Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
1.
Radiother Oncol ; 195: 110240, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38522597

ABSTRACT

BACKGROUND AND PURPOSE: To report mature results for local control and survival in oligometastatic (OM) breast cancer patients treated with stereotactic body radiotherapy (SBRT) on lung and/or liver lesions in a phase II trial. METHODS: This is a prospective non-randomized phase II trial (NCT02581670) which enrolled patients from 2015 to 2021. Eligibility criteria included: age > 18 years, ECOG 0-2, diagnosis of breast cancer, maximum of 4 lung/liver lesions (with a maximum diameter < 5 cm), metastatic disease confined to the lungs and liver or extrapulmonary or extrahepatic disease stable or responding to systemic therapy. The primary end-points were local control (LC) and treatment-related toxicities. The secondary end-points included overall survival (OS), distant metastasis-free survival (DMFS), time to next systemic therapy (TTNS), poly-progression free survival (PPFS). RESULTS: The study included 64 patients with a total of 90 lesions treated with SBRT. LC at 1 and 2 years was 94.9 %, 91 % at 3 years. Median local control was not reached. Median OS was 16.5 months, OS at 1, 2 and 3 years was 87.5 %, 60.9 % and 51.9 %, respectively. Median DMFS was 8.3 months, DMFS at 1, 2 and 3 years was 38.1 %, 20.6 % and 16 % respectively. At univariate analysis, local response to SBRT was found to be statistically linked with better OS, DMFS and STFS. CONCLUSION: SBRT is a safe and valid option in oligometastatic breast cancer patients, with very high rates of local control. An optimal selection of patients is likely needed to improve survival outcomes and reduce the rate of distant progression.


Subject(s)
Breast Neoplasms , Liver Neoplasms , Lung Neoplasms , Radiosurgery , Humans , Female , Breast Neoplasms/pathology , Breast Neoplasms/radiotherapy , Breast Neoplasms/mortality , Radiosurgery/methods , Liver Neoplasms/secondary , Liver Neoplasms/radiotherapy , Liver Neoplasms/mortality , Liver Neoplasms/surgery , Lung Neoplasms/pathology , Lung Neoplasms/radiotherapy , Lung Neoplasms/mortality , Middle Aged , Prospective Studies , Aged , Adult , Aged, 80 and over
2.
Vet Pathol ; 60(2): 185-189, 2023 03.
Article in English | MEDLINE | ID: mdl-36541627

ABSTRACT

Sebaceous carcinomas of the human ocular adnexa commonly exhibit pagetoid spread, mutations in tumor-suppressor genes, and protooncogene copy number gain. Sebaceous carcinomas are rarely reported in other species, and while the Meibomian gland (MG) represents the most common ocular adnexal structure of the canine eyelid to develop neoplasia, most are clinically and histologically benign. The objective of this study was to compare molecular features of canine MG carcinomas and adenomas. Two retrospectively identified MG carcinomas were subject to immunohistochemistry and qPCR. When compared with normal glands, MYC was upregulated in benign and malignant MG neoplasms. Aberrant p53 expression was restricted to the nuclei of intraepithelial neoplastic cells in MG carcinomas. Adipophilin expression was diminished in MG neoplasms compared with the normal MG. Our findings, if confirmed in a larger cohort of cases, could suggest that MG oncogenesis in a dog may exhibit similar molecular features as their human counterparts.


Subject(s)
Adenoma , Carcinoma, Basal Cell , Dog Diseases , Sebaceous Gland Neoplasms , Skin Neoplasms , Humans , Dogs , Animals , Meibomian Glands/metabolism , Meibomian Glands/pathology , Tumor Suppressor Protein p53 , Retrospective Studies , Sebaceous Gland Neoplasms/chemistry , Sebaceous Gland Neoplasms/metabolism , Sebaceous Gland Neoplasms/pathology , Sebaceous Gland Neoplasms/veterinary , Skin Neoplasms/veterinary , Carcinoma, Basal Cell/veterinary , Cell Transformation, Neoplastic , Adenoma/pathology , Adenoma/veterinary , Mutation
3.
J Cancer Res Clin Oncol ; 149(8): 5173-5179, 2023 Jul.
Article in English | MEDLINE | ID: mdl-36357737

ABSTRACT

PURPOSE: Patients with oligo-metastatic disease (OMD) can be safely treated with Stereotactic Radiation Therapy (SRT). Further disease progression is common in these patients. In most cases, patients relapse again with oligo-metastases, however some can experience a poly-progression after a local ablative treatment (LAT). The purpose of this study was to retrospectively identify factors associated with poly-progression in patients receiving SRT for OMD. METHODS: Data from a monocentric database were retrospectively analyzed. Patients treated with SRT for OMD and who developed progression after LAT were selected. Patients were categorized as oligo- or poly-progressive according to the number of new/progressing metastases (≤ or > 5). Herein, we analyzed data about patients' characteristics, oligo-metastatic presentation and radiation treatment characteristics to evaluate their relationship with progression type. RESULTS: From 2013 to 2021, data on 700 patients progressing after LAT were analyzed. Among them, 227 patients (32.4%) experienced a poly-progression; the median time to poly-progression was 7.72 months (range 1-79.6). Five variables associated with poly-progression were found to be statistically significant in the univariate analysis: performance status (p < 0.001), site of the primary tumor (p = 0.016), ablative dose (p = 0.002), treated site (p = 0.002), single or double organ (p = 0.03). Of those, all but the number of involved organs retained their significant predictive value on the multivariate analysis. CONCLUSION: Our study identified four independent factors associated with poly-progression in patients with OMD receiving SRT. Our data may support comprehensive characterization of OMD, better understanding of factors associated with progression.


Subject(s)
Neoplasms , Radiosurgery , Humans , Retrospective Studies , Neoplasms/radiotherapy
4.
J Comp Pathol ; 171: 30-37, 2019 Aug.
Article in English | MEDLINE | ID: mdl-31540623

ABSTRACT

The terminology applied to canine prostatic epithelial lesions, especially carcinomas, is currently not standardized and this hampers the ability of pathologists to study the biological and clinical significance of these lesions. The aim of this review is to present the essential histomorphological diagnostic attributes of a wide spectrum of prostatic epithelial lesions in dogs. In addition to the traditionally recognized prostatic hyperplasia, hormonal atrophy, prostatitis, squamous metaplasia, adenocarcinoma and transitional cell (urothelial) carcinoma, new entities are described and discussed in order to provide veterinary pathologists with a basic atlas of common histological lesions of the canine prostate that is comprehensive and easy to use.


Subject(s)
Dog Diseases/pathology , Prostate/pathology , Prostatic Hyperplasia/veterinary , Prostatic Neoplasms/veterinary , Terminology as Topic , Animals , Dogs , Male , Prostatic Hyperplasia/pathology , Prostatic Neoplasms/pathology
5.
J Comp Pathol ; 158: 1-5, 2018 Jan.
Article in English | MEDLINE | ID: mdl-29422309

ABSTRACT

Proliferative inflammatory atrophy (PIA), which is comprised of highly proliferative but atrophic prostate epithelial cells in association with chronic inflammation, is considered a risk lesion for prostate cancer in men, while its role in canine prostate carcinogenesis is still unknown. We evaluated the value of immunohistochemical labelling for the basal cell marker cytokeratin-5 (CK5) in identifying PIA lesions in 87 samples of formalin-fixed and paraffin wax-embedded canine prostate. Canine PIA showed cytological features identical to the human counterpart and in most cases was associated with chronic lymphoplasmacytic inflammation. PIA lesions were identified in a higher number of CK5-labelled slides (43 out of 87) compared with slides stained by haematoxylin and eosin (HE) (24 out of 87). This lesion was frequently present in normal, hyperplastic and neoplastic canine prostates, although it was underestimated on evaluation of HE-stained slides. Therefore, CK5 can be considered a useful basal cell marker with high sensitivity and specificity for PIA.


Subject(s)
Biomarkers/analysis , Dog Diseases/diagnosis , Keratin-5/analysis , Precancerous Conditions/veterinary , Prostate/pathology , Animals , Atrophy , Dogs , Male
6.
Virchows Arch ; 468(5): 607-17, 2016 May.
Article in English | MEDLINE | ID: mdl-26861919

ABSTRACT

The prognostic value of phosphatase and tensin homolog (PTEN) loss in prostate cancer has primarily been evaluated by either fluorescence in situ hybridization (FISH) or immunohistochemistry (IHC). Previously, we found that PTEN loss by IHC was associated with increased risk of upgrading from biopsy (Gleason 3 + 3) to prostatectomy (Gleason 7+). Now, using an evaluable subset of 111 patients with adjacent biopsy sections, we analyzed the association between PTEN deletion in cancer and the odds of upgrading by a highly sensitive and specific four-color FISH assay. We also compared the concordance of PTEN loss by IHC and PTEN deletion by FISH. PTEN deletion was found in 27 % (12/45) of upgraded cases compared with 11 % (7/66) of controls (P = 0.03). Cancers with PTEN deletions were more likely to be upgraded than those without deletions (adjusting for age odds ratio = 3.40, 95 % confidence interval 1.14-10.11). With respect to concordance, of 93 biopsies with PTEN protein detected by IHC, 89 (96 %) had no PTEN deletion by FISH, and of 18 biopsies without PTEN protein by IHC, 15 had homozygous or hemizygous PTEN deletion by FISH. Only 4 biopsies of the 93 (4 %) with PTEN protein intact had PTEN deletion by FISH. When the regions of uncertainty in these biopsies were systematically studied by FISH, intra-tumoral variation of PTEN deletion was found, which could account for variation in immunoreactivity. Thus, FISH provides a different approach to determining PTEN loss when IHC is uncertain. Both FISH and IHC are concordant, showing consistent positive associations between PTEN loss and upgrading.


Subject(s)
Biomarkers, Tumor/analysis , In Situ Hybridization, Fluorescence , PTEN Phosphohydrolase/metabolism , Prostatic Neoplasms/chemistry , Prostatic Neoplasms/pathology , Aged , Biopsy, Needle , Humans , Immunohistochemistry/methods , In Situ Hybridization, Fluorescence/methods , Male , Middle Aged , Prostatectomy/methods
7.
Prostate Cancer Prostatic Dis ; 18(3): 264-9, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25939516

ABSTRACT

BACKGROUND: Biopsies performed for elevated serum PSA often show inflammatory infiltrates. However, the influence of intraprostatic inflammation on serum PSA in men without biopsy indication and negative for prostate cancer has not been described in detail. METHODS: We studied 224 men in the placebo arm of the Prostate Cancer Prevention Trial (PCPT) who underwent end-of-study biopsy per trial protocol, had PSA <4 ng ml(-1), normal digital rectal examination and a biopsy negative for cancer. We analyzed data from hematoxylin and eosin-stained slides containing a mean of three biopsy cores. Inflammation measures included the extent (percentage of tissue area with inflammation) and intensity (product of scores for extent and grade) of total, acute and chronic inflammation in the entire tissue area examined, and by tissue compartment. We calculated median measures of inflammation by prebiopsy serum PSA tertile (>0 to ≤0.8, >0.8 to ≤1.5 and >1.5 to <4.0 ng ml(-1)). We estimated the association between percentage of tissue area with inflammation and natural logarithm of PSA using linear regression adjusting for age at biopsy. RESULTS: Median percentage of tissue area with inflammation increased from 2 to 5 to 9.5% across PSA tertiles (P-trend <0.0001). For every 5% increase in tissue area with inflammation, log PSA increased by 0.061 ng ml(-1) (P=0.0002). Median extent and intensity scores increased across PSA tertiles in luminal and intraepithelial compartments for acute inflammation and in stromal and intraepithelial compartments for chronic inflammation (all P-trend ≤0.05). CONCLUSIONS: In men without clinical suspicion of prostate cancer, greater overall inflammation, luminal and intraepithelial acute inflammation and stromal and intraepithelial chronic inflammation were associated with higher serum PSA.


Subject(s)
Inflammation/pathology , Prostate-Specific Antigen/blood , Prostate/pathology , Prostatic Neoplasms/blood , Prostatic Neoplasms/pathology , Aged , Aged, 80 and over , Biopsy , Case-Control Studies , Cross-Sectional Studies , Humans , Male , Middle Aged , Risk Factors
8.
Prostate Cancer Prostatic Dis ; 16(2): 209-15, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23545904

ABSTRACT

BACKGROUND: ERG rearrangements and PTEN (phosphatase and tensin homolog deleted on chromosome 10) loss are two of the most common genetic alterations in prostate cancer. However, there is still significant controversy regarding the order of events of these two changes during the carcinogenic process. We used immunohistochemistry (IHC) to determine ERG and PTEN status, and calculated the fraction of cases with homogeneous/heterogeneous ERG and PTEN staining in a given tumor. METHODS: Using a single standard tissue section from the index tumor from radical prostatectomies (N=77), enriched for relatively high grade and stage tumors, we examined ERG and PTEN status by IHC. We determined whether ERG or PTEN staining was homogeneous (all tumor cells staining positive) or heterogeneous (focal tumor cell staining) in a given tumor focus. RESULTS: Fifty-seven percent (N=44/77) of tumor foci showed ERG positivity, with 93% of these (N=41/44) cases showing homogeneous ERG staining in which all tumor cells stained positively. Fifty-three percent (N=41/77) of tumor foci showed PTEN loss, and of these 66% (N=27/41) showed heterogeneous PTEN loss. In ERG homogeneously positive cases, any PTEN loss occurred in 56% (N=23/41) of cases, and of these 65% (N=15/23) showed heterogeneous loss. In ERG-negative tumors, 51.5% (N=17/33) showed PTEN loss, and of these 64.7% (N=11/17) showed heterogeneous PTEN loss. In a subset of cases, genomic deletions of PTEN were verified by fluorescence in situ hybridization in regions with PTEN protein loss as compared with regions with intact PTEN protein, which did not show PTEN genomic loss. CONCLUSIONS: These results support the concept that PTEN loss tends to occur as a subclonal event within a given established prostatic carcinoma clone after ERG gene fusion. The combination of ERG and PTEN IHC staining can be used as a simple test to ascertain PTEN and ERG gene rearrangement status within a given prostate cancer in either a research or clinical setting.


Subject(s)
Adenocarcinoma/metabolism , PTEN Phosphohydrolase/genetics , Prostatic Neoplasms/metabolism , Trans-Activators/genetics , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Adult , Aged , Carcinogenesis/genetics , Carcinogenesis/metabolism , Carcinogenesis/pathology , Disease Progression , Gene Deletion , Homozygote , Humans , Immunohistochemistry , Loss of Heterozygosity , Male , Middle Aged , Neoplasm Grading , Neoplasm Staging , Oncogene Fusion , Oncogene Proteins, Fusion/genetics , Oncogene Proteins, Fusion/metabolism , PTEN Phosphohydrolase/metabolism , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Trans-Activators/metabolism , Transcriptional Regulator ERG
9.
Oncogene ; 31(10): 1254-63, 2012 Mar 08.
Article in English | MEDLINE | ID: mdl-21822302

ABSTRACT

Alterations in nucleoli, including increased numbers, increased size, altered architecture and increased function are hallmarks of prostate cancer cells. The mechanisms that result in increased nucleolar size, number and function in prostate cancer have not been fully elucidated. The nucleolus is formed around repeats of a transcriptional unit encoding a 45S ribosomal RNA (rRNA) precursor that is then processed to yield the mature 18S, 5.8S and 28S RNA species. Although it has been generally accepted that tumor cells overexpress rRNA species, this has not been examined in clinical prostate cancer. We find that indeed levels of the 45S rRNA, 28S, 18S and 5.8S are overexpressed in the majority of human primary prostate cancer specimens as compared with matched benign tissues. One mechanism that can alter nucleolar function and structure in cancer cells is hypomethylation of CpG dinucleotides of the upstream rDNA promoter region. However, this mechanism has not been examined in prostate cancer. To determine whether rRNA overexpression could be explained by hypomethylation of these CpG sites, we also evaluated the DNA methylation status of the rDNA promoter in prostate cancer cell lines and the clinical specimens. Bisulfite sequencing of genomic DNA revealed two roughly equal populations of loci in cell lines consisting of those that contained densely methylated deoxycytidine residues within CpGs and those that were largely unmethylated. All clinical specimens also contained two populations with no marked changes in methylation of this region in cancer as compared with normal. We recently reported that MYC can regulate rRNA levels in human prostate cancer; here we show that MYC mRNA levels are correlated with 45S, 18S and 5.8S rRNA levels. Further, as a surrogate for nucleolar size and number, we examined the expression of fibrillarin, which did not correlate with rRNA levels. We conclude that rRNA levels are increased in human prostate cancer, but that hypomethylation of the rDNA promoter does not explain this increase, nor does hypomethylation explain alterations in nucleolar number and structure in prostate cancer cells. Rather, rRNA levels and nucleolar size and number relate more closely to MYC overexpression.


Subject(s)
DNA Methylation , DNA, Ribosomal/genetics , Promoter Regions, Genetic , Prostatic Neoplasms/genetics , RNA, Ribosomal/physiology , Adult , Aged , Cell Line, Tumor , CpG Islands , Gene Expression Regulation, Neoplastic , Genes, myc , Humans , Male , Middle Aged
10.
Br J Cancer ; 105(5): 602-5, 2011 Aug 23.
Article in English | MEDLINE | ID: mdl-21792196

ABSTRACT

BACKGROUND: We investigated prostate involvement during sexually transmitted infections by measuring serum prostate-specific antigen (PSA) as a marker of prostate infection, inflammation, and/or cell damage in young, male US military members. METHODS: We measured PSA before and during infection for 299 chlamydia, 112 gonorrhoea, and 59 non-chlamydial, non-gonococcal urethritis (NCNGU) cases, and 256 controls. RESULTS: Chlamydia and gonorrhoea, but not NCNGU, cases were more likely to have a large rise (40%) in PSA than controls (33.6%, 19.1%, and 8.2% vs 8.8%, P<0.0001, 0.021, and 0.92, respectively). CONCLUSION: Chlamydia and gonorrhoea may infect the prostate of some infected men.


Subject(s)
Prostate-Specific Antigen/blood , Prostate/physiology , Sexually Transmitted Diseases/etiology , Adult , Case-Control Studies , Chlamydia Infections/blood , Chlamydia Infections/epidemiology , Gonorrhea/blood , Gonorrhea/epidemiology , Humans , Male , Military Personnel/statistics & numerical data , Osmolar Concentration , Prostate/microbiology , Prostate/pathology , Prostate-Specific Antigen/analysis , Sexually Transmitted Diseases/blood , Sexually Transmitted Diseases/epidemiology , Sexually Transmitted Diseases/transmission
11.
Oncogene ; 25(40): 5561-9, 2006 Sep 07.
Article in English | MEDLINE | ID: mdl-16619041

ABSTRACT

Transforming growth factor-beta type 1 (TGF-beta) has been implicated as both a tumor suppressor and a tumor promoter in many solid epithelial cancers. We have previously demonstrated that the cyclin dependent kinase (CDK) inhibitor p21 acts as a molecular switch in determining a growth inhibitory versus growth proliferative response to TGF-beta in the spontaneously immortalized human mammary epithelial cell line MCF-10A. We now demonstrate that this proliferative effect of TGF-beta is mediated through the proinflammatory cytokine, interleukin-1alpha (IL-1alpha). Using gene expression array analysis, we identified IL-1alpha as a cytokine specifically upregulated only in cells lacking p21 and only upon TGF-beta stimulation. Cell proliferation assays verified that recombinant IL-1alpha was capable of inducing a growth proliferative response in p21 null MCF-10A cells, while neutralizing antibodies against IL-1alpha prevented the growth proliferative effects of TGF-beta. Mechanistically, both the CDK and proliferating cell nuclear antigen (PCNA) inhibitory functions of p21 were responsible for preventing TGF-beta induced cell proliferation, but only PCNA inhibition by p21 regulated IL-1alpha gene expression. These studies demonstrate a novel role for IL-1alpha in mediating a proliferative response to TGF-beta signaling, and suggest that therapies directed against IL-1alpha could abate the growth proliferative effects of TGF-beta without compromising its tumor suppressive function.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p21/genetics , Interleukin-1/metabolism , Second Messenger Systems , Transforming Growth Factor beta/metabolism , Cell Line, Tumor , Cell Proliferation , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Dactinomycin/pharmacology , Humans , Interleukin-1/genetics , Nucleic Acid Synthesis Inhibitors/pharmacology , Oligonucleotide Array Sequence Analysis , Proliferating Cell Nuclear Antigen/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Up-Regulation
12.
Urologe A ; 43(5): 573-9, 2004 May.
Article in German | MEDLINE | ID: mdl-15029477

ABSTRACT

BACKGROUND: Prostate cancer is the most commonly diagnosed cancer in men in Europe and North America. Despite its high prevalence, the molecular mechanism of its underlying development and progression is poorly understood. Many studies have revealed multiple molecular alterations during prostate cancer carcinogenesis. GSTP1 CpG island hypermethylation is one of the molecular changes that occur during carcinogenesis. METHODS: We evaluated the role of GSTP1 CpG island hypermethylation in prostatic cancers and discussed its possible role as a molecular biomarker of prostate cancer. RESULTS: Studies haven shown that GSTP1 CpG island hypermethylation is present in about 90% of prostatic carcinomas. The DNA alteration was also detectable in body fluids such as blood, urine, ejaculate, or prostatic secretions. One study showed hypermethylation in histologically unsuspicious lymph nodes in surgical specimens in patients with biochemical PSA (prostate-specific antigen) recurrence. Additionally, it is possible to distinguish between normal prostatic tissue, benign prostatic hyperplasia, and prostate cancer. CONCLUSIONS: The detection of GSTP1 CpG island hypermethylation serves as a molecular marker in prostate cancer screening, detection, and diagnosis. It may even provide information on prostate cancer prognosis. However, prospective trials to evaluate its predictive value are necessary.


Subject(s)
Acyltransferases/genetics , Biomarkers, Tumor/genetics , CpG Islands/genetics , Genetic Testing/methods , Prostatic Neoplasms/diagnosis , Prostatic Neoplasms/genetics , DNA Methylation , Gene Expression Regulation, Neoplastic/genetics , Genetic Predisposition to Disease/genetics , Humans , Male
13.
Cancer Res ; 61(24): 8611-6, 2001 Dec 15.
Article in English | MEDLINE | ID: mdl-11751372

ABSTRACT

Among the many somatic genome alterations present in cancer cells, changes in DNA methylation may represent reversible "epigenetic" lesions, rather than irreversible "genetic" alterations. Cancer cell DNA is typically characterized by increases in the methylation of CpG dinucleotides clustered into CpG islands, near the transcriptional regulatory regions of critical genes, and by an overall reduction in CpG dinucleotide methylation. The transcriptional "silencing" of gene expression associated with such CpG island DNA hypermethylation presents an attractive therapeutic target: restoration of "silenced" gene expression may be possible via therapeutic reversal of CpG island hypermethylation. 5-Aza-cytidine (5-aza-C) and 5-aza-deoxycytidine (5-aza-dC), nucleoside analogue inhibitors of DNA methyltransferases, have been widely used in attempts to reverse abnormal DNA hypermethylation in cancer cells and restore "silenced" gene expression. However, clinical utility of the nucleoside analogue DNA methyltransferase inhibitors has been limited somewhat by myelosuppression and other side effects. Many of these side effects are characteristic of nucleoside analogues that are not DNA methyltransferase inhibitors, offering the possibility that nonnucleoside analogue DNA methyltransferase inhibitors might not possess such side effects. Human prostate cancer (PCA) cells characteristically contain hypermethylated CpG island sequences encompassing the transcriptional regulatory region of GSTP1, the gene encoding the pi-class glutathione S-transferase (GSTP1), and fail to express GSTP1 as a consequence of transcriptional "silencing." Inactivation of GSTP1 by CpG island hypermethylation, the most common somatic genome alteration yet reported for human PCAs, occurs early during human prostatic carcinogenesis and results in a loss of GSTP1 "caretaker" function, leaving prostate cells with inadequate defenses against oxidant and electrophile carcinogens. We report here that the drug procainamide, a nonnucleoside inhibitor of DNA methyltransferases, reversed GSTP1 CpG island hypermethylation and restored GSTP1 expression in LNCaP human PCA cells propagated in vitro or in vivo as xenograft tumors in athymic nude mice.


Subject(s)
DNA Methylation/drug effects , Enzyme Inhibitors/pharmacology , Glutathione Transferase/genetics , Isoenzymes/genetics , Procainamide/pharmacology , Prostatic Neoplasms/genetics , Animals , CpG Islands/physiology , DNA Modification Methylases/antagonists & inhibitors , Enzyme Activation/drug effects , Gene Expression Regulation, Enzymologic/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Gene Silencing/drug effects , Glutathione S-Transferase pi , Glutathione Transferase/biosynthesis , Humans , Isoenzymes/biosynthesis , Male , Mice , Mice, Nude , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/enzymology , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
14.
Cancer Res ; 61(24): 8617-23, 2001 Dec 15.
Article in English | MEDLINE | ID: mdl-11751373

ABSTRACT

Cyclooxygenase-2 (COX-2) is the inducible isoform of the rate-limiting enzymes that convert arachidonic acid to proinflammatory prostaglandins as well as a primary target for nonsteroidal anti-inflammatory drugs. Accumulating evidence suggests that up-regulation of COX-2 is associated with carcinogenesis in multiple organ systems including the large bowel, lung, breast, and prostate. In this report, we examine the expression of COX-2 protein and mRNA in prostate tissue containing various lesions and in prostate cancer cell lines. In the cell lines, LNCaP, DU145, PC-3, and TSU, COX-2 protein expression was undetectable under basal conditions but could be induced transiently by phorbol ester treatment in PC-3 and TSU cells, but not in DU145 and LNCaP cells. Immunohistochemical analysis of 144 human prostate cancer cases suggested that, in contrast to several previous reports, there was no consistent overexpression of COX-2 in established prostate cancer or high-grade prostatic intraepithelial neoplasia, as compared with adjacent normal prostate tissue. Positive staining was seen only in scattered cells (<1%) in both tumor and normal tissue regions but was much more consistently observed in areas of proliferative inflammatory atrophy, lesions that have been implicated in prostatic carcinogenesis. Staining was also seen at times in macrophages. Western blotting and quantitative RT-PCR analyses confirmed these patterns of expression. These results suggest that if nonsteroidal anti-inflammatory drugs are indeed chemopreventive and/or chemotherapeutic for prostate cancer, their effects are likely to be mediated by modulating COX-2 activity in non-PCa cells (either inflammatory cells or atrophic epithelial cells) or by affecting a COX-2-independent pathway.


Subject(s)
Isoenzymes/biosynthesis , Prostaglandin-Endoperoxide Synthases/biosynthesis , Prostate/pathology , Prostatic Neoplasms/enzymology , Atrophy/enzymology , Blotting, Western , Cyclooxygenase 2 , Disease Progression , Epithelium/enzymology , Epithelium/pathology , Humans , Immunohistochemistry , Isoenzymes/genetics , Male , Membrane Proteins , Prostaglandin-Endoperoxide Synthases/genetics , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction , Stromal Cells/enzymology , Stromal Cells/pathology , Tumor Cells, Cultured , Up-Regulation
15.
Am J Pathol ; 159(5): 1815-26, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11696442

ABSTRACT

GSTP1 CpG island hypermethylation is the most common somatic genome alteration described for human prostate cancer (PCA); lack of GSTP1 expression is characteristic of human PCA cells in vivo. We report here that loss of GSTP1 function may have been selected during the pathogenesis of human PCA. Using a variety of techniques to detect GSTP1 CpG island DNA hypermethylation in PCA DNA, we found only hypermethylated GSTP1 alleles in each PCA cell in all but two PCA cases studied. In these two cases, CpG island hypermethylation was present at only one of two GSTP1 alleles in PCA DNA. In one of the cases, DNA hypermethylation at one GSTP1 allele and deletion of the other GSTP1 allele were evident. In the other case, an unmethylated GSTP1 allele was detected, accompanied by abundant GSTP1 expression. GSTP1 CpG island DNA hypermethylation was responsible for lack of GSTP1 expression by LNCaP PCA cells: treatment of the cells with 5-azacytidine (5-aza-C), an inhibitor of DNA methyltransferases, reversed the GSTP1 promoter DNA hypermethylation, activated GSTP1 transcription, and restored GSTP1 expression. GSTP1 promoter activity, assessed via transfection of GSTP1 promoter-CAT reporter constructs in LNCaP cells, was inhibited by SssI-catalyzed CpG dinucleotide methylation. Remarkably, although selection for loss of GSTP1 function may be inferred for human PCA, GSTP1 did not act like a tumor suppressor gene, as LNCaP cells expressing GSTP1, either after 5-aza-C treatment or as a consequence of transfection with GSTP1 cDNA, grew well in vitro and in vivo. Perhaps, GSTP1 inactivation may render prostatic cells susceptible to additional genome alterations, caused by electrophilic or oxidant carcinogens, that provide a selective growth advantage.


Subject(s)
CpG Islands/physiology , Glutathione Transferase/genetics , Glutathione Transferase/metabolism , Isoenzymes/genetics , Isoenzymes/metabolism , Prostatic Neoplasms/metabolism , Alleles , Base Sequence/genetics , Blotting, Southern , Carcinogenicity Tests , Cell Division/physiology , CpG Islands/genetics , DNA, Neoplasm/genetics , Glutathione S-Transferase pi , Glutathione Transferase/antagonists & inhibitors , Glutathione Transferase/deficiency , Humans , Isoenzymes/antagonists & inhibitors , Isoenzymes/deficiency , Male , Methylation , Prostatic Neoplasms/pathology , Reference Values , Tumor Cells, Cultured
16.
Urology ; 58(4): 619-24, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11597556

ABSTRACT

OBJECTIVES: To examine the expression of the p63 protein in normal, preneoplastic, and neoplastic human prostatic tissue. The p63 gene, a member of the p53 gene family, is expressed in the basal epithelial cells of multiple organs. Irregularities in p63 expression have been associated with epithelial carcinogenesis. METHODS: We performed immunohistochemistry with an anti-p63 antibody on specimens from radical prostatectomies, prostate needle biopsies, and metastatic prostate adenocarcinoma. We analyzed p63 expression in regions of normal prostate, benign prostatic hyperplasia, proliferative inflammatory atrophy (PIA), high-grade intraepithelial neoplasia, and adenocarcinoma. RESULTS: Basal epithelial cells in normal, benign prostatic hyperplasia, and high-grade intraepithelial neoplasia tissue stained intensely for the p63 polypeptide, but the vast majority of adenocarcinoma specimens from 233 patients-66 (94%) of 70 radical prostatectomies, 132 (89%) of 148 prostate needle biopsies, and 14 (93%) of 15 metastases-did not. In tumors in which the adenocarcinoma cells were positive, the staining intensity was weak and occurred in less than 1% of the cells. Tumors that stained positive for p63 were more likely to be high grade than those that did not (P <0.0001). Basal cells in PIA expressed p63, but these cells were sparsely distributed relative to the basal cells in the normal glands. Luminal cells in PIA were, in general, negative for p63. CONCLUSIONS: In contrast to normal and preneoplastic prostatic tissue, the vast majority of prostate adenocarcinomas do not express p63. Therefore, p63 immunohistochemistry represents a potential novel adjuvant method for facilitating the pathologic diagnosis of prostate cancer in prostate needle biopsies. The selective expression of p63 in normal basal cells, coupled with the finding that p63 null mice fail to develop prostates, provides strong evidence that the basal cells represent prostatic epithelial stem cells. In addition, these findings suggest that p63 may protect prostatic epithelial cells against neoplastic transformation and support the hypothesis that intermediately differentiated cells in the luminal epithelium of PIA are the targets of neoplastic transformation in the prostate.


Subject(s)
Adenocarcinoma/chemistry , Membrane Proteins , Phosphoproteins/analysis , Prostatic Neoplasms/chemistry , Trans-Activators/analysis , Adenocarcinoma/diagnosis , Adenocarcinoma/secondary , Adult , Aged , Aged, 80 and over , Biopsy, Needle , Bone Neoplasms/secondary , Cell Differentiation , DNA-Binding Proteins , Epithelium/chemistry , Genes, Tumor Suppressor , Humans , Immunohistochemistry , Keratins/analysis , Lymphatic Metastasis , Male , Middle Aged , Precancerous Conditions/diagnosis , Prostate/chemistry , Prostatic Hyperplasia/pathology , Prostatic Neoplasms/diagnosis , Stem Cells/pathology , Transcription Factors , Tumor Suppressor Proteins
17.
Am J Pathol ; 159(3): 837-43, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11549576

ABSTRACT

With the completion of the Human Genome Project and high-throughput screening methods using cDNA array and tissue microarray (TMA) technology, there is a pressing need to manage the voluminous data sets generated from these types of investigations. Herein is described a database model to handle 1) clinical and pathology data, 2) TMA location information, and 3) web-based histology results. The model is useful for managing clinical, pathology, and molecular data on >1300 prostate cancer patients dating back to 1995 from the University of Michigan Specialized Program of Research Excellence for prostate cancer. The key components in this multidatabase model are 1) the TMA database, 2) the TMA-image database (TMA-I DB), and 3) the prostate pathology and clinical information databases. All databases were created in Microsoft Access (Microsoft, Redmond, WA). Desired patient, tissue, block, diagnosis, array location, and respective clinical and pathology information is obtained by linking the unique identifier fields among database tables. The TMA database is comprised of interrelated data from 336 prostate cancer patients transferred into 19 TMA blocks with 5451 TMA biopsy cores. Tissue samples include 1695 normal prostate, 3171 prostate cancer, 464 prostatic intraepithelial neoplasia, and 121 atrophy. All 19 TMA blocks have been analyzed over the Internet for several immunohistochemical biomarkers including E-cadherin, prostate-specific antigen, p27(Kip1), and Ki-67 labeling index. This system facilitates the statistical analysis of high-density TMA data with clinical and pathology information in an efficient and cost-effective manner. Because the review is performed over the Internet, this system is ideal for collaborative multi-institutional studies.


Subject(s)
Databases as Topic , Oligonucleotide Array Sequence Analysis , Pathology/methods , Prostate/pathology , Prostatic Neoplasms/pathology , Humans , Male
18.
Prostate ; 49(1): 30-7, 2001 Sep 15.
Article in English | MEDLINE | ID: mdl-11550208

ABSTRACT

BACKGROUND: Glutathione S-transferases (GSTs), inducible enzymes that catalyze the detoxification of reactive electrophiles and oxidants, protect against neoplastic transformation. Prostatic adenocarcinoma and high-grade prostatic intraepithelial neoplasia (HGPIN) fail to express GSTP1, a major class of GST. This failure of expression is associated with methlyation of the GSTP1 promoter, a somatic alteration proposed to be a critical step in prostatic carcinogenesis. However, simple atrophy and post-atrophic hyperplasia-proliferative lesions associated with chronic inflammation, which we have termed "proliferative inflammatory atrophy" (PIA)-express elevated levels of GSTP1. We postulated that this increase in GSTP1 expression in PIA occurs in response to increased oxidative stress. We examined the expression of another major class of GST, GSTA1, in the human prostate. METHODS: We performed immunohistochemistry against GSTA1 on formalin-fixed radical prostatectomies (n = 45). A stereological grid point counting method was used to estimate the percent of cells staining positive for GSTA1 in normal prostate, PIA, HGPIN, and adenocarcinoma. RESULTS: In contrast to GSTP1, normal peripheral zone epithelium was virtually devoid of GSTA1. Strikingly, though, epithelial cells in PIA demonstrated strong staining for GSTA1 (median of percent of cells staining positive = 44) as compared to those in normal peripheral zone (median = 3.0, P <.00001), HGPIN (median = 2.9, P <.00001), and adenocarcinoma (median = 3.8, P <.00001). Variations in GSTA1 were also detected between normal anatomic zones: the central zone showed an increase in the percentage of cells staining positive (median = 20.9) as compared to the transition (median = 0.47, P <.0002) and the peripheral (P <.0001) zones. CONCLUSIONS: Expression of GSTA1 is increased in PIA, supporting the concept that cells within these lesions are subject to localized increases in oxidative stress. Low levels of GSTA1 and GSTP1 in HGPIN and adenocarcinoma suggest a broad lack of detoxification activity in these cells, which may be associated with carcinogenesis in the prostate.


Subject(s)
Adenocarcinoma/pathology , Gene Expression Regulation, Neoplastic , Glutathione Transferase/biosynthesis , Oxidative Stress/physiology , Prostate/physiology , Prostatic Neoplasms/pathology , Adenocarcinoma/surgery , Glutathione S-Transferase pi , Glutathione Transferase/genetics , Humans , Immunohistochemistry , Isoenzymes/biosynthesis , Isoenzymes/genetics , Male , Precancerous Conditions/pathology , Prostatectomy , Prostatic Intraepithelial Neoplasia/pathology , Prostatic Neoplasms/surgery
19.
Hematol Oncol Clin North Am ; 15(3): 407-21, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11525288

ABSTRACT

The field of prostate cancer research is poised for dramatic improvements in our ability to better diagnose men at risk of prostate cancer and to better predict prognosis and response to treatment. Histopathologic and molecular analyses lie at the heart of these issues. Improvements in our understanding of the mechanisms of prostate carcinogenesis and in determining why the prostate seems to be so highly targeted for cancer development will lead to rational strategies of disease prevention.


Subject(s)
Adenocarcinoma/pathology , Prostate/pathology , Prostatic Neoplasms/pathology , Adenocarcinoma/chemistry , Adenocarcinoma/diagnosis , Adenocarcinoma/epidemiology , Adenocarcinoma/therapy , Age of Onset , Aged , Atrophy , Biomarkers, Tumor/analysis , Biopsy, Needle/methods , Humans , Hyperplasia , Incidence , Male , Middle Aged , Neoplasm Metastasis , Neoplasm Proteins/analysis , Prognosis , Prostate/chemistry , Prostate-Specific Antigen/blood , Prostatic Intraepithelial Neoplasia/chemistry , Prostatic Intraepithelial Neoplasia/diagnosis , Prostatic Intraepithelial Neoplasia/epidemiology , Prostatic Intraepithelial Neoplasia/pathology , Prostatic Neoplasms/chemistry , Prostatic Neoplasms/diagnosis , Prostatic Neoplasms/epidemiology , Prostatic Neoplasms/therapy , Prostatitis/blood , Prostatitis/pathology , Staining and Labeling/methods
20.
Prostate ; 48(3): 128-30, 2001 Aug 01.
Article in English | MEDLINE | ID: mdl-11494327
SELECTION OF CITATIONS
SEARCH DETAIL