1
|
Tossetta G, Fantone S, Gesuita R, Montironi R, Marzioni D, Mazzucchelli R. AT-rich interactive domain 1A (ARID1A) cannot be considered a morphological marker for prostate cancer progression: A pilot study. Acta Histochem 2022; 124:151847. [PMID: 35038591 DOI: 10.1016/j.acthis.2022.151847] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 12/30/2021] [Accepted: 01/10/2022] [Indexed: 12/12/2022]
Abstract
Prostate cancer (PCa) is one of the most common cancers worldwide but it presents many subtypes and patient heterogeneity. It is necessary to discriminate localised not aggressive PCa and metastatic cancer in order to better define the personalised treatment. The identification of an appropriate biomarker to combine with Gleason grading system, that is one of the most important prognostic factors in prostate cancer outcome, remains a major clinical issue. We have tested AT-rich interactive domain 1A (ARID1A) in prostate tissue is order to verify its possible role as morphological marker for prostate cancer progression. ARID1A is a tumour suppressor protein playing a pivotal role in chromatin remodelling during transcriptional regulation. It was decreased in many cancers correlating with tumour aggressiveness. Our data shown that ARID1A had a nuclear staining and that it is significantly decreased in prostate cancers suggesting that it can be involved in this neoplasm but it is not able to discriminate prostate cancer progression.
Collapse
|
2
|
Prostate Cancer Genomics: Recent Advances and the Prevailing Underrepresentation from Racial and Ethnic Minorities. Int J Mol Sci 2018; 19:ijms19041255. [PMID: 29690565 PMCID: PMC5979433 DOI: 10.3390/ijms19041255] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Revised: 04/15/2018] [Accepted: 04/15/2018] [Indexed: 02/07/2023] Open
Abstract
Prostate cancer (CaP) is the most commonly diagnosed non-cutaneous cancer and the second leading cause of male cancer deaths in the United States. Among African American (AA) men, CaP is the most prevalent malignancy, with disproportionately higher incidence and mortality rates. Even after discounting the influence of socioeconomic factors, the effect of molecular and genetic factors on racial disparity of CaP is evident. Earlier studies on the molecular basis for CaP disparity have focused on the influence of heritable mutations and single-nucleotide polymorphisms (SNPs). Most CaP susceptibility alleles identified based on genome-wide association studies (GWAS) were common, low-penetrance variants. Germline CaP-associated mutations that are highly penetrant, such as those found in HOXB13 and BRCA2, are usually rare. More recently, genomic studies enabled by Next-Gen Sequencing (NGS) technologies have focused on the identification of somatic mutations that contribute to CaP tumorigenesis. These studies confirmed the high prevalence of ERG gene fusions and PTEN deletions among Caucasian Americans and identified novel somatic alterations in SPOP and FOXA1 genes in early stages of CaP. Individuals with African ancestry and other minorities are often underrepresented in these large-scale genomic studies, which are performed primarily using tumors from men of European ancestry. The insufficient number of specimens from AA men and other minority populations, together with the heterogeneity in the molecular etiology of CaP across populations, challenge the generalizability of findings from these projects. Efforts to close this gap by sequencing larger numbers of tumor specimens from more diverse populations, although still at an early stage, have discovered distinct genomic alterations. These research findings can have a direct impact on the diagnosis of CaP, the stratification of patients for treatment, and can help to address the disparity in incidence and mortality of CaP. This review examines the progress of understanding in CaP genetics and genomics and highlight the need to increase the representation from minority populations.
Collapse
|
3
|
Abstract
Although prostate cancer is the most common malignancy to affect men in the Western world, the molecular mechanisms underlying its development and progression remain poorly understood. Like all cancers, prostate cancer is a genetic disease that is characterized by multiple genomic alterations, including point mutations, microsatellite variations, and chromosomal alterations such as translocations, insertions, duplications, and deletions. In prostate cancer, but not other carcinomas, these chromosome alterations result in a high frequency of gene fusion events. The development and application of novel high-resolution technologies has significantly accelerated the detection of genomic alterations, revealing the complex nature and heterogeneity of the disease. The clinical heterogeneity of prostate cancer can be partly explained by this underlying genetic heterogeneity, which has been observed between patients from different geographical and ethnic populations, different individuals within these populations, different tumour foci within the same patient, and different cells within the same tumour focus. The highly heterogeneous nature of prostate cancer provides a real challenge for clinical disease management and a detailed understanding of the genetic alterations in all cells, including small subpopulations, would be highly advantageous.
Collapse
|
4
|
Diagnostic value of DNA alteration: loss of heterozygosity or allelic imbalance-promising for molecular staging of prostate cancers. Med Oncol 2013; 30:391. [PMID: 23288724 PMCID: PMC3586396 DOI: 10.1007/s12032-012-0391-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2012] [Accepted: 11/21/2012] [Indexed: 01/12/2023]
Abstract
The biological behavior of prostate cancer is uncertain, and therefore, search for molecular prognostic markers associated with disease progression seems to be essential. We performed microsatellite allelotyping of DNA isolated from primary prostate tumors biopsies (prostate adenocarcinoma, PCa). We evaluated the frequency of allelic imbalance (AI), including loss of heterozygosity and/or microsatellite imbalance (LOH/MSI) as well as the association of these DNA alterations with clinicopathological variables. We assessed the significance of LOH/MSI alterations in selected imprinted and non-imprinted chromosomal regions (IR and NIR) in PCa. A total of 50 biopsies of prostate tumor (containing >75 % tumor cells) were histologically examined confirming prostate carcinoma. Microsatellite allelotyping using 16 microsatellite markers linked to the following chromosomal regions: 1p31.2, 3p21.3–25.3, 7q32.2, 9p21.3, 11p15.5, 12q23.2, and 16q22.1 was performed. The incidence of LOH/MSI alterations in prostate tumor cells was the highest for chromosomal regions 7q32.2 and 16q22.1 (31.25 and 26.60 %, respectively), followed by 1p31.2 and 3p21.3–25.3 (26.50 and 17.40 %, respectively). Statistically significant increase in LOH/MSI alterations has been observed for markers: D1S2137 (1p region; p = 0.00032), D9S974 (9p region; p = 0.0017), and D16S3025 (16q region; p = 0.0017). Statistically significant differences in frequency of LOH/MSI alterations in particular chromosomal regions have been found for 1p31.2, 7q32.2 and 16q22.1 (p = 0.027, p = 0.012 and p = 0.031, respectively). We documented statistically significant association between Fractional Allele Loss (FAL) index and advanced tumor stage (p < 0.05). We suggest that genomic instability of LOH/MSI type is a frequent event in prostate carcinogenesis and assessed as FAL index has clinical value for the molecular staging of prostate cancer in (TRUS)-guided prostate biopsy material.
Collapse
|
5
|
White NMA, Youssef YM, Fendler A, Stephan C, Jung K, Yousef GM. The miRNA-kallikrein axis of interaction: a new dimension in the pathogenesis of prostate cancer. Biol Chem 2012; 393:379-89. [PMID: 22505520 DOI: 10.1515/hsz-2011-0246] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2011] [Accepted: 01/11/2012] [Indexed: 11/15/2022]
Abstract
Kallikrein-related peptidases (KLKs) are a family of serine proteases that were shown to be useful cancer biomarkers. KLKs have been shown to be dysregulated in prostate cancer (PCa). microRNAs (miRNAs) are short RNA nucleotides that negatively regulate gene expression and have been reportedly dysregulated in PCa. We compiled a comprehensive list of 55 miRNAs that are differentially expressed in PCa from previous microarray analysis and published literature. Target prediction analyses showed that 29 of these miRNAs are predicted to target 10 KLKs. Eight of these miRNAs were predicted to target more than one KLK. Quantitative real-time (qRT)-PCR demonstrated that there was an inverse correlation pattern in the expression (normal vs. cancer) between dysregulated miRNAs and their target KLKs. In addition, we experientially validated the miRNA-KLK interaction by transfecting miR-331-3p and miR-143 into a PCa cell line. Decreased expression of targets KLK4 and KLK10, respectively, and decreased cellular growth were observed. In addition to KLKs, dysregulated miRNAs were predicted to target other genes involved in the pathogenesis of PCa. These data show that miRNAs can contribute to KLK regulation in PCa. The miRNA-KLK axis of interaction projects a new element in the pathogenesis of PCa that may have therapeutic implications.
Collapse
Affiliation(s)
- Nicole M A White
- Department of Laboratory Medicine and the Keenan Research Centre in the Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, Ontario, Canada
| | | | | | | | | | | |
Collapse
|
6
|
Guyader C, Céraline J, Gravier E, Morin A, Michel S, Erdmann E, de Pinieux G, Cabon F, Bergerat JP, Poupon MF, Oudard S. Risk of hormone escape in a human prostate cancer model depends on therapy modalities and can be reduced by tyrosine kinase inhibitors. PLoS One 2012; 7:e42252. [PMID: 22879924 PMCID: PMC3412862 DOI: 10.1371/journal.pone.0042252] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2012] [Accepted: 07/05/2012] [Indexed: 11/29/2022] Open
Abstract
Almost all prostate cancers respond to androgen deprivation treatment but many recur. We postulated that risk of hormone escape -frequency and delay- are influenced by hormone therapy modalities. More, hormone therapies induce crucial biological changes involving androgen receptors; some might be targets for escape prevention. We investigated the relationship between the androgen deprivation treatment and the risk of recurrence using nude mice bearing the high grade, hormone-dependent human prostate cancer xenograft PAC120. Tumor-bearing mice were treated by Luteinizing-Hormone Releasing Hormone (LHRH) antagonist alone, continuous or intermittent regimen, or combined with androgen receptor (AR) antagonists (bicalutamide or flutamide). Tumor growth was monitored. Biological changes were studied as for genomic alterations, AR mutations and protein expression in a large series of recurrent tumors according to hormone therapy modalities. Therapies targeting Her-2 or AKT were tested in combination with castration. All statistical tests were two-sided. Tumor growth was inhibited by continuous administration of the LH-RH antagonist degarelix (castration), but 40% of tumors recurred. Intermittent castration or complete blockade induced by degarelix and antiandrogens combination, inhibited tumor growth but increased the risk of recurrence (RR) as compared to continuous castration (RRintermittent: 14.5, RRcomplete blockade: 6.5 and 1.35). All recurrent tumors displayed new quantitative genetic alterations and AR mutations, whatever the treatment modalities. AR amplification was found after complete blockade. Increased expression of Her-2/neu with frequent ERK/AKT activation was detected in all variants. Combination of castration with a Her-2/neu inhibitor decreased recurrence risk (0.17) and combination with an mTOR inhibitor prevented it. Anti-hormone treatments influence risk of recurrence although tumor growth inhibition was initially similar. Recurrent tumors displayed genetic instability, AR mutations, and alterations of phosphorylation pathways. We postulated that Her-2/AKT pathways allowed salvage of tumor cells under castration and we demonstrated that their inhibition prevented tumor recurrence in our model.
Collapse
Affiliation(s)
| | - Jocelyn Céraline
- Signaling and Prostate Cancer Group, Université de Strasbourg, Strasbourg, France
| | - Eléonore Gravier
- Translational Research Department, Institut Curie, Paris, France
- Biostatistics Department, Institut Curie, Paris, France
- U900, INSERM, Paris, France
- Ecole des Mines de Paris, ParisTech, Fontainebleau, France
| | | | - Sandrine Michel
- Biomarker Research and Validation Department, BioMérieux, Marcy l’Etoile, France
| | - Eva Erdmann
- Signaling and Prostate Cancer Group, Université de Strasbourg, Strasbourg, France
| | | | | | - Jean-Pierre Bergerat
- Signaling and Prostate Cancer Group, Université de Strasbourg, Strasbourg, France
| | | | - Stéphane Oudard
- Medical Oncology, Hôpital Européen Georges Pompidou, Paris, France
- Université Paris V René Descartes, Paris, France
- * E-mail:
| |
Collapse
|
7
|
Changes of transthyretin and clusterin after androgen ablation therapy and correlation with prostate cancer malignancy. Transl Oncol 2012; 5:124-32. [PMID: 22496929 DOI: 10.1593/tlo.11259] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2011] [Revised: 12/30/2011] [Accepted: 01/03/2012] [Indexed: 01/07/2023] Open
Abstract
After androgen ablation therapy (AAT), advanced prostate cancer (Pca) eventually progresses to castration-resistant Pca (CRPC); however, the biomarkers that are used to predict its prognosis are limited. In this study, serum samples from four patients with advanced Pca were collected at the time of the initial diagnosis and 3 months after AAT. Proteomic changes were analyzed with two-dimensional differential in-gel electrophoresis and matrix-assisted laser desorption/ionization time-of-flight mass spectrometry. Altogether, nine proteins were differentially expressed in the samples collected at diagnosis and in the samples collected after AAT. Among them, the expression of transthyretin (TTR) was 1.58-fold lower and clusterin (CLU) was 1.51-fold higher in the sera of post-AAT patients compared with those in the sera from pre-AAT patients. The significant changes in serum TTR and CLU in post-AAT patients were further confirmed by a large-scale ELISA. Immunohistochemistical staining revealed that the expression levels of TTR and CLU were significantly higher in Pca tissue than in normal and benign prostate hyperplasia tissue. The expression levels of TTR and CLU in Pca tissue were found to be associated with the grade and stage of Pca. Overall, this study indicated that TTR and CLU might be used to monitor the efficacy of AAT therapy and serve as biomarkers for the prognosis of Pca.
Collapse
|
8
|
Squire JA, Park PC, Yoshimoto M, Alami J, Williams JL, Evans A, Joshua AM. Prostate cancer as a model system for genetic diversity in tumors. Adv Cancer Res 2012; 112:183-216. [PMID: 21925305 DOI: 10.1016/b978-0-12-387688-1.00007-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
This chapter will summarize novel understandings of the early molecular events in prostatic carcinogenesis that may underlie both the genetic and clinical heterogeneity. Areas covered include preneoplasia, stem cell concepts, telomere abnormalities, and the nature of tumor-stromal interactions. The oncogenomics of prostate cancer is reviewed with emphasis on androgen signaling, ETS gene family aberrations, and PTEN deletion. The notion that "field cancerization," coupled with genomic instability may explain both the occurrence of multifocal disease, and the recent observations of genetic diversity of ERG alteration in individual tumors are discussed. Collectively, genomic studies are rapidly moving human prostate cancer closer to the promise of personalized medicine, so that specific genetic profiles of individual tumors will determine the best therapeutic approaches.
Collapse
Affiliation(s)
- Jeremy A Squire
- Department of Pathology and Molecular Medicine, Queen's University, Kingston, ON, Canada
| | | | | | | | | | | | | |
Collapse
|
9
|
Bartuma H, Nord KH, Macchia G, Isaksson M, Nilsson J, Domanski HA, Mandahl N, Mertens F. Gene expression and single nucleotide polymorphism array analyses of spindle cell lipomas and conventional lipomas with 13q14 deletion. Genes Chromosomes Cancer 2011; 50:619-32. [DOI: 10.1002/gcc.20884] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2011] [Revised: 03/24/2011] [Accepted: 03/24/2011] [Indexed: 01/07/2023] Open
|
10
|
Pittman AM, Naranjo S, Jalava SE, Twiss P, Ma Y, Olver B, Lloyd A, Vijayakrishnan J, Qureshi M, Broderick P, van Wezel T, Morreau H, Tuupanen S, Aaltonen LA, Alonso ME, Manzanares M, Gavilán A, Visakorpi T, Gómez-Skarmeta JL, Houlston RS. Allelic variation at the 8q23.3 colorectal cancer risk locus functions as a cis-acting regulator of EIF3H. PLoS Genet 2010; 6:e1001126. [PMID: 20862326 PMCID: PMC2940760 DOI: 10.1371/journal.pgen.1001126] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2010] [Accepted: 08/13/2010] [Indexed: 11/27/2022] Open
Abstract
Common genetic variation at human 8q23.3 is significantly associated with colorectal cancer (CRC) risk. To elucidate the basis of this association we compared the frequency of common variants at 8q23.3 in 1,964 CRC cases and 2,081 healthy controls. Reporter gene studies showed that the single nucleotide polymorphism rs16888589 acts as an allele-specific transcriptional repressor. Chromosome conformation capture (3C) analysis demonstrated that the genomic region harboring rs16888589 interacts with the promoter of gene for eukaryotic translation initiation factor 3, subunit H (EIF3H). We show that increased expression of EIF3H gene increases CRC growth and invasiveness thereby providing a biological mechanism for the 8q23.3 association. These data provide evidence for a functional basis for the non-coding risk variant rs16888589 at 8q23.3 and provides novel insight into the etiological basis of CRC. Common inherited variation on human chromosome 8q23 influences the risk of developing colorectal cancer (CRC). To understand the basis of this association we have compared the frequency of common genetic variants at 8q23 in ∼2,000 CRC cases and ∼2,000 healthy controls. Functional analyses of variants strongly associated with CRC risk showed that the single nucleotide polymorphism rs16888589 underscores the 8q23.3 association. The region of the genome harboring rs16888589 increases the expression of the gene for eukaryotic translation initiation factor 3, subunit H. We show that increased expression of this gene increases CRC growth thereby providing a biological mechanism for the 8q23.3 association. This finding is of particular importance in elucidating the etiological basis of CRC.
Collapse
Affiliation(s)
- Alan M. Pittman
- Section of Cancer Genetics, Institute of Cancer Research, Sutton, United Kingdom
| | - Silvia Naranjo
- Centro Andaluz de Biología del Desarrollo, CSIC-UPO, Seville, Spain
| | - Sanni E. Jalava
- Institute of Medical Technology, University of Tampere and Tampere University Hospital, Tampere, Finland
| | - Philip Twiss
- Section of Cancer Genetics, Institute of Cancer Research, Sutton, United Kingdom
| | - Yussanne Ma
- Section of Cancer Genetics, Institute of Cancer Research, Sutton, United Kingdom
| | - Bianca Olver
- Section of Cancer Genetics, Institute of Cancer Research, Sutton, United Kingdom
| | - Amy Lloyd
- Section of Cancer Genetics, Institute of Cancer Research, Sutton, United Kingdom
| | | | - Mobshra Qureshi
- Section of Cancer Genetics, Institute of Cancer Research, Sutton, United Kingdom
| | - Peter Broderick
- Section of Cancer Genetics, Institute of Cancer Research, Sutton, United Kingdom
| | - Tom van Wezel
- Department of Pathology, Leiden University Medical Center, Leiden, The Netherlands
| | - Hans Morreau
- Department of Pathology, Leiden University Medical Center, Leiden, The Netherlands
| | - Sari Tuupanen
- Department of Medical Genetics, University of Helsinki, Helsinki, Finland
| | - Lauri A. Aaltonen
- Department of Medical Genetics, University of Helsinki, Helsinki, Finland
| | - M. Eva Alonso
- Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain
| | | | - Angela Gavilán
- Centro Nacional de Investigaciones Biomédica en Red Enfermedades Raras (CIBERER), Universidad Pablo de Olavide-CSIC, Seville, Spain
| | - Tapio Visakorpi
- Institute of Medical Technology, University of Tampere and Tampere University Hospital, Tampere, Finland
| | | | - Richard S. Houlston
- Section of Cancer Genetics, Institute of Cancer Research, Sutton, United Kingdom
- * E-mail:
| |
Collapse
|
11
|
Humbert L, Chevrette M. Somatic Molecular Genetics of Prostate Cancer. MALE REPRODUCTIVE CANCERS 2010:143-180. [DOI: 10.1007/978-1-4419-0449-2_5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/02/2023]
|
12
|
Reynolds MA. Molecular alterations in prostate cancer. Cancer Lett 2008; 271:13-24. [PMID: 18554779 DOI: 10.1016/j.canlet.2008.04.047] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2007] [Revised: 12/21/2007] [Accepted: 04/24/2008] [Indexed: 01/07/2023]
Abstract
Prostate tumors display a range of clinical phenotypes, from indolent to aggressively metastatic. Numerous gene expression profiling studies have been conducted toward the potential molecular staging of these pathologies, however the identification of genetic markers that predict aggressive disease has not yet been demonstrated in the clinical setting. A recent survey of the literature has shown that molecular alterations in prostate carcinomas can occur through a variety of different mechanisms, ranging from upstream epigenetic changes and genetic polymorphisms to downstream modulations through alternative splicing and other post-translational processes, some of which could involve noncoding RNAs. A summary of these results and recommendations for future work are the subject of this review.
Collapse
|
13
|
Prostate Molecular Oncogenesis. Prostate Cancer 2008. [DOI: 10.1007/978-1-60327-079-3_4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
|
14
|
Leonetti C, Biroccio A, D'Angelo C, Semple SC, Scarsella M, Zupi G. Therapeutic integration of c-myc and bcl-2 antisense molecules with docetaxel in a preclinical model of hormone-refractory prostate cancer. Prostate 2007; 67:1475-85. [PMID: 17654511 DOI: 10.1002/pros.20636] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
BACKGROUND The response of hormone-refractory prostate cancer (HRPC) to chemotherapy remains modest, necessitating the search for new forms of treatment to improve the prognosis. Since an increased expression of oncogenes, including c-myc and bcl-2, accompanies the transition to HRPC, we evaluated whether the concomitant downregulation of these oncogenes by antisense strategy sensitized HRPC to chemotherapy. METHODS PC-3 prostate cancer cells were exposed in vitro to c-myc (INX-6295) and bcl-2 (G3139) antisense oligodeoxynucleotides (ODNs) and docetaxel given alone or in combination. Therapeutic efficacy of the different treatments was also evaluated in xenografts. RESULTS We show that the triple combination of drugs given in the sequence G3139/docetaxel/INX-6295 was the most active in reducing the survival of PC-3. Likewise, the combination triggered apoptosis in more than 80% of cells. A marked tumor weight inhibition was observed in PC-3 xenografts after G3139/docetaxel/INX-6295 treatment, with a complete tumor regression being noted in half the mice. A 111% overall increase in life survival and a complete cure in two out of eight mice was also reported. This treatment remained effective even when started at a very late stage of tumor growth producing about 80% tumor weight inhibition (TWI), with tumor regression being maintained for 1 month. Finally, the antitumor effect resulted in a significant increase (70%) in mice survival. CONCLUSIONS These data indicate that the combined targeting of genes involved in uncontrolled proliferation and evasion of apoptosis renders HRPC responsive to chemotherapy making this treatment a promising antineoplastic strategy.
Collapse
Affiliation(s)
- Carlo Leonetti
- Experimental Chemotherapy Laboratory, Regina Elena Cancer Institute, Rome, Italy
| | | | | | | | | | | |
Collapse
|
15
|
Schwarzenbach H, Chun FKH, Lange I, Carpenter S, Gottberg M, Erbersdobler A, Friedrich MG, Huland H, Pantel K. Detection of tumor-specific DNA in blood and bone marrow plasma from patients with prostate cancer. Int J Cancer 2007; 120:1465-71. [PMID: 17205532 DOI: 10.1002/ijc.22470] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Tumor tissues, blood plasma and bone marrow (BM) aspirates of 57 prostate cancer patients (PCa) without clinical signs of overt metastases were assessed for LOH (loss of heterozygosity) by a PCR-based fluorescence microsatellite analysis, using a panel of 15 markers. Additionally, micrometastatic tumor cells in BM were monitored by an immunocytological cytokeratin assay. In total, 25 (44%), 32 (56%) and 41 (72%) of the patients had at least 1 LOH in their blood, BM and tumor samples, respectively. Among the informative cases, the frequency of LOH was highest in blood plasma for the markers D8S360 (18%) and D10S1765 (15%), and in BM plasma for THRB (24%) and D8S137 (22%). Comparison of blood plasma and BM with tumors showed discrepant results in 35% and 45% of patients, respectively. Whereas all LOHs at THRB in BM plasma were also detected in the autologous tumor tissues, LOHs at D6S474 and D11S898 in BM were not retrieved in the tumors. The comparison with established risk factors showed a correlation of borderline significance for LOH at D9S1748 in the BM aspirates (p=0.055) and a significant correlation in the tumor samples (p=0.004) with increasing pathologic Gleason scores. Interestingly, 22% of the PCa patients harbored tumor cells in their BM and tended (p=0.065) to have more frequent LOH (16%) in BM plasma compared to patients without tumor cells (9%). These data demonstrate, for the first time, the presence of free tumor-specific DNA in blood and BM of PCa patients and suggest a possible relationship to BM micrometastasis.
Collapse
Affiliation(s)
- Heidi Schwarzenbach
- Institute of Tumor Biology, University Medical Center Hamburg-Eppendorf, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Jemal A, Siegel R, Ward E, Murray T, Xu J, Thun MJ. Cancer statistics, 2007. CA Cancer J Clin 2007; 1785:156-81. [PMID: 17237035 DOI: 10.1016/j.bbcan.2007.12.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2007] [Revised: 12/02/2007] [Accepted: 12/03/2007] [Indexed: 02/06/2023] Open
Abstract
Each year, the American Cancer Society (ACS) estimates the number of new cancer cases and deaths expected in the United States in the current year and compiles the most recent data on cancer incidence, mortality, and survival based on incidence data from the National Cancer Institute, Centers for Disease Control and Prevention, and the North American Association of Central Cancer Registries and mortality data from the National Center for Health Statistics. This report considers incidence data through 2003 and mortality data through 2004. Incidence and death rates are age-standardized to the 2000 US standard million population. A total of 1,444,920 new cancer cases and 559,650 deaths for cancers are projected to occur in the United States in 2007. Notable trends in cancer incidence and mortality rates include stabilization of the age-standardized, delay-adjusted incidence rates for all cancers combined in men from 1995 through 2003; a continuing increase in the incidence rate by 0.3% per year in women; and a 13.6% total decrease in age-standardized cancer death rates among men and women combined between 1991 and 2004. This report also examines cancer incidence, mortality, and survival by site, sex, race/ethnicity, geographic area, and calendar year, as well as the proportionate contribution of selected sites to the overall trends. While the absolute number of cancer deaths decreased for the second consecutive year in the United States (by more than 3,000 from 2003 to 2004) and much progress has been made in reducing mortality rates and improving survival, cancer still accounts for more deaths than heart disease in persons under age 85 years. Further progress can be accelerated by supporting new discoveries and by applying existing cancer control knowledge across all segments of the population.
Collapse
Affiliation(s)
- Ahmedin Jemal
- Cancer Occurrence, Department of Epidemiology and Surveillance Research, American Cancer Society, Atlanta, GA, USA
| | | | | | | | | | | |
Collapse
|
17
|
Sun M, Srikantan V, Ma L, Li J, Zhang W, Petrovics G, Makarem M, Strovel JW, Horrigan SG, Augustus M, Sesterhenn IA, Moul JW, Chandrasekharappa S, Zou Z, Srivastava S. Characterization of frequently deleted 6q locus in prostate cancer. DNA Cell Biol 2006; 25:597-607. [PMID: 17132090 DOI: 10.1089/dna.2006.25.597] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
The long arm of chromosome 6 is frequently deleted in diverse human neoplasms. Our previous study showed a minimum deletion region between markers D6S1056 and D6S300 on chromosome 6q in primary prostate cancer (CaP). In this study, we further refined a 200-kb minimal region of deletion (6qTSG1) centered around D6S1013 marker. The 6qTSG1 transcripts contained complex multiple splicing variants with low or absent expression in CaP cells. None of the transcripts identified contained open reading frames that code for a protein in the NCBI database. The expression of 6qTSG transcripts revealed interesting hormonal regulation relevant to CaP biology. Expression of 6q TSG transcript was induced in LNCaP cells that were cultured in charcoal-stripped serum medium suggesting an upregulation of 6qTSG transcript by androgen ablation and cell growth inhibition/apoptosis. Induction of 6qTSG1 expression in response to androgen ablation was abrogated in androgen-independent derivatives of LNCaP cells. In summary, we have defined a candidate CaP suppressor locus on chromosome 6q16.1, and deletions of this locus are frequently associated with prostate tumorigenesis. In the light of emerging role of noncoding RNAs in cancer biology including CaP, future investigations of 6qTSG11 locus is warranted.
Collapse
Affiliation(s)
- Mei Sun
- Center for Prostate Disease Research, Department of Surgery, Uniformed Services University of the Health Sciences, Rockville, Maryland 20832, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Abstract
PURPOSE OF REVIEW The genetic and molecular basis of prostate-cancer pathogenesis is reviewed. RECENT FINDINGS Several genetic loci have been found that are associated with hereditary predisposition to prostate cancer, but they account for a small fraction of all cases. A number of suppressor genes have been identified that are activated by either complete or partial genetic loss in sporadic prostate cancer. Chromosomal translocation results in transcriptional activation of truncated ETS transcription factors ERG and ETV1, the first candidates for dominant oncogenes for prostate cancer. Lastly, the androgen receptor is active throughout the course of prostate cancer and, in androgen-independent prostate cancer, takes on the role of a dominant oncogene as the target of gene amplification, overexpression, and the activation of mutations. SUMMARY Genetic lesions responsible for familial and sporadic prostate cancer are being revealed and they suggest that prostate cancer often initiates owing to an increased susceptibility to oxidative damage; it then progresses by affecting transcription factors, the PI3 kinase pathway, and other growth stimulatory pathways. The final common pathway after androgen ablation appears to be activation of androgen receptor.
Collapse
Affiliation(s)
- Randi L Shand
- Departments of Oncology and Medicine, Lombardi Comprehensive Cancer Center, Georgetown University, 3800 Reservoir Road NW, Washington, DC 20007, USA
| | | |
Collapse
|
19
|
Ribeiro FR, Henrique R, Martins AT, Jerónimo C, Teixeira MR. Relative copy number gain of MYC in diagnostic needle biopsies is an independent prognostic factor for prostate cancer patients. Eur Urol 2006; 52:116-25. [PMID: 17070983 DOI: 10.1016/j.eururo.2006.09.018] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2006] [Accepted: 09/25/2006] [Indexed: 12/25/2022]
Abstract
OBJECTIVES We have recently shown using comparative genomic hybridization (CGH) that 8q gain is an independent predictor of poor survival for prostate cancer patients. Because CGH may be difficult to implement in the clinical practice, we tested the feasibility of using a three-color fluorescent assay to assess 8q status in diagnostic, paraffin-embedded biopsy samples from prostate cancer patients. METHODS Fluorescence in situ hybridization with a dual-color probe flanking the MYC gene at 8q24 and a control probe for chromosome 18 was performed in a retrospective series of paraffin-embedded biopsies from 60 prostate cancer patients. The prognostic significance of 8q status was assessed by calculating disease-specific survival curves for these patients. RESULTS Whereas 44 (73%) samples displayed copy number gains of the MYC gene, a MYC/CEP18 ratio > or = 1.5 was detected in 36 (60%) samples. Kaplan-Meier curves with log-rank test showed that patients whose tumors displayed MYC/CEP18 ratio > or = 1.5 had a significantly worse disease-specific survival (p=0.003). The dual-color labelling of the MYC probe further allowed us to detect structural rearrangements of this gene in six (10%) carcinomas. CONCLUSIONS We show that a standard fluorescent protocol can successfully be applied to diagnostic needle biopsies to identify relative 8q gain in prostate carcinomas and that patients with a MYC/CEP18 ratio > or = 1.5 present a significantly higher risk of dying from the disease. The prognostic significance of this genetic variable was seen even for patients with Gleason score 7 or clinical stage II/III carcinomas, whose clinical behavior is currently difficult to predict.
Collapse
|
20
|
Müller I, Urban K, Pantel K, Schwarzenbach H. Comparison of Genetic Alterations Detected in Circulating Microsatellite DNA in Blood Plasma Samples of Patients with Prostate Cancer and Benign Prostatic Hyperplasia. Ann N Y Acad Sci 2006; 1075:222-9. [PMID: 17108215 DOI: 10.1196/annals.1368.030] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Prostate cancer is the most frequent malignant disease and the second most frequent cause of death due to cancer in men in the Western world. Since serum prostate-specific antigen (PSA) and its subforms show poor specificity in clinical practice, a molecular marker for the detection and discrimination of prostate cancer (PCa) could be of great interest. To investigate the potential significance of genetic aberrations, such as loss of heterozygosity (LOH), in PCa we identified and characterized allelic losses in circulating tumor-associated DNA in blood from patients with localized PCa. Genomic DNA extracted from cell-free plasma of blood samples drawn from 65 PCa patients was analyzed using a panel of 15 polymorphic microsatellite markers mapping to known tumor-suppressor genes. Comparative analyses were performed with a control group of 36 patients with benign prostatic hyperplasia (BPH). In the current study, we demonstrate that PCa patients had higher DNA concentrations in their blood circulation than BPH patients. In the marker panel studied, LOH was more frequently detected in PCa patients (34%) than in BPH patients (22%). The incidence of LOH in the plasma DNA of PCa patients was highest at chromosomal regions 3p24 (THRB, 22%) and 8p21 (D8S360, 22%) in comparison to the BPH control cohort, which frequently showed LOH at loci 8q21, 8p21, 9p21, and 11q22 (D8S286, D8S360, D9S1748, and D11S898, each 6%). These results indicate that microsatellite analysis using plasma DNA may be an interesting tool for molecular screening of PCa patients.
Collapse
Affiliation(s)
- Imke Müller
- Institute of Tumor Biology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany
| | | | | | | |
Collapse
|
21
|
Johansson B, Pourian MR, Chuan YC, Byman I, Bergh A, Pang ST, Norstedt G, Bergman T, Pousette A. Proteomic comparison of prostate cancer cell lines LNCaP-FGC and LNCaP-r reveals heatshock protein 60 as a marker for prostate malignancy. Prostate 2006; 66:1235-44. [PMID: 16705742 PMCID: PMC7168115 DOI: 10.1002/pros.20453] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2005] [Accepted: 03/16/2006] [Indexed: 11/11/2022]
Abstract
BACKGROUND Androgen-sensitive prostate cancer cell-line LNCaP-FGC and androgen-resistant line LNCaP-r constitute a model for development of androgen resistance in prostate cancer. METHODS Proteins differently expressed in the two cell-lines were identified by two-dimensional (2-D) electrophoresis and mass spectrometry. HSP60, more abundant in LNCaP-r, was studied by RT-PCR and immunohistochemistry in specimens of human prostate cancer. RESULTS HSP60 was upregulated in LNCaP-r, nm23 in LNCaP-FGC, and titin (two isoforms) in either LNCaP-r or LNCaP-FGC. In non-malignant prostate, HSP60-staining was in the glandular compartment, particularly basal epithelial cells. In prostate cancer, most epithelial cells showed moderate-strong staining without apparent correlation between staining intensity and Gleason grade. CONCLUSIONS The LNCaP-FGC/LNCaP-r model, characterized by 2-D electrophoresis, reveals distinct proteomic alterations. With HSP60, results from cell-lines correlated with clinical results, indicating that this model can be used for dissection of mechanisms involved in transformation to androgen resistance and assignment of protein markers in prostate cancer.
Collapse
MESH Headings
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/metabolism
- Blotting, Western
- Cell Line, Tumor
- Cell Transformation, Neoplastic/genetics
- Cell Transformation, Neoplastic/metabolism
- Cell Transformation, Neoplastic/pathology
- Chaperonin 60/genetics
- Chaperonin 60/metabolism
- Electrophoresis, Gel, Two-Dimensional
- Epithelial Cells/metabolism
- Epithelial Cells/pathology
- Gene Expression Regulation, Neoplastic
- Humans
- Immunohistochemistry
- Male
- Mass Spectrometry
- Prognosis
- Prostatic Neoplasms/diagnosis
- Prostatic Neoplasms/genetics
- Prostatic Neoplasms/metabolism
- Prostatic Neoplasms/pathology
- Proteomics
- Reverse Transcriptase Polymerase Chain Reaction
Collapse
Affiliation(s)
- Björn Johansson
- Department of Clinical Neuroscience, Karolinska Hospital, Karolinska Institutet, Stockholm, Sweden.
| | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Chaudhary J, Schmidt M. The impact of genomic alterations on the transcriptome: a prostate cancer cell line case study. Chromosome Res 2006; 14:567-86. [PMID: 16823619 DOI: 10.1007/s10577-006-1055-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2006] [Revised: 03/24/2006] [Accepted: 03/24/2006] [Indexed: 12/13/2022]
Abstract
Genetic instability may lead to the loss/gain of transcriptional control. Here we investigated the effect of genomic instability, that is loss/gain of chromosomal regions on the global transcriptome of prostate cancer cell line DU145. The genomic loss/gain map obtained through BAC array-based CGH was superimposed on the dynamic transcriptome of DU145 cells treated with serum for 0 h (serum starved), 2 h and 12 h. The genomic analysis suggested that in DU145 cells: (1) chromosomal gains are prominent than losses and (2) copy number changes are associated with chromosome-specific and dynamic gene expression regulatory mechanisms. A significant proportion of the genes in the stable regions of the chromosome were up-regulated whereas a higher proportion of genes were down-regulated at 2 and 12 h in the deleted regions of the chromosomes following serum treatment. No change in expression was observed for the genes in the gained regions over a period of time. This analysis led us to propose that loss of heterozygosity leads to an overall transcriptional down-regulation that may further lead to a decrease in the expression of putative tumor suppressors. The genomic profile of DU145 is similar to pathological specimens of prostate cancer, hence the genomic/transcriptomic signature of DU145 can be used to understand the pathology of prostate cancer. It is expected that this analysis will allow a better understanding of transcriptional regulatory mechanisms in the context of genomic loss and gain and may lead to the discovery of novel oncogenes and tumor suppressors and the underlying regulatory pathways.
Collapse
MESH Headings
- Cell Line, Tumor
- Chromosomal Instability/genetics
- Chromosomes, Human, Pair 1
- Chromosomes, Human, Pair 14
- Chromosomes, Human, Pair 18
- Chromosomes, Human, Pair 5
- Chromosomes, Human, Pair 8
- Gene Expression Regulation
- Genomic Instability/genetics
- Humans
- Loss of Heterozygosity
- Male
- Oligonucleotide Array Sequence Analysis
- Prostatic Neoplasms/genetics
- Transcription, Genetic/genetics
Collapse
Affiliation(s)
- J Chaudhary
- 4029D RCST, Department of Biological Sciences, Center for Cancer Research and Therapeutics Development, Clark Atlanta University, Atlanta, GA 30314, USA.
| | | |
Collapse
|
23
|
Buhmeida A, Pyrhönen S, Laato M, Collan Y. Prognostic factors in prostate cancer. Diagn Pathol 2006; 1:4. [PMID: 16759347 PMCID: PMC1479371 DOI: 10.1186/1746-1596-1-4] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2006] [Accepted: 04/03/2006] [Indexed: 02/05/2023] Open
Abstract
Prognostic factors in organ confined prostate cancer will reflect survival after surgical radical prostatectomy. Gleason score, tumour volume, surgical margins and Ki-67 index have the most significant prognosticators. Also the origins from the transitional zone, p53 status in cancer tissue, stage, and aneuploidy have shown prognostic significance. Progression-associated features include Gleason score, stage, and capsular invasion, but PSA is also highly significant. Progression can also be predicted with biological markers (E-cadherin, microvessel density, and aneuploidy) with high level of significance. Other prognostic features of clinical or PSA-associated progression include age, IGF-1, p27, and Ki-67. In patients who were treated with radiotherapy the survival was potentially predictable with age, race and p53, but available research on other markers is limited. The most significant published survival-associated prognosticators of prostate cancer with extension outside prostate are microvessel density and total blood PSA. However, survival can potentially be predicted by other markers like androgen receptor, and Ki-67-positive cell fraction. In advanced prostate cancer nuclear morphometry and Gleason score are the most highly significant progression-associated prognosticators. In conclusion, Gleason score, capsular invasion, blood PSA, stage, and aneuploidy are the best markers of progression in organ confined disease. Other biological markers are less important. In advanced disease Gleason score and nuclear morphometry can be used as predictors of progression. Compound prognostic factors based on combinations of single prognosticators, or on gene expression profiles (tested by DNA arrays) are promising, but clinically relevant data is still lacking.
Collapse
Affiliation(s)
- A Buhmeida
- Departments of Oncology and Radiotherapy, Turku University Hospital, Turku, Finland
| | - S Pyrhönen
- Departments of Oncology and Radiotherapy, Turku University Hospital, Turku, Finland
| | - M Laato
- Departments of Surgery, Turku University Hospital, Turku, Finland
| | - Y Collan
- Departments of Pathology, Turku University Hospital, Turku, Finland
| |
Collapse
|
24
|
Chieffi P, Cozzolino L, Kisslinger A, Libertini S, Staibano S, Mansueto G, De Rosa G, Villacci A, Vitale M, Linardopoulos S, Portella G, Tramontano D. Aurora B expression directly correlates with prostate cancer malignancy and influence prostate cell proliferation. Prostate 2006; 66:326-33. [PMID: 16267859 DOI: 10.1002/pros.20345] [Citation(s) in RCA: 111] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
BACKGROUND Chromosomal instability is one of the most common features of prostate cancer (PC), especially in advanced stages. Recent studies suggest that defects in mitotic checkpoints play a role in carcinogenesis. Lack of mitotic regulation induces aneuploidy in cancer cells acting thereafter as a driving force for malignant progression. Serine/threonine protein kinases of the Aurora genes family play an important throughout the entire cell cycle. In that Aurora B regulates chromosome segregation by ensuring the orientation of sister chromatids. As a consequence, the overexpression of Aurora B in diploid human cells NHDF induces the appearance of multinucleate cells. METHODS Archive samples of normal and neoplastic prostate tissue, and prostate derived cell lines were screened for the expression of Aurora B. RESULTS Immunohistochemical analysis showed increased nuclear expression of Aurora-B in high Gleason grade PCs respect to low and intermediate grade cases and in all cancers in respect to hyperplastic and normal glands. Furthermore, in the high Gleason grade anaplastic cancer tissues Aurora B expression was accompanied by the phosphorylation of the histone H3. In analogy to the in vivo situation, Aurora B was vigorously expressed in the androgen independent PC cell lines PC3 and DU145, while a very modest expression of the kinase was observed in the androgen sensitive LnCap cells and in the EPN cells, a line of epithelial cells derived from normal prostate tissue. In addition, in PC3 cells Aurora B expression is accompanied the by the phosphorylation of the histone H3. The block of Aurora B expression induced by an inhibitor of Aurora kinase activity significantly reduced the growth of prostate carcinoma cells, but not that of non-transformed EPN cells. CONCLUSIONS Our data are the first demonstration of a role of Aurora B in PC progression. In addition, the observation that Aurora B specific inhibitors interfere with PC cell proliferation but not with that of non-transformed prostate epithelial cells suggest that Aurora B is a potential therapeutic target for PC.
Collapse
Affiliation(s)
- Paolo Chieffi
- Dipartimento di Medicina Sperimentale, II Università di Napoli, Naples, Italy.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Pang ST, Weng WH, Flores-Morales A, Johansson B, Pourian MR, Nilsson P, Pousette A, Larsson C, Norstedt G. Cytogenetic and expression profiles associated with transformation to androgen-resistant prostate cancer. Prostate 2006; 66:157-72. [PMID: 16173030 DOI: 10.1002/pros.20328] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
BACKGROUND The mechanisms underlying the progression of prostate cancer to androgen-resistant cancer are still not fully understood. Here, we studied the genetic events associated with this transformation. METHODS The androgen sensitive prostate cancer cells line LNCaP-FGC and its androgen resistant subline LNCaP-r were investigated using SKY, CGH, and cDNA microarray. RESULTS Karyotypically, several additional chromosomal aberrations were seen in LNCaP-r as compared to the parental line. CGH also revealed unique net chromosomal alterations in LNCaP-r compared to LNCaP-FGC, including gain of 2p13-23, 2q21-32, and 13q and loss of 6p22-pter. cDNA microarray analysis identified several genes involved in DNA methylation, such as DNMT2, DNMT3a, and methyl-CpG binding domain protein 2 and 4 that were higher expressed in LNCaP-r. Interestingly, androgen responsiveness of LNCaP-r was restored after treated with DNA methyltransferase inhibitor. CONCLUSIONS Our findings may serve as a basis for molecular dissection of the mechanisms involved in development of androgen resistant prostate cancer.
Collapse
Affiliation(s)
- See-Tong Pang
- Department of Surgery, Division of Urology, Chang Gung Memorial Hospital, Kwei-Shan, Tao Yuan, Taiwan.
| | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Bataller M, Portugal J. Apoptosis and cell recovery in response to oxidative stress in p53-deficient prostate carcinoma cells. Arch Biochem Biophys 2005; 437:151-8. [PMID: 15850555 DOI: 10.1016/j.abb.2005.03.008] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2005] [Revised: 03/08/2005] [Accepted: 03/09/2005] [Indexed: 10/25/2022]
Abstract
We have studied the effects of different concentrations of H(2)O(2) on the proliferation of PC-3 prostate carcinoma cells. Since this cell line lacks functional p53, we sought to characterize whether apoptotic response to the oxidative insult was altered such that, unlike in cells containing functional p53 apoptosis may be reduced and replaced by other mechanisms of cellular arrest and death. We did not observe necrosis in PC-3 cells treated with H(2)O(2) concentrations of up to 500 microM. In the presence of 50 microM H(2)O(2), arrest was observed in the G2-phase of the cell cycle, along with p53-independent apoptosis. In the presence of 500 microM H(2)O(2), addition of l-buthionine sulfoximine increased the percentage of apoptotic cell death. Senescence-associated cell arrest was never observed. Moreover, some of the treated cells seemed to be resistant to oxidative damage. These cells re-entered the cell cycle and proliferated normally. Analysis of the expression of p21(waf1) and of p21 protein levels, as well as the activity of caspase-3 and caspase-8, allowed us to characterize some aspects of the arrest of PC-3 cells in G2 and the apoptotic response to oxidative stress in the absence of functional p53.
Collapse
Affiliation(s)
- Marc Bataller
- Instituto de Biología Molecular de Barcelona, CSIC, Parc Cientific de Barcelona, Josep Samitier 1-5, E-08028 Barcelona, Spain
| | | |
Collapse
|
27
|
Marques RB, Erkens-Schulze S, de Ridder CM, Hermans KG, Waltering K, Visakorpi T, Trapman J, Romijn JC, van Weerden WM, Jenster G. Androgen receptor modifications in prostate cancer cells upon long-termandrogen ablation and antiandrogen treatment. Int J Cancer 2005; 117:221-9. [PMID: 15900601 DOI: 10.1002/ijc.21201] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
To study the mechanisms whereby androgen-dependent tumors relapse in patients undergoing androgen blockade, we developed a novel progression model for prostate cancer. The PC346C cell line, established from a transurethral resection of a primary tumor, expresses wild-type (wt) androgen receptor (AR) and secretes prostate-specific antigen (PSA). Optimal proliferation of PC346C requires androgens and is inhibited by the antiandrogen hydroxyflutamide. Orthotopic injection in the dorsal-lateral prostate of castrated athymic nude mice did not produce tumors, whereas fast tumor growth occurred in sham-operated males. Three androgen-independent sublines were derived from PC346C upon long-term in vitro androgen deprivation: PC346DCC, PC346Flu1 and PC346Flu2. PC346DCC exhibited androgen-insensitive growth, which was not inhibited by flutamide. AR and PSA were detected at very low levels, coinciding with background AR activity in a reporter assay, which suggests that these cells have bypassed the AR pathway. PC346Flu1 and PC346Flu2 were derived by culture in steroid-stripped medium supplemented with hydroxyflutamide. PC346Flu1 strongly upregulated AR expression and showed 10-fold higher AR activation than the parental PC346C. PC346Flu1 proliferation was inhibited in vitro by R1881 at 0.1 nM concentration, consistent with a slower tumor growth rate in intact males than in castrated mice. PC346Flu2 carries the well-known T877A AR mutation, causing the receptor to become activated by diverse nonandrogenic ligands including hydroxyflutamide. Array-based comparative genomic hybridization revealed little change between the various PC346 lines. The common alterations include gain of chromosomes 1, 7 and 8q and loss of 13q, which are frequently found in prostate cancer. In conclusion, by in vitro hormone manipulations of a unique androgen-dependent cell line expressing wtAR, we successfully reproduced common AR modifications observed in hormone-refractory prostate cancer: downregulation, overexpression and mutation.
Collapse
Affiliation(s)
- Rute B Marques
- Department of Urology, Josephine Nefkens Institute, Erasmus Medical Center, Rotterdam, the Netherlands
| | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Tagawa H, Karnan S, Kasugai Y, Tuzuki S, Suzuki R, Hosokawa Y, Seto M. MASL1, a candidate oncogene found in amplification at 8p23.1, is translocated in immunoblastic B-cell lymphoma cell line OCI-LY8. Oncogene 2004; 23:2576-81. [PMID: 14691450 DOI: 10.1038/sj.onc.1207352] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Genetic amplification at chromosome 8p23.1 has been reported in some solid tumors. Translocation of 8p23.1 has also been reported in hematological malignancies and head and neck squamous cell cancer. In an attempt to clarify whether this translocation is implicated in lymphomagenesis, we performed FISH analysis of the immunoblastic B-cell lymphoma cell line OCI-LY8, which has chromosome translocation at 8p23.1, with various BAC clones. We found split signals on BAC, RP11-18L2 where the MASL1 gene is located. This translocation was found to produce a chimeric transcript of MASL1 exon 1 with a cryptic exon from the genome region at 14q21. Our study indicates that MASL1 is not only a target gene for genomic amplification but also for chromosomal translocation. Since tumorigenic activity of the MASL1 has not been proven, its in vitro transforming activity was studied and in vivo nude mice assay were performed. Although no in vitro transforming activity was detected by focus formation, the in vivo tumorigenesis assay with nude mice showed that both MASL1 and chimeric MASL1 possess tumorigenic activity. This suggests that MASL1 is an important oncogene not only for solid tumors but also for hematologic malignancies.
Collapse
MESH Headings
- Animals
- Base Sequence
- Cell Cycle Proteins/chemistry
- Cell Cycle Proteins/genetics
- Cell Line, Tumor
- Cell Transformation, Neoplastic
- Chromosome Banding
- Chromosome Mapping
- Chromosomes, Human, Pair 8
- DNA, Neoplasm/genetics
- DNA-Binding Proteins/chemistry
- DNA-Binding Proteins/genetics
- Gene Amplification
- Humans
- In Situ Hybridization, Fluorescence
- Karyotyping
- Lymphoma, B-Cell/genetics
- Lymphoma, B-Cell/pathology
- Mice
- Mice, Nude
- NIH 3T3 Cells
- Neoplasm Transplantation
- Oncogene Proteins/chemistry
- Oncogene Proteins/genetics
- Oncogenes
- Recombinant Proteins/metabolism
- Translocation, Genetic
- Transplantation, Heterologous
Collapse
Affiliation(s)
- Hiroyuki Tagawa
- Division of Molecular Medicine, Aichi Cancer Center Research Institute, 1-1 Kanokoden, Chikusa-ku, Nagoya 464-8681, Japan
| | | | | | | | | | | | | |
Collapse
|
29
|
Celep F, Karagüzel A, Ozgür GK, Yildiz K. Detection of Chromosomal Aberrations in Prostate Cancer by Fluorescence In Situ Hybridization (FISH). Eur Urol 2003; 44:666-71. [PMID: 14644118 DOI: 10.1016/s0302-2838(03)00414-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
PURPOSE Fluorescence in situ hybridization (FISH) is a powerful tool for quantitative analysis of chromosomes and genes and can be applied in a variety of specimens, including cell cultures, isolated nuclei from fresh and fixed tissues, and histological tissue sections. For detection of numerical chromosome aberrations, we examined prostatic cancer samples at our department. In addition, we also observed primary and secondary aberrations taking part in the initiation and progression of tumours. MATERIALS AND METHODS FISH using chromosome-specific alpha-satellite DNA probes for chromosomes 7, 8, 9, 10, 17, X and Y was performed on 19 prostatic cancer and 19 benign prostatic hyperplasia (BPH) samples obtained from transurethral resection (TUR) and archival paraffin-embedded blocks. RESULTS Numerical aberrations were observed in 41% of the tumours studied. A range of aberrant copy numbers of chromosome 9 (68%), 7 (63%), 8 (58%), 17 (37%), Y (32%) and 10 (26%) was observed. We did not observe significant aberrations in BPH samples. In prostate cancer patients, chromosomes 7 (47%), 8 (58%) and 9 (63%) were monosomic by FISH. Monosomy 8 and 9 were significant differences (p>0.05) between prostate cancer and BPH patients. CONCLUSIONS FISH analysis could be observed an one of strongest methods of analysis in detecting numerical aberrations of individual chromosomes with application to paraffin-block samples, metaphase and, interphase nuclei. To our knowledge, this analysis is firstly studied in Turkish patients. Therefore, results of this analysis may be important for Turkish patients.
Collapse
Affiliation(s)
- Figen Celep
- Karadeniz Technical University, Faculty of Medicine, Department of Medical Biology and Genetics, 61080 Trabzon, Turkey.
| | | | | | | |
Collapse
|
30
|
Kumar-Sinha C, Chinnaiyan AM. Molecular markers to identify patients at risk for recurrence after primary treatment for prostate cancer. Urology 2003; 62 Suppl 1:19-35. [PMID: 14747039 DOI: 10.1016/j.urology.2003.10.007] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Accurate prognostication is a prerequisite for accurate therapeutics and management of prostate cancer because indolent tumors may require no intervention, whereas aggressive tumors lead to patient mortality. There is a critical need to define these subgroups of patients with prostate cancer differing in clinical outcome. Prognostic nomograms based on clinical data provide useful predictions of clinical states and outcomes, but they need further refinements to improve accuracy and universality. Genomic and proteomic analyses have provided many novel markers that may help define prognostic parameters based on the underlying biology of prostate cancer progression at the molecular level. These molecular markers are likely to augment traditional prognostic modalities by providing a set of molecularly defined and quantifiable variables. Encompassing the genome, transcriptome, and proteome of prostate cancer will likely provide "molecular signatures" that will bridge prognostication, prediction, and treatment in a single continuum.
Collapse
Affiliation(s)
- Chandan Kumar-Sinha
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan 48109-0602, USA
| | | |
Collapse
|
31
|
Martel CL, Gumerlock PH, Meyers FJ, Lara PN. Current strategies in the management of hormone refractory prostate cancer. Cancer Treat Rev 2003; 29:171-87. [PMID: 12787712 DOI: 10.1016/s0305-7372(02)00090-7] [Citation(s) in RCA: 71] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Prostate cancer is the most common cancer diagnosed in American males, and is the second leading cause of cancer-related deaths. Most patients who develop metastatic disease will initially respond to androgen deprivation, but response is invariably temporary. Most patients will develop androgen-independent ("hormone-refractory") disease that results in progressive clinical deterioration and ultimately death. This progression to androgen independence is accompanied by increasingly evident DNA instability and alterations in genes and gene expression, including mutations in p53, over-expression of Bcl2, and mutations in the androgen receptor gene, among others. Treatment options for hormone refractory disease include intensive supportive care, radiotherapy, bisphosphonates, second-line hormonal manipulations, cytotoxic chemotherapy and investigational agents. A post-treatment reduction in the level of prostate specific antigen (PSA) by 50% has been shown to correlate with survival and has been accepted by consensus as a valid endpoint in clinical trials. Chemotherapeutic agents such as mitoxantrone, estramustine, and the taxanes have yielded improved response rates and palliative benefit, but not improved survival. Therefore, current efforts must be focused on enrolling patients onto clinical trials of investigational agents with novel mechanisms of action, and on using survival, time to progression, and quality of life as end points in routine clinical practice.
Collapse
Affiliation(s)
- Cynthia L Martel
- Division of Hematology and Oncology, University of California, Davis, Cancer Center, 4501 X Street, Sacramento, CA 95817, USA
| | | | | | | |
Collapse
|
32
|
Dumur CI, Dechsukhum C, Ware JL, Cofield SS, Best AM, Wilkinson DS, Garrett CT, Ferreira-Gonzalez A. Genome-wide detection of LOH in prostate cancer using human SNP microarray technology. Genomics 2003; 81:260-9. [PMID: 12659810 DOI: 10.1016/s0888-7543(03)00020-x] [Citation(s) in RCA: 79] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Loss of heterozygosity (LOH) of chromosomal regions is crucial in tumor progression. In this study we assessed the potential of the Affymetrix GeneChip HuSNP mapping assay for detecting genome-wide LOH in prostate tumors. We analyzed two human prostate cell lines, P69SV40Tag (P69) and its tumorigenic subline, M12, and 11 prostate cancer cases. The M12 cells showed LOH in chromosomes 3p12.1-p22.1, 11q22.1-q24.2, 19p13.12, and 19q13.42. All of the prostate cases with informative single-nucleotide polymorphism (SNP) markers showed LOH in 1p31.2, 10q11.21, 12p13.1, 16q23.1-q23.2, 17p13.3, 17q21.31, and 21q21.2. Additionally, a high percentage of cases showed LOH at 6p25.1-p25.3 (75%), 8p22-p23.2, and 10q22.1 (70%). Several tumor suppressor genes (TSGs) have been mapped in these loci. These results demonstrate that the HuSNP mapping assay can serve as an alternative to comparative genomic hybridization for assessing genome-wide LOH and can identify chromosomal regions harboring candidate TSGs implicated in prostate cancer.
Collapse
Affiliation(s)
- Catherine I Dumur
- Department of Pathology, Virginia Commonwealth University, Richmond, VA23298-0248, USA
| | | | | | | | | | | | | | | |
Collapse
|
33
|
Stathopoulos EN, Dambaki C, Kampa M, Theodoropoulos PA, Anezinis P, Delakas D, Delides GS, Castanas E. Membrane androgen binding sites are preferentially expressed in human prostate carcinoma cells. BMC Clin Pathol 2003; 3:1. [PMID: 12588669 PMCID: PMC149438 DOI: 10.1186/1472-6890-3-1] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2002] [Accepted: 01/30/2003] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Prostate cancer is one of the most frequent malignancies in males. Nevertheless, to this moment, there is no specific routine diagnostic marker to be used in clinical practice. Recently, the identification of a membrane testosterone binding site involved in the remodeling of actin cytoskeleton structures and PSA secretion, on LNCaP human prostate cancer cells has been reported. We have investigated whether this membrane testosterone binding component could be of value for the identification of prostate cancer. METHODS Using a non-internalizable testosterone-BSA-FITC analog, proven to bind on membrane sites only in LNCaP cells, we have investigated the expression of membrane testosterone binding sites in a series of prostate carcinomas (n = 14), morphologically normal epithelia, taken from areas of the surgical specimens far from the location of the carcinomas (n = 8) and benign prostate hyperplasia epithelia (n = 10). Isolated epithelial cells were studied by flow cytometry, and touching preparations, after 10-min incubation. In addition, routine histological slides were assayed by confocal laser microscopy. RESULTS We show that membrane testosterone binding sites are preferentially expressed in prostate carcinoma cells, while BPH and non-malignant epithelial cells show a low or absent binding. CONCLUSIONS Our results indicate that membrane testosterone receptors might be of use for the rapid routine identification of prostate cancer, representing a new diagnostic marker of the disease.
Collapse
Affiliation(s)
| | - Constantina Dambaki
- Department of Pathology, University of Crete, School of Medicine, Heraklion, Greece
| | - Marilena Kampa
- Department of Experimental Endocrinology, University of Crete, School of Medicine, Heraklion, Greece
| | | | | | | | - George S Delides
- Department of Pathology, University of Crete, School of Medicine, Heraklion, Greece
| | - Elias Castanas
- Department of Experimental Endocrinology, University of Crete, School of Medicine, Heraklion, Greece
| |
Collapse
|
34
|
Devi GR, Oldenkamp JR, London CA, Iversen PL. Inhibition of human chorionic gonadotropin beta-subunit modulates the mitogenic effect of c-myc in human prostate cancer cells. Prostate 2002; 53:200-10. [PMID: 12386920 DOI: 10.1002/pros.10151] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
BACKGROUND Amplification of the proto-oncogene c-myc has been identified as one of the most common genetic alterations in prostate cancer, thus making it an attractive therapeutic target. However, certain prostate cancer cells are unresponsive to c-Myc inhibition. The purpose of this study was to test the hypothesis that effective growth inhibition in the refractory cancer cells can be achieved by blocking c-myc along with a growth factor using a novel phosphorodiamidate morpholino antisense oligomer-based approach. Human chorionic gonadotropin, a growth factor implicated in neoplasm, causes activation of c-myc through a G-protein-coupled pathway of signal transduction. METHODS In this study, the effect of inhibition of beta-hCG and c-myc singly or in combination was evaluated in DU145 (RB -/-, p53-/-, androgen-independent) and LNCaP (Rb+/+, p53 +/+, androgen-sensitive) human prostate cancer cell lines and in a DU145 subcutaneous xenograft murine model. RESULTS Antisense phosphorodiamidate morpholino oligomers directed against beta-hCG and c-myc caused a specific decrease of the target protein levels. Unlike LNCaP cells, DU145 cell growth was refractory to c-Myc inhibition. Unresponsiveness to c-myc inhibition in DU145 cells was overcome by targeting both beta-hCG and c-myc genes, resulting in potentiation of the antiproliferative effect seen with inhibition of beta-hCG alone. CONCLUSIONS The inhibition of beta-hCG sensitizes prostate cancer cells to the antiproliferative effects of c-Myc inhibition, including tumors that are refractory to c-Myc decrease alone.
Collapse
|
35
|
Weihua Z, Lathe R, Warner M, Gustafsson JA. An endocrine pathway in the prostate, ERbeta, AR, 5alpha-androstane-3beta,17beta-diol, and CYP7B1, regulates prostate growth. Proc Natl Acad Sci U S A 2002; 99:13589-94. [PMID: 12370428 PMCID: PMC129718 DOI: 10.1073/pnas.162477299] [Citation(s) in RCA: 264] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Epithelial proliferation of the ventral prostate in rodents peaks between 2 and 4 weeks of age, and by week 8, proliferating cells are rare. We have used ERbeta(-/-) and CYP7B1(-/-) mice to investigate the role of ERbeta and one of its ligands, 5alpha-androstane-3beta,17beta-diol (3betaAdiol), in growth of the ventral prostate. Before puberty, ERbeta was found in quiescent but not in proliferating cells, and proliferating cells occurred more frequently in ventral prostates of ERbeta(-/-) mice than in wild-type littermates. Treatment with 3betaAdiol decreased proliferation in wild-type but not in ERbeta(-/-) mice. In rats, treatment with 3betaAdiol from postnatal day 2 to 28 resulted in reduction in growth of ventral prostates. The prostates of CYP7B1(-/-) mice were hypoproliferative before puberty and smaller than those of their wild-type littermates after puberty. Because CYP7B1 represents the major pathway for inactivating 3betaAdiol in the prostate, we suggest that ERbeta, 3betaAdiol, and CYP7B1 are the components of a pathway that regulates growth of the rodent ventral prostate. In this pathway, ERbeta is an antiproliferative receptor, 3betaAdiol is an ERbeta ligand, and CYP7B1 is the enzyme that regulates ERbeta function by regulating the level of 3betaAdiol.
Collapse
Affiliation(s)
- Zhang Weihua
- Department of Medical Nutrition, Karolinska Institute, Novum, S-141 86 Huddinge, Sweden
| | | | | | | |
Collapse
|
36
|
Steiner T, Junker K, Burkhardt F, Braunsdorf A, Janitzky V, Schubert J. Gain in chromosome 8q correlates with early progression in hormonal treated prostate cancer. Eur Urol 2002; 41:167-71. [PMID: 12074404 DOI: 10.1016/s0302-2838(01)00030-6] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
OBJECTIVE Development and especially hormone refractant progression of prostate cancer are incompletely understood. Clinical studies evaluating genetic aberrations of prior therapy biopsies in correlation with progression data in patients receiving hormone therapy for prostate cancer have not been performed until now. METHODS After DNA isolation from histological sections of primary prostate cancer biopsies, comparative genomic hybridization (CGH) was performed according to standard protocols. Primary staging, clinical course and PSA levels of the patients were assessed. RESULTS CGH was performed on 28 primary prostate cancer samples. After a mean follow-up of 36 months 11 (39%) of the patients showed progression of disease under hormonal treatment. In patients without and with progression we found the following results, respectively: losses of 6q (41/36%), 8p (41/45%), 16q (23/18%), 18q (30/9%), and gains of 8q (12/64%; P < 0.0001) and 17 (47/26%). CONCLUSIONS Gain of 8q is found predominantly in primary core biopsies of local advanced or metastasized prostate cancers. It shows in univariate analysis significant correlation with progression in hormone treated prostate cancer. This fact suggests that gain in 8q represents a marker of aggressiveness in prostate cancer.
Collapse
Affiliation(s)
- Thomas Steiner
- Department of Urology, Friedrich-Schiller-University, Jena, Germany.
| | | | | | | | | | | |
Collapse
|