLC cell lines had different sensitivities to PLK1 inhibition, with a minority demonstrating sensitivity to all three inhibitors. PLK1 inhibition led to G2-M arrest, but only treatment-sensitive cell lines underwent substantial apoptosis following PLK1 inhibition. NSCLC lines with high epithelial-mesenchymal transition (EMT) gene signature scores (mesenchymal cell lines) were more sensitive to PLK1 inhibition than epithelial lines (P< 0.02). Likewise, proteomic profiling demonstrated that E-cadherin expression was higher in the resistant cell lines than in the sensitive ones (P< 0.01). Induction of an epithelial phenotype by expression of the miRNA miR-200 increased cellular resistance to PLK1 inhibition. Also, KRAS mutation and alterations in the tight-junction, ErbB, and Rho signaling pathways correlated with drug response of NSCLC. CONCLUSIONS: In this first reported large-scale integrated analysis of PLK1 inhibitor sensitivity, we demonstrated that EMT leads to PLK1 inhibition sensitivity of NSCLC cells. Our findings have important clinical implications for mesenchymal NSCLC, a significant subtype of the disease that is associated with resistance to currently approved targeted therapies.


Apoptosis/drug effects , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Cell Cycle Proteins/antagonists & inhibitors , Epithelial-Mesenchymal Transition , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Proto-Oncogene Proteins/antagonists & inhibitors , Animals , Apoptosis/genetics , Benzimidazoles/pharmacology , Carcinoma, Non-Small-Cell Lung/genetics , Cell Cycle Checkpoints/drug effects , Cell Cycle Checkpoints/genetics , Cell Line, Tumor , Chromosomal Instability , Disease Models, Animal , Drug Resistance, Neoplasm/genetics , Epithelial-Mesenchymal Transition/genetics , Humans , Lung Neoplasms/genetics , Mice , Mutation , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins p21(ras)/genetics , Pteridines/pharmacology , Thiophenes/pharmacology , Tumor Burden/drug effects , Xenograft Model Antitumor Assays , Polo-Like Kinase 1
18.
Oncotarget ; 7(1): 565-79, 2016 Jan 05.
Article En | MEDLINE | ID: mdl-26623721

Improved therapies are greatly needed for non-small cell lung cancer (NSCLC) that does not harbor targetable kinase mutations or translocations. We previously demonstrated that NSCLC cells that harbor kinase-inactivating BRAF mutations (KIBRAF) undergo senescence when treated with the multitargeted kinase inhibitor dasatinib. Similarly, treatment with dasatinib resulted in a profound and durable response in a patient with KIBRAF NSCLC. However, no canonical pathways explain dasatinib-induced senescence in KIBRAF NSCLC. To investigate the underlying mechanism, we used 2 approaches: gene expression and reverse phase protein arrays. Both approaches showed that DNA repair pathways were differentially modulated between KIBRAF NSCLC cells and those with wild-type (WT) BRAF. Consistent with these findings, dasatinib induced DNA damage and activated DNA repair pathways leading to senescence only in the KIBRAF cells. Moreover, dasatinib-induced senescence was dependent on Chk1 and p21, proteins known to mediate DNA damage-induced senescence. Dasatinib also led to a marked decrease in TAZ but not YAP protein levels. Overexpression of TAZ inhibited dasatinib-induced senescence. To investigate other vulnerabilities in KIBRAF NSCLC cells, we compared the sensitivity of these cells with that of WTBRAF NSCLC cells to 79 drugs and identified a pattern of sensitivity to EGFR and MEK inhibitors in the KIBRAF cells. Clinically approved EGFR and MEK inhibitors, which are better tolerated than dasatinib, could be used to treat KIBRAF NSCLC. Our novel finding that dasatinib induced DNA damage and subsequently activated DNA repair pathways leading to senescence in KIBRAF NSCLC cells represents a unique vulnerability with potential clinical applications.


Cellular Senescence/drug effects , DNA Damage , DNA Repair/drug effects , Dasatinib/pharmacology , Mutation , Proto-Oncogene Proteins B-raf/genetics , Blotting, Western , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Cellular Senescence/genetics , Checkpoint Kinase 1 , Comet Assay , Cyclin-Dependent Kinase Inhibitor p21/genetics , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Gene Expression Profiling/methods , Gene Expression Regulation, Neoplastic/drug effects , Humans , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Protein Kinase Inhibitors/pharmacology , Protein Kinases/genetics , Protein Kinases/metabolism , Proto-Oncogene Proteins B-raf/metabolism , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , Signal Transduction/genetics
19.
Article En | MEDLINE | ID: mdl-26170655

