51
|
Höti N, Li Y, Chen CL, Chowdhury WH, Johns DC, Xia Q, Kabul A, Hsieh JT, Berg M, Ketner G, Lupold SE, Rodriguez R. Androgen receptor attenuation of Ad5 replication: implications for the development of conditionally replication competent adenoviruses. Mol Ther 2007; 15:1495-503. [PMID: 17565351 DOI: 10.1038/sj.mt.6300223] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
Conditionally replication competent adenoviruses (CRAds) represent one of the most intensely studied gene therapy strategies for a variety of malignancies, including prostate cancer. These viruses can be generated by placing a tissue or cancer-specific promoter upstream of one or more of the viral genes required for replication (e.g., E1A, E1B). We report here that E1A inhibits androgen receptor (AR) target gene induction and, correspondingly, activated AR inhibits adenoviral replication. This mutual inhibition appears to be an indirect effect, possibly through competition for shared transcriptional co-activators. The net effect is that the oncolytic effect of prostate-specific CRAds is attenuated by these interactions. Fusion of the E1A to AR ameliorates this inhibition, while enhancing specificity. These findings have significant implications in the development of prostate-specific CRAd therapies.
Collapse
Affiliation(s)
- Naseruddin Höti
- James Buchanan Brady Urology Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland 21287-2101, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
52
|
Thompson VC, Morris TGW, Cochrane DR, Cavanagh J, Wafa LA, Hamilton T, Wang S, Fazli L, Gleave ME, Nelson CC. Relaxin becomes upregulated during prostate cancer progression to androgen independence and is negatively regulated by androgens. Prostate 2006; 66:1698-709. [PMID: 16998820 DOI: 10.1002/pros.20423] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
BACKGROUND Relaxin is a potent peptide hormone normally secreted by the prostate. This study characterized relaxin expression during prostate cancer progression to androgen independence (AI), and in response to androgens. METHODS The prostate cancer cell line, LNCaP, was assayed by microarrays and confirmatory Northern analysis to assess changes in relaxin levels due to androgen treatment and in LNCaP xenografts following castration. Relaxin protein levels were examined by immunohistochemistry (IHC) in tissue microarrays of human prostate cancer samples following androgen ablation. RESULTS Relaxin levels decreased in a time and concentration-dependent manner due to androgens in vitro, and increased in xenografts post-castration. Relaxin increased in radical prostatectomy specimens after 6 months of androgen ablation and in AI tumors, was highest in bone metastases. CONCLUSIONS Relaxin is negatively regulated by androgens in vitro and in vivo, which correlates to clinical prostate cancer specimens following androgen ablation. The role of relaxin in angiogenesis and tissue remodeling suggests it may contribute to prostate cancer progression.
Collapse
Affiliation(s)
- Vanessa C Thompson
- The Prostate Centre, Vancouver General Hospital, Vancouver, British Columbia, Canada
| | | | | | | | | | | | | | | | | | | |
Collapse
|
53
|
Watt F, Watanabe R, Yang W, Agren N, Arvidsson Y, Funa K. A novel MASH1 enhancer with N-myc and CREB-binding sites is active in neuroblastoma. Cancer Gene Ther 2006; 14:287-96. [PMID: 17124508 DOI: 10.1038/sj.cgt.7701012] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Neuroblastoma is one of the most common solid tumors in childhood. With the aim of developing a targeting vector for neuroblastoma, we cloned and characterized an enhancer in the 5'-flanking regions of the MASH1 gene by a random-trap method from a 36 kb cosmid DNA. The enhancer-containing clone was identified by the expression of GFP when transfected into neuroblastoma cell lines. The enhancer-luciferase activity is higher in neuroblastoma cell lines, IMR32, BE2 and SH-SY5Y, compared with those in non-neuroblastoma cell lines, U1242 glioma, N417 small cell lung cancer and EOMA hemangioma. The core enhancer was determined within a 0.2 kb fragment, yielding three- to fourfold higher activity than that of the MASH1 promoter alone in IMR32 and BE2. This area possesses GATA- and CREB-binding sites, as well as the E-box. EMSA on this area demonstrated that CREB/ATF could bind the DNA. Chromatin immunoprecipitation assay revealed that N-myc, CREB, and co-activators CBP and PCAF, but not HDAC1, are bound to the core enhancer at the same time as the co-activators and N-myc bind to the promoter. This supports the idea that the commonly overexpressed genes HASH1 and N-myc are regulated in concert, confirming their importance as prognostic markers or targets for therapy.
Collapse
Affiliation(s)
- F Watt
- Children's Cancer Institute Australia for Medical Research, Randwick, New South Wales, Australia
| | | | | | | | | | | |
Collapse
|
54
|
Hattori Y, Maitani Y. Two-step transcriptional amplification-lipid-based nanoparticles using PSMA or midkine promoter for suicide gene therapy in prostate cancer. Cancer Sci 2006; 97:787-98. [PMID: 16800821 PMCID: PMC11159223 DOI: 10.1111/j.1349-7006.2006.00243.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
A two-step transcriptional amplification system (TSTA) was used to enhance the efficacy of suicide gene therapy for treatment of prostate cancer. We designed a TSTA system and constructed two types of plasmid: one containing GAL4-VP16 fusion protein under the control of a tumor-specific promoter, the other containing luciferase or herpes simplex virus thymidine kinase (HSV-tk) under the control of a synthetic promoter. The TSTA systems using nanoparticles based on lipids were evaluated by measuring the amount of induced luciferase activity as a function of prostate-specific membrane antigen (PSMA) and midkine (Mk) promoters, specific for LNCaP and PC-3 prostate cancer cells, respectively. In LNCaP cells that were PSMA-positive, the TSTA system featuring the PSMA enhancer and promoter exhibited activity that was 640-fold greater than a system consisting of one-step transcription with the PSMA promoter. In contrast, this difference in activity did not occur in PSMA-negative PC-3 cells. In Mk-positive PC-3 cells, the TSTA system with the Mk promoter exhibited a five-fold increase in activity over one-step transcription, but such activity was not induced in Mk-negative LNCaP cells. When using HSV-tk for suicide gene therapy, TSTA systems featuring the PSMA or Mk promoter inhibited in vitro cell growth in the presence of ganciclovir. Furthermore, the TSTA system featuring the Mk promoter suppressed in vivo growth of PC-3 tumor xenografts to a greater extent than one-step transcription. These findings show that TSTA systems can enhance PSMA and Mk promoter activities and selectively inhibit PC-3 cell growth in tumors. This suggests that TSTA systems featuring tumor-specific promoters are suitable for cancer treatment by gene therapy.
