Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 84
Filter
1.
Oncogene ; 29(22): 3228-40, 2010 Jun 03.
Article in English | MEDLINE | ID: mdl-20305694

ABSTRACT

We have previously identified an oncogenic role of artemin (ARTN), a member of glial cell derived neurotrophic factor family of ligands, in mammary carcinoma. We herein report that ARTN is an estrogen-inducible gene. Meta-analysis of gene expression data sets showed that ARTN expression is positively correlated to estrogen receptor (ER) status in human mammary carcinoma. Furthermore, in patients with ER-positive mammary carcinoma treated with tamoxifen, high ARTN expression is significantly correlated with decreased survival. Forced expression of ARTN in ER-positive human mammary carcinoma cells increased ER transcriptional activity, promoted estrogen-independent growth and produced resistance to tamoxifen and fulvestrant in vitro and to tamoxifen in xenograft models. ARTN-stimulated resistance to tamoxifen and fulvestrant is mediated by increased BCL-2 expression. Conversely, depletion of endogenous ARTN by small-interfering RNA or functional antagonism of ARTN by antibody enhanced the efficacy of antiestrogens. Tamoxifen decreased ARTN expression in tamoxifen-sensitive mammary carcinoma cells whereas ARTN expression was increased in tamoxifen-resistant cells and not affected by tamoxifen treatment. Antibody inhibition of ARTN in tamoxifen-resistant cells improved tamoxifen sensitivity. Functional antagonism of ARTN therefore warrants consideration as an adjuvant therapy to enhance antiestrogen efficacy in ER-positive mammary carcinoma.


Subject(s)
Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Estradiol/pharmacology , Estrogen Receptor Modulators/pharmacology , Nerve Tissue Proteins/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Drug Resistance, Neoplasm , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Neoplasm Invasiveness , Nerve Tissue Proteins/antagonists & inhibitors , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/metabolism , Proto-Oncogene Proteins c-bcl-2/biosynthesis , Proto-Oncogene Proteins c-bcl-2/genetics , RNA, Small Interfering/genetics , Receptors, Estrogen/biosynthesis , Receptors, Estrogen/genetics , Receptors, Estrogen/metabolism , Tamoxifen/pharmacology , Transcription, Genetic
2.
Br J Cancer ; 100(8): 1358-64, 2009 Apr 21.
Article in English | MEDLINE | ID: mdl-19319135

ABSTRACT

We investigated common genetic variation in the entire ESR1 and EGF genes in relation to endometrial cancer risk, myometrial invasion and endometrial cancer survival. We genotyped a dense set of single-nucleotide polymorphisms (SNPs) in both genes and selected haplotype tagging SNPs (tagSNPs). The tagSNPs were genotyped in 713 Swedish endometrial cancer cases and 1567 population controls and the results incorporated into logistic regression and Cox proportional hazards models. We found five adjacent tagSNPs covering a region of 15 kb at the 5' end of ESR1 that decreased the endometrial cancer risk. The ESR1 variants did not, however, seem to affect myometrial invasion or endometrial cancer survival. For the EGF gene, no association emerged between common genetic variants and endometrial cancer risk or myometrial invasion, but we found a five-tagSNP region that covered 51 kb at the 5' end of the gene where all five tagSNPs seemed to decrease the risk of dying from endometrial cancer. One of the five tagSNPs in this region was in strong linkage disequilibrium (LD) with the untranslated A61G (rs4444903) EGF variant, earlier shown to be associated with risk for other forms of cancer.


Subject(s)
Endometrial Neoplasms/genetics , Epidermal Growth Factor/genetics , Estrogen Receptor alpha/genetics , Genetic Variation , Polymorphism, Single Nucleotide , Aged , Case-Control Studies , Endometrial Neoplasms/epidemiology , Endometrial Neoplasms/mortality , Female , Genotype , Humans , Middle Aged , Neoplasm Invasiveness/genetics , Registries , Risk Factors , Survival Analysis , Sweden
3.
Oncogene ; 26(27): 3998-4008, 2007 Jun 07.
Article in English | MEDLINE | ID: mdl-17213808

ABSTRACT

Expression of homeobox A1 (HOXA1) results in oncogenic transformation of immortalized human mammary epithelial cells with aggressive tumor formation in vivo. However, the mechanisms by which HOXA1 mediates oncogenic transformation is not well defined. To identify molecules that could potentially be involved in HOXA1-mediated oncogenic transformation, microarray analysis was utilized to characterize and compare the gene expression pattern in response to forced expression or depletion of HOXA1 in human mammary carcinoma cells. Gene expression profiling identified that genes involved in the p44/42 mitogen-activated protein (MAP) kinase activation pathway (GRB2, MAP kinase kinase (MEK1) and SDFR1) or p44/42 MAP kinase-regulated genes (IER3, EPAS1, PCNA and catalase) are downstream expression targets of HOXA1. Forced expression of HOXA1 increased GRB2 and MEK1 mRNA and protein expression and increased p44/42 MAP kinase phosphorylation, activity and Elk-1-mediated transcription. Use of a MEK1 inhibitor demonstrated that increased p44/42 MAP kinase activity is required for the HOXA1-mediated increase in cell proliferation, survival, oncogenicity and oncogenic transformation. Thus, modulation of the p44/42 MAP kinase pathway is one mechanism by which HOXA1 mediates oncogenic transformation of the human mammary epithelial cell.


Subject(s)
Homeodomain Proteins/physiology , MAP Kinase Signaling System/physiology , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 3/genetics , Transcription Factors/physiology , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation , Cell Survival/genetics , Cell Survival/physiology , Cluster Analysis , Female , Gene Expression Profiling , Homeodomain Proteins/genetics , Humans , MAP Kinase Signaling System/genetics , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , RNA Interference , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/genetics , Reverse Transcriptase Polymerase Chain Reaction , Transcription Factors/genetics , Transfection , ets-Domain Protein Elk-1/genetics , ets-Domain Protein Elk-1/physiology
4.
J Clin Endocrinol Metab ; 91(10): 3941-6, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16868059

ABSTRACT

CONTEXT: Raloxifene is a promising breast cancer prevention agent in postmenopausal women at increased risk for breast cancer. The effects of raloxifene in premenopausal women are unknown. OBJECTIVE: We evaluated the effect of raloxifene in premenopausal women at increased risk for breast cancer on bone mineral density (BMD). DESIGN: This was a phase II clinical trial. SETTING: This study was conducted at an academic medical center. PARTICIPANTS: Thirty-seven premenopausal women at increased risk for breast cancer enrolled in the trial. Thirty subjects began treatment and 27 were evaluable. INTERVENTION: Raloxifene (60 mg daily) and elemental calcium (500 mg daily) were given for 2 yr. Subjects were followed up off medications for 1 yr. MAIN OUTCOME MEASURE: The primary end point was the intrasubject percent change in BMD at 1 yr measured by dual-energy x-ray absorptiometry. RESULTS: The mean baseline lumbar spine density was 1.027 g/cm(2). Lumbar spine density decreased 2.3% at 1 yr (P < 0.00001) and 3.5% at 2 yr (P < .00001). Percent change from yr 2 to 3 was +1.4%. The mean baseline total hip bone density was 0.905 g/cm(2). Total hip density decreased 0.3% at 1 yr and 1.0% at 2 yr (P = 0.033). Percent change from yr 2 to 3 was +1.7%. CONCLUSIONS: Raloxifene use is associated with a decrease in BMD in premenopausal women at increased risk for breast cancer. The clinical significance of this decrease is unknown and is attenuated with stopping raloxifene.


Subject(s)
Bone Density/drug effects , Breast Neoplasms/prevention & control , Raloxifene Hydrochloride/pharmacology , Selective Estrogen Receptor Modulators/pharmacology , Adult , Female , Fibrinogen/analysis , Humans , Lipids/blood , Middle Aged , Premenopause , Quality of Life , Raloxifene Hydrochloride/adverse effects
7.
Singapore Med J ; 44(7): 375-9, 2003 Jul.
Article in English | MEDLINE | ID: mdl-14620732

ABSTRACT

A 40-year-old man presented with recent increase in size of a painless long-standing soft mass over the right shoulder. Magnetic resonance (MR) imaging showed a lobulated mass located within the subcutaneous layer overlying the deltoid muscle. It was homogeneously T1-hyperintense, T2-hyperintense, and non-enhancing. Signal characteristics were those of a simple lipoma. The diagnosis was confirmed on histopathological examination of the excised specimen. Musculoskeletal lipomas are discussed, with emphasis on their characteristic appearances on various imaging modalities, and illustrated by additional examples.


Subject(s)
Lipoma/diagnosis , Shoulder , Soft Tissue Neoplasms/diagnosis , Adult , Humans , Lipoma/surgery , Magnetic Resonance Imaging , Male , Soft Tissue Neoplasms/surgery
8.
Cytopathology ; 14(6): 309-13, 2003 Dec.
Article in English | MEDLINE | ID: mdl-14632727

ABSTRACT

In solid tumours the predominant genetic mechanism for oncogene activation is through amplification of genes. The HER-2 (also known as ErbB2/c-erbB2/HER-2/neu) oncogene is the most frequently amplified oncogene in breast cancer and is also commonly amplified in other forms of cancer. The HER-2 amplicon also contains other biologically relevant genes with altered copy numbers, among these genes is the topoisomerase IIalpha (TOP2A). TOP2A gene is located adjacent to the HER-2 oncogene at the chromosome location 17q12-q21 and is either amplified or deleted, with equal frequency, in almost 90% of HER-2 amplified primary breast tumours. Recent data suggest that amplification and deletion of TOP2A may account for both sensitivity and resistance to topoII-inhibitor-chemotherapy, depending on the specific genetic defect at the TOP2A locus. In this issue of the Cytopathology, Bofin et al. present preliminary evidence for high prevalance of TOP2A amplification and deletion not only in the HER-2 amplified breast tumours, but also in the primary breast tumours without the HER-2 amplification. This finding together with the concept that TOP2A gene amplification and deletion seem to account for both relative chemosensitivity and resistance to topoII-inhibitor therapy further highlights the importance of screening for TOP2A gene copy number aberrations when topoII-inhibitors are considered either alone or in combination of other chemotherapeutic drugs for the treatment of cancer patients.


Subject(s)
Breast Neoplasms/genetics , DNA Topoisomerases, Type II/genetics , Gene Amplification , Gene Deletion , Antigens, Neoplasm , Breast Neoplasms/pathology , DNA-Binding Proteins , Humans , Poly-ADP-Ribose Binding Proteins
9.
Biotechniques ; 33(3): 664-6, 668, 670, 2002 Sep.
Article in English | MEDLINE | ID: mdl-12238776

ABSTRACT

The rat aortic ring assay has been previously described as a useful ex vivo model for analyzing the biological activity of various inhibitors of angiogenesis. Rat aortic rings are exposed to antiangiogenic agents for a five-day incubation period. Then, the degree of microvessel outgrowth from the rings is analyzed and quantified. In contrast to most in vitro angiogenesis assays, the rat aortic ring model provides a unique microenvironment to evaluate the interaction of various cell types and biological factors for their influence on angiogenesis. Microarray analysis is an accepted method for the evaluation of gene expression profiles and can be used to better understand changes in gene expression that occur when rat aortic rings are exposed to a particular biological agent. Here we describe a method of using microarray technology to evaluate the modulation of gene expression in angiogenesis using the rat aortic ring assay.


Subject(s)
Angiogenesis Inhibitors/analysis , Angiogenesis Inhibitors/genetics , Gene Expression Profiling/methods , Oligonucleotide Array Sequence Analysis/methods , Angiogenesis Inhibitors/administration & dosage , Animals , Aorta, Thoracic/drug effects , Aorta, Thoracic/physiology , Endothelial Growth Factors/administration & dosage , Gene Expression Regulation , Male , Neovascularization, Physiologic/drug effects , Neovascularization, Physiologic/genetics , Rats , Rats, Sprague-Dawley , Triazoles/administration & dosage
10.
Mol Cell Biol ; 21(19): 6626-39, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11533250

ABSTRACT

To investigate the transcriptional program underlying thyroid hormone (T3)-induced cell proliferation, cDNA microarrays were used to survey the temporal expression profiles of 4,400 genes. Of 358 responsive genes identified, 88% had not previously been reported to be transcriptionally or functionally modulated by T3. Partitioning the genes into functional classes revealed the activation of multiple pathways, including glucose metabolism, biosynthesis, transcriptional regulation, protein degradation, and detoxification in T3-induced cell proliferation. Clustering the genes by temporal expression patterns provided further insight into the dynamics of T3 response pathways. Of particular significance was the finding that T3 rapidly repressed the expression of key regulators of the Wnt signaling pathway and suppressed the transcriptional downstream elements of the beta-catenin-T-cell factor complex. This was confirmed biochemically, as beta-catenin protein levels also decreased, leading to a decrease in the transcriptional activity of a beta-catenin-responsive promoter. These results indicate that T3-induced cell proliferation is accompanied by a complex coordinated transcriptional reprogramming of many genes in different pathways and that early silencing of the Wnt pathway may be critical to this event.


Subject(s)
Gene Silencing , Proto-Oncogene Proteins/antagonists & inhibitors , Signal Transduction , Trans-Activators , Triiodothyronine/pharmacology , Zebrafish Proteins , Animals , Cell Division , Cell Line , Cytoskeletal Proteins/physiology , Gene Expression Profiling , Kinetics , Oligonucleotide Array Sequence Analysis , RNA, Messenger/biosynthesis , Rats , Transcriptional Activation , Wnt Proteins , beta Catenin
11.
Blood ; 98(6): 1914-21, 2001 Sep 15.
Article in English | MEDLINE | ID: mdl-11535529

ABSTRACT

Because abnormal erythroid differentiation is the most common manifestation of the myelodysplastic syndromes (MDS), it was hypothesized that erythroid gene expression may be used to illustrate myelodysplastic transcription patterns. Ten normal bone marrow aspirates (NBM) were first analyzed using an erythroid-focused cDNA array to define steady-state transcription levels. Proliferation and differentiation gene subsets were identified by statistically significant differences between NBM and erythroleukemia gene expression. Next, cDNAs from 5 separate MDS aspirates were studied: refractory anemia, refractory anemia with ringed sideroblasts, refractory anemia with excess blasts, refractory anemia with excess blasts in transformation (RAEB-T), and RAEB-T/secondary MDS. A distinct pattern of significantly increased proliferation-associated and reduced differentiation-associated gene activity was established for MDS.


Subject(s)
Gene Expression Regulation, Neoplastic , Myelodysplastic Syndromes/genetics , Myelodysplastic Syndromes/pathology , Aged , Bone Marrow Cells/metabolism , Bone Marrow Cells/pathology , Cell Differentiation , Cell Division , Erythrocytes/cytology , Erythrocytes/metabolism , Female , Gene Expression Profiling , Humans , K562 Cells , Male , Middle Aged , Myelodysplastic Syndromes/metabolism , Oligonucleotide Array Sequence Analysis , RNA, Messenger/biosynthesis , RNA, Neoplasm/biosynthesis
12.
J Biol Chem ; 276(28): 25647-50, 2001 Jul 13.
Article in English | MEDLINE | ID: mdl-11384963

ABSTRACT

BRCA1 gene is a tumor suppressor for breast and ovarian cancers with the putative role in DNA repair and transcription. To characterize the role of BRCA1 in transcriptional regulation, we analyzed gene expression profiles of mouse embryonic stem cells deficient in BRCA1 using microarray technology. We found that loss of BRCA1 correlated with decreased expression of several groups of genes including stress response genes, cytoskeleton genes, and genes involved in protein synthesis and degradation. Previous study showed that BRCA1 is a transcriptional co-activator of p53 protein; however the majority of p53 target genes remained at the same expression levels in BRCA1 knockout cells as in the wild type cells. The only p53 target gene down-regulated with the loss of BRCA1 was 14-3-3 sigma, a major G(2)/M checkpoint control gene. Similar to cells with decreased 14-3-3 sigma activity, BRCA1-deficient cells were unable to sustain G(2)/M growth arrest after exposure to ionizing radiation. We find that BRCA1 induction of 14-3-3 sigma requires the presence of wild type p53 and can be regulated by a minimal p53 response element.


