Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 123
Filter
1.
Oncogene ; 34(22): 2815-22, 2015 May 28.
Article in English | MEDLINE | ID: mdl-25132269

ABSTRACT

DNA damage response (DDR) includes the activation of numerous cellular activities that prevent duplication of DNA lesions and maintain genomic integrity, which is critical for the survival of normal and cancer cells. Specific genes involved in the DDR such as BRCA1/2 and P53 are mutated during prostate cancer progression, while various oncogenic signaling such as Akt and c-Myc are activated, enhancing the replication stress and increasing the genomic instability of cancer cells. These events may render prostate cancer cells particularly sensitive to inhibition of specific DDR pathways, such as PARP in homologous recombination DNA repair and Chk1 in cell cycle checkpoint and DNA repair, creating opportunities for synthetic lethality or synergistic cytotoxicity. Recent reports highlight the critical role of androgen receptor (AR) as a regulator of DDR genes, providing a rationale for combining DNA-damaging agents or targeted DDR inhibitors with hormonal manipulation or AR inhibition as treatment for aggressive disease. The aims of this review are to discuss specific DDR defects in prostate cancer that occur during disease progression, to summarize recent advances in understanding the regulation of DDR in prostate cancer, and to present potential therapeutic opportunities through combinational targeting of the intact components of DDR signaling pathways.


Subject(s)
DNA Damage/genetics , DNA Repair/genetics , Prostatic Neoplasms/genetics , Prostatic Neoplasms/therapy , Animals , Cell Cycle Checkpoints/genetics , DNA Replication , DNA, Neoplasm/biosynthesis , DNA, Neoplasm/genetics , Gene Expression Regulation, Neoplastic , Genomic Instability/genetics , Humans , Male , Receptors, Androgen/physiology
2.
Br J Cancer ; 109(12): 3105-15, 2013 Dec 10.
Article in English | MEDLINE | ID: mdl-24157827

ABSTRACT

BACKGROUND: Glutathione S-transferase mu 3 (GSTM3) has been proven to be downregulated in renal cell carcinoma (RCC). We aimed to characterise the role of GSTM3 and its genetic predisposition on the occurrence and postoperative prognosis of RCC. METHODS: The effect of GSTM3 on RCC aggressiveness was examined using transfection and silencing methods. Glutathione S-transferase mu 3 expression in renal tissues was examined by immunohistochemistry. The associations of rs1332018 (A-63C) and rs7483 (V224I) polymorphisms with RCC risk were examined using 400 RCC patients and 802 healthy controls. The factors contributing to postoperative disease-specific survival of RCC patients were evaluated using the Cox proportional hazard model. RESULTS: Glutathione S-transferase mu 3 silencing increased the invasion and anchorage-independent growth of RCC cell lines. rs1332018 (AC+CC vs AA), which correlated with low expression of GSTM3 in kidney, was associated with RCC risk (odds ratio, 1.446; 95% confidence interval (CI), 1.111-1.882). rs1332018 variants and low GSTM3 expression significantly predicted unfavourable postoperative survivals of RCC patients (P<0.05). rs1332018 variants independently predicted a poor prognosis (hazard ratio, 2.119; 95% CI, 1.043-4.307). CONCLUSION: Glutathione S-transferase mu 3 may function as a tumour suppressor in RCC. rs1332018 genetic variants predispose the host to downregulating GSTM3 expression in kidney, facilitate carcinogenesis, and predict an unfavourable postoperative prognosis of RCC.


Subject(s)
Carcinoma, Renal Cell/enzymology , Carcinoma, Renal Cell/genetics , Glutathione Transferase/genetics , Kidney Neoplasms/enzymology , Kidney Neoplasms/genetics , Carcinoma, Renal Cell/pathology , Case-Control Studies , Female , Gene Expression , Gene Knockdown Techniques , Genetic Predisposition to Disease , Genetic Variation , Glutathione Transferase/biosynthesis , Glutathione Transferase/deficiency , Glutathione Transferase/metabolism , Humans , Immunohistochemistry , Kidney Neoplasms/pathology , Male , Polymorphism, Single Nucleotide , Prognosis , Promoter Regions, Genetic , Risk Factors , Transfection
3.
Oncogene ; 32(49): 5501-11, 2013 Dec 05.
Article in English | MEDLINE | ID: mdl-23752182

ABSTRACT

Prostate cancer is the second-leading cause of cancer-related mortality in men in Western societies. Androgen receptor (AR) signaling is a critical survival pathway for prostate cancer cells, and androgen-deprivation therapy (ADT) remains the principal treatment for patients with locally advanced and metastatic disease. Although a majority of patients initially respond to ADT, most will eventually develop castrate resistance, defined as disease progression despite serum testosterone levels of <20 ng/dl. The recent discovery that AR signaling persists during systemic castration via intratumoral production of androgens led to the development of novel anti-androgen therapies including abiraterone acetate and enzalutamide. Although these agents effectively palliate symptoms and prolong life, metastatic castration-resistant prostate cancer remains incurable. An increased understanding of the mechanisms that underlie the pathogenesis of castrate resistance is therefore needed to develop novel therapeutic approaches for this disease. The aim of this review is to summarize the current literature on the biology and treatment of castrate-resistant prostate cancer.


