Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
1.
Target Oncol ; 16(3): 357-367, 2021 05.
Article in English | MEDLINE | ID: mdl-33826036

ABSTRACT

BACKGROUND: Despite standard-of-care androgen-deprivation therapy and an increasing number of treatment options, the mortality rate for prostate cancer remains high. Progress to metastatic castration-resistant prostate cancer (mCRPC) necessitates additional treatments. Abiraterone acetate plus prednisone or prednisolone (AAP) prolongs survival in chemotherapy-naive and docetaxel-experienced patients. OBJECTIVE: To evaluate the real-world safety and efficacy of AAP as first-line and second-line [post-docetaxel only (AAP-PD)] treatment in patients with mCRPC. PATIENTS AND METHODS: The Prostate Cancer Registry (PCR) was a prospective, international, observational study of patients with mCRPC in routine clinical practice. Men aged ≥ 18 years with confirmed mCRPC were included. Baseline characteristics, safety (treatment-emergent adverse events, treatment-emergent severe adverse events), and efficacy [progression-free survival (PFS) and overall survival (OS)] were analyzed. RESULTS: At baseline, patients who received first-line AAP (n = 754) were generally older than patients who received AAP-PD (n = 354); median age was 76 years and 70 years, respectively. However, the rate of visceral metastasis was higher in the AAP-PD cohort than in the AAP cohort (17.7% vs. 9.6%, respectively). Demographics and disease characteristics of patients with baseline cardiovascular disease were similar to those of the overall registry population. Efficacy outcomes were similar for all patients, regardless of the line of AAP therapy. For first-line AAP and AAP-PD, respectively, the median PFS was 8.9 and 5.8 months for all patients and 9.1 and 6.0 months for patients with cardiovascular comorbidities; median OS was 27.1 and 23.4 months for all patients, and 27.4 and 23.1 months for patients with cardiovascular comorbidities. There were no unexpected adverse events in any patient subgroup. CONCLUSIONS: These real-world data complement the findings from randomized controlled trials, indicating that first- and second-line AAP is well tolerated and effective in patients with mCRPC, including those with underlying CV comorbidities. TRIAL REGISTRATION NUMBER: NCT02236637, registered 8 September 2014.


Subject(s)
Abiraterone Acetate/therapeutic use , Prednisone/therapeutic use , Prostatic Neoplasms, Castration-Resistant/drug therapy , Abiraterone Acetate/pharmacology , Aged , Humans , Male , Prednisone/pharmacology , Registries
2.
Target Oncol ; 15(3): 301-315, 2020 06.
Article in English | MEDLINE | ID: mdl-32500294

ABSTRACT

BACKGROUND: Metastatic prostate cancer has a 30% 5-year survival rate despite recent therapeutic advances. There is a need to improve the clinical understanding and treatment of this disease, particularly in the real-world setting and among patients who are under-represented in clinical trials. OBJECTIVE: We aimed to evaluate the characteristics and clinical outcomes of patients who received their first treatment for metastatic castration-resistant prostate cancer (mCRPC) in routine clinical practice, independent of treatment used, including subgroups with baseline cardiac disease, diabetes mellitus, or visceral metastases. PATIENTS AND METHODS: Prospective, noninterventional analysis of patient record data in the multicenter Prostate Cancer Registry (PCR) of men with mCRPC. The data were collected in 16 countries with the aim of recruiting more than 3000 patients between 2013 and 2016. The study end date was 9 July 2018. Data evaluated included baseline characteristics, treatment exposure, and efficacy outcomes [overall survival (OS) and time to progression (TTP)] of patients treated with abiraterone acetate plus prednisone or prednisolone (collectively, "abiraterone"), enzalutamide, or docetaxel. Descriptive outcomes are reported from the overall patient population and subgroups of patients with baseline cardiovascular disease, diabetes mellitus, or visceral metastases. The treatment effects for time to progression were compared for the overall patient population. RESULTS: The study enrollment period lasted 2.5 years, and each patient was followed for a maximum of 3 years. A total of 1874 patients in the PCR had not received previous mCRPC treatment at baseline, although they had received androgen-deprivation therapy. Prevalent co-morbidities included cardiovascular disease in 65.4% and diabetes mellitus in 17.4% of patients. Baseline characteristics suggested that patients with more advanced disease received docetaxel treatment. In the overall patient population, the median time to progression with abiraterone, enzalutamide, and docetaxel as first-line mCRPC therapy was 9.6, 10.3, and 7.6 months, respectively, and median OS was 27.1, 27.1, and 27.9 months, respectively. Outcomes in the subgroups of patients with cardiovascular disease or diabetes mellitus were similar to those of the whole population in the analysis. As expected, patients with visceral metastases had shorter TTP and OS than patients in the overall population. CONCLUSIONS: This analysis shows, for the first time, the effectiveness in parallel of first-line abiraterone, enzalutamide, and docetaxel in mCRPC, including in patients with co-morbidities such as cardiovascular disease or diabetes mellitus or in patients with visceral metastases. These real-world findings from the PCR provide meaningful information to help manage mCRPC, particularly in patients under-represented in clinical studies. TRIAL REGISTRATION: ClinicalTrials.gov identifier NCT02236637; registered September 2014.


Subject(s)
Prostatic Neoplasms, Castration-Resistant/drug therapy , Adult , Aged , Aged, 80 and over , Humans , Male , Middle Aged , Neoplasm Metastasis , Prospective Studies , Prostatic Neoplasms, Castration-Resistant/mortality , Registries , Survival Rate , Treatment Outcome
3.
Prostate ; 70(7): 735-44, 2010 May 15.
Article in English | MEDLINE | ID: mdl-20333727

ABSTRACT

BACKGROUND: Prostate cancer (PC) is generally believed to have a strong inherited component, but the search for susceptibility genes has been hindered by the effects of genetic heterogeneity. The recently developed sumLINK and sumLOD statistics are powerful tools for linkage analysis in the presence of heterogeneity. METHODS: We performed a secondary analysis of 1,233 PC pedigrees from the International Consortium for Prostate Cancer Genetics (ICPCG) using two novel statistics, the sumLINK and sumLOD. For both statistics, dominant and recessive genetic models were considered. False discovery rate (FDR) analysis was conducted to assess the effects of multiple testing. RESULTS: Our analysis identified significant linkage evidence at chromosome 22q12, confirming previous findings by the initial conventional analyses of the same ICPCG data. Twelve other regions were identified with genome-wide suggestive evidence for linkage. Seven regions (1q23, 5q11, 5q35, 6p21, 8q12, 11q13, 20p11-q11) are near loci previously identified in the initial ICPCG pooled data analysis or the subset of aggressive PC pedigrees. Three other regions (1p12, 8p23, 19q13) confirm loci reported by others, and two (2p24, 6q27) are novel susceptibility loci. FDR testing indicates that over 70% of these results are likely true positive findings. Statistical recombinant mapping narrowed regions to an average of 9 cM. CONCLUSIONS: Our results represent genomic regions with the greatest consistency of positive linkage evidence across a very large collection of high-risk PC pedigrees using new statistical tests that deal powerfully with heterogeneity. These regions are excellent candidates for further study to identify PC predisposition genes.


Subject(s)
Genetic Linkage , Pedigree , Prostatic Neoplasms/genetics , Chromosome Mapping , Chromosomes, Human, Pair 22/genetics , Genetic Predisposition to Disease , Genome, Human , Genome-Wide Association Study , Genotype , Humans , Male
4.
Cancer Res ; 69(2): 640-6, 2009 Jan 15.
Article in English | MEDLINE | ID: mdl-19147579

ABSTRACT

Fusion of the 5'-untranslated region of androgen-regulated TMPRSS2 promoter with ETS transcription factor family members is found frequently in prostate cancers, and recent work suggests that the most common TMPRSS2-ERG fusion is associated with an aggressive clinical phenotype compared with fusion-negative prostate cancer. Thus far, analysis of the fusion has been limited to sporadic cases of prostate cancer. In the current study, we explore for an enrichment of TMPRSS2-ERG fusion in familial prostate cancer. TMPRSS2-ERG fusion was identified using a break-apart fluorescence in situ hybridization assay on tissue microarrays. Presence of TMPRSS2-ERG fusion was associated with higher Gleason scores (P = 0.027). Of 75 patients with established history of prostate cancer, we detected the TMPRSS2-ERG fusion in 44 (59%) patients. Almost three quarters (73%) of fusion-positive patients accumulated within 16 specific families whereas only 27% were single fusion-positive cases within one family. Based on reported prevalence rates, we calculated a sibling recurrence risk ratio of up to 18.9. A subset (63%) of families with uniformly TMPRSS2-ERG-positive prostate cancer underwent a genome-wide linkage scan at 500 markers. This revealed several loci located on chromosomes #9, #18, and X that were suggestive of linkage to the TMPRSS2-ERG fusion-positive prostate cancer phenotype with linkage-of-disease scores up to 2.16 and nonparametric linkage scores up to 2.77. This suggests the presence of an inherited susceptibility to developing the TMPRSS2-ERG fusion. Given the association of TMPRSS2-ERG fusion and aggressive prostate cancer, close surveillance of relatives of patients with established fusion-positive prostate cancer or a family history of prostate cancer in general would be warranted.


Subject(s)
Oncogene Proteins, Fusion/genetics , Prostatic Neoplasms/genetics , Aged , Chromosomes, Human, Pair 21 , Cohort Studies , Genetic Linkage , Genetic Predisposition to Disease , Genome, Human , Humans , In Situ Hybridization, Fluorescence , Male , Middle Aged , Neoplasm Staging , Prostatic Neoplasms/pathology
5.
J Assist Reprod Genet ; 26(1): 41-6, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19030983

ABSTRACT

PURPOSE: To investigate the effects of male aging on sperm quality and sperm DNA fragmentation. METHODS: The ejaculates of 320 unselected men attending a fertility clinic and, as a control, 84 normozoospermic men without any history of ART were analyzed according to WHO guidelines. Sperm DNA fragmentation was measured by flow cytometry after staining with propidiumiodide. RESULTS: The patients were divided into four groups: <30 years, 30-35 years, 36-39 years and >or=40 years. Sperm motility decreased with increasing age whereas sperm concentration, morphology, and DNA fragmentation fluctuated throughout the four groups both among patients and among controls. However, we could not detect any significant correlation between male age and conventional semen parameters or sperm DNA fragmentation, respectively, neither in the patients' group nor among the controls. This also applies to a classification of patients and controls into only two age groups with a cut-off point at 35 years. CONCLUSIONS: Our findings suggest that neither the routinely assessed semen parameters nor the amount of spermatozoa with fragmented DNA are affected by male age.


Subject(s)
Aging/genetics , DNA Fragmentation , Infertility, Male/genetics , Infertility, Male/physiopathology , Spermatozoa/physiology , Adult , Aging/pathology , Ambulatory Care Facilities , DNA/physiology , Flow Cytometry , Humans , Infertility, Male/pathology , Male , Middle Aged , Semen/cytology , Sperm Count , Sperm Motility , Spermatozoa/pathology
6.
Cancer Genet Cytogenet ; 182(2): 103-10, 2008 Apr 15.
Article in English | MEDLINE | ID: mdl-18406871

ABSTRACT

The chromosomal region 7q was repeatedly found to be rearranged in prostate carcinoma. It harbors several well described candidate tumor suppressor and oncogenes. We addressed two genes with opposite roles in cancer; CAV1, a putative tumor suppressor gene at 7q31, and EZH2 at 7q36, which is believed to promote tumor progression. Our primary aim was to assess their expression changes in primary tumors, and then to elucidate the underlying mechanism, assuming that genomic alterations of either locus could affect the other gene as well. In 35 prostate tumor samples, compared with adjacent tissues, CAV1 was overall downregulated (P < 10(-06)), whereas EZH2 was significantly overexpressed (P < 10(-06)). The observed dysregulations were coincident in nearly 70% of the cases. Copy number changes occurred in few tumors. Loss of CAV1 DNA was only marginally associated with reduced expression (P = 0.07), however, and genomic amplification of EZH2 could not explain its upregulation. Through bisulfite sequencing of four tumor samples, CpG-hypermethylation was verified as an alternative mechanism for CAV1 silencing, as reported previously. Moreover, it could also be involved in the reactivation of EZH2.


Subject(s)
Caveolin 1/genetics , Chromosomes, Human, Pair 7/genetics , DNA-Binding Proteins/genetics , Gene Expression Regulation, Neoplastic , Prostatic Neoplasms/genetics , Transcription Factors/genetics , CpG Islands , DNA Methylation , DNA Primers , Enhancer of Zeste Homolog 2 Protein , Gene Dosage , Humans , In Situ Hybridization, Fluorescence , Male , Middle Aged , Polycomb Repressive Complex 2 , Prostatic Neoplasms/pathology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction
7.
Hum Mol Genet ; 16(11): 1271-8, 2007 Jun 01.
Article in English | MEDLINE | ID: mdl-17478474

ABSTRACT

Previously, an analysis of 14 extended, high-risk Utah pedigrees localized in the chromosome 22q linkage region to 3.2 Mb at 22q12.3-13.1 (flanked on each side by three recombinants) contained 31 annotated genes. In this large, multi-centered, collaborative study, we performed statistical recombinant mapping in 54 pedigrees selected to be informative for recombinant mapping from nine member groups of the International Consortium for Prostate Cancer Genetics (ICPCG). These 54 pedigrees included the 14 extended pedigrees from Utah and 40 pedigrees from eight other ICPCG member groups. The additional 40 pedigrees were selected from a total pool of 1213 such that each pedigree was required to contain both at least four prostate cancer (PRCA) cases and exhibit evidence for linkage to the chromosome 22q region. The recombinant events in these 40 independent pedigrees confirmed the previously proposed region. Further, when all 54 pedigrees were considered, the three-recombinant consensus region was narrowed down by more than a megabase to 2.2 Mb at chromosome 22q12.3 flanked by D22S281 and D22S683. This narrower region eliminated 20 annotated genes from that previously proposed, leaving only 11 genes. This region at 22q12.3 is the most consistently identified and smallest linkage region for PRCA. This collaborative study by the ICPCG illustrates the value of consortium efforts and the continued utility of linkage analysis using informative pedigrees to localize genes for complex diseases.


Subject(s)
Chromosomes, Human, Pair 22/genetics , Genetic Predisposition to Disease , Prostatic Neoplasms/genetics , Societies, Medical , Humans , International Cooperation , Lod Score , Male
8.
Cancer Epidemiol Biomarkers Prev ; 15(5): 935-8, 2006 May.
Article in English | MEDLINE | ID: mdl-16702373

ABSTRACT

The Nijmegen breakage syndrome 1 (NBS1) gene, which participates in DNA double strand break repair, has been postulated to be a susceptibility factor for a number of cancers, including prostate cancer. Numerous mutations have been identified in NBS1, including the founder mutation 657del5. In this study, a number of analyses were done to determine whether mutations in NBS1 are associated with an increased risk for prostate cancer. The frequency of the 657del5 mutation in both familial prostate cancer cases (1,819 affected men among 909 families) and sporadic prostate cancer cases (1,218 affected men) collected from five centers participating in the International Consortium for Prostate Cancer Genetics were compared with that found in 697 normal controls. Seven individuals were identified to carry the mutation among the 3,037 cases screened: four in the familial group (three from one family and one from another) and three in the sporadic cases. The carrier frequency was 0.22% (2 of 909) for the probands and 0.25% (3 of 1,218) for the sporadic cases of prostate cancer. The 657del5 mutation was not detected in either the 293 unaffected members of the prostate cancer families or in the 697 control samples tested. The entire NBS1 gene was also sequenced in 20 of the youngest affected individuals from the Finnish group of familial cases to identify the presence of possible mutations in this high-risk group. One rare (D95N) and one common (E185Q) missense alteration was identified. More detailed analyses of the E185Q polymorphism, along with a third rare variant (R215W), failed to show an association with prostate cancer. Because the 657del5 mutation was absent from the control population, we are unable to determine if this alteration predisposes to prostate cancer. However, our data does suggest that mutations within NBS1, and in particular, 657del5, do not significantly contribute to the overall prostate cancer burden within our patient samples.


Subject(s)
Cell Cycle Proteins/genetics , Nijmegen Breakage Syndrome/genetics , Nuclear Proteins/genetics , Prostatic Neoplasms/genetics , Aged , Aged, 80 and over , Case-Control Studies , Genetic Predisposition to Disease , Genotype , Humans , Logistic Models , Male , Middle Aged , Mutation , Nijmegen Breakage Syndrome/epidemiology , Prostatic Neoplasms/epidemiology , Risk Factors
9.
Hum Mutat ; 27(1): 98-102, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16287155

ABSTRACT

The MSR1 gene at 8p22 has been suggested as a candidate gene for hereditary prostate cancer because germline variants have been found to be associated with the disease. Aside from a single nonsense mutation (R293X) that was found repeatedly at low frequencies in several samples, little evidence has been gained by follow-up studies to confirm the gene's relevance for prostate cancer. Prompted by reasonable support for a linkage to 8p22, we sought to determine the mutation spectrum of MSR1 in our family sample. Screening of 139 probands (representing 139 prostate cancer families) revealed 15 novel and a total of 20 sequence variants within the 10 coding exons and their intronic proximities. Aside from the known mutation c.877C>T (R293X) present in two of our families, we identified a second nonsense allele (c.251C>G; S84X) and a splice-site mutation (c.818-1G>A) that results in mRNA instability (each in a single pedigree). The novel missense alleles were c.703C>T (H235Y), c.856C>T (P286S), c.905C>T (P302L), c.1193C>G (A398G), and c.1289A>G (K430R). Of the eight variants that affect the encoded protein (splice site, nonsense, and missense), only R293X as well as the polymorphism c.823C>G (P275A) were additionally present at remarkable frequencies in further samples of sporadic prostate cancer and controls. Of note, carriers of R293X were equally frequent in 367 sporadic prostate cancer cases (1.9%) and in 197 controls (2.0%). To our knowledge, our study is the first to demonstrate further loss of function variants of MSR1 apart from R293X. Nevertheless, the low frequencies of deleterious alleles, in addition to an apparently moderate penetrance, does not support MSR1 as a major susceptibility gene in this family sample.


Subject(s)
Germ-Line Mutation/genetics , Prostatic Neoplasms/ethnology , Prostatic Neoplasms/genetics , Scavenger Receptors, Class A/genetics , Aged , Aged, 80 and over , Alleles , DNA Mutational Analysis , Exons/genetics , Gene Expression Regulation, Neoplastic , Germany , Humans , Introns/genetics , Male , Middle Aged , Neoplasm Proteins/genetics , Pedigree , RNA Splice Sites/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Scavenger Receptors, Class A/metabolism
10.
Prostate ; 65(3): 252-9, 2005 Nov 01.
Article in English | MEDLINE | ID: mdl-16015586

ABSTRACT

BACKGROUND: Several linkage studies have provided evidence for a prostate cancer aggressiveness gene on chromosome 7q. This report details the results of the first mutation screen and association study of EZH2 (located at 7q35) as a potential candidate gene for the development of aggressive prostate cancer. METHODS: In 10 families with linkage of chromosome 7q31-33 to aggressive prostate cancer, we sequenced the promoter region and all 20 exons of EZH2. We genotyped 11 variants in 287 prostate cancer probands and 96 controls. Association between the disease and the variants/haplotypes was evaluated taking into account clinical data and disease recurrence. RESULTS: The individual variation sites did not show significant differences in the allele frequencies between cases and controls. In contrast, one haplotype had a higher frequency in controls, and another haplotype was significantly more frequent in cases with low grade tumors (GI/II) and progression free survival (NED). CONCLUSION: We have possibly identified haplotypes which mark alleles that have a beneficial effect on the development of prostate cancer. Moreover, our results suggest that genetic variations of the EZH2 gene are not responsible for the linkage of 7q to aggressive prostate cancer.


Subject(s)
DNA, Neoplasm/genetics , Prostatic Neoplasms/genetics , Proteins/genetics , Adult , Aged , Aged, 80 and over , Alleles , DNA-Binding Proteins , Enhancer of Zeste Homolog 2 Protein , Genetic Predisposition to Disease , Genotype , Haplotypes , Humans , Linkage Disequilibrium/genetics , Male , Middle Aged , Polycomb Repressive Complex 2 , Polymerase Chain Reaction , Polymorphism, Single Nucleotide , Prostatic Neoplasms/pathology , Sequence Analysis, DNA , Transcription Factors
11.
Prostate ; 65(2): 171-7, 2005 Oct 01.
Article in English | MEDLINE | ID: mdl-15948133

ABSTRACT

BACKGROUND: Multiple lines of evidence have implicated the CAV-1 gene in prostate cancer progression. CAV-1 is located within the prostate cancer aggressiveness locus at 7q31-33, and was identified as being overexpressed in prostate tumors. Mutation screening was performed as well as a case-control study to examine if polymorphisms in CAV-1 are associated with prostate cancer aggressiveness in a German population. METHODS: We sequenced the CAV-1 promoter region and its open reading frame in prostate cancer families with linkage to chromosome 7q31-33. Additionally, 105 unrelated familial prostate cancer probands, 190 sporadic cases, and 191 controls were genotyped at four intronic single nucleotide polymorphisms. Resulting haplotypes were tested for association using age at diagnosis, tumor grade, TNM stage, and follow up information to stratify for aggressive disease. RESULTS: No mutation was found in the CAV-1 coding region or in the promoter. One of the 11 observed haplotypes showed an increased frequency in cases with high tumor stage (P = 0.03). CONCLUSIONS: This is the first report providing evidence for CAV-1 being involved in predisposition to aggressive prostate cancer. The association of a potential risk haplotype agrees well with a role of CAV-1 in tumor progression but needs further confirmation.


Subject(s)
Caveolins/genetics , Caveolins/physiology , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Adult , Age of Onset , Aged , Case-Control Studies , Caveolin 1 , DNA Mutational Analysis , Female , Haplotypes , Humans , Male , Middle Aged , Neoplasm Staging , Pedigree , Phenotype , Polymorphism, Single Nucleotide , Prognosis
12.
Anticancer Res ; 25(2B): 1303-7, 2005.
Article in English | MEDLINE | ID: mdl-15865082

ABSTRACT

BACKGROUND: A thymidine to cytosine transition (designated A2 variant) in the promoter region of CYP17 has previously been associated with a familial history of prostate cancer in North American families. The purpose of the present study was to determine whether this correlation could be replicated in a European population. MATERIALS AND METHODS: Case-control comparisons were performed by modelling a dominant (A1/A2 + A2/A2 vs. A1/A1) and a recessive (A2/A2 vs. A1/A2 + A1/A1) effect of the promoter modification. RESULTS: An insignificant overrepresentation of homozygous carriers of the A2 allele (recessive effect) was found in sporadic cases, as compared to controls. However, the A2 variant was not related to familial disease. CONCLUSION: Our results do not suggest a role of CYP17 as a high-risk susceptibility gene for familial prostate cancer, nor as a modifier for the disease risk in the European population.


Subject(s)
Genetic Predisposition to Disease , Polymorphism, Genetic , Prostatic Neoplasms/genetics , Steroid 17-alpha-Hydroxylase/genetics , Aged , Case-Control Studies , Germany , Humans , Male , Middle Aged , Odds Ratio , Promoter Regions, Genetic , Risk Factors
13.
Eur J Hum Genet ; 13(3): 352-60, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15536476

ABSTRACT

Prostate cancer is a complex disease with a substantial genetic contribution involved in the disease risk. Several genomewide linkage studies conducted so far have demonstrated a strong heterogeneity of susceptibility. In order to assess candidate regions that are particularly relevant for the German population, we performed a genomewide linkage search on 139 prostate cancer families. A nonparametric method (Zlr scores), using GENEHUNTERPLUS, was applied at 500 markers (panel P1400, deCODE), with an average spacing of 7.25 cM. In the entire family collection, linkage was most evident at 8p22 (Zlr=2.47, P=0.0068), close to the previously identified susceptibility gene MSR1. Further local maxima with Zlr>2 (P<0.025) were observed at 1q, 5q and 15q. In a subgroup of 47 families, which matched the Johns Hopkins criteria of hereditary prostate cancer, suggestive linkage was found on 1p31 (Zlr=3.37, P=0.00038), a previously not described candidate region. The remaining 92 pedigrees, with no strong disease history, revealed a maximum Zlr=3.15 (P=0.00082) at 8q13, possibly indicating a gene with reduced penetrance or recessive inheritance. Our results suggest pronounced locus heterogeneity of prostate cancer susceptibility in Germany. In the present study population, the MSR1 gene could play a significant role. Other conspicuous loci, like 1p31 and 8q13, need further investigation in order to verify their relevance and to identify candidate genes.


Subject(s)
Genetic Linkage , Genetic Predisposition to Disease , Genome, Human , Prostatic Neoplasms/genetics , Aged , Family Health , Germany , Humans , Male , Middle Aged , Pedigree , Statistics, Nonparametric
14.
Eur J Hum Genet ; 11(1): 17-22, 2003 Jan.
Article in English | MEDLINE | ID: mdl-12529701

ABSTRACT

It has been suggested that chromosome 7q32 contains genes that influence the progression of prostate cancer from latent to invasive disease. In an attempt to confirm this linkage to prostate cancer aggressiveness, 100 German prostate cancer families were genotyped using a panel of eight polymorphic markers on chromosome 7q. We used a multipoint allele sharing method based upon a likelihood ratio test implemented in GENEHUNTERPLUS v1.2 in order to calculate the nonparametric Z(lr) and the associated LOD scores. We applied the aggressiveness of prostate cancer given by the pathological tumour grade of each individual, and the mean age of onset of a family as covariates, and constructed two weighted models. The first (weight(0-1) model) puts weights on families with at least two cases of GIII prostate cancer. The second (weight(0-2) model) also adds weights to families with early and late onset cancer respectively. The unweighted analysis gave no evidence of linkage to chromosome 7q. The Z(lr) scores increased when including the covariates, to 2.60 (P=0.005) using the weight(0-1) and to 3.02 (P=0.001) using the weight(0-2) model for late onset prostate cancer. The associated LOD scores were respectively 1.47 (P=0.009) and 1.98 (P=0.002). The markers that gave most evidence for linkage were exactly in the range of the published prostate cancer aggressiveness region. Our results support a widespread relevance of this locus and suggest that aggressive and late onset prostate cancer is linked to chromosme 7q31-33 in the German population.


Subject(s)
Chromosomes, Human, Pair 7 , Genetic Linkage , Genetic Predisposition to Disease , Prostatic Neoplasms/genetics , Age of Onset , Genetic Heterogeneity , Germany/epidemiology , Humans , Lod Score , Male , Prostatic Neoplasms/epidemiology , Prostatic Neoplasms/pathology
15.
Prostate ; 52(1): 12-9, 2002 Jun 01.
Article in English | MEDLINE | ID: mdl-11992616

ABSTRACT

BACKGROUND: Several prostate cancer (PCa) susceptibility loci have been reported, but attempts to confirm them in independent data sets have produced inconsistent results. It is not yet clear, how much of this variation is due to differences between different populations. HPCX was originally identified in a combined data set of PCa families from the USA and Scandinavia. Considerable differences in the frequency of linked families were observed in this heterogeneous family sample as well as in following studies. METHODS: In order to estimate the significance of HPCX in the German population, DNA samples from 104 PCa families were genotyped at six polymorphic markers spanning a region of approximately 14 cM on Xq27-28, which includes the proposed HPCX candidate locus. RESULTS: In the entire data set, a maximum NPL Z score of 1.20 (P = 0.11) at marker DXS984 was observed. Statistically significant evidence for linkage was obtained in the subset of 63 families with early-onset disease (i.e., < or = 65 years) with a maximum NPL Z score of 2.32 (P = 0.009) at the same location. CONCLUSION: Our results confirm the existence of a prostate cancer susceptibility gene on Xq27-28 also in the German population.


Subject(s)
Genetic Predisposition to Disease , Prostatic Neoplasms/genetics , X Chromosome , Age Factors , Aged , Chromosome Mapping , DNA, Neoplasm/analysis , Genetic Linkage , Genetic Markers , Genotype , Germany , Humans , Lod Score , Male , Middle Aged , Prostatic Neoplasms/diagnosis
16.
J Urol ; 167(2 Pt 1): 718-23, 2002 Feb.
Article in English | MEDLINE | ID: mdl-11792960

ABSTRACT

PURPOSE: Clonality assays can be used to address different questions of neoplasm development. The study of the X chromosome inactivation pattern in female patients provides a useful and most commonly used indirect approach to demonstrate the monoclonal status of a cell population. We used this approach to examine whether recurrent tumors of the bladder supposed to be of monoclonal origin derive from different, independently transformed cells or whether they arise from 1 primary tumor. MATERIALS AND METHODS: We analyzed 45 archival or fresh frozen bladder tumors from 27 female patients. We assessed the X inactivation status of each tumor by a polymerase chain reaction based method after HpaII digestion. RESULTS: Surprisingly 16 of the 45 tumors revealed a polyclonal pattern. The amount of undigested DNA far exceeded what was explained by contamination with normal cells, as determined on histological sections indicating that these tumors were in fact polyclonal. This polyclonal status was further confirmed by a comprehensive series of controls. CONCLUSIONS: Some bladder tumors are polyclonal in origin. Our findings are at variance with earlier observations and possibilities for explanation are proposed.


Subject(s)
Dosage Compensation, Genetic , Neoplasm Recurrence, Local/genetics , Urinary Bladder Neoplasms/genetics , DNA Methylation , DNA, Neoplasm , Female , Humans , Neoplasm Recurrence, Local/pathology , Polymerase Chain Reaction , Polymorphism, Genetic , Receptors, Androgen/genetics , Urinary Bladder Neoplasms/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...