Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
1.
NPJ Precis Oncol ; 8(1): 121, 2024 May 28.
Article in English | MEDLINE | ID: mdl-38806586

ABSTRACT

Cerebrospinal fluid tumor-derived DNA (CSF-tDNA) analysis is a promising approach for monitoring the neoplastic processes of the central nervous system. We applied a lung cancer-specific sequencing panel (CAPP-Seq) to 81 CSF, blood, and tissue samples from 24 lung cancer patients who underwent lumbar puncture (LP) for suspected leptomeningeal disease (LMD). A subset of the cohort (N = 12) participated in a prospective trial of osimertinib for refractory LMD in which serial LPs were performed before and during treatment. CSF-tDNA variant allele fractions (VAFs) were significantly higher than plasma circulating tumor DNA (ctDNA) VAFs (median CSF-tDNA, 32.7%; median plasma ctDNA, 1.8%; P < 0.0001). Concentrations of tumor DNA in CSF and plasma were positively correlated (Spearman's ρ, 0.45; P = 0.03). For LMD diagnosis, cytology was 81.8% sensitive and CSF-tDNA was 91.7% sensitive. CSF-tDNA was also strongly prognostic for overall survival (HR = 7.1; P = 0.02). Among patients with progression on targeted therapy, resistance mutations, such as EGFR T790M and MET amplification, were common in peripheral blood but were rare in time-matched CSF, indicating differences in resistance mechanisms based on the anatomic compartment. In the osimertinib cohort, patients with CNS progression had increased CSF-tDNA VAFs at follow-up LP. Post-osimertinib CSF-tDNA VAF was strongly prognostic for CNS progression (HR = 6.2, P = 0.009). Detection of CSF-tDNA in lung cancer patients with suspected LMD is feasible and may have clinical utility. CSF-tDNA improves the sensitivity of LMD diagnosis, enables improved prognostication, and drives therapeutic strategies that account for spatial heterogeneity in resistance mechanisms.

2.
Cancer Treat Res Commun ; 33: 100659, 2022.
Article in English | MEDLINE | ID: mdl-36427429

ABSTRACT

INTRODUCTION: Increasing evidence suggests that consolidation durvalumab confers limited benefits for patients with stage III EGFR-mutated NSCLC. Induction or maintenance EGFR tyrosine kinase inhibitors (TKIs) added to concurrent chemoradiotherapy (CRT) may optimize definitive treatment, but there are limited data supporting an induction TKI strategy. METHODS: We evaluated the efficacy and safety of induction EGFR TKIs administered before concurrent CRT in a retrospective series of patients with unresectable locally advanced EGFR-mutated NSCLC. Circulating tumor DNA (ctDNA) analysis was performed on a patient subset using CAPP-seq and correlated with outcomes. RESULTS: Of six patients, three received erlotinib and three osimertinib as induction therapy before CRT. Induction TKIs were administered for a median of 2.5 months. The objective response rate after induction TKI was 83%. One patient had a complete response to induction erlotinib and continued erlotinib for 4 years until local progression, which was treated with CRT. Two patients completed maintenance erlotinib after CRT, and another received consolidation durvalumab. After a median follow-up of 20.5 months, only one patient developed disease recurrence, with rising ctDNA coinciding with recurrence. ctDNA remained undetectable in patients without recurrence, or low-level in a patient receiving maintenance erlotinib. Adverse events were mild and expected, and none developed pneumonitis. CONCLUSION: Induction EGFR TKI before CRT may achieve high disease control rates with promising signs of durability in patients with locally advanced EGFR-mutated NSCLC. ctDNA analysis after CRT can correlate well with clinical outcomes. Prospective studies are needed to define the role of induction EGFR TKIs in this setting.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Humans , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/genetics , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , ErbB Receptors/genetics , Retrospective Studies , Protein Kinase Inhibitors/adverse effects , Neoplasm Recurrence, Local/drug therapy , Erlotinib Hydrochloride/therapeutic use
3.
Cancer Res ; 82(16): 2838-2847, 2022 08 16.
Article in English | MEDLINE | ID: mdl-35748739

ABSTRACT

Genomic profiling of bronchoalveolar lavage (BAL) samples may be useful for tumor profiling and diagnosis in the clinic. Here, we compared tumor-derived mutations detected in BAL samples from subjects with non-small cell lung cancer (NSCLC) to those detected in matched plasma samples. Cancer Personalized Profiling by Deep Sequencing (CAPP-Seq) was used to genotype DNA purified from BAL, plasma, and tumor samples from patients with NSCLC. The characteristics of cell-free DNA (cfDNA) isolated from BAL fluid were first characterized to optimize the technical approach. Somatic mutations identified in tumor were then compared with those identified in BAL and plasma, and the potential of BAL cfDNA analysis to distinguish lung cancer patients from risk-matched controls was explored. In total, 200 biofluid and tumor samples from 38 cases and 21 controls undergoing BAL for lung cancer evaluation were profiled. More tumor variants were identified in BAL cfDNA than plasma cfDNA in all stages (P < 0.001) and in stage I to II disease only. Four of 21 controls harbored low levels of cancer-associated driver mutations in BAL cfDNA [mean variant allele frequency (VAF) = 0.5%], suggesting the presence of somatic mutations in nonmalignant airway cells. Finally, using a Random Forest model with leave-one-out cross-validation, an exploratory BAL genomic classifier identified lung cancer with 69% sensitivity and 100% specificity in this cohort and detected more cancers than BAL cytology. Detecting tumor-derived mutations by targeted sequencing of BAL cfDNA is technically feasible and appears to be more sensitive than plasma profiling. Further studies are required to define optimal diagnostic applications and clinical utility. SIGNIFICANCE: Hybrid-capture, targeted deep sequencing of lung cancer mutational burden in cell-free BAL fluid identifies more tumor-derived mutations with increased allele frequencies compared with plasma cell-free DNA. See related commentary by Rolfo et al., p. 2826.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Cell-Free Nucleic Acids , Lung Neoplasms , Biomarkers, Tumor/genetics , Bronchoalveolar Lavage Fluid , DNA, Neoplasm/genetics , Genomics , High-Throughput Nucleotide Sequencing , Humans , Lung Neoplasms/pathology , Mutation
4.
Genome Biol ; 21(1): 107, 2020 05 07.
Article in English | MEDLINE | ID: mdl-32381040

ABSTRACT

BACKGROUND: Tumors comprise a complex microenvironment of interacting malignant and stromal cell types. Much of our understanding of the tumor microenvironment comes from in vitro studies isolating the interactions between malignant cells and a single stromal cell type, often along a single pathway. RESULT: To develop a deeper understanding of the interactions between cells within human lung tumors, we perform RNA-seq profiling of flow-sorted malignant cells, endothelial cells, immune cells, fibroblasts, and bulk cells from freshly resected human primary non-small-cell lung tumors. We map the cell-specific differential expression of prognostically associated secreted factors and cell surface genes, and computationally reconstruct cross-talk between these cell types to generate a novel resource called the Lung Tumor Microenvironment Interactome (LTMI). Using this resource, we identify and validate a prognostically unfavorable influence of Gremlin-1 production by fibroblasts on proliferation of malignant lung adenocarcinoma cells. We also find a prognostically favorable association between infiltration of mast cells and less aggressive tumor cell behavior. CONCLUSION: These results illustrate the utility of the LTMI as a resource for generating hypotheses concerning tumor-microenvironment interactions that may have prognostic and therapeutic relevance.


Subject(s)
Carcinoma, Non-Small-Cell Lung/metabolism , Cell Communication , Lung Neoplasms/metabolism , Receptor Cross-Talk , Tumor Microenvironment , Adenocarcinoma/metabolism , Cell Line, Tumor , Fibroblasts/metabolism , Humans , Intercellular Signaling Peptides and Proteins/metabolism , Primary Cell Culture
5.
Gastroenterology ; 158(3): 494-505.e6, 2020 02.
Article in English | MEDLINE | ID: mdl-31711920

ABSTRACT

BACKGROUND & AIMS: Biomarkers are needed to risk stratify after chemoradiotherapy for localized esophageal cancer. These could improve identification of patients at risk for cancer progression and selection of additional therapy. METHODS: We performed deep sequencing (CAncer Personalized Profiling by deep Sequencing, [CAPP-Seq]) analyses of plasma cell-free DNA collected from 45 patients before and after chemoradiotherapy for esophageal cancer, as well as DNA from leukocytes and fixed esophageal tumor biopsy samples collected during esophagogastroduodenoscopy. Patients were treated from May 2010 through October 2015; 23 patients subsequently underwent esophagectomy, and 22 did not undergo surgery. We also sequenced DNA from blood samples from 40 healthy control individuals. We analyzed 802 regions of 607 genes for single-nucleotide variants previously associated with esophageal adenocarcinoma or squamous cell carcinoma. Patients underwent imaging analyses 6-8 weeks after chemoradiotherapy and were followed for 5 years. Our primary aim was to determine whether detection of circulating tumor DNA (ctDNA) after chemoradiotherapy is associated with risk of tumor progression (growth of local, regional, or distant tumors, detected by imaging or biopsy). RESULTS: The median proportion of tumor-derived DNA in total cell-free DNA before treatment was 0.07%, indicating that ultrasensitive assays are needed for quantification and analysis of ctDNA from localized esophageal tumors. Detection of ctDNA after chemoradiotherapy was associated with tumor progression (hazard ratio, 18.7; P < .0001), formation of distant metastases (hazard ratio, 32.1; P < .0001), and shorter disease-specific survival times (hazard ratio, 23.1; P < .0001). A higher proportion of patients with tumor progression had new mutations detected in plasma samples collected after chemoradiotherapy than patients without progression (P = .03). Detection of ctDNA after chemoradiotherapy preceded radiographic evidence of tumor progression by an average of 2.8 months. Among patients who received chemoradiotherapy without surgery, combined ctDNA and metabolic imaging analysis predicted progression in 100% of patients with tumor progression, compared with 71% for only ctDNA detection and 57% for only metabolic imaging analysis (P < .001 for comparison of either technique to combined analysis). CONCLUSIONS: In an analysis of cell-free DNA in blood samples from patients who underwent chemoradiotherapy for esophageal cancer, detection of ctDNA was associated with tumor progression, metastasis, and disease-specific survival. Analysis of ctDNA might be used to identify patients at highest risk for tumor progression.


Subject(s)
Adenocarcinoma/therapy , Biomarkers, Tumor/blood , Carcinoma, Squamous Cell/diagnosis , Chemoradiotherapy , Circulating Tumor DNA/blood , Esophageal Neoplasms/therapy , Adenocarcinoma/blood , Adenocarcinoma/diagnosis , Adenocarcinoma/mortality , Aged , Biomarkers, Tumor/isolation & purification , Biopsy , Carcinoma, Squamous Cell/blood , Carcinoma, Squamous Cell/mortality , Carcinoma, Squamous Cell/therapy , Circulating Tumor DNA/isolation & purification , Disease Progression , Esophageal Neoplasms/blood , Esophageal Neoplasms/diagnosis , Esophageal Neoplasms/mortality , Esophagus/diagnostic imaging , Esophagus/pathology , Feasibility Studies , Female , Healthy Volunteers , High-Throughput Nucleotide Sequencing , Humans , Kaplan-Meier Estimate , Male , Middle Aged , Neoplasm, Residual , Progression-Free Survival , Prospective Studies , Retrospective Studies , Risk Assessment/methods , Tomography, X-Ray Computed
6.
Nat Commun ; 7: 13898, 2016 12 23.
Article in English | MEDLINE | ID: mdl-28008921

ABSTRACT

Increasing evidence suggests that ionizing radiation therapy (RT) in combination with checkpoint immunotherapy is highly effective in treating a subset of cancers. To better understand the limited responses to this combination we analysed the genetic, microenvironmental, and immune factors in tumours derived from a transgenic breast cancer model. We identified two tumours with similar growth characteristics but different RT responses primarily due to an antitumour immune response. The combination of RT and checkpoint immunotherapy resulted in cures in the responsive but not the unresponsive tumours. Profiling the tumours revealed that the Axl receptor tyrosine kinase is overexpressed in the unresponsive tumours, and Axl knockout resulted in slower growth and increased radiosensitivity. These changes were associated with a CD8+ T-cell response, which was improved in combination with checkpoint immunotherapy. These results suggest a novel role for Axl in suppressing antigen presentation through MHCI, and enhancing cytokine release, which promotes a suppressive myeloid microenvironment.


Subject(s)
Molecular Targeted Therapy , Neoplasms/enzymology , Neoplasms/immunology , Proto-Oncogene Proteins/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Tumor Microenvironment/immunology , Tumor Microenvironment/radiation effects , Animals , Cell Line, Tumor , Cell Proliferation , Combined Modality Therapy , Cytokines/metabolism , Histocompatibility Antigens Class I/metabolism , Immunity , Immunosuppression Therapy , Immunotherapy , Mice, Inbred C57BL , Mice, Transgenic , Models, Biological , Radiation Tolerance/radiation effects , Axl Receptor Tyrosine Kinase
7.
Proteomics ; 13(10-11): 1608-23, 2013 May.
Article in English | MEDLINE | ID: mdl-23505015

ABSTRACT

Exosomes and microvesicles (MVs) are nanometer-sized, membranous vesicles secreted from many cell types into their surrounding extracellular space and into body fluids. These two classes of extracellular vesicles are regarded as a novel mechanism through which cancer cells, including virally infected cancer cells, regulate their micro-environment via the horizontal transfer of bioactive molecules: proteins, lipids, and nucleic acids (DNA, mRNA, micro-RNAs; oncogenic cargo hence often referred to as oncosomes). In head and neck cancer (HNC), exosomes and MVs have been described in Epstein Barr Virus (EBV)-associated nasopharyngeal cancer (NPC), as well as being positively correlated with oral squamous cell carcinoma (OSCC) progression. It has therefore been suggested that HNC-derived vesicles could represent a useful source for biomarker discovery, enriched in tumor antigens and cargo; hence fundamentally important for cancer progression. This current review offers an overall perspective on the roles of exosomes and MVs in HNC biology, focusing on EBV-associated NPC and OSCC. We also highlight the importance of saliva as a proximal and easily accessible bio-fluid for HNC detection, and propose that salivary vesicles might serve as an alternative model in the discovery of novel HNC biomarkers.


Subject(s)
Biomarkers, Tumor/metabolism , Cell-Derived Microparticles/metabolism , Exosomes/metabolism , Head and Neck Neoplasms/metabolism , Animals , Cell-Derived Microparticles/virology , Exosomes/virology , Head and Neck Neoplasms/virology , Herpesvirus 4, Human/genetics , Humans , MicroRNAs/metabolism , Proteome/isolation & purification , Proteome/metabolism , RNA, Viral/metabolism , Saliva/metabolism
8.
Int J Oncol ; 34(4): 963-70, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19287953

ABSTRACT

Bladder cancer is the ninth most common cancer in the world. Urothelial carcinoma (formerly known as transitional cell carcinoma) comprises the majority of bladder cancers. In order to decipher the genetic alteration leading to the carcinogenesis of urothelial cancer, we performed genome-wide allelotyping analysis using 384 microsatellite markers spanning 22 autosomes together with comparative genomic hybridization (CGH) in 21 urothelial cancer. High frequency of allelic imbalance was observed in chromosome arm 1q (61.9%), 3p (61.9%), 4q (66.67%), 8p (57.14%), 9p (76.2%) and 9q (66.67%). Allelic imbalance with frequency above average was also observed in chromosome arm 2q, 10p, 10q, 11p, 11q, 12q, 13q, 15q, 17p and 19q. The allelic imbalance of each case and fractional allelic loss for each chromosome was associated with higher tumor grade and stage (P<0.05). We have also delineated several minimal deletion regions on chromosome 3p, 4q, 8p, 9p, 9q, 11p, 13q, 16q and 17p. By CGH analysis, common chromosomal alterations included gain of 1p, 1q, 12q, 16p, 17q and 19p as well as loss of 4q and 9p in most of the cases. Our findings may provide valuable information to locate putative oncogenes and tumor suppressor genes in the carcinogenesis of bladder cancer in this locality.


Subject(s)
Allelic Imbalance , Comparative Genomic Hybridization/methods , Mutation , Urinary Bladder Neoplasms/genetics , Urinary Bladder Neoplasms/metabolism , Aged , Aged, 80 and over , Chromosome Deletion , Female , Gene Expression Profiling , Humans , Karyotyping , Loss of Heterozygosity , Male , Microsatellite Repeats , Middle Aged
10.
Cancer Res ; 65(18): 8125-33, 2005 Sep 15.
Article in English | MEDLINE | ID: mdl-16166286

ABSTRACT

Nasopharyngeal carcinoma is highly prevalent in Southern China and Southeast Asia. To unveil the molecular basis of this endemic disease, high-resolution comparative genomic hybridization arrays were used for systematic investigation of genomic abnormalities in 26 nasopharyngeal carcinoma samples. A comprehensive picture of genetic lesions associated with tumorigenesis of nasopharyngeal carcinoma was generated. Consistent chromosomal gains were frequently found on 1q, 3q, 8q, 11q, 12p, and 12q. High incidences of nonrandom losses were identified on chromosomes 3p, 9p, 11q, 14q, and 16q. In addition to previously characterized regions, we have identified several novel minimal regions of gains, including 3q27.3-28, 8q21-24, 11q13.1-13.3, and 12q13, which may harbor candidate nasopharyngeal carcinoma-associated oncogenes. In this study, gain of 11q13.1-13.3 was the most frequently detected chromosomal aberration and a 5.3-Mb amplicon was delineated at this region. Within this 11q13 amplicon, concordant amplification and overexpression of cyclin D1 (CCND1) oncogene was found in nasopharyngeal carcinoma cell lines, xenografts, and primary tumors. Knockdown of cyclin D1 by small interfering RNA in nasopharyngeal carcinoma cell lines led to significant decrease of cell proliferation. The findings suggest that cyclin D1 is a target oncogene at 11q13 in nasopharyngeal carcinoma and its activation plays a significant role in nasopharyngeal carcinoma tumorigenesis.


Subject(s)
Chromosomes, Human, Pair 11/genetics , Genes, bcl-1/genetics , Nasopharyngeal Neoplasms/genetics , Animals , Cell Line, Tumor , Chromosome Deletion , Gene Amplification , Humans , Mice , Nucleic Acid Hybridization , RNA, Small Interfering/genetics , Transfection , Transplantation, Heterologous
11.
Int J Oncol ; 26(1): 49-56, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15586224

ABSTRACT

Nasopharyngeal carcinoma (NPC) is a distinct geographical disease with high incidence in Southeast Asia. Previous CGH studies have located multiple regions of chromosomal gains and losses in NPC. To elucidate the regions of gain and amplification, a high-resolution array comparative genomic hybridization (array CGH) was applied to characterize the common amplicons in NPC cell lines and xenografts. Consistent with our previous CGH findings, frequent gains at chromosomes 1q, 3q, 7, 8, 9q, 12q and 20q were detected. High incidence of gains were identified on chromosome 3q and 12q. The findings were further confirmed by FISH analysis. Using 16 BAC clones on 3q26-28 and 6 BAC clones on 12q13, the smallest regions of gain at 3q and 12q were defined on five NPC cell lines. Chromosome 3q26.33 (RP11-510K16) and 12q13.2-q13.3 (RP11-183H16) showed the highest amplification frequency in FISH analysis with 100 and 66.7%, respectively. PIK3CA, a candidate oncogene located at 3q26.32 adjacent to this 3q amplicon was investigated by real-time quantitative reverse transcriptase-polymerase chain reaction (RT-PCR). Overexpression of this gene was found in 4 NPC cell lines and 3 xenografts. The findings implied that PIK3CA may be a putative oncogene involved in the tumorigenesis of NPC.


Subject(s)
Carcinoma/genetics , Chromosomes, Human, Pair 12 , Chromosomes, Human, Pair 3 , Nasopharyngeal Neoplasms/genetics , Phosphatidylinositol 3-Kinases/genetics , Biomarkers, Tumor/genetics , Class I Phosphatidylinositol 3-Kinases , Gene Amplification/genetics , Humans , Oligonucleotide Array Sequence Analysis , Oncogenes/genetics , Transcriptional Activation
12.
Mol Carcinog ; 38(4): 170-8, 2003 Dec.
Article in English | MEDLINE | ID: mdl-14639656

ABSTRACT

Deletion of 11q23 is a common genetic aberration in nasopharyngeal carcinoma (NPC). Multiple candidate tumor suppressor genes (TSG) were mapped to this region but few of them were investigated in NPC. TSLC1 (tumor suppressor in lung cancer) is recently reported to be a putative TSG on 11q23. This gene was found to be inactivated by promoter hypermethylation in non-small cell lung carcinoma (NSCLC), liver cancer, and breast cancer. To study the role of TSLC1 gene in NPC tumorigenesis, we screened for mutations and aberrant methylation of TSLC1 gene in 5 NPC cell lines, 3 NPC xenografts, and 38 primary NPC cases. No somatic mutations of TSLC1 were detected in the NPC samples, but a 9-bp (CCACCACCA) deletion in exon 8 was found in a primary NPC and its corresponding blood sample. Bisulfite sequencing revealed aberrant methylation of TSLC1 promoter in four NPC cell lines. Loss of TSLC1 gene expression was found in two cell lines (HK-1 and CNE-2) with dense methylation. Expression of this gene was restored in these cell lines after treatment with demethylating agent 5-aza-2'-deoxycytidine. Our results showed that silencing of TSLC1 gene expression in NPC was associated with promoter hypermethylation. Promoter hypermethylation of TSLC1 gene was further illustrated in 34.2% (13/38) of primary NPCs. No aberrant promoter methylation was found in any of the four investigated normal nasopharyngeal epithelia. Frequent epigenetic inactivation of TSLC1 gene in NPC suggested that this gene is one of the target tumor suppressor genes of this endemic cancer.


Subject(s)
Azacitidine/analogs & derivatives , DNA Methylation , Immunoglobulins , Membrane Proteins , Nasopharyngeal Neoplasms/genetics , Promoter Regions, Genetic , Proteins/genetics , Adult , Aged , Animals , Azacitidine/pharmacology , Base Sequence , Cell Adhesion Molecule-1 , Cell Adhesion Molecules , Chromosomes, Human, Pair 11/genetics , DNA Modification Methylases/antagonists & inhibitors , DNA Primers/chemistry , DNA, Neoplasm/genetics , Decitabine , Enzyme Inhibitors/pharmacology , Female , Gene Expression Regulation, Neoplastic , Gene Silencing , Genes, Tumor Suppressor , Humans , Loss of Heterozygosity , Male , Mice , Microsatellite Repeats , Middle Aged , Molecular Sequence Data , Mutation , Nasopharyngeal Neoplasms/metabolism , Nasopharynx/metabolism , Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Sequence Deletion , Transplantation, Heterologous , Tumor Cells, Cultured , Tumor Suppressor Proteins
13.
Cancer Genet Cytogenet ; 134(1): 71-6, 2002 Apr 01.
Article in English | MEDLINE | ID: mdl-11996800

ABSTRACT

Ganglioglioma is a mixed neuronal and glial tumor first described by Perkin in 1926. Because of its rare occurrence in the central nervous system, the pathogenesis of this neoplasm is still largely unknown. Previous studies of ganglioglioma mainly focused on histologic features, immunohistochemical analysis, clinical treatment, and patient outcome. Very few cytogenetic and molecular genetic studies have been reported on this neoplasm. To better understand the mechanism underlying the development of ganglioglioma, we performed comparative genomic hybridization analysis to investigate chromosomal imbalances across the entire genome in five cases of gangliogliomas. Loss of genetic material on the short arm of chromosome 9 was a common genetic alteration found in three of five cases. Overrepresentation of partial or the whole chromosome 7 was another recurrent chromosomal imbalance, confirmed by fluorescence in situ hybridization. Immunohistochemical analysis was performed; all five cases revealed no reaction or low expression for epidermal growth factor receptor antibody. Our study highlights chromosomal regions for further fine mapping and investigation of candidate tumor suppressor genes involved in the pathogenesis of ganglioglioma.


Subject(s)
Brain Neoplasms/genetics , Chromosome Aberrations , Ganglioglioma/genetics , Genome, Human , Nucleic Acid Hybridization/methods , Adolescent , Adult , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Chromosomes, Human, Pair 7/genetics , ErbB Receptors/analysis , Female , Ganglioglioma/metabolism , Ganglioglioma/pathology , Humans , Immunohistochemistry , In Situ Hybridization, Fluorescence , Male
15.
Int J Oncol ; 20(3): 467-73, 2002 Mar.
Article in English | MEDLINE | ID: mdl-11836556

ABSTRACT

We have applied the method of genomic microarray to investigate amplification of oncogenes throughout the genome of nasopharyngeal carcinoma (NPC). Array based comparative genomic hybridization (array CGH) allows simultaneous examination of 58 oncogenes commonly amplified in various human cancers. In the present study, we have examined 15 NPC samples including five cell lines, two xenografts and eight primary tumours with array CGH to reveal the particular oncogenes associated with this cancer. This is the first genome wide survey of multiple oncogene amplifications involved in the development of NPC. Non-random gene amplifications were identified for the first time in NPC on MYCL1 in 1p34.3 and on TERC and PIK3CA at 3q26.3. Other high level amplified oncogenes included NRAS, RAF1, MYB, EGFR, FGF4, EMS1, and D17S167. Highest frequencies of gain of novel oncogenes were detected on MYCL1 (66.7%), TERC (46.7%), ESR (46.7%), PIK3CA (40%), LAMC2 (33.3%), and CSE1L (33.3%).


Subject(s)
Carcinoma/genetics , Genetic Techniques , Nasopharyngeal Neoplasms/genetics , Oligonucleotide Array Sequence Analysis , Oncogenes/genetics , Chromosome Mapping , Down-Regulation , Humans , In Situ Hybridization, Fluorescence , Neoplasm Transplantation , Nucleic Acid Hybridization , Tumor Cells, Cultured , Up-Regulation
16.
Cancer Genet Cytogenet ; 132(1): 14-9, 2002 Jan 01.
Article in English | MEDLINE | ID: mdl-11801302

ABSTRACT

Pleomorphic xanthoastrocytoma (PXA) is a rare, low-grade astrocytic tumor found in the central nervous system. Histologically, the tumor is characterized by markedly pleomorphic and lipidized cells. Although most of the patients have a favorable prognosis, a small number of cases undergoing recurrence or progression to anaplastic astrocytoma were reported. Very few genetic studies have been performed on PXA because of its rarity and the pathogenesis of this neoplasm is largely unknown. In order to provide an overview of genetic alterations in PXA, we performed comparative genomic hybridization to identify chromosomal imbalances (DNA gains and losses) in three cases of PXA. Genetic imbalance was detected on at least one chromosome for each case. One case, which revealed multiple genetic alterations, showed a poor prognosis. DNA gain on chromosome 7 and loss on 8p were demonstrated in two of three cases, suggesting that the candidate gene(s) located on these regions may play a role in the development of PXA. Further studies are needed to identify the residing candidate genes that are involved in the tumorigenesis of PXA. In addition, the histopathological features and previous genetic studies on PXA are reviewed.


Subject(s)
Astrocytoma/genetics , Brain Neoplasms/genetics , Chromosome Aberrations , Adult , Astrocytoma/diagnosis , Astrocytoma/surgery , Brain Neoplasms/diagnosis , Brain Neoplasms/surgery , Child , Chromosome Deletion , DNA, Neoplasm/genetics , Gene Amplification , Humans , Male , Nucleic Acid Hybridization
SELECTION OF CITATIONS
SEARCH DETAIL
...