Subject(s)
Androgen Antagonists/therapeutic use , Prostatic Neoplasms, Castration-Resistant/drug therapy , Receptors, Androgen/metabolism , Castration , Disease Progression , Drug Resistance, Neoplasm , Humans , Male , Neoplasm Metastasis/drug therapy , Receptors, Androgen/genetics
4.
Gene Ther ; 18(10): 969-78, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21512508

ABSTRACT

We previously identified the mouse and human Glipr1 and GLIPR1/RTVP-1 genes, respectively, as direct p53 targets with proapoptotic activities in various cancer cell lines, including prostate cancer (PCa). Intratumoral injection of an adenoviral vector capable of efficient transduction and expression of Glipr1 (AdGlipr1) yielded promising therapeutic results in an orthotopic, metastatic mouse model of PCa. AdGlipr1-transduced macrophages (Mφ/Glipr1) generated greater surface expression of CD40, CD80 and major histocompatibility complex class II molecules and greater production of interleukin 12 (IL-12) and IL-6 in vitro than control macrophages did. Mechanistic analysis indicated that increased production of IL-12 in Mφ/Glipr1 depends on activation of the p38 signaling cascade. Mφ/Glipr1 injected into orthotopic 178-2BMA tumors in vivo resulted in significantly suppressed prostate tumor growth and spontaneous lung metastases and longer survival relative to those observed in control-treated mice. Furthermore, these preclinical data indicate the generation of systemic natural killer cell activity and tumor-specific cytotoxic T lymphocyte responses. Trafficking studies confirmed that intratumorally injected Mφ/Glipr1 could migrate to draining lymph nodes. Overall, our data suggest that this novel gene-modified cell approach is an effective treatment avenue that induces antitumor immune responses in preclinical studies.


Subject(s)
Genetic Therapy/methods , Macrophages/metabolism , Neoplasm Metastasis/prevention & control , Prostatic Neoplasms/genetics , Prostatic Neoplasms/therapy , Proteins/genetics , Adenoviridae , Animals , B7-1 Antigen/metabolism , CD40 Antigens/metabolism , Genetic Vectors/administration & dosage , Interleukin-12/metabolism , Interleukin-6/metabolism , Killer Cells, Natural/immunology , Kinetics , Male , Mice , Prostatic Neoplasms/immunology , T-Lymphocytes, Cytotoxic/immunology
5.
Prostate Cancer Prostatic Dis ; 13(1): 6-11, 2010 Mar.
Article in English | MEDLINE | ID: mdl-19581923

ABSTRACT

Caveolin-1 (cav-1) is reportedly overexpressed in prostate cancer cells and is associated with disease progression. Specific oncogenic activities of cav-1 associated with Akt activation also occur in prostate cancer. A membrane-associated protein, cav-1, is nonetheless secreted by prostate cancer cells; results of recent studies showed that secreted cav-1 can stimulate cell survival and angiogenic activities, defining a role for cav-1 in the prostate cancer microenvironment. Serum cav-1 levels were also higher in prostate cancer patients than in control men without prostate cancer, and the preoperative serum cav-1 concentration had prognostic potential in men undergoing radical prostatectomy. Secreted cav-1 is therefore a potential biomarker and therapeutic target for prostate cancer.


Subject(s)
Caveolin 1/physiology , Prostatic Neoplasms/physiopathology , Biomarkers, Tumor , Caveolin 1/blood , Caveolin 1/metabolism , Disease Progression , Humans , Male , Neovascularization, Pathologic , Prognosis , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Up-Regulation
6.
Prostate Cancer Prostatic Dis ; 12(3): 301-9, 2009.
Article in English | MEDLINE | ID: mdl-19104507

ABSTRACT

We evaluated the potential use of intraoperative gelatin matrix hemostatic sealant (GMHS; FloSeal; Baxter Healthcare) embedded with macrophages (Mphi) transduced with murine interleukin (IL)-12 recombinant adenoviral vector (G/Mphi/AdmIL-12) for prevention of recurrence of prostate cancer following radical prostatectomy. Application of G/Mphi/AdmIL-12 resulted in significant suppression of tumor growth and spontaneous lung metastases, a statistically significant survival advantage of the G/Mphi/AdmIL-12-treated animals, more efficient trafficking of Mphi to lymph nodes draining from the prostate and generation of systemic natural killer cell activity and tumor-specific cytolytic T lymphocyte responses compared to the controls in a preclinical mouse model of residual prostate cancer. Our data recommend this treatment as a novel adjuvant for prevention of local recurrence of prostate cancer following radical prostatectomy.


Subject(s)
Genetic Therapy , Interleukin-12/genetics , Macrophages/physiology , Prostatic Neoplasms/therapy , Adenoviridae/genetics , Animals , Cell Movement , Cell Survival/drug effects , Disease Models, Animal , Gelatin , Hemostatics/pharmacology , Interleukin-12/immunology , Macrophages/immunology , Male , Mice , Prostatic Neoplasms/mortality , Prostatic Neoplasms/pathology
7.
Gene Ther ; 15(10): 787-96, 2008 May.
Article in English | MEDLINE | ID: mdl-18385769

ABSTRACT

There is a critical need to develop new and effective cancer therapies that target bone, the primary metastatic site for prostate cancer and other malignancies. Among the various therapeutic approaches being considered for this application, gene-modified cell-based therapies may have specific advantages. Gene-modified cell therapy uses gene transfer and cell-based technologies in a complementary fashion to chaperone appropriate gene expression cassettes to active sites of tumor growth. In this paper, we briefly review potential cell vehicles for this approach and discuss relevant gene therapy strategies for prostate cancer. We further discuss selected studies that led to the conceptual development and preclinical testing of IL-12 gene-modified bone marrow cell therapy for prostate cancer. Finally, we discuss future directions in the development of gene-modified cell therapy for metastatic prostate cancer, including the need to identify and test novel therapeutic genes such as GLIPR1.


