Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
1.
Acad Pathol ; 8: 23742895211008657, 2021.
Article in English | MEDLINE | ID: mdl-33889720

ABSTRACT

The following fictional case is intended as a learning tool within the Pathology Competencies for Medical Education (PCME), a set of national standards for teaching pathology. These are divided into three basic competencies: Disease Mechanisms and Processes, Organ System Pathology, and Diagnostic Medicine and Therapeutic Pathology. For additional information, and a full list of learning objectives for all three competencies, see http://journals.sagepub.com/doi/10.1177/2374289517715040.1.

2.
Cancers (Basel) ; 12(9)2020 Aug 24.
Article in English | MEDLINE | ID: mdl-32846967

ABSTRACT

Triple-negative breast cancer (TNBC), characterized by the absence or low expression of estrogen receptor (ER), progesterone receptor (PR), and human epidermal growth factor receptor (HER2), is the most aggressive subtype of breast cancer. TNBC accounts for about 15% of breast cancer cases in the U.S., and is known for high relapse rates and poor overall survival (OS). Chemo-resistant TNBC is a genetically diverse, highly heterogeneous, and rapidly evolving disease that challenges our ability to individualize treatment for incomplete responders and relapsed patients. Currently, the frontline standard chemotherapy, composed of anthracyclines, alkylating agents, and taxanes, is commonly used to treat high-risk and locally advanced TNBC. Several FDA-approved drugs that target programmed cell death protein-1 (Keytruda) and programmed death ligand-1 (Tecentriq), poly ADP-ribose polymerase (PARP), and/or antibody drug conjugates (Trodelvy) have shown promise in improving clinical outcomes for a subset of TNBC. These inhibitors that target key genetic mutations and specific molecular signaling pathways that drive malignant tumor growth have been used as single agents and/or in combination with standard chemotherapy regimens. Here, we review the current TNBC treatment options, unmet clinical needs, and actionable drug targets, including epidermal growth factor (EGFR), vascular endothelial growth factor (VEGF), androgen receptor (AR), estrogen receptor beta (ERß), phosphoinositide-3 kinase (PI3K), mammalian target of rapamycin (mTOR), and protein kinase B (PKB or AKT) activation in TNBC. Supported by strong evidence in developmental, evolutionary, and cancer biology, we propose that the K-RAS/SIAH pathway activation is a major tumor driver, and SIAH is a new drug target, a therapy-responsive prognostic biomarker, and a major tumor vulnerability in TNBC. Since persistent K-RAS/SIAH/EGFR pathway activation endows TNBC tumor cells with chemo-resistance, aggressive dissemination, and early relapse, we hope to design an anti-SIAH-centered anti-K-RAS/EGFR targeted therapy as a novel therapeutic strategy to control and eradicate incurable TNBC in the future.

3.
Ann Breast Cancer Ther ; 4(1): 48-57, 2020 May 02.
Article in English | MEDLINE | ID: mdl-32542231

ABSTRACT

Chemo-resistant breast cancer is a major barrier to curative treatment for a significant number of women with breast cancer. Neoadjuvant chemotherapy (NACT) is standard first- line treatment for most women diagnosed with high-risk TNBC, HER2+, and locally advanced ER+ breast cancer. Current clinical prognostic tools evaluate four clinicopathological factors: Tumor size, LN status, pathological stage, and tumor molecular subtype. However, many similarly treated patients with identical residual cancer burden (RCB) following NACT experience distinctly different tumor relapse rates, clinical outcomes and survival. This problem is particularly apparent for incomplete responders with a high-risk RCB classification following NACT. Therefore, there is a pressing need to identify new prognostic and predictive biomarkers, and develop novel curative therapies to augment current standard of care (SOC) treatment regimens to save more lives. Here, we will discuss these unmet needs and clinical challenges that stand in the way of precision medicine and personalized cancer therapy.

