Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Nat Genet ; 45(12): 1483-6, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24185511

ABSTRACT

The diagnosed incidence of small intestine neuroendocrine tumors (SI-NETs) is increasing, and the underlying genomic mechanisms have not yet been defined. Using exome- and genome-sequence analysis of SI-NETs, we identified recurrent somatic mutations and deletions in CDKN1B, the cyclin-dependent kinase inhibitor gene, which encodes p27. We observed frameshift mutations of CDKN1B in 14 of 180 SI-NETs, and we detected hemizygous deletions encompassing CDKN1B in 7 out of 50 SI-NETs, nominating p27 as a tumor suppressor and implicating cell cycle dysregulation in the etiology of SI-NETs.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p27/genetics , Intestinal Neoplasms/genetics , Mutation , Neuroendocrine Tumors/genetics , Cell Cycle/genetics , Cohort Studies , Genes, Tumor Suppressor , Genetic Predisposition to Disease , Humans , Intestinal Neoplasms/epidemiology , Intestinal Neoplasms/pathology , Intestine, Small/pathology , Neuroendocrine Tumors/epidemiology , Neuroendocrine Tumors/pathology , Sequence Analysis, DNA
2.
Sci Transl Med ; 3(99): 99ra86, 2011 Sep 07.
Article in English | MEDLINE | ID: mdl-21900593

ABSTRACT

Cetuximab, an antibody directed against the epidermal growth factor receptor, is an effective clinical therapy for patients with colorectal, head and neck, and non-small cell lung cancer, particularly for those with KRAS and BRAF wild-type cancers. Treatment in all patients is limited eventually by the development of acquired resistance, but little is known about the underlying mechanism. Here, we show that activation of ERBB2 signaling in cell lines, either through ERBB2 amplification or through heregulin up-regulation, leads to persistent extracellular signal-regulated kinase 1/2 signaling and consequently to cetuximab resistance. Inhibition of ERBB2 or disruption of ERBB2/ERBB3 heterodimerization restores cetuximab sensitivity in vitro and in vivo. A subset of colorectal cancer patients who exhibit either de novo or acquired resistance to cetuximab-based therapy has ERBB2 amplification or high levels of circulating heregulin. Collectively, these findings identify two distinct resistance mechanisms, both of which promote aberrant ERBB2 signaling, that mediate cetuximab resistance. Moreover, these results suggest that ERBB2 inhibitors, in combination with cetuximab, represent a rational therapeutic strategy that should be assessed in patients with cetuximab-resistant cancers.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antineoplastic Agents/therapeutic use , Drug Resistance, Neoplasm , ErbB Receptors/metabolism , Neoplasms/drug therapy , Receptor, ErbB-2/metabolism , Signal Transduction/physiology , Animals , Antibodies, Monoclonal, Humanized , Cell Line, Tumor , Cetuximab , ErbB Receptors/genetics , Humans , Mice , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/genetics , Mitogen-Activated Protein Kinase 3/metabolism , Neoplasm Transplantation , Neoplasms/metabolism , Neuregulin-1/genetics , Neuregulin-1/metabolism , Receptor, ErbB-2/genetics , Transplantation, Heterologous
3.
Proc Natl Acad Sci U S A ; 107(34): 15157-62, 2010 Aug 24.
Article in English | MEDLINE | ID: mdl-20696899

ABSTRACT

Surprisingly few pathways signal between cells, raising questions about mechanisms for tissue-specific responses. In particular, Wnt ligands signal in many mammalian tissues, including the intestinal epithelium, where constitutive signaling causes cancer. Genome-wide analysis of DNA cis-regulatory regions bound by the intestine-restricted transcription factor CDX2 in colonic cells uncovered highly significant overrepresentation of sequences that bind TCF4, a transcriptional effector of intestinal Wnt signaling. Chromatin immunoprecipitation confirmed TCF4 occupancy at most such sites and co-occupancy of CDX2 and TCF4 across short distances. A region spanning the single nucleotide polymorphism rs6983267, which lies within a MYC enhancer and confers colorectal cancer risk in humans, represented one of many co-occupied sites. Co-occupancy correlated with intestine-specific gene expression and CDX2 loss reduced TCF4 binding. These results implicate CDX2 in directing TCF4 binding in intestinal cells. Co-occupancy of regulatory regions by signal-effector and tissue-restricted transcription factors may represent a general mechanism for ubiquitous signaling pathways to achieve tissue-specific outcomes.


