Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 136
Filter
1.
Oncogene ; 30(42): 4327-38, 2011 Oct 20.
Article in English | MEDLINE | ID: mdl-21532617

ABSTRACT

Prostate cancer and breast cancer are the most common malignancies in the western world. Androgen receptor (AR) and PTEN both have been well documented to have important roles in prostate carcinogenesis. In contrast, AR and PTEN in breast carcinogenesis have not been well studied. Furthermore, the crosstalk and connection between those two pathways remain unclear. Increased AR expression in prostate cancers, combined with decreased PTEN expression, portends a poor clinical outcome. Paradoxically, both high AR and high PTEN levels, detected by immunohistochemistry, in primary breast carcinomas have been associated with better disease-free survival. Here, we performed in silico analysis of publicly available microarray data sets from prostate or breast carcinomas. We found an inverse correlation between AR and PTEN transcript expression in prostate cancer tissues in contrast to the positive correlation in breast cancer. These data led us to hypothesize that AR may directly affect PTEN transcriptional regulation in prostate and breast cancer cells. Here, we show for the first time that AR inhibits PTEN transcription in prostate cancer cells, whereas AR upregulates PTEN transcription in breast cancer cells, which mechanistically explains both the immunohistochemical PTEN-AR expressional data noted in clinical trials and in our in silico analysis of the transcriptomes of breast and prostate cancers. In addition, we have fine-mapped the AR-binding motif within the PTEN promoter. Here we show that, in patients with Cowden syndrome, an inherited cancer syndrome caused by germline mutations scattered throughout PTEN, point variants affecting the 3' end of the AR-binding motif result in abrogation of androgen-mediated transcriptional regulation of PTEN expression. We may speculate that the differential AR effect on PTEN may begin to explain organ-specific and perhaps sex-specific neoplasia predisposition in Cowden syndrome, as well as why only a fraction of women with germline PTEN mutations develop breast cancer, depending on the androgen steroid milieu and levels.


Subject(s)
Breast Neoplasms/metabolism , Carcinoma/metabolism , PTEN Phosphohydrolase/metabolism , Prostatic Neoplasms/metabolism , Receptors, Androgen/metabolism , Base Sequence , Breast Neoplasms/genetics , Carcinoma/genetics , Cell Line, Tumor , Female , Gene Expression Regulation, Neoplastic , Hamartoma Syndrome, Multiple/genetics , Hamartoma Syndrome, Multiple/metabolism , Humans , Male , Molecular Sequence Data , PTEN Phosphohydrolase/genetics , Point Mutation , Promoter Regions, Genetic , Prostatic Neoplasms/genetics , Sex Factors
2.
Crit Rev Immunol ; 21(1-3): 249-61, 2001.
Article in English | MEDLINE | ID: mdl-11642607

ABSTRACT

Prostate-specific membrane antigen (PSMA) is a potential target in prostate cancer patients because it is very highly expressed and because it has been reported to be upregulated by androgen deprivation. This review discusses the historical background, biochemical characteristics, gene regulation, potential for targeting, tissue localization, and a novel T-body strategy.


Subject(s)
Antigens, Surface , Carboxypeptidases/analysis , Prostatic Neoplasms/therapy , Alternative Splicing , Animals , Antibodies, Monoclonal , Binding Sites , Cancer Vaccines/immunology , Carboxypeptidases/genetics , Cloning, Molecular , Genetic Therapy , Glutamate Carboxypeptidase II , Humans , Male , Mice , Prostate/diagnostic imaging , Prostatic Neoplasms/immunology , Radionuclide Imaging
4.
Urology ; 58(2 Suppl 1): 132-9, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11502468

ABSTRACT

Prostate-specific membrane antigen (PSMA) is a type-2 membrane protein expressed in the prostate, and it is highly expressed in metastatic or poorly differentiated adenocarcinomas. Moreover, PSMA expression is upregulated by androgen deprivation. These advantages make PSMA a useful target for prostate cancer therapy, especially in combination with conventional hormonal treatment. We recently reported that a prostate-specific enhancer is present in the third intron of the PSMA gene. In this study, we have further analyzed the activity of PSMA promoter/enhancer in prostate cancer cells and cells of other tissue origins (breast cancer MCF-7, lung cancer H157, and colorectal cancer HCT8 cells), and we have examined whether this construct could be used for efficient expression of the suicide gene, cytosine deaminase (CD), in vivo. The PSMA promoter/enhancer expressed the luciferase reporter gene in the prostate cancer lines LNCaP and C4-2, with 8- to 20-fold higher expression than the simian virus 40 promoter/enhancer, although it was inactive in the other cell lines. This construct efficiently drove the suicide gene CD, sensitizing C4-2 cells to 5-fluorocytosine (5-FC) with the inhibitory concentration (IC(50)) <300 micromol/L in vitro. Athymic male nude mice bearing the transfected C4-2 cells were treated with intraperitoneal injections of either 5-FC (600 mg/kg) twice a day or saline solution for 3 weeks. C4-2 cell tumors were eliminated by 5-FC when they were expressing our therapeutic construct carrying CD under the regulatory control of the PSMA promoter/enhancer. Our results show the in vivo utility of the PSMA promoter/enhancer in a gene therapy situation targeting prostate cancer.


