Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
1.
Ann Oncol ; 28(8): 1903-1909, 2017 Aug 01.
Article in English | MEDLINE | ID: mdl-28486686

ABSTRACT

BACKGROUND: Prostate cancers (PCs) with similar characteristics at the time of diagnosis can have very different disease outcomes. Conventional biomarkers of PC still lack precision in identifying individuals at high risk of PC recurrence. While many candidate biomarkers are proposed in the literature, few are in clinical practice as they lack rigorous validation. This study prospectively enrolled an independent phase III cohort to evaluate the clinical utility of zinc-alpha 2-glycoprotein (AZGP1) as a prognostic biomarker in localized PC. PATIENTS AND METHODS: In our multicentre, prospective phase III study, AZGP1 status in 347 radical prostatectomy specimens was assayed by immunohistochemistry in a NATA-accredited laboratory. The AZGP1 score was assessed in a multivariable model incorporating established prognostic factors. We also report extended outcomes from our previous phase II study. The primary endpoint was biochemical relapse-free survival (BRFS). Secondary endpoints were metastasis-free survival (MFS) and PC-specific survival (PCSS). RESULTS: In the phase II cohort, with a median follow-up of 15.8 years, low/absent AZGP1 expression was an independent predictor of poor BRFS (HR, 1.4; 95% CI, 1.1-1.9; P = 0.03), MFS (HR, 2.8; 95% CI, 1.2-6.6; P = 0.02) and PCSS (HR, 3.8; 95% CI, 1.5-9.5; P = 0.005). These results were validated in our prospective phase III cohort. Low/absent AZGP1 expression independently predicted for BRFS (HR, 1.9; 95% CI, 1.1-3.3; P = 0.02), with shorter MFS (HR, 2.0; 95% CI, 1.1-3.4; P = 0.02). AZGP1 improved the discriminatory value when incorporated into existing prognostic risk models. CONCLUSION: Our study provides prospective phase III validation that absent/low AZGP1 expression provides independent prognostic value in PC. This study provides robust evidence for the incorporation of this biomarker into clinical practice.


Subject(s)
Biomarkers, Tumor/metabolism , Carrier Proteins/metabolism , Glycoproteins/metabolism , Prostatic Neoplasms/metabolism , Adipokines , Adult , Aged , Biopsy , Cohort Studies , Humans , Male , Middle Aged , Prospective Studies , Prostatic Neoplasms/pathology
2.
Br J Cancer ; 112(8): 1340-8, 2015 Apr 14.
Article in English | MEDLINE | ID: mdl-25867259

ABSTRACT

BACKGROUND: Docetaxel improves symptoms and survival in metastatic castration-resistant prostate cancer (CRPC). However, ∼50% of patients are chemoresistant. This study examined whether changes in cytokine levels predict for docetaxel resistance in vitro and in a clinical cohort. METHODS: PC3 cells or their docetaxel-resistant subline (PC3Rx) were co-cultured with U937 monocytes, with and without docetaxel treatment, and cytokine levels were measured. The circulating levels of 28 cytokines were measured pre-/post cycle 1 of docetaxel from 55 men with CRPC, and compared with prostate-specific antigen (PSA) response. RESULTS: PC3Rx-U937 co-culture expressed more cytokines, chiefly markers of alternative macrophage differentiation, compared with PC3-U937 co-culture. Docetaxel treatment enhanced cytokine production by PC3Rx-U937 co-culture, while reducing cytokine levels in PC3-U937. In patients, changes in the levels of seven circulating cytokines (macrophage inhibitory cytokine 1 (MIC1), interleukin (IL)-1ra, IL-1ß, IL-4, IL-6, IL-12 and IFNγ) after cycle 1 of docetaxel were associated with progressive disease (all P<0.05). The combination of changes in MIC1, IL-4 and IL-6 most strongly predicted PSA response (P=0.002). CONCLUSIONS: In vitro studies suggest docetaxel resistance is mediated, at least in part, by cytokines induced by the interaction between the docetaxel-resistant tumour cells and macrophages. Early changes in circulating cytokine levels were associated with docetaxel resistance in CRPC patients. When considered together, these data suggest a significant role for the inflammatory response and macrophages in the development of docetaxel resistance in CRPC.