Collapse
Affiliation(s)
- Yoshiyuki Hattori
- Institute of Medicinal Chemistry, Hoshi University, Ebara 2-4-41, Shinagawa-ku, Tokyo 142-8501, Japan
| | | |
Collapse
|
55
|
Desai P, Jiménez JA, Kao C, Gardner TA. Future innovations in treating advanced prostate cancer. Urol Clin North Am 2006; 33:247-72, viii. [PMID: 16631463 DOI: 10.1016/j.ucl.2005.12.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Many novel techniques for the treatment of prostate cancer are being aggressively investigated because prostate cancer is prevalent in the population and the current treatments for advanced prostate cancer are woefully inadequate. Although the current treatment options prolong life, most patients will eventually experience local recurrence or develop advanced disease. A greater understanding of the molecular events underlying cancer has enabled investigators to explore gene therapy approaches that are targeted against these molecular events. This article discusses antiangiogenic therapy, immune based therapy, and gene therapy. Any of these experimental modalities could be developed to replace hormone ablation therapy which causes unpleasant side effects, decreases the quality of life of the patient, and only temporarily controls the disease.
Collapse
Affiliation(s)
- Pratik Desai
- Department of Urology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | | | | | | |
Collapse
|
56
|
Russell PJ, Khatri A. Novel gene-directed enzyme prodrug therapies against prostate cancer. Expert Opin Investig Drugs 2006; 15:947-61. [PMID: 16859396 DOI: 10.1517/13543784.15.8.947] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
There is no effective cure for late-stage hormone (androgen) refractory prostate cancer. Although chemotherapy offers palliation to these late-stage patients, it also leads to systemic toxicities leading to poor quality of life. Clearly, the focus is on the development and evaluation of novel biologically relevant alternatives such as cytoreductive gene-directed enzyme prodrug therapy (GDEPT). With the current limitations of effective gene delivery in vivo, the in situ amplification of cytotoxicity due to bystander effects of GDEPT has special attraction for patients with prostate cancer, the prostate being dispensable. This review focuses on the development, application and potential of various GDEPTs for treating prostate cancer. The current status of research related to the issues of enhancement of in situ GDEPT delivery and prostate cancer-specific targeting of vectors (especially viral vectors) is assessed. Finally, the scope and progress of synergies between GDEPT and other treatment modalities, both traditional and alternate, are discussed.
Collapse
Affiliation(s)
- Pamela J Russell
- Oncology Research Centre, Prince of Wales Hospital Sydney, Level 2, Clinical Sciences Building, Barker Street, Randwick, NSW 2031, Australia.
| | | |
Collapse
|
57
|
Chen H, Mutton LN, Prins GS, Bieberich CJ. Distinct regulatory elements mediate the dynamic expression pattern of Nkx3.1. Dev Dyn 2006; 234:961-73. [PMID: 16245334 PMCID: PMC2819389 DOI: 10.1002/dvdy.20596] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Loss of Nkx3.1 function in mice results in defects in prostate development and epithelial hyperplasia, indicating that this gene plays important roles in both the initiation and maintenance of prostate differentiation. In humans, decreased NKX3.1 expression is associated with the progression of prostate cancer. Despite these roles in prostate development and disease, the transcriptional regulation of Nkx3.1 has not been systematically addressed. A reporter gene approach in transgenic mice was used to identify regulatory regions that dictate the expression pattern of Nkx3.1. A 32-kb DNA fragment from the Nkx3.1 locus that specifies the expected expression pattern during embryogenesis and postnatal life has been identified. Deletion analyses demonstrated that cis-regulatory elements that mediate expression in distinct sites are separable. A 5-kb fragment downstream of the Nkx3.1 coding region contains elements that support expression in the prostate and bulbourethral glands, whereas an upstream fragment contains elements that direct expression in somites and testes. Reporter gene expression analyses also revealed several previously unknown sites of Nkx3.1 expression in males, including urethral glands, glandular cells in the urethral diverticulum and basal epithelial cells in the prostate. In addition, these analyses revealed Nkx3.1 expression in female urethral glands. The identification of Nkx3.1 cis-regulatory elements provides a unique starting point to dissect signaling pathways involved in prostate organogenesis and pathogenesis and provides a system to perturb gene expression throughout prostate development.
Collapse
Affiliation(s)
- Hui Chen
- Department of Biological Sciences, University of Maryland Baltimore County, Baltimore, Maryland
| | - Laura N. Mutton
- Department of Biological Sciences, University of Maryland Baltimore County, Baltimore, Maryland
| | - Gail S. Prins
- Department of Urology, University of Illinois at Chicago, 820 Wood Street, M/C 955 Chicago, Illinois
| | - Charles J. Bieberich
- Department of Biological Sciences, University of Maryland Baltimore County, Baltimore, Maryland
- Correspondence to: Charles J. Bieberich, Department of Biological Sciences, University of Maryland Baltimore County, Baltimore, MD 21250.
| |
Collapse
|
58
|
Aggarwal S, Ricklis RM, Williams SA, Denmeade SR. Comparative study of PSMA expression in the prostate of mouse, dog, monkey, and human. Prostate 2006; 66:903-10. [PMID: 16496413 DOI: 10.1002/pros.20413] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
BACKGROUND Intraprostatic PSMA targeted prodrugs/protoxins are under development in our laboratory. Future toxicologic studies of these therapies require identification of animal models that express PSMA within the prostate. METHOD PSMA enzymatic activity and protein expression was determined. PSMA expression in the prostates of mouse, dog, and monkey were compared to humans by real-time PCR analysis. RESULTS No substrate hydrolysis was observed in dog or monkey prostate homogenates. Monkey prostate was negative for PSMA protein expression. No significant PSMA mRNA levels were detected by real time PCR in mouse, dog, or monkey prostate tissue compared to PSMA negative tissues. CONCLUSIONS PSMA is not expressed in any significant amount in the prostates of mouse, beagle dog, or macaque monkeys in this study but is expressed in high levels by human prostate. These non-human species, therefore, are not suitable toxicologic models to assess prostate damage from PSMA-activated intraprostatic prodrug/protoxin therapies.
Collapse
Affiliation(s)
- Saurabh Aggarwal
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University Whiting School of Engineering, Baltimore, Maryland 21231, USA
| | | | | | | |
Collapse
|
59
|
Chapel-Fernandes S, Jordier F, Lauro F, Maitland N, Chiaroni J, de Micco P, Mannoni P, Bagnis C. Use of the PSA enhancer core element to modulate the expression of prostate- and non-prostate-specific basal promoters in a lentiviral vector context. Cancer Gene Ther 2006; 13:919-29. [PMID: 16741521 DOI: 10.1038/sj.cgt.7700966] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Composite promoters combining the prostate-specific antigen (PSA) enhancer core element with promoter elements derived from gene coding for human prostate-specific transglutaminase gene, prostate-specific membrane antigen gene, prostate-specific antigen, rat probasin or phosphoglycerate kinase were characterized for their ability to specifically express the enhanced green fluorescent protein (EGFP) gene in prostate versus non-prostate cancer cell lines when transferred with a human immunodeficiency virus-1-based lentiviral vector. By themselves minimal proximal promoter elements were found to inefficiently promote relevant tissue-specific expression; in all the vectors tested, addition of the PSA enhancer core element markedly improved EGFP expression in LnCaP, a cancer prostate cell line used as a model for prostate cancer. The composite promoter was inactive in HuH7, a hepatocarcinoma cell line used as a model of neighboring non-prostate cancer cells. Among the promoters tested, the combination of the PSA enhancer and the rat probasin promoter showed both high specificity and a strong EGFP expression. Neither a high viral input nor the presence of the cPPT/CTS sequence affected composite promoter behavior. Our data suggest that composite prostate-specific promoters constructed by combining key elements from various promoters can improve and/or confer tissue specific expression in a lentiviral vector context.
Collapse
|
60
|
Kaneko M, Yang W, Matsumoto Y, Watt F, Funa K. Activity of a novel PDGF beta-receptor enhancer during the cell cycle and upon differentiation of neuroblastoma. Exp Cell Res 2006; 312:2028-39. [PMID: 16624290 DOI: 10.1016/j.yexcr.2006.03.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2005] [Revised: 02/27/2006] [Accepted: 03/06/2006] [Indexed: 11/17/2022]
Abstract
PDGF acts as an autocrine and paracrine factor in certain tumors through upregulation of the PDGF beta-receptor expression. In order to elucidate the control mechanism for the receptor expression, we have isolated an enhancer from two P1 clones that together contain a 102 kb NotI region covering the entire human PDGFRB gene. They were partially digested with TspI and cloned into the PDGFRB enhancer trap vector to make a library for identification of enhancers. The digested DNA containing enhancer was identified by expression of GFP when transfected in PDGF beta-receptor expressing cells. One of the enhancer clones was further examined by making several deletion mutants in a luciferase vector. This enhancer was most active in neuroblastoma cells, IMR32 and BE2, but less active in hemangioma and in smooth muscle cell lines. Chip assay revealed that SP1, AP2, and GATA2 bound the enhancer in BE2 cells. Their interaction occurred dependently of the cell cycle and synchronously with their binding to the promoter. Transfection of GATA2 alone or with Ets, which binds adjacent to GATA, resulted in differentiation of BE2 cells in parallel with increased PDGF beta-receptor expression. Furthermore, over-expression of the PDGF beta-receptor in BE2 cells induced neurite extension.
Collapse
Affiliation(s)
- Masaharu Kaneko
- Institute of Biomedicine, Department of Medical Chemistry and Cell Biology, Göteborg University, Box 420, SE-405 30 Gothenburg, Sweden
| | | | | | | | | |
Collapse
|
61
|
Magee JA, Chang LW, Stormo GD, Milbrandt J. Direct, androgen receptor-mediated regulation of the FKBP5 gene via a distal enhancer element. Endocrinology 2006; 147:590-8. [PMID: 16210365 DOI: 10.1210/en.2005-1001] [Citation(s) in RCA: 136] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Androgen signaling via the androgen receptor (AR) transcription factor is crucial to normal prostate homeostasis and prostate tumorigenesis. Current models of AR function are predominantly based on studies of prostate-specific antigen regulation in androgen-responsive cell lines. To expand on these in vitro paradigms, we used the mouse prostate to elucidate the mechanisms through which AR regulates another direct target, FKBP5, in vivo. FKBP5 encodes an immunophilin that has been previously implicated in glucocorticoid and progestin signaling pathways and that likely influences prostate physiology in the presence of androgens. In this work, we show that androgens directly regulate FKBP5 via an interaction between the AR and a distal enhancer located 65 kb downstream of the transcription start site in the fifth intron of the FKBP5 gene. We have found that AR selectively recruits cAMP response element-binding protein to this enhancer. These interactions, in turn, result in chromatin remodeling that affects the enhancer proper but not the FKBP5 locus as a whole. Furthermore, in contrast to prostate-specific antigen-regulatory mechanisms, we show that transactivation of the FKBP5 gene does not rely on a single looping complex to mediate communication between the distal enhancer and proximal promoter. Rather, the distal enhancer complex and basal transcription apparatus communicate indirectly with one another, implicating a regulatory mechanism that has not been previously appreciated for AR target genes.
Collapse
Affiliation(s)
- Jeffrey A Magee
- Department of Pathology, Washington University School of Medicine, St. Louis, Missouri 63110, USA
| | | | | | | |
Collapse
|
62
|
Zeng H, Wu Q, Li H, Wei Q, Lu Y, Li X, Wang F, Zhao F, Ding Z, Yang Y. Construction of prostate-specific expressed recombinant plasmids with high transcriptional activity of prostate-specific membrane antigen (PSMA) promoter/enhancer. ACTA ACUST UNITED AC 2005; 26:215-21. [PMID: 15713827 DOI: 10.1002/j.1939-4640.2005.tb01088.x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
To screen different combinations of prostate-specific membrane antigen (PSMA) promoter/enhancer with the strongest transcriptional activity in prostate-specific cells, we used PSMA regulatory elements to control specific expression of the target gene in gene therapy of prostate adenocarcinoma. PSMA promoter and enhancer DNA sequences were amplified from the LNCaP human prostate cancer cell line by polymerase chain reaction, then recombinant plasmids of the enhanced green fluorescent protein (EGFP: pEGFP-PSMA(Pro), pEGFP-PSMA(E-P), pEGFP-PSMA(E(r)-P), pEGFP-PSMA(E(d)-P), and pEGFP-PSMA(E(t)-P)) were constructed with molecular clonal techniques. At the same time, all experimental cell lines were analyzed for the expression of PSMA with the use of PSMA monoclonal antibody and the ABC immunohistochemical assay kit. After plasmids were transfected via liposome, we observed the expression of the reporter gene (EGFP) under a fluorescent microscope and compared the different levels of EGFP expression with reverse transcriptase polymerase chain reaction and flow cytometry so that we could choose the one with the highest transcriptional activity. Only the LNCaP cell line expressed PSMA positively with immunohistochemical stain. The PSMA promoter/enhancer had transcriptional activity in PSMA(+) cell lines and no activity in PSMA(-) cell lines. PSMA(E-P) achieved the strongest activity in different PSMA promoter/enhancer combinations. We confirmed the specific expression of PSMA in prostate cells again. Similarly, transcriptional activity of the PSMA promoter/enhancer was prostate specific. PSMA(E-P) achieved the strongest transcriptional activity among PSMA promoter/enhancer combinations, which could be used in advanced research for tissue-specific treatment.
Collapse
Affiliation(s)
- Hao Zeng
- Department of Urology, West China Hospital, Sichuan University, 37 Guoxue Xiang Street, Chengdu, Sichuan, 610041, P.R. China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
63
|
Laidler P, Dulińska J, Lekka M, Lekki J. Expression of prostate specific membrane antigen in androgen-independent prostate cancer cell line PC-3. Arch Biochem Biophys 2005; 435:1-14. [PMID: 15680901 DOI: 10.1016/j.abb.2004.12.003] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2004] [Revised: 12/07/2004] [Indexed: 11/19/2022]
Abstract
During the progression of prostate cancer from androgen-dependence or sensitivity to androgen-independence, the overall expression of prostate specific membrane antigen (PSMA) increases with its appearance in plasma membrane. However, surprisingly some androgen-independent metastatic prostate cancer cell lines do not express this protein. Estradiol (E2) and basic fibroblast growth factor (bFGF) due to their recognized and strong involvement in prostate growth, development, and pathology were selected with the aim of restoring the expression of PSMA in markedly dedifferentiated prostate cancer PC-3 cells and in Du 145. E2 (10(-7)-10(-11)M) and bFGF (10ng/ml) stimulated the expression of mRNAs for PSMA (2- to 4-fold increase) that apparently were further translated and processed to its membrane form in LNCaP, PC-3, and Du 145 cells. The values of interaction force between the same anti-PSMA antibodies and all studied cells were almost identical (45-64pN), indicating antigenic similarity of the membrane form of PSMA expressed in LNCaP, PC-3, and Du 145 cells.
Collapse
Affiliation(s)
- Piotr Laidler
- Institute of Medical Biochemistry, Jagiellonian University, Medical College, ul. Kopernika 7, 31-034 Kraków, Poland.
| | | | | | | |
Collapse
|
64
|
Cheng WS, Kraaij R, Nilsson B, van der Weel L, de Ridder CMA, Tötterman TH, Essand M. A novel TARP-promoter-based adenovirus against hormone-dependent and hormone-refractory prostate cancer. Mol Ther 2005; 10:355-64. [PMID: 15294182 DOI: 10.1016/j.ymthe.2004.05.022] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2004] [Accepted: 05/13/2004] [Indexed: 11/19/2022] Open
Abstract
TARP (T cell receptor gamma-chain alternate reading frame protein) is a protein that in males is uniquely expressed in prostate epithelial cells and prostate cancer cells. We have previously shown that the transcriptional activity of a chimeric sequence comprising the TARP promoter (TARPp) and the PSA enhancer (PSAe) is strictly controlled by testosterone and highly restricted to cells of prostate origin. Here we report that a chimeric sequence comprising TARPp and the PSMA enhancer (PSMAe) is highly active in testosterone-deprived prostate cancer cells, while a regulatory sequence comprising PSAe, PSMAe, and TARPp (PPT) has high prostate-specific activity both in the presence and in the absence of testosterone. Therefore, the PPT sequence may, in a gene therapy setting, be beneficial to prostate cancer patients that have been treated with androgen withdrawal. A recombinant adenovirus vector with the PPT sequence, shielded from interfering adenoviral sequences by the mouse H19 insulator, yields high and prostate-specific transgene expression both in cell cultures and when prostate cancer, PC-346C, tumors were grown orthotopically in nude mice. Intravenous virus administration reveals both higher activity and higher selectivity for the insulator-shielded PPT sequence than for the immediate-early CMV promoter. Therefore, we believe that an adenovirus with therapeutic gene expression controlled by an insulator-shielded PPT sequence is a promising candidate for gene therapy of prostate cancer.
Collapse
Affiliation(s)
- Wing-Shing Cheng
- Clinical Immunology, Rudbeck Laboratory, Uppsala University, SE-75185, Uppsala, Sweden
| | | | | | | | | | | | | |
Collapse
|
65
|
Wang XY, Martiniello-Wilks R, Shaw JM, Ho T, Coulston N, Cooke-Yarborough C, Molloy PL, Cameron F, Moghaddam M, Lockett TJ, Webster LK, Smith IK, Both GW, Russell PJ. Preclinical evaluation of a prostate-targeted gene-directed enzyme prodrug therapy delivered by ovine atadenovirus. Gene Ther 2005; 11:1559-67. [PMID: 15343359 DOI: 10.1038/sj.gt.3302308] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Gene-directed enzyme prodrug therapy (GDEPT) based on the Escherichia coli enzyme, purine nucleoside phosphorylase (PNP), provides a novel strategy for treating slowly growing tumors like prostate cancer (CaP). PNP converts systemically administered prodrug, fludarabine phosphate, to a toxic metabolite, 2-fluoroadenine, that kills PNP-expressing and nearby cells by inhibiting DNA, RNA and protein synthesis. Reporter gene expression directed by a hybrid prostate-directed promoter and enhancer, PSMEPb, was assayed after plasmid transfection or viral transduction of prostate and non-CaP cell lines. Androgen-sensitive (AS) LNCaP-LN3 and androgen-independent (AI) PC3 human CaP xenografts in nude mice were injected intratumorally with an ovine atadenovirus vector, OAdV623, that carries the PNP gene under PSMEPb, formulated with cationic lipid for enhanced infectivity. Fludarabine phosphate was then given intraperitoneally for 5 days at 75 mg/m2/day. PNP expression was evaluated by enzymic conversion of its substrate using reverse phase HPLC. OAdV623 showed excellent in vitro transcriptional specificity for CaP cells. In vivo, expression of PNP persisted for > 6 days after OAdV623 injection and a single treatment provided 100% increase in tumor doubling time and > 50% inhibition of tumor growth for both LNCaP-LN3 and PC3 lines, with increased tumor necrosis and apoptosis and decreased tumor cell proliferation. OAdV623 significantly suppressed the growth of AS and AI human CaP xenografts in mice.
Collapse
Affiliation(s)
- X Y Wang
- Oncology Research Centre, Prince of Wales Hospital Clinical School of Medicine, The University of New South Wales, Randwick, Australia
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
66
|
Lee SJ, Zhang Y, Lee SD, Jung C, Li X, Kim HS, Bae KH, Jeng MH, Kao C, Gardner T. Targeting Prostate Cancer with Conditionally Replicative Adenovirus Using PSMA Enhancer. Mol Ther 2004; 10:1051-8. [PMID: 15564137 DOI: 10.1016/j.ymthe.2004.08.028] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2004] [Accepted: 08/30/2004] [Indexed: 12/15/2022] Open
Abstract
Prostate cancer is the second most commonly diagnosed cancer in men and accounts for significant mortality and morbidity in the United States. Initially androgen-dependent, prostate cancer ultimately becomes androgen-independent, which makes the disease extremely difficult to cure. In this study, we examined the use of conditionally replication-competent adenovirus for the treatment of hormone-independent prostate cancer. We utilized PSME, an enhancer element for prostate-specific PSMA expression, to control viral E1A protein expression and achieve exclusive virus replication in prostate. Western blotting confirmed that PSME mediated high E1A protein expression in PSMA-positive, androgen-independent prostate cancer cells (C4-2 and CWR22rv), but was much less active in PSMA-negative cancer cells (PC-3 and A549). Consistent with E1A protein expression, the recombinant adenovirus Ad5-PSME-E1a replicated in C4-2 and CWR22rv almost as efficiently as wild type with low levels of androgen, but its replication was significantly attenuated in PSMA-negative cells. In the in vitro killing assay, Ad5-PSME-E1a lysed all C4-2 and CWR22rv cells 5 days after infection, with minimal effect on PSMA-negative cells. In addition, injections of 1.7 x 10(8) plaque-forming units in a CWR22rv xenograft model in nude mice induced significant tumor growth delay, with a substantial necrotic area. These studies suggest that PSME-driven replication-competent adenovirus may be a new therapeutic modality for prostate cancer patients after hormone ablation therapy.
Collapse
Affiliation(s)
- Sang-Jin Lee
- Department of Urology, Indiana University, Indianapolis, IN 46202, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
67
|
Both GW. Ovine atadenovirus: a review of its biology, biosafety profile and application as a gene delivery vector. Immunol Cell Biol 2004; 82:189-95. [PMID: 15061773 DOI: 10.1046/j.0818-9641.2004.01223.x] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The ovine adenovirus isolate OAdV287 is the prototype of the newly recognized genus of atadenoviruses. Although not as well studied as human mastadenoviruses, a substantial amount of work has now been carried out with this virus and an understanding of its interesting and unique properties is beginning to emerge. In this article the biology and biosafety profile of the virus is reviewed. This knowledge underpins the exploitation of the virus as a gene delivery vector. Its potential as a vaccine vector and its application to the treatment of prostate cancer is summarized and discussed.
Collapse
Affiliation(s)
- Gerald W Both
- CSIRO Molecular Science, North Ryde, NSW 2113, Australia.
| |
Collapse
|
68
|
O'Keefe DS, Bacich DJ, Heston WDW. Comparative analysis of prostate-specific membrane antigen (PSMA) versus a prostate-specific membrane antigen-like gene. Prostate 2004; 58:200-10. [PMID: 14716746 DOI: 10.1002/pros.10319] [Citation(s) in RCA: 73] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
BACKGROUND Currently prostate-specific membrane antigen (PSMA) is showing promise both as an imaging and therapeutic target for occult prostate cancer metastases. First generation antibodies against PSMA are used for the FDA approved Prostascint trade mark monoclonal antibody scan and second generation antibodies are being developed for therapeutic targeting as well as imaging 1. However, there have been reports describing PSMA expression in non-prostatic tissues including kidney, liver, and brain. As we had previously showed the existence of a human PSMA homolog, we set out to determine if this non-prostatic expression was due to expression of the PSMA or another gene. MATERIALS AND METHODS The PSMA homolog (PSMA-like) cDNA was cloned by screening a liver cDNA library. mRNA expression of the PSMA and PSMA-like genes was determined via Northern blot analysis using two different probes and protein expression confirmed in some tissues via Western blot analysis. Transcriptional regulation of the two genes was examined using reporter constructs driving luciferase expression. RESULTS The PSMA-like gene possesses 98% identity to the PSMA gene at the nucleotide level and is expressed in kidney and liver under the control of a different promoter to the PSMA gene. The PSMA gene is expressed in several human tissues and is most abundant in the nervous system and the prostate. CONCLUSION The non-prostatic expression of PSMA should be taken into consideration when designing clinical strategies targeting PSMA.
Collapse
Affiliation(s)
- Denise S O'Keefe
- The Department of Urology, University of Pittsburgh Cancer Institute, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | | | | |
Collapse
|
69
|
Sato M, Johnson M, Zhang L, Zhang B, Le K, Gambhir SS, Carey M, Wu L. Optimization of adenoviral vectors to direct highly amplified prostate-specific expression for imaging and gene therapy. Mol Ther 2004; 8:726-37. [PMID: 14599805 PMCID: PMC2820502 DOI: 10.1016/j.ymthe.2003.08.016] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Gene expression-based imaging coupled to gene therapy will permit the prediction of therapeutic outcome. A significant challenge for successful gene therapy is to achieve a high-level of specific gene expression; however, tissue-specific promoters are weak. We postulate that if the weak activity of tissue-specific promoters can be amplified to the levels of strong viral promoters, which have been successful in preclinical scenarios, while retaining specificity, the therapeutic index of gene therapy can be greatly augmented. With this in mind, we developed a two-step transcriptional activation (TSTA) system. In this two-tiered system, a modified prostate-specific antigen promoter was employed to drive a potent synthetic transcriptional activator, GAL4-VP2. This, in turn, activated the expression of a GAL4-dependent reporter or therapeutic gene. Here we demonstrate that recombinant adenoviral vectors (Ads) in which we have incorporated prostate-targeted TSTA expression cassettes retain cell specificity and androgen responsiveness in cell culture and in animal models, as measured by noninvasive optical bioluminescence imaging. We investigated the mechanism of TSTA in different adenoviral configurations. In one configuration, both the activator and the reporter components are inserted into a single Ad (AdTSTA-FL). The activity of AdTSTA-FL exceeds that of a cytomegalovirus promoter-driven vector (AdCMV-FL), while maintaining tissue specificity. When the activator and reporter components are placed in two separate Ads, androgen induction is more robust than for the single AdTSTA-FL. Based on these findings, we hope to refine the TSTA Ads further to improve the efficacy and safety of prostate cancer gene therapy.
Collapse
Affiliation(s)
- Makoto Sato
- Department of Urology, University of California at Los Angeles School of Medicine, Los Angeles, California 90095
| | - Mai Johnson
- Department of Urology, University of California at Los Angeles School of Medicine, Los Angeles, California 90095
| | - Liqun Zhang
- Department of Biological Chemistry, University of California at Los Angeles School of Medicine, Los Angeles, California 90095
| | - Baohui Zhang
- Department of Urology, University of California at Los Angeles School of Medicine, Los Angeles, California 90095
| | - Kim Le
- Department of Biological Chemistry, University of California at Los Angeles School of Medicine, Los Angeles, California 90095
| | - Sanjiv S. Gambhir
- Crump Institute of Molecular Imaging and Department of Molecular and Medical Pharmacology, University of California at Los Angeles School of Medicine, Los Angeles, California 90095
| | - Michael Carey
- Crump Institute of Molecular Imaging and Department of Molecular and Medical Pharmacology, University of California at Los Angeles School of Medicine, Los Angeles, California 90095
- Department of Biological Chemistry, University of California at Los Angeles School of Medicine, Los Angeles, California 90095
| | - Lily Wu
- Department of Urology, University of California at Los Angeles School of Medicine, Los Angeles, California 90095
- Crump Institute of Molecular Imaging and Department of Molecular and Medical Pharmacology, University of California at Los Angeles School of Medicine, Los Angeles, California 90095
- To whom correspondence and reprint requests should be addressed. Fax: (310) 206-5343.
| |
Collapse
|
70
|
Lee SJ, Lee K, Yang X, Jung C, Gardner T, Kim HS, Jeng MH, Kao C. NFATc1 with AP-3 site binding specificity mediates gene expression of prostate-specific-membrane-antigen. J Mol Biol 2003; 330:749-60. [PMID: 12850144 DOI: 10.1016/s0022-2836(03)00640-5] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Prostate-specific-membrane-antigen (PSMA) is a marker protein expressed primarily in prostate epithelium. Its prostate-specific expression is conferred by PSMA enhancer (PSME), localized within the third intron of PSMA-encoding gene FOLH1. We recently reported that the 5'-end 90 bp of PSME harbored crucial enhancer elements for high PSMA expression. Deletion of this 90 bp sequence, called PSME(del3), significantly diminished PSME activity. We have further analyzed the regulatory elements in this 90 bp by transient transfection of linker scanning mutants. Two mutants, LN17 and 18, which harbored an AP-1 site and an AP-3 site, respectively, exhibited significantly lower enhancer activity. Subsequent site-directed mutagenesis changing the AP-3 site abolished the enhancer activity of PSME but not AP-1, indicating that AP-3 was the key cis-element enabling high PSMA expression. In addition, a 12 bp AP-3 site was able to enhance PSME(del3) activity by almost 40% higher compared to full-length PSME. However, AP-3 alone retained just the basal level of activity, indicating that the action by AP-3 was mediated by cooperation with other transcription factors binding to the PSME(del3) region. Transcription factor NFATc1 isoforms in nuclear extract were co-precipitated with the biotinylated AP-3 site by immobilized agarose beads and the genomic DNA containing PSME was precipitated by antibodies reactive to NFATc1, demonstrating that NFATc1 isoforms bound to the AP-3 site in PSME in vivo. Furthermore, ionomycin (calcium ionophore) and TPA augmented the enhancer activity of PSME, implying that calcium is an important regulator for PSMA expression in prostate cancer cell.
Collapse
Affiliation(s)
- Sang-Jin Lee
- Department of Urology, Indiana University, Indianapolis, IN 46202, USA
| | | | | | | | | | | | | | | |
Collapse
|
71
|
Abstract
Cancer gene therapy has been one of the most exciting areas of therapeutic research in the past decade. In this review, we discuss strategies to restrict transcription of transgenes to tumour cells. A range of promoters which are tissue-specific, tumour-specific, or inducible by exogenous agents are presented. Transcriptional targeting should prevent normal tissue toxicities associated with other cancer treatments, such as radiation and chemotherapy. In addition, the specificity of these strategies should provide improved targeting of metastatic tumours following systemic gene delivery. Rapid progress in the ability to specifically control transgenes will allow systemic gene delivery for cancer therapy to become a real possibility in the near future.
Collapse
Affiliation(s)
- Tracy Robson
- School of Biomedical Sciences, University of Ulster, Newtownabbey, Co. Antrim, BT37 0QB, Northern Ireland, UK
| | - David G. Hirst
- School of Biomedical Sciences, University of Ulster, Newtownabbey, Co. Antrim, BT37 0QB, Northern Ireland, UK
| |
Collapse
|
72
|
Denmeade SR, Sokoll LJ, Dalrymple S, Rosen DM, Gady AM, Bruzek D, Ricklis RM, Isaacs JT. Dissociation between androgen responsiveness for malignant growth vs. expression of prostate specific differentiation markers PSA, hK2, and PSMA in human prostate cancer models. Prostate 2003; 54:249-57. [PMID: 12539223 DOI: 10.1002/pros.10199] [Citation(s) in RCA: 102] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
BACKGROUND A detailed understanding is evolving as to how androgen receptor (AR) functions as a transcriptional regulator via its binding to androgen response elements (ARE) within promoter and enhancer regions of prostate-specific differentiation markers such as PSA, hK2, and PSMA. It has been assumed that an understanding of regulation of expression of these marker proteins would also provide an understanding of the mechanisms whereby AR interactions regulate proliferation and survival of malignant prostate cells. In order to validate this hypothesis, we used a series of human prostate cancer models [i.e., LAPC-4, CWR22Rv1, MDA PCA-2b, LNCaP, and C4-2B (derived from LNCaP)] to test whether there is a consistent concordance between androgen responsive regulation for malignant growth vs. regulation of expression of prostate differentiation specific markers PSA, hK2, and PSMA. METHODS In order to define androgen growth responsiveness in vivo, human prostate cancer cell lines were inoculated as xenografts into intact vs. surgically castrated adult male nude mice and the subsequent tumor growth response monitored. To assess androgen regulation of PSA and hK2 expression in these cell lines, the concentration of PSA and hK2 in the conditioned standard media and charcoal stripped media +/- androgen from each cell line was determined using an immunoassay system. PSMA enzymatic activity was determined using the PSMA substrate (3)H N-acetylaspartylglutamate ((3)H NAAG). RESULTS Wild-type AR expressing LAPC-4 cells are androgen responsive for their in vivo growth. This cell line is also androgen sensitive for the expression of both PSA and hK2 in vitro and express PSMA. CWR22Rv1 cells have a mutated AR and are androgen responsive for growth in vivo and androgen sensitive for hk2 but not PSA expression. CWR22Rv1 produce approximately 1.4-fold more PSA, approximately 18-fold more hK2, and have 21-fold higher PSMA activity than LAPC-4 cells. MDA PCA-2b cells are androgen responsive for growth in vivo and androgen sensitive for PSA expression. MDA PCA-2b cells produce approximately 250-fold more PSA but almost equivalent amounts of hK2 compared to LAPC-4 and have approximately 19-fold higher PSMA activity. Both late passage LNCaP and C4-2B are androgen independent for growth in vivo but remain androgen sensitive for both PSA and hK2 expression. LNCaP cells produce approximately 50-fold more PSA, approximately 35-fold more hK2, and have 28-fold higher PSMA activity compared to LAPC-4. C4-2B cells produce approximately 80-fold higher levels of PSA, approximately 250-fold higher levels of hK2. C4-2B also the highest PSMA activity of the cell lines with 105-fold higher PSMA activity than LAPC-4 and approximately 4-fold higher activity than late passage LNCaP cells. CONCLUSIONS Androgen can coordinately regulate both the tumor growth and expression of prostate specific marker genes as observed for the LAPC-4 human prostate cancer cells. Such coordinated regulation, however, is not universal. In all of the other cell lines, there is a dissociation between androgen responsive regulation of malignant growth vs. regulation of expression of prostate specific markers PSA and hK2. In addition, PSMA activity in these cell lines increases as cells become more androgen independent for growth in vivo. These results emphasize that tumor growth and the expression of the specific secretory genes are independently regulated molecular events even if they share a requirement for androgen and/or AR function. Additional independent mechanisms occur in prostate cancer cells for regulation of expression for even the highly related PSA and hK2 genes. Further studies are needed to clarify the mechanisms for androgen ligand-independent, AR-dependent regulation of the genes that directly effect the growth of androgen (i.e., ligand) independent prostate cancer cells. Unfortunately, the data in this present report do not validate the use of the PSA or hK2 gene as surrogates for a model system for such critically important mechanistic studies. Prostate prostate cancer cells. Unfortunately, the data in this present report do not validate the use of the PSA or hK2 gene as surrogates for a model system for such critically important mechanistic studies.
Collapse
Affiliation(s)
- Samuel R Denmeade
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, Maryland 21231, USA.
| | | | | | | | | | | | | | | |
Collapse
|
73
|
Adenovirus Mediated Prostate Specific Enzyme Prodrug Gene Therapy Using Prostate Specific Antigen Promoter Enhanced by the Cre-loxP System. J Urol 2002. [DOI: 10.1097/00005392-200212000-00097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
74
|
Yoshimura I, Ikegami S, Suzuki S, Tadakuma T, Hayakawa M. Adenovirus mediated prostate specific enzyme prodrug gene therapy using prostate specific antigen promoter enhanced by the Cre-loxP system. J Urol 2002; 168:2659-64. [PMID: 12442005 DOI: 10.1016/s0022-5347(05)64239-5] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Tissue or tumor specific gene delivery is crucial for achieving successful results in suicide gene therapy. Prostate specific antigen (PSA) promoter is known to be highly specific in prostate tissue but its promoter activity is much weaker than that of constitutive viral promoters. In the current study we enhanced PSA promoter activity by combining it with the Cre-loxP system. We also applied this system to adenovirus mediated suicide gene therapy with the cytosine deaminase (CD) gene. MATERIALS AND METHODS The Cre-loxP DNA recombination system was used to enhance PSA promoter. A plasmid with the PSA promoter-enhancer combination was constructed to drive Cre recombinase. Another plasmid contained the cytomegalovirus promoter-loxP-flanked stop signal-luciferase gene. LNCaP human prostate cancer cells were co-transfected with these 2 plasmids and luciferase activity was measured to assess promoter activities. Adenoviral vectors with the CD suicide gene were constructed in similar fashion and tested in LNCaP cells in in vitro/in vivo prostate cancer models. RESULTS Promoter activity of the combined PSA promoter/enhancer and Cre-loxP system was 3 times stronger than that of PSA promoter/enhancer alone. It was further enhanced 7-fold in the presence of testosterone. Application of this system to CD suicide gene therapy by adenoviral vectors inhibited subcutaneous LNCaP tumor growth in nude mice. CONCLUSIONS Combining the Cre-loxP system with PSA promoter/enhancer amplified promoter activity and was found to inhibit the growth of PSA producing prostate cancer cells in vivo.
Collapse
Affiliation(s)
- Ichiro Yoshimura
- Department of Urology, National Defence Medical College, Tokorozawa, Saitama, Japan
| | | | | | | | | |
Collapse
|
75
|
Lee SJ, Kim HS, Yu R, Lee K, Gardner TA, Jung C, Jeng MH, Yeung F, Cheng L, Kao C. Novel prostate-specific promoter derived from PSA and PSMA enhancers. Mol Ther 2002; 6:415-21. [PMID: 12231179 DOI: 10.1006/mthe.2002.0682] [Citation(s) in RCA: 68] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The expression of prostate-specific membrane antigen (PSMA) and prostate-specific antigen (PSA), two well characterized marker proteins, remains highly active in the hormone refractory stage of prostate cancer. In this study, an artificial chimeric enhancer (PSES) composed of two modified regulatory elements controlling the expression of PSA and PSMA genes was tested for its promoter activity and tissue specificity using the reporter system. As a result, this novel PSES promoter remained silent in PSA- and PSMA-negative prostate and non-prostate cancer cell lines, but mediated high levels of luciferase in PSA- and PSMA-expressing prostate cancer cell lines in the presence and absence of androgen. To determine whether PSES could be used for in vivo gene therapy of prostate cancer, a recombinant adenovirus, Ad-PSES-luc, was constructed. Luciferase activity in prostate cancer cell lines mediated by Ad-PSES-luc was 400- to 1000-fold higher than in several other non-prostate cell lines, suggesting the high tissue-specificity of the PSES promoter in an adenoviral vector. Finally, recombinant virus Ad-PSES-luc was injected into mice to evaluate the tissue-discriminatory promoter activity in an experimental animal. Unlike Ad-CMV-luc, the luciferase activity from systemic injection of Ad-PSES-luc was fairly low in all major organs. However, when injected into prostate, Ad-PSES-luc drove high luciferase activity almost exclusively in prostate and not in other tissues. Our results demonstrated the potential use of PSES for the treatment of androgen-independent prostate cancer patients.
Collapse
Affiliation(s)
- Sang-Jin Lee
- Department of Urology, Indiana University, Indianapolis, Indiana 46202, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
76
|
Voeks D, Martiniello-Wilks R, Madden V, Smith K, Bennetts E, Both GW, Russell PJ. Gene therapy for prostate cancer delivered by ovine adenovirus and mediated by purine nucleoside phosphorylase and fludarabine in mouse models. Gene Ther 2002; 9:759-68. [PMID: 12040457 DOI: 10.1038/sj.gt.3301698] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2001] [Accepted: 02/19/2002] [Indexed: 11/09/2022]
Abstract
A gene-directed enzyme pro-drug therapy (GDEPT) based on purine nucleoside phosphorylase (PNP), that converts the prodrug, fludarabine to 2-fluoroadenine, has been described, but studies are limited compared with other GDEPTs. We investigated the in vitro and in vivo efficacies of PNP-GDEPT for treating androgen-independent (AI) prostate cancer. The PNP gene controlled by Rous sarcoma virus (RSV) constitutive promoter was delivered using a recombinant ovine adenovirus vector (OAdV220) that uses a different receptor from human adenovirus type 5. In vitro, OAdV220 provided increased transgene expression over a comparable human Ad5 vector in infected AI, murine RM1 prostate cancer cells. Subsequent in vivo testing was therefore confined to OAdV220. Transduction of RM1 cells with OAdV220 before implantation in immunocompetent mice dramatically inhibited subcutaneous (s.c.) tumor growth when fludarabine phosphate was administered systemically and increased mouse survival in a dose-dependent manner. In tumor-bearing C57BL/6 mice, a single intratumoral injection of OAdV220 produced detectable PNP activity for at least 6 days and with prodrug, retarded the growth of aggressive RM1 s.c. tumors by 35% at day 14. There was a consistent trend to reduction of pre-established intraprostatic RM1 tumors. A similar regimen induced significant therapeutic efficacy in human PC3 xenografts. Thus, ovine adenovirus-mediated GDEPT using the PNP system was effective in vivo against AI prostate cancers, the aggressive murine RM1, and the human PC3 lines. Methods that improve viral dissemination and stimulate the immune system in vivo may further improve efficacy.
Collapse
Affiliation(s)
- D Voeks
- Oncology Research Centre, Prince of Wales Hospital, Sydney, Australia
| | | | | | | | | | | | | |
Collapse
|
77
|
Martiniello-Wilks R, Tsatralis T, Russell P, Brookes DE, Zandvliet D, Lockett LJ, Both GW, Molloy PL, Russell PJ. Transcription-targeted gene therapy for androgen-independent prostate cancer. Cancer Gene Ther 2002; 9:443-52. [PMID: 11961667 DOI: 10.1038/sj.cgt.7700451] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2002] [Indexed: 11/08/2022]
Abstract
The Escherichia coli enzyme (purine nucleoside phosphorylase, PNP) gene is delivered directly into PC3 tumors by one injection of replication-deficient human type-5 adenovirus (Ad5). Expressed PNP converts the systemically administered prodrug, 6MPDR, to a toxic purine, 6MP, causing cell death. We sought to increase the specificity of recombinant Ad vectors by controlling PNP expression with the promoter region from the androgen-dependent, prostate-specific rat probasin (Pb) gene. To increase its activity, the promoter was combined with the SV40 enhancer (SVPb). Cell lines were transfected with plasmids containing both a reporter gene, under SVPb control, and a reference gene cassette to allow normalization of expression levels. Plasmids expressed approximately 20-fold more reporter in prostate cancer than in other cells, but surprisingly, the SVPb element was both androgen-independent and retained substantial prostate specificity. Killing by Ad5-SVPb-PNP vector of cell lines cultured with 6MPDR for 6 days was 5- to 10-fold greater in prostate cancer than in liver or lung cells. In vivo, a single intratumoral injection of Ad5-SVPb-PNP (4 x 10(8) pfu), followed by 6MPDR administration twice daily for 6 days, significantly suppressed the growth of human prostate tumors in nude mice and increased their survival compared to control animals. Thus, the androgen-independent, prostate-targeting Ad5 vector reduces human prostate cancer growth significantly in vitro and in vivo. This first example of an androgen-independent vector points the way toward treatment of emerging androgen-independent prostate cancer in conjunction with hormone ablation therapy at a time when the tumor burden is low.
Collapse
|
78
|
Noss KR, Wolfe SA, Grimes SR. Upregulation of prostate specific membrane antigen/folate hydrolase transcription by an enhancer. Gene 2002; 285:247-56. [PMID: 12039052 DOI: 10.1016/s0378-1119(02)00397-9] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Prostate specific membrane antigen (PSMA), also known as folate hydrolase (FOLH1), is a 100 kDa glycoprotein with elevated expression in prostate epithelial tissue. Expression of PSMA is upregulated as prostate tumor grade increases and is found in the vasculature of many tumors, with no presence in benign tissues. Due to the potential of the regulatory elements of the PSMA promoter and enhancer to be used in gene therapy and as biomarkers for prostate cancer under conditions of androgen ablation during treatment, we sequenced and analyzed the ability of 5.5 kb of PSMA promoter/leader region to promote transcription. A recently discovered enhancer, found in the third intron of the PSMA gene, FOLH1, was also studied. The promoter/leader region sequence provided basal expression in transcription assays, while addition of the enhancer activated transcription 41-fold in transient transfections and 144-fold in stable transfections of the LNCaP prostate cell line. This enhancement of transcription was not found in nonprostate cell lines or prostate cell lines that do not express PSMA. An analysis of the ability of androgens to act via the PSMA promoter/leader region and enhancer to activate transcription in transiently transfected LNCaP cells revealed no significant androgen response using the FOLH1 promoter/leader region and a downregulation of 42% with addition of the enhancer. In stably transfected LNCaP cells, the FOLH1 promoter/leader region produced a 21% downregulation in response to androgens, while addition of the enhancer resulted in a 45% downregulation. These results demonstrate significant upregulation of transcription by the PSMA promoter/enhancer, with specificity for the LNCaP prostate cell line, and downregulation of transcription in response to physiological levels of androgen.
Collapse
Affiliation(s)
- Kenneth R Noss
- Research Service (151), Overton Brooks Veterans Administration Medical Center, Shreveport, LA 71101-4295, USA
| | | | | |
Collapse
|
79
|
Kaminski JM, Nguyen K, Buyyounouski M, Pollack A. Prostate cancer gene therapy and the role of radiation. Cancer Treat Rev 2002; 28:49-64. [PMID: 12027414 DOI: 10.1053/ctrv.2002.0250] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Even though prostate cancer is detected earlier than in the pre-PSA era, prostate cancer is the second leading cause of cancer mortality in the American male. Prostate cancer therapy is not ideal, especially for high-risk localized and metastatic cancer; therefore, investigators have sought new therapeutic modalities such as angiogenesis inhibitors, inhibitors of the cell signaling pathway, vaccines, and gene therapy. Gene therapy has emerged as potential therapy for both localized and systemic prostate cancer. Gene therapy has been shown to work supra-additively with radiation in controlling prostate cancer in vivo. With further technological advances in radiation therapy, gene therapy, and the understanding of prostate cancer biology, gene therapy will potentially have an important role in prostate cancer therapy.
Collapse
Affiliation(s)
- J M Kaminski
- Department of Radiation Oncology, Fox Chase Cancer Center, 7701 Burolme Avenue, Philadelphia, PA 19111, USA.
| | | | | | | |
Collapse
|
80
|
Uchida A, O'Keefe DS, Bacich DJ, Molloy PL, Heston WD. In vivo suicide gene therapy model using a newly discovered prostate-specific membrane antigen promoter/enhancer: a potential alternative approach to androgen deprivation therapy. Urology 2001; 58:132-9. [PMID: 11502468 DOI: 10.1016/s0090-4295(01)01256-0] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
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.
Collapse
Affiliation(s)
- A Uchida
- George M. O'Brien Urology Research Center, Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, Ohio 44195, USA
| | | | | | | | | |
Collapse
|