Subject(s)
Biomarkers, Tumor , Exonucleases , Genes, BRCA1 , Neoplasm Proteins , Proteins/genetics , Stem Cells/physiology , 14-3-3 Proteins , Animals , Exoribonucleases , Gene Expression Regulation, Developmental/physiology , Genes, p53 , Mice , Transcription, Genetic/physiology
13.
Cancer Res ; 61(12): 4797-808, 2001 Jun 15.
Article in English | MEDLINE | ID: mdl-11406555

ABSTRACT

To identify changes in gene expression with transformation and metastasis, we investigated differential gene expression in a squamous carcinoma model established in syngeneic mice. We used mRNA differential display (DD) to detect global differences and cDNA arrays enriched for cancer-associated genes using mRNA from primary keratinocytes, transformed Pam 212 squamous carcinoma cells, and metastases of Pam 212. After DD, 72 candidate cDNAs expressed primarily in transformed and metastatic cells were selected and cloned. Fifty-seven were detected, and 32 were confirmed to be differentially expressed by Northern blot analysis. mRNA expression profiles were also generated using a mouse cDNA array composed of 4000 elements representing known genes and expressed sequence tags plus the 57 DD candidate cDNAs detected by Northern analysis to facilitate data validation. cDNA array detected 76.9% of the differentially expressed mRNAs selected from DD and confirmed by Northern blot, whereas low-abundance mRNAs did not reach the threshold for detection by the lower-sensitivity array method. Clustering analysis of DD and array results from transformed and metastatic cells identified genes that exhibited decreased or increased expression with transformation and metastasis. Alterations in the expression of several genes detected during tumor progression were consistent with their functional activities involving growth (p21, p27, and cyclin D1), resistance and apoptosis (glutathione-S-transferase, cIAP-1, PEA-15, and Fas ligand), inflammation and angiogenesis [chemokine growth-regulated oncogene 1 (also called KC)], and signal transduction (c-Met, yes-associated protein, and syk). Strikingly, 10 of 22 genes in the cluster expressed in metastases have been associated with activation of the nuclear factor (NF)-kappaB signal pathway. The NF-kappaB-inducible cytokine Gro-1 was recently shown to promote tumor growth, metastasis, and angiogenesis of squamous cell carcinomas in vivo (Loukinova et al., Oncogene, 19: 3477-3486, 2000). The results demonstrate that early response genes related to NF-kappaB contribute to metastatic tumor progression. Comparison of cell lines and tumor tissue revealed a concordance of approximately 50% by array, and 70% for Northern-confirmed, metastasis-related genes. Functional genomic approaches comparing expression among cell lines and tumor tissue may promote a better understanding of the genes expressed by malignant and host cells during tumor progression and metastasis.


Subject(s)
Apoptosis/genetics , Carcinoma, Squamous Cell/genetics , NF-kappa B/physiology , Neovascularization, Pathologic/genetics , Signal Transduction/genetics , Skin Neoplasms/genetics , Animals , Blotting, Northern , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Cell Division/genetics , Cell Line, Transformed , Cell Transformation, Neoplastic/genetics , Cluster Analysis , Disease Progression , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Keratinocytes/cytology , Keratinocytes/physiology , Male , Mice , Mice, Inbred BALB C , NF-kappa B/biosynthesis , NF-kappa B/genetics , Neoplasm Metastasis , Oligonucleotide Array Sequence Analysis , Skin Neoplasms/metabolism , Skin Neoplasms/pathology , Up-Regulation
14.
Cancer Gene Ther ; 8(3): 231-9, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11332994

ABSTRACT

Recombinant adenoviruses expressing human BRCA1 (AdBRCA1), murine Brca1 (AdBrca1), three clinically relevant human mutant BRCA1 proteins (t340, C61G, and 1853Stop), or a murine Brca1 C-terminal deletion mutant were constructed and evaluated in vitro. These recombinants were capable of transducing high-level transgene expression to a wide variety of cell lines in vitro. Three independent methods were utilized to monitor cell growth following transduction with these recombinants. High-level expression of either the human or mouse wild-type BRCA1 protein was incompatible with maximal levels of cell growth. AdBRCA1 transduction inhibited the outgrowth of several human breast and ovarian cell lines in colony formation assays. Flow cytometric analysis revealed an accumulation of the transduced cells in the G0/G1 phase of the cell cycle. This BRCA1-mediated accumulation of cells in G0/G1 was accompanied by an increase in the cellular level of hypophosphorylated pRB. Ad mutant BRCA1 t340, C61G, and 1853Stop viruses were impaired, to varying degrees, in their ability to transduce a growth-arrested state to the target cells. Using these same three criteria, overexpression of murine Brca1 by AdBrca1 was also capable of transducing a growth-arrested state to human cells. Deletion of the C-terminus of Brca1 diminished this activity. This panel of adenoviruses may be useful reagents as part of an approach to understand the function of BRCA1/Brca1 in normal breast and ovary and help to define the tumor suppressor defect (s) conferred by clinical BRCA1 mutations in breast and ovarian cell tumorigenesis.


Subject(s)
Adenoviridae/genetics , Cell Cycle/genetics , Cell Division/genetics , Electroporation/methods , Genes, BRCA1/genetics , beta-Galactosidase/metabolism , Adenoviridae/metabolism , Animals , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Carcinoma/genetics , Carcinoma/pathology , Cell Cycle/physiology , Cell Division/physiology , DNA Restriction Enzymes/metabolism , Female , Green Fluorescent Proteins , Humans , Luminescent Proteins/analysis , Luminescent Proteins/genetics , Mammary Neoplasms, Animal/genetics , Mammary Neoplasms, Animal/pathology , Mice , Mutation, Missense , Recombination, Genetic , Transduction, Genetic
15.
Breast Cancer Res ; 3(1): 11-3, 2001.
Article in English | MEDLINE | ID: mdl-11250741

ABSTRACT

Recent work by Mak et al demonstrates that mice carrying a T-cell-specific disruption of the brca1 gene display markedly impaired T-lymphocyte development and proliferation in the absence of any increased tendency for the formation of tumors. Interestingly, the extent of these defects was found to be highly dependent on cellular context. Contrasting the rather broad tissue expression pattern of brca1 against its exquisitely selective etiologic role in cancers of the breast and ovary, many of us are left to ponder--where is the specificity?


Subject(s)
BRCA1 Protein/physiology , T-Lymphocytes/immunology , Animals , Cell Lineage , Gene Expression Regulation/immunology , Gene Rearrangement, T-Lymphocyte/immunology , Mice , Mice, Knockout , Neoplasms/immunology
16.
Cancer Res ; 60(21): 5922-8, 2000 Nov 01.
Article in English | MEDLINE | ID: mdl-11085504

ABSTRACT

c-Myc functions through direct activation or repression of transcription. Using cDNA microarray analysis, we have identified c-Myc-responsive genes by comparing gene expression profiles between c-myc null and c-myc wild-type rat fibroblast cells and between c-myc null and c-myc null cells reconstituted with c-myc. From a panel of 4400 cDNA elements, we found 198 genes responsive to c-myc when comparing wild-type or reconstituted cells with the null cells. The plurality of the named c-Myc-responsive genes that were up-regulated, including 30 ribosomal protein genes, are involved in macromolecular synthesis and metabolism, suggesting a major role of c-Myc in the regulation of protein synthetic and metabolic pathways. When ectopically overexpressed, c-Myc induced a different and smaller set of c-Myc-responsive genes as compared with the physiologically expressed c-Myc condition. Thus, these results from expression profiling suggest a new primary function for c-Myc and raise the possibility that the physiological and transforming functions of c-myc may be separable.


Subject(s)
Gene Expression Profiling , Genes, myc/physiology , Proto-Oncogene Proteins c-myc/physiology , Animals , Cell Line , DNA, Complementary/genetics , Down-Regulation , Fibroblasts/physiology , Gene Expression Regulation/physiology , Oligonucleotide Array Sequence Analysis , Protein Biosynthesis , Proteins/metabolism , Proto-Oncogene Proteins c-myc/biosynthesis , Proto-Oncogene Proteins c-myc/genetics , Rats , Up-Regulation
17.
J Clin Oncol ; 18(20): 3471-9, 2000 Oct 15.
Article in English | MEDLINE | ID: mdl-11032587

