Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
1.
Sci Rep ; 7(1): 5124, 2017 07 11.
Article in English | MEDLINE | ID: mdl-28698647

ABSTRACT

Prostate cancer is the most common cancer among western men, with a significant mortality and morbidity reported for advanced metastatic disease. Current understanding of metastatic disease is limited due to difficulty of sampling as prostate cancer mainly metastasizes to bone. By analysing prostate cancer bone metastases using high density microarrays, we found a common genomic copy number loss at 6q16.1-16.2, containing the FBXL4 gene, which was confirmed in larger series of bone metastases by fluorescence in situ hybridisation (FISH). Loss of FBXL4 was also detected in primary tumours and it was highly associated with prognostic factors including high Gleason score, clinical stage, prostate-specific antigen (PSA) and extent of disease, as well as poor patient survival, suggesting that FBXL4 loss contributes to prostate cancer progression. We also demonstrated that FBXL4 deletion is detectable in circulating tumour cells (CTCs), making it a potential prognostic biomarker by 'liquid biopsy'. In vitro analysis showed that FBXL4 plays a role in regulating the migration and invasion of prostate cancer cells. FBXL4 potentially controls cancer metastasis through regulation of ERLEC1 levels. Therefore, FBXL4 could be a potential novel prostate cancer suppressor gene, which may prevent cancer progression and metastasis through controlling cell invasion.


Subject(s)
Bone Neoplasms/genetics , Bone Neoplasms/secondary , F-Box Proteins/genetics , F-Box Proteins/metabolism , Prostatic Neoplasms/genetics , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Bone Neoplasms/metabolism , Bone Neoplasms/pathology , Cell Line, Tumor , Cell Movement , Disease Progression , Down-Regulation , Gene Deletion , Gene Dosage , Gene Expression Regulation, Neoplastic , Genetic Association Studies , Genetic Predisposition to Disease , Humans , Lectins/metabolism , Male , Neoplasm Staging , Neoplastic Cells, Circulating/metabolism , Polymorphism, Single Nucleotide , Prognosis , Prostate-Specific Antigen/metabolism , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Survival Analysis
2.
Clin Cancer Res ; 23(17): 5112-5122, 2017 Sep 01.
Article in English | MEDLINE | ID: mdl-28615267

ABSTRACT

Purpose: To develop an approach for the investigation of different subtypes of circulating tumor cells (CTC) and other cells to evaluate their potential prognostic value of prostate cancer.Experimental Design: Malignancy of CTCs undergoing epithelial-to-mesenchymal transition (EMT) was confirmed by repeated FISH. Subgroups of CTCs in 81 patients with prostate cancer (43 castration resistant and 38 untreated localized) were correlated to disease aggressiveness parameters. AUC analysis was applied to compare the performance for metastasis prediction between serum PSA level alone and a combined risk score using both PSA and EMTing CTC count. Circulating megakaryocytes and cancer patient survival association was performed using Cox model.Results: The majority of vimentin (VIM)+/CD45- cells were malignant, with genomic alterations in several genomic regions. The number of cytokeratin (CK)-/VIM+/CD45- CTCs correlated with disease burden, tumor aggressiveness, and poorer survival. Meanwhile, CK+/VIM+/CD45- CTCs were associated with metastases better than other subtypes of CTCs in these limited samples. Combination of PSA level and the number of CK+/VIM+/CD45- CTCs enhanced the prediction of cancer metastases [AUC, 0.921; 95% confidence interval (CI), 0.858-0.985]. The number of circulating megakaryocytes was potentially associated with good patient survival in advanced prostate cancer (HR, 0.849; 95% CI, 0.628-1.146, per cell increase), and the difference between the number of mesenchymal CTCs and megakaryocytes strongly correlated to poor survival (HR, 10.17; 95% CI, 2.164-47.789, if score ≥2.0).Conclusions: This CTC analysis approach and the potential association of megakaryocytes with cancer prognosis may greatly enhance our ability to investigate the cancer metastasis process and to predict/monitor cancer progression. Clin Cancer Res; 23(17); 5112-22. ©2017 AACR.


Subject(s)
Biomarkers, Tumor/blood , Megakaryocytes/pathology , Neoplastic Cells, Circulating/pathology , Prostatic Neoplasms/blood , Aged , Aged, 80 and over , Cell Line, Tumor , Disease-Free Survival , Epithelial-Mesenchymal Transition/genetics , Humans , In Situ Hybridization, Fluorescence , Male , Middle Aged , Prognosis , Prostate-Specific Antigen/blood , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology
3.
J Pathol ; 232(5): 566-77, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24407904

ABSTRACT

Genomic changes affecting tumour suppressor genes are fundamental to cancer. We applied SNP array analysis to a panel of testicular germ cell tumours to search for novel tumour suppressor genes and identified a frequent small deletion on 6q25.3 affecting just one gene, ZDHHC14. The expression of ZDHHC14, a putative protein palmitoyltransferase with unknown cellular function, was decreased at both RNA and protein levels in testicular germ cell tumours. ZDHHC14 expression was also significantly decreased in a panel of prostate cancer samples and cell lines. In addition to our findings of genetic and protein expression changes in clinical samples, inducible overexpression of ZDHHC14 led to reduced cell viability and increased apoptosis through the classic caspase-dependent apoptotic pathway and heterozygous knockout of ZDHHC14 increased [CORRECTED] cell colony formation ability. Finally, we confirmed our in vitro findings of the tumour suppressor role of ZDHHC14 in a mouse xenograft model, showing that overexpression of ZDHHC14 inhibits tumourigenesis. Thus, we have identified a novel tumour suppressor gene that is commonly down-regulated in testicular germ cell tumours and prostate cancer, as well as given insight into the cellular functional role of ZDHHC14, a potential protein palmitoyltransferase that may play a key protective role in cancer.


Subject(s)
Acyltransferases/genetics , Genes, Tumor Suppressor , Neoplasms, Germ Cell and Embryonal/genetics , Prostatic Neoplasms/genetics , Testicular Neoplasms/genetics , Acyltransferases/metabolism , Animals , Apoptosis , Cell Line, Tumor , Cell Proliferation , Cell Survival , Down-Regulation , Gene Deletion , Gene Expression Profiling/methods , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , HEK293 Cells , Humans , Male , Mice , Mice, Nude , Neoplasms, Germ Cell and Embryonal/enzymology , Neoplasms, Germ Cell and Embryonal/pathology , Oligonucleotide Array Sequence Analysis , Polymorphism, Single Nucleotide , Prostatic Neoplasms/enzymology , Prostatic Neoplasms/pathology , Prostatic Neoplasms/prevention & control , RNA Interference , Testicular Neoplasms/enzymology , Testicular Neoplasms/pathology , Time Factors , Transfection , Tumor Burden
4.
Genes Chromosomes Cancer ; 51(6): 579-89, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22334418

ABSTRACT

Many human cancers present as multifocal lesions. Understanding the clonal origin of multifocal cancers is of both etiological and clinical importance. The molecular basis of multifocal prostate cancer has previously been explored using a limited number of isolated markers and, although independent origin is widely believed, the clonal origin of multifocal prostate cancer is still debatable. We attempted to address clonal origin using a genome-wide copy-number analysis of individual cancer and high-grade prostatic intraepithelial neoplasia (HGPIN) lesions. Using Affymetrix array 6.0 copy-number analysis, we compared the genomic changes detected in 48 individual cancer and HGPIN lesions, isolated from 18 clinically localized prostate cancer cases. Identical genomic copy-number changes, shared by all same-case cancer foci, were detected in all 13 informative cases displaying multiple tumor foci. In addition, individual HGPIN lesions in the two multifocal-HGPIN cases available shared identical genomic changes. Commonly known genomic alterations, including losses at 6q15, 8p21.3-8p21.2, 10q23.2-10q23.31, 16q22.3, 16q23.2-16q23.3 and 21q22.2-21q22.3 regions and gain of 8q24.3 were the most frequently detected changes in this study and each was detected in all same-case foci in at least one case. Microarray data were confirmed by fluorescence in situ hybridization in selected foci. Our high-resolution genome-wide copy-number data suggest that many multifocal cases derive from a single prostate cancer precursor clone and that this precursor may give rise to separate HGPIN foci and may further progress to multifocal invasive prostate cancer. These findings, which demonstrate the monoclonal origin of multifocal prostate cancer, should significantly enhance our understanding of prostate carcinogenesis.


Subject(s)
Gene Dosage , Genome, Human , Prostatic Intraepithelial Neoplasia/genetics , Prostatic Neoplasms/genetics , Aged , Clonal Evolution , Humans , In Situ Hybridization, Fluorescence , Male , Middle Aged , Neoplastic Processes , Oligonucleotide Array Sequence Analysis , Prostatic Intraepithelial Neoplasia/pathology , Prostatic Neoplasms/pathology , Reproducibility of Results
5.
Cancer Res ; 70(23): 9544-8, 2010 Dec 01.
Article in English | MEDLINE | ID: mdl-20947519

ABSTRACT

Fusion genes play important roles in tumorigenesis. The identification of the high-frequency TMPRSS2 fusion with ERG and other ETS family genes in prostate cancer highlights the importance of fusion genes in solid tumor development and progression. However, the mechanisms leading to these fusions are unclear. We investigated whether androgen, through stimulating its receptor, could promote spatial genome reorganization and contribute to the generation of the TMPRSS2:ERG fusion. We show that treatment with androgen can induce the TMPRSS2:ERG fusion in both malignant and nonmalignant prostate epithelial cells. Although the fusion could be detected in malignant cells following 24-hour treatment, prolonged exposure to androgen was required to detect the fusion transcript in nonmalignant cells. We associated the fusion incidence with genetic factors, including androgen-induced gene proximity, androgen receptor exon1 CAG repeat length and expression of the PIWIL1 gene. This study demonstrates that fusions can be induced prior to malignant transformation and generation of the fusion is associated with both gene proximity and loss of the ability to prevent double-strand breaks.


Subject(s)
Dihydrotestosterone/pharmacology , Epithelial Cells/drug effects , Gene Fusion/drug effects , Oncogene Proteins, Fusion/genetics , Androgens/pharmacology , Argonaute Proteins , Cell Line, Transformed , Cell Line, Tumor , Dose-Response Relationship, Drug , Epithelial Cells/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Humans , In Situ Hybridization, Fluorescence , Male , Prostate/cytology , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Proteins/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Androgen/genetics , Receptors, Androgen/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Time Factors
6.
Cancer Res ; 70(13): 5207-12, 2010 Jul 01.
Article in English | MEDLINE | ID: mdl-20516122

ABSTRACT

Prostate cancer is significantly more common in Western men than in Asian men, but the basis for this difference remains unknown. Because genomic studies of Asian prostate cancer are very limited, we used a genome-wide approach to reveal the genomic alterations in Chinese prostate cancers. We found a significant reduction in the frequency of certain somatic genomic changes that are commonly found in Western prostate cancers, including the 21q22.2-22.3 deletion, which involves the TMPRSS2:ERG fusion gene, and 10q deletion, which causes PTEN inactivation. Array results were confirmed by PCR-based molecular copy-number counting in selected samples. The different frequencies of these genomic changes were further evaluated by fluorescent in situ hybridization and immunohistochemistry analyses of tissue microarray samples. These alterations might be key genetic changes underlying the regional/ethnic difference in clinical incidence and might be induced by specific environmental and/or genetic risk factors that Western men are exposed to. Our findings suggest that tumors arise in Western and Chinese populations by alternative pathogenetic mechanisms.


Subject(s)
Asian People/genetics , Prostatic Neoplasms/ethnology , Prostatic Neoplasms/genetics , White People/genetics , China , Gene Rearrangement , Genome, Human , Humans , Male , Oncogene Proteins, Fusion/genetics , PTEN Phosphohydrolase/genetics , Polymorphism, Single Nucleotide , Prostatic Neoplasms/pathology , Trans-Activators/genetics , Transcriptional Regulator ERG , United Kingdom
7.
Am J Pathol ; 176(6): 2607-15, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20395447

ABSTRACT

Development of chemoresistance limits the clinical efficiency of platinum-based therapy. Although many resistance mechanisms have been demonstrated, genetic/molecular alterations responsible for drug resistance in the majority of clinical cases have not been identified. We analyzed three pairs of testicular germ cell tumor cell lines using Affymetrix expression microarrays and revealed a limited number of differentially expressed genes across the cell lines when comparing the parental and resistant cells. Among them, CCND1 was the most significantly differentially expressed gene. Analysis of testicular germ cell tumor clinical samples by quantitative reverse transcription PCR analysis revealed that overall expression of CCND1 was significantly higher in resistant cases compared with sensitive samples (P < 0.0001). We also found that CCND1 was dramatically overexpressed both in induced and intrinsically resistant samples of ovarian and prostate cancer. Finally combined CCND1 knockdown using small-interfering RNA and cisplatin treatment inhibited cell growth in vitro significantly more effectively than any of these single treatments. Therefore, deregulation of CCND1 may be a major cause of cisplatin resistance in testicular germ cell tumors and may also be implicated in ovarian and prostate cancers. CCND1 could be potentially used as a marker for treatment stratification and as a molecular target to improve the treatment of platinum-resistant tumors.


Subject(s)
Antineoplastic Agents/therapeutic use , Cisplatin/therapeutic use , Cyclin D1/genetics , Drug Resistance, Neoplasm/genetics , Neoplasms, Germ Cell and Embryonal , Testicular Neoplasms , Cell Line, Tumor , Cell Proliferation , Cell Survival , Comparative Genomic Hybridization , Cyclin D1/metabolism , Female , Gene Expression Profiling , Humans , Male , Microarray Analysis , Neoplasms, Germ Cell and Embryonal/drug therapy , Neoplasms, Germ Cell and Embryonal/pathology , Neoplasms, Germ Cell and Embryonal/physiopathology , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/pathology , Ovarian Neoplasms/physiopathology , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/pathology , Prostatic Neoplasms/physiopathology , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Testicular Neoplasms/drug therapy , Testicular Neoplasms/pathology , Testicular Neoplasms/physiopathology
10.
Genes Chromosomes Cancer ; 47(7): 604-13, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18384131

ABSTRACT

Since the introduction of cisplatin into the clinic, the treatment of patients with a variety of solid tumors including testicular germ cell tumors, ovarian and lung cancers, has dramatically improved. One of the main causes for therapeutic failure in these malignancies is the development of drug resistance. Testicular germ cell tumors (TGCTs), the most common malignancy in young men, exhibit extreme sensitivity to cisplatin-based chemotherapy, making them an ideal model for investigating the mechanisms of cisplatin chemo-sensitivity and resistance. TGCT development and pathogenesis have been well studied but little is known about the genetic background in chemo-resistant cases. We investigated genomic differences between three TGCT parental cell lines and their cisplatin resistant derivatives. Using 10K single nucleotide polymorphism (SNP) microarray analysis, we identified two small chromosomal regions with consistent copy number changes across all three pairs of resistant cell lines. These were an 8.7 Mb region at 6q26-27, which displayed consistent copy number gain and a 0.3 Mb deletion involving 4 SNPs at 10p14. Both the chromosomal gain and loss were confirmed by fluorescence in situ hybridization. The significance of these regions should be further investigated as they may contain key genes involved in the development of chemo- resistance to cisplatin-based treatment in TGCTs and other cancers.


Subject(s)
Antineoplastic Agents/therapeutic use , Chromosome Aberrations , Cisplatin/therapeutic use , Drug Resistance, Neoplasm/genetics , Testicular Neoplasms/drug therapy , Testicular Neoplasms/genetics , Gene Dosage , Humans , In Situ Hybridization, Fluorescence , Male , Microarray Analysis , Polymorphism, Single Nucleotide , Tumor Cells, Cultured
11.
Asian J Androl ; 10(3): 467-73, 2008 May.
Article in English | MEDLINE | ID: mdl-18385909

ABSTRACT

AIM: To investigate the existence of TMPRSS2:ERG fusion gene in circulating tumor cells (CTC) from prostate cancer patients and its potential in monitoring tumor metastasis. METHODS: We analyzed the frequency of TMPRSS2:ERG and TMPRSS2:ETV1 transcripts in 27 prostate cancer biopsies from prostatectomies, and TMPRSS2:ERG transcripts in CTC isolated from 15 patients with advanced androgen independent disease using reverse transcription polymerase chain reaction (RT-PCR). Fluorescence in situ hybridization (FISH) was applied to analyze the genomic truncation of ERG, which is the result of TMPRSS2:ERG fusion in 10 of the 15 CTC samples. RESULTS: TMPRSS2:ERG transcripts were found in 44% of our samples, but we did not detect expression of TMPRSS2:ETV1. Using FISH analysis we detected chromosomal rearrangements affecting the ERG gene in 6 of 10 CTC samples, including 1 case with associated TMPRSS2:ERG fusion at the primary site. However, TMPRSS2:ERG transcripts were not detected in any of the 15 CTC samples, including the 10 cases analyzed by FISH. CONCLUSION: Although further study is required to address the association between TMPRSS2:ERG fusion and prostate cancer metastasis, detection of genomic truncation of the ERG gene by FISH analysis could be useful for monitoring the appearance of CTC and the potential for prostate cancer metastasis.


Subject(s)
Oncogene Proteins, Fusion/genetics , Prostatic Neoplasms/genetics , Base Sequence , DNA Primers , Humans , In Situ Hybridization, Fluorescence , Male , Neoplastic Cells, Circulating , Prostatic Neoplasms/blood , Reverse Transcriptase Polymerase Chain Reaction
12.
Eur Urol ; 54(6): 1333-43, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18262716

ABSTRACT

OBJECTIVES: Androgen-deprivation therapy effectively shrinks hormone-naïve prostate cancer, both in the prostate and at sites of distant metastasis. However prolonged androgen deprivation generally results in relapse and androgen-independent tumour growth, which is inevitably fatal. The molecular events that enable prostate cancer cells to proliferate in reduced androgen conditions are poorly understood. Here we investigate the role of Hedgehog signalling in androgen-independent prostate cancer (AIPC). METHODS: Activity of the Hedgehog signalling pathway was analysed in cultured prostate cancer cells, and circulating prostate tumour cells were isolated from blood samples of patients with AIPC. RESULTS: AIPC cells were derived through prolonged culture in reduced androgen conditions, modelling hormone therapy in patients, and expressed increased levels of Hedgehog signalling proteins. Exposure of cultured AIPC cells to cyclopamine, which inhibits Hedgehog signalling, resulted in inhibition of cancer cell growth. The expression of the Hedgehog receptor PTCH and the highly prostate cancer-specific gene DD3(PCA3) was significantly higher in circulating prostate cancer cells isolated from patients with AIPC compared with samples prepared from normal individuals. There was an association between PTCH and DD3(PCA3) expression and the length of androgen-ablation therapy. CONCLUSIONS: Our data are consistent with reports implicating overactivity of Hedgehog signalling in prostate cancer and suggest that Hedgehog signalling contributes to the androgen-independent growth of prostate cancer cells. As systemic anti-Hedgehog medicines are developed, the Hedgehog pathway will become a potential new therapeutic target in advanced prostate cancer.


Subject(s)
Hedgehog Proteins/physiology , Prostatic Neoplasms/etiology , Signal Transduction , Aged , Aged, 80 and over , Androgens/physiology , Humans , Male , Middle Aged
13.
BJU Int ; 101(5): 570-4, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18257857

ABSTRACT

OBJECTIVE: To examine the nodal (N+) vs extranodal (M+) staging in each of the International Germ Cell Consensus Classification Group (IGCCCG) subgroups in an audit of 437 patients treated in The Anglian Germ Cell Cancer Group, where chemotherapy was the primary management, as there is an increasingly earlier presentation of patients with less advanced disease who thus face potentially unnecessary treatment. PATIENTS AND METHODS: Clinicians from seven centres prospectively registered patients in a central database, and the follow-up was coordinated by one of the authors. RESULTS: Between 1982 and 2002, 436 patients (median follow-up 60 months) were registered; 63% of IGCCCG good risk (298), 42% of intermediate (62) and 8% poor risk (77) were stage II; 79% of N+M0 intermediate and poor risk cases (29) were alive, vs only 60% of M+ stage IV cases (92, P < 0.05). The trend was similar in IGCCCG good risk patients, with 92% of N+ stage II (156) alive vs only 85% (94) of stage IV M+ (not significant). The frequency of retroperitoneal lymph node dissection after chemotherapy increased from 26% (1983-1993) to 34% (1994-2002), and survival from 89% to 94%. There were no relapses in eight patients who elected to stop treatment after two courses. Four of six patients with positive findings on positron emission tomography had a durable complete response, assessed by standard uptake values, when tested at 72-96 h. CONCLUSION: Extra-lymphatic spread, although prognostically important within the IGCCCG subgroups, is only statistically significant for intermediate and poor risk combined. The observation that there might be N+ patients cured by two chemotherapy courses alone suggests that there might be opportunities to reduce the morbidity of treatment.


Subject(s)
Antineoplastic Agents/administration & dosage , Neoplasms, Germ Cell and Embryonal/drug therapy , Positron-Emission Tomography , Testicular Neoplasms/drug therapy , Adult , Cohort Studies , Humans , Male , Middle Aged , Neoplasm Recurrence, Local , Neoplasm Staging , Neoplasms, Germ Cell and Embryonal/diagnostic imaging , Neoplasms, Germ Cell and Embryonal/mortality , Neoplasms, Germ Cell and Embryonal/pathology , Pilot Projects , Prospective Studies , Risk Factors , Survival Analysis , Testicular Neoplasms/diagnostic imaging , Testicular Neoplasms/mortality , Testicular Neoplasms/pathology , Tomography, X-Ray Computed , Treatment Outcome
14.
J Urol ; 177(5): 1907-12, 2007 May.
Article in English | MEDLINE | ID: mdl-17437846

ABSTRACT

PURPOSE: We developed and describe a practical method by which primary prostate cancer specimens can be screened for recurrent chromosomal translocations, which is a potential source of fusion genes, as well as a process by which identified translocations can be mapped to define the genes involved. MATERIALS AND METHODS: A series of 7 prostate cancer cell lines and 25 transiently established primary cell cultures, which were sourced from tissue harvested at 16 radical prostatectomies and 9 channel transurethral prostate resections, were screened for chromosomal translocations using multiplex-fluorescence in situ hybridization technology. A series of fluorescence in situ hybridization based breakpoint mapping experiments were performed to identify candidate genes involved in regions associated with recurrent translocation. RESULTS: Our analysis identified the repetition of 2 translocations in prostate cancer lines, that is t(1;15) and t(4;6), at a frequency of 28% and 57%, respectively. More significantly 4 of the 25 subsequently established primary cultures (16%) also revealed a t(4;6) translocation. Using the LNCaP cell line the breakpoints involved were mapped to the t(4;6)(q22;q15) region and a number of candidate genes were identified. CONCLUSIONS: We found that the t(4;6) translocation is also a repeat event in primary cell cultures from malignant prostate cancer. Breakpoint mapping showed that the gene UNC5C loses its promoter and first exon as a direct result of the translocation in the 4q22 region. As such, we identified it as a possible contributor to a putative fusion gene in prostate cancer.


Subject(s)
Chromosomes, Human, Pair 4 , Chromosomes, Human, Pair 6 , Prostatic Neoplasms/genetics , Translocation, Genetic/genetics , Cell Line, Tumor , Chromosome Breakage , Chromosome Mapping/methods , Humans , In Situ Hybridization, Fluorescence , Male , Prostatic Neoplasms/pathology , Terminal Repeat Sequences
15.
Genes Chromosomes Cancer ; 46(7): 675-83, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17431877

ABSTRACT

Many techniques have been developed in recent years for genome-wide analysis of genetic alterations, but no current approach is capable of rapidly identifying all chromosome rearrangements with precise definition of breakpoints. Combining multiple color fluorescent in situ hybridization and high-density single nucleotide polymorphism array analyses, we present here an approach for high resolution karyotyping and fast identification of chromosome breakpoints. We characterized all of the chromosome amplifications and deletions, and most of the chromosome translocation breakpoints of three prostate cancer cell lines at a resolution which can be further analyzed by sequence-based techniques. Genes at the breakpoints were readily determined and potentially fused genes identified. Using high-density exon arrays we simultaneously confirmed altered exon expression patterns in many of these breakpoint genes.


Subject(s)
Chromosome Fragile Sites , Humans , In Situ Hybridization, Fluorescence , Karyotyping , Polymorphism, Single Nucleotide
17.
BJU Int ; 96(6): 819-21, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16153209

ABSTRACT

OBJECTIVE: To assess the size and stage of testicular tumours on presentation in the period 1984-2002. PATIENTS AND METHODS: Demographic details and information on staging on 550 patients treated at St. Bartholomew's and the Royal London Hospital in the period 1984-2002 were collected prospectively in the departmental database. Information on testicular size was obtained by reviewing the histopathology records, and the maximum dimension of the tumour as measured in the gross specimen was taken as the size of the testicular tumour. RESULTS: The period 1984-2002 was divided into three intervals, i.e. 1984-95, 1996-98 and 1999-2002. The mean testicular tumour size in the three intervals decreased from 4 cm (162 tumours) to 3.2 cm (85) and 2.5 cm (72; P = 0.002, Student's t-test). The proportion of tumours of <2 cm on presentation also increased, from 11% to 14% and 23% in the three intervals, respectively, while the proportion of patients with stage 1 disease increased from 57%, to 63% and 77%, respectively. CONCLUSIONS: The size of testicular tumours on presentation has shown a consistent decline in the last two decades, the mean size now being 2.5 cm. That 23% are now <2 cm raises the possibility of testis-preserving surgery in this young group of patients, who have an excellent prognosis, and therefore in the long-term issues such as psychological morbidity and natural fertility assume greater importance. There is a need for a randomized controlled trial to evaluate these issues.


Subject(s)
Germinoma/pathology , Testicular Neoplasms/pathology , Early Diagnosis , Humans , Male , Neoplasm Staging , Prospective Studies
18.
Nat Clin Pract Urol ; 2(12): 590-1, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16474544
19.
Cancer Causes Control ; 15(3): 277-83, 2004 Apr.
Article in English | MEDLINE | ID: mdl-15090722

ABSTRACT

OBJECTIVE: To investigate the role of a range of maternal and pre-natal characteristics as potential risk factors for testicular cancer. METHODS: A population-based case-control study of testicular cancer. Mothers of participants completed a questionnaire about their reproductive and obstetric history. RESULTS: The risk of testicular cancer was approximately doubled for sons of mothers aged 15-19 years at conception compared with mothers with older ages at conception. Nausea or vomiting during the first trimester of pregnancy was associated with a reduced risk of testicular cancer (odds ratio of 0.73, 95% confidence interval 0.53-1.00). There was also a borderline reduction in risk in men who had been breastfed for 6 months or more (odds ratio 0.65, 95% confidence interval 0.41-1.04). Men who had low birthweights (< 2500 g) or had been born two or more weeks early had slightly increased risks, as did men whose mothers had used oral contraception in the 12 months before their conception. CONCLUSIONS: These findings support previous reports of increased risks in men born early or with low birthweight, but the direction of the association with maternal age is contrary to some other studies. The suggestion of a protective effect of breastfeeding requires further confirmation.


Subject(s)
Infant, Low Birth Weight , Maternal Age , Testicular Neoplasms/etiology , Adolescent , Adult , Birth Weight , Case-Control Studies , Female , Humans , Infant, Newborn , Male , Medical History Taking , Middle Aged , Nausea , Pregnancy , Pregnancy Complications , Pregnancy Trimester, First , Risk Factors , Vomiting
SELECTION OF CITATIONS
SEARCH DETAIL
...