Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
1.
BMC Cancer ; 24(1): 441, 2024 Apr 10.
Article in English | MEDLINE | ID: mdl-38594604

ABSTRACT

BACKGROUND: We recently found that epiplakin 1 (EPPK1) alterations were present in 12% of lung adenocarcinoma (LUAD) cases and were associated with a poor prognosis in early-stage LUAD when combined with other molecular alterations. This study aimed to identify a probable crucial role for EPPK1 in cancer development. METHODS: EPPK1 mRNA and protein expression was analyzed with clinical variables. Normal bronchial epithelial cell lines were exposed to cigarette smoke for 16 weeks to determine whether EPPK1 protein expression was altered after exposure. Further, we used CRISPR-Cas9 to knock out (KO) EPPK1 in LUAD cell lines and observed how the cancer cells were altered functionally and genetically. RESULTS: EPPK1 protein expression was associated with smoking and poor prognosis in early-stage LUAD. Moreover, a consequential mesenchymal-to-epithelial transition was observed, subsequently resulting in diminished cell proliferation and invasion after EPPK1 KO. RNA sequencing revealed that EPPK1 KO induced downregulation of 11 oncogenes, 75 anti-apoptosis, and 22 angiogenesis genes while upregulating 8 tumor suppressors and 12 anti-cell growth genes. We also observed the downregulation of MYC and upregulation of p53 expression at both protein and RNA levels following EPPK1 KO. Gene ontology enrichment analysis of molecular functions highlighted the correlation of EPPK1 with the regulation of mesenchymal cell proliferation, mesenchymal differentiation, angiogenesis, and cell growth after EPPK1 KO. CONCLUSIONS: Our data suggest that EPPK1 is linked to smoking, epithelial to mesenchymal transition, and the regulation of cancer progression, indicating its potential as a therapeutic target for LUAD.


Subject(s)
Adenocarcinoma of Lung , Adenocarcinoma , Lung Neoplasms , Humans , Lung Neoplasms/pathology , Epithelial-Mesenchymal Transition/genetics , Prognosis , Adenocarcinoma of Lung/pathology , Adenocarcinoma/pathology , Cell Proliferation/genetics , Gene Expression Regulation, Neoplastic , Cell Line, Tumor
3.
Cancer Biomark ; 33(4): 449-465, 2022.
Article in English | MEDLINE | ID: mdl-35491773

ABSTRACT

The Early Detection Research Network's (EDRN) purpose is to discover, develop and validate biomarkers and imaging methods to detect early-stage cancers or at-risk individuals. The EDRN is composed of sites that fall into four categories: Biomarker Developmental Laboratories (BDL), Biomarker Reference Laboratories (BRL), Clinical Validation Centers (CVC) and Data Management and Coordinating Centers. Each component has a crucial role to play within the mission of the EDRN. The primary role of the CVCs is to support biomarker developers through validation trials on promising biomarkers discovered by both EDRN and non-EDRN investigators. The second round of funding for the EDRN Lung CVC at Vanderbilt University Medical Center (VUMC) was funded in October 2016 and we intended to accomplish the three missions of the CVCs: To conduct innovative research on the validation of candidate biomarkers for early cancer detection and risk assessment of lung cancer in an observational study; to compare biomarker performance; and to serve as a resource center for collaborative research within the Network and partner with established EDRN BDLs and BRLs, new laboratories and industry partners. This report outlines the impact of the VUMC EDRN Lung CVC and describes the role in promoting and validating biological and imaging biomarkers.


Subject(s)
Biomarkers, Tumor , Early Detection of Cancer , Lung Neoplasms , Humans , Lung Neoplasms/diagnosis , Validation Studies as Topic
4.
JTO Clin Res Rep ; 3(5): 100306, 2022 May.
Article in English | MEDLINE | ID: mdl-35434667

ABSTRACT

The application of KRAS G12C inhibitors in the setting of NSCLC represents a major milestone for a previously "undruggable" target. Here, we present the second reported case of de novo KRAS G12C-mutant primary SCLC. Would our patient benefit from a KRAS G12C inhibitor?