Subject(s)
Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Homeodomain Proteins/metabolism , Intestinal Mucosa/metabolism , Transcription Factors/metabolism , Base Sequence , Binding Sites/genetics , CDX2 Transcription Factor , Caco-2 Cells , Colonic Neoplasms/genetics , Colonic Neoplasms/metabolism , Genetic Complementation Test , Homeodomain Proteins/genetics , Humans , Molecular Sequence Data , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Neoplasm/genetics , RNA, Neoplasm/metabolism , Regulatory Sequences, Nucleic Acid , Signal Transduction , Transcription Factor 4 , Wnt Proteins/metabolism
4.
PLoS One ; 4(11): e7887, 2009 Nov 18.
Article in English | MEDLINE | ID: mdl-19924296

ABSTRACT

BACKGROUND: Detection of critical cancer gene mutations in clinical tumor specimens may predict patient outcomes and inform treatment options; however, high-throughput mutation profiling remains underdeveloped as a diagnostic approach. We report the implementation of a genotyping and validation algorithm that enables robust tumor mutation profiling in the clinical setting. METHODOLOGY: We developed and implemented an optimized mutation profiling platform ("OncoMap") to interrogate approximately 400 mutations in 33 known oncogenes and tumor suppressors, many of which are known to predict response or resistance to targeted therapies. The performance of OncoMap was analyzed using DNA derived from both frozen and FFPE clinical material in a diverse set of cancer types. A subsequent in-depth analysis was conducted on histologically and clinically annotated pediatric gliomas. The sensitivity and specificity of OncoMap were 93.8% and 100% in fresh frozen tissue; and 89.3% and 99.4% in FFPE-derived DNA. We detected known mutations at the expected frequencies in common cancers, as well as novel mutations in adult and pediatric cancers that are likely to predict heightened response or resistance to existing or developmental cancer therapies. OncoMap profiles also support a new molecular stratification of pediatric low-grade gliomas based on BRAF mutations that may have immediate clinical impact. CONCLUSIONS: Our results demonstrate the clinical feasibility of high-throughput mutation profiling to query a large panel of "actionable" cancer gene mutations. In the future, this type of approach may be incorporated into both cancer epidemiologic studies and clinical decision making to specify the use of many targeted anticancer agents.


Subject(s)
Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Mutation , Neoplasms/metabolism , Algorithms , Codon , DNA Mutational Analysis , DNA Primers/genetics , Genotype , Glioma/genetics , Humans , Polymerase Chain Reaction , Proto-Oncogene Proteins B-raf/genetics , Reproducibility of Results , Sensitivity and Specificity
5.
Clin Cancer Res ; 15(14): 4665-73, 2009 Jul 15.
Article in English | MEDLINE | ID: mdl-19584150

ABSTRACT

PURPOSE: The homeodomain transcription factor CDX2 is a relatively specific immunohistochemical marker for gastrointestinal carcinoma. However, no study has comprehensively examined the relationship between CDX2 expression in colon cancer and clinical, pathologic, prognostic, and molecular features, including microsatellite instability and CpG island methylator phenotype (CIMP). EXPERIMENTAL DESIGN: Utilizing 621 colorectal cancers with clinical outcome and molecular data, CDX2 loss was detected in 183 (29%) tumors by immunohistochemistry. RESULTS: In multivariate logistic regression analysis, CDX2 loss was associated with female gender [odds ratio (OR), 3.32; P < 0.0001], CIMP-high (OR, 4.42; P = 0.0003), high tumor grade (OR, 2.69; P = 0.0085), stage IV disease (OR, 2.03; P = 0.019), and inversely with LINE-1 hypomethylation (for a 30% decline; OR, 0.33; P = 0.0031), p53 expression (OR, 0.55; P = 0.011), and beta-catenin activation (OR, 0.60; P = 0.037), but not with body mass index, tumor location, microsatellite instability, BRAF, KRAS, PIK3CA, p21, or cyclooxygenase-2. CDX2 loss was not independently associated with patient survival. However, the prognostic effect of CDX2 loss seemed to differ according to family history of colorectal cancer (P(interaction) = 0.0094). CDX2 loss was associated with high overall mortality (multivariate hazard ratio, 2.40; 95% CI, 1.28-4.51) among patients with a family history of colorectal cancer; no such association was present (multivariate hazard ratio, 0.97; 95% CI, 0.66-1.41) among patients without a family history of colorectal cancer. CONCLUSIONS: CDX2 loss in colorectal cancer is independently associated with female gender, CIMP-high, high-level LINE-1 methylation, high tumor grade, and advanced stage. CDX2 loss may be associated with poor prognosis among patients with a family history of colorectal cancer.