4.
EBioMedicine ; 11: 183-198, 2016 Sep.
Article in English | MEDLINE | ID: mdl-27569656

ABSTRACT

BACKGROUND: Metastatic breast cancer exhibits diverse and rapidly evolving intra- and inter-tumor heterogeneity. Patients with similar clinical presentations often display distinct tumor responses to standard of care (SOC) therapies. Genome landscape studies indicate that EGFR/HER2/RAS "pathway" activation is highly prevalent in malignant breast cancers. The identification of therapy-responsive and prognostic biomarkers is paramount important to stratify patients and guide therapies in clinical oncology and personalized medicine. METHODS: In this study, we analyzed matched pairs of tumor specimens collected from 182 patients who received neoadjuvant systemic therapies (NST). Statistical analyses were conducted to determine whether EGFR/HER2/RAS pathway biomarkers and clinicopathological predictors, alone and in combination, are prognostic in breast cancer. FINDINGS: SIAH and EGFR outperform ER, PR, HER2 and Ki67 as two logical, sensitive and prognostic biomarkers in metastatic breast cancer. We found that increased SIAH and EGFR expression correlated with advanced pathological stage and aggressive molecular subtypes. Both SIAH expression post-NST and NST-induced changes in EGFR expression in invasive mammary tumors are associated with tumor regression and increased survival, whereas ER, PR, and HER2 were not. These results suggest that SIAH and EGFR are two prognostic biomarkers in breast cancer with lymph node metastases. INTERPRETATION: The discovery of incorporating tumor heterogeneity-independent and growth-sensitive RAS pathway biomarkers, SIAH and EGFR, whose altered expression can be used to estimate therapeutic efficacy, detect emergence of resistant clones, forecast tumor regression, differentiate among partial responders, and predict patient survival in the neoadjuvant setting, has a clear clinical implication in personalizing breast cancer therapy. FUNDING: This work was supported by the Dorothy G. Hoefer Foundation for Breast Cancer Research (A.H. Tang); Center for Innovative Technology (CIT)-Commonwealth Research Commercialization Fund (CRCF) (MF14S-009-LS to A.H. Tang), and National Cancer Institute (CA140550 to A.H. Tang).


Subject(s)
Breast Neoplasms/metabolism , Breast Neoplasms/mortality , ErbB Receptors/metabolism , Nuclear Proteins/metabolism , Signal Transduction , Ubiquitin-Protein Ligases/metabolism , ras Proteins/metabolism , Biomarkers, Tumor , Breast Neoplasms/pathology , Breast Neoplasms/therapy , Disease Progression , ErbB Receptors/genetics , Female , Gene Expression , Humans , Immunohistochemistry , Kaplan-Meier Estimate , Models, Biological , Neoadjuvant Therapy , Neoplasm Grading , Neoplasm Metastasis , Neoplasm Staging , Nuclear Proteins/genetics , Prognosis , Proportional Hazards Models , Treatment Outcome , Ubiquitin-Protein Ligases/genetics , ras Proteins/genetics
5.
Clin Lab Int ; 40: 18-23, 2016 Nov.
Article in English | MEDLINE | ID: mdl-28579913

ABSTRACT

Metastatic breast cancer is a highly heterogeneous, rapidly evolving and devastating disease that challenges our ability to find curative therapies. RAS pathway activation is an understudied research area in breast cancer. EGFR/RAS pathway activation is prevalent in breast cancer with poor prognosis. The prognostic RAS pathway biomarkers can be used to identify resistant tumour clones, stratify patients and guide therapies.

6.
Breast Cancer Res Treat ; 153(3): 635-45, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26364297

ABSTRACT

Mutations in the p53 gene are among the most frequent genetic events in human cancer and may be triggered by environmental and occupational exposures. We examined the association of clinical and pathological characteristics of breast tumors and breast cancer risk factors according to the prevalence and type of p53 mutations. Using tumor blocks from incident cases from a case-control study in western New York, we screened for p53 mutations in exons 2-11 using the Affymetrix p53 Gene Chip array and analyzed case-case comparisons using logistic regression. The p53 mutation frequency among cases was 28.1 %; 95 % were point mutations (13 % of which were silent) and the remainder were single base pair deletions. Sixty seven percent of all point mutations were transitions; 24 % of them are G:C>A:T at CpG sites. Positive p53 mutation status was associated with poorer differentiation (OR, 95 % CI 2.29, 1.21-4.32), higher nuclear grade (OR, 95 % CI 1.99, 1.22-3.25), and increased Ki-67 status (OR, 95 % CI 1.81, 1.10-2.98). Cases with P53 mutations were more likely to have a combined ER-positive and PR-negative status (OR, 95 % CI 1.65, 1.01-2.71), and a combined ER-negative and PR-negative status (OR, 95 % CI 2.18, 1.47-3.23). Body mass index >30 kg/m(2), waist circumference >79 cm, and waist-to-hip ratio >0.86 were also associated with p53 status; obese breast cancer cases are more likely to have p53 mutations (OR, 95 % CI 1.78, 1.19-2.68). We confirmed that p53 mutations are associated with less favorable tumor characteristics and identified an association of p53 mutation status and adiposity.


