Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 88
Filter
1.
Eur J Cancer ; 42(16): 2802-6, 2006 Nov.
Article in English | MEDLINE | ID: mdl-16963262

ABSTRACT

Several linkage and loss of heterozygosity (LOH) analyses suggest that the region 3p21-p26, which is a chromosomal location of MLH1, could harbour a susceptibility gene for prostate cancer (PRCA). Furthermore, in a recent candidate single nucleotide polymorphism (SNP) analysis the I219V variation of the MLH1 gene was associated with PRCA. Microsatellite instability (MSI) and germ-line MLH1 mutations were originally demonstrated in hereditary non-polyposis colorectal cancer (HNPCC) but MSI and loss of MLH1 function have also been detected in PRCA. To assess the contribution of MLH1 germline mutations to the development of PRCA in Finland different approaches were used. First, the samples from 11 PRCA-colon cancer patients were screened for MLH1, MSH2 and MSH6 protein expression by immunohistochemistry (IHC). IHC revealed one patient with a putative MLH1 aberration and sequencing of this sample revealed five sequence variants including two missense variants P434L and I219V. Second, the samples from Finnish hereditary prostate cancer (HPC) families were used for the screening of MLH1 mutations which produced twelve MLH1 sequence variants including two missense mutations, I219V, as in the PRCA-colon cancer patient, and V647M. P434L and V647 were both novel, rare variants. Carrier frequencies of the I219V mutation were compared between hereditary prostate cancer (HPC) patients, unselected PRCA cases, patients with benign prostate hyperplasia and controls, but no differences between the sample groups were found. P434L was not present in this study population and V647M was a very rare variant found only in one HPC family. According to the present results, MLH1 does not have a major role in PRCA causation in Finland.


Subject(s)
Carrier Proteins/genetics , Germ-Line Mutation/genetics , Mutation, Missense/genetics , Nuclear Proteins/genetics , Prostatic Neoplasms/genetics , Adaptor Proteins, Signal Transducing , Adult , Aged , Aged, 80 and over , Humans , Immunohistochemistry , Male , Middle Aged , MutL Protein Homolog 1 , Pedigree , Registries
2.
J Med Genet ; 43(6): 523-6, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16155190

ABSTRACT

Hereditary leiomyomatosis and renal cell cancer (HLRCC) is a tumour predisposition syndrome caused by heterozygous germline mutations in the fumarate hydratase (FH) gene. The condition is characterised by predisposition to benign leiomyomas of the skin and the uterus, renal cell carcinoma (RCC), and uterine leiomyosarcoma (ULMS). To comprehensively examine the cancer risk and tumour spectrum in Finnish FH mutation positive families, genealogical and cancer data were obtained from 868 individuals. The cohort analysis of the standardised incidence ratios (SIR) was analysed from 256 individuals. FH mutation status was analysed from all available individuals (n = 98). To study tumour spectrum in FH mutation carriers, loss of the wild type allele was analysed from all available tumours (n = 22). The SIR was 6.5 for RCC and 71 for ULMS. The overall cancer risk was statistically significantly increased in the age group of 15-29 years, consistent with features of cancer predisposition families in general. FH germline mutation was found in 55% of studied individuals. Most RCC and ULMS tumours displayed biallelic inactivation of FH, as did breast and bladder cancers. In addition, several benign tumours including atypical uterine leiomyomas, kidney cysts, and adrenal gland adenomas were observed. The present study confirms with calculated risk ratios the association of early onset RCC and ULMS with FH germline mutations in Finns. Some evidence for association of breast and bladder carcinoma with HLRCC was obtained. The data enlighten the organ specific malignant potential of HLRCC.


Subject(s)
Fumarate Hydratase/genetics , Genetic Predisposition to Disease , Germ-Line Mutation , Neoplasms/genetics , Adolescent , Adult , Age Factors , Aged , Carcinoma, Renal Cell/diagnosis , Carcinoma, Renal Cell/genetics , Cohort Studies , Female , Finland , Humans , Kidney Neoplasms/diagnosis , Kidney Neoplasms/genetics , Leiomyomatosis/diagnosis , Leiomyomatosis/genetics , Male , Middle Aged , Neoplasms/diagnosis , Phenotype , Risk Factors
4.
Am J Pathol ; 159(6): 2089-94, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11733359

ABSTRACT

Gain of the long arm of chromosome 8 (8q) is one of the most common gains found in the advanced prostate cancer by comparative genomic hybridization. We have previously identified a putative target gene for the 8q gain, EIF3S3, that encodes a p40 subunit of eukaryotic translation initiation factor 3 (eIF3). Here, we studied the frequency of the EIF3S3 amplification in different stages of prostate cancer and co-amplification of EIF3S3 and oncogene MYC. In addition, prognostic utility of the EIF3S3 copy number alteration was evaluated. The analyses were done with fluorescence in situ hybridization and tissue microarray technology. High-level amplification of EIF3S3 was found in 11 of 125 (9%) of pT1/pT2 tumors, 12 of 44 (27%) of pT3/pT4 tumors, and 8 of 37 (22%) of lymph node metastases as well as in 26 of 78 (33%) and 15 of 30 (50%) of hormone refractory locally recurrent tumors and metastases, respectively. The amplification was associated with high Gleason score (P < 0.001). One of the 79 tumors with EIF3S3 amplification had only two copies of MYC, whereas all tumors with amplification of MYC had also amplification of EIF3S3 indicating common co-amplification of the genes. Gain of EIF3S3 was associated with poor cancer-specific survival in incidentally found prostate carcinomas (P = 0.023). In the analyses of prostatectomy-treated patients, the amplification was not statistically significantly associated with progression-free time. In conclusion, amplification of EIF3S3 gene is common in late-stage prostate cancer suggesting that it may be functionally involved in the progression of the disease.


Subject(s)
Peptide Initiation Factors/genetics , Prostatic Neoplasms/pathology , Aged , Aged, 80 and over , Eukaryotic Initiation Factor-3 , Gene Amplification , Gene Dosage , Humans , In Situ Hybridization, Fluorescence , Male , Middle Aged , Neoplasm Staging , Prostatic Neoplasms/genetics , Prostatic Neoplasms/mortality , Survival Analysis , Survival Rate
5.
Cancer Genet Cytogenet ; 131(1): 74-8, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11734323

ABSTRACT

The genetic mechanisms of prostate cancer recurrence during hormonal therapy are largely unknown. So far, data from conventional karyotype analysis on hormone-refractory prostate carcinomas have not been published, mainly because of the difficulties in obtaining fresh hormone-refractory prostate carcinoma samples and getting metaphases from them. Here, we have studied chromosomal changes in 12 locally recurrent, hormone-refractory prostate carcinomas using karyotyping and CGH that revealed genetic aberrations in all tumors. Loss of the Y chromosome was the most common (89%) finding, and tetraploidy or near-tetraploidy was detected in all tumors. Also non-random translocations were found in 56% of the tumors. The present study indicates that clonal chromosomal aberrations in hormone-refractory prostate carcinomas are more common than in untreated primary tumors, and also, further studies on the frequency and significance of translocations in prostate carcinoma progression during hormonal therapy are warranted.


Subject(s)
Androgens/pharmacology , Chromosome Aberrations , Neoplasm Recurrence, Local/genetics , Prostatic Neoplasms/genetics , Humans , Karyotyping , Male , Neoplasm Recurrence, Local/classification , Neoplasm Recurrence, Local/pathology , Nucleic Acid Hybridization , Prostatic Neoplasms/classification , Prostatic Neoplasms/pathology , Tumor Cells, Cultured
6.
Lab Invest ; 81(12): 1647-51, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11742035

ABSTRACT

Mechanisms of prostate cancer (CaP) recurrence during a combined androgen blockade (CAB) are poorly understood. Previously, the role of androgen receptor (AR) gene mutations underlying the CAB therapy relapse has been raised. To investigate the hypothesis that AR gene aberrations are involved in CAB relapse, 11 locally recurrent CaP samples from patients treated with orchiectomy and bicalutamide were analyzed for copy number changes and DNA sequence alterations of the AR gene by fluorescence in situ hybridization and single-strand conformation polymorphism, respectively. Altogether, base changes were detected in four tumors (36%). Three of them were missense mutations (G166S, W741C, M749I) and two were silent polymorphisms. Interestingly, none of the tumors had AR amplification. These data suggest that different AR variants are developed and selected for during various types of hormonal treatments, and also, that CAB achieved by orchiectomy and bicalutamide does not act as a selective force for AR amplification.


Subject(s)
Androgen Antagonists/therapeutic use , Anilides/therapeutic use , Mutation , Orchiectomy , Prostatic Neoplasms/genetics , Prostatic Neoplasms/therapy , Receptors, Androgen/genetics , Combined Modality Therapy , Humans , Male , Neoplasm Recurrence, Local , Nitriles , Tosyl Compounds
7.
Clin Cancer Res ; 7(11): 3465-71, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11705864

ABSTRACT

In our recent cancer registry-based study, the incidence of gastric carcinoma was increased up to 5-fold in male relatives of early-onset prostate cancer (PCA) patients. This association may reflect the influence of genetic factors predisposing individuals to both tumor types. Germ-line mutations of the CDH1 gene at 16q have recently been associated with familial gastric cancer. Furthermore, two genome-wide linkage studies of PCA recently reported positivity at 16q. We therefore identified families and individual patients with both gastric and PCA and investigated whether the CDH1 gene mutations were involved in cancer predisposition in these cases. Fifteen of the 180 Finnish hereditary PCA families (8.3%) had one or more gastric cancer cases. No truncating or splice site CDH1 mutations were identified by PCR single-strand conformational polymorphism in these families or in eight individual patients who had both prostate and gastric cancer. However, a novel S270A missense mutation in exon 6 of the CDH1 gene was seen in a single family with four prostate and two gastric cancers. A large-scale population-based survey indicated a higher prevalence of S270A among both familial PCA cases (3.3%; n = 120; P = 0.01) and unselected PCA patients (1.5%; n = 472; P = 0.12) as compared with blood donors serving as population controls (0.5%; n = 923). We conclude that individual rare mutations and polymorphisms in the CDH1 gene, such as S270A, may contribute to the onset of PCA and warrant further investigations in other populations. However, the CDH1 gene does not appear to explain the link between prostate and gastric cancer.


Subject(s)
Cadherins/genetics , Germ-Line Mutation , Prostatic Neoplasms/genetics , Adaptor Proteins, Signal Transducing , Aged , Aged, 80 and over , Carrier Proteins , Colorectal Neoplasms, Hereditary Nonpolyposis/genetics , DNA Mutational Analysis , DNA, Neoplasm/chemistry , DNA, Neoplasm/genetics , Family Health , Female , Genetic Testing , Humans , Male , MutL Protein Homolog 1 , Mutation , Mutation, Missense , Neoplasm Proteins/genetics , Nuclear Proteins , Pedigree , Prostatic Neoplasms/diagnosis , Stomach Neoplasms/genetics
8.
Cancer Res ; 61(16): 6038-41, 2001 Aug 15.
Article in English | MEDLINE | ID: mdl-11507049

ABSTRACT

The ELAC2/HPC2 gene at 17p11 is the first candidate gene identified for human prostate cancer (PRCA) based on linkage analysis and positional cloning (S. V. Tavtigian et al. Nat. Genet., 27:172-180, 2001). A truncating mutation was found in one hereditary prostate cancer (HPC) family, whereas two missense variants, Ser217Leu and Ala541Thr, were reported to be associated with increased PRCA risk in the general population. Here, we screened for mutations of the ELAC2/HPC2 gene in 66 Finnish HPC families. Several sequence variants, including a new exonic variant (Glu622Val) were found, but none of the mutations were truncating. We then analyzed the frequency of the three found missense variants in 1365 individuals, including hereditary (n = 107) and unselected (n = 467) PRCA, benign prostatic hyperplasia (n = 223), and population controls (568 healthy male blood donors). Ser217Leu and Ala541Thr variants carried no significantly elevated risk for HPC or PRCA, although the latter variant was associated with benign prostatic hyperplasia. The previously undescribed Glu622Val variant had a 1.0% population prevalence, but a significantly higher frequency in PRCA cases (3.0% odds ratio, 2.94; 95% confidence interval, 1.05-8.23). We conclude that ELAC2/HPC2 truncating mutations are rare in HPC, but that rare variants of the ELAC2/HPC2 require additional study as risk factors for PRCA in the general population.


Subject(s)
Neoplasm Proteins/genetics , Prostatic Neoplasms/genetics , Aged , Aged, 80 and over , DNA/genetics , DNA, Neoplasm/genetics , Exons/genetics , Finland , Genetic Predisposition to Disease/genetics , Genetic Variation , Humans , Male , Middle Aged , Mutation, Missense , Polymorphism, Restriction Fragment Length , Polymorphism, Single-Stranded Conformational
9.
Cancer Causes Control ; 12(3): 223-30, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11405327

ABSTRACT

OBJECTIVE: Five to ten percent of prostate cancers may be caused by inherited genetic defects. In order to explore the nature of inherited cancer risks in the genetically homogeneous Finnish population, we investigated the incidence of prostate cancer and other cancers in first-degree relatives of prostate cancer patients by linking the population-based parish records on relatives with the Finnish Cancer Registry (FCR) data. METHODS: The study population was composed of first-degree relatives of two groups of prostate cancer patients diagnosed in Finland during 1988-1993: (1) all early-onset (<60 years) patients (n = 557) from the entire country, (2) a sample (n = 989) of prostate cancer patients diagnosed at an age of > 60 years. A total of 11,427 first-degree relatives were identified through parish records, and their cancer incidence was determined based on a total of 299,970 person-years. Standardized incidence ratios (SIR) were calculated based on expected cancer rates in the general population. RESULTS: The SIR of prostate cancer was increased in both Cohort 1 (2.5, 95% CI 1.9-3.2) and Cohort 2 (1.7, 95% CI 1.4 2.1). The risk of prostate cancer was high for relatives of patients diagnosed at an early age, and then leveled off for patients in the median age of prostate cancer diagnosis (70-79 years). However, the prostate cancer risk for relatives of patients diagnosed > or = 80 years was again statistically significantly elevated (SIR 1.8, 95% CI 1.3-2.6), suggesting a contribution of genetic factors to prostate cancer also at a late age of onset. Gastric cancer was the only other cancer type with a significantly elevated risk among the relatives. Increased risk of gastric cancer was seen only in male relatives of prostate cancer patients diagnosed at an early age, with the highest risk detected for the male relatives of prostate cancer patients diagnosed at an age of 55 years or less (SIR 5.0, 95% CI 2.8-8.2). CONCLUSIONS: Our population-based study indicates that hereditary factors may play an important role in the development of prostate cancer among the relatives of men diagnosed both at younger and older ages. This finding is relevant in the context of our observations that HPCX (hereditary prostate cancer susceptibility locus on Xq27-28) linkage in Finland is found exclusively among families with late age of onset. The association of gastric cancer with prostate cancer has not been reported previously, and may reflect the effects of a novel predisposition locus, which increases the risk to both of these common tumor types.


Subject(s)
Neoplastic Syndromes, Hereditary/genetics , Prostatic Neoplasms/genetics , Stomach Neoplasms/genetics , Adult , Age Factors , Aged , Cluster Analysis , Cohort Studies , Databases, Factual , Female , Finland/epidemiology , Humans , Incidence , Male , Middle Aged , Neoplastic Syndromes, Hereditary/epidemiology , Odds Ratio , Pedigree , Prostatic Neoplasms/epidemiology , Registries , Risk Assessment , Stomach Neoplasms/epidemiology
10.
Chem Biol Interact ; 135-136: 363-72, 2001 Jun 01.
Article in English | MEDLINE | ID: mdl-11397401

ABSTRACT

Epoxy metabolites of 1,3-butadiene are electrophilic and can bind to nucleophilic sites in DNA forming DNA adducts. In this study, guanine N7 adducts of epoxy butene and guanine N7 adducts of epoxy butanediol were measured in lung tissues of mice inhalation exposed to various concentrations of 1,3-butadiene. 32P-postlabeling of DNA adducts were used to demonstrate that the DNA adducts derived from epoxybutene and epoxybutanediol were formed in a dose dependent manner. More than 98% of all adducts detected were formed from epoxybutanediol. Enantiomeric distribution of the adducts formed in vivo differs from that of in vitro experiments demonstrated before. In the case of epoxybutene most of the adducts were formed to the terminal carbon of the S-epoxybutene enantiomer. Most of the adducts derived from epoxybutanediol were formed from the 2S-3R enantiomer. The data demonstrates that enzymatic processes involved with activation and/or detoxification of the metabolites are enantiospecific and/or DNA repair machinery repairs the damage with stereochemical considerations. These are the crucial factors if interspecies differences in tumor sensitiveness is concerned.


Subject(s)
Butadienes/metabolism , DNA Adducts/metabolism , Lung/metabolism , Administration, Inhalation , Animals , Butadienes/administration & dosage , Butadienes/chemistry , DNA Adducts/chemistry , Epoxy Compounds/chemistry , Epoxy Compounds/metabolism , Glycols/chemistry , Glycols/metabolism , Guanine/chemistry , Mice , Stereoisomerism
11.
Br J Cancer ; 84(10): 1344-7, 2001 May 18.
Article in English | MEDLINE | ID: mdl-11355945

ABSTRACT

Prostatic steroid 5-alpha-reductase gene (SRD5A2) encodes a critical enzyme involved in the conversion of testosterone to dihydrotestosterone. A germline mis-sense substitution (A49T) leads to a variant SRD5A2 protein, which has a 5-fold higher in vitro V(max)than the wild-type protein (Ross et al, 1998; Makridakis et al, 1999). The A49T variant was recently associated with 2.5 to 3.28-fold increased risk of prostate cancer (PC) in African-American and Hispanic men (Makridakis et al, 1999). Also, Jaffe et al (2000) reported an association between A49T and more aggressive disease among Caucasian patients. Here, we report that the prevalence of the A49T variant in 449 Finnish PC patients was 6.0%, not significantly different from 6.3% observed in 223 patients with benign prostatic hyperplasia or 5.8% in 588 population-based controls (odds ratio for PC 1.04, 95% C.I. 0.62-1.76, P = 0.89). There was no association between A49T and the family history of the patients nor with tumour stage or grade. Our results argue against a prominent role of the A49T variant as a genetic risk factor for prostate cancer development and progression in the Finnish population.


Subject(s)
3-Oxo-5-alpha-Steroid 4-Dehydrogenase/genetics , Mutation, Missense , Prostatic Hyperplasia/genetics , Prostatic Neoplasms/genetics , Confidence Intervals , Finland , Gene Frequency , Genetic Linkage , Genetic Variation , Humans , Male , Neoplasm Staging , Odds Ratio , Prostatic Hyperplasia/enzymology , Prostatic Neoplasms/enzymology , Prostatic Neoplasms/pathology
12.
Chest ; 119(5): 1610-2, 2001 May.
Article in English | MEDLINE | ID: mdl-11348979

ABSTRACT

We report on a sister and brother with recurrent spontaneous pneumothorax without underlying connective tissue disease and without other affected relatives. The occurrence of spontaneous pneumothorax in two siblings from a large Finnish family raises the possibility of autosomal recessive inheritance of this disorder in some patients.


Subject(s)
Pneumothorax/genetics , Adult , Female , Genes, Recessive , Humans , Male , Pedigree , Recurrence
13.
Proc Natl Acad Sci U S A ; 98(8): 4575-80, 2001 Apr 10.
Article in English | MEDLINE | ID: mdl-11287641

ABSTRACT

The ANX7 gene is located on human chromosome 10q21, a site long hypothesized to harbor a tumor suppressor gene(s) (TSG) associated with prostate and other cancers. To test whether ANX7 might be a candidate TSG, we examined the ANX7-dependent suppression of human tumor cell growth, stage-specific ANX7 expression in 301 prostate specimens on a prostate tissue microarray, and loss of heterozygosity (LOH) of microsatellite markers at or near the ANX7 locus. Here we report that human tumor cell proliferation and colony formation are markedly reduced when the wild-type ANX7 gene is transfected into two prostate tumor cell lines, LNCaP and DU145. Consistently, analysis of ANX7 protein expression in human prostate tumor microarrays reveals a significantly higher rate of loss of ANX7 expression in metastatic and local recurrences of hormone refractory prostate cancer as compared with primary tumors (P = 0.0001). Using four microsatellite markers at or near the ANX7 locus, and laser capture microdissected tumor cells, 35% of the 20 primary prostate tumors show LOH. The microsatellite marker closest to the ANX7 locus showed the highest rate of LOH, including one homozygous deletion. We conclude that the ANX7 gene exhibits many biological and genetic properties expected of a TSG and may play a role in prostate cancer progression.


Subject(s)
Annexin A7/genetics , Genes, Tumor Suppressor , Prostatic Neoplasms/genetics , Cell Division/genetics , Chromosomes, Human, Pair 10 , Genetic Markers , Humans , Immunohistochemistry , Loss of Heterozygosity , Male , Oligonucleotide Array Sequence Analysis , Polymerase Chain Reaction , Polymorphism, Genetic , Prostatic Neoplasms/pathology
14.
Prostate ; 46(3): 233-9, 2001 Feb 15.
Article in English | MEDLINE | ID: mdl-11170152

ABSTRACT

BACKGROUND: Germline mutations in recessive cancer predisposition genes are uncovered by somatic genetic deletions during tumor development. Analysis of genetic changes in tumor tissues from patients with an inherited predisposition may therefore highlight regions of the genome containing susceptibility or modifier genes. Our aim was to characterize genetic changes in familial prostate cancer METHODS: Twenty-one primary prostate cancers from 19 Finnish prostate cancer families were analyzed for somatic genetic changes by comparative genomic hybridization (CGH). RESULTS: The average number of genetic alterations per tumor was 4.0 +/- 1.9, distributed equally among losses and gains. The most common losses were found at chromosomal regions 13q14-q22 (29%), 8p12-pter (24%), and 6q13-q16 (14%), and the most common gains at 19p (25%), 19q (14%) and 7q (14%). CONCLUSIONS: These results suggest that prostate cancers in genetically predisposed individuals arise for the most part through similar somatic genetic progression pathways as sporadic prostate cancers. This also implies that the biological properties of tumors from the two groups may not be different from one another.


Subject(s)
Carcinoma/genetics , Prostatic Neoplasms/genetics , Aged , Aged, 80 and over , Chromosome Aberrations , Genetic Predisposition to Disease/genetics , Humans , Karyotyping , Male , Middle Aged , Nucleic Acid Hybridization
15.
Cancer Res ; 60(22): 6479-81, 2000 Nov 15.
Article in English | MEDLINE | ID: mdl-11103816

ABSTRACT

Mutations of the androgen receptor (AR) gene have been reported in prostate cancer, usually from tumor tissue specimens from late-stage, androgen-independent cancer. Occasionally, germ-line mutations have been found, but a link between AR mutations and predisposition to human prostate cancer has not been firmly established. Recently, two independent studies reported the same germ-line mutation at codon 726 in exon E (CGC to CTC) in two apparently unrelated Finnish prostate cancer patients. This arginine to leucine substitution was reported to alter the transactivational specificity of the AR protein. In the present study, the R726L mutation was analyzed by allele-specific oligohybridization in DNA specimens from 418 consecutive prostate cancer patients who reported a negative family history (sporadic group) and from 106 patients with a positive family history (hereditary group). The population frequency of the R726L mutation in blood donors was 3 of 900 (0.33%). In contrast, eight (1.91%) mutations (odds ratio = 5.8; P = 0.006) were found in the sporadic group, and two (1.89%) mutations were found in the hereditary group (odds ratio = 5.8; P = 0.09). Suggestive evidence of the segregation of the mutation with prostate cancer was seen in these two families. The present study indicates that the R726L substitution in the AR may confer an up to 6-fold increased risk of prostate cancer and may contribute to cancer development in up to 2% of Finnish prostate cancer patients. These results warrant additional large-scale studies of the significance of rare mutations and polymorphisms in candidate genes along the androgen signaling pathway as risk factors for prostate cancer.


Subject(s)
Germ-Line Mutation , Prostatic Neoplasms/genetics , Receptors, Androgen/genetics , Binding Sites , Female , Hormones/metabolism , Humans , Male , Protein Structure, Tertiary/genetics , Receptors, Androgen/metabolism , Repetitive Sequences, Nucleic Acid
16.
J Urol ; 164(6): 1992-5, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11061898

ABSTRACT

PURPOSE: Amplification of the androgen receptor gene has been found in a third of hormone refractory prostate carcinomas. It is possible that amplification facilitates cell growth ability in low concentrations of androgens remaining in the serum after androgen deprivation therapy. We evaluate whether androgen receptor gene amplification at primary progression is associated with response to second line combined androgen blockade for prostate cancer. MATERIALS AND METHODS: A total of 77 patients with prostate cancer were treated initially with androgen deprivation monotherapy followed by combined androgen blockade after the first progression. After initiation of second line combined androgen blockade patients were followed every 3 months to evaluate treatment responses. Biopsies were taken from the prostate at the first progression under endocrine monotherapy. Androgen receptor gene copy number was determined by fluorescence in situ hybridization. RESULTS: Androgen receptor gene amplification was found in 10 of the 77 cases (13%) at the primary disease progression, and was associated with a favorable response to second line combined androgen blockade. Only 1 of 34 (3%) patients classified as nonresponders had androgen receptor gene amplification, whereas 9 of 41 (21%) classified as having either stable disease or response had amplification (p = 0.016). Patients with androgen receptor gene amplification also had a decrease in prostate specific antigen more often after combined androgen blockade than those with no amplification (p = 0.079). However, androgen receptor gene amplification was not associated with patient survival after the first progression. CONCLUSIONS: Androgen receptor gene amplification detected in tumors progressing during androgen deprivation monotherapy is associated with favorable treatment response to second line combined androgen blockade. This finding suggests that at least some androgen receptor amplified tumors retain a high degree of dependency on residual androgens remaining in serum after monotherapy.


Subject(s)
Androgen Antagonists/therapeutic use , Antineoplastic Agents, Hormonal/therapeutic use , Gene Amplification , Orchiectomy , Prostatic Neoplasms/therapy , Receptors, Androgen/genetics , Adult , Aged , Aged, 80 and over , Biopsy , Combined Modality Therapy , Disease Progression , Humans , Male , Middle Aged , Neoplasm Recurrence, Local/therapy , Pilot Projects , Prospective Studies , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Treatment Failure
17.
Cancer Res ; 60(21): 6111-5, 2000 Nov 01.
Article in English | MEDLINE | ID: mdl-11085535

ABSTRACT

NKX3.1 is a prostate-specific homeobox gene located on chromosome 8p21. In the mouse, Nkx3.1 has growth-suppressive and differentiating effects on prostatic epithelium. Mutations of the coding region of NKX3.1 were not found in human prostate cancer, failing to support the notion that NKX3.1 was a tumor suppressor gene. To study the expression o NKX3.1 protein in human tissues and prostate cancer, we derived a rabbit antiserum against purified recombinant NKX3.1. Among normal human tissues, NKX3.1 expression was seen in testis, in rare pulmonary mucous glands, and in isolated regions of transitional epithelium of the ureter. NKX3.1 was uniformly expressed in nuclei of normal prostate epithelial cells in 61 histological sections from radical prostatectomy specimens. We analyzed 507 samples of neoplastic prostate epithelium, most of which were contained on a tissue microarray that contained samples from different stages of prostatic neoplasia. We observed complete loss of NKX3.1 expression in 5% of benign prostatic hyperplasias, 20% of high-grade prostatic intraepithelial neoplasias, 6% of T1a/b samples, 22% of T3/4 samples, 34% of hormone-refractory prostate cancers, and 78% of metastases. Our data show that NKX3.1 expression is highly, but not exclusively, specific for the prostate. Loss of NKX3.1 expression is strongly associated with hormone-refractory disease and advanced tumor stage in prostate cancer (P < 0.0001).


Subject(s)
Gene Silencing , Homeodomain Proteins/genetics , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Transcription Factors/genetics , Animals , Disease Progression , Epithelium/metabolism , Gene Expression , Gene Expression Regulation, Neoplastic , Genes, Tumor Suppressor , Homeodomain Proteins/biosynthesis , Homeodomain Proteins/immunology , Humans , Immune Sera , Immunohistochemistry , Male , Prostatic Neoplasms/metabolism , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Rabbits , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Recombinant Fusion Proteins/isolation & purification , Testis/metabolism , Transcription Factors/biosynthesis , Transcription Factors/immunology
18.
Oncol Rep ; 7(5): 1141-4, 2000.
Article in English | MEDLINE | ID: mdl-10948353

ABSTRACT

Proliferative and apoptotic activities, as well as p53 protein expression, of ten untreated primary prostate carcinomas that showed extremely poor response to hormonal therapy (primary androgen independent prostate carcinomas) were compared with the stage- and grade-matched primary tumor specimens with favorable response to hormonal therapy (androgen dependent prostate carcinomas). The mean proliferative activity measured by Ki-67 immunohistochemistry was slightly higher in the primary androgen independent prostate carcinomas (8.70+/-5.24) than in the androgen dependent prostate carcinomas (7.09+/-2.68; p=0.27). The mean apoptotic activity by in situ end-labeling technique in the primary androgen independent prostate carcinomas (0.96+/-1.03) was less than half of that in the androgen dependent prostate carcinomas (2.75+/-0.98; p=0.0001). Ten percent of the androgen dependent prostate tumors showed p53 protein expression, whereas 30% of the primary androgen independent prostate tumors were immunopositive for p53 (p=0.30). In summary, we have shown that apoptotic activity in the primary androgen independent prostate carcinomas is significantly lower than in the matched androgen dependent prostate carcinomas while the proliferative activity remains unaffected. These results suggest that primary androgen independent prostate carcinomas may have genetic properties, such as inactivation of the p53 gene, that enable them to escape apoptosis caused by androgen ablation.


Subject(s)
Androgens/physiology , Apoptosis/physiology , Neoplasms, Hormone-Dependent/pathology , Prostatic Neoplasms/pathology , Aged , Aged, 80 and over , Androgen Antagonists/therapeutic use , Antineoplastic Agents, Hormonal/therapeutic use , Cell Division/physiology , Humans , Immunohistochemistry , Male , Middle Aged , Neoplasms, Hormone-Dependent/drug therapy , Prostatic Neoplasms/drug therapy , Survival Analysis , Tumor Suppressor Protein p53/biosynthesis , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/immunology
19.
BJU Int ; 86(3): 240-7, 2000 Aug.
Article in English | MEDLINE | ID: mdl-10930923

ABSTRACT

OBJECTIVES: To determine the survival and investigate the prognostic significance of immunohistochemical variables and clinical factors in patients with hormone-resistant prostate cancer (HRPC) and symptomatic pelvic tumours, in whom preliminary observations indicated that survival exceeded the median 8-10 months of patients with HRPC and painful bone metastases. PATIENTS AND METHODS: Seventy-five patients with HRPC referred for palliative pelvic radiotherapy between 1980 and 1996 were identified. For all patients at least two prostate biopsies had been obtained, one before primary hormone treatment and at least one after clinical progression despite androgen deprivation (HRPC biopsy). Bone scans at the time of referral were assessed. The medical records were reviewed for clinical variables of possible prognostic significance. Histological grade was recorded, and prostate-specific antigen (PSA), androgen receptors (ARs), Ki-67 and p53 determined immunohistochemically. In 18 HRPC specimens the degree of AR amplification was analysed. RESULTS: Positive staining for ARs was high in the HRPC biopsies, although there was no association with AR amplification. Ki-67 positivity increased after the development of HRPC. The median (range) survival was 14 (1-141) months; age < 65 years was associated with increased survival. In a multivariate analysis the following variables remained independent prognostic factors for survival from the time of the HRPC biopsy: bone metastases (0-10 vs > 10 lesions, P < 0.001), low Ki-67 score (0 vs 1-3, P = 0.006) and low p53 positivity score (0 vs 1-3, P = 0.014) in the HRPC biopsy. CONCLUSIONS: The median survival of patients with HRPC and pelvic tumours requiring palliation seems to exceed that of patients with HRPC and dominating painful bone metastases by at least 4-6 months. Simple clinical (bone metastases) and immunohistochemical variables (Ki-67, p53) enable patients with particularly long survival times to be identified, and in whom palliative treatment needs to be improved.


Subject(s)
Prostatic Neoplasms/mortality , Aged , Aged, 80 and over , Antineoplastic Agents, Hormonal/therapeutic use , Biopsy/methods , Bone Neoplasms/secondary , Drug Resistance, Neoplasm , Humans , Immunohistochemistry , In Situ Hybridization, Fluorescence , Ki-67 Antigen/analysis , Male , Middle Aged , Neoplasm Proteins/analysis , Palliative Care/methods , Prognosis , Prostate/pathology , Prostatic Neoplasms/pathology , Prostatic Neoplasms/therapy , Survival Analysis
20.
Proc Natl Acad Sci U S A ; 97(17): 9603-8, 2000 Aug 15.
Article in English | MEDLINE | ID: mdl-10944226

ABSTRACT

A significant proportion of familial breast cancers cannot be explained by mutations in the BRCA1 or BRCA2 genes. We applied a strategy to identify predisposition loci for breast cancer by using mathematical models to identify early somatic genetic deletions in tumor tissues followed by targeted linkage analysis. Comparative genomic hybridization was used to study 61 breast tumors from 37 breast cancer families with no identified BRCA1 or BRCA2 mutations. Branching and phylogenetic tree models predicted that loss of 13q was one of the earliest genetic events in hereditary cancers. In a Swedish family with five breast cancer cases, all analyzed tumors showed distinct 13q deletions, with the minimal region of loss at 13q21-q22. Genotyping revealed segregation of a shared 13q21 germ-line haplotype in the family. Targeted linkage analysis was carried out in a set of 77 Finnish, Icelandic, and Swedish breast cancer families with no detected BRCA1 and BRCA2 mutations. A maximum parametric two-point logarithm of odds score of 2.76 was obtained for a marker at 13q21 (D13S1308, theta = 0.10). The multipoint logarithm of odds score under heterogeneity was 3.46. The results were further evaluated by simulation to assess the probability of obtaining significant evidence in favor of linkage by chance as well as to take into account the possible influence of the BRCA2 locus, located at a recombination fraction of 0.25 from the new locus. The simulation substantiated the evidence of linkage at D13S1308 (P < 0.0017). The results warrant studies of this putative breast cancer predisposition locus in other populations.


Subject(s)
Breast Neoplasms/genetics , Chromosome Deletion , Chromosomes, Human, Pair 13/genetics , Genetic Predisposition to Disease/genetics , Aged , BRCA2 Protein , Breast Neoplasms/pathology , Chromosome Mapping , Disease Progression , Female , Genes, BRCA1/genetics , Genome, Human , Genotype , Germ-Line Mutation/genetics , Haplotypes/genetics , Humans , Hybrid Cells , Lod Score , Male , Middle Aged , Models, Genetic , Neoplasm Proteins/genetics , Nucleic Acid Hybridization , Pedigree , Transcription Factors/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...