Subject(s)
Colorectal Neoplasms/pathology , Homeodomain Proteins/metabolism , Aged , CDX2 Transcription Factor , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , CpG Islands/genetics , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Cyclooxygenase 2/metabolism , DNA Methylation , Female , Humans , Immunohistochemistry , Logistic Models , Male , Microsatellite Instability , Middle Aged , Multivariate Analysis , Neoplasm Staging , Odds Ratio , Phosphatidylinositol 3-Kinases/metabolism , Prognosis , Proto-Oncogene Proteins B-raf/metabolism , Survival Analysis , Tumor Suppressor Protein p53/metabolism , beta Catenin/metabolism
6.
Genes Chromosomes Cancer ; 47(7): 591-603, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18383209

ABSTRACT

Carcinoid tumors of the small intestine are characterized by an indolent clinical course, secretion of neuropeptides, and resistance to standard cytotoxic chemotherapy. To evaluate the molecular events underlying carcinoid tumorigenesis, we used high-resolution arrays of single nucleotide polymorphisms to study chromosomal gains and losses in 24 primary and metastatic small bowel carcinoid tumors derived from 18 patients. Regions of gain or loss comprising whole chromosomes or large chromosomal regions constituted the most common class of anomalies. Loss of all or most of chromosome 18 was the commonest finding, evident in 11 of the 18 cases. Heterozygosity was also lost on chromosome arms 9p and 16q. The amplitude of observed gains was modest in comparison to those reported in some other tumor types. One focal region of recurrent gain on 14q mapped to the locus of the gene encoding the antiapoptotic protein DAD1, and immunohistochemical staining confirmed DAD1 protein expression in tumor samples. This detailed study of an uncommon neoplasm provides a basis to investigate putative oncogenes and tumor suppressor genes in intestinal carcinoid tumors.


Subject(s)
Chromosome Aberrations , Intestinal Neoplasms/genetics , Intestine, Small , Liver Neoplasms/genetics , Malignant Carcinoid Syndrome/genetics , Neoplasm Recurrence, Local/genetics , Adult , Aged , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Chromosomes, Human, Pair 9/genetics , Female , Humans , Immunoenzyme Techniques , Intestinal Neoplasms/pathology , Liver Neoplasms/secondary , Male , Membrane Proteins/genetics , Membrane Proteins/metabolism , Microsatellite Repeats , Middle Aged , Neoplasm Recurrence, Local/pathology , Polymorphism, Single Nucleotide/genetics , Prognosis
7.
J Mol Diagn ; 7(3): 422-6, 2005 Aug.
Article in English | MEDLINE | ID: mdl-16049315

ABSTRACT

We present the case of a 6-year-old male who received an allogeneic bone marrow transplant as part of treatment for acute lymphoblastic leukemia. The patient relapsed 5 months after transplantation and received additional chemotherapy. He acquired an angioinvasive fungal infection that required transfusion of granulocytes. Approximately 5 weeks after relapsing (181 days after transplant), a bone marrow specimen was taken for molecular engraftment analysis and flow cytometry to assess graft loss as well as residual disease. The engraftment results generated by the multiple short tandem repeat loci tested were inconsistent, and alleles were present at several loci that were of neither patient nor donor origin. An error in specimen identification was initially considered. Further investigation into the circumstances surrounding procurement of the patient's bone marrow aspirate revealed that the patient had received a granulocyte transfusion approximately 10 hours before the bone marrow specimen was taken. In addition, morphological and flow cytometric analyses of the same bone marrow aspirate demonstrated a significant degree of peripheral blood contamination. We determined that the unknown alleles in the bone marrow engraftment specimen were derived from the donor of the transfused granulocytes. This case illustrates that white cell transfusion can lead to erroneous bone marrow engraftment results, particularly if only one microsatellite locus is used to monitor engraftment.


Subject(s)
Bone Marrow Transplantation , DNA/analysis , Graft Survival/genetics , Leukocyte Transfusion , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Child , DNA/genetics , Flow Cytometry , Granulocytes/transplantation , Humans , Male , Microsatellite Repeats , Precursor Cell Lymphoblastic Leukemia-Lymphoma/surgery , Transplantation, Homologous
8.
Oncogene ; 24(22): 3652-6, 2005 May 19.
Article in English | MEDLINE | ID: mdl-15806175

ABSTRACT

Germline BRCA2 mutations predispose to the development of pancreatic cancer. A polymorphic stop codon in the coding region of BRCA2 (K3326X) has been described, and although an initial epidemiological study suggested it was not disease causing, subsequent studies have been inconclusive. To investigate the biological significance of the K3326X polymorphism, we determined its prevalence in patients with sporadic and familial pancreatic cancer. Using a case-control design, we studied 250 patients with resected sporadic pancreatic adenocarcinomas, 144 patients with familial pancreatic adenocarcinoma, 115 spouses of patients with pancreatic cancer, and a disease control group of 135 patients without a personal history of cancer who had undergone cholecystectomy for non-neoplastic disease. The K3326X polymorphism was detected using heteroduplex analysis and DNA sequencing. The BRCA2 K3326X polymorphism was significantly more prevalent in individuals with familial pancreatic cancer: 8/144 (5.6%) vs 3/250 controls (1.2%) (odds ratio, 4.84; 95% CI, 1.27-18.55, P<0.01). One K3326X carrier with familial pancreatic cancer carried an alteration (IVS 16-2A>G) suspected to be deleterious. Excluding this case did not alter the significance of the association (OR: 4.24, P<0.01). In contrast, there was no difference in prevalence among individuals with sporadic pancreatic cancer - 7/250 (OR: 2.37, 95% CI: 0.61-9.27). The increased prevalence of the BRCA2 K3326X polymorphism in patients with familial pancreatic cancer suggests that this polymorphism is deleterious and contributes to pancreatic cancer risk.


Subject(s)
Adenocarcinoma/epidemiology , Adenocarcinoma/genetics , Genes, BRCA2 , Pancreatic Neoplasms/epidemiology , Pancreatic Neoplasms/genetics , Adult , Aged , Case-Control Studies , Codon, Terminator , DNA Mutational Analysis , Female , Genetic Predisposition to Disease , Humans , Male , Middle Aged , Polymerase Chain Reaction , Polymorphism, Genetic , Prevalence
9.
Mol Diagn ; 7(1): 35-40, 2003.
Article in English | MEDLINE | ID: mdl-14529318

ABSTRACT

AIM: The Factor V Leiden mutation (G1691A) is a clinically important polymorphism that results in an increased risk of thrombosis. The goal of this study was to compare a temperature gradient capillary electrophoresis (TGCE) platform for the detection of Factor V gene mutations to a conventional restriction fragment length polymorphism (RFLP) assay. METHODS: Three hundred and four samples were analyzed by both TGCE and a common clinical Mnl I/RFLP assay. Concordance of results between the two assays was observed for 302/304 (99.3%) of the samples. RESULTS: All of the Leiden mutants (23/23, 100%) were identified by TGCE. Of the two discrepant results, one was caused by low peak heights in the TGCE output data and was easily rectified by the addition of a minimum peak height threshold. The second discrepancy resulted from the presence of a G-->A transition 95 bp downstream of the Leiden mutation site. This polymorphism represents a previously unreported alteration of the Factor V gene. CONCLUSIONS: The TGCE assay is less labor-intensive and has a higher throughput capacity than the Mnl I/RFLP assay. TGCE is a less specific assay than the Mnl I/RFLP assay that allows for the detection of novel polymorphisms, but also creates the need for all positive TGCE results to be confirmed by an alternate method such as sequencing. Our results demonstrate that TGCE is a highly sensitive method for mutation detection and has utility for mutation discovery analysis.


Subject(s)
Factor V/genetics , DNA Mutational Analysis , Electrophoresis, Capillary , Humans , Mutation , Polymerase Chain Reaction , Polymorphism, Restriction Fragment Length
SELECTION OF CITATIONS
SEARCH DETAIL
...