Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 121
Filter
1.
Oncogene ; 35(48): 6235-6245, 2016 12 01.
Article in English | MEDLINE | ID: mdl-27641331

ABSTRACT

Restoration of tumor suppression is an attractive onco-therapeutic approach. It is particularly relevant when a tumor suppressor is excessively degraded by an overactive oncogenic E3 ligase. We previously discovered that the E6-associated protein (E6AP; as classified in the human papilloma virus context) is an E3 ligase that has an important role in the cellular stress response, and it directly targets the tumor-suppressor promyelocytic leukemia protein (PML) for proteasomal degradation. In this study, we have examined the role of the E6AP-PML axis in prostate cancer (PC). We show that knockdown (KD) of E6AP expression attenuates growth of PC cell lines in vitro. We validated this finding in vivo using cell line xenografts, patient-derived xenografts and mouse genetics. We found that KD of E6AP attenuates cancer cell growth by promoting cellular senescence in vivo, which correlates with restoration of tumor suppression by PML. In addition, we show that KD of E6AP sensitizes cells to radiation-induced death. Overall, our findings demonstrate a role for E6AP in the promotion of PC and support E6AP targeting as a novel approach for PC treatment, either alone or in combination with radiation.


Subject(s)
Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Ubiquitin-Protein Ligases/genetics , Animals , Cell Cycle/genetics , Cell Line, Tumor , Cell Proliferation , Cell Survival/genetics , Cellular Senescence/genetics , Disease Models, Animal , Down-Regulation , Gene Expression , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Heterografts , Humans , Male , Mice , Prognosis , Promyelocytic Leukemia Protein/genetics , Promyelocytic Leukemia Protein/metabolism , Prostatic Neoplasms/mortality , RNA, Small Interfering/genetics , Stress, Physiological , Tumor Burden
2.
Br J Cancer ; 106(4): 768-74, 2012 Feb 14.
Article in English | MEDLINE | ID: mdl-22240788

ABSTRACT

BACKGROUND: The aim of this study was to investigate the hypothesis that changes in circulating microRNAs (miRs) represent potentially useful biomarkers for the diagnosis, staging and prediction of outcome in prostate cancer. METHODS: Real-time polymerase chain reaction analysis of 742 miRs was performed using plasma-derived circulating microvesicles of 78 prostate cancer patients and 28 normal control individuals to identify differentially quantified miRs. RESULTS: A total of 12 miRs were differentially quantified in prostate cancer patients compared with controls, including 9 in patients without metastases. In all, 11 miRs were present in significantly greater amounts in prostate cancer patients with metastases compared with those without metastases. The association of miR-141 and miR-375 with metastatic prostate cancer was confirmed using serum-derived exosomes and microvesicles in a separate cohort of patients with recurrent or non-recurrent disease following radical prostatectomy. An analysis of five selected miRs in urine samples found that miR-107 and miR-574-3p were quantified at significantly higher concentrations in the urine of men with prostate cancer compared with controls. CONCLUSION: These observations suggest that changes in miR concentration in prostate cancer patients may be identified by analysing various body fluids. Moreover, circulating miRs may be used to diagnose and stage prostate cancer.


Subject(s)
MicroRNAs/blood , Prostatic Neoplasms/genetics , Aged , Biomarkers, Tumor/genetics , Humans , Male , MicroRNAs/urine , Neoplasm Metastasis , Prognosis , Prostate-Specific Antigen/analysis , Prostatic Neoplasms/blood , Prostatic Neoplasms/pathology
3.
Oncogene ; 31(45): 4759-67, 2012 Nov 08.
Article in English | MEDLINE | ID: mdl-22266865

ABSTRACT

Reactivation of the androgen receptor (AR) during androgen depletion therapy (ADT) underlies castration-resistant prostate cancer (CRPCa). Alternative splicing of the AR gene and synthesis of constitutively active COOH-terminally truncated AR variants lacking the AR ligand-binding domain has emerged as an important mechanism of ADT resistance in CRPCa. In a previous study, we demonstrated that altered AR splicing in CRPCa 22Rv1 cells was linked to a 35-kb intragenic tandem duplication of AR exon 3 and flanking sequences. In this study, we demonstrate that complex patterns of AR gene copy number imbalances occur in PCa cell lines, xenografts and clinical specimens. To investigate whether these copy number imbalances reflect AR gene rearrangements that could be linked to splicing disruptions, we carried out a detailed analysis of AR gene structure in the LuCaP 86.2 and CWR-R1 models of CRPCa. By deletion-spanning PCR, we discovered a 8579-bp deletion of AR exons 5, 6 and 7 in the LuCaP 86.2 xenograft, which provides a rational explanation for synthesis of the truncated AR v567es AR variant in this model. Similarly, targeted resequencing of the AR gene in CWR-R1 cells led to the discovery of a 48-kb deletion in AR intron 1. This intragenic deletion marked a specific CWR-R1 cell population with enhanced expression of the truncated AR-V7/AR3 variant, a high level of androgen-independent AR transcriptional activity and rapid androgen independent growth. Together, these data demonstrate that structural alterations in the AR gene are linked to stable gain-of-function splicing alterations in CRPCa.


Subject(s)
Alternative Splicing , Gene Deletion , Gene Expression Regulation, Neoplastic , Prostatic Neoplasms/genetics , Receptors, Androgen/genetics , Animals , Cell Line, Tumor , DNA Copy Number Variations , Disease Models, Animal , Exons , Gene Order , Humans , Introns , Male , Mice , Orchiectomy , Prostatic Neoplasms/metabolism , RNA Stability , Receptors, Androgen/metabolism , Transplantation, Heterologous
4.
Oncogene ; 31(17): 2153-63, 2012 Apr 26.
Article in English | MEDLINE | ID: mdl-21909140

ABSTRACT

Androgen receptor (AR) is overexpressed in the majority of castration-resistant prostate cancers (CRPCs). Our goal was to study the effect of AR overexpression on the chromatin binding of the receptor and to identify AR target genes that may be important in the emergence of CRPC. We have established two sublines of LNCaP prostate cancer (PC) cell line, one overexpressing AR 2-3-fold and the other 4-5-fold compared with the control cells. We used chromatin immunoprecipitation (ChIP) and deep-sequencing (seq) to identify AR-binding sites (ARBSs). We found that the number of ARBSs and the AR-binding strength were positively associated with the level of AR when cells were stimulated with low concentrations of androgens. In cells overexpressing AR, the chromatin binding of the receptor took place in 100-fold lower concentration of the ligand than in control cells. We confirmed the association of AR level and chromatin binding in two PC xenografts, one containing AR gene amplification with high AR expression, and the other with low expression. By combining the ChIP-seq and expression profiling, we identified AR target genes that are upregulated in PC. Of them, the expression of ZWINT, SKP2 (S-phase kinase-associated protein 2 (p45)) and FEN1 (flap structure-specific endonuclease 1) was demonstrated to be increased in CRPC, while the expression of SNAI2 was decreased in both PC and CRPC. FEN1 protein expression was also associated with poor prognosis in prostatectomy-treated patients. Finally, the knock-down of FEN1 with small interfering RNA inhibited the growth of LNCaP cells. Our data demonstrate that the overexpression of AR sensitizes the receptor binding to chromatin, thus, explaining how AR signaling pathway is reactivated in CRPC cells.


Subject(s)
Chromatin/metabolism , Prostatic Neoplasms/metabolism , Receptors, Androgen/metabolism , Animals , Binding Sites , Cell Line, Tumor , Flap Endonucleases/genetics , Gene Amplification , Gene Expression Profiling , Humans , Intracellular Signaling Peptides and Proteins/genetics , Male , Mice , Nuclear Proteins/genetics , Nucleic Acid Amplification Techniques , Prostatic Neoplasms/genetics , Receptors, Androgen/genetics , S-Phase Kinase-Associated Proteins/genetics , Transplantation, Heterologous
5.
Oncogene ; 31(35): 3939-48, 2012 Aug 30.
Article in English | MEDLINE | ID: mdl-22139082

ABSTRACT

A clearer definition of the molecular determinants that drive the development and progression of prostate cancer (PCa) is urgently needed. Efforts to map recurrent somatic deletions in the tumor genome, especially homozygous deletions (HODs), have provided important positional information in the search for cancer-causing genes. Analyzing HODs in the tumors of 244 patients from two independent cohorts and 22 PCa xenografts using high-resolution single-nucleotide polymorphism arrays, herein we report the identification of CHD1, a chromatin remodeler, as one of the most frequently homozygously deleted genes in PCa, second only to PTEN in this regard. The HODs observed in CHD1, including deletions affecting only internal exons of CHD1, were found to completely extinguish the expression of mRNA of this gene in PCa xenografts. Loss of this chromatin remodeler in clinical specimens is significantly associated with an increased number of additional chromosomal deletions, both hemi- and homozygous, especially on 2q, 5q and 6q. Together with the deletions observed in HEK293 cells stably transfected with CHD1 small hairpin RNA, these data suggest a causal relationship. Downregulation of Chd1 in mouse prostate epithelial cells caused dramatic morphological changes indicative of increased invasiveness, but did not result in transformation. Indicating a new role of CHD1, these findings collectively suggest that distinct CHD1-associated alterations of genomic structure evolve during and are required for the development of PCa.


Subject(s)
Chromatin Assembly and Disassembly , DNA Helicases/genetics , DNA Helicases/physiology , DNA-Binding Proteins/genetics , DNA-Binding Proteins/physiology , Gene Deletion , Prostatic Neoplasms/genetics , Animals , Cell Line , Down-Regulation , HEK293 Cells , Homozygote , Humans , Male , Mice , Neoplasm Transplantation , Oligonucleotide Array Sequence Analysis , PTEN Phosphohydrolase/genetics , Polymorphism, Single Nucleotide , RNA Interference , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/genetics , Transplantation, Heterologous
6.
Br J Cancer ; 101(2): 263-8, 2009 Jul 21.
Article in English | MEDLINE | ID: mdl-19603032

ABSTRACT

BACKGROUND: Dasatinib is a small molecule kinase inhibitor that has recently been shown to inhibit Src family kinases (SFK) and also has activity against CaP. Of importance to metastatic CaP, which frequently metastasises to bone, SFK are also vital to the regulation of bone remodelling. We sought to determine the ability of dasatinib to inhibit growth of CaP in bone. METHODS: C4-2B CaP cells were injected into tibiae of SCID mice and treated with dasatinib, alone or in combination with docetaxel. Serum prostate-specific antigen levels, bone mineral density, radiographs and histology were analysed. RESULTS: Treatment with dasatinib alone significantly lowered sacrifice serum prostate-specific antigen levels compared to control, 2.3+/-0.4 vs 9.2+/-2.1 (P=0.004). Combination therapy improved efficacy over dasatinib alone (P=0.010). Dasatinib increased bone mineral density in tumoured tibiae by 25% over control tumoured tibiae (P<0.001). CONCLUSION: Dasatinib inhibits growth of C4-2B cells in bone with improved efficacy when combined with docetaxel. Additionally, dasatinib inhibits osteolysis associated with CaP. These data support further study of dasatinib in clinical trials for men with CaP bone metastases.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Bone Neoplasms/drug therapy , Bone Neoplasms/secondary , Osteolysis/drug therapy , Prostatic Neoplasms/drug therapy , Protein Kinase Inhibitors/pharmacology , Pyrimidines/pharmacology , Thiazoles/pharmacology , Animals , Bone Density/drug effects , Bone Neoplasms/blood , Cell Line, Tumor , Dasatinib , Docetaxel , Humans , Male , Mice , Mice, SCID , Osteolysis/blood , Prostate-Specific Antigen/blood , Prostatic Neoplasms/blood , Prostatic Neoplasms/pathology , Protein Kinase Inhibitors/adverse effects , Pyrimidines/adverse effects , Taxoids/administration & dosage , Thiazoles/adverse effects , Xenograft Model Antitumor Assays , src-Family Kinases/antagonists & inhibitors
7.
J Pathol ; 212(4): 395-401, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17503439

ABSTRACT

BAG-1L (Bcl-2-associated anthanogene 1) has been found to interact with androgen receptor (AR), and has been suggested to be involved in the development of prostate cancer. In order to determine the presence of genetic and/or expression alterations of BAG-1L in prostate cancer, we analysed human prostate cancer cell lines and xenografts as well as patient samples of untreated, hormone-naïve, and hormone-refractory prostate carcinomas for sequence variations using denaturing high-performance liquid chromatography (DHPLC), for gene copy number using fluorescence in situ hybridization (FISH), and for expression using both quantitative RT-PCR and immunostaining. Only one sequence variation was found in all 37 cell lines and xenografts analysed. BAG-1 gene amplification was detected in two xenografts. In addition, gene amplification was found in 6 of 81 (7.4%) hormone-refractory clinical tumours, whereas no amplification was found in any of the 130 untreated tumours analysed. Additionally, gain of the BAG-1 gene was observed in 27.2% of the hormone-refractory tumours and in 18.5% of the untreated carcinomas. In a set of 263 patient samples, BAG-1L protein expression was significantly higher in hormone-refractory tumours than in primary tumours (p = 0.002). Altogether, these data suggest that amplification and overexpression of BAG-1L may be involved in the progression of prostate cancer.


Subject(s)
DNA-Binding Proteins/metabolism , Neoplasm Proteins/metabolism , Prostatic Neoplasms/metabolism , Transcription Factors/metabolism , Antineoplastic Agents, Hormonal/therapeutic use , Cell Line, Tumor , Chromatography, High Pressure Liquid/methods , DNA Mutational Analysis/methods , DNA-Binding Proteins/genetics , Follow-Up Studies , Gene Expression Regulation, Neoplastic , Humans , Male , Neoplasm Proteins/genetics , Neoplasm Transplantation , Orchiectomy , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Prostatic Neoplasms/therapy , RNA, Messenger/genetics , RNA, Neoplasm/genetics , Receptors, Androgen/metabolism , Reverse Transcriptase Polymerase Chain Reaction/methods , Transcription Factors/genetics , Transplantation, Heterologous , Treatment Failure
8.
Oncogene ; 26(16): 2386-94, 2007 Apr 05.
Article in English | MEDLINE | ID: mdl-17016436

ABSTRACT

The gene for E3 ubiquitin ligase WWP1 is located at 8q21, a region frequently amplified in human cancers, including prostate cancer. Recent studies have shown that WWP1 negatively regulates the TGFbeta tumor suppressor pathway by inactivating its molecular components, including Smad2, Smad4 and TbetaR1. These findings suggest an oncogenic role of WWP1 in carcinogenesis, but direct supporting evidence has been lacking. In this study, we examined WWP1 for gene dosage, mRNA expression, mutation and functions in a number of human prostate cancer samples. We found that the WWP1 gene had copy number gain in 15 of 34 (44%) xenografts and cell lines from prostate cancer and 15 of 49 (31%) clinical prostate cancer samples. Consistently, WWP1 was overexpressed in 60% of xenografts and cell lines from prostate cancer. Mutation of WWP1 occurred infrequently in prostate cancer. Functionally, WWP1 overexpression promoted colony formation in the 22Rv1 prostate cancer cell line. In PC-3 prostate cancer cells, WWP1 knockdown significantly suppressed cell proliferation and enhanced TGFbeta-mediated growth inhibition. These findings suggest that WWP1 is an oncogene that undergoes genomic amplification at 8q21 in human prostate cancer, and WWP1 overexpression is a common mechanism involved in the inactivation of TGFbeta function in human cancer.


Subject(s)
Prostatic Neoplasms/genetics , Ubiquitin-Protein Ligases/genetics , Animals , Cell Line, Tumor , Chromosomes, Human, Pair 8 , Gene Amplification , Gene Dosage , Gene Expression Regulation, Neoplastic , Humans , Male , RNA, Messenger/genetics , Transforming Growth Factor beta/antagonists & inhibitors , Transplantation, Heterologous
9.
Prostate Cancer Prostatic Dis ; 8(3): 253-9, 2005.
Article in English | MEDLINE | ID: mdl-15999121

ABSTRACT

Zoledronic acid (ZA) has been shown to inhibit prostate tumor growth in vitro and have beneficial effects in patients with advanced prostate cancer (CaP). The aim of this study was to determine whether ZA exhibits direct anti-tumor effects on CaP cells in vivo. To distinguish the effects of inhibition of osteolysis and direct anti-tumor activity of ZA in vivo, we compared the results of treatment with ZA and osteoprotegerin (Fc-OPG), which inhibits osteolysis, but without significant direct anti-tumor effects. In vitro Fc-OPG had no significant effects on C4-2 proliferation, whereas ZA decreased proliferation. However, both agents decreased tumor growth in bone. Moreover, both increased bone volume and prevented the overall decreases in BMD associated with growth of C4-2 cells in bone. Our study provides novel and significant observations that the in vivo effects of ZA are consistent with indirect effects mediated by osteoclasts.


Subject(s)
Antibodies/therapeutic use , Antineoplastic Agents/therapeutic use , Bone Neoplasms/drug therapy , Bone Neoplasms/secondary , Diphosphonates/pharmacology , Glycoproteins/chemistry , Glycoproteins/immunology , Imidazoles/pharmacology , Immunoglobulin Fc Fragments/therapeutic use , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/pathology , Receptors, Cytoplasmic and Nuclear/chemistry , Receptors, Cytoplasmic and Nuclear/immunology , Receptors, Tumor Necrosis Factor/chemistry , Receptors, Tumor Necrosis Factor/immunology , Animals , Apoptosis , Bone Density , Bone Density Conservation Agents/pharmacology , Cell Proliferation , Densitometry , Diphosphonates/chemistry , Glycoproteins/pharmacology , Humans , Imidazoles/chemistry , In Vitro Techniques , Male , Mice , Mice, SCID , Neoplasm Metastasis , Osteoclasts/metabolism , Osteolysis , Osteoprotegerin , Tibia/pathology , Time Factors , Zoledronic Acid
10.
J Clin Pathol ; 57(3): 321-3, 2004 Mar.
Article in English | MEDLINE | ID: mdl-14990610

ABSTRACT

Conventional prostate adenocarcinomas consist mainly of tumour cells of luminal immunophenotype with scattered neuroendocrine (NE) cells. NE cells are defined by chromogranin A (CGA) immunoreactivity. Unlike luminal cells, NE cells lack androgen receptor (AR) and prostate specific antigen (PSA) immunoreactivity. This report describes the first case of conventional prostate adenocarcinoma expressing CGA, PSA, and AR as determined by immunohistochemistry. A 64 year old man was diagnosed with conventional prostate adenocarcinoma in 1993; he underwent cystoprostatectomy in 1994; he developed an iliac bone metastasis in 1997 and mediastinal lymph node metastases in 1999. All specimens obtained during the progression of the disease consisted primarily of luminal cells with only scattered NE cells. In contrast, in samples of non-osseous and osseous metastases obtained at necropsy in 2001, greater than 80% of tumour cells were shown to express PSA, AR, and CGA. This suggests that during tumour progression, conventional prostate adenocarcinomas may evolve into an NE cell phenotype.


Subject(s)
Adenocarcinoma/chemistry , Chromogranins/analysis , Prostatic Neoplasms/chemistry , Receptors, Androgen/analysis , Adenocarcinoma/pathology , Adenocarcinoma/secondary , Biomarkers, Tumor/analysis , Bone Neoplasms/secondary , Chromogranin A , Fatal Outcome , Humans , Immunohistochemistry , Lymphatic Metastasis , Male , Middle Aged , Neuroendocrine Tumors/pathology , Prostate-Specific Antigen/analysis , Prostatic Neoplasms/pathology
11.
J Cell Biochem ; 91(1): 151-60, 2004 Jan 01.
Article in English | MEDLINE | ID: mdl-14689587

ABSTRACT

Prostate cancer is the most commonly diagnosed malignancy in men and is often associated with bone metastases. Prostate cancer bone lesions can be lytic or schlerotic, with the latter predominating. Bone morphogenetic proteins (BMPs) are a family of growth factors, which may play a role in the formation of prostate cancer osteoblastic bone metastases. This study evaluated the effects of BMPs on prostate cancer cell lines. We observed growth inhibitory effects of BMP-2 and -4 on LNCaP, while PC-3 was unaffected. Flow cytometric analysis determined that LNCaP cell growth was arrested in G(1) after bone morphogenetic protein-2 treatment. Treatment of LNCaP and PC-3 with BMP-2 and -4 activated downstream signaling pathways involving SMAD-1, up-regulation of p21(CIP1/WAF1) and changes in retinoblastoma (Rb) phosphorylation. Interestingly, bone morphogenetic protein-2 treatment stimulated a 2.7-fold increase in osteoprotegerin (OPG), a molecule, which inhibits osteoclastogenesis, production in PC-3.


Subject(s)
Bone Morphogenetic Proteins/pharmacology , Bone Neoplasms/metabolism , Prostatic Neoplasms/metabolism , Signal Transduction/physiology , Transforming Growth Factor beta , Bone Morphogenetic Protein 2 , Bone Morphogenetic Protein 4 , Bone Neoplasms/secondary , Cell Division/drug effects , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/metabolism , DNA-Binding Proteins/metabolism , Flow Cytometry , G1 Phase/physiology , Glycoproteins/metabolism , Humans , Male , Osteoprotegerin , Phosphorylation/drug effects , Receptors, Cytoplasmic and Nuclear/metabolism , Receptors, Tumor Necrosis Factor , Retinoblastoma Protein/metabolism , Smad Proteins , Smad1 Protein , Trans-Activators/metabolism , Tumor Cells, Cultured , Up-Regulation/drug effects
12.
Clin Exp Metastasis ; 21(5): 381-7, 2004.
Article in English | MEDLINE | ID: mdl-15672862

ABSTRACT

Osteoprotegerin (OPG) plays a central role in controlling bone resorption. Exogenous administration of OPG has been shown to be effective in preventing osteolysis and limiting the growth of osteolytic metastasis. The objective of this study was to investigate the effects of OPG on osteoblastic prostate cancer (CaP) metastases in an animal model. LuCaP 23.1 cells were injected intra-tibially and Fc-OPG (6.0 mg/kg) was administered subcutaneously three times a week starting either 24 hours prior to cell injection (prevention regimen) or at 4 weeks post-injection (treatment regimen). Changes in bone mineral density at the tumor site were determined by dual x-ray absorptiometry. Tumor growth was monitored by evaluating serum prostate specific antigen (PSA). Fc-OPG did not inhibit establishment of osteoblastic bone lesions of LuCaP 23.1, but it decreased growth of the tumor cells, as determined by decreases in serum PSA levels of 73.0 +/- 44.3% (P < 0.001) and 78.3 +/- 25.3% (P < 0.001) under the treatment and prevention regimens, respectively, compared to the untreated tumor-bearing animals. Administration of Fc-OPG decreased the proliferative index by 35.0% (P = 0.1838) in the treatment group, and 75.2% (P = 0.0358) in the prevention group. The results of this study suggest a potential role for OPG in the treatment of established osteoblastic CaP bone metastases.


Subject(s)
Bone Neoplasms/therapy , Glycoproteins/administration & dosage , Osteoblasts/metabolism , Prostatic Neoplasms/therapy , Receptors, Cytoplasmic and Nuclear/administration & dosage , Tibia/pathology , Animals , Bone Density , Bone Neoplasms/secondary , Injections, Subcutaneous , Lymphatic Metastasis/pathology , Male , Mice , Mice, Nude , Mice, SCID , Osteoprotegerin , Prostate-Specific Antigen/blood , Prostatic Neoplasms/pathology , Receptors, Tumor Necrosis Factor/administration & dosage , Tibia/metabolism , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
13.
Clin Exp Metastasis ; 20(2): 171-80, 2003.
Article in English | MEDLINE | ID: mdl-12705638

ABSTRACT

Bisphosphonates (BisP) are non-metabolized compounds with high bone affinity used in bone metastasis diagnosis and treatment. Currently, BisP are used to treat hypercalcemia of malignancy as well as to prevent, minimize, or delay skeletal morbidity. These compounds have a long half-life in bone. Thus long-term BisP treatment might saturate bone and interfere with a single-dose scanning agent used for bone scintigraphy when visualizing bone metastases. In an effort to answer this question, this study evaluated the concordance of histology and Technetium99 methylene diophosphonate (Tc99 MDP) bone scintigraphy in the diagnosis of bone metastases in prostate cancer patients. We assessed the concordance of findings between bone scintigraphy and histology using 188 bone biopsies from 11 autopsied patients who died with metastatic prostate cancer, 5 of whom were treated with pamidronate for 2 to 13 months before death. Overall agreement between histology and bone scintigraphy was 84%, 86% in non-pamidronate-treated patients and 82% in pamidronate-treated patients. Scintigraphic bone metastases without histological metastasis (false negatives = 12.7%) were observed in 24 anatomic locations; half of these were in one patient who had been treated with pamidronate and had no histological bone response to the carcinoma. There were only 4 sites where a positive bone scan was not associated with histologic metastasis (false positives = 2.21%). There was no statistical difference between the treated and non-treated group for concordance, specificity, sensitivity, positive and negative predictive values of bone scintigraphy and prevalence of histological abnormality. Long-term pamidronate treatment of prostate cancer bone metastases does not generally affect the ability to detect bone metastases with Tc99 MDP bone scintigraphy.


Subject(s)
Antineoplastic Agents/therapeutic use , Bone Neoplasms/diagnostic imaging , Bone Neoplasms/secondary , Diphosphonates/therapeutic use , Prostatic Neoplasms/diagnostic imaging , Prostatic Neoplasms/pathology , Autopsy , Bone Neoplasms/drug therapy , Humans , Infusions, Intravenous , Male , Pamidronate , Predictive Value of Tests , Prostate-Specific Antigen/blood , Prostatic Neoplasms/drug therapy , Radionuclide Imaging , Radiopharmaceuticals , Sensitivity and Specificity , Technetium Tc 99m Medronate
14.
J Bone Miner Res ; 18(2): 222-30, 2003 Feb.
Article in English | MEDLINE | ID: mdl-12568399

ABSTRACT

Prostate cancer (CaP) is the most commonly diagnosed malignancy in men and is often associated with bone metastases, which cause much of the morbidity associated with CaP. Lesions associated with CaP generally exhibit increased bone formation and resorption. Increased bone resorption may release factors from the extracellular matrix that contribute to tumor growth. Cathepsin K (cat K) is a cysteine protease that exhibits strong degradative activity against the extracellular matrix and is involved in osteoclast-mediated bone destruction. In this study, we analyzed the expression of cat K in CaP cell lines and patient samples. Cat K message was detected in CaP cell lines by reverse transcription-polymerase chain reaction (RT-PCR) and in primary CaP and metastases by in situ hybridization. Immunohistochemistry revealed variable expression of cat K in primary CaP samples, as well as nonosseous metastases, whereas expression in bone metastases was significantly higher than in primary CaP, and normal prostate tissues were negative. Cat K protein was detected in CaP cell lines by Western blotting after immunoprecipitation. Cat K enzymatic activity was also detected in CaP cell lines by a fluorogenic assay and by an assay for degradation of collagen type I. Increased levels of NTx, a marker of bone matrix degradation mediated primarily by cat K, were also detected in sera of patients with CaP bone metastases. We hypothesize that CaP-expressed cat K may contribute to the invasive potential of CaP, while increased expression in bone metastases is consistent with a role in matrix degradation.


Subject(s)
Cathepsins/metabolism , Prostatic Neoplasms/metabolism , RNA, Messenger/metabolism , Blotting, Western , Bone Resorption , Cathepsin K , Collagen/metabolism , Collagen Type I/metabolism , Cysteine Endopeptidases/metabolism , Disease Progression , Electrophoresis, Polyacrylamide Gel , Enzyme-Linked Immunosorbent Assay , Humans , Immunohistochemistry , In Situ Hybridization , Male , Neoplasm Metastasis , Precipitin Tests , Prostatic Neoplasms/pathology , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured
15.
Hybridoma ; 20(4): 231-6, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11604108

ABSTRACT

The monoclonal antibody (MAb) A6H, originally developed to fetal renal tissues, was found to be highly reactive to renal cell carcinoma and was subsequently demonstrated to co-stimulate a subpopulation of T cells. The A6H antigen had not been identified heretofore. Antigen from detergent extracts of renal cell carcinoma cells (7860) was immunoabsorbed with A6H-agarose, and the resin-bound proteins were separated by sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE). The antigen had a molecular weight of approximately 120 kDa as determined by Western blots. The 120-kDa protein band was excised and subjected to in-gel tryptic digestion, and the resulting peptides were separated and analyzed by liquid chromatography tandem mass spectrometry (LC MS\MS). The tandem mass spectra of the eluting peptides were used in combination with the SEQUEST computer program to search a human National Cancer Institute (NCI) protein database for the identity of the protein. The target antigen was shown to be dipeptidyl peptidase IV (DPP IV), which is also known as the cluster differentiation antigen CD26. Flow analysis of the expression of the A6H antigen and of CD26 on 7860 cells and on peripheral blood lymphocytes supported the identification of the A6H antigen as DPP IV. Recognition that the A6H antigen is DPP IV/CD26 afforded the opportunity to compare previous studies on A6H with those on other anti-CD26 antibodies in terms of expression in cancer cell lines and various tissues and as co-stimulators of T-cell activation.


Subject(s)
Antibodies, Monoclonal/immunology , Antigens, Neoplasm/immunology , Dipeptidyl Peptidase 4/immunology , Amino Acid Sequence , Antigens, Neoplasm/isolation & purification , Blotting, Western , Carcinoma, Renal Cell/enzymology , Cells, Cultured , Chromatography, Liquid/methods , Electrophoresis, Polyacrylamide Gel , Flow Cytometry , Humans , Kidney/enzymology , Kidney Neoplasms/enzymology , Lymphocyte Activation , Mass Spectrometry/methods , Molecular Sequence Data , T-Lymphocytes/immunology
16.
Clin Cancer Res ; 7(10): 2977-83, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11595685

ABSTRACT

PURPOSE: Osteoprotegerin (OPG) is a soluble osteoclastogenesis inhibitor that regulates bone turnover. We reported recently that OPG protein expression is significantly increased in prostate cancer (CaP) cells present in bone metastases. The aim of this study was to determine serum OPG levels in patients at different stages of CaP and correlate the results with disease status. EXPERIMENTAL DESIGN: OPG levels were examined in patients with benign prostatic hyperplasia, clinically localized CaP, early recurrence of CaP, and advanced CaP and evidence of bone metastases. Serum OPG levels were measured by sandwich ELISA assays. The serum Crosslaps (sCTX) assay was used to quantify bone resorption in the advanced CaP group. RESULTS: Serum OPG levels were increased significantly in the advanced CaP group versus all other groups. There was no significant correlation between serum OPG levels and PSA levels either in the advanced CaP group or within any of three treatment subclasses of this group: no Tx, those not treated; Tx, those treated; and R, those treated with resorption blockers. Levels of OPG were negatively correlated with sCTX levels only in the advanced CaP Tx group. sCTX levels correlated with prostate-specific antigen levels in the advanced CaP Tx and R groups but not in the no-Tx group. CONCLUSIONS: Our data show that serum OPG levels are increased with advanced CaP. We hypothesize that OPG levels are related to CaP progression and suggest that further studies of the biological effects of OPG on CaP metastases are warranted.


Subject(s)
Glycoproteins/blood , Prostatic Neoplasms/pathology , Receptors, Cytoplasmic and Nuclear/blood , Adult , Aged , Aged, 80 and over , Bone Neoplasms/blood , Bone Neoplasms/secondary , Collagen/blood , Enzyme-Linked Immunosorbent Assay , Humans , Male , Middle Aged , Neoplasm Recurrence, Local/blood , Neoplasm Recurrence, Local/pathology , Osteoprotegerin , Peptide Fragments/blood , Prostatic Hyperplasia/blood , Prostatic Hyperplasia/pathology , Prostatic Neoplasms/blood , Receptors, Tumor Necrosis Factor
17.
BJU Int ; 88(3): 221-5, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11488733

ABSTRACT

OBJECTIVE: To determine whether the percentage of free/total prostate-specific antigen (f/tPSA) can predict the pathological features in patients with clinically localized prostate cancer before radical prostatectomy. PATIENTS AND METHODS: Univariate and multivariate logistic regression was used to analyse data from 171 untreated patients who underwent radical prostatectomy. Variables included the total PSA (tPSA), fPSA, f/tPSA, biopsy Gleason score, clinical stage and patient age. RESULTS: In 115 patients with pathologically organ-confined tumours ( pT2N0) the mean (SD) tPSA value was 6.9 (5.6) ng/mL; in 56 patients with extracapsular disease ( pT3pN0/N+) it was 10.2 (7.6) ng/mL; the respective f/tPSA values were 14.9 (8.1)% and 14.2 (12.9)%. In the univariate and multivariate analysis, tPSA and biopsy Gleason score were highly significant in predicting extracapsular disease (P < 0.001 and 0.002) but the f/tPSA was not (P = 0.18). There was no significant difference between the mean f/tPSA and final Gleason scores. CONCLUSION: The f/tPSA does not predict extracapsular disease in patients with clinically localized prostate cancer before radical prostatectomy. Knowing the f/tPSA provides no significant additional information in predicting extracapsular disease when the biopsy Gleason score and tPSA are known.


Subject(s)
Prostate-Specific Antigen/metabolism , Prostatectomy/methods , Prostatic Neoplasms/diagnosis , Biopsy/methods , Humans , Lymph Node Excision , Male , Middle Aged , Multivariate Analysis , Neoplasm Staging/methods , Preoperative Care/methods , Prostatic Neoplasms/pathology , Retrospective Studies
18.
J Urol ; 166(1): 93-8; discussion 98-9, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11435831

ABSTRACT

PURPOSE: The free-to-total prostate specific antigen (PSA) ratio and complexed PSA have been introduced as adjuncts to total PSA for prostate cancer screening. Little data exist on the use of these tests for serial PSA screening. We compared serial total PSA, the free-to-total PSA ratio and calculated complexed PSA in men diagnosed with prostate cancer and matched controls in a population based study. MATERIALS AND METHODS: We identified 90 men diagnosed with prostate cancer between 1988 and 1996 with at least 3 serial serum samples obtained at 2-year intervals who were participants in the beta-Carotene and Retinol Efficacy Trial for the prevention of lung cancer. Samples were available up to 10 years before diagnosis. A total of 90 age matched men from the same cohort without prostate carcinoma were identified as controls. Free and total PSA was measured by the Abbott AxSYM system. RESULTS: Baseline demographics of cases and controls were similar. At baseline and diagnosis the men with prostate cancer had higher total and complexed PSA, and a lower free-to-total PSA ratio than controls. Mean followup was 5.2 years in cases and 5.5 in controls. The yearly change in PSA parameters in cases versus controls was 20.7% versus 3.5% for total, -3.4% versus 0.2% for free-to-total and 21.5% versus 3.4% for complexed PSA (p <0.0001). At diagnosis PSA alone was estimated to perform with more than 90% specificity in our model. CONCLUSIONS: In this population based study total PSA was superior to the free-to-total PSA ratio for predicting the development of prostate cancer. While serial changes in free-to-total PSA ratios with time were statistically significantly different in men diagnosed with prostate cancer and controls, the magnitude of these serial changes were slight enough to render them clinically insignificant.


Subject(s)
Adenocarcinoma/diagnosis , Biomarkers, Tumor/analysis , Mass Screening/methods , Prostate-Specific Antigen/blood , Prostatic Neoplasms/diagnosis , Adenocarcinoma/epidemiology , Adult , Age Distribution , Aged , Aged, 80 and over , Case-Control Studies , Cohort Studies , Confidence Intervals , Humans , Incidence , Logistic Models , Male , Middle Aged , Predictive Value of Tests , Prostatic Neoplasms/epidemiology , Reference Values , Risk Factors , Sensitivity and Specificity
19.
Int J Cancer ; 93(1): 47-52, 2001 Jul 01.
Article in English | MEDLINE | ID: mdl-11391620

ABSTRACT

The second most common target of prostate-cancer metastasis is bone, and the phenomenon of skeletal metastasis represents the incurable stage of disease. Histologically, skeletal metastasis from prostate cancer is distinctive due to its osteoblastic nature. The osteoblastic bone metastasis shows extensive new bone formation, with possible involvement of the soluble growth factors secreted by tumor cells, such as bone morphogenetic proteins (BMPs). In the present study, we analyzed the gene expression of one of the new members of the BMP family, placental bone morphogenetic protein (PLAB). In situ hybridization studies showed high levels of this gene in normal prostate. However, the gene is down-regulated during the progression of cancer at the primary site. The most significant finding was re-expression of the PLAB gene in osseous metastatic lesions. Our results demonstrate that tumor cells, when released from the primary site and after re-growth elsewhere, are capable of re-expressing specific genes that may play a different role at metastatic sites than at the primary site.


Subject(s)
Bone Morphogenetic Proteins/genetics , Precancerous Conditions/genetics , Prostate/metabolism , Prostatic Neoplasms/genetics , Bone Morphogenetic Proteins/analysis , Disease Progression , Humans , In Situ Hybridization , Male , Neoplasm Invasiveness , Neoplasm Metastasis , Precancerous Conditions/pathology , Prostate/cytology , Prostate/pathology , Prostatic Neoplasms/pathology , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction , Transcription, Genetic , Tumor Cells, Cultured
20.
Int J Cancer ; 92(6): 871-6, 2001 Jun 15.
Article in English | MEDLINE | ID: mdl-11351309

ABSTRACT

Recently we described the generation of the prostate tissue-specific monoclonal antibody (MAb) 107-1A4, its expression pattern and preliminary targeting of human prostate cancer xenografts. In this report we demonstrate that the target antigen for MAb 107-1A4 is prostate-specific membrane antigen (PSMA) using immunoaffinity absorption followed by SDS-PAGE and mass spectrometric analysis of peptides produced by in-gel tryptic digestion. The identity of the antigen has been confirmed by Western blots using MAbs of known specificity. MAb 107-1A4 is not reactive on Western blots. The conformational epitope for 107-1A4 is on the extracellular domain of PSMA. In competition studies, the binding of MAb 107-1A4 to LNCaP cells is inhibited by itself but not by any other of several other anti-PSMA MAbs, suggesting that the epitope may be unique. These results suggest that 107-1A4 is reactive to a conformational epitope in the external domain of PSMA that is unique among the panel of anti-PSMA MAbs in this study. Furthermore this work demonstrates the ability of mass spectroscopy to elucidate antibody-ligand interaction.


Subject(s)
Antibodies, Monoclonal/chemistry , Antigens, Surface , Carboxypeptidases/chemistry , Mass Spectrometry/methods , Oligonucleotide Array Sequence Analysis , Antibodies, Monoclonal/analysis , Binding, Competitive , Blotting, Western , Electrophoresis, Agar Gel , Electrophoresis, Polyacrylamide Gel , Epitopes , Glutamate Carboxypeptidase II , Humans , Ligands , Precipitin Tests , Protein Binding , Protein Conformation , Protein Structure, Tertiary , Trypsin/metabolism , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...