Subject(s)
Cytokines/blood , Drug Resistance, Neoplasm , Kallikreins/blood , Macrophages/metabolism , Prostate-Specific Antigen/blood , Prostatic Neoplasms, Castration-Resistant/metabolism , Aged , Aged, 80 and over , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Coculture Techniques , Docetaxel , Humans , Male , Middle Aged , Prostatic Neoplasms, Castration-Resistant/drug therapy , Taxoids/pharmacology
3.
Oncogene ; 28(43): 3847-56, 2009 Oct 29.
Article in English | MEDLINE | ID: mdl-19684615

ABSTRACT

GATA-2, a member of the GATA family of transcription factors, is involved in androgen receptor (AR) signaling, however, little is known regarding its role in prostate cancer. Here, we report that GATA-2 is expressed in a substantial proportion of prostate cancers and that high expression of GATA-2 is associated with biochemical recurrence and distant metastatic progression in a validation set of 203 cancers. In vitro data show that GATA-2 is directly recruited to the promoter region of the AR upon androgen stimulation of LNCaP prostate cancer cells with 5alpha-dihydroxytestosterone (DHT) for 24 h. Ectopic GATA-2 expression causes the induction of AR transcript levels under androgen-depleted conditions (P<0.05). The expression of the AR target gene, AZGP1, is induced upon androgen stimulation and this effect is repressed by GATA-2. In contrast, GATA-2 significantly increases transcript levels of KLK2, which increases further in a time-dependent manner on DHT treatment and in the presence of GATA-2. These results indicate that upregulation of GATA-2 may contribute to the progression to aggressive prostate cancer through modulation of expression of AR and key androgen-regulated genes, one of which, AZGP1, is associated with the progression to metastatic disease.


Subject(s)
GATA2 Transcription Factor/physiology , Gene Expression Regulation, Neoplastic , Prostatic Neoplasms/pathology , Adipokines , Carrier Proteins/genetics , GATA2 Transcription Factor/analysis , Glycoproteins/genetics , Humans , Male , Phenotype , Promoter Regions, Genetic , Prostatic Neoplasms/genetics , Receptors, Androgen/analysis , Receptors, Androgen/genetics , Tissue Kallikreins/genetics
4.
Oncogene ; 28(15): 1812-20, 2009 Apr 16.
Article in English | MEDLINE | ID: mdl-19287456

ABSTRACT

Aberrant expression of cyclin D1 protein is a common feature of breast cancer. However, the CCND1 gene encodes two gene products, cyclin D1a and cyclin D1b, which have discrete mechanisms of regulation and impact on cell behavior. A polymorphism at nucleotide 870 in the CCND1 gene, rs603965, influences the relative production of the encoded proteins and can impart increased risk for tumor development. Here, the impact of both the G/A870 polymorphism and cyclin D1b protein production on breast cancer risk, disease phenotype and patient outcome was analysed. In a large multiethnic case-control study, the G/A870 polymorphism conferred no significant risk for breast cancer overall or by stage or estrogen receptor (ER) status. However, the cyclin D1b protein was found to be upregulated in breast cancer, independent of cyclin D1a levels, and exhibited heterogeneous levels in breast cancer specimens. High cyclin D1a expression inversely correlated with the Ki67 proliferation marker and was not associated with clinical outcome. In contrast, elevated cyclin D1b expression was independently associated with adverse outcomes, including recurrence, distant metastasis and decreased survival. Interestingly, cyclin D1b was particularly associated with poor outcome in the context of ER-negative breast cancer. Thus, specific cyclin D1 isoforms are associated with discrete forms of breast cancer and high cyclin D1b protein levels hold prognostic potential.


Subject(s)
Breast Neoplasms/chemistry , Cyclin D1/analysis , Breast Neoplasms/etiology , Breast Neoplasms/mortality , Cyclin D1/genetics , Genes, erbB-2 , Humans , Immunohistochemistry , Ki-67 Antigen/analysis , Polymorphism, Genetic , Prognosis , Protein Isoforms , Receptors, Estrogen/analysis
5.
Br J Cancer ; 100(1): 123-33, 2009 Jan 13.
Article in English | MEDLINE | ID: mdl-19066611