ABSTRACT

PURPOSE: An association between the overexpression of proto-oncogene HER-2/neu and resistance to tamoxifen in estrogen receptor (ER)-positive primary and metastatic breast cancer has been suggested. We examine a possible interaction between HER-2/neu or p53 expression and tamoxifen effectiveness in patients with ER-positive, node-positive disease treated with cyclophosphamide, doxorubicin, and fluorouracil in a large adjuvant chemotherapy trial (Cancer and Leukemia Group B [CALGB] 8541). Tamoxifen assignment was not randomized-physician discretion was used for premenopausal and postmenopausal women. Trial protocol then specified assignment to postmenopausal women with ER-positive tumors, although not all took tamoxifen. PATIENTS AND METHODS: CALGB 8541 assessed HER-2/neu expression in patients with ER-positive disease by immunohistochemistry (IHC) and fluorescent in situ hybridization (FISH) and amplification by differential polymerase chain reaction (PCR). IHC assessed expression of p53. Univariate and multivariate proportional hazards models assessed tamoxifen-HER-2/neu status interactions and tamoxifen-p53 status interactions. RESULTS: HER-2/neu status was available for 651 patients with ER-positive disease; 650, 608, and 353 patients were assessed by IHC, PCR, and FISH, respectively. Approximately one half received tamoxifen. Reduction in risk of disease recurrence or death resulting from tamoxifen was approximately 37% (32% with overexpression and 39% with normal expression of HER-2/neu; n = 155 by IHC). The tamoxifen-HER-2/neu status interaction was not significant in multivariate analysis of all three HER-2/neu assessment methods. Tamoxifen-p53 interaction did not significantly predict outcome. CONCLUSION: Disease-free and overall survival benefit of tamoxifen in patients with ER-positive, node-positive breast cancer does not depend on HER-2/neu or p53 status. Our data suggest that neither HER-2/neu nor p53 expression should be used to determine assignment of tamoxifen.


Subject(s)
Antineoplastic Agents, Hormonal/therapeutic use , Breast Neoplasms/drug therapy , Receptor, ErbB-2/biosynthesis , Receptors, Estrogen/physiology , Tamoxifen/therapeutic use , Tumor Suppressor Protein p53/biosynthesis , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cyclophosphamide/administration & dosage , Doxorubicin/administration & dosage , Drug Resistance, Neoplasm , Female , Fluorouracil/administration & dosage , Gene Expression , Humans , Immunohistochemistry , In Situ Hybridization, Fluorescence , Lymphatic Metastasis , Middle Aged , Multicenter Studies as Topic , Polymerase Chain Reaction , Postmenopause/physiology , Proportional Hazards Models , Proto-Oncogene Mas , Randomized Controlled Trials as Topic , Receptor, ErbB-2/analysis , Tumor Suppressor Protein p53/analysis
18.
Nat Biotechnol ; 18(4): 457-9, 2000 Apr.
Article in English | MEDLINE | ID: mdl-10748532

ABSTRACT

The completion of the Human Genome Project has made possible the comprehensive analysis of gene expression, and cDNA microarrays are now being employed for expression analysis in cancer cell lines or excised surgical specimens. However, broader application of cDNA microarrays is limited by the amount of RNA required: 50-200 microg of total RNA (T-RNA) and 2-5 microg poly(A) RNA. To broaden the use of cDNA microarrays, some methods aiming at intensifying fluorescence signal have resulted in modest improvement. Methods devoted to amplifying starting poly(A) RNA or cDNA show promise, in that detection can be increased by orders of magnitude. However, despite the common use of these amplification procedures, no systematic assessment of their limits and biases has been documented. We devised a procedure that optimizes amplification of low-abundance RNA samples by combining antisense RNA (aRNA) amplification with a template-switching effect (Clonetech, Palo Alto, CA). The fidelity of aRNA amplified from 1:10,000 to 1:100,000 of commonly used input RNA was comparable to expression profiles observed with conventional poly(A) RNA- or T-RNA-based arrays.


Subject(s)
Gene Amplification , Gene Expression Profiling , RNA, Messenger/genetics , Gene Expression Profiling/methods , Humans , Leukemia, Myeloid, Acute , Melanoma , RNA, Antisense/genetics , Reproducibility of Results , Tumor Cells, Cultured
19.
Cancer Epidemiol Biomarkers Prev ; 9(1): 65-71, 2000 Jan.
Article in English | MEDLINE | ID: mdl-10667465

ABSTRACT

We examined risk factors for breast cancer after subdividing cases based on the presence of HER-2/neu oncogene amplification in their tumors. Data were from the Carolina Breast Cancer Study, a population-based, case-control study of 577 invasive breast cancer patients, diagnosed during 1993-1996 and ages 20-74 years, and 790 controls frequency-matched on race and age. Information on breast cancer risk factors was obtained from structured personal interviews. About 20% of paraffin-embedded tissues from the breast cancers of cases were identified as positive for HER-2/neu amplification (HER-2/neu+) by differential PCR. Early age at menarche, higher waist:hip ratio, and family history of breast or ovarian cancer were associated with elevated odds ratios (ORs) for both HER-2/neu+ and HER-2/neu- breast cancers. Breastfeeding for at least 1 year was inversely associated with HER-2/neu+ breast cancer [OR, 0.3; 95% confidence interval (CI), 0.1-0.7] more so than HER-2/neu- breast cancer (OR, 0.8; 95% CI, 0.5-1.2). Most of the remaining risk factors had ORs around 1.0 for both HER-2/neu+ and HER-2/neu- breast cancers, although a few exhibited possible associations with one disease subtype in analyses stratified by menopausal status. These study results suggest that most recognized breast cancer risk factors do not operate through HER-2/neu amplification in breast carcinogenesis. Differential effects of long-term breastfeeding by HER-2/neu amplification status have been observed in earlier studies and are provocative; however, the direction and magnitude of the associations have not been consistent.


Subject(s)
Breast Neoplasms/etiology , Gene Amplification/genetics , Genes, erbB-2/genetics , Receptor, ErbB-2/genetics , Adult , Age Factors , Aged , Body Constitution , Breast Feeding , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Case-Control Studies , Confidence Intervals , Female , Humans , Interviews as Topic , Menarche , Menopause , Middle Aged , Neoplasm Invasiveness , Odds Ratio , Ovarian Neoplasms/genetics , Paraffin Embedding , Population Surveillance , Risk Factors
20.
J Clin Oncol ; 18(2): 267-74, 2000 Jan.
Article in English | MEDLINE | ID: mdl-10637239

ABSTRACT

PURPOSE: The purpose of this study was to determine whether the presence of HER-2/neu gene amplification and/or overexpression in benign breast disease was associated with an increased risk of subsequent breast cancer. PATIENTS AND METHODS: We conducted a nested case-control study of a cohort of women who were diagnosed with benign breast disease at the Mayo Clinic and who were subsequently observed for the development of breast cancer. Patients who developed breast cancer formed the case group, and a matched sample from the remaining cohort served as controls. Benign tissue samples from 137 cases and 156 controls and malignant tissues from 99 cases provided DNA or tissue for evaluation of HER-2/neu amplification and protein overexpression. RESULTS: Among the controls, seven benign tissues (4.5%) demonstrated low-level HER-2/neu amplification, whereas 13 benign (9.5%) and 18 malignant (18%) tissue specimens from cases exhibited amplification. HER-2/neu amplification in benign breast biopsies was associated with an increased risk of breast cancer (odds ratio ¿OR = 2.2; 95% confidence interval ¿CI, 0.9 to 5.8); this association approached statistical significance. The risks for breast cancer associated with benign breast histopathologic diagnoses were OR = 1.1 (95% CI, 0.6 to 1.9) for lesions exhibiting proliferation without atypia and OR = 1.5 (95% CI, 0.4 to 5.6) for the diagnosis of atypical ductal hyperplasia. For women having both HER-2/neu amplification and a proliferative histopathologic diagnosis (either typical or atypical), the risk of breast cancer was more than seven-fold (OR = 7.2; 95% CI, 0.9 to 60.8). Overexpression of the HER-2/neu protein product, defined as membrane staining in 10% or more of epithelial cells, was found in 30% of the breast tumors but was not detected in any of the benign breast tissues. Case patients who had HER-2/neu gene amplification in their malignant tumor were more likely to have had HER-2/neu amplification in their prior benign biopsy (P =.06, Fisher's exact test). CONCLUSION: Women with benign breast biopsies demonstrating both HER-2/neu amplification and a proliferative histopathologic diagnosis may be at substantially increased risk for subsequent breast cancer.


Subject(s)
Breast Diseases/genetics , Breast Neoplasms/genetics , Gene Amplification , Receptor, ErbB-2/genetics , Adult , Breast Diseases/pathology , Breast Neoplasms/etiology , Breast Neoplasms/pathology , Case-Control Studies , Female , Gene Expression Regulation, Neoplastic , Humans , Receptor, ErbB-2/biosynthesis , Risk Assessment
SELECTION OF CITATIONS
SEARCH DETAIL
...