5.
PLoS One ; 17(3): e0265427, 2022.
Article in English | MEDLINE | ID: mdl-35294486

ABSTRACT

BACKGROUND: 18F-fluorodeoxyglucose (FDG) PET/CT is recommended for evaluation of intermediate-risk indeterminate pulmonary nodules (IPNs). While highly sensitive, the specificity of FDG remains suboptimal for differentiating malignant from benign nodules, particularly in areas where fungal lung diseases are prevalent. Thus, a cancer-specific imaging probe is greatly needed. In this study, we tested the hypothesis that a PET radiotracer (S)-4-(3-[18F]-fluoropropyl)-L-glutamic acid (FSPG) improves the diagnostic accuracy of IPNs compared to 18F-FDG PET/CT. METHODS: This study was conducted at a major academic medical center and an affiliated VA medical center. Twenty-six patients with newly discovered IPNs 7-30mm diameter or newly diagnosed lung cancer completed serial PET/CT scans utilizing 18F-FDG and 18F-FSPG, without intervening treatment of the lesion. The scans were independently reviewed by two dual-trained diagnostic radiology and nuclear medicine physicians. Characteristics evaluated included quantitative SUVmax values of the pulmonary nodules and metastases. RESULTS: A total of 17 out of 26 patients had cancer and 9 had benign lesions. 18F-FSPG was negative in 6 of 9 benign lesions compared to 7 of 9 with 18F-FDG. 18F-FSPG and 18F-FDG were positive in 14 of 17 and 12 of 17 malignant lesions, respectively. 18F-FSPG detected brain and intracardiac metastases missed by 18F-FDG PET in one case, while 18F-FDG detected a metastasis to the kidney missed by 18F-FSPG. CONCLUSION: In this pilot study, there was no significant difference in overall diagnostic accuracy between 18F-FSPG and 18F-FDG for the evaluation of IPNs and staging of lung cancer. Additional studies will be needed to determine the clinical utility of this tracer in the management of IPNs and lung cancer.


Subject(s)
Lung Neoplasms , Multiple Pulmonary Nodules , Fluorodeoxyglucose F18 , Glutamic Acid , Humans , Lung Neoplasms/diagnostic imaging , Multiple Pulmonary Nodules/diagnostic imaging , Pilot Projects , Positron Emission Tomography Computed Tomography/methods , Positron-Emission Tomography/methods , Radiopharmaceuticals , Sensitivity and Specificity
6.
Sci Rep ; 11(1): 14424, 2021 07 13.
Article in English | MEDLINE | ID: mdl-34257356

ABSTRACT

Lung adenocarcinoma (ADC) is a heterogeneous group of tumors associated with different survival rates, even when detected at an early stage. Here, we aim to investigate whether CyTOF identifies cellular and molecular predictors of tumor behavior. We developed and validated a CyTOF panel of 34 antibodies in four ADC cell lines and PBMC. We tested our panel in a set of 10 ADCs, classified into long- (LPS) (n = 4) and short-predicted survival (SPS) (n = 6) based on radiomics features. We identified cellular subpopulations of epithelial cancer cells (ECC) and their microenvironment and validated our results by multiplex immunofluorescence (mIF) applied to a tissue microarray (TMA) of LPS and SPS ADCs. The antibody panel captured the phenotypical differences in ADC cell lines and PBMC. LPS ADCs had a higher proportion of immune cells. ECC clusters (ECCc) were identified and uncovered two ADC groups. ECCc with high HLA-DR expression were correlated with CD4+ and CD8+ T cells, with LPS samples being enriched for those clusters. We confirmed a positive correlation between HLA-DR expression on ECC and T cell number by mIF staining on TMA slides. Spatial analysis demonstrated shorter distances from T cells to the nearest ECC in LPS. Our results demonstrate a distinctive cellular profile of ECC and their microenvironment in ADC. We showed that HLA-DR expression in ECC is correlated with T cell infiltration, and that a set of ADCs with high abundance of HLA-DR+ ECCc and T cells is enriched in LPS samples. This suggests new insights into the role of antigen presenting tumor cells in tumorigenesis.


Subject(s)
Adenocarcinoma of Lung , HLA-DR Antigens , Leukocytes, Mononuclear , CD4-Positive T-Lymphocytes , CD8-Positive T-Lymphocytes , Humans
7.
Mod Pathol ; 34(9): 1614-1633, 2021 09.
Article in English | MEDLINE | ID: mdl-34031537

ABSTRACT

The severe acute respiratory syndrome Coronavirus-2 (SARS-CoV-2) pandemic has had devastating effects on global health and worldwide economy. Despite an initial reluctance to perform autopsies due to concerns for aerosolization of viral particles, a large number of autopsy studies published since May 2020 have shed light on the pathophysiology of Coronavirus disease 2019 (COVID-19). This review summarizes the histopathologic findings and clinicopathologic correlations from autopsies and biopsies performed in patients with COVID-19. PubMed and Medline (EBSCO and Ovid) were queried from June 4, 2020 to September 30, 2020 and histopathologic data from autopsy and biopsy studies were collected based on 2009 Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines. A total of 58 studies reporting 662 patients were included. Demographic data, comorbidities at presentation, histopathologic findings, and virus detection strategies by organ system were collected. Diffuse alveolar damage, thromboembolism, and nonspecific shock injury in multiple organs were the main findings in this review. The pathologic findings emerging from autopsy and biopsy studies reviewed herein suggest that in addition to a direct viral effect in some organs, a unifying pathogenic mechanism for COVID-19 is ARDS with its known and characteristic inflammatory response, cytokine release, fever, inflammation, and generalized endothelial disturbance. This study supports the notion that autopsy studies are of utmost importance to our understanding of disease features and treatment effect to increase our knowledge of COVID-19 pathophysiology and contribute to more effective treatment strategies.


Subject(s)
COVID-19/pathology , Respiratory Distress Syndrome/pathology , Respiratory Distress Syndrome/virology , Humans , SARS-CoV-2
8.
Am J Clin Pathol ; 156(2): 313-319, 2021 07 06.
Article in English | MEDLINE | ID: mdl-33609098

ABSTRACT

OBJECTIVES: Mucinous adenocarcinoma arising in unresected congenital pulmonary airway malformation (CPAM) is rare. Underlying driver mutations in addition to KRAS gain-of-function mutations in this setting and the long-term outcomes of these patients are unknown. METHODS: We report a case of metastatic mucinous adenocarcinoma harboring both KRAS and GNAS mutations arising in a type 1 CPAM of a 14-year-old male. A literature review was performed. RESULTS: Next-generation sequencing revealed identical KRAS (G12V) mutations in both the CPAM and metastatic adenocarcinoma and a missense mutation in the GNAS (R201C) gene in the metastatic adenocarcinoma only. Median survival was 23 and 4 years for patients with localized (no or limited spread within the same lobe of CPAM) and distant involvement (spread to any different lobe of CPAM) of mucinous cells, respectively (95% confidence interval, 23-23 and 1.5-22 years, respectively; P = .017). CONCLUSIONS: Mucinous cell proliferation associated with type 1 CPAM has exceptionally good long-term outcomes if confined within the same lobe of CPAM. A second oncogenic mutation in the GNAS gene may be necessary for progression to malignancy and distant spread.


Subject(s)
Adenocarcinoma, Mucinous/pathology , Chromogranins/genetics , Cystic Adenomatoid Malformation of Lung, Congenital/pathology , GTP-Binding Protein alpha Subunits, Gs/genetics , Lung Neoplasms/pathology , Proto-Oncogene Proteins p21(ras)/genetics , Adenocarcinoma of Lung/genetics , Adenocarcinoma of Lung/pathology , Adenocarcinoma, Mucinous/genetics , Adolescent , Cystic Adenomatoid Malformation of Lung, Congenital/genetics , Humans , Lung Neoplasms/genetics , Male , Mutation
9.
Ann Am Thorac Soc ; 18(7): 1227-1234, 2021 07.
Article in English | MEDLINE | ID: mdl-33400907

ABSTRACT

Rationale: A prospective longitudinal cohort of individuals at high risk of developing lung cancer was established to build a biorepository of carefully annotated biological specimens and low-dose computed tomography (LDCT) chest images for derivation and validation of candidate biomarkers for early detection of lung cancer.Objectives: The goal of this study is to characterize individuals with high risk for lung cancer, accumulating valuable biospecimens and LDCT chest scans longitudinally over 5 years.Methods: Participants 55-80 years of age with a 5-year estimated risk of developing lung cancer >1.5% were recruited and enrolled from clinics at the Vanderbilt University Medical Center, Veteran Affairs Medical Center, and Meharry Medical Center. Individual demographic characteristics were assessed via questionnaire at baseline. Participants underwent an LDCT scan, spirometry, sputum cytology, and research bronchoscopy at the time of enrollment. Participants will be followed yearly for 5 years. Positive LDCT scans are followed-up according to standard of care. The clinical, imaging, and biospecimen data are collected prospectively and stored in a biorepository. Participants are offered smoking cessation counseling at each study visit.Results: A total of 480 participants were enrolled at study baseline and consented to sharing their data and biospecimens for research. Participants are followed with yearly clinic visits to collect imaging data and biospecimens. To date, a total of 19 cancers (13 adenocarcinomas, four squamous cell carcinomas, one large cell neuroendocrine, and one small-cell lung cancer) have been identified.Conclusions: We established a unique prospective cohort of individuals at high risk for lung cancer, enrolled at three institutions, for whom full clinical data, well-annotated LDCT scans, and biospecimens are being collected longitudinally. This repository will allow for the derivation and independent validation of clinical, imaging, and molecular biomarkers of risk for diagnosis of lung cancer.Clinical trial registered with ClinicalTrials.gov (NCT01475500).


Subject(s)
Early Detection of Cancer , Lung Neoplasms , Biomarkers , Humans , Lung Neoplasms/diagnostic imaging , Mass Screening , Prospective Studies , Tomography, X-Ray Computed
10.
Int J Cancer ; 144(5): 1104-1114, 2019 03 01.
Article in English | MEDLINE | ID: mdl-30152518

ABSTRACT

Somatostatin receptor 2 (SSTR2) is overexpressed in a majority of neuroendocrine neoplasms, including small-cell lung carcinomas (SCLCs). SSTR2 was previously considered an inhibitory receptor on cell growth, but its agonists had poor clinical responses in multiple clinical trials. The role of this receptor as a potential therapeutic target in lung cancer merits further investigation. We evaluated the expression of SSTR2 in a cohort of 96 primary tumors from patients with SCLC and found 48% expressed SSTR2. Correlation analysis in both CCLE and an SCLC RNAseq cohort confirmed high-level expression and identified an association between NEUROD1 and SSTR2. There was a significant association with SSTR2 expression profile and poor clinical outcome. We tested whether SSTR2 expression might contribute to tumor progression through activation of downstream signaling pathways, using in vitro and in vivo systems and downregulated SSTR2 expression in lung cancer cells by shRNA. SSTR2 downregulation led to increased apoptosis and dramatically decreased tumor growth in vitro and in vivo in multiple cell lines with decreased AMPKα phosphorylation and increased oxidative metabolism. These results demonstrate a role for SSTR2 signaling in SCLC and suggest that SSTR2 is a poor prognostic biomarker in SCLC and potential future therapeutic signaling target.


Subject(s)
Cell Proliferation/genetics , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Receptors, Somatostatin/genetics , Small Cell Lung Carcinoma/genetics , Small Cell Lung Carcinoma/pathology , AMP-Activated Protein Kinases/genetics , Animals , Apoptosis/genetics , Basic Helix-Loop-Helix Transcription Factors/genetics , Biomarkers, Tumor/genetics , Cell Line, Tumor , Disease Progression , Down-Regulation/genetics , Humans , Mice , Mice, Nude , Nerve Tissue Proteins/genetics , RNA, Messenger/genetics , Signal Transduction/genetics
11.
Oncogene ; 37(36): 5007-5019, 2018 09.
Article in English | MEDLINE | ID: mdl-29789716

ABSTRACT

Many tumors increase uptake and dependence on glucose, cystine or glutamine. These basic observations on cancer cell metabolism have opened multiple new diagnostic and therapeutic avenues in cancer research. Recent studies demonstrated that smoking could induce the expression of xCT (SLC7A11) in oral cancer cells, suggesting that overexpression of xCT may support lung tumor progression. We hypothesized that overexpression of xCT occurs in lung cancer cells to satisfy the metabolic requirements for growth and survival. Our results demonstrated that 1) xCT was highly expressed at the cytoplasmic membrane in non-small cell lung cancer (NSCLC), 2) the expression of xCT was correlated with advanced stage and predicted a worse 5-year survival, 3) targeting xCT transport activity in xCT overexpressing NSCLC cells with sulfasalazine decreased cell proliferation and invasion in vitro and in vivo and 4) increased dependence on glutamine was observed in xCT overexpressed normal airway epithelial cells. These results suggested that xCT regulate metabolic requirements during lung cancer progression and be a potential therapeutic target in NSCLC.


Subject(s)
Amino Acid Transport System y+/metabolism , Carcinoma, Non-Small-Cell Lung/metabolism , Lung Neoplasms/metabolism , 3T3 Cells , A549 Cells , Animals , Cell Line , Cell Line, Tumor , Cell Proliferation/physiology , Cell Survival/physiology , Cystine/metabolism , Cytoplasm/metabolism , Disease Progression , Female , Glutamine/metabolism , Humans , Male , Mice , Middle Aged
12.
J Proteome Res ; 16(9): 3266-3276, 2017 09 01.
Article in English | MEDLINE | ID: mdl-28731711

ABSTRACT

We hypothesized that distinct protein expression features of benign and malignant pulmonary nodules may reveal novel candidate biomarkers for the early detection of lung cancer. We performed proteome profiling by liquid chromatography-tandem mass spectrometry to characterize 34 resected benign lung nodules, 24 untreated lung adenocarcinomas (ADCs), and biopsies of bronchial epithelium. Group comparisons identified 65 proteins that differentiate nodules from ADCs and normal bronchial epithelium and 66 proteins that differentiate ADCs from nodules and normal bronchial epithelium. We developed a multiplexed parallel reaction monitoring (PRM) assay to quantify a subset of 43 of these candidate biomarkers in an independent cohort of 20 benign nodules, 21 ADCs, and 20 normal bronchial biopsies. PRM analyses confirmed significant nodule-specific abundance of 10 proteins including ALOX5, ALOX5AP, CCL19, CILP1, COL5A2, ITGB2, ITGAX, PTPRE, S100A12, and SLC2A3 and significant ADC-specific abundance of CEACAM6, CRABP2, LAD1, PLOD2, and TMEM110-MUSTN1. Immunohistochemistry analyses for seven selected proteins performed on an independent set of tissue microarrays confirmed nodule-specific expression of ALOX5, ALOX5AP, ITGAX, and SLC2A3 and cancer-specific expression of CEACAM6. These studies illustrate the value of global and targeted proteomics in a systematic process to identify and qualify candidate biomarkers for noninvasive molecular diagnosis of lung cancer.


Subject(s)
Adenocarcinoma/diagnosis , Biomarkers, Tumor/genetics , Gene Expression Regulation, Neoplastic , Lung Neoplasms/diagnosis , Neoplasm Proteins/genetics , Solitary Pulmonary Nodule/diagnosis , 5-Lipoxygenase-Activating Proteins/genetics , 5-Lipoxygenase-Activating Proteins/metabolism , Adenocarcinoma/genetics , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Adenocarcinoma of Lung , Adult , Aged , Antigens, CD/genetics , Antigens, CD/metabolism , Arachidonate 5-Lipoxygenase/genetics , Arachidonate 5-Lipoxygenase/metabolism , Biomarkers, Tumor/metabolism , CD11 Antigens/genetics , CD11 Antigens/metabolism , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/metabolism , Diagnosis, Differential , Female , GPI-Linked Proteins/genetics , GPI-Linked Proteins/metabolism , Glucose Transporter Type 3/genetics , Glucose Transporter Type 3/metabolism , Humans , Integrin alpha Chains/genetics , Integrin alpha Chains/metabolism , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Male , Middle Aged , Neoplasm Proteins/metabolism , Proteomics/methods , Respiratory Mucosa/metabolism , Respiratory Mucosa/pathology , Solitary Pulmonary Nodule/genetics , Solitary Pulmonary Nodule/metabolism , Solitary Pulmonary Nodule/pathology , Tandem Mass Spectrometry , Tissue Array Analysis , Transcriptome
13.
Sci Rep ; 7: 45828, 2017 04 07.
Article in English | MEDLINE | ID: mdl-28387221

ABSTRACT

Triple negative breast cancers (TNBC) are aggressive tumors, with high rates of metastatic spread and targeted therapies are critically needed. We aimed to assess the prognostic and predictive value of a 3q 19-gene signature identified previously from lung cancer in a collection of 4,801 breast tumor gene expression data. The 3q gene signature had a strong association with features of aggressiveness such as high grade, hormone receptor negativity, presence of a basal-like or TNBC phenotype and reduced distant metastasis free survival. The 3q gene signature was strongly associated with lung metastasis only in TNBC (P < 0.0001, Hazard ratio (HR) 1.44, 95% confidence interval (CI), 1.31-1.60), significantly associated with brain but not bone metastasis regardless of TNBC status. The association of one 3q driver gene FXR1 with distant metastasis in TNBC (P = 0.01) was further validated by immunohistochemistry. In addition, the 3q gene signature was associated with better response to neoadjuvant chemotherapy in TNBC (P < 0.0001) but not in non-TNBC patients. Our study suggests that the 3q gene signature is a novel prognostic marker for lung and/or brain metastasis and a predictive marker for the response to neoadjuvant chemotherapy in TNBC, implying a potential role for 3q genes in the mechanism of organ-specific metastasis.


Subject(s)
Chromosomes, Human, Pair 3/genetics , Neoplasm Metastasis/diagnosis , Neoplasm Metastasis/genetics , Triple Negative Breast Neoplasms/diagnosis , Triple Negative Breast Neoplasms/genetics , Biomarkers, Tumor/genetics , Chemotherapy, Adjuvant , Disease-Free Survival , Female , Gene Expression Regulation, Neoplastic , Genetic Predisposition to Disease , Humans , Neoplasm Metastasis/drug therapy , RNA-Binding Proteins/metabolism , Triple Negative Breast Neoplasms/drug therapy
14.
J Mol Diagn ; 19(1): 57-64, 2017 01.
Article in English | MEDLINE | ID: mdl-27810331

ABSTRACT

Colorectal (CRCs) and endometrioid (EMCs) cancers in patients with Lynch syndrome exhibit microsatellite instability (MSI) detected by PCR or immunohistochemistry (IHC). While both assays are equally sensitive for CRCs, some suggest that PCR has a higher false-negative rate than IHC in EMCs. We assessed the MSI profiles of 91 EMC and 311 CRC specimens using five mononucleotide repeat markers: BAT25, BAT26, NR21, NR24, and MONO27. EMCs with high MSI (MSI-H) showed a mean left shift of 3 nucleotides (nt), which was significantly different from 6 nt in CRCs. A shift of 1 nt was observed in multiple markers in 76% of MSI-H EMCs, whereas only 12% of MSI-H CRCs displayed a 1-nt shift in one of five markers. IHC against four mismatch repair proteins was performed in 78 EMCs. Loss of staining in one or more proteins was detected in 18 of 19 tumors that were MSI-H by PCR. When EMC tumor cell burden was diluted to <30%, MSI-H was no longer observed in two of three EMCs with a mean nucleotide shift of 1 nt. These results indicate that EMC and CRC MSI profiles are different and that caution should be exercised when interpreting the results, as subtle, 1-nt changes may be missed. These findings provide a potential cause of previously reported discordant MSI and IHC results in EMCs.


Subject(s)
Biomarkers, Tumor/genetics , Carcinoma, Endometrioid/genetics , Colorectal Neoplasms, Hereditary Nonpolyposis/genetics , Endometrial Neoplasms/genetics , Microsatellite Instability , Adult , Aged , Aged, 80 and over , Carcinoma, Endometrioid/metabolism , Carcinoma, Endometrioid/pathology , Colorectal Neoplasms, Hereditary Nonpolyposis/metabolism , Colorectal Neoplasms, Hereditary Nonpolyposis/pathology , Endometrial Neoplasms/metabolism , Endometrial Neoplasms/pathology , False Negative Reactions , Female , Humans , Male , Microsatellite Repeats , Middle Aged , MutS DNA Mismatch-Binding Protein/metabolism , Retrospective Studies , Young Adult
15.
JCI Insight ; 1(19): e88814, 2016 11 17.
Article in English | MEDLINE | ID: mdl-27882349

ABSTRACT

The molecular determinants of lung cancer risk remain largely unknown. Airway epithelial cells are prone to assault by risk factors and are considered to be the primary cell type involved in the field of cancerization. To investigate risk-associated changes in the bronchial epithelium proteome that may offer new insights into the molecular pathogenesis of lung cancer, proteins were identified in the airway epithelial cells of bronchial brushing specimens from risk-stratified individuals by shotgun proteomics. Differential expression of selected proteins was validated by parallel reaction monitoring mass spectrometry in an independent set of individual bronchial brushings. We identified 2,869 proteins, of which 312 proteins demonstrated a trend in expression. Pathway analysis revealed enrichment of carbohydrate metabolic enzymes in high-risk individuals. Glucose consumption and lactate production were increased in human bronchial epithelial BEAS2B cells treated with cigarette smoke condensate for 7 months. Increased lipid biosynthetic capacity and net reductive carboxylation were revealed by metabolic flux analyses of [U-13C5] glutamine in this in vitro model, suggesting profound metabolic reprogramming in the airway epithelium of high-risk individuals. These results provide a rationale for the development of potentially new chemopreventive strategies and selection of patients for surveillance programs.


Subject(s)
Epithelial Cells/metabolism , Proteome/analysis , Respiratory Mucosa/pathology , Smoke/adverse effects , Bronchi , Cell Line , Gene Expression Profiling , Humans , Lipid Metabolism , Lung Neoplasms/metabolism , Metabolomics , Respiratory Mucosa/cytology , Smoking
16.
J Thorac Oncol ; 10(10): 1430-6, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26301800

ABSTRACT

INTRODUCTION: The mutational profile of non-small-cell lung cancer (NSCLC) has become an important tool in tailoring therapy to patients, with clear differences according to the population of origin. African Americans (AAs) have higher lung cancer incidence and mortality than Caucasians, yet discrepant results have been reported regarding the frequency of somatic driver mutations. We hypothesized that NSCLC has a distinct mutational profile in this group. METHODS: We collected NSCLC samples resected from self-reported AAs in five sites from Tennessee, Michigan, and Ohio. Gene mutations were assessed by either SNaPshot or next generation sequencing, and ALK translocations were evaluated by fluorescence in situ hybridization. RESULTS: Two hundred sixty patients were included, mostly males (62.3%) and smokers (86.6%). Eighty-one samples (31.2%) were squamous cell carcinomas. The most frequently mutated genes were KRAS (15.4%), epidermal growth factor receptor (EGFR, 5.0%), PIK3CA (0.8%), BRAF, NRAS, ERBB2, and AKT1 (0.4% each). ALK translocations were detected in two nonsquamous tumors (1.7%), totaling 61 cases (23.5%) with driver oncogenic alterations. Among 179 nonsquamous samples, 54 (30.2%) presented a driver alteration. The frequency of driver alterations altogether was lower than that reported in Caucasians, whereas no difference was detected in either EGFR or KRAS mutations. Overall survival was longer among patients with EGFR mutations. CONCLUSIONS: We demonstrated that NSCLC from AAs has a different pattern of somatic driver mutations than from Caucasians. The majority of driver alterations in this group are yet to be described, which will require more comprehensive panels and assessment of noncanonical alterations.


Subject(s)
Black or African American/genetics , Carcinoma, Non-Small-Cell Lung/ethnology , Carcinoma, Non-Small-Cell Lung/genetics , Lung Neoplasms/ethnology , Lung Neoplasms/genetics , Mutation , Adult , Aged , Aged, 80 and over , Carcinoma, Non-Small-Cell Lung/enzymology , Carcinoma, Non-Small-Cell Lung/pathology , ErbB Receptors/genetics , Female , Genotype , Humans , Lung Neoplasms/enzymology , Lung Neoplasms/pathology , Male , Middle Aged , Retrospective Studies
17.
Int J Cancer ; 137(7): 1587-97, 2015 Oct 01.
Article in English | MEDLINE | ID: mdl-25821004

ABSTRACT

We previously elucidated the pleotropic role of solute carrier family A1 member 5 (SLC1A5) as the primary transporter of glutamine (Gln), a modulator of cell growth and oxidative stress in non-small cell lung cancer (NSCLC). The aim of our study was to evaluate SLC1A5 as a potential new therapeutic target and candidate biomarker predictive of survival and response to therapy. SLC1A5 targeting was examined in a panel of NSCLC and human bronchial cell lines by RNA interference and by a small molecular inhibitor, gamma-l-glutamyl-p-nitroanilide (GPNA). The effects of targeting SLC1A5 on cell growth, Gln uptake, ATP level, autophagy and cell death were examined. Inactivation of SLC1A5 genetically or pharmacologically decreased Gln consumption, inhibited cell growth, induced autophagy and apoptosis in a subgroup of NSCLC cell lines that overexpress SLC1A5. Targeting SLC1A5 function decreased tumor growth in NSCLC xenografts. A multivariate Cox proportional hazards analysis indicates that patients with increased SLC1A5 mRNA expression have significantly shorter overall survival (p = 0.01, HR = 1.24, 95% CI: 1.05-1.46), adjusted for age, gender, smoking history and disease stage. In an immunohistochemistry study on 207 NSCLC patients, SLC1A5 protein expression remained highly significant prognostic value in both univariate (p < 0.0001, HR = 1.45, 95% CI: 1.15-1.50) and multivariate analyses (p = 0.04, HR = 1.22, 95% CI: 1.01-1.31). These results position SLC1A5 as a new candidate prognostic biomarker for selective targeting of Gln-dependent NSCLC.


Subject(s)
Amino Acid Transport System ASC/antagonists & inhibitors , Amino Acid Transport System ASC/metabolism , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/therapy , Glutamine/metabolism , Lung Neoplasms/metabolism , Lung Neoplasms/therapy , Amino Acid Transport System ASC/biosynthesis , Amino Acid Transport System ASC/genetics , Animals , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Gene Knockdown Techniques , Humans , Lung Neoplasms/pathology , Mice , Minor Histocompatibility Antigens , Molecular Targeted Therapy , Prognosis , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/genetics , Xenograft Model Antitumor Assays
18.
Proc Natl Acad Sci U S A ; 112(11): 3469-74, 2015 Mar 17.
Article in English | MEDLINE | ID: mdl-25733852

ABSTRACT

Aberrant expression of RNA-binding proteins has profound implications for cellular physiology and the pathogenesis of human diseases such as cancer. We previously identified the Fragile X-Related 1 gene (FXR1) as one amplified candidate driver gene at 3q26-29 in lung squamous cell carcinoma (SCC). FXR1 is an autosomal paralog of Fragile X mental retardation 1 and has not been directly linked to human cancers. Here we demonstrate that FXR1 is a key regulator of tumor progression and its overexpression is critical for nonsmall cell lung cancer (NSCLC) cell growth in vitro and in vivo. We identified the mechanisms by which FXR1 executes its regulatory function by forming a novel complex with two other oncogenes, protein kinase C, iota and epithelial cell transforming 2, located in the same amplicon via distinct binding mechanisms. FXR1 expression is a candidate biomarker predictive of poor survival in multiple solid tumors including NSCLCs. Because FXR1 is overexpressed and associated with poor clinical outcomes in multiple cancers, these results have implications for other solid malignancies.


Subject(s)
Chromosomes, Human, Pair 3/genetics , Lung Neoplasms/genetics , RNA-Binding Proteins/metabolism , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , Cell Proliferation , DNA Copy Number Variations/genetics , Gene Expression Regulation, Neoplastic , Humans , Isoenzymes/genetics , Isoenzymes/metabolism , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Prognosis , Protein Kinase C/genetics , Protein Kinase C/metabolism , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA-Binding Proteins/genetics , Survival Analysis , Treatment Outcome
19.
Am J Otolaryngol ; 36(3): 475-8, 2015.
Article in English | MEDLINE | ID: mdl-25701460

ABSTRACT

Primary middle ear neoplasms are rare, and commonly cause conductive hearing loss. When aural masses result in sensorineural hearing loss, malignant etiologies are often suspected. Fibroepithelial polyps are benign lesions commonly found in the skin and genitourinary tract and are not commonly identified as primary lesions of the middle ear. Here, we present a case of a benign fibroepithelial polyp of the middle ear causing sensorineural hearing loss and describe its surgical management. Imaging and histologic characteristics are reviewed.


Subject(s)
Ear Diseases/pathology , Ear, Middle , Hearing Loss, Sensorineural/etiology , Polyps/pathology , Ear Diseases/complications , Ear Diseases/surgery , Female , Humans , Middle Aged , Polyps/complications , Polyps/surgery
20.
Mol Cancer Ther ; 14(2): 542-52, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25477325

ABSTRACT

Metastatic EGFR-mutant lung cancers are sensitive to the first- and second-generation EGFR tyrosine kinase inhibitors (TKIs) gefitinib, erlotinib, and afatinib, but resistance develops. Acquired resistance to gefitinib or erlotinib occurs most commonly (>50%) via the emergence of a second-site EGFR mutation, T790M. Two strategies to overcome T790M-mediated resistance are dual inhibition of EGFR with afatinib plus the anti-EGFR antibody cetuximab (A+C), or mutant-specific EGFR inhibition with AZD9291. A+C and AZD9291 are now also being tested as first-line therapies, but whether these therapies will extend progression-free survival or induce more aggressive forms of resistance in this setting remains unknown. We modeled resistance to multiple generations of anti-EGFR therapies preclinically to understand the effects of sequential treatment with anti-EGFR agents on drug resistance and determine the optimal order of treatment. Using a panel of erlotinib/afatinib-resistant cells, including a novel patient-derived cell line (VP-2), we found that AZD9291 was more potent than A+C at inhibiting cell growth and EGFR signaling in this setting. Four of four xenograft-derived A+C-resistant cell lines displayed in vitro and in vivo sensitivity to AZD9291, but four of four AZD9291-resistant cell lines demonstrated cross-resistance to A+C. Addition of cetuximab to AZD9291 did not confer additive benefit in any preclinical disease setting. This work, emphasizing a mechanistic understanding of the effects of therapies on tumor evolution, provides a framework for future clinical trials testing different treatment sequences. This paradigm is applicable to other tumor types in which multiple generations of inhibitors are now available.


Subject(s)
ErbB Receptors/genetics , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Molecular Targeted Therapy , Mutation/genetics , Acrylamides/pharmacology , Acrylamides/therapeutic use , Afatinib , Aged , Aniline Compounds/pharmacology , Aniline Compounds/therapeutic use , Animals , Antibodies, Monoclonal, Humanized/pharmacology , Antibodies, Monoclonal, Humanized/therapeutic use , Cell Line, Tumor , Cell Proliferation/drug effects , Cetuximab , Drug Resistance, Neoplasm/drug effects , Humans , Male , Mice, Nude , Quinazolines/pharmacology , Quinazolines/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL
...