Subject(s)
Adiposity , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Genes, p53 , Mutation , Adult , Aged , Alleles , Biomarkers, Tumor , Breast Neoplasms/epidemiology , Case-Control Studies , Female , Genotype , Humans , Middle Aged , Neoplasm Grading , Neoplasm Staging , New York/epidemiology , Population Surveillance , Risk Factors , Tumor Burden
7.
Radiol Case Rep ; 8(3): 425, 2013.
Article in English | MEDLINE | ID: mdl-27330632

ABSTRACT

We present a case of a dedifferentiated liposarcoma with osteosarcomatous degeneration in a 46-year-old male. The mass, palpated during a routine physical exam, was analyzed with dynamic, contrast-enhanced CT and CT-guided core-needle biopsy before surgical resection.

8.
Alcohol ; 45(7): 689-99, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21168302

ABSTRACT

The mechanism for the observed association of alcohol consumption breast cancer risk is not known; understanding that mechanism could improve understanding of breast carcinogenesis and optimize prevention strategies. Alcohol may impact breast malignancies or tumor progression by altering DNA methylation. We examined promoter methylation of three genes, the E-cadherin, p16, and retinoic acid-binding receptor-ß2 (RAR-ß2) genes in archived breast tumor tissues from participants in a population-based case-control study. Real time methylation-specific PCR was performed on 803 paraffin-embedded samples, and lifetime alcohol consumption was queried. Unordered polytomous and unconditional logistic regression were used to derive adjusted odds ratios (ORs) and 95% confidence intervals (CIs). RAR-ß2 methylation was not associated with drinking. Among premenopausal women, alcohol consumption was also not associated with promoter methylation for E-cadherin and p16 genes. In case-case comparisons of postmenopausal breast cancer, compared with lifetime never drinkers, promoter methylation likelihood was increased for higher alcohol intake for E-cadherin (OR=2.39; 95% CI, 1.15-4.96), in particular for those with estrogen receptor-negative tumors (OR=4.13; 95% CI, 1.16-14.72), and decreased for p16 (OR=0.52; 95% CI, 0.29-0.92). There were indications that the association with p16 was stronger for drinking at younger ages. Methylation was also associated with drinking intensity independent of total consumption for both genes. We found alcohol consumption was associated with DNA methylation in postmenopausal breast tumors, suggesting that the association of alcohol and breast cancer may be related, at least in part, to altered methylation, and may differ by drinking pattern.


Subject(s)
Alcohol Drinking/genetics , Breast Neoplasms/genetics , DNA Methylation , Adult , Aged , Breast Neoplasms/etiology , Cadherins/genetics , Case-Control Studies , Female , Genes, p16 , Humans , Middle Aged , Odds Ratio , Postmenopause , Promoter Regions, Genetic/genetics , Receptors, Retinoic Acid/genetics
9.
Breast Cancer Res Treat ; 121(2): 453-60, 2010 Jun.
Article in English | MEDLINE | ID: mdl-19847642

ABSTRACT

Mitochondrial DNA (mtDNA) mutations are frequent in breast tumors, but the etiology of these mutations is unknown. We hypothesized that these mutations are associated with exposures that affect oxidative stress such as alcohol metabolism. Using archived tumor blocks from incident breast cancer cases in a case control study, the Western New York Exposures and Breast Cancer (WEB) study, analysis of mtDNA mutations was conducted on 128 breast cancer cases selected based on extremes of alcohol intake. Temporal temperature gradient gel electrophoresis (TTGE) was used to screen the entire mtDNA genome and sequencing was completed for all TTGE positive samples. Case-case comparisons were completed using unconditional logistic regression to determine the relative prevalence of the mutations by exposures including alcohol consumption, manganese superoxide dismutase (MnSOD) genotype, nutrient intake related to oxidative stress and established breast cancer risk factors. Somatic mtDNA mutations were found in 60 of the 128 tumors examined. There were no differences in the prevalence of mtDNA mutations by alcohol consumption, MnSOD genotype or dietary intake. The likelihood of mtDNA mutations was reduced among those with a positive family history for breast cancer (OR = 0.33, CI = 0.12-0.92), among postmenopausal women who used hormone replacement therapy (OR = 0.46, CI = 0.19-1.08, P = 0.08) and was increased for ER negative tumors (OR = 2.05, CI = 0.95-4.43, P = 0.07). Consistent with previous studies, we found that mtDNA mutations are a frequent occurrence in breast tumors. An understanding of the etiology of mtDNA mutations may provide insight into breast carcinogenesis.


Subject(s)
Alcohol Drinking/adverse effects , Breast Neoplasms/etiology , Breast Neoplasms/genetics , DNA, Mitochondrial/genetics , Breast Neoplasms/epidemiology , Case-Control Studies , DNA Mutational Analysis , Female , Genetic Predisposition to Disease , Genotype , Humans , Immunohistochemistry , Middle Aged , Mutation , Polymerase Chain Reaction , Superoxide Dismutase/genetics
10.
Cancer Epidemiol Biomarkers Prev ; 18(3): 998-1002, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19240236

ABSTRACT

Aberrant promoter methylation is recognized as an important feature of breast carcinogenesis. We hypothesized that genetic variation of genes for methylenetetrahydrofolate reductase (MTHFR) and methionine synthase (MTR), two critical enzymes in the one-carbon metabolism, may alter DNA methylation levels and thus influence DNA methylation in breast cancer. We evaluated case-control association of MTHFR C677T, A1298C, and MTR A2756G polymorphisms for cases strata-defined by promoter methylation status for each of three genes, E-cadherin, p16, and RAR-beta2 in breast cancer; in addition, we evaluated case-case comparisons of the likelihood of promoter methylation in relation to genotypes using a population-based case-control study conducted in Western New York State. Methylation was evaluated with real-time methylation-specific PCRs for 803 paraffin-embedded breast tumor tissues from women with primary, incident breast cancer. We applied unordered polytomous regression and unconditional logistic regression to derive adjusted odds ratios and 95% confidence intervals. We did not find any association of MTHFR and MTR polymorphisms with breast cancer risk stratified by methylation status nor between polymorphisms and likelihood of promoter methylation of any of the genes. There was no evidence of difference within strata defined by menopausal status, estrogen receptor status, folate intake, and lifetime alcohol consumption. Overall, we found no evidence that these common polymorphisms of the MTHFR and MTR genes are associated with promoter methylation of E-cadherin, p16, and RAR-beta2 genes in breast cancer.


Subject(s)
5-Methyltetrahydrofolate-Homocysteine S-Methyltransferase/genetics , Breast Neoplasms/enzymology , Breast Neoplasms/genetics , DNA Methylation , Methylenetetrahydrofolate Reductase (NADPH2)/genetics , Polymorphism, Genetic , Analysis of Variance , Case-Control Studies , Chi-Square Distribution , Cyclin-Dependent Kinase Inhibitor p16 , Female , Genotype , Humans , Neoplasm Proteins/genetics , Receptors, Retinoic Acid/genetics , Ubiquitin-Protein Ligases/genetics
11.
Breast Cancer Res Treat ; 114(3): 559-68, 2009 Apr.
Article in English | MEDLINE | ID: mdl-18463976

ABSTRACT

Aberrant DNA hypermethylation of gene promoter regions has been increasingly recognized as a common molecular alteration in carcinogenesis. We evaluated the association between major clinicopathological features and hypermethylation of genes in tumors among 803 incidence breast cancer cases from a large population-based case-control study conducted in Western New York State. DNA samples were isolated from archive paraffin embedded tumor tissue and were analyzed for hypermethylation status of the E-cadherin, p16, and RAR-beta(2) genes using real time methylation-specific polymerase chain reaction. The frequencies of hypermethylation were 20.0% for E-cadherin, 25.9% for p16, and 27.5% for RAR-beta(2) genes. For postmenopausal women, hypermethylation of E-cadherin tended to be more likely in progesterone receptor (PR) negative than in PR-positive tumors (odds ratio (OR), 1.41; 95% confidence interval (CI), 0.91-2.18). Hypermethylation of p16 tended to be more frequent among estrogen receptor (ER) negative cases than ER-positive cases (OR, 1.51; 95% CI, 1.01-2.32). Hypermethylation of RAR-beta(2) gene was inversely associated with histological and nuclear grade of breast cancer.


Subject(s)
Breast Neoplasms/etiology , Breast Neoplasms/genetics , DNA Methylation , Gene Expression Regulation, Neoplastic , Adult , Aged , Breast Neoplasms/epidemiology , Cadherins/biosynthesis , Cadherins/genetics , Case-Control Studies , Cyclin-Dependent Kinase Inhibitor p16 , Humans , Middle Aged , Neoplasm Metastasis , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , New York , Postmenopause , Receptors, Progesterone/biosynthesis , Receptors, Progesterone/genetics , Receptors, Retinoic Acid/biosynthesis , Receptors, Retinoic Acid/genetics , Reverse Transcriptase Polymerase Chain Reaction
12.
Ann Surg Oncol ; 14(11): 3117-24, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17701256

ABSTRACT

BACKGROUND: The lack of prognostic factors in ductal carcinoma in situ (DCIS) that reliably identifies biologically aggressive tumors adversely affects optimal management. The urokinase-type plasminogen activator (uPA) system, comprised of its receptor, uPAR, and its inhibitor (PAI-1), are critical elements for tumor invasion and their expression in invasive breast cancer can predict clinical outcome. Expression of the uPA system in DCIS may be relevant in defining histological subsets of DCIS with invasive potential. METHODS: Localization of uPA, uPAR, and PAI-1 was investigated immunohistochemically in 60 DCIS tumors. FISH experiments were performed to determine whether uPA was present in cancer cells themselves or derived from stromal elements. RESULTS: uPA was ubiquitously expressed in the malignant ductal epithelium of 95% (57/60) of DCIS tumors studied. uPA-mRNA was detected in the malignant ductal epithelium but not the adjacent normal ductal epithelium and stromal elements. uPAR was expressed in 27% (6/22) of high-grade and 24% (9/38) of non-high-grade DCIS. In comparing coexpression, uPA and uPAR were coexpressed in only 25% (15/60) of tumors. PAI-1 was infrequently expressed in high grade (3/22) and absent in non-high-grade DCIS. CONCLUSIONS: This study identifies the presence of uPA, uPAR, and PAI-1 in both high-grade and non-high-grade DCIS. It may be speculated that coexpression of uPA and its receptor may identify subsets of DCIS with an increased risk for progression to invasive disease. If so, then expression of uPA system components may have prognostic and therapeutic significance in DCIS.


Subject(s)
Breast Neoplasms/metabolism , Carcinoma, Intraductal, Noninfiltrating/metabolism , Plasminogen Activator Inhibitor 1/metabolism , Receptors, Cell Surface/metabolism , Urokinase-Type Plasminogen Activator/metabolism , Breast/metabolism , Breast/pathology , Breast Neoplasms/pathology , Carcinoma, Intraductal, Noninfiltrating/pathology , Female , Humans , Immunoenzyme Techniques , In Situ Hybridization, Fluorescence , Receptors, Urokinase Plasminogen Activator , Urokinase-Type Plasminogen Activator/genetics
13.
Breast Cancer Res Treat ; 102(1): 19-30, 2007 Mar.
Article in English | MEDLINE | ID: mdl-16897429

ABSTRACT

Previous studies using immunohistochemistry suggest that loss of the expression of the prostate-derived Ets transcription factor (PDEF) is a strong indicator for cancer cell malignancy. However, the underlying mechanism for this has not been well elucidated. We determined the role of PDEF in breast cancer cell growth and tumor formation using a series of experiments including Western blotting, promoter-luciferase reporter assay, RNA interference technology and a mouse xenograft model. We also determined the relationship between PDEF expression in human breast tumor specimen and cancer patient survivability. These studies revealed that PDEF expression is inversely associated with survivin expression and breast cancer cell xenograft tumor formation. PDEF-specific shRNA-mediated silencing of PDEF expression resulted in the upregulation of survivin expression in MCF-7 cells, which was associated with increased cell growth and resistance to drug-induced DNA fragmentation (apoptosis). In contrast, survivin-specific siRNA-mediated silencing of survivin expression decreased MCF-7 cell growth. Ectopic expression of PDEF inhibited both survivin promoter activity and endogenous survivin expression. Importantly, shRNA-mediated silencing of PDEF expression in MCF-7 breast cancer cells enhanced survivin expression and xenograft tumor formation in vivo. Furthermore, loss of PDEF expression in breast cancer tissues tends to be associated with unfavorable prognosis. These studies provide new information for the role of PDEF and survivin in breast cancer cell growth and tumor formation.


Subject(s)
Breast Neoplasms/therapy , Microtubule-Associated Proteins/antagonists & inhibitors , Neoplasm Proteins/antagonists & inhibitors , Proto-Oncogene Proteins c-ets/physiology , Adult , Animals , Breast Neoplasms/pathology , Down-Regulation , Female , Humans , Inhibitor of Apoptosis Proteins , Mice , Mice, SCID , Microtubule-Associated Proteins/genetics , Middle Aged , Neoplasm Proteins/genetics , Neoplasm Transplantation , Promoter Regions, Genetic , Proto-Oncogene Proteins c-ets/antagonists & inhibitors , Proto-Oncogene Proteins c-ets/genetics , RNA, Messenger/analysis , RNA, Small Interfering/physiology , Survivin , Transplantation, Heterologous
14.
Breast Cancer Res Treat ; 97(1): 107-10, 2006 May.
Article in English | MEDLINE | ID: mdl-16319977

ABSTRACT

We have measured genomic instability in invasive breast carcinomas and assessed the relationship of genomic instability to known tumor prognostic factors. DNAs from tumors and adjacent normal tissue of 18 breast cancer patients were subjected to inter-Simple Sequence Repeat (inter-SSR) PCR for quantitation of tumor genomic instability. Associations between genomic instability level and known breast cancer prognostic factors were evaluated using the Pearson Product Moment Correlation, the Kruskal-Wallis test of independent samples and the Mann-Whitney non-parametric test. Genomic instability was detected by inter-SSR PCR in over 90% of the breast tumors. The mean instability index was 3.08% (0-7.59%), approximately the same mean value observed in studies of colorectal and thyroid carcinomas. Significantly higher levels of instability were associated with tumors exhibiting necrosis. Genomic instability as measured is detected in the majority of breast cancers at levels comparable to other tumor types. Hypoxia, such as that observed in necrotic regions of tumors, has been associated with elevated genomic damage. We hypothesize that the higher levels of genomic instability detected in necrotic tumors is a consequence of hypoxia-associated DNA damage.


Subject(s)
Breast Neoplasms/genetics , Genomic Instability , Neoplasm Invasiveness/genetics , Repetitive Sequences, Nucleic Acid , Adult , Aged , Aged, 80 and over , Allelic Imbalance , DNA, Neoplasm/genetics , Demography , Female , Humans , Lymphatic Metastasis/genetics , Middle Aged , Polymerase Chain Reaction
16.
Am J Clin Pathol ; 121 Suppl: S33-49, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15298149

ABSTRACT

The HER-2/neu oncogene is located on chromosome 17q and encodes a transmembrane glycoprotein with intracellular tyrosine kinase activity. Several studies have shown an association of HER-2 gene amplification or protein overexpression with prognosis and predictor of therapeutic response. Most important, the presence of amplification or overexpression is the basis of eligibility for trastuzumab therapy. However, there are several methods of determining HER-2 status, each measuring a different aspect such as DNA content, gene copy number, protein expression, expression of RNA, and circulating HER-2 extracellular domain protein. There is no consensus with regard to the optimal test for HER-2 assessment. This review examines the various methods used in an attempt to define the most accurate and reliable test. The most widely used assays are immunohistochemical analysis and fluorescence in situ hybridization (FISH), which measure protein expression and gene amplification, respectively. Based on current data, FISH is the most accurate and reproducible test with a better correlation with prognosis and response to therapy.


Subject(s)
Breast Neoplasms/chemistry , Receptor, ErbB-2/analysis , Algorithms , Breast Neoplasms/therapy , Female , Humans , Immunohistochemistry , In Situ Hybridization, Fluorescence , Prognosis
17.
Breast J ; 10(3): 200-3, 2004.
Article in English | MEDLINE | ID: mdl-15125745

ABSTRACT

Mammographic-pathologic correlation of suspicious microcalcifications is essential for optimal diagnosis and local staging of early breast carcinoma. Loss of microcalcifications during histologic sectioning has been suggested as one reason for the occasional lack of microscopic visualization of microcalcifications in routinely processed breast biopsy specimens obtained for suspicious mammographic microcalcifications. Two case reports utilizing radiography of histologic shavings of stereotactic core biopsies and surgical excisional biopsies of mammographic microcalcifications provide concrete evidence of the loss of large calcific particles during the microtome process.


Subject(s)
Breast Neoplasms , Calcinosis , Carcinoma, Intraductal, Noninfiltrating , Microtomy , Breast Neoplasms/diagnostic imaging , Breast Neoplasms/pathology , Breast Neoplasms/surgery , Calcinosis/diagnostic imaging , Calcinosis/pathology , Carcinoma, Intraductal, Noninfiltrating/diagnostic imaging , Carcinoma, Intraductal, Noninfiltrating/pathology , Carcinoma, Intraductal, Noninfiltrating/surgery , Diagnosis, Differential , Female , Humans , Mammography/methods , Microtomy/methods , Sensitivity and Specificity , Specimen Handling/methods
18.
Oncol Rep ; 11(2): 453-8, 2004 Feb.
Article in English | MEDLINE | ID: mdl-14719083

ABSTRACT

We previously reported that prostate derived Ets transcription factor (PDEF) is a breast tumor-associated molecule. To obtain further insights into PDEF expression in other human tumor types, a cDNA library database from human adult normal and tumor tissues was compiled and searched for PDEF distribution. The results showed that PDEF is present at relative higher frequencies in the cDNA libraries from brain, breast, lung and ovarian tumors in comparison to those from the corresponding normal tissues. RT/PCR analysis of PDEF expression in ovarian tumors confirmed that PDEF is expressed in 36 out of 51 (71%) ovarian tumors. Further comparison of the distribution of PDEF with other widely recognized cancer-associated molecules showed that PDEF has more restricted distributions than Her-2/neu, Bcl-2, survivin or telomerase in cDNA libraries from normal human tissues and more increased distribution than Her-2/neu, CA-125, Bcl-2, survivin and telomerase in cDNA libraries from brain (except survivin), breast, lung and ovarian tumors. These data together show a better tumor-association for PDEF and suggest that PDEF is a more suitable target for developing specific cancer therapies.


Subject(s)
Proto-Oncogene Proteins/metabolism , Transcription Factors/metabolism , DNA, Complementary/genetics , Databases, Factual , Female , Gene Library , Humans , Male , Ovarian Neoplasms , Ovary , Prostate , Protein Binding , Protein-Tyrosine Kinases/isolation & purification , Protein-Tyrosine Kinases/metabolism , Proto-Oncogene Proteins/isolation & purification , Proto-Oncogene Proteins c-ets , Reverse Transcriptase Polymerase Chain Reaction , Transcription Factors/isolation & purification
19.
Diagn Mol Pathol ; 12(4): 201-11, 2003 Dec.
Article in English | MEDLINE | ID: mdl-14639106

ABSTRACT

Regarding HER-2/neu expression (gene or protein level) in lung cancer, several studies with inconsistent results have been recently reported, partially due to variable techniques used and/or heterogeneous populations examined. The objective of this study was to examine HER-2/neu expression in a well-defined cohort of non-small-cell lung cancers (NSCLC) and in nonneoplastic lung tissue utilizing a combination of high-density tissue microarray, immunohistochemistry (IHC), and fluorescent in situ hybridization (FISH) under uniform test conditions. One hundred forty stage I-IIIA primary NSCLCs and 38 non-neoplastic lung samples were examined. IHC, using an FDA-approved Hercept monoclonal antibody kit, was performed and HER-2/neu gene alteration was assessed by FISH. The association of expression of HER-2/neu with clinicopathologic parameters was analyzed. Ninety-four percent of tumor samples (131/140) were fully interpretable after tissue processing. Twenty-five of them (19%) overexpressed (2+, 3+) HER-2/neu, while 106 (81%) had no or weak expression. All thirty-four interpretable non-neoplastic lung samples were negative for HER-2/neu alteration at protein and gene level. HER-2/neu protein overexpression correlated well with HER-2/neu gene amplification (r =.83, P < 0.001). HER-2/neu overexpression was significantly associated with histologic subtype: 19 adenocarcinomas (19/82, 23%) versus 4 squamous cell carcinomas (4/44, 9%) overexpressed Her-2/neu (P = 0.04). Statistical significance was observed between HER-2/neu expression and tumor differentiation, with strong positive (3+) expression observed more frequently in poorly differentiated tumors (P = 0.01). Patients with HER-2/neu abnormalities, particularly HER-2/neu gene amplification, exhibited a shorter survival (P = 0.043). The statistically significant difference (P < 0.005) between HER-2/neu alteration in tumor samples(25/131, 19%) and in the nonneoplastic tissue (0/34, 0%) implies that HER-2/neu may have a role in the carcinogenesis of NSCLC. The findings provide evidence supporting the hypothesis that the HER-2/neu receptor may represent a useful molecular target in the treatment of NSCLC. The significant association of HER-2/neu expression and gene amplification with poorly differentiated carcinoma compared with well differentiated carcinoma suggests that HER-2/neu may be involved in NSCLC tumor evolution. Patients with HER-2/neu gene amplification and strong positive expression of HER-2/neu protein showed a strong tendency toward shorter survival.


Subject(s)
Carcinoma, Non-Small-Cell Lung/genetics , DNA, Neoplasm/genetics , Gene Expression Regulation, Neoplastic , Lung Neoplasms/genetics , Receptor, ErbB-2/genetics , Adult , Aged , Aged, 80 and over , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/mortality , Carcinoma, Non-Small-Cell Lung/secondary , Female , Humans , Immunoenzyme Techniques , In Situ Hybridization, Fluorescence , Lung/metabolism , Lung/pathology , Lung Neoplasms/metabolism , Lung Neoplasms/mortality , Lung Neoplasms/pathology , Male , Middle Aged , Neoplasm Staging , Oligonucleotide Array Sequence Analysis , Proportional Hazards Models , Receptor, ErbB-2/metabolism , Survival Rate
20.
Head Neck ; 25(10): 825-32, 2003 Oct.
Article in English | MEDLINE | ID: mdl-12966506

ABSTRACT

OBJECTIVES: The object of this study is to measure genomic instability in papillary thyroid cancer and correlate these measurements with known clinical prognosticators such as patient age, tumor size, histologic subtype, and three commonly used thyroid risk assessment indices. A secondary objective of this study was to use the measurements of genomic instability to estimate the number of mutational events present in the papillary thyroid cancer genome. METHODS: Inter-simple sequence repeat polymerase chain reaction (ISSR-PCR) is a rapid and reproducible technique for quantitation of genomic instability, or the degree of genome alteration, in solid tumors. This includes quantitation of amplifications, deletions, translocations, and insertions. Twenty-eight papillary carcinomas were evaluated by ISSR-PCR. RESULTS: Evaluation of 28 papillary carcinomas by ISSR-PCR demonstrated a wide range of genomic instability. Of the panel of clinicopathologic factors examined, only patient age was significantly associated with genomic instability. The mean genomic instability index value was greatest in the youngest age group, which was significantly different from the median value measured in the oldest age group (3.7, 2.5, respectively, p =.05). The mean value in the intermediate age group fell between the younger and older groups (3.1). By use of ISSR-PCR, we have calculated 15,000 individual genomic events as having occurred in each papillary tumor cell. CONCLUSIONS: Despite its generally indolent biologic behavior, papillary thyroid cancer exhibits a high degree of genomic instability comparable to that seen in colorectal cancer. These results suggest that elevated genomic instability, as measured by ISSR-PCR, may not be sufficient to enable thyroid tumor progression to less indolent disease and that this process is severely constrained by some additional essential factor such as the differentiated state of the tissue or the need to bring into play an additional form of genomic destabilization.


Subject(s)
Carcinoma, Papillary/genetics , DNA, Neoplasm/analysis , Genomic Instability/genetics , Polymerase Chain Reaction , Thyroid Neoplasms/genetics , Adolescent , Adult , Age Factors , Aged , Aged, 80 and over , Carcinoma, Papillary/pathology , Carcinoma, Papillary/surgery , Chromosome Aberrations , Female , Humans , Male , Middle Aged , Neoplasm Staging , Polymerase Chain Reaction/methods , Prognosis , Risk Factors , Thyroid Neoplasms/pathology , Thyroid Neoplasms/surgery , Thyroidectomy
SELECTION OF CITATIONS
SEARCH DETAIL
...