BACKGROUND: The objective of the study is to develop a scoring system for predicting a 90-day re-exacerbation in hospitalized patients with acute exacerbations of chronic obstructive pulmonary disease (AECOPD). METHODS: A total of 176 consecutive hospitalized patients with AECOPD were included. The sociodemographic characteristics, status before acute exacerbation (AE), presentations of and treatment for the current AE, and the re-exacerbation in 90 days after discharge from hospital were collected. RESULTS: The re-exacerbation rate in 90 days was 48.9% (86 out of 176). It was associated with the degree of lung function impairment (Global initiative for chronic Obstructive Lung Disease [GOLD] grades), frequency of AE in the previous year, and parameters of the current AE, including pleural effusion, use of accessory respiratory muscles, inhaled long-acting ß-2-agonists, inhaled corticosteroids, controlled oxygen therapy, noninvasive mechanical ventilation, and length of hospital stay, but was not associated with body mass index, modified Medical Research Council scale, or chronic obstructive pulmonary disease assessment test. A subgroup of ten variables was selected and developed into the re-exacerbation index scoring system (age grades, GOLD grades, AE times in the previous year, pleural effusion, use of accessory respiratory muscles, noninvasive mechanical ventilation, controlled oxygen therapy, inhaled long-acting ß-2-agonists and inhaled corticosteroids, and length of hospital stay). The re-exacerbation index showed good discrimination for re-exacerbation, with a C-statistic of 0.750 (P<0.001). CONCLUSION: A comprehensive assessment integrating parameters of stable chronic obstructive pulmonary disease, clinical presentations at exacerbation, and treatment showed a strong predictive capacity for short-term outcome in patients with AECOPD. Further studies are required to verify these findings.


Decision Support Techniques , Health Status Indicators , Pulmonary Disease, Chronic Obstructive/diagnosis , Aged , Area Under Curve , Disease Progression , Female , Health Status , Hospitalization , Humans , Male , Middle Aged , Predictive Value of Tests , Prognosis , Prospective Studies , Pulmonary Disease, Chronic Obstructive/physiopathology , Pulmonary Disease, Chronic Obstructive/therapy , ROC Curve , Risk Assessment , Risk Factors , Severity of Illness Index , Time Factors
20.
Anticancer Drugs ; 26(8): 835-42, 2015 Sep.
Article En | MEDLINE | ID: mdl-26053277

Treatments for recurrent or metastatic head and neck squamous cell carcinoma (HNSCC) have limited efficacy. One potential therapeutic target for HNSCC is the RAS/RAF/MEK/ERK cascade, which is one of the major signaling pathways for HNSCC cell survival. In HNSCC, RAS can be activated either by HRAS mutation or by upstream signaling. The ABL inhibitor nilotinib acts as a weak RAF inhibitor that induces RAF dimerization and subsequent activation of MEK/ERK in other cancer cell lines with activated RAS, leading to an unexpected dependence on MEK/ERK for cell survival. We hypothesized that nilotinib and the MEK inhibitor MEK162 would be synergistic in HNSCC cell lines owing to the frequent activation of RAS. We treated HNSCC cell lines with nilotinib and performed immunoblotting and cell-viability experiments. We used an orthotopic mouse model to assess synergistic effects in vivo. Nilotinib induced significant BRAF-CRAF heterodimerization and ERK activation irrespective of RAS mutation status. In cell-viability assays, nilotinib synergized with MEK162. MEK162 alone induced G1 arrest that was minimally enhanced by nilotinib. In the mouse model, treatment with MEK162 alone or combined with nilotinib led to tumor growth inhibition. In HNSCC, nilotinib-induced RAF dimerization is independent of RAS mutation status, but this dimerization does not lead to MEK dependence for cell survival in all HNSCC cell lines. MEK inhibition alone leads to decreased proliferation both in vitro and in vivo. Although nilotinib has some synergistic effects with MEK162, other agents may be more effective against HNSCC when combined with MEK162.


Antineoplastic Agents/pharmacology , Head and Neck Neoplasms/pathology , Mitogen-Activated Protein Kinases/metabolism , Neoplasms, Squamous Cell/pathology , raf Kinases/metabolism , ras Proteins/metabolism , Animals , Antineoplastic Agents/therapeutic use , Benzimidazoles/pharmacology , Benzimidazoles/therapeutic use , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Drug Synergism , Head and Neck Neoplasms/drug therapy , Heterografts , Humans , Mice, Nude , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Mutation , Neoplasm Transplantation , Neoplasms, Squamous Cell/drug therapy , Protein Multimerization , Proto-Oncogene Proteins c-raf/metabolism , Pyrimidines/pharmacology , Pyrimidines/therapeutic use , ras Proteins/genetics
...