Subject(s)
Adenocarcinoma/therapy , Genetic Therapy/methods , Prostate-Specific Antigen/genetics , Prostatic Neoplasms/therapy , Adenocarcinoma/genetics , Animals , Cytosine Deaminase , Flucytosine/therapeutic use , Gene Expression , Genes, Reporter/genetics , Humans , Luciferases/genetics , Male , Mice , Nucleoside Deaminases/genetics , Nucleoside Deaminases/metabolism , Prodrugs , Promoter Regions, Genetic/genetics , Prostatic Neoplasms/genetics , Simian virus 40/genetics , Tumor Cells, Cultured/metabolism
5.
Urology ; 57(6): 1179-83, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11377343

ABSTRACT

OBJECTIVES: To compare the immunohistochemical properties of the 7E11 anti-prostate-specific membrane antigen (anti-PSMA) monoclonal antibody (mAb) with the recently developed anti-PSMA mAb, PM2J004.5, and with other common immunomarkers in metastatic prostate cancer. PSMA is a type II integral membrane glycoprotein highly expressed in prostate cancer cells. The mAb 7E11 is currently used in the radioisotopic evaluation of prostate cancer, and its immunohistochemical properties have been examined in primary prostate cancer specimens. METHODS: We examined 23 formalin-fixed, paraffin-embedded, metastatic prostate carcinoma specimens from various anatomic sites, including bone, lymph node, liver, lung, and soft tissue. Using the biotin-streptavidin method, we performed immunohistochemical reactions with the anti-PSMA mAbs 7E11 and PM2J004.5 and with antibodies to prostate-specific antigen and prostatic acid phosphatase. The immunoreactions were scored by pathologists unaware of the clinical and pathologic data according to a staining intensity scale and the percentage of cells stained. RESULTS: All four mAbs consistently stained the metastatic prostate cancer specimens. In 2 (8.7%) of 23 cases, however, the prostate-specific antigen immunoreaction was negative but the anti-PSMA mAbs had positive staining. Although 7E11 and PM2J004.5 had a similar staining intensity and percentage of cells stained for most specimens, in 3 (13%) of 23 specimens, 7E11 had less intense staining. None of the specimens were negative for all four antibodies. CONCLUSIONS: Anti-PSMA mAbs consistently immunoreacted with metastatic prostate cancer specimens and were positive in instances when prostate-specific antigen staining was negative. The anti-PSMA mAbs demonstrated similar staining patterns; however, in select cases, the PM2J004.5 mAb did show more intense staining. The anti-PSMA mAbs 7E11 and PM2J004.5 are useful in the pathologic evaluation of paraffin-embedded metastatic prostate cancer specimens.


Subject(s)
Antibodies, Monoclonal , Antigens, Surface , Biomarkers, Tumor/immunology , Carboxypeptidases/immunology , Prostatic Neoplasms/pathology , Glutamate Carboxypeptidase II , Humans , Immunohistochemistry , Male , Paraffin Embedding , Prostatic Neoplasms/immunology
7.
Genomics ; 73(3): 243-54, 2001 May 01.
Article in English | MEDLINE | ID: mdl-11350116

ABSTRACT

Prostate-specific membrane antigen (PSMA) is an integral membrane protein that is highly expressed on the surface of prostate epithelial cells. It is also expressed on the vascular endothelium of a number of tumor types. We have used an enhancer trap approach with randomly cleaved overlapping DNA fragments from an approximately 55-kb P1 cosmid insert encompassing the 5' half and upstream sequences of the PSMA gene (FOLH1) to isolate an enhancer that strongly activates the FOLH1 core promoter region. The enhancer (PSME) is located in the third intron about 12 kb downstream from the start site of transcription and is characterized by a 72-bp direct repeat within a 331-bp core region. The PSME activates transcription from its own and heterologous promoters in prostate cell lines; enhancement is greatest in the PSMA-expressing cell line LNCaP (>250-fold). The PSME shows essentially no activity in five nonprostate cell lines. PSME-enhanced expression is repressed in the presence of androgen, mimicking the repression of the endogenous FOLH1 gene. The data demonstrate that both cell-type specificity and androgen regulation are intrinsic properties of the enhancer. These properties make the PSME an excellent candidate for regulation of gene expression in gene therapy approaches to prostate cancer.


Subject(s)
Antigens, Surface , Carboxypeptidases/genetics , Enhancer Elements, Genetic/genetics , Gene Expression Regulation , Prostate/metabolism , Androgens/pharmacology , Base Sequence , Cell Line , Cloning, Molecular , Gene Expression Regulation/drug effects , Glutamate Carboxypeptidase II , Humans , Introns/genetics , Male , Molecular Sequence Data , Organ Specificity , Promoter Regions, Genetic/genetics , Restriction Mapping , Sequence Analysis, DNA , Sequence Deletion/genetics , Transfection , Tumor Cells, Cultured
8.
Urology ; 57(4): 801-5, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11306418

ABSTRACT

OBJECTIVES: To determine whether the tumor-associated neovasculature of metastatic prostate and metastatic conventional (clear cell) renal carcinoma express prostate-specific membrane antigen (PSMA). PSMA is a type II integral membrane glycoprotein highly expressed in prostate cancer cells and also recently discovered to be expressed in the neovasculature of non-prostatic primary malignancies. METHODS: We examined metastatic prostate carcinoma (22 patients) and metastatic conventional (clear cell) renal carcinoma (20 patients) in various anatomic sites, including bone, lymph nodes, liver, lung, and soft tissue. Using the biotin-streptavidin method, we performed immunohistochemical reactions with the anti-PSMA monoclonal antibodies (mAbs) 7E11 and PM2J004.5 and with the anti-endothelial cell mAb CD34. RESULTS: Metastatic conventional (clear cell) renal carcinoma consistently expressed PSMA. The PM2J004.5 mAb was positive in 20 of 20 specimens, and the 7E11 mAb was positive in 15 of 20. The anti-PSMA immunoreactions with the neovasculature were confirmed by similar staining by the anti-CD34 mAb (20 of 20). Although the metastatic prostatic cancer cells expressed PSMA in all the specimens, only 2 of 22 had neovasculature PSMA expression. CONCLUSIONS: As in primary prostatic adenocarcinomas, the neovasculature of metastatic prostate cancer, regardless of site, rarely express PSMA. The neovascular endothelial cells of metastatic clear cell renal carcinoma, however, express PSMA. This expression may make PSMA an effective target for mAb-based antineovasculature therapy in metastatic renal carcinoma.


Subject(s)
Carcinoma, Renal Cell/chemistry , Carcinoma, Renal Cell/secondary , Endothelium, Vascular/chemistry , Kidney Neoplasms/chemistry , Kidney Neoplasms/secondary , Prostate-Specific Antigen/analysis , Adenocarcinoma, Clear Cell/blood supply , Adenocarcinoma, Clear Cell/chemistry , Adenocarcinoma, Clear Cell/secondary , Carcinoma, Renal Cell/blood supply , Humans , Immunohistochemistry , Kidney Neoplasms/blood supply , Lymphatic Metastasis , Male , Prostatic Neoplasms/pathology
9.
Mamm Genome ; 12(2): 117-23, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11210180

ABSTRACT

Human Prostate Specific Membrane Antigen (PSMA), also known as folate hydrolase I (FOLH1), is a 750-amino acid type II membrane glycoprotein, which is primarily expressed in normal human prostate epithelium and is upregulated in prostate cancer, including metastatic disease. We have cloned and sequenced the mouse homolog of PSMA, which we have termed Folh1, and have found that it is not expressed in the mouse prostate, but primarily in the brain and kidney. We have demonstrated that Folh1, like its human counterpart, is a glutamate-preferring carboxypeptidase, which has at least two enzymatic activities: (1) N-acetylated alpha-linked L-amino dipeptidase (NAALADase), an enzyme involved in regulation of excitatory signaling in the brain, and (2) a gamma-glutamyl carboxypeptidase (folate hydrolase). The 2,256-nt open reading frame of Folh1 encodes for a 752-amino acid protein, with 86% identity and 91% similarity to the human PSMA amino acid sequence. Cells transfected with Folh1 gained both NAALADase and folate hydrolase activities. Examination of tissues for NAALADase activity correlated with the mRNA expression pattern for Folh1. Fluorescent in situ hybridization (FISH) revealed Folh1 maps to only one locus in the mouse genome, Chromosome 7D1-2.


Subject(s)
Carboxypeptidases/genetics , Genome , Amino Acid Sequence , Animals , Antigens, Surface/genetics , Antigens, Surface/metabolism , Base Sequence , Blotting, Northern , Carboxypeptidases/metabolism , Cell Line , Cloning, Molecular , Gene Expression Regulation , Glutamate Carboxypeptidase II , In Situ Hybridization, Fluorescence , Mice , Molecular Sequence Data , Open Reading Frames , Organ Specificity , RNA, Messenger/analysis , Reverse Transcriptase Polymerase Chain Reaction , Sequence Analysis, DNA , Transfection
10.
Prostate ; 45(4): 304-14, 2000 Dec 01.
Article in English | MEDLINE | ID: mdl-11102955

ABSTRACT

BACKGROUND: This study determined the effects of S-allylmercaptocysteine (SAMC), a phytoconstituent from garlic, on the expression of androgen-responsive biomarkers, prostate specific antigen (PSA), and prostate specific membrane antigen (PSMA), in human prostatic carcinoma cells (LNCaP). METHODS: Secretion of PSA was determined as well as the activity of PSMA measured as a function of its ability to hydrolyze poly-gamma-glutamated folate and N-acetylaspartylglutamate (NAAG). Folate hydrolase capacity was also determined in SAMC-treated cells grown in charcoal stripped fetal calf serum (CS-FCS). In addition, testosterone disappearance was measured from culture media of SAMC-treated LNCaP and PC-3 cells as well as from cell free lysates. RESULTS: PSA secretions were significantly decreased compared to control values at 1 day (8.4 +/- 2.6 vs. 18.9 +/- 1.7, P < 0.01), 4 days (18.9 +/- 5.3 vs. 73.8 +/- 4. 4, P < 0.001), and 6 days (35.6 +/- 2.1 vs. 96.5 +/- 17.9 ng/10(5) cells, P < 0.01; mean +/- SD). By contrast, PSMA activity measured as either folate hydrolase or NAAG dipeptidase (NAALADase) activity increased in cells treated with SAMC. PSMA-folate hydrolase activity in SAMC-treated cells grown in CS-FCS increased beyond that observed in cells grown in CS-FCS alone. Pre-exposure of LNCaP cells to SAMC resulted in enhanced rate of testosterone disappearance from culture media at 6 hr (P < 0.01) and at 48 hr (P < 0.001) compared to media from cells not previously exposed to SAMC. Results similar to these were also observed in androgen-independent PC-3 cells treated with SAMC. In lysates of SAMC-treated LNCaP cells, the rate of testosterone catabolism was twice that from phosphate buffered saline (PBS)-treated cells. SAMC-treated LNCaP cells grown in media supplemented with testosterone temporarily exhibited enhanced growth over a 2 day period but cell numbers declined later to levels similar to those of SAMC treatment. CONCLUSIONS: These results show that SAMC exhibits differential effects on recognized biomarkers for LNCaP cells similar to those produced by androgen deprivation and strongly suggests that this effect may be mediated, in part, by diminishing the trophic effects of testosterone, likely by converting it to metabolites less reactive toward androgen receptors.


Subject(s)
Adenocarcinoma/metabolism , Antigens, Surface , Biomarkers, Tumor/biosynthesis , Cysteine/analogs & derivatives , Cysteine/pharmacology , Prostatic Neoplasms/metabolism , Testosterone/metabolism , Adenocarcinoma/immunology , Adenocarcinoma/pathology , Antigens, Neoplasm/biosynthesis , Carboxypeptidases/biosynthesis , Carboxypeptidases/metabolism , Cell Division/drug effects , Culture Media , Cysteine/metabolism , Drug Interactions , Garlic/chemistry , Glutamate Carboxypeptidase II , Growth Inhibitors/pharmacology , Humans , Male , Plants, Medicinal , Prostate-Specific Antigen/biosynthesis , Prostate-Specific Antigen/metabolism , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/immunology , Receptors, Androgen/metabolism , Secretory Rate/drug effects , Testosterone/pharmacokinetics , Testosterone/pharmacology , Tumor Cells, Cultured/drug effects , gamma-Glutamyl Hydrolase/metabolism
11.
Mol Urol ; 4(3): 217-22;discussion 223, 2000.
Article in English | MEDLINE | ID: mdl-11062377

ABSTRACT

Prostate-specific membrane antigen (PSMA) is a potential target in prostate cancer patients because it is very highly expressed and because it has been reported to be upregulated by androgen deprivation. This overview addresses the expression of the PSMA gene in terms of the promoter and enhancer and how that may play a role in gene therapy. We also review PSMA as a target for antibodies for imaging and treatment and the development of a novel hybrid T-cell receptor that combines the specificity of anti-PSMA antibodies with that of T-cell receptor activation when introduced into primary lymphocytes by retroviral-mediated gene transfer. We also discuss our recent findings on the expression of a PSMA-like gene and how that understanding allows specific targeting of PSMA.


Subject(s)
Antigens, Neoplasm/metabolism , Carboxypeptidases/metabolism , Prostatic Neoplasms/enzymology , Prostatic Neoplasms/therapy , Animals , Antibodies, Neoplasm/immunology , Antigens, Neoplasm/genetics , Antigens, Neoplasm/immunology , Antigens, Surface/genetics , Antigens, Surface/immunology , Antigens, Surface/metabolism , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Carboxypeptidases/genetics , Carboxypeptidases/immunology , Enhancer Elements, Genetic , Enzyme Inhibitors/pharmacology , Female , Genetic Therapy , Glutamate Carboxypeptidase II , Humans , Male , Prodrugs/metabolism , Promoter Regions, Genetic , Prostatic Neoplasms/blood supply , Prostatic Neoplasms/genetics , Receptors, Antigen, T-Cell/immunology , Tumor Cells, Cultured
12.
Prostate ; 45(2): 149-57, 2000 Oct 01.
Article in English | MEDLINE | ID: mdl-11027414

ABSTRACT

BACKGROUND: Prostate-specific membrane antigen (PSMA) is abundantly expressed in virtually 100% of prostate cancers and metastases. In addition, unlike prostate-specific antigen (PSA), PSMA is upregulated under conditions of androgen deprivation. Therefore, PSMA is an attractive therapeutic target for advanced prostate cancer. Recently, both the promoter and the enhancer driving prostate-specific expression of the PSMA gene were cloned. We describe here our analysis of the PSMA enhancer for the most active region(s) and present a way of using the enhancer in combination with the E. coli cytosine deaminase gene for suicide-driven gene therapy that converts the nontoxic prodrug 5-fluorocytosine (5-FC) into the cytotoxic drug 5-fluorouracil (5-FU) in prostate cancer cells. METHODS: Deletion constructs of the full-length PSMA enhancer were subcloned into a luciferase reporter vector containing either the PSMA or SV-40 promoter. The most active portion of the enhancer was then determined via luciferase activity in the C4-2 cell line. We then replaced the luciferase gene with the E. coli cytosine deaminase gene in the subclone that showed the most luciferase activity. The specificity of this technique was examined in vitro, using the prostate cancer cell line LNCaP, its androgen-independent derivative C4-2, and a number of nonprostatic cell lines. The toxicity of 5-FC and 5-FU on transiently transfected cell lines was then compared. RESULTS: The enhancer region originally isolated from the PSMA gene was approximately 2 kb. Deletion constructs revealed that at least two distinct regions seem to contribute to expression of the gene in prostate cancer cells, and therefore the best construct for prostate-specific expression was determined to be 1, 648 bp long. The IC(50) of 5-FC was similar in all cell lines tested (>10 mM). However, transfection with the 1648 nt PSMA enhancer and the PSMA promoter to drive the cytosine deaminase gene enhanced toxicity in a dose-dependent manner more than 50-fold, while cells that did not express the PSMA gene were not significantly sensitized by transfection. CONCLUSIONS: Suicide gene therapy using the PSMA enhancer may be of benefit to patients who have undergone androgen ablation therapy and are suffering a relapse of disease.


Subject(s)
Antigens, Surface , Carboxypeptidases/genetics , Enhancer Elements, Genetic , Escherichia coli/enzymology , Genetic Therapy , Nucleoside Deaminases/genetics , Promoter Regions, Genetic , Base Sequence , Cell Division/drug effects , Cytosine Deaminase , DNA, Complementary , Escherichia coli/genetics , Flucytosine/pharmacology , Fluorouracil/pharmacology , Genes, Reporter , Genetic Therapy/methods , Glutamate Carboxypeptidase II , Green Fluorescent Proteins , Humans , Luminescent Proteins/genetics , Male , Molecular Sequence Data , Nucleoside Deaminases/metabolism , Prodrugs/pharmacology , Transfection , Tumor Cells, Cultured
13.
Clin Cancer Res ; 6(7): 2913-20, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10914741

ABSTRACT

We have developed a novel liposome-mediated immunogene therapy using interleukin 2 (IL-2) and B7.1 in a murine bladder cancer model. A carcinogen-induced murine bladder cancer cell line, MBT-2, was transfected with cationic liposome 1,2-dimyristyloxypropyl-3-dimethyl-hydroxyethyl ammonium bromide/dioleolylphosphatidylethanolamine and IL-2 plasmid. The optimized transfection condition generated IL-2 levels of 245-305 ng/10(6) cells/24 h, 100-fold higher than the levels seen with retrovirus transfection. Ninety percent of the peak level of IL-2 production was maintained for up to 11 days after transfection. Animal studies were conducted in C3H/HeJ female mice with 2 x 10(4) MBT-2 cells implanted orthotopically on day 0. Multiple vaccination schedules were performed with i.p. injection of 5 x 10(6) IL-2 and/or B7.1 gene-modified cell preparations. The greatest impact on survival was observed with the day 5, 10, and 15 regimen. Control animals receiving retrovirally gene-modified MBT-2/IL-2 cell preparations had a median survival of 29 days. Animals receiving the IL-2 liposomally gene-modified cell preparation alone had a median survival of 46 days. Seventy-five percent of animals receiving IL-2 followed by B7.1 gene-modified tumor vaccines were the only group to show complete tumor-free survival at day 60. All of these surviving animals rejected the parental MBT-2 tumor rechallenge and survived at day 120 with a high CTL response. In conclusion, liposome-mediated transfection demonstrates a clear advantage as compared with the retroviral system in the MBT-2 model. Multi-agent as opposed to single-agent cytokine gene-modified tumor vaccines were beneficial. These "targeted" sequential vaccinations using IL-2 followed by B7.1 gene-modified tumor cells significantly increased a systemic immune response that translated into increased survival.


Subject(s)
B7-1 Antigen/therapeutic use , Cancer Vaccines/therapeutic use , Genetic Therapy/methods , Interleukin-2/genetics , Phosphatidylethanolamines , Urinary Bladder Neoplasms/therapy , Animals , Combined Modality Therapy , Disease-Free Survival , Drug Administration Schedule , Drug Carriers , Female , Glycerophospholipids , Interleukin-2/therapeutic use , Lipids , Liposomes , Mice , Mice, Inbred C3H , Quaternary Ammonium Compounds , Recombinant Proteins/genetics , Recombinant Proteins/therapeutic use , T-Lymphocytes, Cytotoxic/immunology , Transfection/methods , Tumor Cells, Cultured , Urinary Bladder Neoplasms/immunology
14.
Gene Ther ; 7(10): 844-51, 2000 May.
Article in English | MEDLINE | ID: mdl-10845722

ABSTRACT

Using a novel orthotopic MBT-2 murine bladder tumor model, we evaluated the feasibility of intravesical gene therapy utilizing a cationic liposome, DMRIE/DOPE. Superficial bladder tumors were consistently established by intravesical instillation of 5x10(5) MBT-2 cells in syngeneic C3H female mice. In situ gene transfer to bladder tumors was accomplished via intravesical instillation of plasmid DNA/DMRIE/DOPE lipoplex. Beta-Galactosidase (beta-gal) gene expression was preferentially evident in bladder tumors and was present for at least 7 days after a single 30 min in situ transfection. Murine interleukin-2 (IL-2) gene was used for treatment of 3-day-old pre-established bladder tumors. Forty percent of animals treated with IL-2 gene were completely free of tumors by 60 days following the initial tumor implantation, while all control groups treated with beta-gal gene died. Those animals initially cured of pre-established tumors were completely resistant to a subsequent tumor re-challenge and their splenocyte-derived cytotoxic T lymphocytes were shown to be specific to MBT-2 cells, indicating that immunological memory against MBT-2 tumors was elicited by the treatment. These results demonstrate the possibility of an effective clinical application of this in situ intravesical IL-2 gene delivery system to high-risk superficial bladder tumors, obviating a need for tumor procurement and ex vivo gene transfer.


Subject(s)
Genetic Therapy/methods , Interleukin-2/genetics , Urinary Bladder Neoplasms/therapy , Administration, Intravesical , Animals , Cytotoxicity, Immunologic , Disease Models, Animal , Feasibility Studies , Female , Genes, Reporter , Immunologic Memory , Liposomes , Mice , Mice, Inbred C3H , Neoplasm Transplantation , Transfection , Tumor Cells, Cultured , Urinary Bladder Neoplasms/immunology , Urinary Bladder Neoplasms/prevention & control
16.
Histol Histopathol ; 15(2): 365-74, 2000 04.
Article in English | MEDLINE | ID: mdl-10809354

ABSTRACT

The tyrosine kinase receptor c-kit and its ligand [kit ligand (KL) or stem cell factor (SCF)] exert a broad range of biological activities during organogenesis and normal cell development. Recent studies have revealed that altered c-kit levels occur in a variety of malignancies and cancer cell lines. KL has also been shown to stimulate the growth of malignant cells, as well as to promote chemotaxis. We had previously reported expression of KL in stroma cells of normal human prostate. The present study was undertaken in order to analyze the patterns of expression of c-kit and KL in a well characterized set of prostatic tissues, including normal prostate (n=4), benign prostatic hyperplasia (BPH) (n=53) and adenocarcinoma (n=46) samples. The distribution of c-kit and KL proteins was studied by immunohistochemical analyses, while transcript levels were determined by in situ hybridization with specific RNA probes on a subset of the benign and malignant tissues referred above. In addition, reverse-transcriptase polymerase chain reaction (RT-PCR) was performed to determine levels of c-kit and KL expression in cultures of epithelial and stroma cells, as well as in the prostate cancer cell lines LNCaP, DU145 and PC3. c-kit protein in normal prostate was exclusively detected in mast cells by immunohistochemistry and in situ hybridization. However, c-kit transcripts, but not c-kit protein, were detected in low levels and with an heterogeneous pattern in basal epithelial cells of ducts and acini. c-kit in BPH was detected in epithelial cells in 9 of 53 (17%) specimens. c-kit protein expression in malignant epithelial cells was identified in 1 of 46 (2%) tumors. However, c-kit transcripts were detected in low levels by in situ hybridization in most of the tumors analyzed. KL protein and transcripts in normal prostate were detected in high levels in stroma cells. However, epithelial cells were unreactive for anti-KL antibody, but showed low levels of KL transcripts mainly in cells of the basal layer. Basal epithelial cells in hyperplastic glands showed KL expression in 13 of 53 (24%) specimens. KL protein in tumor cells was noted in 18 of 46 (39%) cases. c-kit transcripts were not found in normal prostate and in the 3 cancer cell lines analyzed by RT-PCR, however, it was present in cultured epithelial cells of BPH, and in cultures of stroma cells from both normal and BPH. The majority of cultured cell lines of epithelial and stromal origin displayed considerable levels of KL. In addition all prostate cell lines studied showed significant levels of KL transcripts. In summary, co-expression of c-kit and KL in a subset of BPH cases may suggest an autocrine mode of signaling. Data from this study reveals that altered patterns of c-kit and KL expression are associated with BPH and adenocarcinoma of prostate. It appears that KL induces mast cells proliferation and maturation and enhances their release of protease. This could explain the accumulation of mast cells at tumor sites, a phenomenon that was not observed in normal prostate or BPH samples.


Subject(s)
Adenocarcinoma/metabolism , Prostate/metabolism , Prostatic Hyperplasia/metabolism , Prostatic Neoplasms/metabolism , Proto-Oncogene Proteins c-kit/metabolism , Stem Cell Factor/metabolism , Adenocarcinoma/pathology , Adult , Animals , Humans , Immunoenzyme Techniques , In Situ Hybridization/methods , Ligands , Male , Mice , Phenotype , Prostate/pathology , Prostatic Hyperplasia/pathology , Prostatic Neoplasms/pathology , Proto-Oncogene Proteins c-kit/genetics , Reverse Transcriptase Polymerase Chain Reaction/methods , Stem Cell Factor/genetics , Tumor Cells, Cultured
17.
Clin Cancer Res ; 6(3): 838-46, 2000 Mar.
Article in English | MEDLINE | ID: mdl-10741705

ABSTRACT

The evaluation of new therapies in prostate cancer requires unique end points for agents with diverse mechanisms of action. Because retinoic acid may have a confounding effect on prostate-specific antigen, we incorporated a pathological end point into the outcome assessment of two sequential clinical trials using all-trans-retinoic acid (ATRA) and the combination of 13-cis-retinoic acid and IFN-2a (cRA¿IFN). Pre- and posttherapy tumor biopsy specimens were studied for histological changes, apoptosis (terminal deoxynucleotidyl transferase-mediated nick end labeling assay), and proliferation index (Ki67). Prostate-specific membrane antigen (PSMA) expression was also evaluated using two different monoclonal antibodies to its intracellular domain (Cytogen 7E11 and Hybritech PM2). Fourteen patients with androgen-independent disease were treated with ATRA (50 mg/m2 p.o. every 8 h daily) and 16 androgen-independent and 4 androgen-dependent patients were treated with cRA¿IFN (10 mg/kg/day cRA plus 3, 6, or 9 million units daily IFN). Both therapies were well tolerated, with fatigue and cheilitis being the most common adverse events. Clinical activity, assessed by radiographs and serum prostate-specific antigen, was minimal, and the majority of patients progressed within 3 months. One patient with androgen-dependent disease had prolonged stabilization for >1 year. The majority of cases (95%) showed no gross histological changes and no difference in apoptotic or proliferative indices. Increased PSMA immunoreactivity was seen in seven of nine (78%) cases using PM2 antibody and in two of nine (22%) cases using the 7E11 antibody. Although antitumor effects were modest, the results suggest a role for retinoids in modulating the expression of PSMA on prostate cancer cells.


Subject(s)
Antigens, Surface , Antineoplastic Agents/therapeutic use , Prostatic Neoplasms/drug therapy , Tretinoin/therapeutic use , Aged , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/adverse effects , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Biopsy , Bone Neoplasms/drug therapy , Bone Neoplasms/secondary , Carboxypeptidases/analysis , Cheilitis/chemically induced , Dyspnea/chemically induced , Exanthema/chemically induced , Fatigue/chemically induced , Glutamate Carboxypeptidase II , Hematologic Diseases/chemically induced , Humans , Interferon alpha-2 , Interferon-alpha/administration & dosage , Interferon-alpha/adverse effects , Ki-67 Antigen/analysis , Liver/enzymology , Male , Middle Aged , Prostate/chemistry , Prostate/drug effects , Prostate/pathology , Prostate-Specific Antigen/blood , Prostate-Specific Antigen/drug effects , Prostatic Neoplasms/pathology , Recombinant Proteins , Transaminases/drug effects , Transaminases/metabolism , Treatment Outcome , Tretinoin/administration & dosage , Tretinoin/adverse effects
18.
Cancer ; 88(2): 407-15, 2000 Jan 15.
Article in English | MEDLINE | ID: mdl-10640975

ABSTRACT

BACKGROUND: Prostate specific membrane antigen (PSMA) is a transmembrane glycoprotein highly expressed in benign prostate secretory-acinar epithelium and prostate carcinoma. The results of several studies suggest that PSMA expression is increased in prostate carcinoma cell lines subjected to androgen deprivation and in androgen-independent tumors. The authors studied the effects of short term (3-month) androgen deprivation on PSMA expression in prostate carcinoma specimens using two anti-PSMA monoclonal antibodies (mAbs), 7E11 and PM2J004.5. METHODS: The study included patients with clinically localized prostate carcinoma who were prospectively randomized into 1 of 2 treatment groups: 3 months of neoadjuvant androgen deprivation therapy followed by radical prostatectomy (ADT/RP), or radical prostatectomy (RP) alone. Representative formalin fixed, paraffin embedded prostate sections were immunostained with the anti-PSMA mAbs 7E11 and PM2J004.5 by the streptavidin-biotin method. The authors recorded the staining intensity and the percentage of positive cells stained in benign epithelium, high grade prostatic intraepithelial neoplasia (PIN), and prostate carcinoma. They compared the results of 7E11 with those of PM2J004.5 in benign epithelium, high grade prostate, and carcinoma and also compared the results between the two treatment groups (ADT/RP vs. RP alone). RESULTS: Both anti-PSMA mAbs stained benign secretory-acinar epithelium, high grade PIN, and prostate carcinoma. In both treatment groups, PM2J004.5 reacted with a significantly greater percentage of cells (P < 0.001) and with significantly greater intensity (P < 0.001) compared with 7E11 in benign epithelium and prostate carcinoma. With both anti-PSMA mAbs, the percentage of cells stained and the intensity of staining in high grade PIN was similar to that in prostate carcinoma. In the group that received RP alone, the percentage of cells stained and the intensity of staining with 7E11 were significantly greater in high grade PIN and prostate carcinoma compared with benign epithelium (P < 0.001), and the intensity of staining with the PM2J004.5 was significantly greater in high grade PIN and prostate carcinoma compared with benign epithelium (P < 0.001). In the ADT/RP group, the percentage of cells stained and the intensity of staining with 7E11 and PM2J004.5 were significantly greater in prostate carcinoma compared with benign epithelium (P < 0.006). PSMA staining did not correlate with either Gleason score (in the group that received RP alone) or pathologic stage (in both the RP-alone and ADT/RP groups) and did not differ between the two treatment groups. CONCLUSIONS: Short term neoadjuvant ADT does not affect PSMA expression in benign prostate secretory-acinar epithelium, high grade PIN, or prostate carcinoma. Prostate carcinoma and high grade PIN express significantly higher levels of PSMA than benign prostate secretory-acinar epithelium. Compared with 7E11, the PM2J004.5 anti-PSMA mAb is a more sensitive immunohistochemical marker of prostate carcinoma in formalin fixed, paraffin embedded tissue.


Subject(s)
Androgen Antagonists/pharmacology , Antigens, Surface , Carboxypeptidases/biosynthesis , Prostatic Neoplasms/drug therapy , Aged , Androgen Antagonists/therapeutic use , Antibodies, Monoclonal , Carboxypeptidases/analysis , Glutamate Carboxypeptidase II , Humans , Immunohistochemistry , Male , Middle Aged , Neoadjuvant Therapy , Prostatectomy , Prostatic Neoplasms/pathology , Prostatic Neoplasms/surgery , Sensitivity and Specificity
19.
Curr Opin Urol ; 9(5): 391-5, 1999 Sep.
Article in English | MEDLINE | ID: mdl-10579076

ABSTRACT

Over the past two decades, monoclonal antibody technology has had an increasing impact on clinical diagnostic and therapeutic options, and this is true in the realm of managing prostate cancer. Several targets such as prostate-specific antigen and prostatic acid phosphatase as well as, more recently, angiogenic antigens such as vascular endothelial growth factor have been examined for therapy. Prostate-specific membrane antigen, a type II integral membrane glycoprotein initially characterized by the monoclonal antibody 7E11, has shown promise. Recent evidence suggests that prostate-specific membrane antigen is also expressed in tumor-associated neovasculature of a wide variety of malignant neoplasms. With its expression in prostate secretory-acinar epithelium and the prostate and in the neovasculature associated with tumors, prostate-specific membrane antigen represents an excellent antigenic target for monoclonal antibody diagnostic and therapeutic options. As research continues, the role of monoclonal antibody imaging and therapy will become increasingly important in the management of prostate cancer.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antigens, Neoplasm/immunology , Antigens, Surface/immunology , Prostate-Specific Antigen/immunology , Prostatic Neoplasms/diagnosis , Prostatic Neoplasms/drug therapy , Antibodies, Monoclonal/immunology , Antibody Specificity , Humans , Male , Organ Specificity
20.
Cancer Res ; 59(20): 5194-201, 1999 Oct 15.
Article in English | MEDLINE | ID: mdl-10537297

ABSTRACT

Cell lines derived from human prostate cancer are regarded as relatively resistant to both radiation-induced clonogenic death and apoptosis. Here we attempted to modulate the response of LNCaP prostate cancer cells to radiation therapy (XRT) by pretreatment with 12-O-tetradecanoylphorbol acetate (TPA), a known apoptogenic agent in LNCaP cells. Using plateau-phase cultures, we investigated the response of these cells to XRT, TPA, and a combination of XRT and TPA. LNCaP irradiation did not result in ceramide generation or apoptosis. However, pretreatment with TPA enabled XRT to generate ceramide via activation of the enzyme ceramide synthase and signal apoptosis. Apoptosis was abrogated by the competitive inhibitor of ceramide synthase, fumonisin B1. Furthermore, when transplanted orthotopically into the prostate of nude mice, LNCaP cells produced tumors that recapitulated the responses of LNCaP cells in vitro. XRT or TPA failed to signal apoptosis in LNCaP tumors, whereas a combination of the two resulted in substantial (20-25%) apoptosis within 24 h. There was an additional benefit associated with this regimen because TPA pretreatment protected the adjacent rectum from radiation-induced apoptosis. This represents the first description of signaling-based therapy designed to overcome one form of radiation resistance expressed preferentially in LNCaP human prostate cancer cells.


Subject(s)
Apoptosis , Oxidoreductases/physiology , Prostatic Neoplasms/radiotherapy , Radiation-Sensitizing Agents/pharmacology , Tetradecanoylphorbol Acetate/pharmacology , Animals , Apoptosis/drug effects , Ceramides/biosynthesis , Enzyme Activation/drug effects , Humans , Male , Mice , Mice, Nude , Oxidoreductases/drug effects , Oxidoreductases/radiation effects , Prostatic Neoplasms/enzymology , Prostatic Neoplasms/pathology , Radiation Tolerance , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL