Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 34
1.
Ann Oncol ; 31(2): 274-282, 2020 02.
Article En | MEDLINE | ID: mdl-31959344

BACKGROUND: The etiology and the molecular basis of lung adenocarcinomas (LuADs) in nonsmokers are currently unknown. Furthermore, the scarcity of available primary cultures continues to hamper our biological understanding of non-smoking-related lung adenocarcinomas (NSK-LuADs). PATIENTS AND METHODS: We established patient-derived cancer cell (PDC) cultures from metastatic NSK-LuADs, including two pairs of matched EGFR-mutant PDCs before and after resistance to tyrosine kinase inhibitors (TKIs), and then performed whole-exome and RNA sequencing to delineate their genomic architecture. For validation, we analyzed independent cohorts of primary LuADs. RESULTS: In addition to known non-smoker-associated alterations (e.g. RET, ALK, EGFR, and ERBB2), we discovered novel fusions and recurrently mutated genes, including ATF7IP, a regulator of gene expression, that was inactivated in 5% of primary LuAD cases. We also found germline mutations at dominant familiar-cancer genes, highlighting the importance of genetic predisposition in the origin of a subset of NSK-LuADs. Furthermore, there was an over-representation of inactivating alterations at RB1, mostly through complex intragenic rearrangements, in treatment-naive EGFR-mutant LuADs. Three EGFR-mutant and one EGFR-wild-type tumors acquired resistance to EGFR-TKIs and chemotherapy, respectively, and histology on re-biopsies revealed the development of small-cell lung cancer/squamous cell carcinoma (SCLC/LuSCC) transformation. These features were consistent with RB1 inactivation and acquired EGFR-T790M mutation or FGFR3-TACC3 fusion in EGFR-mutant tumors. CONCLUSIONS: We found recurrent alterations in LuADs that deserve further exploration. Our work also demonstrates that a subset of NSK-LuADs arises within cancer-predisposition syndromes. The preferential occurrence of RB1 inactivation, via complex rearrangements, found in EGFR-mutant tumors appears to favor SCLC/LuSCC transformation under growth-inhibition pressures. Thus RB1 inactivation may predict the risk of LuAD transformation to a more aggressive type of lung cancer, and may need to be considered as a part of the clinical management of NSK-LuADs patients.


ErbB Receptors , Lung Neoplasms , Adenocarcinoma of Lung , Drug Resistance, Neoplasm/genetics , ErbB Receptors/genetics , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Microtubule-Associated Proteins , Mutation , Protein Kinase Inhibitors/pharmacology , Retinoblastoma Binding Proteins , Ubiquitin-Protein Ligases
2.
Clin. transl. oncol. (Print) ; 20(7): 881-888, jul. 2018. ilus, tab, graf
Article En | IBECS | ID: ibc-173639

Purpose: Aberrant activation of MET as a result of exon 14-skipping (METex14) mutations or gene amplification is an oncogenic mechanism in non-small cell lung carcinoma (NSCLC) and a potential therapeutic target. The purpose of this study was to characterize MET alterations in a cohort of NSCLC patients treated with surgery. Methods and patients: 157 NSCLCs of various histopathologies, including pulmonary sarcomatoid carcinomas (PSC), were tested for MET alterations. METex14 mutations, MET copy number alterations and the levels of MET protein were determined by Sanger sequencing, fluorescence in situ hybridization and immunohistochemistry, respectively. Concurrent alterations of other important cancer genes and immunostaining of the downstream effector, phopho-S6, were also determined. Results: METex14 mutations and MET amplification were detected in seven tumors. MET genetic alterations were found predominantly in the lung adenocarcinoma (ADC) and PSC histopathologies. High levels of MET protein were found in most MET-amplified tumors, but not in all METex14-mutated tumors. Strong phopho-S6 staining was observed in about half of the MET-activated tumors. One tumor with METex14 exhibited concurrent ERBB2 amplification. Conclusions: MET activation, by either METex14 mutations or amplification, is characteristic of a subset of early stage NSCLCs and may coexist with ERBB2 amplification. This may have potential therapeutic implications. The presence of METex14 mutations was associated with low levels of MET protein, which may limit the use of total MET immunostaining as a marker for preselecting patients for MET-targeted therapies


No disponible


Humans , Male , Female , Adult , Middle Aged , Aged , Aged, 80 and over , Carcinoma, Non-Small-Cell Lung/genetics , Lung Neoplasms/genetics , Proto-Oncogene Proteins c-met/genetics , Genetic Testing/methods , Gene Amplification/genetics , Mutation/genetics , Carcinoma, Squamous Cell/pathology , Carcinoma, Large Cell/pathology
3.
Clin Transl Oncol ; 20(7): 881-888, 2018 Jul.
Article En | MEDLINE | ID: mdl-29139039

PURPOSE: Aberrant activation of MET as a result of exon 14-skipping (METex14) mutations or gene amplification is an oncogenic mechanism in non-small cell lung carcinoma (NSCLC) and a potential therapeutic target. The purpose of this study was to characterize MET alterations in a cohort of NSCLC patients treated with surgery. METHODS AND PATIENTS: 157 NSCLCs of various histopathologies, including pulmonary sarcomatoid carcinomas (PSC), were tested for MET alterations. METex14 mutations, MET copy number alterations and the levels of MET protein were determined by Sanger sequencing, fluorescence in situ hybridization and immunohistochemistry, respectively. Concurrent alterations of other important cancer genes and immunostaining of the downstream effector, phopho-S6, were also determined. RESULTS: METex14 mutations and MET amplification were detected in seven tumors. MET genetic alterations were found predominantly in the lung adenocarcinoma (ADC) and PSC histopathologies. High levels of MET protein were found in most MET-amplified tumors, but not in all METex14-mutated tumors. Strong phopho-S6 staining was observed in about half of the MET-activated tumors. One tumor with METex14 exhibited concurrent ERBB2 amplification. CONCLUSIONS: MET activation, by either METex14 mutations or amplification, is characteristic of a subset of early stage NSCLCs and may coexist with ERBB2 amplification. This may have potential therapeutic implications. The presence of METex14 mutations was associated with low levels of MET protein, which may limit the use of total MET immunostaining as a marker for preselecting patients for MET-targeted therapies.


Biomarkers, Tumor/genetics , Carcinoma, Non-Small-Cell Lung/genetics , Gene Amplification , Genetic Testing , Lung Neoplasms/genetics , Mutation , Proto-Oncogene Proteins c-met/genetics , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Adult , Aged , Aged, 80 and over , Biomarkers, Tumor/metabolism , Carcinoma, Large Cell/genetics , Carcinoma, Large Cell/pathology , Carcinoma, Non-Small-Cell Lung/pathology , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/pathology , Exons , Female , Follow-Up Studies , Humans , Lung Neoplasms/pathology , Male , Middle Aged , Prognosis , Proto-Oncogene Proteins c-met/metabolism , Receptor, ErbB-2/genetics , Receptor, ErbB-2/metabolism
4.
Oncogene ; 36(9): 1287-1296, 2017 03 02.
Article En | MEDLINE | ID: mdl-27593925

Components of the SWI/SNF chromatin remodeling complex, including BRG1 (also SMARCA4), are inactivated in cancer. Among other functions, SWI/SNF orchestrates the response to retinoid acid (RA) and glucocorticoids (GC) involving downregulation of MYC. The epigenetic drugs SAHA and azacytidine, as well as RA and GC, are currently being used to treat some malignancies but their therapeutic potential in lung cancer is not well established. Here we aimed to determine the possible therapeutic effects of azacytidine and SAHA (A/S) alone or in combination with GC plus RA (GC/RA) in lung cancers with either BRG1 inactivation or MYC amplification. In vitro, responses to GC/RA treatment were more effective in MYC-amplified cells. These effects were mediated by BRG1 and involved a reprogramming towards prodifferentiation gene expression signatures and downregulation of MYC. In MYC-amplified cells, administration of GC/RA enhanced the cell growth inhibitory effects of A/S which, in turn, accentuated the prodifferentiation features promoted by GC/RA. Finally, these treatments improved overall survival of mice orthotopically implanted with MYC-amplified, but not BRG1-mutant, cells and reduced tumor cell viability and proliferation. We propose that the combination of epigenetic treatments with retinoids and corticoids of MYC-driven lung tumors constitute a strategy for therapeutic intervention in this otherwise incurable disease.


Azacitidine/pharmacology , Drug Resistance, Neoplasm/drug effects , Glucocorticoids/pharmacology , Hydroxamic Acids/pharmacology , Lung Neoplasms/drug therapy , Proto-Oncogene Proteins c-myc/metabolism , Tretinoin/pharmacology , Animals , Antimetabolites, Antineoplastic/pharmacology , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Cell Proliferation/drug effects , DNA Helicases/genetics , DNA Methylation , Histone Deacetylase Inhibitors/pharmacology , Humans , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Male , Mice , Mice, Nude , Mutation/genetics , Nuclear Proteins/genetics , Proto-Oncogene Proteins c-myc/genetics , Transcription Factors/genetics , Tumor Cells, Cultured , Vorinostat , Xenograft Model Antitumor Assays
5.
Oncogene ; 35(39): 5179-90, 2016 09 29.
Article En | MEDLINE | ID: mdl-26996667

Neuroblastoma (NB) is a neoplasm of the sympathetic nervous system, and is the most common solid tumor of infancy. NBs are very heterogeneous, with a clinical course ranging from spontaneous regression to resistance to all current forms of treatment. High-risk patients need intense chemotherapy, and only 30-40% will be cured. Relapsed or metastatic tumors acquire multi-drug resistance, raising the need for alternative treatments. Owing to the diverse mechanisms that are responsible of NB chemoresistance, we aimed to target epigenetic factors that control multiple pathways to bypass therapy resistance. We found that the SWI/SNF-related, matrix-associated, actin-dependent regulator of chromatin, subfamily a, member 4 (SMARCA4/BRG1) was consistently upregulated in advanced stages of NB, with high BRG1 levels being indicative of poor outcome. Loss-of-function experiments in vitro and in vivo showed that BRG1 is essential for the proliferation of NB cells. Furthermore, whole-genome transcriptome analysis revealed that BRG1 controls the expression of key elements of oncogenic pathways such as PI3K/AKT and BCL2, which offers a promising new combination therapy for high-risk NB.


Cell Survival/genetics , DNA Helicases/genetics , Neuroblastoma/genetics , Nuclear Proteins/genetics , Transcription Factors/genetics , Transcriptome/genetics , Cell Death/genetics , Cell Line, Tumor , Cell Proliferation/genetics , Female , Gene Expression Regulation, Neoplastic , Humans , Male , Neuroblastoma/pathology , Phosphatidylinositol 3-Kinases/genetics , Proto-Oncogene Proteins c-bcl-2/genetics , Signal Transduction/genetics
6.
Oncogene ; 33(21): 2681-9, 2014 May 22.
Article En | MEDLINE | ID: mdl-23752187

Our rapidly growing knowledge about cancer genetics attests to the widespread occurrence of alterations at genes encoding different components of the SWI/SNF complex. This reveals an important new feature that sustains cancer development: the blockade of chromatin remodeling. Here, we provide an overview of our current knowledge on the gene alterations of chromatin-remodeling factors, and how they relate to cancer and human developmental diseases. We also consider the functional repercussions, particularly how the inactivation of the SWI/SNF complex impairs the appropriate cell response to nuclear receptor signaling, which, in turn, prevents cell differentiation and sustains cell growth independently of the environment.


Cell Differentiation , Chromosomal Proteins, Non-Histone/genetics , Neoplasms/genetics , Transcription Factors/genetics , Animals , Child Development Disorders, Pervasive/genetics , Chromatin Assembly and Disassembly , Gene Expression Regulation, Neoplastic , Germ-Line Mutation , Humans , Receptors, Cytoplasmic and Nuclear/physiology , Signal Transduction
7.
Eur Respir J ; 37(1): 136-42, 2011 Jan.
Article En | MEDLINE | ID: mdl-20817702

The objective of the present study was to elaborate a survival model that integrates anatomic factors, according to the 2010 seventh edition of the tumour, node and metastasis (TNM) staging system, with clinical and molecular factors. Pathologic TNM descriptors (group A), clinical variables (group B), laboratory parameters (group C) and molecular markers (tissue microarrays; group D) were collected from 512 early-stage nonsmall cell lung cancer (NSCLC) patients with complete resection. A multivariate analysis stepped supervised learning classification algorithm was used. The prognostic performance by groups was: areas under the receiver operating characteristic curve (C-index): 0.67 (group A), 0.65 (Group B), 0.57 (group C) and 0.65 (group D). Considering all variables together selected for each of the four groups (integrated group) the C-index was 0.74 (95% CI 0.70-0.79), with statistically significant differences compared with each isolated group (from p = 0.006 to p < 0.001). Variables with the greatest prognostic discrimination were the presence of another ipsilobar nodule and tumour size > 3 cm, followed by other anatomical and clinical factors, and molecular expressions of phosphorylated mammalian target of rapamycin (phospho-mTOR), Ki67cell proliferation index and phosphorylated acetyl-coenzyme A carboxylase. This study on early-stage NSCLC shows the benefit from integrating pathological TNM, clinical and molecular factors into a composite prognostic model. The model of the integrated group classified patients with significantly higher accuracy compared to the TNM 2010 staging.


Carcinoma, Non-Small-Cell Lung/diagnosis , Carcinoma, Non-Small-Cell Lung/genetics , Lung Neoplasms/diagnosis , Lung Neoplasms/genetics , Neoplasm Staging/methods , Aged , Algorithms , Area Under Curve , Carcinoma, Non-Small-Cell Lung/therapy , Cohort Studies , Humans , Ki-67 Antigen/biosynthesis , Lung Neoplasms/therapy , Medical Oncology/methods , Middle Aged , Neoplasm Metastasis , Probability , Prognosis , Time Factors
8.
Clin Transl Oncol ; 11(5): 263-9, 2009 May.
Article En | MEDLINE | ID: mdl-19451058

Lung cancer is the leading cause of death due to cancer in most western countries and, as tobacco consumption is not significantly decreasing worldwide, will remain so in the coming decades. Thus, in addition to preventing uptake and encouraging cessation of the smoking habit, it is important to invest in understanding the biology of this type of cancer. Of particular interest are the recent efforts directed towards characterising the entire set of gene alterations in lung cancer. The present review describes the catalogue of known genetic alterations in lung cancer, their biological role and their use in clinical management.


Lung Neoplasms/genetics , Smoking/adverse effects , Genomics , Humans
9.
Clin. transl. oncol. (Print) ; 11(5): 263-269, mayo 2009. ilus
Article En | IBECS | ID: ibc-123630

Lung cancer is the leading cause of death due to cancer in most western countries and, as tobacco consumption is not significantly decreasing worldwide, will remain so in the coming decades. Thus, in addition to preventing uptake and encouraging cessation of the smoking habit, it is important to invest in understanding the biology of this type of cancer. Of particular interest are the recent efforts directed towards characterising the entire set of gene alterations in lung cancer. The present review describes the catalogue of known genetic alterations in lung cancer, their biological role and their use in clinical management (AU)


Humans , Male , Female , Lung Neoplasms/genetics , Smoking/adverse effects , Genomics/methods , Genomics/trends , Smoking/epidemiology , Smoking/prevention & control
10.
Rev. patol. respir ; 11(4): 161-167, oct.-nov. 2008. tab
Article Es | IBECS | ID: ibc-142861

Objetivos: Evaluar un potencial sesgo de selección poblacional en un estudio de expresión proteica en carcinoma broncogénico no microcítico resecado (CBNM) y realizar un análisis de la expresión molecular global y la específica según la estirpe histológica. Población y métodos: estudio observacional, de cohorte, concurrente, de todos los pacientes con CBNM tratados quirúrgicamente en nuestro Hospital entre octubre de 1993 y septiembre de 1997. Elaboramos matrices de tejido en tumor resecado y la expresión proteica se estudió mediante inmunohistoquímica. La población se dividió entre los que se pudo efectuar estudio molecular (población A) y el resto (población B). El análisis de sesgos se realizó con el test Chi cuadrado para variables cualitativas, la T de Student para cuantitativas y el Kaplan-Meier para curvas de supervivencia. Las diferencias de expresión proteica según la histología se analizaron con el test Chi cuadrado. Resultados: 180 casos totales. En 146 existía material suficiente para el estudio (población A) y en 34 no (población B). No hay diferencias significativas entre las poblaciones excepto que los casos de la población B eran más pequeños (3,3 cm vs 4,5 cm) (p= 0,04) con menos neumonectomías (t 1,8% vs 3t,6%) (p= 0,02). La estirpe más frecuente es taepidennoide (68%), De las 32 proteínas estudiadas, 8 se expresan más en epidennoides (ciclina A, CDK6, RB, p63, survivina N, EGFR, mTORp, p53) y 3 en adenocarcinomas (survivina C, ligando FAS y Cdc6). Conclusiones: Nuestra muestra representa a la población seleccionada sin que exista un sesgo de selección inicial de la suficiente magnitud como para comprometer sus resultados. En este estudio, 11 marcadores se expresan de forma diferente en función de la estirpe histológica del CBNM (AU)


To evaluate a population’s potential selection bias in a study of protein expression in resected NSCLC and to perform an analysis of global and specific molecular expression according to histological type. Population and methods: An observational, cohort, concurrent study in all patients with NSCLC surgically treated in our hospital between October 1993 and September 1997. Tissue arrays were designed from samples of resected tissue. The study method used for the evaluation of protein expression was immunohistochemistry. Population was divided into groups: patients with a molecular study (population A) and the rest (population B). Bias analyses was performed using the Chi square test for qualitative variables and the student T test for quantitative variables; the Kaplan-Meier test was used to compare survival curves. The Chi square test was used to analyse expression dijferences in proteins depending on the histological type. Results: Of total 180 cases, 146 had enough material for molecular study (population A) and 34 did not (population B). There were no differences between both populations, with the exception that tumours in population B were smaller (3.3 cm vs 4.5 cm) (p= 0.04) and with a lower frequency of pneumonectomies (11.8% vs 31.6%) (p= 0.02). The epidermoid histological type is the most frequent (68%). Of the 32 proteins studied, 8 are more frequently expressed in epidermoid types (cyclin A. CDK6, RB, p63, survivin N, EGFR, mTORp, p53) and 3 in adenoearcinomas (survivin C, FAS ligand and Cdc6). Conclusions: Our sample is representative of the selected population with no initial selection bias large enough to compromise the results. In this study, 11I markers were expressed differently according to the histological type of the NSCLC (AU)


Humans , Carcinoma, Bronchogenic/pathology , Lung Neoplasms/pathology , Carcinoma, Non-Small-Cell Lung/pathology , Biomarkers, Tumor/analysis , Risk Factors
11.
J Pathol ; 214(3): 347-56, 2008 Feb.
Article En | MEDLINE | ID: mdl-17992665

The development of targeted therapies creates a need to discriminate tumours accurately by their histological and genetic characteristics. Here, we aim to identify gene expression profiles and single markers that recapitulate the pathological and genetic background of non-small cell lung cancer (NSCLC). We performed cDNA microarray analysis on a series of 69 NSCLCs, with known mutation status for important genes, and six normal lung tissues. Unsupervised cluster analysis segregated normal lungs from lung tumours and lung tumours according to their histopathology and the presence of EGFR mutations. Several transcripts were highly overexpressed (by approximately 20 times) in squamous cell carcinomas (SCCs) relative to adenocarcinomas (ACs) and confirmed by immunohistochemistry in an independent cohort of 75 lung tumours. Expression of 13 genes constituted the most prominent hallmarks of EGFR-mutant tumours, including increased levels of proline dehydrogenase (PRODH) and down-regulation of X-box binding protein 1 (XBP1). No genes were differentially expressed, with a fold change >or= 4 or

Carcinoma, Non-Small-Cell Lung/genetics , Gene Expression Profiling , Genetic Markers , Lung Neoplasms/genetics , Oligonucleotide Array Sequence Analysis , Adenocarcinoma/genetics , Carcinoma, Squamous Cell/genetics , Case-Control Studies , Class I Phosphatidylinositol 3-Kinases , Cluster Analysis , ErbB Receptors/genetics , Gene Amplification , Genes, ras , Humans , Immunohistochemistry , In Situ Hybridization, Fluorescence , Mutation , Phosphatidylinositol 3-Kinases/genetics
12.
Oncogene ; 26(57): 7825-32, 2007 Dec 13.
Article En | MEDLINE | ID: mdl-17599048

Germline LKB1 mutations are responsible for Peutz-Jeghers syndrome (PJS). Tumors at several locations frequently arise in these patients, confirming that LKB1 is linked to cancer predisposition and is therefore a bona fide tumor-suppressor gene. In humans, the LKB1 gene is located in the short arm of chromosome 19, which is frequently deleted in many tumors of sporadic origin. However, LKB1 alterations in tumors other than those of PJS are rarely reported. Notably, this is not the case for non-small-cell lung cancer, where nearly half of the tumors harbor somatic and homozygous inactivating mutations in LKB1. The present review considers the frequency and pattern of LKB1 gene mutations in sporadic cancers of various origins, and the role of the encoded protein in cancer development.


Lung Neoplasms/genetics , Mutation , Peutz-Jeghers Syndrome/genetics , Protein Serine-Threonine Kinases/genetics , AMP-Activated Protein Kinase Kinases , Genetic Predisposition to Disease , Humans , Protein Serine-Threonine Kinases/physiology
13.
Oncogene ; 26(11): 1616-25, 2007 Mar 08.
Article En | MEDLINE | ID: mdl-16953221

LKB1, mutated in Peutz-Jeghers and in sporadic lung tumours, phosphorylates a group of protein kinases named AMP-activated protein kinase (AMPK)-related kinases. Among them is included the AMPK, a sensor of cellular energy status. To investigate the relevance of LKB1 in lung carcinogenesis, we study several lung cancer cells with and without LKB1-inactivating mutations. We report that LKB1-mutant cells are deficient for AMPK activity and refractory to mTOR inhibition upon glucose depletion but not growth-factor deprivation. The requirement for wild-type LKB1 to properly activate AMPK is further demonstrated in genetically modified cancer cells. In addition, LKB1-deficient lung primary tumours had diminished AMPK activity, assessed by complete absence or low level of phosphorylation of its critical substrate, acetyl-CoA carboxylase. We also demonstrate that LKB1 wild-type cells are more resistant to cell death upon glucose withdrawal than their mutant counterparts. Finally, modulation of AMPK activity did not affect PI3K/AKT signalling, an advantage for the potential use of AMPK as a target for cancer therapy in LKB1 wild-type tumours. Thus, sustained abrogation of cell energetic checkpoint control, through alterations at key genes, appear to be an obligatory step in the development of some lung tumours.


Cell Survival , Lung Neoplasms/enzymology , Multienzyme Complexes/metabolism , Protein Kinases/metabolism , Protein Serine-Threonine Kinases/metabolism , Signal Transduction , AMP-Activated Protein Kinase Kinases , AMP-Activated Protein Kinases , Cell Division , Humans , Immunohistochemistry , Lung Neoplasms/pathology , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins c-akt/metabolism , TOR Serine-Threonine Kinases
14.
Cancer Lett ; 230(1): 141-8, 2005 Dec 08.
Article En | MEDLINE | ID: mdl-16253770

In spite of much effort, no good markers have yet been found for predicting prognosis or response to therapy in advanced head and neck squamous cell carcinoma (HNSCCs) patients. beta-catenin, a protein involved in the cytoskeleton, cell-cell adhesion and gene transcription, is a factor associated with tumour progression. Recently, an interaction has been reported between beta-catenin, and NF-kappaB coupled with an inverse association of beta-catenin, and FAS (CD95/APO-1) protein expression in breast and colorectal tumours. To confirm these observations and to test their clinical impact in HNSCCs we have evaluated the expression of beta-catenin, NF-kappaB and FAS proteins. We used tissue microarrays to simultaneously analyse the levels of these proteins immunohistochemically in 118 HNSCCs. Among the 113 tumours evaluable for beta-catenin, increased and decreased levels were detected in 41 (36%) and 62 (55%) of the tumours, respectively. beta-catenin, protein staining was mainly membranous but 10 tumours (9%) showed the clear presence of protein in the cytoplasm, and none in the nucleus. Moreover, 81% of the tumours had decreased FAS protein expression, indicating that loss of FAS protein is a common feature of HNSCCs. Abnormal or nuclear NF-kappaB staining was observed in 24% of the tumours. No association was detected between the expression levels of the proteins evaluated. Regarding clinical associations, tumours from the hypopharynx had significantly lower levels of beta-catenin expression than those from other locations (P<0.05). Moreover, our data revealed that patients whose tumours had low levels of beta-catenin protein expression had decreased survival probability (24.8 months vs. NR, P=0.03) and reduced response to therapy (15.4 vs. 43 months; P=0.01) compared with patients whose tumours had high levels of beta-catenin. Taken together, our observations indicate that beta-catenin, NF-kappaB and FAS expression are independent events during HNSCC development and that levels of beta-catenin protein may identify subsets of advanced HNSCCs patients with different prognosis and response to therapy capabilities.


Carcinoma, Squamous Cell/physiopathology , Head and Neck Neoplasms/physiopathology , NF-kappa B/biosynthesis , Receptors, Tumor Necrosis Factor/biosynthesis , beta Catenin/biosynthesis , Biomarkers, Tumor/analysis , Gene Expression Profiling , Humans , Immunohistochemistry , NF-kappa B/analysis , Oligonucleotide Array Sequence Analysis , Prognosis , Receptors, Tumor Necrosis Factor/analysis , Survival Analysis , beta Catenin/analysis , fas Receptor
15.
Cancer ; 92(6): 1525-30, 2001 Sep 15.
Article En | MEDLINE | ID: mdl-11745231

BACKGROUND: The majority of lung carcinoma cases occur in current or former smokers. K-ras gene mutations are common in lung adenocarcinoma and have been associated with cigarette smoking, asbestos exposure, and female gender. METHODS: In the current study, the authors examined the contribution of cigarette smoking to K-ras gene mutations in patients with primary lung adenocarcinoma. Smoking histories were obtained from 106 prospectively enrolled patients with primary adenocarcinoma of the lung. RESULTS: K-ras mutations were detected in the primary tumor using an allele-specific ligation assay. Ninety-two of the 106 patients (87%) with lung adenocarcinoma were smokers. Nonsmokers with this tumor were more likely to be women (11 of 14; 79%), whereas the majority of smokers (57%) were men. K-ras mutations were detected in 40 of 106 tumors (38%) and were significantly more common in smokers compared with nonsmokers (43% vs. 0%; P = 0.001). CONCLUSIONS: The results of the current study confirm and extend previous observations that smokers with adenocarcinoma of the lung are more likely to have K-ras mutant tumors compared with nonsmokers. The strong link between cigarette smoking and K-ras mutations in adenocarcinoma of the lung supports the role of specific tobacco carcinogens in the etiology of this malignancy.


Adenocarcinoma/immunology , Genes, ras/immunology , Lung Neoplasms/immunology , Mutation , Smoking/adverse effects , Aged , Female , Humans , Male , Prospective Studies , Sex Factors
16.
Cancer Res ; 61(19): 7015-9, 2001 Oct 01.
Article En | MEDLINE | ID: mdl-11585726

Mitochondrial DNA (mtDNA) mutations scattered through coding and noncoding regions have been reported in cancer. The mechanisms that generate such mutations and the importance of mtDNA mutations in tumor development are still not clear. Here we present the identification of a specific and highly polymorphic homopolymeric C stretch (D310), located within the displacement (D) loop, as a mutational hotspot in primary tumors. Twenty-two % of the 247 primary tumors analyzed harbored somatic deletions/insertions at this mononucleotide repeat. Moreover, these alterations were also present in head and neck preneoplastic lesions. We further characterized the D310 variants that appeared in the lung and head and neck tumors. Most of the somatic alterations found in tumors showed deletion/insertions of 1- or 2-bp generating D310 variants identical to constitutive polymorphisms described previously. Sequencing analysis of individual clones from lymphocytes revealed that patients with D310 mutations in the tumors had statistically significant higher levels of D310 heteroplasmy (more than one length variant) in the lymphocyte mtDNA as compared with the patients without D310 mutations in the tumor mtDNA. On the basis of our observations, we propose a model in which D310 alterations are already present in normal cells and achieve homoplasmy in the tumor through a restriction/amplification event attributable to random genetic drift and clonal expansion.


DNA, Mitochondrial/genetics , Microsatellite Repeats/genetics , Neoplasms/genetics , Carcinoma, Squamous Cell/blood , Carcinoma, Squamous Cell/genetics , Female , Germ-Line Mutation , Head and Neck Neoplasms/blood , Head and Neck Neoplasms/genetics , Humans , Lung Neoplasms/genetics , Lymphocytes/physiology , Male , Neoplasms/blood , Polymorphism, Genetic , Precancerous Conditions/blood , Precancerous Conditions/genetics , Sequence Analysis, DNA
17.
Cancer Res ; 61(20): 7623-6, 2001 Oct 15.
Article En | MEDLINE | ID: mdl-11606403

To determine the frequency and distribution of mitochondrial DNA mutations in breast cancer, 18 primary breast tumors were analyzed by direct sequencing. Twelve somatic mutations not present in matched lymphocytes and normal breast tissues were detected in 11 of the tumors screened (61%). Of these mutations, five (42%) were deletions or insertions in a homopolymeric C-stretch between nucleotides 303-315 (D310) within the D-loop. The remaining seven mutations (58%) were single-base substitutions in the coding (ND1, ND4, ND5, and cytochrome b genes) or noncoding regions (D-loop) of the mitochondrial genome. In three cases (25%), the mutations detected in coding regions led to amino acid substitutions in the protein sequence. We then screened an additional 46 primary breast tumors with a rapid PCR-based assay to identify poly-C alterations in D310, and we found seven more cancers with alterations. Using D310 mutations as clonal marker, we detected identical changes in five of five matched fine-needle aspirates and in four of four metastases-positive lymph nodes. The high frequency of D310 alterations in primary breast cancer combined with the high sensitivity of the PCR-based assays provides a new molecular tool for cancer detection.


Breast Neoplasms, Male/genetics , Breast Neoplasms/genetics , DNA, Mitochondrial/genetics , Mutation , Biopsy, Needle , Breast Neoplasms/pathology , Breast Neoplasms, Male/pathology , Female , Genetic Markers/genetics , Humans , Male
18.
Clin Cancer Res ; 7(9): 2727-30, 2001 Sep.
Article En | MEDLINE | ID: mdl-11555585

Novel approaches for the early detection and management of prostate cancer are urgently needed. Clonal genetic alterations have been used as targets for the detection of neoplastic cells in bodily fluids from many cancer types. A similar strategy for molecular diagnosis of prostate cancer requires a common and/or early genetic alteration as a specific target for neoplastic prostate cells. Hypermethylation of regulatory sequences at the glutathione S-transferase pi (GSTP1) gene locus is found in the majority (>90%) of primary prostate carcinomas, but not in normal prostatic tissue or other normal tissues. We hypothesized that urine from prostate cancer patients might contain shed neoplastic cells or debris amenable to DNA analysis. Matched specimens of primary tumor, peripheral blood lymphocytes (normal control), and simple voided urine were collected from 28 patients with prostate cancer of a clinical stage amenable to cure. Genomic DNA was isolated from the samples, and the methylation status of GSTP1 was examined in a blinded manner using methylation-specific PCR. Decoding of the results revealed that 22 of 28 (79%) prostate tumors were positive for GSTP1 methylation. In 6 of 22 (27%) cases, the corresponding urine-sediment DNA was positive for GSTP1 methylation, indicating the presence of neoplastic DNA in the urine. Furthermore, there was no case where urine-sediment DNA harbored methylation when the corresponding tumor was negative. Although we only detected GSTP1 methylation in under one-third of voided urine samples, we have demonstrated that molecular diagnosis of prostate neoplasia in urine is feasible. Larger studies focusing on carcinoma size, location in the prostate, and urine collection techniques, as well as more sensitive technology, may lead to the useful application of GSTP1 hypermethylation in prostate cancer diagnosis and management.


DNA Methylation , Glutathione Transferase/genetics , Isoenzymes/genetics , Prostatic Neoplasms/urine , DNA, Neoplasm/genetics , DNA, Neoplasm/metabolism , DNA, Neoplasm/urine , Glutathione S-Transferase pi , Glutathione Transferase/urine , Humans , Isoenzymes/urine , Male , Polymerase Chain Reaction , Prostatic Neoplasms/diagnosis , Prostatic Neoplasms/enzymology
19.
Clin Cancer Res ; 7(7): 1963-8, 2001 Jul.
Article En | MEDLINE | ID: mdl-11448911

Genetic alterations at critical chromosome loci have been shown to be predictors of the progression of oral premalignancy-to-invasive cancer. We obtained a unique group of oral biopsies, initially collected during a prospective study designed to test the ability of OraTest (toluidine blue), to identify recurrent oral neoplastic lesions in patients with definite therapy for head and neck or upper aerodigestive tract (UADT) cancer. A total of 46 cases, including 13 squamous cell carcinoma (SCC), 11 carcinoma-in situ or dysplasia, and 22 morphologically normal oral biopsies, were analyzed for loss of heterozygosity (LOH) at 9p21, 3p21, and 17p13(TP53) by microsatellite analysis. LOH at one or more tested markers in at least one biopsy was detected in 76% (35 of 46) cases. All of the SCC and carcinoma-in situ cases showed LOH, and, strikingly, more than one-half (69%, 13 of 22) of morphologically normal epithelia also harbored LOH in at least one tested marker. The most frequent LOH was found on chromosome 9p21 (69%, 31 of 45). LOH was observed at 3p21, 17p13(TP53), or in multiple chromosomal arms significantly more often in SCC than in normal epithelia. In the majority of cases, two oral biopsies, one from an OraTest-staining positive area and another from a negative area adjacent to the stain, were collected. Among 25 LOH positive cases with two biopsies, identical allelic losses were confirmed between stained and nonstained biopsies in 16 cases. In the remaining nine cases with discordant LOH patterns between two biopsies, eight cases showed LOH at more genetic loci in OraTest-stained areas. Our data confirm that clonal genetic alterations, especially 9p21 deletion, are often present in the oral epithelia of patients with previous UADT malignancy and, combined with previous studies, suggest that genetic analysis will help stratify patients at risk of developing a secondary oral cancer. In addition to detecting cancer, our study suggests that OraTest can detect clinically occult lesions in the progression pathway to oral cancer.


Carcinoma in Situ/genetics , Carcinoma, Squamous Cell/genetics , Head and Neck Neoplasms/genetics , Loss of Heterozygosity , Adult , Aged , Biopsy , Carcinoma in Situ/pathology , Carcinoma, Squamous Cell/pathology , Chromosomes, Human, Pair 17/genetics , Chromosomes, Human, Pair 3/genetics , Chromosomes, Human, Pair 9/genetics , DNA, Neoplasm/genetics , Head and Neck Neoplasms/pathology , Humans , Microsatellite Repeats , Middle Aged , Mouth Mucosa/pathology , Tolonium Chloride
20.
Cancer Res ; 61(5): 2092-6, 2001 Mar 01.
Article En | MEDLINE | ID: mdl-11280771

Epidemiological studies have demonstrated a causal association between tobacco use and carcinoma of the lung, and some genetic targets of the carcinogens in cigarette smoke have been defined recently. We further examined the effect of cigarette smoking on the frequency of allelic losses on chromosome 9p21 and the incidence of p16 inactivation. Chromosomal loss at 9p21-24 was determined by microsatellite analysis using 14 markers in 47 patients with non-small cell lung cancer. In addition, p16 gene inactivation was determined by DNA sequence analysis, methylation-specific PCR, and immunohistochemistry. Tumors from a group of nonsmokers (n = 14) were compared with tumors from a group of smokers (n = 33) matched for cell type, tumor stage, and gender. Allelic loss encompassing the p16 locus was present significantly (P = 0.01) more often in smokers (23 of 33 smokers, 70%) than in nonsmokers (4 of 14 nonsmokers, 28%). No significant differences in the frequency of p16 inactivation were observed between smokers and nonsmokers (45% versus 36%). However, homozygous deletion of the p16 gene locus and point mutation of p16 gene were only observed in tumors from smokers, whereas the p16 gene was inactivated in tumors from nonsmokers only through promoter hypermethylation. Thus, inactivation of the p16 gene is a common event in all non-small cell lung cancer, but the mechanism of gene alteration differs between smokers and nonsmokers. The significant link between tobacco and loss of the p16 locus identifies additional genetic targets of smoking in the pathogenesis of lung cancer.


Carcinoma, Non-Small-Cell Lung/genetics , Chromosomes, Human, Pair 9 , Genes, p16/genetics , Lung Neoplasms/genetics , Smoking/genetics , Aged , Carcinoma, Non-Small-Cell Lung/etiology , Chromosome Deletion , Cyclin-Dependent Kinase Inhibitor p16/biosynthesis , DNA Methylation , Female , Gene Expression Regulation, Neoplastic , Gene Silencing , Humans , Immunohistochemistry , Loss of Heterozygosity , Lung Neoplasms/etiology , Male , Microsatellite Repeats/genetics , Point Mutation , Smoking/adverse effects
...