Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Appl Immunohistochem Mol Morphol ; 22(4): 308-16, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24717231

ABSTRACT

A formalin-fixed paraffin-embedded tissue-based prognostic assay to assess the risk for recurrence in stage II colon cancer has recently been clinically validated. This study describes the analytical performance and quality control measures of the assay. The reportable range was determined to be [-1.129, 1.414] in risk score units. The accuracy was evaluated with a split sample comparison within the production lab and between the production lab and a reference lab. The concordance between the replicates within the production lab was 79% (95% confidence interval, 64%-91%). There was no evidence of bias, and the concordance was 78% (95% confidence interval, 61%-90%) between the labs. The lab-to-lab concordance was further evaluated by simulating risk scores from the full reportable range. The simulation suggested a higher concordance. The sensitivity study demonstrated that the percentage of tumor tissue did not impact the risk score and that RNA concentration of 9.5 ng/µL was a conservative determination of the analyte lower limit of quantification. From the precision study, the repeatability and reproducibility estimates were 0.1267 and 0.0548 in risk score units, respectively. Furthermore, multifaceted quality control measures were implemented, such as proper tissue processing steps, high-risk and low-risk controls, nontemplate control, and a gene expression-based classifier to evaluate the cDNA amplification kit, a key reagent in the assay. In conclusion, this study demonstrates the strong analytical performance of the assay and further supports its use as an objective standardized prognostic test for stage II colon cancer.


Subject(s)
Biomarkers, Tumor/genetics , Colonic Neoplasms/diagnosis , DNA, Neoplasm/analysis , Gene Expression Regulation, Neoplastic , Neoplasm Recurrence, Local/diagnosis , Reagent Kits, Diagnostic/standards , Colonic Neoplasms/genetics , Colonic Neoplasms/pathology , DNA, Complementary/analysis , DNA, Complementary/genetics , DNA, Neoplasm/genetics , Formaldehyde , Humans , Neoplasm Recurrence, Local/genetics , Neoplasm Recurrence, Local/pathology , Neoplasm Staging , Observer Variation , Paraffin Embedding , Prognosis , Quality Control , Reproducibility of Results , Sensitivity and Specificity , Tissue Fixation
2.
J Natl Cancer Inst ; 106(1): djt335, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24402422

ABSTRACT

BACKGROUND: There is no method routinely used to predict response to anthracycline and cyclophosphamide-based chemotherapy in the clinic; therefore patients often receive treatment for breast cancer with no benefit. Loss of the Fanconi anemia/BRCA (FA/BRCA) DNA damage response (DDR) pathway occurs in approximately 25% of breast cancer patients through several mechanisms and results in sensitization to DNA-damaging agents. The aim of this study was to develop an assay to detect DDR-deficient tumors associated with loss of the FA/BRCA pathway, for the purpose of treatment selection. METHODS: DNA microarray data from 21 FA patients and 11 control subjects were analyzed to identify genetic processes associated with a deficiency in DDR. Unsupervised hierarchical clustering was then performed using 60 BRCA1/2 mutant and 47 sporadic tumor samples, and a molecular subgroup was identified that was defined by the molecular processes represented within FA patients. A 44-gene microarray-based assay (the DDR deficiency assay) was developed to prospectively identify this subgroup from formalin-fixed, paraffin-embedded samples. All statistical tests were two-sided. RESULTS: In a publicly available independent cohort of 203 patients, the assay predicted complete pathologic response vs residual disease after neoadjuvant DNA-damaging chemotherapy (5-fluorouracil, anthracycline, and cyclophosphamide) with an odds ratio of 3.96 (95% confidence interval [Cl] =1.67 to 9.41; P = .002). In a new independent cohort of 191 breast cancer patients treated with adjuvant 5-fluorouracil, epirubicin, and cyclophosphamide, a positive assay result predicted 5-year relapse-free survival with a hazard ratio of 0.37 (95% Cl = 0.15 to 0.88; P = .03) compared with the assay negative population. CONCLUSIONS: A formalin-fixed, paraffin-embedded tissue-based assay has been developed and independently validated as a predictor of response and prognosis after anthracycline/cyclophosphamide-based chemotherapy in the neoadjuvant and adjuvant settings. These findings warrant further validation in a prospective clinical study.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , DNA Damage/drug effects , DNA, Neoplasm/drug effects , Fanconi Anemia/metabolism , Adult , Aged , Anthracyclines/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Breast Neoplasms/genetics , Chemotherapy, Adjuvant , Cyclophosphamide/administration & dosage , Disease-Free Survival , Epirubicin/administration & dosage , Fanconi Anemia/genetics , Female , Fluorouracil/administration & dosage , Humans , Middle Aged , Neoadjuvant Therapy/methods , Odds Ratio , Oligonucleotide Array Sequence Analysis , Prospective Studies
3.
J Clin Oncol ; 29(35): 4620-6, 2011 Dec 10.
Article in English | MEDLINE | ID: mdl-22067406

ABSTRACT

PURPOSE: Current prognostic factors are poor at identifying patients at risk of disease recurrence after surgery for stage II colon cancer. Here we describe a DNA microarray-based prognostic assay using clinically relevant formalin-fixed paraffin-embedded (FFPE) samples. PATIENTS AND METHODS: A gene signature was developed from a balanced set of 73 patients with recurrent disease (high risk) and 142 patients with no recurrence (low risk) within 5 years of surgery. RESULTS: The 634-probe set signature identified high-risk patients with a hazard ratio (HR) of 2.62 (P < .001) during cross validation of the training set. In an independent validation set of 144 samples, the signature identified high-risk patients with an HR of 2.53 (P < .001) for recurrence and an HR of 2.21 (P = .0084) for cancer-related death. Additionally, the signature was shown to perform independently from known prognostic factors (P < .001). CONCLUSION: This gene signature represents a novel prognostic biomarker for patients with stage II colon cancer that can be applied to FFPE tumor samples.


Subject(s)
Colonic Neoplasms/pathology , Neoplasm Recurrence, Local/pathology , Paraffin Embedding/methods , Aged , Colonic Neoplasms/genetics , Female , Formaldehyde , Gene Expression Profiling , Humans , Male , Middle Aged , Neoplasm Recurrence, Local/genetics , Neoplasm Staging , Oligonucleotide Array Sequence Analysis/methods , Paraffin Embedding/standards , Prognosis , Retrospective Studies , Risk Factors , Tissue Fixation
4.
Nucleic Acids Res ; 39(22): 9536-48, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21880590

ABSTRACT

A role for BRCA1 in the direct and indirect regulation of transcription is well established. However, a comprehensive view of the degree to which BRCA1 impacts transcriptional regulation on a genome-wide level has not been defined. We performed genome-wide expression profiling and ChIP-chip analysis, comparison of which revealed that although BRCA1 depletion results in transcriptional changes in 1294 genes, only 44 of these are promoter bound by BRCA1. However, 27% of these transcripts were linked to transcriptional regulation possibly explaining the large number of indirect transcriptional changes observed by microarray analysis. We show that no specific consensus sequence exists for BRCA1 DNA binding but rather demonstrate the presence of a number of known and novel transcription factor (TF)- binding sites commonly found on BRCA1 bound promoters. Co-immunoprecipitations confirmed that BRCA1 interacts with a number of these TFs including AP2-α, PAX2 and ZF5. Finally, we show that BRCA1 is bound to a subset of promoters of genes that are not altered by BRCA1 loss, but are transcriptionally regulated in a BRCA1-dependent manner upon DNA damage. These data suggest a model, whereby BRCA1 is present on defined promoters as part of an inactive complex poised to respond to various genotoxic stimuli.


Subject(s)
BRCA1 Protein/physiology , Transcriptome , BRCA1 Protein/antagonists & inhibitors , BRCA1 Protein/metabolism , Cell Line, Tumor , Chromatin Immunoprecipitation , Gene Expression Profiling , Gene Expression Regulation , Humans , Oligonucleotide Array Sequence Analysis , Stress, Physiological/genetics , Transcription Factors/metabolism
5.
BMC Med Genomics ; 1: 20, 2008 May 30.
Article in English | MEDLINE | ID: mdl-18513400

ABSTRACT

BACKGROUND: Non-small cell lung cancer (NSCLC) is the leading cause of cancer mortality worldwide. At present no reliable biomarkers are available to guide the management of this condition. Microarray technology may allow appropriate biomarkers to be identified but present platforms are lacking disease focus and are thus likely to miss potentially vital information contained in patient tissue samples. METHODS: A combination of large-scale in-house sequencing, gene expression profiling and public sequence and gene expression data mining were used to characterise the transcriptome of NSCLC and the data used to generate a disease-focused microarray - the Lung Cancer DSA research tool. RESULTS: Built on the Affymetrix GeneChip platform, the Lung Cancer DSA research tool allows for interrogation of ~60,000 transcripts relevant to Lung Cancer, tens of thousands of which are unavailable on leading commercial microarrays. CONCLUSION: We have developed the first high-density disease specific transcriptome microarray. We present the array design process and the results of experiments carried out to demonstrate the array's utility. This approach serves as a template for the development of other disease transcriptome microarrays, including non-neoplastic diseases.

6.
Clin Cancer Res ; 13(24): 7413-20, 2007 Dec 15.
Article in English | MEDLINE | ID: mdl-18094425

ABSTRACT

PURPOSE: We investigated whether BRCA1 mRNA expression levels may represent a biomarker of survival in sporadic epithelial ovarian cancer following chemotherapy treatment. EXPERIMENTAL DESIGN: The effect of loss of BRCA1 expression on chemotherapy response in ovarian cancer was measured in vitro using dose inhibition assays and Annexin V flow cytometry. Univariate and multivariate analyses were done to evaluate the relationship between BRCA1 mRNA expression levels and survival after chemotherapy treatment in 70 fresh frozen ovarian tumors. RESULTS: We show that inhibition of endogenous BRCA1 expression in ovarian cancer cell lines results in increased sensitivity to platinum therapy and decreased sensitivity to antimicrotubule agents. In addition, we show that patients with low/intermediate levels of BRCA1 mRNA have a significantly improved overall survival following treatment with platinum-based chemotherapy in comparison with patients with high levels of BRCA1 mRNA (57.2 versus 18.2 months; P = 0.0017; hazard ratio, 2.9). Furthermore, overall median survival for higher-BRCA1-expressing patients was found to increase following taxane-containing chemotherapy (23.0 versus 18.2 months; P = 0.12; hazard ratio, 0.53). CONCLUSIONS: We provide evidence to support a role for BRCA1 mRNA expression as a predictive marker of survival in sporadic epithelial ovarian cancer.


Subject(s)
BRCA1 Protein/biosynthesis , Drug Resistance, Neoplasm/genetics , Neoplasms, Glandular and Epithelial/genetics , Ovarian Neoplasms/genetics , RNA, Messenger/analysis , Antineoplastic Agents/therapeutic use , Apoptosis/genetics , BRCA1 Protein/genetics , Biomarkers, Tumor/analysis , Biomarkers, Tumor/genetics , Blotting, Western , Female , Flow Cytometry , Humans , Kaplan-Meier Estimate , Middle Aged , Neoplasms, Glandular and Epithelial/drug therapy , Neoplasms, Glandular and Epithelial/mortality , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/mortality , Polymerase Chain Reaction , Survival Analysis
7.
J Natl Cancer Inst ; 99(22): 1683-94, 2007 Nov 21.
Article in English | MEDLINE | ID: mdl-18000219

ABSTRACT

BACKGROUND: BRCA1-mutant breast tumors are typically estrogen receptor alpha (ER alpha) negative, whereas most sporadic tumors express wild-type BRCA1 and are ER alpha positive. We examined a possible mechanism for the observed ER alpha-negative phenotype of BRCA1-mutant tumors. METHODS: We used a breast cancer disease-specific microarray to identify transcripts that were differentially expressed between paraffin-embedded samples of 17 BRCA1-mutant and 14 sporadic breast tumors. We measured the mRNA levels of estrogen receptor 1 (ESR1) (the gene encoding ER alpha), which was differentially expressed in the tumor samples, by quantitative polymerase chain reaction. Regulation of ESR1 mRNA and ER alpha protein expression was assessed in human breast cancer HCC1937 cells that were stably reconstituted with wild-type BRCA1 expression construct and in human breast cancer T47D and MCF-7 cells transiently transfected with BRCA1-specific short-interfering RNA (siRNA). Chromatin immunoprecipitation assays were performed to determine if BRCA1 binds the ESR1 promoter and to identify other interacting proteins. Sensitivity to the antiestrogen drug fulvestrant was examined in T47D and MCF-7 cells transfected with BRCA1-specific siRNA. All statistical tests were two-sided. RESULTS: Mean ESR1 gene expression was 5.4-fold lower in BRCA1-mutant tumors than in sporadic tumors (95% confidence interval [CI] = 2.6-fold to 40.1-fold, P = .0019). The transcription factor Oct-1 recruited BRCA1 to the ESR1 promoter, and both BRCA1 and Oct-1 were required for ER alpha expression. BRCA1-depleted breast cancer cells expressing exogenous ER alpha were more sensitive to fulvestrant than BRCA1-depleted cells transfected with empty vector (T47D cells, the mean concentration of fulvestrant that inhibited the growth of 40% of the cells [IC40] for empty vector versus ER alpha: >10(-5) versus 8.0 x 10(-9) M [95% CI = 3.1 x 10(-10) to 3.2 x 10(-6) M]; MCF-7 cells, mean IC40 for empty vector versus ER alpha: >10(-5) versus 4.9 x 10(-8) M [95% CI = 2.0 x 10(-9) to 3.9 x 10(-6) M]). CONCLUSIONS: BRCA1 alters the response of breast cancer cells to antiestrogen therapy by directly modulating ER alpha expression.


Subject(s)
Antineoplastic Agents, Hormonal/pharmacology , Breast Neoplasms/chemistry , Breast Neoplasms/genetics , Estradiol/analogs & derivatives , Estrogen Receptor Modulators/pharmacology , Estrogen Receptor alpha/deficiency , Gene Silencing , Genes, BRCA1 , Mutation , Blotting, Northern , Breast Neoplasms/drug therapy , Cell Line, Tumor , Estradiol/pharmacology , Estrogen Receptor alpha/genetics , Female , Fulvestrant , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/drug effects , Humans , Immunoblotting , Immunoprecipitation , RNA, Messenger/analysis , RNA, Small Interfering , Research Design , Reverse Transcriptase Polymerase Chain Reaction , Transcription, Genetic
8.
Mol Cancer Res ; 5(3): 261-70, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17374731

ABSTRACT

BRCA1 encodes a tumor suppressor gene that is mutated in the germ line of women with a genetic predisposition to breast and ovarian cancer. BRCA1 has been implicated in a number of important cellular functions including DNA damage repair, transcriptional regulation, cell cycle control, and ubiquitination. Using an Affymetrix U95A microarray, IRF-7 was identified as a BRCA1 transcriptional target and was also shown to be synergistically up-regulated by BRCA1 specifically in the presence of IFN-gamma, coincident with the synergistic induction of apoptosis. We show that BRCA1, signal transducer and activator of transcription (STAT)-1, and STAT2 are all required for the induction of IRF-7 following stimulation with IFN-gamma. We also show that the induction of IRF-7 by BRCA1 and IFN-gamma is dependent on the type I IFNs, IFN-alpha and IFN-beta. We show that BRCA1 is required for the up-regulation of STAT1, STAT2, and the type I IFNs in response to IFN-gamma. We show that BRCA1 is localized at the promoters of the molecules involved in type I IFN signaling leading to their up-regulation. Blocking this intermediary type I IFN step using specific antisera shows the requirement for IFN-alpha and IFN-beta in the induction of IRF-7 and apoptosis. Finally, we outline a mechanism for the BRCA1/IFN-gamma regulation of target genes involved in the innate immune response, which is dependent on type I IFN signaling.


Subject(s)
BRCA1 Protein/metabolism , Immunity, Innate/genetics , Interferon Regulatory Factor-7/genetics , Interferon Type I/metabolism , Interferon-gamma/pharmacology , Apoptosis , BRCA1 Protein/analysis , Cell Line, Tumor , Cell Proliferation , Humans , Interferon Type I/antagonists & inhibitors , Interferon Type I/pharmacology , Promoter Regions, Genetic , STAT1 Transcription Factor/genetics , STAT1 Transcription Factor/metabolism , STAT2 Transcription Factor/genetics , STAT2 Transcription Factor/metabolism , Signal Transduction/drug effects , Signal Transduction/immunology , Up-Regulation
9.
Mol Pharmacol ; 70(1): 60-70, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16571652

ABSTRACT

We have demonstrated previously that certain members of a series of novel pyrrolo-1,5-benzoxazepine (PBOX) compounds potently induce apoptosis in a variety of human chemotherapy-resistant cancer cell lines and in primary ex vivo material derived from cancer patients. A better understanding of the molecular mechanisms underlying the apoptotic effects of these PBOX compounds is essential to their development as antineoplastic therapeutic agents. This study sought to test the hypothesis that proapoptotic PBOX compounds target the microtubules. We show that a representative proapoptotic PBOX compound, PBOX-6, induces apoptosis in both the MCF-7 and K562 cell lines. An accumulation of cells in G2/M precedes apoptosis in response to PBOX-6. PBOX-6 induces prometaphase arrest and causes an accumulation of cyclin B1 levels and activation of cyclin B1/CDK1 kinase in a manner similar to that of two representative antimicrotubule agents, nocodazole and paclitaxel. Indirect immunofluorescence demonstrates that both PBOX-6 and another pro-apoptotic PBOX compound, PBOX-15, cause microtubule depolymerization in MCF-7 cells. They also inhibit the assembly of purified tubulin in vitro, whereas a nonapoptotic PBOX compound (PBOX-21) has no effect on either the cellular microtubule network or on the assembly of purified tubulin. This suggests that the molecular target of the pro-apoptotic PBOX compounds is tubulin. PBOX-6 does not bind to either the vinblastine or the colchicine binding site on tubulin, suggesting that it binds to an as-yet-uncharacterised novel site on tubulin. The ability of PBOX-6 to bind tubulin and cause microtubule depolymerization confirms it as a novel candidate for antineoplastic therapy.


Subject(s)
Apoptosis/drug effects , Oxazepines/pharmacology , Pyrroles/pharmacology , Tubulin/metabolism , Antineoplastic Agents, Phytogenic/metabolism , Antineoplastic Agents, Phytogenic/pharmacology , Binding Sites , Binding, Competitive , Blotting, Western , CDC2 Protein Kinase/metabolism , Carbamates/pharmacology , Cell Division , Cell Line, Tumor , Colchicine/metabolism , Colchicine/pharmacology , Cyclin B/metabolism , Cyclin B1 , Dose-Response Relationship, Drug , G2 Phase , Humans , K562 Cells , Metaphase/drug effects , Microtubules/drug effects , Microtubules/metabolism , Oxazepines/metabolism , Pyrroles/metabolism , Time Factors , Vinblastine/metabolism , Vinblastine/pharmacology
10.
Biochim Biophys Acta ; 1639(1): 43-52, 2003 Sep 01.
Article in English | MEDLINE | ID: mdl-12943967

ABSTRACT

The present study examines the molecular mechanisms by which a member of a novel series of pyrrolo-1,5-benzoxazepines, PBOX-21, induces G1 arrest in 1321N1 cells. PBOX-21-induced G1 arrest is preceded by both a decrease in CDK2 kinase activity, which is critical for the G1/S transition, and a downregulation in cyclin D(3) protein expression levels, suggesting that these two events may be crucially involved in the mediation of the cell cycle arrest. The decrease in CDK2 activity may be due to an observed decrease in CDK2 protein levels following PBOX-21 treatment. Coinciding with the arrest is a reduction in the activity of CDK4, due to either the observed PBOX-21 induced downregulation in CDK4 expression, or a reduction in complex formation between cyclin D(3)-CDK4 leading to a decrease in the levels of active cyclin D(3)-CDK4 complexes with kinase activity. The level of CDK6 activity was also seen to be reduced following PBOX-21 treatment, also possibly due to a reduction in complex formation with cyclin D(3). However, this reduction in CDK6 kinase activity was not seen until after PBOX-21-induced G1 arrest has reached its maximum, and therefore may be viewed as a consequence of, and a method of maintaining the PBOX-21-induced arrest, rather than a cause. Also in parallel with the G1 arrest elicited by PBOX-21 is an upregulation in the universal CDK inhibitor, p21. Furthermore, the retinoblastoma protein (Rb), a substrate of CDK2 and CDK6, whose phosphorylation is necessary for cell cycle progression, becomes hypophosphorylated. These results indicate that PBOX-21 exerts its growth inhibitory effects through the modulation of the expression and activity of several key G1 regulatory proteins.


Subject(s)
Antineoplastic Agents/pharmacology , Astrocytoma/enzymology , Benzodiazepines/pharmacology , CDC2-CDC28 Kinases , Cyclin-Dependent Kinases/metabolism , G1 Phase/physiology , Proto-Oncogene Proteins , Astrocytoma/drug therapy , Astrocytoma/metabolism , Cyclin-Dependent Kinase 2 , Cyclin-Dependent Kinase 4 , Cyclin-Dependent Kinase 6 , Humans , Phosphorylation , Protein Serine-Threonine Kinases/metabolism , Retinoblastoma Protein/metabolism , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL
...