ABSTRACT

BAG-1 (bcl-2-associated athanogene) enhances oestrogen receptor (ER) function and may influence outcome and response to endocrine therapy in breast cancer. We determined relationships between BAG-1 expression, molecular phenotype, response to tamoxifen therapy and outcome in a cohort of breast cancer patients and its influence on tamoxifen sensitivity in MCF-7 breast cancer cells in vitro. Publically available gene expression data sets were analysed to identify relationships between BAG-1 mRNA expression and patient outcome. BAG-1 protein expression was assessed using immunohistochemistry in 292 patients with invasive ductal carcinoma and correlated with clinicopathological variables, therapeutic response and disease outcome. BAG-1-overexpressing MCF-7 cells were treated with antioestrogens to assess its effects on cell proliferation. Gene expression data demonstrated a consistent association between high BAG-1 mRNA and improved survival. In ER+ cancer (n=189), a high nuclear BAG-1 expression independently predicted improved outcome for local recurrence (P=0.0464), distant metastases (P=0.0435), death from breast cancer (P=0.009, hazards ratio 0.29, 95% CI: 0.114-0.735) and improved outcome in tamoxifen-treated patients (n=107; P=0.0191). BAG-1 overexpression in MCF-7 cells augmented antioestrogen-induced growth arrest. A high BAG-1 expression predicts improved patient outcome in ER+ breast carcinoma. This may reflect both a better definition of the hormone-responsive phenotype and a concurrent increased sensitivity to tamoxifen.


Subject(s)
Breast Neoplasms/drug therapy , Carcinoma, Ductal, Breast/drug therapy , DNA-Binding Proteins/physiology , Estrogen Antagonists/therapeutic use , Receptors, Estrogen/analysis , Tamoxifen/therapeutic use , Transcription Factors/physiology , Breast Neoplasms/chemistry , Breast Neoplasms/pathology , Carcinoma, Ductal, Breast/chemistry , Carcinoma, Ductal, Breast/pathology , Cell Line, Tumor , DNA-Binding Proteins/analysis , DNA-Binding Proteins/genetics , Female , Humans , Immunohistochemistry , Neoplasm Invasiveness , RNA, Messenger/analysis , Transcription Factors/analysis , Transcription Factors/genetics
6.
Br J Cancer ; 98(6): 1085-93, 2008 Mar 25.
Article in English | MEDLINE | ID: mdl-18349819

ABSTRACT

Despite a high initial response rate to first-line platinum/paclitaxel chemotherapy, most women with epithelial ovarian cancer relapse with recurrent disease that becomes refractory to further cytotoxic treatment. We have previously shown that the E3 ubiquitin ligase, EDD, a regulator of DNA damage responses, is amplified and overexpressed in serous ovarian carcinoma. Given that DNA damage pathways are linked to platinum resistance, the aim of this study was to determine if EDD expression was associated with disease recurrence and platinum sensitivity in serous ovarian cancer. High nuclear EDD expression, as determined by immunohistochemistry in a cohort of 151 women with serous ovarian carcinoma, was associated with an approximately two-fold increased risk of disease recurrence and death in patients who initially responded to first-line chemotherapy, independently of disease stage and suboptimal debulking. Although EDD expression was not directly correlated with relative cisplatin sensitivity of ovarian cancer cell lines, sensitivity to cisplatin was partially restored in platinum-resistant A2780-cp70 ovarian cancer cells following siRNA-mediated knockdown of EDD expression. These results identify EDD as a new independent prognostic marker for outcome in serous ovarian cancer, and suggest that pathways involving EDD, including DNA damage responses, may represent new therapeutic targets for chemoresistant ovarian cancer.


Subject(s)
Cisplatin/pharmacology , Drug Resistance, Neoplasm , Ovarian Neoplasms/metabolism , Ubiquitin-Protein Ligases/metabolism , Cell Line, Tumor , Cystadenocarcinoma, Serous , Female , Humans , Middle Aged , Neoplasm Recurrence, Local , Ovarian Neoplasms/drug therapy , Prognosis , Retrospective Studies
7.
Br J Cancer ; 98(3): 537-41, 2008 Feb 12.
Article in English | MEDLINE | ID: mdl-18231110

ABSTRACT

Identification of a biomarker of prognosis and response to therapy that can be assessed preoperatively would significantly improve overall outcomes for patients with pancreatic cancer. In this study, patients whose tumours exhibited high LMO4 expression had a significant survival advantage following operative resection, whereas the survival of those patients whose tumours had low or no LMO4 expression was not significantly different when resection was compared with operative biopsy alone.


Subject(s)
Adenocarcinoma/metabolism , Carcinoma, Pancreatic Ductal/metabolism , Homeodomain Proteins/metabolism , Pancreatic Neoplasms/metabolism , Transcription Factors/metabolism , Adaptor Proteins, Signal Transducing , Adenocarcinoma/surgery , Adult , Aged , Aged, 80 and over , Biomarkers, Tumor/analysis , Carcinoma, Pancreatic Ductal/surgery , Cohort Studies , Female , Humans , LIM Domain Proteins , Male , Middle Aged , Pancreatectomy , Pancreatic Neoplasms/surgery , Prognosis , Survival Analysis , Treatment Outcome
8.
Br J Cancer ; 94(6): 904-13, 2006 Mar 27.
Article in English | MEDLINE | ID: mdl-16508639

ABSTRACT

Mucinous epithelial ovarian cancers (MOC) are clinically and morphologically distinct from the other histological subtypes of ovarian cancer. To determine the genetic basis of MOC and to identify potential tumour markers, gene expression profiling of 49 primary ovarian cancers of different histological subtypes was performed using a customised oligonucleotide microarray containing >59 000 probesets. The results show that MOC express a genetic profile that both differs and overlaps with other subtypes of epithelial ovarian cancer. Concordant with its histological phenotype, MOC express genes characteristic of mucinous carcinomas of varying epithelial origin, including intestinal carcinomas. Differences in gene expression between MOC and other histological subtypes of ovarian cancer were confirmed by RT-PCR and/or immunohistochemistry. In particular, galectin 4 (LGALS4) was highly and specifically expressed in MOC, but expressed at lower levels in benign mucinous cysts and borderline (atypical proliferative) tumours, supporting a malignant progression model of MOC. Hence LGALS4 may have application as an early and differential diagnostic marker of MOC.


Subject(s)
Adenocarcinoma, Mucinous/genetics , Gene Expression Profiling , Genetic Markers , Ovarian Neoplasms/genetics , Adenocarcinoma, Mucinous/pathology , Cell Transformation, Neoplastic , Disease Progression , Female , Galectin 4/biosynthesis , Humans , Immunohistochemistry , Middle Aged , Oligonucleotide Array Sequence Analysis , Ovarian Neoplasms/pathology , Phenotype , Reverse Transcriptase Polymerase Chain Reaction
9.
Gut ; 50(6): 861-8, 2002 Jun.
Article in English | MEDLINE | ID: mdl-12010891

ABSTRACT

BACKGROUND AND AIMS: Intraductal papillary mucinous tumours (IPMT) of the pancreas constitute a unique pathological entity with an overall incidence of associated invasive malignancy of 20%. The malignant potential of an individual IPMT cannot be accurately predicted. Preoperative estimation of the risk of associated invasive malignancy with IPMT would be of significant clinical benefit. As aberrations in cell cycle regulatory genes are associated with the progression of precursor pancreatic ductal lesions to invasive adenocarcinoma, we examined expression of key cell cycle regulatory genes in the cyclin D1/retinoblastoma pathway and the transforming growth factor beta/Smad4 signalling pathway in a cohort of patients with surgically resected IPMT. METHODS: Sections of formalin fixed paraffin embedded pancreatic tissue from a cohort of 18 patients with IPMT were examined using immunohistochemistry for protein expression of cell cycle regulatory genes p16(INK4A), p21(CIP1), p27(KIP1), cyclin D1, pRb, and p53, as well as the cell signalling molecule Smad4. A comparison of expression levels was made between adenoma/borderline IPMT (10 patients) and intraductal papillary mucinous carcinoma (IPMC) (eight patients, four of whom harboured invasive carcinoma). Statistical analysis was performed using the chi(2) and Fisher's exact tests. RESULTS: Aberrant expression of the proteins examined increased in frequency from adenoma/borderline IPMT to IPMC. Specifically, there was a significantly greater incidence of loss of p16(INK4A) expression in IPMC: 8/8 lesions (100%) compared with 1/10 (10%) adenoma/borderline IPMT (p<0.001). Similarly, loss of Smad4 expression was associated with IPMC: 3/8 (38%) versus adenoma/borderline IPMT 0/10 (p<0.03). Loss of Smad4 expression within the IPMT was the best marker for the presence of invasive carcinoma (p<0.001). CONCLUSIONS: These data indicate that loss of p16(INK4A) and Smad4 expression occur more frequently in IPMC alone, or with associated invasive carcinoma, compared with adenoma/borderline IPMT. Aberrant protein expression of these cell cycle regulatory genes in IPMT and pancreatic intraepithelial neoplasia in the current model of pancreatic cancer progression suggest similarities in their development and may also represent the subsequent risk of invasive carcinoma.


Subject(s)
Adenocarcinoma, Mucinous/metabolism , Carcinoma in Situ/metabolism , Carcinoma, Pancreatic Ductal/metabolism , Cyclin-Dependent Kinase Inhibitor p16/metabolism , DNA-Binding Proteins/metabolism , Pancreatic Neoplasms/metabolism , Trans-Activators/metabolism , Aged , Aged, 80 and over , Cell Cycle Proteins/metabolism , Cyclin D1/metabolism , Cyclin-Dependent Kinase Inhibitor p21 , Cyclin-Dependent Kinase Inhibitor p27 , Cyclins/metabolism , Female , Humans , Immunohistochemistry , Male , Middle Aged , Retinoblastoma Protein/metabolism , Smad4 Protein , Tumor Cells, Cultured , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Proteins/metabolism
10.
Cancer Res ; 61(24): 8830-7, 2001 Dec 15.
Article in English | MEDLINE | ID: mdl-11751405

ABSTRACT

Pancreatic cancer (PC) is thought to develop through a series of duct lesions termed pancreatic intraepithelial neoplasia (PanIN). Characterization of the molecular pathology of these lesions may lead to additional understanding of pancreatic ductal carcinogenesis. We examined the protein expression of four functionally related genes, p21(WAF1/CIP1) (CDKN1A), p53, cyclin D1 (CCND1), and DPC4/Smad4 (MADH4), aberrations of which are associated with PC, within 451 PanIN lesions present in the pancreata of 60 patients. p21(WAF1/CIP1) overexpression was present in the normal ducts of 9% of patients and increased progressively to 16% of patients with PanIN-1A lesions, to 32% of patients with PanIN-1B lesions, 56% of patients with PanIN-2 lesions, 80% of patients with PanIN-3 lesions, and 85% of patients with invasive carcinomas (P < 0.01). p53 and cyclin D1 overexpression occurred predominantly in PanIN-3 lesions (P < 0.01), and loss of DPC4/Smad4 expression occurred predominantly in PanIN-3 lesions and invasive carcinoma (P < 0.01). In addition, p21(WAF1/CIP1) overexpression occurred independently of p53 and DPC4/Smad4 expression within invasive carcinoma and PanIN-3 lesions. Cyclin D1 overexpression or loss of DPC4/Smad4 expression was apparent in 85% of invasive carcinomas but in only 14% of PanIN-2 lesions. These data demonstrate that overexpression of p21(WAF1/CIP1) occurs early in the development of PanIN, before aberrations in p53, cyclin D1, and DPC4/Smad4 expression. p21(WAF1/CIP1) overexpression, independent of p53 and/or DPC4/Smad4 expression, may reflect increased Ras activity, either directly through activating K-ras mutations or as a consequence of HER-2/neu (ERBB2) overexpression, both of which are common in PC and in early events in the development of PanIN. These data support further the current progression model for PC and demonstrate that aberrant expression of key cell cycle regulatory genes may be important in the early development and progression of PanIN.


Subject(s)
Carcinoma, Pancreatic Ductal/metabolism , Cyclins/biosynthesis , Pancreatic Neoplasms/metabolism , Precancerous Conditions/metabolism , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/pathology , Cyclin D1/biosynthesis , Cyclin D1/genetics , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/genetics , DNA-Binding Proteins/biosynthesis , DNA-Binding Proteins/genetics , Disease Progression , Gene Expression Regulation, Neoplastic , Humans , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Precancerous Conditions/genetics , Precancerous Conditions/pathology , Smad4 Protein , Trans-Activators/biosynthesis , Trans-Activators/genetics , Tumor Suppressor Protein p53/biosynthesis , Tumor Suppressor Protein p53/genetics
11.
J Clin Oncol ; 19(16): 3692-705, 2001 Aug 15.
Article in English | MEDLINE | ID: mdl-11504751

ABSTRACT

PURPOSE: Although predicting outcome for men with clinically localized prostate cancer (PC) has improved, the staging system and nomograms used to do this are based on results from the North American health system. To be internationally applicable, these models require testing in cohorts from a variety of different health systems based on the predominant PC case identification methods used. PATIENTS AND METHODS: We studied 732 men with localized PC treated with radical prostatectomy and no preoperative therapy between 1986 and 1999 at one Australian institution to determine the effect of clinicopathologic features on disease-free survival. RESULTS: Preoperative serum prostate-specific antigen (PSA) concentration, Gleason score, pathologic stage, and year of surgery were independent predictors of outcome. Although margin status demonstrated only a trend toward significance in multivariate modeling overall, it proved to be independent in subgroups based on later year of surgery (1986 to 1994 v 1995 to 1998), preoperative PSA of less than 10 ng/mL, and Gleason score > or = 7. Adjuvant radiation therapy improved disease-free survival rates in patients with multiple surgical margin involvement. CONCLUSION: This work confirms the prognostic significance of pathologic stage, Gleason score, and preoperative serum PSA. In the context of a contemporaneous screening effect in Australia, these findings may have implications for methods that predict outcome following surgery as screening becomes more prevalent in a population. The independent prognostic effect of margin status may alter with an increase in the proportion of screening-identified PCs. Staging systems and nomograms that predict outcome following surgery require validation in cohorts with different health practices before being universally applied.


Subject(s)
Adenocarcinoma/mortality , Adenocarcinoma/pathology , Neoplasm Staging/standards , Prostatic Neoplasms/mortality , Prostatic Neoplasms/pathology , Adenocarcinoma/surgery , Adult , Aged , Cohort Studies , Disease-Free Survival , Humans , Male , Middle Aged , New South Wales/epidemiology , Predictive Value of Tests , Prognosis , Prostate-Specific Antigen/blood , Prostatectomy , Prostatic Neoplasms/surgery , Survival Analysis
12.
Cancer Res ; 61(14): 5331-5, 2001 Jul 15.
Article in English | MEDLINE | ID: mdl-11454669

ABSTRACT

The role of estrogen and its receptors in the etiology and progression of prostate cancer (PC) is poorly understood. In normal and malignant human prostate, estrogen receptor-alpha is expressed only in the stroma, whereas estrogen receptor-beta (ERbeta) is present in both normal stroma and epithelium. Because loss of ERbeta expression is associated with prostate hyperplasia in ERbeta-null mice, this study determined patterns of ERbeta expression in normal, hyperplastic, and malignant human prostate and associations with clinical outcome. Five normal prostates from organ donors and 159 radical prostatectomy specimens from patients with clinically localized PC were assessed for ERbeta expression using immunohistochemistry. ERbeta-positivity was defined as > or =5% of cells demonstrating nuclear immunoreactivity. All of the five normal prostates showed strong ERbeta-nuclear staining in >95% of the epithelium and 35% of the stromal cells. The number of ERbeta-positive cases declined to 24.2% (38/157) in hyperplasia adjacent to carcinoma and 11.3% (18/159) in PCs. ERbeta-positivity was related to decreased relapse-free survival (log-rank P = 0.04). Thus, loss of ERbeta expression is associated with progression from normal prostate epithelium to PC, whereas those cancers that retained ERbeta expression were associated with a higher rate of recurrence. These data identify the need to further investigate the potential role of ERbeta in the regulation of prostate epithelial cell proliferation and the functional consequences of decreased ERbeta expression in the evolution of PC.


Subject(s)
Prostatic Neoplasms/pathology , Receptors, Estrogen/biosynthesis , Adult , Aged , Disease-Free Survival , Estrogen Receptor alpha , Estrogen Receptor beta , Humans , Hyperplasia/metabolism , Hyperplasia/pathology , Immunohistochemistry , Male , Middle Aged , Prostate/chemistry , Prostate/pathology , Prostatic Neoplasms/metabolism
13.
Clin Cancer Res ; 7(3): 544-50, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11297246

ABSTRACT

Prostate cancer (PC) is the most commonly diagnosed male cancer in industrialized societies. No molecular markers of PC progression or outcome with proven clinical utility have been described. Because the loss of normal cell cycle control is an early event in the evolution of cancer, we sought to determine whether changes in expression of the cyclin-dependent kinase inhibitor, p16INK4A, predicted outcome in this disease. We screened a cohort of 206 patients with clinically localized PC treated with radical prostatectomy for overexpression of the INK4A gene, the product of which inactivates the G1-phase cyclin dependent kinases, Cdk4 and Cdk6. p16INK4A protein expression was evaluated by immunohistochemistry in areas of high-grade intraepithelial neoplasia (HGPIN), a precursor to invasive disease, and of cancer in the same specimen. Data were evaluated for disease relapse using the Kaplan-Meier method and in a Cox proportional hazards model by assessing p16INK4A status in areas of HGPIN and cancer with other variables of known clinical relevance. Overexpression of p16INK4A in HGPIN and cancer was correlated with, but independent of, pathological stage and was associated with early relapse in PC patients treated with radical prostatectomy (log-rank test, P < 0.001). In a multivariate model adjusted for Gleason grade, pretreatment prostate-specific antigen levels, pathological stage, and margin status, overexpression of p16INK4A in HGPIN was an independent predictor of disease relapse and increased the risk of recurrence 2.24-fold (95% confidence interval, 1.28-3.93). These data provide the first evidence for a prognostic marker in HGPIN. The clinical utility of p16INK4A status in stratifying patients for aggressive treatment very early in the disease process, potentially several years prior to the onset of invasive disease, requires further investigation.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p16/biosynthesis , Prostatic Neoplasms/metabolism , Recurrence , Age Factors , Aged , Cohort Studies , Disease Progression , Disease-Free Survival , Humans , Immunohistochemistry , Male , Middle Aged , Multivariate Analysis , Proportional Hazards Models , Prostate-Specific Antigen/biosynthesis , Prostatic Neoplasms/diagnosis , Prostatic Neoplasms/pathology , Time Factors
14.
Cancer Res ; 61(2): 423-7, 2001 Jan 15.
Article in English | MEDLINE | ID: mdl-11212224

ABSTRACT

The molecular basis of androgen-independent prostate cancer is unknown; however, functional androgen receptor (AR) signaling is maintained after the acquisition of hormone-refractory disease. Because normal and malignant prostate epithelial cell proliferation is regulated by androgen stimulation via both the AR-positive stroma and epithelium, we sought to evaluate patterns of AR expression in these cells and to determine any relationships with prostate cancer progression. AR expression in the malignant epithelium and associated periepithelial and nonperiepithelial stroma was measured in a cohort of 96 patients with clinically localized prostate cancer treated with radical prostatectomy. Data were evaluated for disease relapse using the Kaplan-Meier method and in a Cox proportional hazards model with other variables of known clinical relevance, including Gleason score, pathological stage, clinical stage, and pretreatment prostate-specific antigen concentration. Concurrent overexpression of AR (> or = 70% positive nuclei) in the malignant epithelium and loss of AR immunoreactivity in the adjacent periepithelial stroma (< or = 30%) was associated with higher clinical stage (P = 0.01), higher pretreatment prostate-specific antigen level (P = 0.03), and earlier relapse after radical prostatectomy (log-rank P = 0.009). These data identify a pattern of AR expression in malignant epithelium and adjacent stroma that is associated with a poor clinical outcome in prostate cancer. Equally important, they identify the need to further investigate the mechanistic basis of loss of AR expression in the malignant stroma and its potential role in deregulation of prostate epithelial cell proliferation.


Subject(s)
Epithelial Cells/chemistry , Prostatic Neoplasms/metabolism , Receptors, Androgen/biosynthesis , Stromal Cells/chemistry , Aged , Disease-Free Survival , Epithelial Cells/pathology , Humans , Immunohistochemistry , Male , Middle Aged , Neoplasm Recurrence, Local , Prostate-Specific Antigen/blood , Prostatectomy , Prostatic Neoplasms/pathology , Prostatic Neoplasms/surgery , Stromal Cells/pathology
15.
Cancer Res ; 60(6): 1585-94, 2000 Mar 15.
Article in English | MEDLINE | ID: mdl-10749127

ABSTRACT

The role of p53 in the pathogenesis of, and as a predictive biomarker for, localized prostate cancer (PCa) is contested. Recent work has suggested that patterns of p53 nuclear accumulation determined by immunohistochemistry are prognostic, whereas studies using other methods question the role of p53 mutations in predicting outcome. We studied 263 men with localized PCa treated with radical prostatectomy to determine whether p53 nuclear accumulation predicts relapse and disease-specific mortality. We combined two p53 immunohistochemistry scoring systems: (a) percentage of p53-positive tumor nuclei in all major foci of cancer within the prostate; and (b) clustering, where the presence of 12 or more p53-positive cells within a x 200 power field was deemed "cluster positive." Analysis was undertaken using chi2, Kruskal-Wallis, and Mann-Whitney tests for clinicopathological variables and the Kaplan-Meier method, log-rank test, and univariate and multivariate Cox regression modeling for evaluation of contribution to relapse and disease-specific survival. At mean follow-up of 55.1 months (range, 4.9-123.0 months), 39% (102 of 263) of patients had relapsed and 2.3% (6 of 253) had died of PCa. Pretreatment serum prostate-specific antigen concentration, pathological tumor stage, lymph node involvement, Gleason score, and p53 nuclear accumulation, as determined by either percentage score or cluster status, were independent predictors of relapse in multivariate analysis. Clustering of p53-positive cells distinguished between favorable and poor prognosis patients within the lowest p53-positive stratum (>0 to <2%) and was the most discriminatory threshold for predicting relapse in the entire cohort. p53 status predicted outcome in patients with a Gleason score of 5 and above but not those with a score of 4 and below. In patients treated with neoadjuvant hormonal therapy, p53 cluster positivity carried a 90% (19 of 21) risk of relapse by 36 months. All six patients who died from PCa in the period of the study exhibited p53 nuclear accumulation in 20% or more tumor nuclei. This study demonstrates strong relationships between p53 nuclear accumulation and relapse and disease-specific mortality in a large series of localized PCas. Furthermore, the presence of clusters of p53-positive nuclei delineates a group of patients with poor prognosis not identified by traditional scoring methods and supports the hypothesis that p53 dysfunction within PCa may exist in foci of tumor cells that are clonally expanded in metastases.


Subject(s)
Cell Nucleus/chemistry , Prostatectomy , Prostatic Neoplasms/metabolism , Tumor Suppressor Protein p53/metabolism , Aged , Cohort Studies , Follow-Up Studies , Humans , Immunohistochemistry , Male , Middle Aged , Multivariate Analysis , Neoplasm Recurrence, Local , Neoplasm Staging , Prognosis , Prostate-Specific Antigen/blood , Prostatic Neoplasms/pathology , Prostatic Neoplasms/surgery , Survival Analysis
16.
Clin Cancer Res ; 4(4): 847-54, 1998 Apr.
Article in English | MEDLINE | ID: mdl-9563877

ABSTRACT

The cell cycle regulatory gene cyclin D1 is a candidate oncogene in breast cancer. It is overexpressed in 30-50% of invasive primary breast cancers and plays a key role in mediating mitogenic responses to steroids and growth factors in breast cancer cells in vitro. Because the role of cyclin D1 in the proliferative and early noninvasive stages of breast cancer is largely unknown, we examined normal breast epithelium (NBE), proliferative disease (PD), ductal carcinoma in situ (DCIS), and invasive carcinoma (IC) to evaluate the timing and possible importance of cyclin D1 expression in the development of breast cancer. Using immunohistochemistry, we examined cyclin D1 protein expression in 471 breast tissue samples. A quantitative scoring system for immunohistochemistry based on percentage of positive cells was developed that correlated with Western blot analysis of antigen concentration in paired samples (r2 = 0.91, P = 0.003). A sample was considered positive if >5% of relevant epithelial cells demonstrated nuclear staining. Cyclin D1 positivity was observed in 11.7% (7 of 60) samples of NBE, 25% (11 of 44) of PD without atypia, 39.4% (13 of 33) of atypical ductal hyperplasia, 43.6% (17 of 39) of low-grade DCIS, 47.9% (23 of 48) of high-grade DCIS, and 48.3% (99 of 205) of IC. Cyclin D1 expression was significantly higher in PD than NBE (P = 0.006) and in DCIS than PD (P = 0.038). There was no significant increase from DCIS to IC (P = 0.52). The increase in cyclin D1 expression in the overall progression from NBE to IC was also highly significant (P = 0.0001). Therefore, cyclin D1 expression was detected at levels significantly greater than in NBE in the earliest proliferative epithelial lesions of the breast with a further significant increase accompanying the progression to any form of cancer. This suggests that overexpression of cyclin D1 protein is important at the earliest stages of breast oncogenesis and continues to have a crucial role throughout the development of malignancy.


Subject(s)
Breast Neoplasms/metabolism , Carcinoma, Intraductal, Noninfiltrating/metabolism , Cyclin D1/metabolism , Neoplasm Proteins/metabolism , Breast Neoplasms/pathology , Carcinoma, Intraductal, Noninfiltrating/pathology , Female , Humans , Hyperplasia/metabolism , Hyperplasia/pathology , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL