Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 134
Filter
1.
Am J Epidemiol ; 165(8): 874-81, 2007 Apr 15.
Article in English | MEDLINE | ID: mdl-17244633

ABSTRACT

Volunteers for prevention or screening trials are generally healthier and have lower mortality than the general population. The Prostate, Lung, Colorectal, and Ovarian Cancer Screening Trial (PLCO) is an ongoing, multicenter, randomized trial that randomized 155,000 men and women aged 55-74 years to a screening or control arm between 1993 and 2001. The authors compared demographics, mortality rates, and cancer incidence and survival rates of PLCO subjects during the early phase of the trial with those of the US population. Incidence and mortality from PLCO cancers (prostate, lung, colorectal, and ovarian) were excluded because they are the subject of the ongoing trial. Standardized mortality ratios for all-cause mortality were 46 for men, 38 for women, and 43 overall (100 = standard). Cause-specific standardized mortality ratios were 56 for cancer, 37 for cardiovascular disease, and 34 for both respiratory and digestive diseases. Standardized mortality ratios for all-cause mortality increased with time on study from 31 at year 1 to 48 at year 7. Adjusting the PLCO population to a standardized demographic distribution would increase the standardized mortality ratio only modestly to 54 for women and 55 for men. Standardized incidence ratios for all cancer were 84 in women and 73 in men, with a large range of standardized incidence ratios observed for specific cancers.


Subject(s)
Health Status , Mass Screening , Neoplasms/epidemiology , Voluntary Programs , Aged , Colorectal Neoplasms/epidemiology , Female , Humans , Lung Neoplasms/epidemiology , Male , Middle Aged , Ovarian Neoplasms/epidemiology , Prostatic Neoplasms/epidemiology , United States/epidemiology
2.
Cell Death Differ ; 10(2): 240-8, 2003 Feb.
Article in English | MEDLINE | ID: mdl-12700652

ABSTRACT

The sphingomyelin metabolites ceramide and sphingosine are mediators of cell death induced by gamma-irradiation. We studied the production of ceramide and the effects of exogenous ceramide on apoptosis in LNCaP prostate cancer cells that are highly resistant to gamma-irradiation-induced cell death. LNCaP cells can be sensitized to gamma-irradiation by tumor necrosis factor alpha (TNF-alpha) and, to a lesser degree, by the agonistic FAS antibody CH-11. TNF-alpha activated intrinsic and extrinsic apoptosis pathways and increased ceramide and sphingosine levels in irradiated LNCaP cells. CH-11 activated only the extrinsic apoptosis pathways and had a negligible effect on ceramide and sphingosine levels in irradiated LNCaP cells. Exogenous ceramide and bacterial sphingomyelinase sensitized LNCaP cells to radiation-induced apoptosis and had a synergistic effect on cell death after irradiation with TNF-alpha, but not with CH-11. Cell death effects after exposure to ceramide and irradiation were blocked by the serine protease inhibitor TLCK (Na-p-tosyl-L-lysine-chloromethylketone), but not by the caspase inhibitor z-VAD (2-val-Ala-Asp(oMe)-CH(2)F). During LNCaP cell apoptosis induced by exogenous ceramide, we observed activation of caspase-9, but not caspases-8, -3, or -7. The effect of ceramide occurred largely via the intrinsic mitochondrial apoptosis pathway and enhanced TNF-alpha, but not CH-11 effects on irradiated cells. The data show that ceramide enhanced activation of the intrinsic apoptotic pathway and enhanced cell death induced by TNF-alpha with or without gamma-irradiation. TNF-alpha and gamma-irradiation elevated levels of endogenous ceramide and activated the intrinsic cell death pathway.


Subject(s)
Apoptosis/radiation effects , Ceramides/metabolism , Prostatic Neoplasms/pathology , Adenocarcinoma/pathology , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Apoptosis/physiology , Caspases/physiology , Cell Line, Tumor , Ceramides/pharmacology , Drug Synergism , Enzyme Activation , Humans , Male , Serine Proteinase Inhibitors/pharmacology , Sphingomyelin Phosphodiesterase/pharmacology , Sphingosine/metabolism , Tosyllysine Chloromethyl Ketone/pharmacology , Tumor Necrosis Factor-alpha/pharmacology
3.
Cell Death Differ ; 9(9): 972-80, 2002 Sep.
Article in English | MEDLINE | ID: mdl-12181748

ABSTRACT

LNCaP prostate cancer cells are resistant to induction of apoptosis by gamma-irradiation and partially sensitive to TNF-alpha or FAS antibody, irradiation sensitizes cells to apoptosis induced by FAS antibody or TNF-alpha. LNCaP cell clones stably expressing IkappaBalpha super repressor were resistant to apoptosis induced by death ligands in the presence or absence of irradiation. IkappaBalpha super repressor expression also increased clonogenic survival after exposure to TNF-alpha+irradiation, but had no effect on survival after irradiation alone. IkappaBalpha super repressor expression blocked the increase of whole cell and cell surface FAS expression induced by TNF-alpha, but did not effect induction of FAS expression and cell surface FAS expression that resulted from irradiation. In cells expressing IkappaBalpha super repressor there was diminished activation of caspases-8 and -7 and diminished production of proscaspases-8 and -7, usually required for death induction in LNCaP cells. Peptide inhibitors of caspase activation complemented the IkappaBalpha super repressor inhibition of apoptosis, but peptide inhibitors of serine proteases had no effect on LNCaP cells expressing IkappaBalpha super repressor. Moreover, cleavage of a serine protease substrate was induced by treatment of LNCaP cells with TNF-alpha and irradiation. The data suggest that in LNCaP cells NF-kappaB mediates a proapoptotic pathway that leads to activation of proapoptotic serine proteases.


Subject(s)
Apoptosis/physiology , Carcinoma/enzymology , NF-kappa B/metabolism , Prostatic Neoplasms/enzymology , Serine Endopeptidases/metabolism , Tumor Cells, Cultured/enzymology , Apoptosis/drug effects , Apoptosis/radiation effects , Carcinoma/drug therapy , Carcinoma/radiotherapy , Caspases/drug effects , Caspases/metabolism , Caspases/radiation effects , Drug Resistance, Neoplasm/genetics , Enzyme Inhibitors/pharmacology , Gamma Rays , Gene Expression Regulation/drug effects , Gene Expression Regulation/physiology , Gene Expression Regulation/radiation effects , Humans , I-kappa B Proteins/antagonists & inhibitors , I-kappa B Proteins/metabolism , Male , Mutation/drug effects , Mutation/genetics , Mutation/radiation effects , NF-KappaB Inhibitor alpha , NF-kappa B/drug effects , NF-kappa B/radiation effects , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/radiotherapy , Radiation Tolerance/drug effects , Radiation Tolerance/genetics , Repressor Proteins/genetics , Repressor Proteins/metabolism , Tumor Cells, Cultured/drug effects , Tumor Cells, Cultured/radiation effects , Tumor Necrosis Factor-alpha/metabolism , Tumor Necrosis Factor-alpha/pharmacology , fas Receptor/genetics , fas Receptor/metabolism
4.
Urology ; 58(4): 561-6, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11597539

ABSTRACT

OBJECTIVES: To characterize the role of demographic and clinical parameters in the measurements of prostate-specific antigen (PSA), free PSA (fPSA), and percent free PSA (%fPSA). METHODS: This was a cohort study of volunteers to a randomized screening trial. A central laboratory determined PSA and fPSA for the Prostate, Lung, Colorectal and Ovarian (PLCO) Cancer Screening Trial. A baseline evaluation of free and total PSA was done for 7183 white, black, Asian, Hispanic, and other male volunteers, aged 55 to 74 years. Comparisons were made across racial and ethnic groups and across a set of clinical parameters from a baseline questionnaire. RESULTS: The median levels of serum PSA were less than 2.1 ng/mL in each age-race grouping of the study participants. The levels of free and total PSA were higher in black (n = 868, 12%) participants than in white (n = 4995, 70%) and Asian (n = 849, 11.8%) participants. Individuals who identified themselves as ethnically Hispanic (n = 339, 4.7%) had median PSA levels higher than whites who were not Hispanic. The free and total PSA levels increased with age, particularly among men 70 to 74 years old. However, the %fPSA levels showed less variation among the four racial groups or by age. The free and total PSA levels were higher among those who had a history of benign prostatic disease. CONCLUSIONS: Demographic (age and race/ethnicity) and clinical (history of benign prostatic disease) variables had a moderate effect on the measures of PSA and fPSA and very little effect on %fPSA.


Subject(s)
Mass Screening/standards , Prostate-Specific Antigen/blood , Prostate-Specific Antigen/genetics , Aged , Asian People/genetics , Black People/genetics , Cohort Studies , Hispanic or Latino/genetics , Hispanic or Latino/statistics & numerical data , Humans , Male , Middle Aged , Prostate-Specific Antigen/standards , Sensitivity and Specificity , White People/genetics
5.
Cancer Res ; 61(14): 5611-8, 2001 Jul 15.
Article in English | MEDLINE | ID: mdl-11454715

ABSTRACT

Androgen plays a critical role in the promotion and growth of prostate cancer. Androgen ablation has an expanding role in prostate cancer treatment and is now used to improve the efficacy of radiation therapy in addition to its role in treatment of metastatic disease. Here we show that androgen interferes with induction of prostate cancer cell death induced by a variety of stimuli. The effect of androgen on cell death occurs predominantly by interference with caspase activation and the inhibition of caspase cleavage in both the extrinsic and intrinsic cell death pathways. Androgen inhibited apoptosis induced by both tumor necrosis factor alpha (TNF-alpha) and by Fas activation with or without concomitant irradiation. An antiapoptotic effect was seen in the presence of R1881, dihydrotestosterone, and also 17beta-estradiol within 24 h of death induction. Sustained inhibition of apoptosis at 72 h was seen only with R1881, dihydrotestosterone, cyproterone acetate, and hydroxyflutamide. Androgen treatment inhibited activation of caspases-8, -7, and -9 by TNF-alpha +/- irradiation. Androgen attenuated BAX expression and blocked appearance of the proapoptotic p18 fragment of BAX. Androgen also abrogated BID cleavage induced by TNF-alpha + irradiation that contributed to a decrease in cytochrome c egress from mitochondria induced by TNF-alpha +/- irradiation. There was also decreased mitochondrial depolarization in response to TNF-alpha + irradiation. Production of the proapoptotic lipid metabolite ceramide was not affected by androgen, but androgen acted downstream from ceramide generation because R1881 blocked cell-death induction by bacterial sphingomyelinase. Inhibition of phosphoinositol-3-kinase activity by wortmannin induced apoptosis that was also blocked by androgen, but there was no effect on protein levels or phosphorylation of AKT, indicating that R1881 did not interact with survival signaling of phosphoinositol-3-kinase. Lastly, androgen inhibited activation of nuclear factor-kappaB during death induction, but the effect of androgen on cell death was not mediated by interference with the nuclear factor-kappaB pathway. The data suggest that androgen induced blockade of caspase activation in both intrinsic and extrinsic cell death pathways and thereby was able to protect prostate cancer cells from apoptosis induced by diverse stimuli.


Subject(s)
Androgens/pharmacology , Apoptosis/drug effects , Prostatic Neoplasms/pathology , Caspases/metabolism , Cell Death/drug effects , Cell Death/radiation effects , Ceramides/metabolism , Enzyme Activation/drug effects , Humans , Male , Metribolone/pharmacology , NF-kappa B/drug effects , NF-kappa B/metabolism , Neoplasms, Hormone-Dependent/drug therapy , Neoplasms, Hormone-Dependent/pathology , Phosphatidylinositol 3-Kinases/metabolism , Prostatic Neoplasms/drug therapy , Signal Transduction/drug effects , Testosterone Congeners/pharmacology , Tumor Cells, Cultured/drug effects , Tumor Cells, Cultured/metabolism , Tumor Cells, Cultured/radiation effects , Tumor Necrosis Factor-alpha/pharmacology
6.
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
7.
Cancer Res ; 60(17): 4709-13, 2000 Sep 01.
Article in English | MEDLINE | ID: mdl-10987273

ABSTRACT

beta-Catenin is a multifunctional molecule with important roles in intercellular adhesion and signal transduction. We reported previously that beta-catenin is mutated in human prostate cancer. In this study, we investigated the role of beta-catenin mutations on androgen receptor (AR) signaling. beta-Catenin significantly enhanced androgen-stimulated transcriptional activation by the AR. beta-Catenin also increased AR transcriptional activation by androstenedione and estradiol and diminished the antagonism of bicalutamide. Coimmunoprecipitation of beta-catenin with AR from LNCaP prostate cancer cells showed that the two molecules are present in the same complex. The amount of beta-catenin in complex with AR was increased by androgen. These findings implicate beta-catenin in the regulation of AR function and support a role for beta-catenin mutations in the pathogenesis of prostate cancer.


Subject(s)
Cytoskeletal Proteins/physiology , Receptors, Androgen/physiology , Trans-Activators , Transcriptional Activation/physiology , Androgen Antagonists/pharmacology , Androgens/physiology , Cytoskeletal Proteins/genetics , Cytoskeletal Proteins/metabolism , Humans , Ligands , Male , Mutation , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Receptors, Androgen/genetics , Receptors, Androgen/metabolism , Substrate Specificity , Tumor Cells, Cultured , beta Catenin
8.
Cancer Res ; 60(16): 4468-74, 2000 Aug 15.
Article in English | MEDLINE | ID: mdl-10969794

ABSTRACT

Ceramide has been implicated as an important component of radiation-induced apoptosis of human prostate cancer cells. We examined the role of the sphingolipid metabolites--ceramide, sphingosine, and sphingosine-1-phosphate--in susceptibility to radiation-induced apoptosis in prostate cancer cell lines with different sensitivities to gamma-irradiation. Exposure of radiation-sensitive TSU-Pr1 cells to 8-Gy irradiation led to a sustained increase in ceramide, beginning after 12 h of treatment and increasing to 2.5- to 3-fold within 48 h. Moreover, irradiation of TSU-Pr1 cells also produced a marked and rapid 50% decrease in the activity of sphingosine kinase, the enzyme that phosphorylates sphingosine to form sphingosine-1-phosphate. In contrast, the radiation-insensitive cell line, LNCaP, had sustained sphingosine kinase activity and did not produce elevated ceramide levels on 8-Gy irradiation. Although LNCaP cells are highly resistant to gamma-irradiation-induced apoptosis, they are sensitive to the death-inducing effects of tumor necrosis factor alpha, which also increases ceramide levels in these cells (K. Kimura et al., Cancer Res., 59: 1606-1614, 1999). Moreover, we found that although irradiation alone did not increase sphingosine levels in LNCaP cells, tumor necrosis factor alpha plus irradiation induced significantly higher sphingosine levels and markedly reduced intracellular levels of sphingosine-1-phosphate. The elevation of sphingosine levels either by exogenous sphingosine or by treatment with the sphingosine kinase inhibitor N,N-dimethylsphingosine induced apoptosis and also sensitized LNCaP cells to gamma-irradiation-induced apoptosis. Our data suggest that the relative levels of sphingolipid metabolites may play a role in determining the radiosensitivity of prostate cancer cells, and that the enhancement of ceramide and sphingosine generation could be of therapeutic value.


Subject(s)
Apoptosis/radiation effects , Prostatic Neoplasms/pathology , Radiation Tolerance/physiology , Sphingosine/analogs & derivatives , Sphingosine/physiology , Apoptosis/drug effects , Apoptosis/physiology , Caspases/metabolism , Ceramides/metabolism , Ceramides/physiology , Enzyme Activation , Enzyme Inhibitors/pharmacology , Humans , Male , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Phosphotransferases (Alcohol Group Acceptor)/physiology , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/radiotherapy , Radiation Tolerance/drug effects , Radiation-Sensitizing Agents/pharmacology , Sphingomyelin Phosphodiesterase/pharmacology , Sphingosine/metabolism , Sphingosine/pharmacology , Tumor Cells, Cultured/radiation effects
9.
Nucleic Acids Res ; 28(12): 2389-95, 2000 Jun 15.
Article in English | MEDLINE | ID: mdl-10871372

ABSTRACT

NKX3.1 is a member of the NK class of homeodomain proteins and is most closely related to Drosophila NK-3. NKX3.1 has predominantly prostate-specific expression in the adult human. Previous studies suggested that NKX3.1 exerts a growth-suppressive effect on prostatic epithelial cells and controls differentiated glandular functions. Using a binding site selection assay with recombinant NKX3.1 protein we identified a TAAGTA consensus binding sequence that has not been reported for any other NK class homeoprotein. By electromobility shift assay we demonstrated that NKX3.1 preferentially binds the TAAGTA sequence rather than the binding site for Nkx2.1 (CAAGTG) or Msx1 (TAATTG). Using mutated binding sites in competitive gel shift assays, we analyzed the nucleotides in the TAAGTA consensus sequence that are important for NKX3.1 binding. The consensus binding site of a naturally occurring polymorphic NKX3.1 protein with arginine replaced by cysteine at position 52 was identical to the wild-type binding sequence. The binding affinities of wild-type and polymorphic NKX3.1 for the TAAGTA consensus site were very similar, with values of 20 and 22 nM, respectively. Wild-type and polymorphic NKX3.1 specifically repressed transcription of luciferase from a reporter vector with three copies of the NKX3.1-binding site upstream from a thymidine kinase promoter. The data show that among NK family proteins NKX3.1 binds a novel DNA sequence and can behave as an in vitro transcriptional repressor.


Subject(s)
DNA/chemistry , DNA/metabolism , Homeodomain Proteins/metabolism , Oligodeoxyribonucleotides/chemistry , Spermatozoa/metabolism , Transcription Factors/metabolism , Base Sequence , Binding Sites , Consensus Sequence , Genes, Tumor Suppressor , Humans , Kinetics , Male , Oligodeoxyribonucleotides/metabolism , Recombinant Proteins/metabolism
10.
J Biol Chem ; 275(12): 8610-7, 2000 Mar 24.
Article in English | MEDLINE | ID: mdl-10722700

ABSTRACT

Activation of either tumor necrosis factor receptor 1 or Fas induces a low level of programmed cell death in LNCaP human prostate cancer cells. We have shown that LNCaP cells are entirely resistant to gamma-radiation-induced apoptosis, but can be sensitized to irradiation by TNF-alpha. Fas activation also sensitized LNCaP cells to irradiation, causing nearly 40% cell death 72 h after irradiation. Caspase-8 was cleaved and activated after exposure to tumor necrosis factor (TNF)-alpha. However, after exposure to anti-Fas antibody caspase-8 cleavage occurred only between the 26-kDa N-terminal prodomain and the 28-kDa C-terminal region that contains the protease components. Although anti-Fas antibody plus irradiation induced apoptosis that could be blocked by the pancaspase inhibitor zVAD, there was no measurable caspase-8 activity after exposure to anti-Fas antibody. The effector caspases-6 and -7, and to a lesser extent caspase-3, were activated by TNF-alpha, but not by anti-Fas antibody. Anti-Fas antibody, like TNF-alpha also activated serine proteases that contributed to cell death. Exposure of LNCaP cells simultaneously to TNF-alpha and anti-Fas antibody CH-11 resulted in marked enhancement of apoptosis that occurred very rapidly and was still further augmented by irradiation. Rapid apoptosis that ensued from combined treatment with TNF-alpha, anti-Fas antibody, and irradiation was completely blocked either by zVAD or expression of dominant negative Fas-associated death domain. Our data shows that there are qualitative differences in caspase activation resulting from either TNF receptor 1 or Fas. Simultaneous activation of these receptors was synergistic and caused rapid epithelial cell apoptosis mediated by the caspase cascade.


Subject(s)
Adaptor Proteins, Signal Transducing , Apoptosis/physiology , Gamma Rays/adverse effects , Prostatic Neoplasms/metabolism , Tumor Necrosis Factor-alpha/pharmacology , fas Receptor/pharmacology , Amino Acid Chloromethyl Ketones/pharmacology , Antigens, CD/metabolism , Carrier Proteins/metabolism , Caspase 8 , Caspase 9 , Caspases/metabolism , Drug Synergism , Enzyme Activation , Fas-Associated Death Domain Protein , Humans , Ligands , Male , Radiation Tolerance , Receptors, Tumor Necrosis Factor/metabolism , Receptors, Tumor Necrosis Factor, Type I , Signal Transduction , Tosyllysine Chloromethyl Ketone/pharmacology , Tumor Cells, Cultured , fas Receptor/immunology
11.
Control Clin Trials ; 21(6 Suppl): 273S-309S, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11189684

ABSTRACT

The objectives of the Prostate, Lung, Colorectal and Ovarian Cancer Screening Trial are to determine in screenees ages 55-74 at entry whether screening with flexible sigmoidoscopy (60-cm sigmoidoscope) can reduce mortality from colorectal cancer, whether screening with chest X-ray can reduce mortality from lung cancer, whether screening men with digital rectal examination (DRE) plus serum prostate-specific antigen (PSA) can reduce mortality from prostate cancer, and whether screening women with CA125 and transvaginal ultrasound (TVU) can reduce mortality from ovarian cancer. Secondary objectives are to assess screening variables other than mortality for each of the interventions including sensitivity, specificity, and positive predictive value; to assess incidence, stage, and survival of cancer cases; and to investigate biologic and/or prognostic characterizations of tumor tissue and biochemical products as intermediate endpoints. The design is a multicenter, two-armed, randomized trial with 37,000 females and 37,000 males in each of the two arms. In the intervention arm, the PSA and CA125 tests are performed at entry, then annually for 5 years. The DRE, TVU, and chest X-ray exams are performed at entry and then annually for 3 years. Sigmoidoscopy is performed at entry and then at the 5-year point. Participants in the control arm follow their usual medical care practices. Participants will be followed for at least 13 years from randomization to ascertain all cancers of the prostate, lung, colorectum, and ovary, as well as deaths from all causes. A pilot phase was undertaken to assess the randomization, screening, and data collection procedures of the trial and to estimate design parameters such as compliance and contamination levels. This paper describes eligibility, consent, and other design features of the trial, randomization and screening procedures, and an outline of the follow-up procedures. Sample-size calculations are reported, and a data analysis plan is presented.


Subject(s)
Colorectal Neoplasms/diagnosis , Lung Neoplasms/diagnosis , Mass Screening , Ovarian Neoplasms/diagnosis , Prostatic Neoplasms/diagnosis , Randomized Controlled Trials as Topic , Research Design , Colorectal Neoplasms/prevention & control , Female , Humans , Lung Neoplasms/prevention & control , Male , Multicenter Studies as Topic , Ovarian Neoplasms/prevention & control , Prostatic Neoplasms/prevention & control
12.
Clin Cancer Res ; 5(9): 2596-604, 1999 Sep.
Article in English | MEDLINE | ID: mdl-10499638

ABSTRACT

7-Hydroxystaurosporine (UCN-01), a protein kinase inhibitor in clinical development, demonstrates potent antineoplastic activity. To determine whether specific genetic abnormalities would modulate the response to UCN-01, a model of human non-small cell lung carcinoma (NSCLC) cell lines with differential abnormalities of p16CDKN2, RB, and p53 was used for these studies. Cell growth was measured by the 3-[4,5-dimethylthiazol-2-yl]-2,5-diphenyltetrazolium bromide assay, and cell cycling was studied using flow cytometric analysis of DNA content. Changes in protein levels and phosphorylation were assessed by Western blotting. In cell lines expressing wild-type RB (A549 and Calul), UCN-01 treatment resulted in dose-dependent growth inhibition, arrest of cells in G1, and a reduction of cells in S phase. p16CDKN2-null cells showed similar growth inhibition to normal fetal lung fibroblasts. UCN-01-induced growth arrest was accompanied by induction of p21CDKN1 and a shift of Rb to the hypophosphorylated state in both p53 wild-type and mutant cell lines. In contrast, UCN-01 treatment of the RB-null cell line H596 resulted in less growth inhibition. To test the role of RB in response to UCN-01, effects of treatment were examined in two human isogenic models of RB expression: the bladder cancer cell line 5637 (RB-null) and the prostate cancer cell line DU-145 (RB-mutant). In the Rb-expressing 5637 subline (RB5), UCN-01 treatment resulted in Rb hypophosphorylation and an accumulation in G1 in contrast to the parent line. Similarly, the wild-type Rb-expressing DU-145 sublines (DU1.1 and B5) showed increased G1 arrest compared with the parent cells. We conclude that UCN-01-induced G1 arrest can occur in cells null for p53 and p16CDKN2, and that RB status influences the ability of UCN-01 to induce a G1 arrest. These data suggest that the molecular profile of cell cycle regulating genes in individual tumors may predict responsiveness and provide insight into optimal therapeutic application of this new antineoplastic agent.


Subject(s)
Alkaloids/pharmacology , Antineoplastic Agents/pharmacology , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/genetics , Genes, Retinoblastoma/drug effects , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Carcinoma, Non-Small-Cell Lung/metabolism , Cyclin-Dependent Kinase Inhibitor p16/biosynthesis , Cyclin-Dependent Kinase Inhibitor p16/genetics , Enzyme Inhibitors/pharmacology , G1 Phase/drug effects , Genes, p53/drug effects , Growth Inhibitors/pharmacology , Humans , Lung Neoplasms/metabolism , Phosphorylation , Retinoblastoma Protein/biosynthesis , Retinoblastoma Protein/genetics , Staurosporine/analogs & derivatives , Tumor Cells, Cultured , Tumor Suppressor Protein p53/biosynthesis , Tumor Suppressor Protein p53/genetics
13.
Cell Death Differ ; 6(5): 394-401, 1999 May.
Article in English | MEDLINE | ID: mdl-10381629

ABSTRACT

Cells differ in the time required to execute cell death after receipt of a death signal. One reason may be the requirement for de novo synthesis of components of the death pathway. TSU-Pr1 prostate cancer cells treated with okadaic acid demonstrated activation of caspase-3, PARP cleavage, and nuclear fragmentation by 24 h and apoptosis by 72 h. Levels of procaspase-3 and procaspase-7, the precursor molecules of two effector caspases, were not depleted during apoptosis. Levels of procaspase-3 and -7 mRNA increased steadily in TSU-Pr1 cells up to 72 h after exposure to okadaic acid. Nuclear run-off experiments showed that the increase in mRNA was not due to transcriptional activation of caspase-3 and -7 mRNA. Antisense caspase-3 and caspase-7 oligodeoxynucleotides caused a depletion of procaspases-3 and -7 and a delay in apoptosis of TSU-Pr1 cells. Caspase antisense oligodeoxynucleotides inhibited apoptosis to a similar extent as peptide inhibitors of cysteine proteases. Synthesis of procaspases-3 and -7 was necessary to sustain programmed cell death in TSU-Pr1 prostate cancer cells.


Subject(s)
Apoptosis , Caspases/biosynthesis , Enzyme Precursors/biosynthesis , Caspase 3 , Caspase 7 , Enzyme Activation , Humans , Male , Okadaic Acid/pharmacology , Oligodeoxyribonucleotides, Antisense , Prostatic Neoplasms , Tumor Cells, Cultured
14.
Cancer Res ; 59(7): 1606-14, 1999 Apr 01.
Article in English | MEDLINE | ID: mdl-10197636

ABSTRACT

LNCaP prostate cancer cells are highly resistant to induction of programmed cell death by y-irradiation and somewhat sensitive to the death-inducing effects of tumor necrosis factor (TNF)-alpha. Simultaneous exposure of LNCaP cells to TNF-alpha and 8 Gy of irradiation was synergistic and resulted in a 3-fold increase of apoptotic cells within 72 h compared to TNF-alpha alone. It appeared that TNF-alpha sensitized the cells to irradiation because, when cells were irradiated 24 h after exposure to TNF-alpha, increased cell death was observed. In contrast, irradiation delivered 24 h prior to TNF-alpha exposure did not result in more cell death than after TNF-alpha alone. TNF-alpha induced expression of its own mRNA, but TNF-alpha mRNA induction was neither induced nor enhanced by irradiation. Activation of the transcription factor nuclear factor kappaB can be induced by TNF-alpha and has a modulating antiapoptotic effect. But enhancement of TNF-alpha-induced cell death by irradiation did not result from altered activation of nuclear factor kappaB. TNF-alpha treatment of LNCaP cells resulted in partial activation of caspase-8 and -6 but not caspase-3. There was only minimal poly(ADP-ribose) polymerase cleavage seen in LNCaP cells after exposure to both TNF-alpha and irradiation at 72 h, a time when 60% of the cells were apoptotic. Experiments with peptide inhibitors of cysteine and serine proteases suggested that caspases were the predominant mediators of apoptosis induced by TNF-alpha alone but that serine proteases contributed significantly to cell death induced by TNF-alpha plus irradiation. TNF-alpha increased production of ceramide in LNCaP cells 48 h after exposure. Although irradiation alone had no effect on ceramide production in LNCaP cells, TNF-alpha plus irradiation induced significantly more ceramide than TNF-alpha alone. Ceramide production did not occur immediately after exposure to TNF-alpha, but rather was delayed such that ceramide levels were increased only 24 h after exposure to apoptotic stimuli. Moreover, non-toxic levels of exogenous C2-ceramide sensitized LNCaP cells to irradiation similarly to TNF-alpha, suggesting that one mechanism by which LNCaP cells were sensitized to irradiation was by increased intracellular ceramide. Hence, ceramide generation is a critical component in radiation-induced apoptosis in human prostate cancer cells. Inhibition of ceramide generation may provide a selective advantage in the development of radioresistance in prostate cancer.


Subject(s)
Apoptosis/radiation effects , Prostatic Neoplasms/pathology , Tumor Necrosis Factor-alpha/pharmacology , Apoptosis/drug effects , Caspases/physiology , Ceramides/biosynthesis , Gamma Rays , Humans , Male , NF-kappa B/metabolism , Prostatic Neoplasms/radiotherapy , Radiation Tolerance , Tumor Necrosis Factor-alpha/biosynthesis , fas Receptor/physiology
15.
Cancer Res ; 58(15): 3275-81, 1998 Aug 01.
Article in English | MEDLINE | ID: mdl-9699655

ABSTRACT

The role of the retinoblastoma gene product, RB, in transmitting the signals of apoptosis is unclear, but RB is considered to be antiapoptotic because RB mediates cell cycle arrest that also can interrupt intracellular signaling pathways leading to apoptosis. Gamma-radiation can cause apoptosis, the process of programmed cell death, via several mechanisms including DNA damage, ceramide production, and the generation of free radical oxygen species. We investigated the effect of RB on radiation-induced apoptosis by restoring normal RB expression in DU-145 prostate cancer cells that have one deleted and one truncated RB gene. DU-145 cells are highly resistant to apoptosis induced either by radiation or by the addition of ceramide. Two independently derived RB-positive DU-145 derivative cell lines underwent apoptosis after irradiation or exposure to the cell permeable C2-ceramide. Apoptosis in the RB-positive cell lines was not associated with major changes in the cell cycle response to irradiation. RB-mediated apoptosis occurred in the absence of expression of caspases 8, 6, 3, and 7 and without detectable cleavage of poly(ADP)ribose polymerase. However, a specific inhibitor of serine proteases, Na-p-Tosyl-L-lysyl-chloromethyl ketone, inhibited radiation-induced apoptosis in DU-145 cells expressing RB. Radiation-induced apoptosis was preceded by an increase in JUN protein expression and accompanied by activation of the stress-related JUN kinase. Our data show that RB is proapoptotic in DU-145 cells and acts upstream of JUN expression and JNK activation.


Subject(s)
Apoptosis/physiology , Apoptosis/radiation effects , Caspases , JNK Mitogen-Activated Protein Kinases , Mitogen-Activated Protein Kinase Kinases , Retinoblastoma Protein/physiology , Caspase 3 , Cysteine Endopeptidases/metabolism , Enzyme Activation , Humans , MAP Kinase Kinase 4 , Male , Oncogene Protein p65(gag-jun)/metabolism , Phosphorylation , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Prostatic Neoplasms/radiotherapy , Protein Kinases/metabolism , Retinoblastoma Protein/genetics , Serine Endopeptidases/metabolism , Transfection , Tumor Cells, Cultured/radiation effects
16.
Cancer Res ; 58(12): 2520-3, 1998 Jun 15.
Article in English | MEDLINE | ID: mdl-9635571

ABSTRACT

Beta-catenin plays essential roles in both intercellular adhesion and signal transduction. As a signaling molecule, beta-catenin supplies an activating domain to the T-cell factor/lymphoid enhancer-binding factor family of DNA-binding proteins and activates gene transcription. Posttranslational stabilization of beta-catenin, leading to elevated protein levels and constitutive gene activation, has been proposed as an important step in oncogenesis. Stabilization of beta-catenin can occur through mutation to highly conserved amino acids encoded in exon 3 of the beta-catenin gene (CTNNB1). To determine whether this pathway of malignant transformation is important in prostate cancer, we analyzed 104 prostate cancer tissue specimens, 4 prostate cancer cell lines, and 3 prostate tumor xenografts for activating mutations in exon 3 of CTNNB1. Mutations were detected in 5 of the 104 prostate cancer tissue samples. Four of the five mutations involved serine or threonine residues implicated in the degradation of beta-catenin. A fifth tumor had a mutation at codon 32, changing a highly conserved aspartic acid to a tyrosine. Mutational analysis of multiple regions from several tumor samples showed that the beta-catenin mutations were present focally and therefore may occur during tumor progression.


Subject(s)
Cytoskeletal Proteins/genetics , Neoplasm Proteins/genetics , Point Mutation/genetics , Prostatic Neoplasms/genetics , Trans-Activators , Exons/genetics , Humans , Male , Polymorphism, Single-Stranded Conformational , Prostatic Neoplasms/chemistry , beta Catenin
17.
J Clin Oncol ; 16(2): 779-92, 1998 Feb.
Article in English | MEDLINE | ID: mdl-9469370

ABSTRACT

PURPOSE: To review the preclinical and clinical data on the carcinogenic potential of tamoxifen. DESIGN: A MEDLINE search on the carcinogenicity of tamoxifen was conducted and the literature reviewed. RESULTS: Because tamoxifen has estrogen-like effects on some tissues, such as the human uterus, there has been concern that tamoxifen could promote endometrial cancers in women on chronic tamoxifen therapy. Observations in some randomized trials of adjuvant tamoxifen therapy are consistent with a small, but real, increased risk of endometrial cancer in women who take tamoxifen. Since increased endometrial cancer incidence has not been observed in all studies of chronic tamoxifen therapy, there may be an element of detection bias. Laboratory studies have demonstrated that tamoxifen is hepatocarcinogenic in laboratory rats, but not in other species. This carcinogenicity in rats has been linked to the formation of DNA adducts. CONCLUSION: The incidence of endometrial cancer is increased in women who take tamoxifen. The data suggest that tamoxifen might be a tumor promoter in human endometrium. However, on the basis of the number of tumors seen by endometrial sampling of tamoxifen-treated women, the impact of tamoxifen as a tumor promoter is small. Women on chronic tamoxifen therapy should have routine annual gynecologic examinations and receive endometrial sampling only in the event of uterine bleeding. Unlike the data in rats, there is no conclusive evidence to link tamoxifen with an increased rate of hepatocellular cancer in humans; the contrasting carcinogenic potential may be attributed to substantial interspecies differences in the metabolism of tamoxifen.


Subject(s)
Antineoplastic Agents, Hormonal/adverse effects , Endometrial Neoplasms/chemically induced , Tamoxifen/adverse effects , Animals , Female , Humans , Risk Factors
19.
Cancer Res ; 57(20): 4455-9, 1997 Oct 15.
Article in English | MEDLINE | ID: mdl-9377551

ABSTRACT

Loss of heterozygosity at chromosome 8p21-22 is common in human prostate cancer, suggesting the presence of one or more tumor suppressor genes at this locus. A homeobox gene that is expressed specifically in adult human prostate, NKX3.1, the expression of which is regulated by androgen, maps to chromosome 8p21. Fine structure in situ mapping showed that NKX3.1 is proximal to MSR32 (macrophage scavenger receptor type II) and LPL (human lipoprotein lipase) and very close to NEFL (human neurofilament light chain) on 8p21. Single-strand conformational polymorphism analysis of 48 radical prostatectomy cancer specimens and 3 metastases for the entire coding region of NKX3.1 showed no tumor-specific sequence alterations in 50 specimens and total absence of the gene in 1 specimen known to have a biallelic deletion of 8p21. NKX3.1 was found to have a polymorphism at nucleotide 154 in codon 52 that resulted in a CGC-->TGC sequence change and an Arg-->Cys amino acid alteration (R52C). This polymorphism was present in 20% of DNA samples. If NKX3.1 is a target of the 8p21 LOH, it is not via disruption of the coding region of the gene.


Subject(s)
Chromosomes, Human, Pair 8 , Homeodomain Proteins/genetics , Point Mutation , Polymorphism, Single-Stranded Conformational , Prostate/metabolism , Prostatic Neoplasms/genetics , Sequence Deletion , Transcription Factors/genetics , Adult , Amino Acid Substitution , Arginine , Base Sequence , Chromosome Mapping , Cystine , DNA, Neoplasm/chemistry , Exons , Homeodomain Proteins/biosynthesis , Humans , Male , Molecular Sequence Data , Polymerase Chain Reaction , Prostatectomy , Prostatic Neoplasms/surgery , Transcription Factors/biosynthesis
20.
J Urol ; 158(1): 212-6, 1997 Jul.
Article in English | MEDLINE | ID: mdl-9186362

ABSTRACT

PURPOSE: Benign prostatic hyperplasia (BPH) is related to advancing age and the presence of androgens and occurs in virtually all older men. BPH causes morbidity, most often by urinary obstruction, in a substantial fraction of men over sixty. Both finasteride and androgen ablation induce partial diminution in BPH that occurs over weeks to months. This is in contrast to the often rapid involution seen in both normal prostatic epithelium and prostatic carcinoma in response to androgen withdrawal. This study was performed to analyze the response of prostatic cells, and in particular BPH, to acute androgen ablation. MATERIALS AND METHODS: We subjected a cohort of 26 men to androgen ablation with goserelin, a gonadotrophin releasing hormone agonist, for 3-4 weeks prior to radical prostatectomy for prostate cancer. Preablation biopsy specimens and prostatectomy specimens were immunohistochemically stained for apoptotic cells and for expression of apoptosis regulatory proteins Bcl-2, Bax, Bcl-x, and Bak. RESULTS: Normal prostatic epithelial cells and prostate cancer responded to hormone deprivation by undergoing apoptosis, but in 19/26 specimens prostatic hyperplasia had a total absence of apoptosis. In all 26 specimens, benign prostatic hyperplasia demonstrated increased expression of the Bcl-2 protein, but no change in the expression of Bax, Bcl-x, and Bak. In contrast, adjacent normal and malignant prostatic epithelium showed positive staining for apoptosis and did not alter Bcl-2 expression in response to androgen ablation. CONCLUSIONS: BPH demonstrated increased staining for Bcl-2 after androgen deprivation that may render hyperplastic epithelium relatively resistant to apoptosis induced acutely by androgen withdrawal.


Subject(s)
Apoptosis , Gonadotropin-Releasing Hormone/agonists , Goserelin/therapeutic use , Prostatic Hyperplasia/drug therapy , Prostatic Hyperplasia/pathology , Proto-Oncogene Proteins c-bcl-2/biosynthesis , Apoptosis/drug effects , Humans , Male , Membrane Proteins/biosynthesis , Prostatic Hyperplasia/metabolism , Proto-Oncogene Proteins/biosynthesis , Proto-Oncogene Proteins c-bcl-2/analysis , Up-Regulation , bcl-2 Homologous Antagonist-Killer Protein , bcl-2-Associated X Protein , bcl-X Protein
SELECTION OF CITATIONS
SEARCH DETAIL
...