Subject(s)
Bone Marrow Transplantation/methods , Bone Neoplasms/secondary , Bone Neoplasms/therapy , Genetic Therapy/methods , Prostatic Neoplasms/therapy , Bone Marrow Cells/immunology , Genetic Engineering , Humans , Immunotherapy/methods , Interleukin-12/genetics , Male , Oncolytic Virotherapy/methods
8.
Cancer Gene Ther ; 14(10): 819-27, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17627292

ABSTRACT

To investigate the immunomodulatory effects of interleukin-12 (IL-12) for treatment of metastatic prostate cancer, we administered adult bone marrow cells (BMC) that were genetically modified by retroviral vector-mediated IL-12 gene transduction in an experimental mouse model of prostate cancer metastasis. This therapy produced significant anti-metastatic effects in bone and lung and prolonged animal survival. Flow cytometric analysis indicated donor BMC could effectively home to bone and lung after treatment. Intensive infiltration of CD4 and CD8T cells in lung metastases and increased systemic natural killer and cytotoxic T lymphocyte activities indicated induction of a significant anti-metastatic immune response after treatment with IL-12 transduced BMC. Our results demonstrate the therapeutic potential of gene-modified BMC gene therapy.


Subject(s)
Bone Marrow Cells/physiology , Bone Neoplasms/prevention & control , Disease Models, Animal , Interleukin-12/genetics , Lung Neoplasms/prevention & control , Prostatic Neoplasms/prevention & control , Animals , Bone Neoplasms/immunology , Bone Neoplasms/secondary , Gene Expression , Genetic Vectors , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Killer Cells, Natural/immunology , Lung Neoplasms/immunology , Lung Neoplasms/secondary , Male , Mice , Prostatic Neoplasms/immunology , Prostatic Neoplasms/pathology , Retroviridae/genetics , Survival Rate , T-Lymphocytes, Cytotoxic/immunology , Transduction, Genetic
9.
Gene Ther ; 14(3): 227-36, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17024109

ABSTRACT

We investigated the potential benefits of combining adenoviral vector mediated in situ interleukin-12 (AdmIL-12) gene therapy with radiation therapy (XRT) to enhance therapeutic efficacy. In a metastatic mouse prostate cancer cell line, 178-2 BMA, AdmIL-12+XRT demonstrated enhanced therapeutic activities in vitro as determined by clonogenic survival, apoptosis, and mIL-12 levels. At the molecular level, increased expression of tumor necrosis factor-alpha mRNA was specific for the combined therapy. In a subcutaneous 178-2 BMA in vivo model, the combination of AdmIL-12+XRT produced statistically significant tumor growth suppression compared to control vector Adbetagal, Adbetagal XRT, or AdmIL-12 as monotherapy. In addition, significant prolongation of survival was demonstrated for the combination of AdmIL-12+XRT. The combination of AdmIL-12+XRT significantly suppressed both spontaneous and pre-established lung metastases, and led to a prolonged elevation of serum IL-12 and significantly increased natural killer (NK) activities. Importantly, in vivo depletion of NK cells resulted in significant attenuation of the antimetastatic activities of AdmIL-12 alone or AdmIL-12+XRT. These combined effects suggest that AdIL-12 gene therapy together with radiotherapy may achieve maximal tumor control (both local and systemic) in selected prostate cancer patients via radio-gene therapy induced local cytotoxicity and local and systemic antitumor immunity.


Subject(s)
Adenoviridae/genetics , Genetic Therapy/methods , Genetic Vectors/administration & dosage , Interleukin-12/genetics , Lung Neoplasms/secondary , Prostatic Neoplasms/therapy , Animals , Cell Line, Tumor , Combined Modality Therapy , Genetic Vectors/genetics , Humans , Injections, Intralesional , Interleukin-12/blood , Killer Cells, Natural/immunology , Lung Neoplasms/radiotherapy , Lung Neoplasms/therapy , Male , Mice , Mice, Mutant Strains , Prostatic Neoplasms/immunology , Prostatic Neoplasms/radiotherapy , Transduction, Genetic/methods , Tumor Necrosis Factor-alpha/metabolism
10.
Cancer Gene Ther ; 13(7): 658-63, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16485011

ABSTRACT

We previously identified a novel p53 target gene, RTVP-1, that possesses unique cytotoxic and immunostimulatory activities which make it potentially useful for cancer gene therapy. To test the therapeutic potential of RTVP-1 in a gene-modified tumor cell-based vaccine model, we used an adenoviral vector capable of efficient transduction and expression of RTVP-1 (AdRTVP-1), together with a highly metastatic mouse prostate cancer cell line (178-2 BMA). A vaccine was prepared with 178-2 BMA cells transduced with AdRTVP-1 or a control adenoviral vector expressing beta-galactosidase (Adbetagal). After irradiation of the cells, syngeneic 129/Sv mice were vaccinated three times at weekly intervals. After 3 weeks, they were challenged with orthotopic 178-2 BMA cells. After 21 days, fewer than 60% of the RTVP-1-cell-vaccinated mice developed tumors compared to 100% of the control mice. The RTVP-1-cell vaccine significantly reduced primary tumor wet weight compared with control Adbetagal-cell vaccine (P<0.0001 at 7 and 14 days). Experimental metastasis to lung was also significantly reduced (P=0.0377), and survival significantly increased (P=0.0002). In addition, significantly increased NK and CTL activities were demonstrated in the AdRTVP-1-cell-vaccinated mice. These findings indicate that RTVP-1 gene-modified cell-based vaccines may be useful in the prevention of recurrent prostate cancer.


Subject(s)
Adenoviridae , Cancer Vaccines/therapeutic use , Neoplasm Proteins , Nerve Tissue Proteins , Prostatic Neoplasms/prevention & control , Animals , Cancer Vaccines/genetics , Cancer Vaccines/immunology , Cell Line, Tumor , Humans , Male , Membrane Proteins , Mice , Neoplasm Proteins/immunology , Neoplasms, Experimental/genetics , Neoplasms, Experimental/immunology , Neoplasms, Experimental/pathology , Neoplasms, Experimental/prevention & control , Nerve Tissue Proteins/immunology , Prostatic Neoplasms/genetics , Prostatic Neoplasms/immunology , Prostatic Neoplasms/pathology , Transduction, Genetic
11.
Cancer Gene Ther ; 13(1): 91-8, 2006 Jan 01.
Article in English | MEDLINE | ID: mdl-16052232

ABSTRACT

We developed a preclinical prostate cancer model to study the feasibility of adoptive immunotherapy for residual tumor following neo-adjuvant in situ adenoviral-vector-mediated interleukin 12 (AdIL-12) gene therapy. Splenocytes were obtained from mice with orthotopic 178-2 BMA metastatic mouse prostate cancers treated previously with AdIL-12, or a vector with the IL-12 genes plus the costimulatory gene B7-1 (AdIL-12/B7), or a control gene (Adbetagal). The splenocytes were subsequently injected intravenously into syngeneic mice bearing orthotopic 178-2 BMA tumors generated 3 days previously. Significant orthotopic tumor growth suppression was achieved with splenocytes derived from mice whose tumors had been injected with AdIL-12 compared to splenocytes from control Adbetagal mice (P = 0.0005) and splenocytes from AdIL-12/B7-treated mice significantly suppressed spontaneous lung metastases compared to splenocytes from control mice (P = 0.0356). Adoptive transfer of splenocytes from either AdIL-12 (P = 0.004) or AdIL-12/B7 (P = 0.009)-treated mice significantly prolonged survival relative to controls. Transfer of NK and tumor-specific CTL activities was detected and depletion of CD4+ and CD8+ T cells by in vitro antibody-mediated complement lysis of the splenocytes prior to injection abrogated the effects. Systemic IL-12 administration delivered by intramuscular AdIL-12 injection enhanced the antitumor effects of adoptive splenocyte transfer and boosted the CTL response. Our data provide evidence that this form of adoptive immunotherapy can enhance the effectiveness of neo-adjuvant in situ IL-12 gene therapy in cases of persistent malignancy.


Subject(s)
Adoptive Transfer/methods , Genetic Therapy , Interleukin-12/metabolism , Prostatic Neoplasms/therapy , Spleen/metabolism , Animals , B7-1 Antigen/metabolism , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/metabolism , Immunotherapy , Male , Mice , Neoplasm Metastasis , Prostatic Neoplasms/metabolism , Spleen/cytology , T-Lymphocytes/metabolism , Time Factors
12.
Prostate Cancer Prostatic Dis ; 8(2): 108-18, 2005.
Article in English | MEDLINE | ID: mdl-15724144

ABSTRACT

The PI3-K-Akt pathway plays a central role in the development and progression of prostate cancer and other malignancies. We review original studies and summarize relevant sections of previous reviews concerning the relationships between abnormalities in the PI3-K-Akt pathway and prostate cancer progression. We discuss laboratory and clinical data that indicate gene perturbation and dysregulation of PI3-K-Akt pathway is common in prostate cancer and other malignancies. We further discuss the critical role of the PI3-K-Akt pathway in the oncogenic signaling network and provide examples that establish the PI3-K-Akt pathway as a focal point for the future development of informative biomarkers and effective therapies for prostate cancer.


Subject(s)
Gene Expression Regulation, Neoplastic , Phosphatidylinositol 3-Kinases/physiology , Prostatic Neoplasms/enzymology , Prostatic Neoplasms/physiopathology , Protein Serine-Threonine Kinases/physiology , Proto-Oncogene Proteins/physiology , Biomarkers, Tumor/analysis , Disease Progression , Humans , Male , Phosphatidylinositol 3-Kinases/biosynthesis , Phosphatidylinositol 3-Kinases/genetics , Protein Serine-Threonine Kinases/biosynthesis , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins/biosynthesis , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-akt , Signal Transduction
13.
Prostate Cancer Prostatic Dis ; 5(4): 316-25, 2002.
Article in English | MEDLINE | ID: mdl-12627218

ABSTRACT

Adenoviral vector delivery of the Herpes simplex virus thymidine kinase (HSV-tk) gene in combination with the prodrug ganciclovir (GCV) has been tested in phase I clinical trials for prostate cancer and found to exhibit a satisfactory toxicity profile. We have developed additional adenoviral vectors with differing promoters to optimize the expression profile and in the present study evaluate the potential systemic toxicity of these vectors. Four recombinant adenoviral vectors that express the HSV-tk gene were generated using three different promoters: CMV (leftward orientation); RSV (both rightward and leftward orientation); and the mouse caveolin-1 (cav-1) promoter (leftward orientation). Efficacy was determined in vitro by cytotoxicity assays in a mouse prostate cancer cell line, RM-9, and in vivo by treating orthotopic tumors. Potential toxicity was evaluated from liver histology and apoptotic cell counts and enzyme levels in the serum following intravenous adenoviral vector injection. Although there were differences in HSV-tk expression at the protein level among the four vectors there were no significant differences in in-vitro cytotoxicity studies with GCV or in vivo in tumor growth suppression of an orthotopic mouse prostate cancer model in GCV treated mice. Intravenous delivery of high doses of all adenoviral vectors lead to abnormalities in liver function as measured by specific serum markers and histological evaluation of liver tissue and increased levels of apoptosis in the liver. These abnormalities were most prevalent with the vector containing the CMV promoter and the rightward oriented RSV promoter. They were least prevalent in the vector regulated by the cav-1 promoter. Upregulation of specific chemokines, MIP-2 and MIP-1beta was correlated with apoptotic counts. Our results demonstrate that comprehensive toxicological analysis of adenoviral vectors provides internally consistent information that can differentiate vectors with comparable efficacy based on toxicity. In these studies vectors with the cav-1 promoter-driven and leftward RSV-driven HSV-tk gene demonstrated minimal toxicities with cytotoxic effectiveness comparable to more toxic vectors. Our studies further suggest that promoter selection can influence the toxic effects of an adenoviral gene therapy vector.


Subject(s)
Adenocarcinoma/therapy , Adenoviridae/genetics , Avian Sarcoma Viruses/genetics , Caveolins/genetics , Chemokines/biosynthesis , Cytomegalovirus/genetics , Defective Viruses/genetics , Genes, Viral , Genetic Therapy , Genetic Vectors/toxicity , Hepatitis, Viral, Animal/etiology , Promoter Regions, Genetic , Prostatic Neoplasms/therapy , Simplexvirus/genetics , Thymidine Kinase/genetics , Viral Proteins/genetics , Animals , Apoptosis , Caveolin 1 , Chemokine CCL4 , Chemokine CCL5/biosynthesis , Chemokine CCL5/genetics , Chemokine CXCL10 , Chemokine CXCL2 , Chemokines/genetics , Chemokines, CXC/biosynthesis , Chemokines, CXC/genetics , Gene Expression , Gene Expression Regulation, Viral , Genes, Synthetic , Genetic Vectors/genetics , Genetic Vectors/therapeutic use , Injections, Intravenous , Liver Function Tests , Macrophage Inflammatory Proteins/biosynthesis , Macrophage Inflammatory Proteins/genetics , Male , Mice , Mice, Inbred C57BL , Monokines/biosynthesis , Monokines/genetics , Reverse Transcriptase Polymerase Chain Reaction , Simplexvirus/enzymology , Thymidine Kinase/antagonists & inhibitors , Tumor Cells, Cultured/pathology , Tumor Cells, Cultured/virology , Viral Proteins/antagonists & inhibitors
14.
Clin Cancer Res ; 7(12): 4272-9, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11751529

ABSTRACT

Caveolin-1, a structural component of caveolae, is overexpressed in metastatic and androgen-resistant prostate cancer and highly expressed in tumor-associated endothelial cells. The mouse cav-1 promoter was cloned and placed upstream of the HSV-tk gene in an adenoviral vector (Adcav-1tk) and compared with a cytomegalovirus (CMV) or Rous sarcoma virus (RSV) promoter-driven HSV-tk, AdCMVtk and AdRSVtk vectors, respectively. Mouse and human prostate cancer cells and mouse endothelial cells were infected with Adcav-1tk, AdCMVtk or control vectors without the HSV-tk gene (Adcav-1 and AdCMV) and subsequently treated with ganciclovir (GCV). GCV-mediated in vitro cytotoxicity induced by the Adcav-1tk vector was comparable to that for AdCMVtk in multiple mouse and human prostate cancer cell lines. To evaluate the activity of Adcav-1tk in vivo, orthotopic mouse prostate cancer tumors were generated with RM-9 cells and injected in situ with Adcav-1tk, AdCMVtk, AdRSVtk, or AdCMVbetagal (control) and treated with GCV. All three HSV-tk transducing vectors produced statistically significant reductions in wet weight and increased apoptotic indices compared with the control vector. However, only Adcav-1tk produced significant necrosis, and only Adcav-1tk and AdRSVtk caused significant decreases in microvessel density. In conclusion, Adcav-1tk demonstrated efficacy in vitro and in vivo in preclinical models of prostate cancer. Our results suggest that the cav-1 promoter may have unique benefits in targeting gene therapy to prostate cancer and its associated vasculature.


Subject(s)
Adenoviridae/genetics , Caveolins/genetics , Genetic Therapy/methods , Genetic Vectors , Promoter Regions, Genetic , Prostatic Neoplasms/pathology , Animals , Caveolin 1 , Cell Survival , Humans , Male , Mice , Mice, Inbred C57BL , Prostatic Neoplasms/genetics , Simplexvirus/genetics , Thymidine Kinase/genetics , Tumor Cells, Cultured
15.
Expert Opin Biol Ther ; 1(3): 481-95, 2001 May.
Article in English | MEDLINE | ID: mdl-11727520

ABSTRACT

The development of effective treatments for prostate cancer is thwarted by the natural history of the disease. The biological and clinical potential of most individual cancers is uncertain. In many cases the disease will not progress to clinical significance but experimental and clinical studies indicate that prostate cancer can and may metastasis early in the course of the disease from relatively small foci (i.e., not necessarily the largest or index cancer). Localised prostate cancer is potentially curable with localised therapies (radical prostatectomy or irradiation therapy). However, there are no curative therapies for metastatic prostate cancer. Gene therapy, especially those approaches with an immunomodulatory component, may provide additional therapeutic options with the potential to affect both localised and systemic disease. We have pioneered the development and application of in situ gene therapy protocols using adenoviral vectors to transduce specific genes that generate cytotoxic activity and/or a systemic antitumour immune response. In addition we have completed initial studies that demonstrate the therapeutic potential of adenoviral vector-mediated gene modified cell-based vaccines. Our review discusses preclinical studies focused on the development of immunostimulatory in situ gene therapy approaches that hopefully will provide novel and effective treatments for localised and metastatic prostate cancer.


Subject(s)
Genetic Therapy/methods , Prostatic Neoplasms/therapy , Animals , Humans , Male , Mice , Prostatic Neoplasms/pathology , Prostatic Neoplasms/secondary
16.
Oncogene ; 20(46): 6707-17, 2001 Oct 11.
Article in English | MEDLINE | ID: mdl-11709705

ABSTRACT

Numerous LOH and mutation analysis studies in different tumor tissues, including prostate, indicate that there are multiple tumor suppressor genes (TSGs) present within the human chromosome 8p21-22 and 10q23-24 regions. Recently, we showed that LZTS1 (or FEZ1), a putative TSG located on 8p22, has the potential to function as a cell growth modulator. We report here the cloning, gene organization, cDNA sequence characterization and expression analysis of LAPSER1, an LZTS1-related gene. This gene maps within a subregion of human chromosome 10q24.3 that has been reported to be deleted in various cancers, including prostate tumors, as frequently as the neighboring PTEN locus. The complete LAPSER1 cDNA sequence encodes a predicted protein containing various domains resembling those typically found in transcription factors (P-Box, Q-rich and multiple leucine zippers). LAPSER1 is expressed at the highest levels in normal prostate and testis, where multiple isoforms are seen, some of which are either undetectable or differentially expressed in some prostate tumor tissues and cell lines. Over-expression of LAPSER1 cDNA strongly inhibited cell growth and colony-forming efficiencies of most cancer cells assessed. Together these data suggest that LAPSER1 is another gene involved in the regulation of cell growth whose loss of function may contribute to the development of cancer.


Subject(s)
Chromosomes, Human, Pair 10 , DNA-Binding Proteins/genetics , DNA-Binding Proteins/physiology , Genes, Tumor Suppressor , Tumor Suppressor Proteins , Amino Acid Sequence , Animals , Blotting, Northern , Cell Cycle Proteins , Cell Division , Cell Line , Chromosome Mapping , Cloning, Molecular , DNA Mutational Analysis , DNA, Complementary/metabolism , Databases as Topic , Gene Deletion , Gene Expression Regulation, Neoplastic , Humans , Loss of Heterozygosity , Male , Models, Genetic , Molecular Sequence Data , Prostate/metabolism , Prostatic Neoplasms/metabolism , Protein Structure, Tertiary , Rats , Reverse Transcriptase Polymerase Chain Reaction , Sequence Homology, Amino Acid , Testis/metabolism , Tissue Distribution , Transfection
17.
Hum Gene Ther ; 12(16): 1955-67, 2001 Nov 01.
Article in English | MEDLINE | ID: mdl-11686937

ABSTRACT

In an extended phase I/II study we evaluated 36 prostate cancer patients with local recurrence after radiotherapy who received single or repeated cycles of replication-deficient adenoviral vector (ADV)-mediated herpes simplex virus-thymidine kinase (HSV-tk) plus ganciclovir (GCV) in situ gene therapy with respect to serum PSA levels, alterations in immune cells, and numbers of apoptotic cells in needle biopsies. An initial cycle of HSV-tk plus GCV gene therapy caused a significant prolongation of the mean serum PSA-doubling time (PSADT) from 15.9 to 42.5 months (p = 0.0271) and in 28 of the injected patients (77.8%) there was a mean PSA reduction (PSAR) of 28%. It took a mean of 8.5 months for the PSA to return to the initial PSA (TR-PSA) value. A repeated cycle of gene therapy failed to significantly extend PSADT but did result in significant increases in PSAR (29.4%) and TR-PSA (10.5 months). Moderately increased serum adenovirus antibody titers were generally observed 2 weeks after initial vector injection. Also at this time there was a statistically significant increase in the mean percent of CD8(+) T cells positive for the HLA-DR marker of activation in peripheral blood (p = 0.0088). Studies using prostate biopsies obtained at the same time point demonstrated that vector DNA was detectable by PCR in most samples yet all patients remained positive for prostate cancer in at least one biopsy core. Further analysis demonstrated a correlation between the level of CD8(+) cells and the number of apoptotic cells in biopsies containing cancer cells (p = 0.042). We conclude that repeated cycles of in situ HSV-tk plus GCV gene therapy can be administered to prostate cancer patients who failed radiotherapy and have a localized recurrence. Biological responses to this experimental therapy including increases in PSADT, PSAR, and TR-PSA, and activated CD8(+) T cells present in the peripheral blood, were demonstrated. Interestingly, the density of CD8(+) cells in posttreatment biopsies correlated with the number of apoptotic cells.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Genetic Therapy , Lymphocyte Activation , Prostate-Specific Antigen/blood , Prostatic Neoplasms/therapy , Adenoviridae/genetics , Aged , Antibodies, Viral/blood , Antiviral Agents/administration & dosage , Base Sequence , DNA Primers , Ganciclovir/administration & dosage , Genetic Vectors , Humans , Immunophenotyping , Male , Neoplasm Recurrence, Local , Prostatic Neoplasms/immunology , Prostatic Neoplasms/radiotherapy , Simplexvirus/enzymology , Thymidine Kinase/genetics
18.
Int J Radiat Oncol Biol Phys ; 51(4): 1008-17, 2001 Nov 15.
Article in English | MEDLINE | ID: mdl-11704325

ABSTRACT

PURPOSE: Standard therapies for breast cancer lack tumor specificity and have significant risk for recurrence and toxicities. Herpes simplex virus-thymidine kinase (HSV-tk) gene therapy combined with radiation therapy (XRT) may be effective because of complementary mechanisms and distinct toxicity profiles. HSV-tk gene therapy followed by systemic administration of ganciclovir (GCV) enhances radiation-induced DNA damage by generating high local concentrations of phosphorylated nucleotide analogs that increase radiation-induced DNA breaks and interfere with DNA repair mechanisms. In addition, radiation-induced membrane damage enhances the "bystander effect" by facilitating transfer of nucleotide analogs to neighboring nontransduced cells and by promoting local and systemic immune responses. This study assesses the effect of single and multiple courses of HSV-tk gene therapy in combination with ionizing radiation in a mouse mammary cancer model. METHODS AND MATERIALS: Mouse mammary TM40D tumors transplanted s.c. in syngeneic immunocompetent BALB-c mice were treated with either adenoviral-mediated HSV-tk gene therapy or local radiation or the combination of gene and radiation therapy. A vector consisting of a replication-deficient (E1-deleted) adenovirus type 5 was injected intratumorally to administer the HSV-tk gene, and GCV was initiated 24 h later for a total of 6 days. Radiation was given as a single dose of 5 Gy 48 h after the HSV-tk injection. A metastatic model was developed by tail vein injection of TM40D cells on the same day that the s.c. tumors were established. Systemic antitumor effect was evaluated by counting the number of lung nodules after treating only the primary tumors with gene therapy, radiation, or the combination of gene and radiation therapy. To assess the therapeutic efficacy of multiple courses of this combinatorial approach, one, two, and three courses of HSV-tk + GCV gene therapy, in combination with radiation, were compared to HSV-tk or XRT alone and to sham-treated animals. (Treatments were repeated at 7-day intervals from the HSV-tk injection.) RESULTS: Both single-therapy modalities reduced tumor growth by 11% compared to controls, while the combined therapy resulted in a decrease of 29%. Median survival was 36 days in the combined therapy group, compared to 33 days in the monotherapy groups and 26 days in the control group. In the metastatic model, the number of lung nodules was reduced by 59.5% after HSV-tk gene therapy, whereas radiotherapy had no effect on metastatic growth. Combined therapy led to an additional 66.7% reduction in lung colonization. Compared to controls, local tumor growth was maximally suppressed by three courses of combined therapy (51.5%), followed by two courses of combined therapy (37.2%), and three sessions of XRT alone (35.6%). Median survival was also significantly prolonged to 58 days with the three courses of combined therapy, followed by two courses, to 45 days. All other treatment groups demonstrated median survival times between 26 and 35 days, while controls had a median survival of 24 days. CONCLUSIONS: These results indicate that multiple courses of HSV-tk therapy in combination with radiation improve the therapeutic efficacy of this approach and may provide therapeutic implications for the treatment of human breast cancer and other solid tumors.


Subject(s)
Antiviral Agents/therapeutic use , Ganciclovir/therapeutic use , Genetic Therapy/methods , Herpes Simplex/genetics , Mammary Neoplasms, Experimental/therapy , Thymidine Kinase/genetics , Adenoviridae , Animals , Antiviral Agents/administration & dosage , Combined Modality Therapy , Ganciclovir/administration & dosage , Genetic Vectors/therapeutic use , Herpes Simplex/enzymology , Lung Neoplasms/secondary , Mammary Neoplasms, Experimental/radiotherapy , Mice , Mice, Inbred BALB C , Radiobiology , Radiotherapy Dosage , Survival Analysis , Tumor Cells, Cultured
19.
J Natl Cancer Inst ; 93(13): 998-1007, 2001 Jul 04.
Article in English | MEDLINE | ID: mdl-11438565

ABSTRACT

BACKGROUND: Immunization with modified tumor cells carrying recombinant immunomodulatory genes is being explored as cancer immunotherapy. In this study, we examine whether canarypox ALVAC viruses carrying immunostimulatory cytokine genes (granulocyte-macrophage colony-stimulating factor, interleukin 2, interleukin 12, and tumor necrosis factor-alpha) can induce antitumor immunity (to rechallenge) in the RM-1 model of a highly aggressive, weakly immunogenic murine prostate cancer. METHODS: For antitumor activity studies, RM-1 murine prostate cancer cells were infected with the parental ALVAC virus or one or two recombinant ALVAC-cytokine viruses and then injected into male C57BL/6 mice. For rechallenge studies, other mice were first given an injection subcutaneously with irradiated (nonproliferating) recombinant ALVAC-infected RM-1 cells and then (10 days later) with untreated RM-1 cells. For the determination of which immune cells were required for antitumor activity, mice were immunodepleted of CD4, CD8, or natural killer (NK) NK1.1 cells with the corresponding monoclonal antibodies and were then given an injection of ALVAC-cytokine-infected RM-1 cells. For all experiments, tumor outgrowth and animal survival were monitored. RESULTS: After subcutaneous injection into mice, RM-1 cells infected with one (except ALVAC-interleukin 2) or two ALVAC-cytokine recombinants had statistically significantly greater antitumor activity than RM-1 cells infected with parental ALVAC (P<.001 for all; two-sided test). The antitumor activity of RM-1 cells infected with any two ALVAC-cytokine recombinants was greater than, but not statistically significantly different from, that of RM-1 cells infected with any one ALVAC-cytokine recombinant. NK1.1 cells were necessary for antitumor activity, but tumor-specific CD4(+) regulatory T cells were also induced that inhibited CD8(+) RM-1-specific cytotoxic T cells, resulting in the lack of immunity to a rechallenge by RM-1 cells. DISCUSSION: Canarypox viruses can transfer immunostimulatory cytokine genes into RM-1 prostate cancer cells. When such cells were injected into mice, the cytokines induced an antitumor response against this highly aggressive, weakly immunogenic tumor. This response, however, did not protect the mouse against a rechallenge with RM-1 cells because suppressor CD4(+) T cells were induced that inhibited tumor-specific CD8(+) cytotoxic T cells.


Subject(s)
Avipoxvirus/genetics , Prostatic Neoplasms/therapy , Proteins , Animals , Antibodies, Monoclonal/metabolism , Antigens/biosynthesis , Antigens, Ly , Antigens, Surface , CD4 Antigens/biosynthesis , CD8 Antigens/biosynthesis , Flow Cytometry , Gene Transfer Techniques , Genetic Therapy , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Interleukin-12/genetics , Interleukin-2/genetics , Lectins, C-Type , Male , Mice , Mice, Inbred C57BL , NK Cell Lectin-Like Receptor Subfamily B , Neoplasm Transplantation , Prostatic Neoplasms/immunology , Protein Biosynthesis , Recombinant Proteins/metabolism , T-Lymphocytes, Cytotoxic/metabolism , Time Factors , Tumor Cells, Cultured , Tumor Necrosis Factor-alpha/genetics
20.
Oncogene ; 20(31): 4169-79, 2001 Jul 12.
Article in English | MEDLINE | ID: mdl-11464283

ABSTRACT

Deletions in the 8p21-22 region of the human genome are among the most common genetic alterations in prostate carcinomas. Several studies in different tumor tissues, including prostate, indicate that there are probably multiple tumor suppressor genes (TSGs) present in this region. To identify candidate TSGs on 8p22 a YAC contig spanning this region was assembled and YAC clones retrofitted with a selectable marker (neo) were transferred into rat prostate AT6.2 cells. Two overlapping YAC clones showed greatly reduced colony-forming efficiency, indicating they may carry a TSG. Two BAC clones encompassing the overlapping region also appeared to exert suppressive effects on the growth of AT6.2 cells. Database searches for genes mapped to the critical region identified a gene known as FEZ1 (LZTS1) as a potential candidate suppressor gene. Subsequent experiments showed that over-expression of LZTS1 cDNA inhibited stable colony-forming efficiencies of AT6.2, HEK-293 and LNCaP cells. In contrast, LZTS1-transfected Rat-1 and RM1 cells were growth-stimulated. Database searches also identified additional isoforms of the LZTS1 mRNA, as well as LZTS1 protein domains reminiscent of those found in transcription factors. Together these data suggest that the LZTS1 gene is involved in the regulation of cell growth and its loss of function may contribute to the development of prostatic carcinomas, as well as other cancers.


Subject(s)
Chromosomes, Human, Pair 8 , Genes, Tumor Suppressor , Oncogenes , Prostatic Neoplasms/genetics , Animals , Cell Division , Cell Line , Chromosome Mapping , Chromosomes, Artificial, Bacterial , Chromosomes, Artificial, Yeast , Exons , Humans , Male , Plasmids , Prostate/cytology , Prostatic Neoplasms/pathology , Rats , Reverse Transcriptase Polymerase Chain Reaction , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL